7 research outputs found

    Ratiometric spectral classification images of HerDox-treated tumors <i>in situ:</i> (A) Ratiometric spectral classification images (middle and right-panel) of HerDox-treated tumors.

    No full text
    <p>Ratiometric spectral classification was performed using our previously developed program. Green: autofluorescence (Autofl.), Red: 0.2 Dox+0.8 autofl., Cyan: 0.3 Dox+0.7 autofl., and Blue: 0.1 Dox+0.9 autofl.. The reference spectral signatures used to generate the spectrally classified image are shown in the left-panel. (B) Spectral unmixed image. Scale bar: 80 µm.</p

    Fluorescence intensity image of harvested tissue <i>and in situ</i> confocal fluorescence of tumors: (A) Fluorescence image (left-panel; Ex: 532 nm; Em: 590±30 nm) of organs and tumors extracted from mice receiving HerDox (0.004 mg/kg/day; see Methods).

    No full text
    <p>Confocal fluorescence images of the tumors extracted from HerDox-treated (A, right-panel), Dox-treated (B, right), and untreated mice (B, left) were acquired at different z-depths (a total of thickness: 32 µm) with a step size of 2 µm. Maximum intensity z-projection of confocal fluorescence images of HerDox-treated and untreated tumors was performed. HerDox and Dox fluorescence in (A) and (B) are indicated by a color shift toward 255 on the scale bar in (A). The magnified image (A, right-panel) shows HerDox distribution in the tumor. Arrows indicate nuclei-localized fluorescence. (C) Mean fluorescence intensity of HerDox-treated, Dox-treated and untreated tumor. The error bar indicates standard deviation. *: p<0.01.</p

    TUNEL and H&E stained images of untreated, HerDox-treated, and Dox-treated tumors: (A) TUNEL images of untreated tumors (left), HerDox-treated tumors (middle) (injection drug dosage: 0.004 mg/kg×6 injections), Dox-treated tumors (right) (injection drug dosage: 0.04 mg/kg×6 injections) were acquired using confocal imaging (ex: 488 nm, em: 510–560 nm, and 10×) of tumor specimens.

    No full text
    <p>The arrows indicate the boundary between apoptotic and normal cells. Insets represent magnified images. (B) Average fluorescence of the TUNEL images of untreated, Dox-treated, HerDox-treated tumor sections (*: p<0.01) (C) H&E images of untreated (left), HerGa-treated tumors (middle), and Dox-treated tumors (right) were obtained using the Olympus microscope incorporating a CCD camera with a RGB filter (10×). The scale bar represents 200 µm. Arrows indicate nuclear shrinkage and fragmentation.</p

    A Mechanistic Study of Tumor-Targeted Corrole Toxicity

    No full text
    HerGa is a self-assembled tumor-targeted particle that bears both tumor detection and elimination activities in a single, two-component complex (Agadjanian et al. <i>Proc. Natl. Acad. Sci. U.S.A.</i> <b>2009</b>, <i>106</i>, 6105–6110). Given its multifunctionality, HerGa (composed of the fluorescent cytotoxic corrole macrocycle, S2Ga, noncovalently bound to the tumor-targeted cell penetration protein, HerPBK10) has the potential for high clinical impact, but its mechanism of cell killing remains to be elucidated, and hence is the focus of the present study. Here we show that HerGa requires HerPBK10-mediated cell entry to induce toxicity. HerGa (but not HerPBK10 or S2Ga alone) induced mitochondrial membrane potential disruption and superoxide elevation, which were both prevented by endosomolytic-deficient mutants, indicating that cytosolic exposure is necessary for corrole-mediated cell death. A novel property discovered here is that corrole fluorescence lifetime acts as a pH indicator, broadcasting the intracellular microenvironmental pH during uptake in live cells. This feature in combination with two-photon imaging shows that HerGa undergoes early endosome escape during uptake, avoiding compartments of pH < 6.5. Cytoskeletal disruption accompanied HerGa-mediated mitochondrial changes whereas oxygen scavenging reduced both events. Paclitaxel treatment indicated that HerGa uptake requires dynamic microtubules. Unexpectedly, low pH is insufficient to induce release of the corrole from HerPBK10. Altogether, these studies identify a mechanistic pathway in which early endosomal escape enables HerGa-induced superoxide generation leading to cytoskeletal and mitochondrial damage, thus triggering downstream cell death
    corecore