Atiprimod Blocks STAT3 and STAT5 Phosphorylation, Inhibits Proliferation, and Induces Apoptosis in Acute Myeloid Leukemia (AML) but Not Normal Cells.

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 ◽  
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 ◽  
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.


Biomolecules ◽  
2019 ◽  
Vol 9 (9) ◽  
pp. 476 ◽  
Author(s):  
Chia-Jung Li ◽  
Pei-Yi Chu ◽  
Giou-Teng Yiang ◽  
Meng-Yu Wu

The transforming growth factor-β (TGF-β) signaling pathway plays multiple regulatory roles in the tumorigenesis and development of cancer. TGF-β can inhibit the growth and proliferation of epithelial cells and induce apoptosis, thereby playing a role in inhibiting breast cancer. Therefore, the loss of response in epithelial cells that leads to the inhibition of cell proliferation due to TGF-β is a landmark event in tumorigenesis. As tumors progress, TGF-β can promote tumor cell invasion, metastasis, and drug resistance. At present, the above-mentioned role of TGF-β is related to the interaction of multiple signaling pathways in the cell, which can attenuate or abolish the inhibition of proliferation and apoptosis-promoting effects of TGF-β and enhance its promotion of tumor progression. This article focuses on the molecular mechanisms through which TGF-β interacts with multiple intracellular signaling pathways in tumor progression and the effects of these interactions on tumorigenesis.


2021 ◽  
Vol 2021 ◽  
pp. 1-15
Author(s):  
Chao Hu ◽  
Xiaobin Zhu ◽  
Taogen Zhang ◽  
Zhouming Deng ◽  
Yuanlong Xie ◽  
...  

Introduction. Osteosarcoma is a malignant tumor associated with high mortality rates due to the toxic side effects of current therapeutic methods. Tanshinone IIA can inhibit cell proliferation and promote apoptosis in vitro, but the exact mechanism is still unknown. The aims of this study are to explore the antiosteosarcoma effect of tanshinone IIA via Src kinase and demonstrate the mechanism of this effect. Materials and Methods. Osteosarcoma MG-63 and U2-OS cell lines were stable transfections with Src-shRNA. Then, the antiosteosarcoma effect of tanshinone IIA was tested in vitro. The protein expression levels of Src, p-Src, p-ERK1/2, and p-AKt were detected by Western blot and RT-PCR. CCK-8 assay and BrdU immunofluorescence assay were used to detect cell proliferation. Transwell assay, cell scratch assay, and flow cytometry were used to detect cell invasion, migration, and cell cycle. Tumor-bearing nude mice with osteosarcoma were constructed. The effect of tanshinone IIA was detected by tumor HE staining, tumor inhibition rate, incidence of lung metastasis, and X-ray. Results. The oncogene role of Src kinase in osteosarcoma is reflected in promoting cell proliferation, invasion, and migration and in inhibiting apoptosis. However, Src has different effects on cell proliferation, apoptosis, and cell cycle regulation among cell lines. At a cellular level, the antiosteosarcoma effect of tanshinone IIA is mediated by Src downstream of the MAPK/ERK and PI3K/AKt signaling pathways. At the animal level, tanshinone IIA played a role in resisting osteosarcoma formation by Src downstream of the MAPK/ERK and PI3K/AKt signaling pathways. Conclusion. Tanshinone IIA plays an antiosteosarcoma role in vitro and in vivo and inhibits the progression of osteosarcoma mediated by Src downstream of the MAPK/ERK and PI3K/AKt signaling pathways.


Blood ◽  
1988 ◽  
Vol 72 (5) ◽  
pp. 1487-1491 ◽  
Author(s):  
T Sakurai ◽  
H Hara ◽  
K Nagai

Abstract A new anti-K562 cell monoclonal antibody, RTF8X, a cytotoxic IgM, recognized a surface antigen on erythroblasts from patients with erythroleukemia and polycythemia vera. RTF8X, which is highly specific to K562 cells, did not react with the other 14 hematopoietic cell lines and the seven nonhematopoietic cell lines. RTF8X antigen was not detected in normal peripheral blood, but was found in less than 1% of normal marrow cells. RTF8X did not inhibit in vitro colony formation of CFU-E and BFU-E in a complement-dependent cytotoxicity assay. Cell- sorting analysis showed that, morphologically, the RTF8X-positive marrow cells from the patients and normal volunteers contained more than 60% erythroblasts and that CFU-E and BFU-E were not demonstrated in cells with RTF8X antigen. Enzyme treatment suggested that RTF8X antigen was a sialoglycolipid. These results indicate that RTF8X may recognize the surface antigen found increasingly in association with tumors of erythroid lineage. RTF8X should be useful for studies of erythroid differentiation and proliferation in patients.


Pharmaceutics ◽  
2020 ◽  
Vol 12 (12) ◽  
pp. 1198
Author(s):  
Sathishbabu Paranthaman ◽  
Meghana Goravinahalli Shivananjegowda ◽  
Manohar Mahadev ◽  
Afrasim Moin ◽  
Shivakumar Hagalavadi Nanjappa ◽  
...  

A paradigm shift in treating the most aggressive and malignant form of glioma is continuously evolving; however, these strategies do not provide a better life and survival index. Currently, neurosurgical debulking, radiotherapy, and chemotherapy are the treatment options available for glioma, but these are non-specific in action. Patients invariably develop resistance to these therapies, leading to recurrence and death. Receptor Tyrosine Kinases (RTKs) are among the most common cell surface proteins in glioma and play a significant role in malignant progression; thus, these are currently being explored as therapeutic targets. RTKs belong to the family of cell surface receptors that are activated by ligands which in turn activates two major downstream signaling pathways via Rapidly Accelerating Sarcoma/mitogen activated protein kinase/extracellular-signal-regulated kinase (Ras/MAPK/ERK) and phosphatidylinositol 3-kinase/a serine/threonine protein kinase/mammalian target of rapamycin (PI3K/AKT/mTOR). These pathways are critically involved in regulating cell proliferation, invasion, metabolism, autophagy, and apoptosis. Dysregulation in these pathways results in uncontrolled glioma cell proliferation, invasion, angiogenesis, and cancer progression. Thus, RTK pathways are considered a potential target in glioma management. This review summarizes the possible risk factors involved in the growth of glioblastoma (GBM). The role of RTKs inhibitors (TKIs) and the intracellular signaling pathways involved, small molecules under clinical trials, and the updates were discussed. We have also compiled information on the outcomes from the various endothelial growth factor receptor (EGFR)–TKIs-based nanoformulations from the preclinical and clinical points of view. Aided by an extensive literature search, we propose the challenges and potential opportunities for future research on EGFR–TKIs-based nanodelivery systems.


2010 ◽  
Vol 207 (3) ◽  
pp. 309-317 ◽  
Author(s):  
M Arvigo ◽  
F Gatto ◽  
M Ruscica ◽  
P Ameri ◽  
E Dozio ◽  
...  

Somatostatin analogues inhibit in vitro cell proliferation via specific membrane receptors (SSTRs). Recent studies on transfected cell lines have shown a ligand-induced formation of receptor dimers. The aim of this study is 1) to evaluate the role of specific ligands in modulating receptor interactions in the androgen-dependent prostate cancer cell line, LNCaP, and in the non-small cell lung cancer line, Calu-6, by co-immunoprecipitation and immunoblot; and 2) to correlate the antiproliferative effect of these compounds with their ability in modulating receptor interactions. In LNCaP, we have demonstrated the constitutive presence of sstr1/sstr2, sstr2/sstr5, sstr5/dopamine (DA) type 2 receptor (D2R), and sstr2/D2R dimers. BIM-23704 (sstr1- and sstr2-preferential compound) increased the co-immunoprecipitation of sstr1/sstr2 and significantly inhibited proliferation (−30.98%). BIM-23244 (sstr2–sstr5 selective agonist) significantly increased the co-immunoprecipitation of sstr2/sstr5, and induced a −41.36% inhibition of proliferation. BIM-23A760, a new somatostatin/DA chimeric agonist with a high affinity for sstr2 and D2R and a moderate affinity for sstr5, significantly increased the sstr5/D2R and sstr2/D2R complexes and was the most powerful in inhibiting proliferation (−42.30%). The chimeric compound was also the most efficient in modulating receptor interaction in Calu-6, increasing the co-immunoprecipitation of D2R/sstr5 and inhibiting cell proliferation (−30.54%). However, behind BIM-23A760, BIM-53097 (D2R-preferential compound) also significantly inhibited Calu-6 proliferation (−17.71%), suggesting a key role for D2R in receptor cross talk and in controlling cell growth. Indeed, activation of monomeric receptors did not affect receptor co-immunoprecipitation, whereas cell proliferation was significantly inhibited when the receptors were synergistically activated. In conclusion, our data show a dynamic ligand-induced somatostatin and DA receptor interaction, which may be crucial for the antiproliferative effects of the new analogues.


PPAR Research ◽  
2008 ◽  
Vol 2008 ◽  
pp. 1-11 ◽  
Author(s):  
Giulia Cantini ◽  
Adriana Lombardi ◽  
Elisabetta Piscitelli ◽  
Giada Poli ◽  
Elisabetta Ceni ◽  
...  

Rosiglitazone (RGZ), a thiazolidinedione ligand of the peroxisome proliferator-activated receptor (PPAR)-γ, has been recently described as possessing antitumoral properties. We investigated RGZ effect on cell proliferation in two cell line models (SW13 and H295R) of human adrenocortical carcinoma (ACC) and its interaction with the signaling pathways of the activated IGF-I receptor (IGF-IR). We demonstrate a high expression of IGF-IR in the two cell lines and in ACC. Cell proliferation is stimulated by IGF-I in a dose- and time-dependent manner and is inhibited by RGZ. The analysis of the main intracellular signaling pathways downstream of the activated IGF-IR, phosphatidyl inositol 3-kinase (PI3K)-Akt, and extracellular signal-regulated kinase (ERK1/2) cascades reveals that RGZ rapidly interferes with the Akt and ERK1/2 phosphorylation/activation which mediates IGF-I stimulated proliferation. In conclusion, our results suggest that RGZ exerts an inhibitory effect on human ACC cell proliferation by interfering with the PI3K/Akt and ERK1/2 signaling pathways downstream of the activated IGF-IR.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3352-3352
Author(s):  
Klaus Podar ◽  
Melissa Simoncini ◽  
Yu-Tzu Tai ◽  
Martin Sattler ◽  
Kenji Ishitsuka ◽  
...  

Abstract The tyrosine kinase inhibitor adaphostin is a member of the tyrophostin family of small molecules that interfere with peptide binding rather, than targeting the kinase ATP-binding site. Adaphostin has therefore been examined as an alternative to the 2-phenylaminopyrimidine derivate imatinib mesylate, with remarkable efficacy in the treatment of chronic myeloic leukemia (CML). Previous studies show that adaphostin induces apoptosis: (1) in Bcr/Abl+ cells more rapidly than imatinib mesylate; (2) in imatinib mesylate resistant cells; and (3) in Bcr/ Abl - cells. Imatinib mesylate has minimal, if any activity in MM; the efficacy of adaphostin in multiple myeloma (MM) is unknown. Here we compare the effects of adaphostin and imatinib mesylate against human MM cells. Our results show concentration-dependent apoptosis in MM.1S, U266, OPM-2, INA-6, RPMI8226 and RPMI-Dox40 MM cells after treatment with adaphostin, but not with imatinib mesylate. Imatinib mesylate induced more than 50% apoptosis in K562 cells using concentrations as low as 1mM, which served as a positive control. Moreover, adaphostin, but not imatinib mesylate, induced caspase-9, caspase-8, and PARP cleavage, as well as downregulation of Mcl-1, in MM cells. Further results demonstrated that adaphostin induces peroxide production and DNA strand breaks after long-term treatment. Importantly MM cell proliferation induced by MM cell binding to BMSCs was abrogated by adaphostin- treatment. IL-6 and IGF-1 signaling and sequelae triggered by these cytokines are important growth, survival, and drug resistance factors in MM; conversely, adaphostin but not imatinib mesylate, inhibited phosphorylation of Src tyrosine kinase family, Akt-1, and ERK. Taken together, our studies in MM cells show that (1) adaphostin- inhibits IGF-1- and IL-6- triggered signaling pathways as well as (2) induces reactive oxygen species and apoptosis. These studies therefore provide the preclinical framework for its clinical evaluation to improve patient outcome in MM.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 952-952 ◽  
Author(s):  
Shinichi Kitada ◽  
Edward Monosov ◽  
Sharon Chandler ◽  
Esther D. Avery ◽  
Thomas J. Kipps ◽  
...  

Abstract Altered expression of Bcl-2-family protein plays a central role in apoptosis dysregulation in cancer and leukemia, promoting malignant cell expansion and contributing to chemoresistance. Anti-apoptotic Bcl-2-family proteins Bcl-2 and Bcl-XL have been thoroughly validated as drug discovery targets for cancer, and strategies for inhibiting these proteins have been devised based on mimicking their endogenous antagonists, the BH3-only proteins. CLL (chronic lymphocytic leukemia) is a quintessential example of a human malignancy caused by defective programmed cell death, representing the most common form of adult leukemia in North America and Europe. Over-expression of the Bcl-2 protein is one of the most consistent and prominent etiological factors associated with this disease. In this study, we evaluated biological effects of ABT-737, a novel fully synthetic Bcl-2/Bcl-XL antagonist developed at Abbott Laboratories, on CLL cells and B-lymphoma cell lines in vitro. ABT-737 induced a striking dose-dependent apoptosis in all CLL cells tested, with a Lethal Dose 50% (LD50) of 3–10 nM for 10 of 11 CLL samples. ABT-737 exhibited similar activity against chemo-naive cells as well as extensively treated, relapsed-disease. In contrast, an enantiomer of ABT-737 with little affinity for Bcl-2 and Bcl-XL was inactive at concentrations up to 1 μM, demonstrating specificity. Similarly, ABT-737 induced striking dose-dependent apoptosis in B-lymphoma cell lines, such as 380 lymphoma cell line which over-expressed Bcl-2 as a result of t14:18 translocation, with the LD50 ratio for ABT-737:Enantiomer control of approximately 1,000. At concentrations up to 0.1 μM, ABT-737 had no cytotoxic action on normal T-lymphocytes isolated from peripheral blood collected from healthy individuals. In addition, CLL cells were at least 10 fold more sensitive to ABT-737 than normal CD19-positive B-lymphocytes isolated from healthy individuals. Moreover, ABT-737 induced caspase 3 activation and PARP cleavage within 2 hours in CLL B-cells, while zVAD-fmk completely blocked caspase 3 activation and PARP-cleavage. The mechanism of ABT-737 was further validated by confocal time-lapsed microscopy experiments, where the active compound (but not enantiomer control) was demonstrated to displace a Green Fluorescent Protein (GFP)-tagged BH3-containing protein from wild-type Bcl-XL localized at mitochondrial surfaces in intact tumor cell lines. In contrast, active compound failed to displace GFP-BH3 protein from the mitochondrial surface of cells expressing mutants of Bcl-XL purposely engineered to be incapable of binding ABT-737. Taken together, these data strongly suggest that ABT-737 is a mechanism-based inhibitor of Bcl-2 and Bcl-XL that warrants further evaluation for the possible treatment of CLL and other malignancies linked to over-expression of Bcl-2 or Bcl-XL, where chemorefractory states represent a barrier to successful eradication of cancer.


Sign in / Sign up

Export Citation Format

Share Document