13 research outputs found

    APOPTOSIS INDUCTION OF CENTELLA ASIATICA ON HUMAN BREAST CANCER CELLS

    Get PDF
    The present study evaluated the ability of methanolic extract of Centella asiatica (Linn) Urban (Umbelliferae) to induce apoptosis in different cancer cell lines. MCF-7 cells emerged as the most sensitive cell line for in vitro growth inhibitory activity. C. asiatica extract induced apoptosis in MCF-7 cells as indicated by nuclear condensation, increased annexin staining, loss of mitochondrial membrane potential and induction of DNA breaks identified by TUNEL reactivity. It is possible that the use of C. asiatica extract as a component in herbal medicines could be justifiable

    Supplementary Material for: Aloe Emodin Induces G2/M Cell Cycle Arrest and Apoptosis via Activation of Caspase-6 in Human Colon Cancer Cells

    No full text
    Aloe emodin (AE), a natural anthraquinone, is reported to have antiproliferative activity in various cancer cell lines. In this study, we analyzed the molecular mechanisms involved in the growth-inhibitory activity of this hydroxyanthraquinone in colon cancer cell, WiDr. In our observation AE inhibited cell proliferation by arresting the cell cycle at the G2/M phase and inhibiting cyclin B1. AE appreciably induced cell death specifically through the induction of apoptosis and by activating caspases 9/6. Apoptotic execution was found to be solely dependent on caspase-6 rather than caspase-3 or caspase-7. This is the first study indicating that the AE induces apoptosis specifically through the activation of caspase-6

    Apoptosis Induction Of Centella Asiatica On Human Breast Cancer Cells

    No full text
    The present study evaluated the ability of methanolic extract of Centella asiatica (Linn) Urban (Umbelliferae) to induce apoptosis in different cancer cell lines. MCF-7 cells emerged as the most sensitive cell line for in vitro growth inhibitory activity. C. asiatica extract induced apoptosis in MCF-7 cells as indicated by nuclear condensation, increased annexin staining, loss of mitochondrial membrane potential and induction of DNA breaks identified by TUNEL reactivity. It is possible that the use of C. asiatica extract as a component in herbal medicines could be justifiable

    Metformin reduces ECM remodeling, EMT, and metastasis in a PDAC mouse model.

    No full text
    <p>(A) Expression of genes associated with extra-cellular matrix (ECM) remodeling, epithelial-to-mesenchymal transition (EMT) and inflammation in AK4.4 tumors from control and metformin-treated mice. Data normalized to control group. 3–4 samples per group pooled in one single PCR array plate. Metformin reduces the expression of pro-tumor genes and increases the expression of anti-tumor genes. (B-i) Representative Western blots showing the effect of metformin (300 mg/Kg) on MMPs and EMT markers in AK4.4 tumors. (B-ii) Densitometric analysis of protein expression normalized to ß-actin. Metformin decreases the expression of MMP-9 and vimentin and increases the expression of e-cadherin in AK4.4 tumors. (C) MMP activity in AK4.4 tumor protein extracts from control and metformin-treated mice (n = 3–4). Metformin decreases the activity of MMPs. (D) Effect of metformin on the percentage of mice affected (incidence) with mesenteric (peritoneal) and abdominal wall (retroperitoneal) metastasis in AK4.4 and PAN02 models (n = 3–8). Metformin reduced the percentage of mice that develop wall metastasis in the more metastatic model (PAN02 model) and induced a tendency for reduced wall as well as mesenteric metastasis in the less metastatic AK4.4 model. (E) Effect of metformin on the number (average) of mesenteric (peritoneal) and abdominal wall (retroperitoneal) metastasis per mouse in the AK4.4 and PAN02 models (n = 3–8). Metformin reduced the number of wall metastasis in the PAN02 model. There were also trends for fewer mesenteric metastasis in AK4.4 and PAN02 tumors. Data in B, C and E are presented as the mean ± standard error. *p < 0.05 vs. control.</p

    Metformin treatment associates with reduced hyaluronan levels in human pancreatic cancers in overweight/obese patients.

    No full text
    <p>(i) Representative histology images showing the effect of metformin on tumor hyaluronan levels in normal weight [Body mass index (BMI)<25)] or overweight/obese patients (BMI>25) (n = 22 controls, 7 metformin). (ii) Immunohistochemical analysis of total tumor hyaluronan levels. Metformin decreases the hyaluronan-positive area fraction (%) in overweight/obese patients. Data are presented as the mean ± standard error. * p < 0.05 vs. control in patients with BMI >25.</p

    Metformin reprograms TAMs and reduces inflammation in tumors.

    No full text
    <p>(A-i) Representative immunocytochemistry images showing the effect of metformin metformin on the expression of F4/80 (immunofluorescence) in AK4.4 tumors (percentage of viable tumor area) (n = 4). (A-ii) Metformin-treated tumors (300 mg/kg in drinking water) had significantly reduced levels of F4/80-positive tumor-associated macrophages (TAMs). (B) Effect of metformin (0–0.2mM) on the gene expression (qPCR) of M1 (i) and M2 (ii) markers in RAW 264.7 cells (mouse leukaemic monocyte-macrophages) <i>in vitro</i>. Clinically relevant doses (0.05 mM) of metformin treatment reduces expression of M2 markers in macrophages <i>in vitro</i>, including Arg-1 and IL-10. (C) Effect of metformin on the gene expression (qPCR) of M1 (i) and M2 (ii) markers in TAMs isolated from PAN02 tumors <i>in vivo</i> (n = 3). Metformin treatment reduced expression of the M2 markers Arg-1 and IL-10 in TAMs <i>in vivo</i>. (D) Representative Western blots for the expression of signaling proteins in RAW 264.7 cells treated with metformin at 0, 0.05, 0.1, 0.2 and 0.4 mM. Metformin decreases the activation of signaling pathways and increased activation of AMPKα on RAW cells. Densitometric analysis of protein expression normalized to ß-actin or total protein (in the case of phosphorylated protein) is depicted as numbers below the representative bands. (E) Effect of metformin on the protein expression of major cytokines in AK4.4 tumors (n = 4–5) using multiplex protein array. Metformin treatment associated with reduced IL-1ß and CXCL1 expression in tumors. Data are presented as mean ± standard error in A, C and E. * p < 0.05, ** p < 0.01 vs. control.</p

    Metformin reprograms pancreatic stellate cells (PSCS) and tumor-associated macrophages (TAMs), alleviates the fibro-inflammatory tumor microenvironment and reduces metastasis.

    No full text
    <p>Metformin treatment reduces collagen-I and HA production by PSCs, leading to decreased fibrosis in PDACs. Metformin treatment also reduces cytokine production, infiltration and M2 polarization of TAMs, leading to decreased inflammation. This associated with improved desmoplasia and reduced extracellular matrix (ECM) remodeling, epithelial-to-mesenchymal transition (EMT), and metastasis.</p

    Metformin reduces collagen-I/hyaluronan production by pancreatic stellate cells.

    No full text
    <p>(A) PSCs were incubated <i>in vitro</i> with metformin (1 mM) for 48h. (A-i) Representative immunocytochemistry images showing the effect of metformin on tumor hyaluronan and collagen-I levels in human pancreatic stellate cells (PSCs) <i>in vitro</i> (n = 2). (A-ii) Quantification of hyaluronan expression in PSCs. Metformin decreases the expression of hyaluronan in PSCs. (A-iii) Quantification of the expression of collagen-I in PSCs. Metformin decreases the expression of collagen-I in PSCs. αSMA denotes activated PSCs. (B) Representative Western blots for the expression of fibrosis-related markers and signaling proteins in PSCs treated with metformin at 0, 0.1, 1 and 10mM. Metformin decreases the expression of fibrosis-related markers and signaling proteins in PSCs. Densitometric analysis of protein expression normalized to ß-actin or total protein (in the case of phosphorylated proteins) is depicted as numbers below the representative bands. Data in A are presented as the mean ± standard error. *p < 0.05 vs. control.</p

    PlGF/VEGFR-1 Signaling Promotes Macrophage Polarization and Accelerated Tumor Progression in Obesity

    No full text
    Obesity promotes pancreatic and breast cancer progression via mechanisms that are poorly understood. Although obesity is associated with increased systemic levels of placental growth factor (PlGF), the role of PlGF in obesity-induced tumor progression is not known. PlGF and its receptor VEGFR-1 have been shown to modulate tumor angiogenesis and promote tumor-associated macrophage (TAM) recruitment and activity. Here, we hypothesized that increased activity of PlGF/VEGFR-1 signaling mediates obesity-induced tumor progression by augmenting tumor angiogenesis and TAM recruitment/activity.status: publishe
    corecore