Effects of EGF-R on Transcription Factor Activation in Hematopoietic Cells: EGF-R Activates Multiple Transcription Factors Which Induce Proliferation in the Absence of Autocrine Cytokines.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4185-4185
Author(s):  
John G. Shelton ◽  
Linda S. Steelman ◽  
Martin McMahon ◽  
James A. McCubrey

Abstract v-ErbB is an oncogene related to the Epidermal Growth Factor Receptor (EGF-R) that was initially discovered in the genome of avian erythoblastosis virus. v-ErbB will abrogate the requirement of erythroid progenitor cells for erythropoietin and stem cell factor and block terminal differentiation. EGF-R overexpression has been observed in many pathological situations and the EGF-R gene is amplified in certain tumors. Moreover, there is a truncated form of EGF-R referred to as EGFvIII which resembles v-ErbB in biological properties. One problem frequently encountered in studying the effects of EGF-R overexpression in many tumors is that EGF-R expression is often constitutive and in the presence of increased expression of other oncogenes or in the absence of certain tumor suppressor genes. To circumvent these problems, we subcloned v-ErbB into a vector which contained the estrogen receptor hormone binding domain which renders the v-ErbB protein dependent upon the addition of beta-estradiol or 4 OH tamoxifen for activity. The v-ErbB:ER oncogene will conditionally abrogate the cytokine dependence of human (TF-1) and murine (FL5.12 and FDC-P1) hematopoietic cells efficiently. This construct has allowed us to examine the transcription factors pathways activated by v-ErbB:ER in hematopoietic cells which are required for proliferation in the absence of previously required cytokines. By determining which signal transduction pathways were activated in response to either v-ErbB:ER activation or IL-3 addition in the presence and absence of specific small molecular weight membrane permeable kinase inhibitors, we could ascertain that v-ErbB:ER expression activated the STAT5, Elk, CREB, Jun, and Forkhead (Foxo) family of transcription factors in FL/v-ErbB:ER cells. The activation of these transcription factors was blocked by the respective kinase inhibitors. Thus v-ErbB:ER activated a broad spectrum of transcription factors. Treatment of v-ErbB:ER cells with the EGF-R inhibitor AG1478 very efficiently induced apoptosis in these cells at 100 to 1000 fold lower concentrations than MEK, PI3K or JNK inhibitors and activation of all these transcription factor inhibitors was inhibited. In contrast, when the cells were treated with MEK, PI3K or JNK inhibitors, only the transcription factors specific for the individual pathways were inhibited. FL5.12 cells conditionally transformed to grow in response to activated Raf and Akt, (FL/Akt:ER+Raf-1:AR) were also isolated. Activation of STAT5 by either Raf or Akt did not occur in FL/Akt:ER+Raf-1:AR cells, but did occur when the cells were treated with IL-3. Furthermore, Elk activation occurred in response to Raf activation but not IL-3 stimulation in the FL/Akt:ER+Raf-1:AR cells which grew in response to Raf and Akt. Thus oncogenes such as v-ErbB:ER, which are more effective in their ability to transform hematopoietic cells than oncogenes such as Raf and Akt, can induce multiple transcription factors, only some of which are required for growth in tissue culture systems.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1264-1264
Author(s):  
John G. Shelton ◽  
Linda S. Steelman ◽  
Martin McMahon ◽  
James A. McCubrey

Abstract v-ErbB is an oncogene related to the Epidermal Growth Factor Receptor (EGF-R) that was initially discovered in the genome of avian erythoblastosis virus. v-ErbB will abrogate the requirement of erythroid progenitor cells for erythropoietin and stem cell factor and block terminal differentiation. EGF-R overexpression has been observed in many pathological situations and there is a truncated form of EGF-R referred to as EGFvIII which resembles v-ErbB in biological properties. EGF-R expression is often constitutive and may occur in the presence of expression of other oncogenes or in the absence of certain tumor suppressor genes. To circumvent these problems, we subcloned v-ErbB into a vector which contained the estrogen receptor hormone binding domain which renders the v-ErbB protein dependent upon the addition of beta-estradiol for activity. v-ErbB:ER conditionally abrogated the cytokine dependence of human (TF-1) and murine (FL5.12 and FDC-P1) hematopoietic cells efficiently. This construct allowed examination of the signal transduction and anti-apoptotic pathways activated by v-ErbB:ER in hematopoietic cells. By determining which signal transduction pathways were activated in response to either v-ErbB:ER or IL-3 in the presence and absence of inhibitors, we could ascertain that v-ErbB:ER expression activated the Jak/STAT, Raf/MEK/ERK, PI3K/Akt/mTOR/p70S6K and JNK pathways. Thus v-ErbB:ER activated a broad spectrum of signal transduction pathways some of which were linked to the prevention of apoptosis. Apoptosis was measured by annexin V/PI binding and activation of caspases 3, 8 & 9. Treatment of v-ErbB:ER cells with the EGF-R inhibitor AG1478 efficiently induced apoptosis in these cells at 100 to 1000 fold lower concentrations than MEK, PI3K/mTOR or JNK inhibitors, and activation of all these signal transduction pathways was inhibited with the EGFR inhibitor. Induction of apoptosis by the EGF-R inhibitor was only observed when the cells were grown in response to v-ErbB:ER activation demonstrating specificity. In contrast, other inhibitors suppressed kinase activation and induced some apoptosis when the cells were grown in response to v-ErbB:ER or IL-3. When the cells were treated with MEK, PI3K/mTOR or JNK inhibitors, only the kinases specific for those pathways were inhibited. When MEK and PI3K/mTOR inhibitors were added a synergistic apoptotic response was observed. In contrast, combinations of Jak or JNK and either MEK or PI3K/mTOR inhibitors did not result in a synergistic apoptotic response. Thus the Raf/MEK/ERK and PI3K/Akt/mTOR/p70S6K pathways are the most important pathways in preventing apoptosis in these cells. FL5.12 cells conditionally transformed to grow in response to activated Raf and Akt, (FL/Akt:ER+Raf-1:AR) were also isolated. Activation of STAT5 by either Raf or Akt did not occur in these cells, but did occur upon IL-3 treatment. Furthermore, Elk activation occurred in response to Raf activation but not IL-3 stimulation in the cells which grew in response to Raf and Akt. v-ErbB:ER is more effective in its ability to abrogate cytokine dependence of hematopoietic cells than Raf or Akt, and it induces multiple signal transduction pathways, only some of which are required for growth and the prevention of apoptosis in tissue culture systems.


2000 ◽  
Vol 47 (2) ◽  
pp. 339-347 ◽  
Author(s):  
E Radziszewska ◽  
K Piwocka ◽  
A Bielak-Zmijewska ◽  
J Skierski ◽  
E Sikora

UVC-induced apoptotic symptoms such as morphological changes, DNA fragmentation, Bcl-2 and Bax protein expression were examined in primary splenocyte cultures from young (3 months) and old (24 months) rats. The activities of AP-1 and CRE transcription factors in UVC-irradiated splenocytes were also assessed. At 24 h after UVC irradiation 40% of cells derived from young rats were found to be apoptotic, which was twice as much as in splenocytes from old rats. Apoptosis in cells from old rats did not give typical symptoms like a "DNA ladder" and Bcl-2 protein downregulation, in contrast to splenocytes from young rats. No AP-1 transcription factor activity was found in UVC-irradiated splenocytes from old animals and only a trace activity in splenocytes from young animals. This indicates that, UVC-induced apoptosis in rat splenocytes is practically AP-1 independent and that cells from old rats are less sensitive to UVC irradiation than splenocytes from young rats.


Blood ◽  
2006 ◽  
Vol 107 (10) ◽  
pp. 3983-3991 ◽  
Author(s):  
Elena E. Solomou ◽  
Keyvan Keyvanfar ◽  
Neal S. Young

In aplastic anemia, immune destruction of hematopoietic cells results in bone marrow failure. Type 1 cytokines, especially IFN-γ, have been implicated in the pathophysiology of T-cell–mediated, Fas-mediated stem cell apoptosis of hematopoietic cells. Here, we show that the transcription factor T-bet (T-box expressed in T cells) is increased in T cells from patients with aplastic anemia. Patients' T-bet bound directly to the proximal site of the IFN-γ promoter without any prior stimulation, in contrast to healthy controls. Increased levels of Itk kinase participated in T-bet up-regulation and active transcription of the IFN-γ gene observed in these patients. Blocking PKC-θ, a kinase that lies downstream of Itk kinase, decreased T-bet protein and IFN-γ intracellular levels. These data suggest that the increased IFN-γ levels observed in aplastic anemia patients are the result of active transcription of the IFN-γ gene by T-bet. Blocking the transcription of the IFN-γ gene with kinase inhibitors might lead to the development of novel therapeutic agents for patients with aplastic anemia and other autoimmune diseases.


2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 13032-13032 ◽  
Author(s):  
F. Morgillo ◽  
W. K. Hong ◽  
H. Lee

13032 Background: Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) have been used to treat lung cancers, but resistance to these agents has been observed. This study was designed to investigate whether the insulin-like growth factor (IGF)-mediated signaling pathway induces resistance to the EGFR TKIs in lung cancer. Methods: The antitumor activities and action mechanisms of EGFR inhibitors (erlotinib, gefinitib, cetuximab), single or in combination with IGF-IR inhibitors, were assessed in vitro in a subset of non-small-cell lung cancer (NSCLC) cell lines by the MTT assay, flow cytometry-based TUNEL assay, soft agar, confocal microscopy, metabolic labeling, coimmunoprecipitation, and northern and western blot analyses, and in vivo in animal models. Correlation of EGFR and IGF-1R expression was assessed using lung tissues from patients with NSCLC. Results: EGFR TKI inhibited the proliferation and anchorage-dependent and -independent colony-forming ability of NSCLC cells, which induced apoptosis, only when IGF-1R signaling was blocked. Treatment with EGFR TKIs, but not with the EGFR antibody, induced EGFR:IGF-1R heterodimerization on cell surface and activation of the IGF-1R, resulting in the stimulation of PI3K/Akt/ mTOR pathway, promoting the de novo protein biosynthesis of survivin and EGFR, resulting in the survival of NSCLC cells. When IGF-IR and mTOR were blocked, treatment of EGFR-TKIs induced apoptosis in NSCLC cells, resulting in suppression of the NSCLC tumor growth. When we tested the expression of IGF-R and EGFR in human lung tissue, 9/14 tumor samples exhibited increased expression of EGFR and 7/9 samples showed a correlative increases in IGF-IR protein levels compared to their paired normal counterparts. Conclusions: These findings suggest that simultaneous targeting of EGFR and IGF-1R signaling pathways might be an effective therapeutic strategy against NSCLC. No significant financial relationships to disclose.


Blood ◽  
1996 ◽  
Vol 88 (8) ◽  
pp. 2917-2926 ◽  
Author(s):  
GW Henkel ◽  
SR McKercher ◽  
H Yamamoto ◽  
KL Anderson ◽  
RG Oshima ◽  
...  

Transcription factors play an important role choreographing lineage commitment and expansion of blood cells. Nuclear factors that are expressed primarily or exclusively in hematopoietic cells are likely candidates for regulating blood cell development. The transcription factor PU.1 is found only in hematopoietic cells, whereas ets-2, a related family member, is ubiquitously expressed. To compare the role of these two transcription factors in macrophage development, embryonic stem (ES) cells with a homozygous disruption of either the PU.1 or the ets-2 gene were generated. The ability of both knockout ES cells to differentiate to macrophages was tested. Normal development of macrophages, as determined by histochemical and immunohistochemical analysis, from PU.1 knockout ES cells was significantly blocked. Furthermore, the expression of known markers associated with macrophages, such as c-fms, CD11b, CD18 and granulocyte-macrophage colony-stimulating factor receptor, were not detected by reverse transcriptase-polymerase chain reaction. In contrast to the PU.1 knockout ES cells, macrophages, development from the ets-2 knockout ES cells was normal. Although both PU.1 and ets-2 are found in macrophages, these data show a distinct role for the lineage-restricted PU.1 transcription factor in macrophage development.


2021 ◽  
Vol 28 ◽  
Author(s):  
Yan Si ◽  
Xinxin Pei ◽  
Xiangfang Wang ◽  
Qianqian Han ◽  
Changzhi Xu ◽  
...  

Background: Acquired resistance to epidermal growth factor receptor–tyrosine kinase inhibitors (EGFR-TKIs) is a recurrent phenomenon during clinical therapy of non‑small-cell lung cancer (NSCLC). Studies have shown that HER2 is a key factor contributing to drug resistance in a variety of cancers. Furthermore, we have observed that HER2 is overexpressed in PC-9 NSCLC cells with acquired gefitinib-resistance (PC-9/GR) as compared to that in PC-9 cells. Objective: We hypothesized that blocking both EGFR and HER2 may serve as a potential strategy for treatment of NSCLC with acquired gefitinib-resistance. Methods: To target both EGFR and HER2 simultaneously, we developed a bispecific antibody HECrossMAb, which was derived from a humanized Cetuximab and Trastuzumab. The binding affinity of HECrossMAb for EGFR and HER2 was measured using enzyme-linked immunosorbent assay. The MTT assay was used to determine the effect of HECrossMAb on the proliferation of PC‑9 and PC‑9/GR cells in vitro. Finally, the effect of HECrossMAb on PI3K/AKT signaling and associated transcription factors was measured using western blot analysis. Results: Our results showed that HECrossMAb exerts enhanced cytotoxicity in both PC-9 and PC-9/GR cells by inhibiting the activation of PI3K/AKT signaling and expression of relevant transcription factors such as AEG-1, c-Myc, and c-Fos. Conclusion: Our results suggest that HECrossMAb may function as a potential therapeutic agent for the treatment of NSCLC overexpressing EGFR and HER2.


2018 ◽  
Author(s):  
Remah Ali ◽  
Wells Brown ◽  
Stephen Conner Purdy ◽  
V. Jo Davisson ◽  
Michael K. Wendt

AbstractInhibition of EGFR signaling by small molecule kinase inhibitors and monocloncal antibodies has proven effective in the treatment of multiple cancers. In contrast, metastatic breast cancers (BC) derived from EGFR-expressing mammary tumors are inherently resistant to EGFR-targeted therapies. Mechanisms that contribute to this inherent resistance remain poorly defined. Here we show that in contrast to primary tumors, ligand-mediated activation of EGFR in metastatic BC is dominated by STAT1 signaling. This change in downstream signaling leads to apoptosis and growth inhibition in response to EGF in metastatic BC cells. Mechanistically, these changes in downstream signaling result from an increase in the internalized pool of EGFR in metastatic cells, increasing physical access to the nuclear pool of STAT1. Along these lines, an EGFR mutant that is defective in endocytosis is unable to elicit STAT1 phosphorylation and apoptosis. Additionally, inhibition of endosomal signaling using an EGFR inhibitor linked to a nuclear localization signal specifically prevents EGF-induced STAT1 phosphorylation and cell death, without affecting EGFR:ERK1/2 signaling. Pharmacologic blockade of ERK1/2 signaling through the use of the allosteric MEK1/2 inhibitor, trametinib, dramatically biases downstream EGFR signaling toward a STAT1 dominated event, resulting in enhanced EGF-induced apoptosis in metastatic BC cells. Importantly, combined administration of trametinib and EGF also facilitated an apoptotic switch in EGFR-transformed primary tumor cells, but not normal mammary epithelial cells. These studies reveal a fundamental distinction for EGFR function in metastatic BC. Furthermore, the data demonstrate that pharmacological biasing of EGFR signaling toward STAT1 activation is capable of revealing the apoptotic function of this critical pathway.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2700-2700
Author(s):  
Heather M. Rogers ◽  
Constance Tom Noguchi

Abstract An important treatment strategy for sickle cell anemia is increasing fetal hemoglobin (HbF) in circulating erythrocytes. OSI-2040 (Apicidin), a fungus-derived cyclic tetra-peptide, induces g-globin production in K562 cells. The effect of increasing doses of OSI-2040 (from 7.5 to 750 nM) was determined on cultures of primary human hematopoietic progenitor cells stimulated with erythropoietin (EPO). Cell proliferation and differentiation, globin production, and erythroid transcription factors expression were examined. At concentrations 7.5 nM - 75 nM there was minimal decrease in cell proliferation with little change in % benzidine positive cells after 12 days of culture with EPO. As OSI-2040 concentration increased above 75 nM, cell proliferation and % benzidine positive cells decreased, with concentrations of 300 and 750 nM being highly toxic, reducing cell number by 75% or more. Analysis of globin gene expression indicates that low to mid concentrations of OSI-2040 increase g-globin, with the peak increase occurring at 75 nM, while the highest concentrations (300 and 750 nM) suppress g-globin. OSI-2040 decreases b-globin expression with the highest concentrations resulting in the greatest decreases. The g/(g+b) ratio increases with increasing OSI-2040 concentration reaching a value of 4-fold and greater for concentrations of 75 nM or more, partially a consequence of the suppression of b-globin expression, particularly at higher concentrations. Although the g/(g+b) ratio is relatively high at the highest concentrations of OSI-2040 (300 and 750 nM), it is at a cost in overall globin production and cell toxicity. Hemoglobin expression is determined primarily at the transcription level. We found that OSI-2040 affects expression of select transcription factors, GATA-1, GATA-2, SCL/Tal-1 and EKLF, which are critical for erythroid differentiation. Peak EPO induction of GATA-1, a zinc-finger transcription factor essential for survival and differentiation of erythroid progenitor cells, is delayed with OSI-2040 treatment. OSI-2040 also delays expression of SCL/Tal-1, a basic-helix-loop-helix transcription factor that positively regulates erythroid differentiation and is required for the production of mature erythrocytes. In addition, there is a delay in the induction of EKLF, a zinc-finger transcription factor necessary for induction of b-globin in adult erythroid cells that acts by direct binding to the b-globin promoter. With increasing OSI-2040 concentrations, there is a dose-dependent decrease in overall levels of GATA-1, SCL/Tal-1 and EKLF. GATA-2, a member of the GATA-family that plays a critical role in the survival of early erythroid progenitor cells and is down-regulated with EPO stimulation, shows a slight delay in its reduction at 75 and 150 nM but overall is not greatly affected by OSI-2040. Thus, OSI-2040 concentration is crucial in optimizing the production of HbF. As we have also observed with hydroxyurea, the greatest increase in the g/g+b ratio is at high concentrations of OSI-2040 (300 and 750 nM), up to 25-fold, and is a consequence of reductions in both b- and g-globin. In contrast, a mid-level concentration (75 nM) yields a 2.5–4 fold increase in the g/(g+b) ratio with little or no cytotoxicity. These data suggest that like hydroxyurea, OSI-2040 may be effective in inducing HbF and may be a useful therapeutic alternative.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 54-54
Author(s):  
Kunhua Qin ◽  
Peng Huang ◽  
Anran Huang ◽  
Ruopeng Feng ◽  
Thiyagaraj Mayuranathan ◽  
...  

Sickle cell disease and some forms of b-thalassemia are disorders, which can be improved by therapies that elevate HbF, the fetal form of hemoglobin. It is currently thought that in adult type erythroid cells, silencing of the fetal type b-globin-like genes HBG1 and HBG2 is accomplished predominantly by two major transcription factors: BCL11A and ZBTB7A. However, it is unknown whether there are additional transcription factors that contribute to the repression of the HBG1/2 genes. Here, using a DNA-binding domain focused CRISPR-Cas9 screening approach, we identified NFIA as a novel regulator of HbF expression. NFIA belongs to the NFI transcription factor family, which is composed of NFIA, NFIB, NFIC and NFIX. NFIA and NFIX are the predominantly expressed forms in human erythroid cells, and their expression is higher in adult erythroblast cells when compared to fetal erythroid cells. CRISPR-Cas9 mediated disruption of NFIA in an immortalized human umbilical cord blood-derived erythroid progenitor cell line 2 (HUDEP2) or CD34+ hematopoietic stem and progenitor cell-derived primary erythroblast cells led to a modest reactivation of HBG1/2 mRNA expression, whereas disruption of NFIX had little effect. However, combined NFIA and NFIX disruption produced a substantial increase in HBG1/2 expression, suggesting that these factors function in a partially redundant manner. ChIP-seq and RNA-seq studies showed that NFIA and NFIX have comparable chromatin binding and activity profiles in human erythroid cells. ChIP-seq failed to detect NFI protein occupancy at or near the HBG1/2 genes. However, given the known difficulty in detecting repressor molecules by ChIP at the silent HBG1/2 genes [Martyn et al., Nature Genetics 2018; Liu et al., Cell 2018], we tested a direct involvement of NFI proteins by disrupting putative NFI binding sites near the HBG1/2 genes. Perturbation of one such NFI motif residing upstream of the transcription start site in the HBG1/2 promoters both in HUDEP2 and primary human erythroid cells markedly increased HBG1/2 mRNA levels, comparable to those achieved by combined disruption of NFIA and NFIX. Mutation of another putative NFI motif within intron1 in the HBG1/2 gene also significantly raised HbF levels. While these results implicate NFI proteins in the direct silencing of the HBG1/2 genes, the identity of the bound factors at the NFI motifs remains to be established. Studies are currently ongoing that use alternative approaches such as Cleavage Under Target & Release Using Nuclease (CUT & RUN) to map the chromatin occupancy of NFI factors at the HBG1/2 genomic region. We will also discuss results from ongoing studies of NFI factors in NBSGW mice models. In sum, we uncovered NFI transcription factors as a novel HbF regulator suggesting that the silencing of HbF involves a transcription factor network that is more complex than previously appreciated. Disclosures Weiss: Rubius Inc.: Consultancy, Current equity holder in private company; Cellarity Inc.: Consultancy, Current equity holder in private company; Novartis: Consultancy, Current equity holder in private company; Esperion Therapeutics: Consultancy, Current equity holder in private company; Beam Therapeuticcs: Consultancy, Current equity holder in private company. Blobel:Pfizer: Research Funding; Fulcrum Therapeutics: Consultancy.


2018 ◽  
Author(s):  
Yanrong Li ◽  
Ying Liao ◽  
Qiaona Niu ◽  
Feng Gu ◽  
Yingjie Sun ◽  
...  

ABSTRACTNewcastle disease virus (NDV) causes severe infectious disease in poultry, and selectively kills tumor cells by inducing apoptosis. In this report, we revealed the mechanisms underlying NDV-induced apoptosis via investigation of endoplasmic reticulum (ER) stress-related unfolded protein response (UPR) in HeLa cells. We found that NDV infection induced the expression of pro-apoptotic transcription factor CHOP via PKR-eIF2α pathway. Knock down and exogenous expression studies showed that CHOP promoted cell apoptosis by down-regulation of anti-apoptotic protein BCL-2 and MCL-1, promotion of pro-apoptotic JNK and p38 signaling, and suppression of pro-survival AKT signaling. Meanwhile, CHOP facilitated NDV proliferation. Furthermore, virus infection activated IRE1α, another ER stress sensor, thereby promoting the mRNA splicing of XBP1 and resulting in the translation of transcription factor XBP1s. XBP1s entered into cell nucleus, promoted the expression of ER chaperones and components of ER associated degradation (ERAD). Exogenous expression of XBP1s helped IBV proliferation, and silence of XBP1s reduced virus proliferation. Meanwhile, exogenous expression and knock down studies demonstrated that IRE1α activated pro-apoptotic JNK signaling, promoted apoptosis and inflammation. In conclusion, our current study demonstrates that the induction of CHOP and activation of IRE1α-XBP1/JNK signaling cascades promote apoptosis and benefit NDV proliferation.IMPORTANCEIt is well known that NDV kills host animal and tumor cells by inducing cell apoptosis. Although several studies investigate the apoptotic phenomena in NDV-infected tumor cells, the molecular mechanisms underlying this oncolytic virus induced apoptosis is not well understood yet. In this study, we focus on characterization of the ER stress responses in NDV-infected tumor cells, and find that virus induces apoptosis by up-regulation or activation of several unfolded protein responses (UPR) related transcription factors and signaling: such as ATF4, CHOP and XBP1s, and pro-apoptotic kinases (IRE1α, JNK, p38). Moreover, activation of these transcription factors and signaling cascades helps virus proliferation. Our study dissects the UPR induced apoptosis in NDV-infected tumor cells, and provides the evidence that UPR favors NDV proliferation.


Sign in / Sign up

Export Citation Format

Share Document