Share this post on:

S PK2-induced migration of myeloid cells from both human and murine origins. To assess the impact of PKRA7 on migration/infiltration of mouse macrophages into the microenvironment of xenograft tumors formed by human pancreatic cancer cells, we measured accumulation in the tumors of luciferase-labeled RAW264.7 macrophage cells 24 hours following their IP injection into the nude mice 30 days after subcutaneous implantation of AsPc-1 cancer cells. As shown in GGTI298 chemical information Figure 3E, a significant decrease in luminescent signal emitted by the mouse macrophage cells was observed in mice treated with PKRA7 in comparison to that of the control mice. These results indicate that PKRA7 is able to block macrophage migration/infiltration into the tumor site in an in vivo setting, thus inhibiting the ability of the macrophages to Genz-644282 chemical information positively 23115181 contribute to the growth of xenograft tumors. To further examine the mechanism by which PKRA7 blocks PK2-induced macrophage migration, we performed a cytokine array using quantitative-real time PCR on THP-1 cells that were induced to differentiate into macrophage cells. Among an array of 95 human chemokine/cytokine ligands and their receptors, five displayed a significant induction in their expression after treatment with PK2 including CCL27, CCR10, CCR4, CCR5, and CCR6 (Figure S4). Importantly, at least four of these induced molecules are known to be involved in enhancing the migration of myeloid cells and all of their induction by PK2 was blunted by PKRA7 (Figure 3F), strongly suggesting that suppression of the PK2induced production of these chemokines and receptors underlies the primary mechanism of anti-tumor activity of PKRA7 in the context of pancreatic cancer.PKRA7 Enhances the Efficacy of Standard Therapies for Glioblastoma and Pancreatic Cancer in Xenograft ModelsAlthough PKRA7 displayed strong anti-tumor activities in the contexts of both glioblastoma and pancreatic cancer, it is unlikely to be developed into a therapeutic agent used alone. To test whether PKRA7 could increase the efficacy of standard chemotherapeutic treatment for glioblastoma, we examined the effect of this compound in combination with temozolomide that is currently used in the clinic for this disease [26?7]. Following an established experimental procedure for evaluating the effect of combinational therapy in xenograft mouse models [28?9], 16104 D456MG cells were implanted intracranially and followed by treatment with 10 mg/kg temozolomide for five days, and then with or without PKRA7 administration during the remaining days of the experiments. As shown in Figure 4A, treatment with 1662274 both temozolomide and PKRA7 prolonged the onset of neurological signs of tumor burden compared to mice receiving control, temozolomide or PKRA7 alone, indicating an enhanced effect of combinational therapy with the agents in inhibiting intracranial glioma growth in nude mice (mean survival of 49.8 days forPK2/Bv8/PROK2 Antagonist Suppresses TumorigenesisFigure 2. PKRA7 decreases subcutaneous pancreatic cancer xenograft tumor growth. (A) AsPc-1 cells were SC injected into nude mice, and control (n = 4) or PKRA7 (n = 5) treatment was commenced when tumors became visually detectable (9 days). Measurements were taken every 2?3 days. (B) Average tumor weight of control and PKRA7-treated mouse tumors after removal (*p#0.05). (C) Representative H E slides from control and PKRA7 treated tumors. (D) Quantification of necrotic regions from 5 slides of each tumor per treatment gro.S PK2-induced migration of myeloid cells from both human and murine origins. To assess the impact of PKRA7 on migration/infiltration of mouse macrophages into the microenvironment of xenograft tumors formed by human pancreatic cancer cells, we measured accumulation in the tumors of luciferase-labeled RAW264.7 macrophage cells 24 hours following their IP injection into the nude mice 30 days after subcutaneous implantation of AsPc-1 cancer cells. As shown in Figure 3E, a significant decrease in luminescent signal emitted by the mouse macrophage cells was observed in mice treated with PKRA7 in comparison to that of the control mice. These results indicate that PKRA7 is able to block macrophage migration/infiltration into the tumor site in an in vivo setting, thus inhibiting the ability of the macrophages to positively 23115181 contribute to the growth of xenograft tumors. To further examine the mechanism by which PKRA7 blocks PK2-induced macrophage migration, we performed a cytokine array using quantitative-real time PCR on THP-1 cells that were induced to differentiate into macrophage cells. Among an array of 95 human chemokine/cytokine ligands and their receptors, five displayed a significant induction in their expression after treatment with PK2 including CCL27, CCR10, CCR4, CCR5, and CCR6 (Figure S4). Importantly, at least four of these induced molecules are known to be involved in enhancing the migration of myeloid cells and all of their induction by PK2 was blunted by PKRA7 (Figure 3F), strongly suggesting that suppression of the PK2induced production of these chemokines and receptors underlies the primary mechanism of anti-tumor activity of PKRA7 in the context of pancreatic cancer.PKRA7 Enhances the Efficacy of Standard Therapies for Glioblastoma and Pancreatic Cancer in Xenograft ModelsAlthough PKRA7 displayed strong anti-tumor activities in the contexts of both glioblastoma and pancreatic cancer, it is unlikely to be developed into a therapeutic agent used alone. To test whether PKRA7 could increase the efficacy of standard chemotherapeutic treatment for glioblastoma, we examined the effect of this compound in combination with temozolomide that is currently used in the clinic for this disease [26?7]. Following an established experimental procedure for evaluating the effect of combinational therapy in xenograft mouse models [28?9], 16104 D456MG cells were implanted intracranially and followed by treatment with 10 mg/kg temozolomide for five days, and then with or without PKRA7 administration during the remaining days of the experiments. As shown in Figure 4A, treatment with 1662274 both temozolomide and PKRA7 prolonged the onset of neurological signs of tumor burden compared to mice receiving control, temozolomide or PKRA7 alone, indicating an enhanced effect of combinational therapy with the agents in inhibiting intracranial glioma growth in nude mice (mean survival of 49.8 days forPK2/Bv8/PROK2 Antagonist Suppresses TumorigenesisFigure 2. PKRA7 decreases subcutaneous pancreatic cancer xenograft tumor growth. (A) AsPc-1 cells were SC injected into nude mice, and control (n = 4) or PKRA7 (n = 5) treatment was commenced when tumors became visually detectable (9 days). Measurements were taken every 2?3 days. (B) Average tumor weight of control and PKRA7-treated mouse tumors after removal (*p#0.05). (C) Representative H E slides from control and PKRA7 treated tumors. (D) Quantification of necrotic regions from 5 slides of each tumor per treatment gro.

Share this post on:

Author: Cholesterol Absorption Inhibitors