13 research outputs found

    Targeting intracellular B2 receptors using novel cell-penetrating antagonists to arrest growth and induce apoptosis in human triple-negative breast cancer

    Get PDF
    G protein-coupled receptors (GPCRs) are integral cell-surface proteins having a central role in tumor growth and metastasis. However, several GPCRs retain an atypical intracellular/nuclear location in various types of cancer. The pathological significance of this is currently unknown. Here we extend this observation by showing that the bradykinin B2R (BK-B2R) is nuclearly expressed in the human triple-negative breast cancer (TNBC) cell line MDA-MB-231 and in human clinical specimens of TNBC. We posited that these "nuclearized" receptors could be involved in oncogenic signaling linked to aberrant growth and survival maintenance of TNBC. We used cell-penetrating BK-B2R antagonists, including FR173657 and novel transducible, cell-permeable forms of the peptide B2R antagonist HOE 140 (NG68, NG134) to demonstrate their superior efficacy over impermeable ones (HOE 140), in blocking proliferation and promoting apoptosis of MDA-MB-231 cells. Some showed an even greater antineoplastic activity over conventional chemotherapeutic drugs in vitro. The cell-permeable B2R antagonists had less to no anticancer effects on B2R shRNA-knockdown or non-B2R expressing (COS-1) cells, indicating specificity in their action. Possible mechanisms of their anticancer effects may involve activation of p38kinase/p27Kip1pathways. Together, our data support the existence of a possible intracrine signaling pathway via internal/nuclear B2R, critical for the growth of TNBC cells, and identify new chemical entities that enable to target the corresponding intracellular GPCRs

    Induction of Selective Blood-Tumor Barrier Permeability and Macromolecular Transport by a Biostable Kinin B1 Receptor Agonist in a Glioma Rat Model

    Get PDF
    Treatment of malignant glioma with chemotherapy is limited mostly because of delivery impediment related to the blood-brain tumor barrier (BTB). B1 receptors (B1R), inducible prototypical G-protein coupled receptors (GPCR) can regulate permeability of vessels including possibly that of brain tumors. Here, we determine the extent of BTB permeability induced by the natural and synthetic peptide B1R agonists, LysdesArg9BK (LDBK) and SarLys[dPhe8]desArg9BK (NG29), in syngeneic F98 glioma-implanted Fischer rats. Ten days after tumor inoculation, we detected the presence of B1R on tumor cells and associated vasculature. NG29 infusion increased brain distribution volume and uptake profiles of paramagnetic probes (Magnevist and Gadomer) at tumoral sites (T1-weighted imaging). These effects were blocked by B1R antagonist and non-selective cyclooxygenase inhibitors, but not by B2R antagonist and non-selective nitric oxide synthase inhibitors. Consistent with MRI data, systemic co-administration of NG29 improved brain tumor delivery of Carboplatin chemotherapy (ICP-Mass spectrometry). We also detected elevated B1R expression in clinical samples of high-grade glioma. Our results documented a novel GPCR-signaling mechanism for promoting transient BTB disruption, involving activation of B1R and ensuing production of COX metabolites. They also underlined the potential value of synthetic biostable B1R agonists as selective BTB modulators for local delivery of different sized-therapeutics at (peri)tumoral sites

    NG29 increases transvascular delivery, distribution and accumulation of Gadomer within brain tumor tissues of F98-bearing rats.

    No full text
    <p>(A) Representative axial Gadomer-enhanced <i>T</i><sub>1</sub>-weighted MR images depicting the brain of an F98-implanted rat before and after intracarotid NG29 treatment (10 nmol/kg/min for 5 min i.c.) (upper panels). Note the increase of the signal intensity at the tumor (white arrows). CADV in function of time calculated from the corresponding set of images (1 image/51 s for 50 min) (bottom panel). (B) Dose-, B1R-, PGs-dependence of NG29-induced BTB permeability. Numbers in parenthesis represent infusion rates in nmol/kg/min, for 5 min. The B2R antagonist HOE140 and the B1R antagonist R892 (both at 20 nmol/kg/min, for 5 min, i.c.) were infused simultaneously with NG29 while the non-selective nitric oxide synthase (NOS) inhibitors L-NA (5 mg/kg, i.v.) and L-NAME (20 mg/kg, i.v.), and the non-selective cyclooxygenase (COX) inhibitors Meclofenamate (5 mg/kg, i.v.) and Indomethacin (2.5 mg/kg, i.v.) were administered 30 min before the infusion of NG29. Note the effectiveness of NG29 administered by the i.v. (intrajugular) route. <sup>+</sup>p<0.05 compared to vehicle-treated animals; *p<0.05 compared to NG29 (10 nmol/kg/min)-treated animals. (C) Duration of the increase in BTB permeability caused by NG29 as detemined by relative CADV values. Gadomer was injected 3 min, 60 or 120 min following the start of the infusion (10 nmol/kg/min) of NG29 over 5 min. Each bar represents the mean ± S.E.M. for 3 to 7 animals. *p<0.05 compared to respective vehicle-treated animals. (D) Representative time course of Gadomer uptake in the ipsilateral (tumor-implanted) hemisphere and the contralateral hemisphere, before and after treatment with NG29 (10 nmol/kg/min for 5 min) (left panel). Histographic representation of average maximal Gadomer concentrations in the ipsilateral (tumor-implanted) and the contralateral hemispheres, and the facial muscle following NG29 treatment (right panel). As observed for Gd-DTPA, levels of Gadomer were higher in the ipsi- (tumors) than in contralateral (normal) tissues in CTL animals (not illustrated, ***p<0.001). *p<0.05 compared to untreated (Gadomer alone) ipsilateral groups. Value represents the mean ± S.E.M. obtained from 3 animals.</p

    Overexpression of B1R in human glioma tissues.

    No full text
    <p>(<b>A</b>) Detection of B1R in various human glioma cell lines and nontransformed counterparts, normal human astrocytes, by WB analysis using the anti-B1R antibody RC72. β-actin serves as a loading control in the lower panel. The doublet band in U138-MG cells may indicate degradation of B1R. (<b>B</b>) Comparative quantification of B1R mRNA levels among normal and glioma brain tissue samples was normalized against that of the corresponding β-actin. (<b>C</b>) Expression of B1R in normal versus tumoral tissue specimens (left panel) or in paired primary glioma (T) and peritumoral tissue biopsies (P) (right panel), with each pair obtained from a same patient. Western blot analysis was performed using the anti-B1R antiserum AS434. (<b>D</b>) Representative images from IHC assay of paraffin-embedded specimens of primary glioma tissue biopsies (WHO grades II–IV) exposed to pre-immune serum or AS434 antiserum. Optical magnification is indicated in the bottom-right corner of each image.</p

    Systemic infusion of NG29 increases permeability and uptake of albumin within peritumoral tissue.

    No full text
    <p>(<b>A</b>) Direct immunological staining of endogenous albumin in brain tissues from F98-implanted rats treated or not with intracarotid NG29 (50 nmol/kg/min for 5 min). Representative coronal sections of CTL-, vehicle- and NG29-treated rats immunostained with sheep anti-rat albumin HRP conjugated are shown in the upper panels. Scale bar: 1 mm. Histographic representation of integrated optical density (IOD) values (left) and stained surface areas (right) of immunoreactive albumin in respective animal groups (bottom panels). ***p<0.001 vs CTL. (<b>B</b>) Semi-quantitative measurement of Evans blue content (mg/g wet weight tissue) in several tissues after systemic intravenous (femoral) injection of saline vehicle or NG29 (5 µmol/kg i.v.) in F98 glioma bearing rats. Data are presented as means ± S.E.M. n = 5 to 7 animals per group. *p<0.05 vs respective vehicle group.</p

    NG29 enhances transvascular delivery, distribution and accumulation of Magnevist within brain tumor tissues of F98-bearing rats.

    No full text
    <p>(A) MRI contrast-enhanced detection of glial brain tumors in rats at day 3, 5, 7, 10 14 and 17 post-inoculation. Note the rapidly growing tumor over a 2-week time and the appearance of a necrotic center on the 17 day-image (white arrow) outgrowing its blood supply. Assessment of BTB disruption by MRI monitoring was conducted on the same animal on day 10 post-inoculation, corresponding to mid stage development of the tumor. (B) Representative axial Magnevist-enhanced <i>T</i><sub>1</sub>-weighted MR images depicting the brain of an F98-implanted rat before and after NG29 treatment (10 nmol/kg/min for 5 min i.c.) (left panel). Note the increase in the signal intensity at the tumor (white arrows). Temporal CADV calculated from the corresponding sets of images (1 image/51 s for 50 min) (bottom panel). (C) Relative CADV in percent determined following the infusion of the vehicle (saline), LDBK, NG29 (10 nmol/kg/min for 5 min) or NG29 (10 nmol/kg/min, 5 min) + R892 (20 nmol/kg/min, 5 min). Each bar represents the mean ± S.E.M. for 4 to 6 animals. **p<0.01 compared to vehicle-treated animals; <sup>††</sup>p<0.01 compared to NG29-treated animals. (D) Histographic representation of average maximal Gd-DTPA concentrations in the ipsilateral (tumor-implanted) and the contralateral hemispheres following saline vehicle or NG29 treatment (10 nmol/kg/min)). Note the superior levels of Gd-DTPA (reflecting greater basal permeability) in the ipsi- versus contralateral tissues of vehicle-treated animals (not illustrated, ***p<0.001). **p<0.01 compared to vehicle-treated ipsilateral groups. Value represents the mean ± S.E.M. obtained with 3 animals.</p
    corecore