A Functional Screen of Genes Expressed in AML Patients Identifies Novel Transforming Properties of IL27R.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1454-1454
Author(s):  
Anuradha Pradhan ◽  
Que Lambert ◽  
Gary Reuther

Abstract A thorough understanding of the signaling pathways required for AML formation is necessary in order to develop much needed therapeutic treatments for AML patients. While there are many known mutations that contribute to AML, additional unknown genes likely exist, since the leukemic cells of a significant number of AML patients are cytogenetically normal and AML likely forms as a result of two mutations in different classes of oncogenes. Therefore, in order to identify novel mutations and signaling pathways that contribute to AML, we undertook functional genetic screening for genes, expressed in the leukemic cells of AML patients, that could transform myeloid cells in culture. To do this we generated cDNA libraries from cytogenetically normal AML patients and screened 32D myeloid cells for cytokine independent growth. We did not identify any genes capable of transforming 32D cells to cytokine independence by this approach, likely because the apoptotic response upon cytokine removal is very rapid. We then performed screens in 32D cells that exogenously express the anti-apoptotic Bcl2 protein in order to attempt to sensitize these cells to transformation by unknown oncogenes. These cells undergo cell death much slower than parental 32D cells in the absence of cytokine. From these screens, we identified IL27Ra (also known as TCCR and WSX1) as a gene that can induce the cytokine-independent growth of 32D cells. Interestingly, IL27R is capable of transforming 32D cells that lack exogenous Bcl2 expression, suggesting that our use of Bcl2 sensitized 32D cells to transformation in the context of expressing a library of genes in these cells. Therefore, this approach may allow for the identification of AML oncogenes in a relevant cell system. IL27Ra (IL27R) is a type I cytokine receptor that functions as the ligand-binding component for the receptor for IL-27 and functions with the gp130 co-receptor to induce signal transduction in response to IL-27. Cytokine independent 32D/IL27R cells contain elevated levels of activated forms of various signaling proteins, including JAK1, JAK2, STAT1, STAT5, ERK1/2, and SHP-2. Activation of these signaling proteins is dependent on the kinase activity of JAK family proteins as the pan JAK inhibitor, JAK inhibitor I, blocked activation of these signaling proteins. JAK inhibitor I also induced apoptotic cell death in 32D cells transformed to cytokine independence by IL27R, suggesting the transforming properties of IL27R are dependent on the activity of JAK family members. In addition, IL27R can transform BaF3 pro-B cells to cytokine independence. Since BaF3 cells lack expression of the gp130 co-receptor for IL-27, this suggests that IL27R-mediated transformation of hematopoietic cells is gp130-independent. This is a novel finding for IL27R-dependent signal transduction. IL27R has primarily been studied in the context of T-cell regulation. Our results are very exciting because our work is the first to suggest that IL27R may function as a hematopoietic cell oncogene. This is also interesting because type I cytokine receptors are required for activation of a JAK2 mutant (V617F) found in various myeloproliferative disorders (MPDs). We also show that IL27R is capable of activating JAK2-V617F in cells. Our data demonstrate that IL27R possesses hematopoietic cell transforming properties and suggest that type I cytokine receptors, such as IL27R, may play unappreciated roles in MPDs, including AML.

2000 ◽  
Vol 151 (2) ◽  
pp. 199-208 ◽  
Author(s):  
Thomas Harder ◽  
Marina Kuhn

Activation of T cell antigen receptor (TCR) induces tyrosine phosphorylations that mediate the assembly of signaling protein complexes. Moreover, cholesterol-sphingolipid raft membrane domains have been implicated to play a role in TCR signal transduction. Here, we studied the assembly of TCR with signal transduction proteins and raft markers in plasma membrane subdomains of Jurkat T leukemic cells. We employed a novel method to immunoisolate plasma membrane subfragments that were highly concentrated in activated TCR–CD3 complexes and associated signaling proteins. We found that the raft transmembrane protein linker for activation of T cells (LAT), but not a palmitoylation-deficient non-raft LAT mutant, strongly accumulated in TCR-enriched immunoisolates in a tyrosine phosphorylation–dependent manner. In contrast, other raft-associated molecules, including protein tyrosine kinases Lck and Fyn, GM1, and cholesterol, were not highly concentrated in TCR-enriched plasma membrane immunoisolates. Many downstream signaling proteins coisolated with the TCR/LAT-enriched plasma membrane fragments, suggesting that LAT/TCR assemblies form a structural scaffold for TCR signal transduction proteins. Our results indicate that TCR signaling assemblies in plasma membrane subdomains, rather than generally concentrating raft-associated membrane proteins and lipids, form by a selective protein-mediated anchoring of the raft membrane protein LAT in vicinity of TCR.


Genomics ◽  
2013 ◽  
Vol 101 (2) ◽  
pp. 101-112 ◽  
Author(s):  
Fransiskus X. Ivan ◽  
K.S. Tan ◽  
M.C. Phoon ◽  
Bevin P. Engelward ◽  
Roy E. Welsch ◽  
...  

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2711-2711
Author(s):  
Ravi Dashnamoorthy ◽  
Frederick Lansigan ◽  
Wilson L Davis ◽  
Nancy Kuemmerle ◽  
William B Kinlaw ◽  
...  

Abstract Abstract 2711 Background: Fatty acid synthase (FASN) is a key enzyme of fatty acid synthesis and is upregulated in many cancers. Increased FASN in cancer is associated with poor prognosis, while inhibition of FASN results in cancer cell death. The MEK/ERK signal transduction is one of the primary pathways that activate tumor-related FASN. Lipoprotein lipase (LPL) is also involved in fatty acid metablishm as it releases free fatty acid (FFA) from circulating lipoproteins, making them available for cellular uptake. Notably, these concepts have emerged primarily from solid tumor studies; there is a comparative paucity of data in lymphoma. We examined the functional roles of FASN and LPL in DLBCL cells and their interaction with oncogenic signal transduction pathways including MEK/ERK and an upstream target, hypoxia inducible factor-1 alpha (HIF-1a). We also investigated potential therapeutic implications of targeting fatty acid metabolism for the treatment of DLBCL. Methods: We used the DLBCL cell lines OCI-LY3, OCI-LY19, SUDHL4, and SUDHL10 in normoxic or hypoxic (0.2% O2) conditions. Cerulenin (FASN inhibitor) and Orlistat (FASN and LPL inhibitor) were utilized to examine the effect of fatty acid enzyme inhibition on cell signaling and cell death. We assessed cell viability with the MTT assay and apoptosis by flow cytometric analysis of Annexin-V/propidium iodide (PI). FASN and LPL mRNAs were quantified in DLBCL cell lines by RT-PCR as well as through gene expression profiling (GEP) analysis (by cell of origin) using the CaBIG dataset. Further, FASN and associated signaling pathways (MEK, ERK, and HIF-1a) were analyzed by Western blot. Finally, for investigation of potential interactions between FASN and HIF-1a, or MAPK signaling, we utilized short hairpin RNA interference (shRNA) to knock down (KD) pathways of interest. Results: FASN protein expression was readily detectable in all DLBCL cell lines in normoxia, while the expression of LPL was barely detectable in most cells, except in SUDHL10 and only in hypoxic conditions. RT-PCR showed that all DLBCL cell lines tested expressed high levels of FASN mRNA, while minimal levels of LPL could be detected; GEP showed that FASN was expressed more prominently in germinal center (GC) DLBCL (p=0.0006 vs GC control and p=0.0001 vs non-GC DLBCL), whereas LPL was preferentially expressed in non-GC DLBCL (p<0.0001 vs non-GC control and GC DLBCL). We next examined FASN expression following KD of MEK, ERK, or HIF-1a using shRNA in OCI-LY3 and SUDHL10 cells. HIF-1a KD significantly decreased FASN expression; this result was most prominent in OCI-LY3 cells, although it was also evident in SUDHL10. Interestingly, MEK and ERK KDs had minimal effect on FASN or LPL. Pharmacologic treatment with cerulenin, however, resulted in inhibition of MEK and ERK phosphorylation in OCI-LY3 cells. Additionally, treatment with Cerulenin or Orlistat (0.25–4 μg/mL for 48 hours) resulted in dose-dependent cytotoxicity across several DLBCL cell lines (OCI-LY3, SUDHL4, and SUDHL10) with an approximate IC50 of 1μg/mL in all lines. Furthermore, treatment with Cerulenin resulted in induction of apoptosis, which was mediated by caspase cleavage (caspases 3, 8 and 9) in SUDHL4 and OCI-LY3 cells. Conclusions: We demonstrated that FASN is constitutively activated in DLBCL with expression in part dependent on cell of origin, while LPL protein or message were mostly down-regulated. HIF-1a is a constitutively activated oncogenic pathway in DLBCL (Evens AM, et al. Br J Haematol 2008) and it appeared here to directly regulate FASN expression. In addition, we showed that targeting fatty acid metabolism may be harnessed as a potential therapeutic strategy. Further investigations are required to delineate the mechanisms through which MAPK and HIF-1a regulate FASN expression and to determine the in vivo implications of FASN inhibition on DLBCL tumor growth. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 8 ◽  
pp. 204993612110149
Author(s):  
Sandra-Georgina Solano-Gálvez ◽  
Diego-Abelardo Álvarez-Hernández ◽  
Laila Gutiérrez-Kobeh ◽  
Rosalino Vázquez-López

The maintenance of homeostasis in living systems requires the elimination of unwanted cells which is performed, among other mechanisms, by type I cell death or apoptosis. This type of programmed cell death involves several morphological changes such as cytoplasm shrinkage, chromatin condensation (pyknosis), nuclear fragmentation (karyorrhexis), and plasma membrane blebbing that culminate with the formation of apoptotic bodies. In addition to the maintenance of homeostasis, apoptosis also represents an important defense mechanism for cells against intracellular microorganisms. In counterpart, diverse intracellular pathogens have developed a wide array of strategies to evade apoptosis and persist inside cells. These strategies include the manipulation of signaling pathways involved in the inhibition of apoptosis where mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) play a key role. Leishmania is an intracellular protozoan parasite that causes a wide spectrum of diseases known as leishmaniasis. This parasite displays different strategies, including apoptosis inhibition, to down-regulate host cell defense mechanisms in order to perpetuate infection.


2005 ◽  
Vol 23 (10) ◽  
pp. 2411-2422 ◽  
Author(s):  
Florence Lefranc ◽  
Jacques Brotchi ◽  
Robert Kiss

Purpose The present review aims to emphasize that malignant gliomas are characterized by the diffuse invasion of distant brain tissue by a myriad of single migrating cells that exhibit decreased levels of apoptosis (programmed cell death type I), thus a resistance to cytotoxic insult. Methods The present review surveys the molecular mechanisms of migration in malignant gliomas and potential issues arising from treatments, in addition to relationships between glioma cell migration and resistance to apoptosis in terms of the molecular signaling pathways. Results Clinical and experimental data demonstrate that glioma cell migration is a complex combination of multiple molecular processes, including the alteration of tumor cell adhesion to a modified extracellular matrix, the secretion of proteases by the cells, and modifications to the actin cytoskeleton. Intracellular signaling pathways involved in the acquisition of resistance to apoptosis by migrating glioma cells concern PI3K, Akt, mTOR, NF-κB, and autophagy (programmed cell death type II). Conclusion A number of signaling pathways can be constitutively activated in migrating glioma cells, thus rendering these cells resistant to cytotoxic insults. However, these pathways are not all constitutively activated at the same time in any one glioma. Particular inhibitors should therefore only be chosen if the target is present in the tumor tissue, but this is only possible if individual patients are submitted to the molecular profiling of their tumors before undergoing any treatment to combat their migratory glioma cells. Specific antimigratory compounds should be added to conventional radio- and/or chemotherapy.


Blood ◽  
1991 ◽  
Vol 78 (4) ◽  
pp. 953-960 ◽  
Author(s):  
J Lotem ◽  
EJ Jr Cragoe ◽  
L Sachs

Growth factor-independent clones of myeloid leukemic cells can regain a growth factor-dependent state during differentiation. Loss of viability in these differentiating leukemic cells in the absence of growth factor was associated with DNA fragmentation and morphologic changes typical of programmed cell death (apoptosis). The differentiating leukemic cells could be rescued from apoptosis by a hematopoietic growth factor such as interleukin-3 (IL-3) and by the tumor-promoting phorbol ester 12-O-tetra-decanoyl-phorbol-13-acetate (TPA), but not by the nonpromoting phorbol ester 4-alpha-TPA. IL-3 and TPA rescued differentiating myeloid leukemic cells by different pathways and also rescued normal myeloid precursor cells from apoptosis. The rescue of differentiating leukemic and normal myeloid cells by IL-3 or TPA was blocked by amiloride inhibitors of the Na+/H+ antiporter. We suggest that TPA may act as a tumor promoter by inhibiting programmed cell death.


Blood ◽  
1991 ◽  
Vol 78 (4) ◽  
pp. 953-960 ◽  
Author(s):  
J Lotem ◽  
EJ Jr Cragoe ◽  
L Sachs

Abstract Growth factor-independent clones of myeloid leukemic cells can regain a growth factor-dependent state during differentiation. Loss of viability in these differentiating leukemic cells in the absence of growth factor was associated with DNA fragmentation and morphologic changes typical of programmed cell death (apoptosis). The differentiating leukemic cells could be rescued from apoptosis by a hematopoietic growth factor such as interleukin-3 (IL-3) and by the tumor-promoting phorbol ester 12-O-tetra-decanoyl-phorbol-13-acetate (TPA), but not by the nonpromoting phorbol ester 4-alpha-TPA. IL-3 and TPA rescued differentiating myeloid leukemic cells by different pathways and also rescued normal myeloid precursor cells from apoptosis. The rescue of differentiating leukemic and normal myeloid cells by IL-3 or TPA was blocked by amiloride inhibitors of the Na+/H+ antiporter. We suggest that TPA may act as a tumor promoter by inhibiting programmed cell death.


2021 ◽  
Author(s):  
Marina Saisana ◽  
S. Michael Griffin ◽  
Felicity E. B. May

Abstract Background Gastric adenocarcinoma is common and consequent mortality high. Presentation and mortality are increased in obese individuals, many of whom have elevated circulating insulin concentrations. High plasma insulin concentrations may promote, and increase mortality from, gastric adenocarcinoma. Tumour promotion activities of insulin and its receptor are untested in gastric cancer cells. Methods Tumour gene amplification and expression were computed from sequencing and microarray data. Associations with patient survival were assessed. Insulin-dependent signal transduction, growth, apoptosis and anoikis were analysed in metastatic cells from gastric adenocarcinoma patients and in cell lines. Receptor involvement was tested by pharmacological inhibition and genetic knockdown. RNA was analysed by RT-PCR and proteins by western transfer and immunofluorescence. Results INSR expression was higher in tumour than in normal gastric tissue. High tumour expression was associated with worse patient survival. Insulin receptor was detected readily in metastatic gastric adenocarcinoma cells and cell lines. Isoforms B and A were expressed. Pharmacological inhibition prevented cell growth and division, and induced caspase-dependent cell death. Rare tumour INS expression indicated tumours would be responsive to pancreatic or therapeutic insulins. Insulin stimulated gastric adenocarcinoma cell PI3-kinase/Akt signal transduction, proliferation, and survival. Insulin receptor knockdown inhibited proliferation and induced programmed cell death. Type I IGF receptor knockdown did not induce cell death. Conclusions The insulin and IGF signal transduction pathway is dominant in gastric adenocarcinoma. Gastric adenocarcinoma cell survival depends upon insulin receptor. That insulin has direct cancer-promoting effects on tumour cells has implications for clinical management of obese and diabetic cancer patients.


Sign in / Sign up

Export Citation Format

Share Document