6 research outputs found

    AICAR suppressed proliferation and induced apoptosis of retinoblastoma in vivo.

    No full text
    <p>(A, B) Immunofluorescent analysis for Ki67 of tumors of Y79 cells isolated from control mice (A) and AICAR-treated mice (B). Nuclei were stained with propidium iodide (red). (C) Quantitative analysis of Ki67 (+) cells/PI (+) cells ratio in tumors. Values are significantly lower in the AICAR-treated mice group than in the control mice group (**p<0.01). (D,E) Apoptotic cells in retinoblastoma xenografts. Typical photomicrographs of apoptotic cells using TUNEL assay (green) in Y79 xenografts. Nuclei were stained with propidium iodide (red). Y79 cells isolated from control mice (D) and AICAR-treated mice (E). (F) Quantitative analysis of the apoptotic cell percentage in tumors. Note that the number of TUNEL (+) cells was significantly higher in the AICAR-treated mice group than in the control mice group (**p<0.01). Each column represents the mean ± SEM. Scale bars (A, B, D, E), 200 µm.</p

    AICAR does not alter the levels of cyclins A, D and E in retinoblastoma in vivo.

    No full text
    <p>Quantitative RT-PCR analysis of tumors treated with AICAR in comparison with control shows no significant difference. Each column represents the mean ± SEM.</p

    Proposed mechanism of action for AICAR in human retinoblastoma in an in-vivo xenograft model.

    No full text
    <p>AICAR administration leads to activation of AMPK decreased tumor vessel density and decreased CD11b (macrophage) infiltration. Activated AMPK inhibits mTOR pathway, protein synthesis and cell growth. In addition, AICAR administration results in decreased levels of p21, which was recently found to inhibit apoptosis and promote cell proliferation. Overall signaling changes leads to loss of viability due to apoptosis, proliferation block and inhibition of tumor progression.</p

    AICAR treatment of retinoblastoma is associated with activation of AMPK, inhibition of mTORC1 and decrease of p21.

    No full text
    <p><b>A.</b> AICAR treatment of retinoblastoma is associated with activation of AMPK. Western blot analysis of phosphorylated ACC (Ser-79) (a downstream effector of AMPK) showed significant increase of pACC in tumours from AICAR treated mice comparing to control (**p<0.01, n = 19). <b>B and C.</b> Treatment with AICAR resulted in the inhibition of the mTORC1 pathway. Western blot analysis of tumor xenografts harvested from mice treated with AICAR showed significant decrease of mTOR pathway downstreams, pS6RP (Ser235/236) and the p4E-BP1 (Ser65) when compared to PBS-treated mice (***p<0.001 for both, n = 17 for pS6RP and n = 23 for p4EBP1). <b>D.</b> AICAR down-regulates p21WAF1/Cip1 in AICAR treated tumors as shown via Western blot analysis (*p<0.05, n = 23). Density values bands are graphically expressed relative to control. GAPDH was used as a loading control in all panels. Multiple bands represent separate biological samples. Each column represents the mean ± SEM.</p

    AICAR suppressed tumor angiogenesis and inflammatory cells infiltration.

    No full text
    <p>(A, B) Microvessel density in tumor tissues was determined by immunofluorescent staining by an endothelial-specific antibody CD31. (A) Control group and (B) AICAR-treated group. (C) Quantitative analysis of fluorescent-positive area (per 4000 µm<sup>2</sup>) in tumors. Vessel density was significantly suppressed in AICAR-treated mice group (**p<0.01). (D, E) Macrophage- and neutrophil- infiltration in Y79 xenografts. Typical photomicrographs of immunofluorescent staining for CD11b (red) in Y79 xenografts. Nuclei were stained with propidium iodide (blue). Y79 cells isolated from control mice (D) and AICAR-treated mice (E). (F) Quantitative analysis of the CD11b (+) cells/DAPI (+) cells ratio in tumors. The number of CD11b (+) cells was significantly lower in the AICAR-treated mice group than in the control mice group (**p<0.01). Each column represents the mean ± SEM. Scale bars (A, B, D, E), 200 µm.</p

    Dysregulated claudin-5 cycling in the inner retina causes retinal pigment epithelial cell atrophy

    No full text
    Age-related macular degeneration (AMD) is the leading cause of central retinal vision loss worldwide, with an estimated 1 in 10 people over the age of 55 showing early signs of the condition. There are currently no forms of therapy available for the end stage of dry AMD, geographic atrophy (GA). Here, we show that the inner blood-retina barrier (iBRB) is highly dynamic and may play a contributory role in GA development. We have discovered that the gene CLDN5, which encodes claudin-5, a tight junction protein abundantly expressed at the iBRB, is regulated by BMAL1 and the circadian clock. Persistent suppression of claudin-5 expression in mice exposed to a cholesterolenriched diet induced striking retinal pigment epithelium (RPE) cell atrophy, and persistent targeted suppression of claudin-5 in the macular region of nonhuman primates induced RPE cell atrophy. Moreover, fundus fluorescein angiography in human and nonhuman primate subjects showed increased retinal vascular permeability in the evening compared with the morning. These findings implicate an inner retina-derived component in the early pathophysiological changes observed in AMD, and we suggest that restoring the integrity of the iBRB may represent a novel therapeutic target for the prevention and treatment of GA secondary to dry AMD
    corecore