APCK110, a Novel and Potent Inhibitor of c-Kit, Blocks Phosphorylation of AKT and STAT3, Induces Apoptosis, and Inhibits Proliferation of Acute Myeloid Leukemia (AML) Cells.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 153-153 ◽  
Author(s):  
Stefan Faderl ◽  
William Bornmann ◽  
David Maxwell ◽  
Ashutosh Pal ◽  
Zheng-Hong Peng ◽  
...  

Abstract Tight control of protein tyrosine kinase (TK) activity is crucial for the regulation and maintenance of vital cellular functions such as proliferation, differentiation, and apoptosis. c-KIT is a TK and transmembrane receptor for stem cell factor (SCF). Binding of SCF to c-KIT results in activation of marrow precursors and other blood cells. Activating mutations of c-KIT associated with amino acid Asp-816 (D816) have been identified in leukemic cells of patients with AML and are thought to play an important pathophysiologic role in leukemogenesis. Identification of activating c-KIT mutations and development of novel compounds targeting these mutations may therefore be of therapeutic benefit in AML. Based on the 3-dimensional structure of c-KIT we have generated a number of compounds with activity against c-KIT mutated cells. Here we present initial results of the activity and mechanism of action of the novel c-KIT inhibitor APCK110 in AML cell lines and primary samples from patients with AML. Using an MTT assay, we first studied the antiproliferative effect of APCK110 in the AML cell lines OCI/AML3 and the SCF-responsive cell line OCIM2. Cells were incubated for 72 hours without or with APCK110 at concentrations of 50, 100, 250, and 500 nM, then harvested and their metabolic activity and viability determined as optical density. Next we determined expression of phospho-AKT and -STAT3 in the mastocytosis cell line HMC1.1 and phospho-c-KIT in the AML cell line OCI/AML3 by Western Immunoblotting. We further analyzed induction of caspase 3 and PARP cleavage in OCI/AML3 cells at APCK110 concentrations of 50, 100, 250, and 500 nM using Western Immunoblotting. To demonstrate the effect of APCK110 on primary AML cells, we incubated diagnostic marrow cells from 3 patients with AML with increasing concentrations of APCK110 and used the blast colony assay to measure inhibition of proliferation. We then compared the antiproliferative effect of APCK110 with that of dasatinib and imatinib in OCI/AML3 cells. We show that 1) APCK110 strongly inhibits proliferation of AML cells with 80% inhibition at 500 nM; 2) similar to cell lines, APCK110 also inhibits AML colony growth of primary samples in a dose-dependent manner of up to 80% at 500 nM concentration; 3) APCK110 blocks activation of phospho-AKT, phospho-STAT3, and phospho-c-KIT; 4) APCK110 induces apoptosis by cleavage of caspase 3 and PARP; and 5) APCK110 demonstrates more potent inhibition (up to 100% at 500 nM) of AML cell proliferation than dasatinib (60% at 500 nM) and dasatinib (none at 500 nM). In summary, APCK110 is a novel and potent inhibitor of mutated c-KIT that inhibits AML cell proliferation, blocks activation of intracellular signaling molecules, and induces caspase-dependent apoptosis. Further development of APCK110 for clinical trials of patients with AML should be pursued.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2462-2462
Author(s):  
Stefan Faderl ◽  
Alessandra Ferrajoli ◽  
David Harris ◽  
Quin Van ◽  
Hagop M. Kantarjian ◽  
...  

Abstract Cytokines and growth factors stimulate AML cell proliferation by activating various signaling pathways and high levels of cytokines have been associated with poor prognosis. Blocking signal transduction of cytokine-stimulated pathways may therefore inhibit AML growth and proliferation. Atiprimod is a cationic amphiphilic azaspirane. Although their mechanisms of action are not completely understood, azaspiranes were demonstrated among others to downregulate various cytokine receptors such as for interleukin (IL)-1, IL-2, IL-6, interferon-γ, and tumor necrosis factor-α. Owing to this spectrum of activities, we hypothesized that atiprimod inhibits activation of intracellular signaling pathways in AML cells resulting in apoptosis and growth inhibition. We first studied the antiproliferative effect of atiprimod on 5 AML cell lines (K562, HL-60, KG-1, OCIM2, OCI/AML3) using the MTT assay. Cells were incubated for 72 hours without and with increasing concentrations of atiprimod (1, 2, 3, and 4 μM), then harvested and their metabolic activity and viability determined as optical density measurements. To corroborate the results, we also studied the effect of atiprimod on OCIM2 cells using a clonogenic cell line assay. Next we determined expression of Stat3 and Stat5, as well as Phospho-stat3 and phospho-stat5 in the K562 cells by Western Immunoblotting where cells were incubated in the absence and presence of increasing atiprimod concentrations (0.5, 1, 2, 3, 4 μM). We further evaluated induction of apoptosis of OCIM2 and K562 cells following incubation with atiprimod at 1 and 4 μM using the annexin V-FITC assay and finally analyzed caspase 3 and PARP cleavage in K562 cells at atiprimod concentrations of 0.5, 1, 2, 3, and 4 μM using Western Immunoblotting. To demonstrate the effect of atiprimod on marrow cells from AML patients and healthy volunteers we incubated marrow cells with atprimod at increasing concentrations and used the blast colony assay to measure inhibition of proliferation. Our results demonstrate that: 1) atiprimod inhibits proliferation of AML cell lines and AML blast proliferation from patient samples, but not significantly normal hematopoietic progenitors from samples of healthy controls; 2) atiprimod inhibits phosphorylation of Stat 3 and 5; and 3) atiprimod induces apoptosis in OCIM2 and K562 cells by cleavage of caspase 3 and PARP. In summary, our data suggest that atiprimod inhibits phopshorylation of Stat 3 and 5, induces caspase-dependent apoptosis, and blocks AML cell proliferation. Further evaluation of atiprimod in clinical trials of AML and MDS should be considered.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2528-2528
Author(s):  
Stefan Faderl ◽  
Alessandra Ferrajoli ◽  
David Harris ◽  
Quin Van ◽  
Waldemar Priebe ◽  
...  

Abstract Proliferation and growth of AML cells result from stimulation by cytokines and high levels of cytokines are associated with poor prognosis in AML. Cytokines act through cellular receptors that are associated with members of the Jak family of protein tyrosine kinases. Upon phosphorylation and activation of Jak, proteins bound to Jak initiate signaling pathways including those regulated by Stat proteins. Since constitutive activation of Jak-Stat has been associated with leukemogenesis, we hypothesize that inhibition of Jak-Stat inhibits proliferation of AML cells. To do this, we studied the effects of WP-1034, a novel and potent inhibitor of Jak-Stat, in the OCIM2 AML cell line and fresh samples from AML patients. OCIM2 cells were deprived of serum for 2 hours and then incubated with 1 to 5 μM WP-1034 to investigate its effect on OCIM2 cell proliferation. After incubation of the cells without and with 1, 2.5, 5, 7.5, and 10 μM WP-1034 for 1 hour, and at 5 μM for 0, 20 min, 40 min, and 1, 2, 3, and 4 hours, we determined expression of Stat 1, 3, and 5, as well as Phospho-stat 1, 3, and 5 in the cells by Western Immunoblotting. In addition, we analyzed cell cycle status by PI staining and flow cytometry. We further evaluated induction of apoptosis of OCIM2 cells following incubation with WP-1034 at 3, 5, and 6 μM using the annexin V-CY5 assay and analyzed caspase 3 and PARP cleavage using Western Immunoblotting. To demonstrate the effect of WP-1034 on marrow cells from AML patients and healthy volunteers we incubated marrow cells with WP-1034 at increasing concentrations and used the blast colony assay to measure inhibition of proliferation. Our results show that: i) WP-1034 effectively inhibits proliferation of OCIM2 cells and AML blast proliferation from patient samples; ii) WP-1034 blocks activation of Stat 3 and 5 by decreasing the amount of Phospho-stat 3 and 5 in OCIM2 cells; iii) WP-1034 causes cell cycle arrest in sub-G0 phase and is able to induce apoptosis in OCIM2 cells; and iv) WP-1034 induces apoptosis involving cleavage of caspase 3 and PARP. Our data suggest that WP-1034, a potent inhibitor of Jak-Stat, inhibits proliferation of AML cells by inhibition of Stat 3 and 5 and induction of caspase-dependent apoptosis.


Blood ◽  
1998 ◽  
Vol 92 (9) ◽  
pp. 3381-3387 ◽  
Author(s):  
Nathalie Voorzanger-Rousselot ◽  
M.-C. Favrot ◽  
Jean-Yves Blay

Abstract The modulation of the cytotoxic effects of an anthracyclin by CD40L was investigated in five non-Hodgkin’s lymphoma (NHL) cell lines (Daudi, Raji, BJAB, BL36, BL70). Incubation with doxorubicin (DOX) increased in a dose-dependent manner the percentage of apoptosis in NHL cells. Coculture with irradiated L cells expressing CD40L (CD40L L cells), but not CDw32 (CDw32 L cells), significantly reduced (33% to 89%) the percentage of apoptosis in all five cell lines treated with 0.1 to 0.5 μg/mL of DOX, but in only three cell lines at 1 μg/mL. Interleukin-10 (IL-10), IL-6, IL-2, or tumor necrosis factor (TNF) induced no additive protective effects with CD40L L cells. In all five cell lines, DOX induced a concentration-dependent increase of the activity of the cysteine-protease caspase 3. Coculture with CD40L L cells, but not with CDw32 L cells, inhibited (38% to 100%) the activation of caspase 3 induced by 0.1 to 0.5 μg/mL of DOX in all five NHL cell lines, but in only two cell lines at 1 μg/mL. Finally, the antiproliferative effect of 0.1 to 0.5 μg/mL concentrations of DOX was also partially abrogated on coculture with CD40L L cells in all five cell lines, but in only two cell lines at 1 μg/mL. Cytokines, either alone or in combination with CD40L L cells, did not affect DOX-induced inhibition of proliferation. These results indicate that CD40L inhibits the apoptosis and antiproliferative effect induced by DOX and interferes with caspase 3 activation in B NHL cell lines. © 1998 by The American Society of Hematology.


Blood ◽  
1998 ◽  
Vol 92 (9) ◽  
pp. 3381-3387 ◽  
Author(s):  
Nathalie Voorzanger-Rousselot ◽  
M.-C. Favrot ◽  
Jean-Yves Blay

The modulation of the cytotoxic effects of an anthracyclin by CD40L was investigated in five non-Hodgkin’s lymphoma (NHL) cell lines (Daudi, Raji, BJAB, BL36, BL70). Incubation with doxorubicin (DOX) increased in a dose-dependent manner the percentage of apoptosis in NHL cells. Coculture with irradiated L cells expressing CD40L (CD40L L cells), but not CDw32 (CDw32 L cells), significantly reduced (33% to 89%) the percentage of apoptosis in all five cell lines treated with 0.1 to 0.5 μg/mL of DOX, but in only three cell lines at 1 μg/mL. Interleukin-10 (IL-10), IL-6, IL-2, or tumor necrosis factor (TNF) induced no additive protective effects with CD40L L cells. In all five cell lines, DOX induced a concentration-dependent increase of the activity of the cysteine-protease caspase 3. Coculture with CD40L L cells, but not with CDw32 L cells, inhibited (38% to 100%) the activation of caspase 3 induced by 0.1 to 0.5 μg/mL of DOX in all five NHL cell lines, but in only two cell lines at 1 μg/mL. Finally, the antiproliferative effect of 0.1 to 0.5 μg/mL concentrations of DOX was also partially abrogated on coculture with CD40L L cells in all five cell lines, but in only two cell lines at 1 μg/mL. Cytokines, either alone or in combination with CD40L L cells, did not affect DOX-induced inhibition of proliferation. These results indicate that CD40L inhibits the apoptosis and antiproliferative effect induced by DOX and interferes with caspase 3 activation in B NHL cell lines. © 1998 by The American Society of Hematology.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3460-3460 ◽  
Author(s):  
Yu-Tzu Tai ◽  
Xian-Feng Li ◽  
Iris Breitkreutz ◽  
Weihua Song ◽  
Peter Burger ◽  
...  

Abstract Activation of the extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase (ERK1/2 MAPK) signaling pathway mediates tumor cell growth in many cancers, including human multiple myeloma (MM). Specifically, this pathway mediates MM cell growth and survival induced by cytokines/growth factors (i.e. IL-6, IGF-1, CD40, BAFF) and adhesion to bone marrow stromal cells (BMSCs), thereby conferring resistance to apoptosis in the bone marrow (BM) milieu. In this study, we therefore examined the effect of the MEK1/2 inhibitor AZD6244 (ARRY-142886), on human MM cell lines, freshly isolated patient MM cells and MM cells adhered to BMSCs. AZD6244, inhibits constitutive and cytokine (IL-6, IGF-1, CD40)-stimulated ERK1/2, but not AKT phosphorylation. Importantly, AZD6244 inhibits the proliferation and survival of human MM cell lines, regardless of sensitivity to conventional chemotherapy, as well as freshly isolated patient MM cells. AZD6244 induces apoptosis in patient MM cells even in the presence of BMSCs, as evidenced by caspase 3 activity and PARP cleavage at concentrations as low as 20 nM. AZD6244 overcomes resistance to apoptosis in MM cells conferred by IL-6 and BMSCs, and inhibits IL-6 secretion induced by MM adhesion to BMSCs. AZD6244 suppresses MM cell survival/growth signaling pathways (i.e., STAT3, Bcl-2, cyclin E1, CDK1, CDK3, CDK7, p21/Cdc42/Rac1-activated kinase 1, casein kinase 1e, IRS1, c-maf) and up-regulates proapoptotic cascades (i.e., BAX, BINP3, BIM, BAG1, caspase 3, 8, 6). AZD6244 also upregulates proteins triggering cell cycle arrest (i.e. p16INK4A, p18INK4C, p21/WAF1 [Cdkn1a], p27 [kip1], p57). In addition, AZD6244 inhibits adhesion molecule expression in MM cells (i.e. integrin a4 [VLA-4], integrin b7, ICAM-1, ICAM-2, ICAM-3, catenin a1, c-maf) associated with decreased MM adhesion to BMSCs. These pleiotropic proapoptotic, anti-survival, anti-adhesion and -cytokine secretion effects of AZD6244 abrogate BMSC-derived protection of MM cells, thereby sensitizing them to both conventional (dexamethasone) and novel (perifosine, lenalidomide, and bortezomib) therapies. In contrast, AZD6244 has minimal cytotoxicity in BMSCs and does not inhibit DNA synthesis in CD40 ligand-stimulated CD19 expressing B-cells derived from normal donors at concentrations toxic to MM cells (between 0.02–2 mM). Furthermore, AZD6244 inhibits the expression/secretion of osteoclast (OC)-activating factors (i.e., macrophage inflammatory protein (MIP)-1a, MIP-1b, IL-1b, VEGF) from MM cells. It also downregulates MM growth and survival factors (IL-6, BAFF, APRIL) in OC cultures derived from MM patient peripheral blood mononuclear cells (PBMCs). Significantly, AZD6244 inhibits OC differentiation from MM PBMCs (n=10) in a dose-dependent manner. Together these results provide the preclinical basis for clinical trials with AZD6244 (ARRY-142886) in MM.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4824-4824
Author(s):  
Yiqing Li ◽  
Songmei Yin ◽  
Shuangfeng Xie ◽  
Danian Nie ◽  
Liping Ma ◽  
...  

Abstract Abstract 4824 Recent studies have shown that prostaglandin E2 (PGE2) may play a key role in the tumorigenesis and tumor development. Membrane-bound prostaglandin E2 synthase-1 (mPGES-1), an inducible enzyme that acts downstream of cyclooxygenase (COX) and specifically catalyzes the conversion of prostaglandin H2 (PGH2) to PGE2, was over-expression in a variety of solid tumor cells and tissues such as nonsmall-cell lung cancer, colon carcinoma, gastric carcinoma and breast cancer. MK886, a small molecular inhibitor, is a reasonable potency as an inhibitor of mPGES-1 in vitro experiment. In this study, we examined effects of MK886 on expression of mPGES-1 and PGE2 synthesis in human acute myeloid leukemia cell line (HL-60), observed cell proliferation and apoptosis after 24-h treatment with MK886, and tried to explore the possible mechanisms by checking some protein belong AKT cell singling pathway such as P-AKT, Bax and Bcl-2. We found that the expression levels of mPGES-1 mRNA and protein were higher in HL-60 cells than in normal mononuclearcells (MNC). MK886 inhibited mPGES-1 mRNA and protein expression and reduced PGE2 secretion in HL-60 cells in a dose-dependent manner. The cell proliferation was inhibited and the IC50 was 132.16μmol/L. With the increase of MK886 concentration, the cell apoptosis rate assayed by flow cytometry increased and the apparent apoptotic bodies increased when staining by Hoechst 33258. After treated with MK886 for 24h, protein was extracted and assayed by western blot. The results showed that the expression levels of P-AKT, Bcl-2 and c-myc decreased while the Bax protein expression increased in a dose-dependent manner. The caspase-3 activity, determined by colorimetric detection, also increased dose-dependently. These results indicated that mPGES-1 over-expressed in leukemia cell line HL-60, MK886 could induce apoptosis in HL-60 cells via reducing mPGES-1 expression and PGE2 synthesis dose-dependently, thereby regulate the AKT pathway including Bcl-2 family and the activity of caspase-3. It suggested that mPGES-1 inhibitor might emerge as an important therapeutic tool for leukemia treatment. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 22 (11) ◽  
pp. 5792
Author(s):  
Laura Francesca Pisani ◽  
Gian Eugenio Tontini ◽  
Carmine Gentile ◽  
Beatrice Marinoni ◽  
Isabella Teani ◽  
...  

Interleukin (IL)-33 is a member of the interleukin (IL)-1 family of cytokines linked to the development of inflammatory conditions and cancer in the gastrointestinal tract. This study is designed to investigate whether IL-33 has a direct effect on human gastric epithelial cells (GES-1), the human gastric adenocarcinoma cell line (AGS), and the gastric carcinoma cell line (NCI-N87) by assessing its role in the regulation of cell proliferation, migration, cell cycle, and apoptosis. Cell cycle regulation was also determined in ex vivo gastric cancer samples obtained during endoscopy and surgical procedures. Cell lines and tissue samples underwent stimulation with rhIL-33. Proliferation was assessed by XTT and CFSE assays, migration by wound healing assay, and apoptosis by caspase 3/7 activity assay and annexin V assay. Cell cycle was analyzed by means of propidium iodine assay, and gene expression regulation was assessed by RT-PCR profiling. We found that IL-33 has an antiproliferative and proapoptotic effect on cancer cell lines, and it can stimulate proliferation and reduce apoptosis in normal epithelial cell lines. These effects were also confirmed by the analysis of cell cycle gene expression, which showed a reduced expression of pro-proliferative genes in cancer cells, particularly in genes involved in G0/G1 and G2/M checkpoints. These results were confirmed by gene expression analysis on bioptic and surgical specimens. The aforementioned results indicate that IL-33 may be involved in cell proliferation in an environment- and cell-type-dependent manner.


2020 ◽  
Vol 11 (2) ◽  
pp. 174-180
Author(s):  
Hesam Saghaei Bagheri ◽  
Seyed Hossein Rasta ◽  
Seyedeh Momeneh Mohammadi ◽  
Ali Akbar Rahim Rahimi ◽  
AliAkbar Movassaghpour ◽  
...  

Introduction: Laser radiation is a promising strategy against various malignancies. Recent studies have shown that the application of low-power laser therapy (LPLT) at different doses and exposure times could modulate the growth dynamic of tumor cells. Based on the type of laser, LPLT could potentially trigger cell proliferation, differentiation, and apoptosis in different cell lines. Methods: In this study, MTT assay was used to monitor the effect of low and high laser intensities on the viability of normal and cancer lymphocytes. The protein levels of Ki-67 (a proliferation marker) and Caspase-3 (an apoptosis factor) were measured in human peripheral mononuclear cells (PBMCs) and the B-lymphoblastic cell line (Nalm-6) using flow cytometry after being-exposed to 630-nm LPLT at low (2, 4, 6, and 10 J/cm2 ) and high (15, 30, 60, and 120 J/cm2 ) energy densities in a continuous mode for 48 and 72 hours. Results: By using higher energy densities, 60 and 120 J/cm2 , a significant decrease was shown in the viability of Nalm-6 cells, which reached 6.6 and 10.1% after 48 hours compared to the control cells (P<0.05). Notably, Cell exposure to doses 30, 60, and 120 J/cm2 yielded 7.5, 12.9, and 21.6 cell viability reduction after 72 hours. The collected data showed that the high-intensity parameters of LPLT (15 to 120 J/cm2 ) promoted significant apoptotic changes in the exposed cells coincided with the activation of Caspase-3 compared to the none-treated control cells (P<0.05). The data further showed the stimulation of the Ki-67 factor both in primary PBMCs and the lymphoblastic cell line treated with LPLT at energy densities of 4 and 6 J/cm2 (P<0.05), indicating enhanced cell proliferation. Similar to Nalm-6 cells, primary PBMCs showed apoptosis after 48 hours of being exposed to doses 60, and 120 J/cm2 , indicated by increased Caspase-3 levels (P<0.05). As expected, the Nalm-6 cells were resistant to cytotoxic effects of laser irradiation in the first 48 hours (P>0.05) compared to normal PBMCs. The exposure of Nalm-6 cells to low-intensity laser intensities increased a proliferation rate compared to the PBMCs treated with the same doses. Conclusion: We showed the potency of LPLT in the induction of apoptosis and proliferation in human primary PBMCs and Nalm-6 cells in a dose and time-dependent manner after 72 hours.


2016 ◽  
Vol 44 (6) ◽  
pp. 1395-1402 ◽  
Author(s):  
Yuewen Sun ◽  
Hanchen Qiu ◽  
Mingchun Ou ◽  
Runli Chen ◽  
Gang Liang

Objective Taccaoside, a steroidal saponin, has been shown to be cytotoxic, although the mechanism of cytotoxicity remains unclear. This study examined the effect of taccaoside on the human hepatocellular carcinoma (HCC) cell lines SMMC-7721 and Bel-7404. Methods The antiproliferative effect of taccaoside were measured using the 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide assay. Cells were stained with Hoechst 33258 to observe morphology. Cell cycle and apoptosis were analysed by flow cytometry. Caspase activation was detected using specific assays, and PARP, Bax and Bcl-2 expression were analysed using western blotting. Results Taccaoside showed antiproliferative effect on HCC cell lines growth in a concentration- and time-dependent manner. Taccaoside arrested cell cycle in the G2/M phase and induced caspase-dependent apoptosis. Western blotting indicated that taccaoside upregulated Bax expression and downregulated Bcl-2 expression. PARP cleavage was observed following taccaoside treatment. Conclusions This study showed that taccaoside may inhibit HCC cell proliferation by inducing apoptosis.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4615-4615 ◽  
Author(s):  
Azhar R. Hussain ◽  
Naif A. Al-Jomah ◽  
Abdul K. Siraj ◽  
Manugaran S. Pulicat ◽  
Khaled A. Al-Hussein ◽  
...  

Abstract Primary effusion lymphoma (PEL) is an aggressive and fatal type of cancer. PEL cells produce a variety of autocrine cytokines and growth factors, which provides cyto-protection against conventional chemotherapeutic agents. In efforts to identify novel approaches to block the proliferation of PEL cells, we found that Sanguinarine, a natural compound isolated from the root plant Sanguinaria canadendid, that is being used as an anti-microbial agent, inhibited cell proliferation and induced apoptosis in a dose dependent manner in several PEL cell lines through a bax-dependent signaling pathway. Five PEL cell lines used in this study were treated with various doses of Sanguinarine ranging between 0.5–4μM inhibited cell proliferation in all the cell lines in a dose dependent manner (BC1 40–97%, BC3 46–93%, BCBL1 11–94%, BCP1 20–97% and HBL6 7–95%). Treatment with varying doses of Sanguinarine also induced apoptosis in all cell lines as determined by cell cycle analysis, annexinV/PI dual staining, TUNEL assay and DNA laddering. Sanguinarine treatment resulted in up-regulation of death receptor 5 (DR5) expression, activation of caspase-8 and Bid leading to Bax conformational changes and translocation to the mitochondrial causing loss of mitochondrial membrane potential as measured by JC1 staining and release of cytochrome c to the cytosole. Sanguinarine induced release of cytochrome c resulted in activation of caspase-3, followed by polyadenosin-5′-diphosphate-ribose polymerase (PARP) cleavage leading to inhibition of proliferation and induction of caspase-dependent apoptosis. Furthermore, pre-treatment of PEL cells with z-VAD-fmk, a universal inhibitor of caspases, abrogated caspase-3 and PARP activation and prevented cell death induced by Sanguinarine. Inhibitor of apoptosis proteins (IAPs), play an important role in protecting cells against apoptosis through their direct action on caspases-9 and -3. Treatment of PEL cells with Sanguinarine down-regulated the expression of IAPs; XIAP, cIAP1 and cIAP2. Taken altogether, our findings suggest that Sanguinarine induces apoptosis via up-regulation of DR5, activation of Bax in a caspase-dependent pathway and down-regulation of IAPs. These results provide the molecular basis and preliminary data for new treatment strategies that may incorporate Sanguinarine in regimens for primary effusion lymphoma treatment.


Sign in / Sign up

Export Citation Format

Share Document