Open Access

Nuclear lymphocyte-specific protein tyrosine kinase and its interaction with CR6-interacting factor 1 promote the survival of human leukemic T cells

  • Authors:
    • Shahrooz Vahedi
    • Fu-Yu Chueh
    • Sujoy Dutta
    • Bala Chandran
    • Chao-Lan Yu
  • View Affiliations

  • Published online on: May 19, 2015     https://doi.org/10.3892/or.2015.3990
  • Pages: 43-50
  • Copyright: © Vahedi et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 3.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Overexpression and hyperactivation of lymphocyte-specific protein tyrosine kinase (Lck) have been associated with leukemia development. We previously showed that, other than its known function as a cytoplasmic signal transducer, Lck also acts as a nuclear transcription factor in mouse leukemic cells. In the present study, we demonstrated the presence of nuclear Lck in human leukemic T cells and in primary cells. We further established a positive correlation between Lck nuclear localization and its kinase activity. Proteomic analysis identified CR6-interacting factor 1 (CRIF1) as one of the Lck-interacting proteins. CRIF1 and Lck association in the nucleus was confirmed both by immunofluorescence microscopy and co-immunoprecipitation in human leukemic T cells. Close-range interaction between Lck and CRIF1 was validated by in situ proximity ligation assay (PLA). Consistent with the role of nuclear CRIF1 as a tumor suppressor, CRIF1 silencing promotes leukemic T cell survival in the absence of growth factors. This protective effect can be recapitulated by endogenous Lck or reconstituted Lck in leukemic T cells. All together, our results support a novel function of nuclear Lck in promoting human leukemic T cell survival through interaction with a tumor suppressor. It has important implications in defining a paradigm shift of non-canonical protein tyrosine kinase signaling.

Introduction

Protein tyrosine kinases (PTKs) are a family of enzymes regulating diverse cellular functions that are associated with cancer hallmarks, such as cell survival (1). Many PTKs are plasma membrane-associated signal transducers that transmit signals into the nucleus in response to environmental cues. However, accumulating evidence indicates that PTKs also translocate to the nucleus and exert additional functions. Nuclear PTKs are known to change the stability and activity of nuclear residing proteins as well as to regulate nuclear gene expression (2). For example, epidermal growth factor receptor (EGFR) can regulate nuclear activity either by functioning as a transcription factor or by phosphorylating histone H4 (3,4). EGFR is present in the nuclei of proliferating liver cells and breast cancer cells (5,6). Nuclear EGFR binds to the cyclin D1 promoter region and upregulates cyclin D1 expression to promote breast cancer cell cycle progression (6). In breast cancer cells, ErbB2 also interacts with and phosphorylates Cdc2 in the nucleus to confer resistance to Taxol-induced apoptosis (7). In addition to EGFR, other receptor and non-receptor PTKs have been detected in the nuclei of solid tumors (8,9). However, the role of nuclear PTKs in blood cancer is largely unknown.

Lymphocyte-specific protein tyrosine kinase (Lck) is a Src family kinase (SFK) predominantly expressed in T cells and plays a pivotal role in normal T cell development and homeostasis (10,11). The gene coding for Lck is localized near the chromosomal region with a high frequency of translocation (12). Overexpression and hyperactivation of Lck have been reported in both acute and chronic leukemias (13). Lck overexpression is also linked to poor clinical outcome to prednisone treatment in acute B lymphoblastic leukemia patients (14). In addition to blood malignancies, abnormally high expression and activity of Lck have been reported in solid tumors, such as colorectal and prostate cancers (15,16). Under physiological conditions, Lck is associated with plasma membrane and propagates signals initiated from the T cell receptors (17). However, immunohistochemical analysis of specimens from breast cancer patients revealed the presence of nuclear Lck (18). It suggests that nuclear localization of Lck may also be associated with malignant progression of hematopoietic cells.

Our previous study demonstrated that, in mouse LSTRA leukemia, Lck upregulated the expression of the LIM domain only 2 gene through direct binding to its promoter region (19). We further provided evidence supporting the mouse LSTRA leukemic cell line as a model for the aggressive form of human large granular lymphocyte leukemia (20). These findings led us to hypothesize that Lck may also exhibit additional functions in the nuclear compartment of human leukemic cells. In the present study, we used the well-defined human T leukemic cell line Jurkat to examine the biological outcome and underlying mechanism of Lck nuclear translocation.

Materials and methods

Cell lines and reagents

Human Jurkat E6.1 and Jcam 1.6 T cell lines and the mouse LSTRA T cell line were maintained as described previously (21). The Jcam 1.6 cell line transfected with an expression vector containing the wild-type Lck (Jcam/Lck) was a generous gift from Dr Steven Burakoff (Icahn School of Medicine at Mount Sinai, New York City, NY, USA). CR6-interacting factor 1 (CRIF1)-knockdown stable cell lines were generated from Jcam cells using lentiviral transduction. CRIF1 shRNA (sc-97804-V) and scrambled shRNA control (sc-108080) lentiviral particles were purchased from Santa Cruz Biotechnology (Dallas, TX, USA). After 24-h serum starvation, 104 Jcam cells were harvested and resuspended in 50 μl of freshly thawed virus mixture (2×105 infectious units). After a 6-h incubation, 500 μl of complete RPMI-1640 was added. After 1 day of recovery, puromycin was added to a final concentration of 14 μg/ml and cells were selected for 1 week.

Mouse splenocyte isolation

Splenocytes were isolated from BALB/c mouse spleens as described previously (20). Briefly, the spleen was dissociated by gently pressing it through a 70-μm cell strainer with a syringe plunger. Red blood cells were lysed by resuspending the cells in ammonium-chloride-potassium (ACK) lysis buffer at room temperature for 5 min. Splenocytes were then washed with cold RPMI-1640 and kept on ice for further analysis. The use of animals was approved by the Institutional Animal Care and Use Committee (IACUC) of the university.

Pervanadate activation and dasatinib treatment

Mouse splenocytes and human T cell lines were stimulated with freshly prepared pervanadate at the final concentration of 100 μM at 37°C for 5 min as described previously (22). Dasatinib was purchased from LC Laboratories (Woburn, MA, USA) and kept at −20°C as a 10-mM stock solution in DMSO. For dasatinib treatment, 107 Jurkat cells were treated with DMSO as control or 500 nM of dasatinib at 37°C for 2 h. The reactions were stopped by quick cool down on ice. Pervanadate, dasatinib and DMSO control were then removed by washing cells in ice-cold phosphate-buffered saline.

Subcellular fractionation

Cells were lysed by swelling in hypotonic buffer followed by passing through a 27-gauge needle. Light microscopy was used to ensure cell rupture before proceeding to the next step. The nuclear fraction was collected by differential centrifugation as described previously (19). Fraction purity was verified by immunoblotting of specific markers.

Immunoprecipitation and immunoblotting

Whole cell and nuclear lysates were prepared by solubilizing whole cell and nuclear pellets in RIPA buffer, respectively. For co-immuno-precipitation experiments, proteins were extracted from the nuclear pellets using high-salt buffer (19). Target proteins were either immunoprecipitated or directly detected from total lysates after SDS-PAGE using specific antibodies according to manufacturers’ instructions. Antibodies specific for Lck, CRIF1 and epidermal growth factor receptor substrate 15 (Eps15) were purchased from Santa Cruz Biotechnology. Antibodies specific for phospho-Src family (Tyr416) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) were purchased from Cell Signaling Technology (Danvers, MA, USA). Anti-lamin B1 antibody was purchased from Abcam (Cambridge, MA, USA). Appropriate secondary antibodies conjugated with horseradish peroxidase were used to detect signals by enhanced chemiluminescence system. For signal quantitation, the bands were digitalized and analyzed by ImageJ software.

Confocal immunofluorescence microscopy

The cells were adhered to 10-well slides, fixed, and permeabilized as previously described (21). The cells were blocked with Image-iT FX Signal Enhancer (Life Technologies, Grand Island, NY, USA) for 15 min at room temperature, and then either singly or doubly stained with primary antibodies. Subsequent labeling with Alexa Fluor-conjugated secondary antibodies and DAPI counterstain (Life Technologies) were performed to visualize primary antibodies and nuclei, respectively. Stained cells were viewed using the Olympus FV10i fluorescence confocal microscope. Images were analyzed using FluoView software (Olympus, Melville, NY, USA).

In situ proximity ligation assay (PLA) microscopy

PLA was performed using the DuoLink PLA kit (Sigma-Aldrich, St. Louis, MO, USA) according to the manufacturer’s instructions. Briefly, 104 cells were seeded on each well of 10-well slides. The adhered cells were fixed with 4% paraformaldehyde and then permeabilized with 0.2% Triton X-100. After treatment with DuoLink blocking buffer, the cells were incubated with diluted primary antibodies for Lck and CRIF1. After washing, the cells were incubated with species-specific PLA probes and two additional oligonucleotides under hybridization conditions. Hybridization occurs when PLA probes are in close proximity, which can be subsequently ligated to form a closed circle. A rolling-circle amplification step follows with polymerase to generate a concatemeric product, which can be visualized with fluorephore-labeled oligonucleotides after hybridization. The slides were counterstained with DAPI and analyzed by fluorescence microscope.

Transient transfection

Approximately 104 HEK293T cells were plated in a 10-cm dish in 10 ml of DMEM supplemented with 10% fetal bovine serum. Cells under 70% confluency were transiently transfected with 18 μg of plasmids using calcium phosphate precipitation protocol (23). The cells were harvested between 42–46 h after transfection. Wild-type Lck construct as well as constitutively active (Y505F), kinase dead (Y505F/K273R), and basal kinase (Y505F/Y394F) Lck mutant constructs have been described previously (23).

Cell number and viability

Human T cell lines were washed in plain RPMI-1640 and then incubated in serum-free RPMI-1640 at 37°C with 5% CO2 for 48 h. Cell viability was determined by incubation of a small fraction of cells in 0.1% trypan blue dye. Both viable and dead cells were counted with a hemocytometer under light microscope to calculate the percentage of dead cells.

Statistical analysis

Data are presented as mean ± SE from at least three independent experiments. The significance of differences was analyzed using a Student’s t-test (SigmaPlot 11; SPSS Inc., Chicago, IL, USA). Differences were considered significant at P<0.05.

Results

Lck is present in the nuclear compartment of leukemia cells

LSTRA is a mouse leukemic cell line that overexpresses active Lck kinase (24). We previously showed that a large percentage of endogenous Lck was accumulated in LSTRA nuclei (Fig. 1A, lane 1) (19). To determine whether Lck trans-locates to the nuclei in primary cells, we examined mouse splenocytes by immunofluorescence microscopy using an anti-Lck antibody. As shown in Fig. 1C, Lck staining was mostly cytoplasmic in the resting cells (upper panel). To maximally activate Lck tyrosine phosphorylation, we stimulated mouse splenocytes with pervanadate, which is a potent tyrosine phosphatase inhibitor. As shown in Fig. 1C, a significant amount of Lck translocated to the nucleus of the pervanadate-stimulated cells (lower panel). This result confirms the presence of nuclear Lck in primary cells and further suggests the involvement of Lck phosphorylation in its nuclear translocation.

The Jurkat cell line was isolated from an acute T lympho-blastic leukemia patient and is one of the best characterized human leukemic T cell lines (25). We examined the presence of nuclear Lck by subcellular fractionation of Jurkat cells to isolate nuclear proteins (Fig. 1A, lane 2). Subsequent immunoblotting detected the presence of nuclear Lck (top panel) in the absence of cytoplasmic contamination (bottom panel). Confocal microscopy after staining with the anti-Lck antibody further confirmed localization of endogenous Lck in the Jurkat nucleus (Fig. 1B). These findings support a potential role of nuclear Lck in both mouse and human T cell leukemias.

Kinase activity enhances Lck nuclear translocation

Similar to other SFKs, Lck activity is tightly regulated by phosphorylation of two key regulatory tyrosine residues (26). Phosphorylation of the negative regulatory Y505 results in a closed conformation and reduced Lck kinase activity. On the other hand, phosphorylation of the positive regulatory Y394 confers a fully active Lck kinase with an open conformation. To establish a correlation between Lck kinase activity and its nuclear translocation, we analyzed wild-type and three mutant Lck proteins. The Y505F mutant is locked in an open conformation and becomes constitutively active. Additional mutation on Y394 (Y505F/Y394F) reduces the kinase activity; while mutation on the key residue in kinase domain (K273R) abolishes the kinase activity. All four constructs were transfected into HEK293T cells that lack endogenous Lck. Lck immunoblotting of nuclear versus total proteins (Fig. 2A) and subsequent quantitation (Fig. 2B) showed a step-wise reduction in nuclear Lck accumulation with decreasing kinase activity.

To further establish the role of Lck kinase activity in its nuclear translocation in human T cells, we examined both Jurkat and its Lck-deficient derivative Jcam cell lines. In the Jcam cells, Lck is inactive due to truncation and is expressed at a very low level (27). Phosphorylation of the positive regulatory Y394 is indicative of Lck kinase activity. We determined the level of Y394 phosphorylation by Lck immunoprecipitation and subsequent immunoblotting using an antibody that specifically recognizes phosphorylation of this conserved tyrosine residue in all SFKs. As shown in Fig. 3A, in the Jcam cells, the truncated kinase-dead Lck (lanes 7 and 8) could not translocate to the nucleus either without (lane 4) or with (lane 3) pervanadate stimulation. On the other hand, pervanadate stimulation of Jurkat cells greatly enhanced Y394 phosphorylation and nuclear translocation of Lck (Fig. 3A, lanes 1 and 2). This was consistent with pervanadate-induced Lck translocation into the nuclei of primary cells (Fig. 1C). As an independent confirmation, we treated Jurkat cells with an SFK inhibitor, dasatinib. As shown in Fig. 3B, in the absence of SFK activity (compare lanes 1 and 2), the amount of nuclear Lck was greatly diminished (compare lanes 5 and 6) in the Jurkat cells.

Lck interacts with CRIF1

We previously showed that Lck may function as a transcription factor by binding to specific target genes (19). To identify additional Lck-interacting partners, we performed mass spectrometry on Lck immunoprecipitates prepared from LSTRA lysates. Our previous proteomic analysis identified CRIF1 as one of the top candidates (unpublished data). To confirm the interaction between Lck and CRIF1 in human leukemic T cells, we performed immunofluorescence microscopy and detected the co-localization of Lck and CRIF1 in the Jurkat nucleus (Fig. 4A). Co-immunoprecipitation between Lck and CRIF1 in nuclear extracts prepared from Jurkat cells also confirmed the association of Lck and CRIF1 (Fig. 4B, lane 2).

To further validate the close-range interaction between Lck and CRIF1, we performed in situ PLA microscopy. A positive PLA result relies on two molecules in the proximity of 16 nm or below, which reflects true protein-protein interaction. As shown in Fig. 4C, a PLA signal was detected in the Jurkat nucleus (right panels). Additional PLA staining was observed outside the nuclei of Jurkat cells (Fig. 4C, right panels). This is consistent with our earlier observation of Lck interaction with CRIF1 in mitochondria (unpublished data). As a negative control, no PLA signal was detected in the Lck-deficient Jcam cells (Fig. 4C, left panels). All together, these results support a close interaction between Lck and CRIF1 in the nuclear compartment of Jurkat cells.

Lck promotes leukemic T cell survival by inhibiting CRIF1 function

Nuclear CRIF1 is known as a tumor suppressor by blocking cell cycle progression and reducing cell survival. The tumor-suppressing activities of CRIF1 are largely mediated by its interaction with distinct nuclear proteins, such as cyclin-dependent kinase 2 (CDK2) and Nur77 (28,29). To determine the role of CRIF1 in human leukemic T cell survival in the absence of Lck, we knocked down CRIF1 expression in Jcam cells by RNA interference (Fig. 5A, lane 2). Both the control and CRIF1-knockdown Jcam cells were subjected to serum starvation to compare their sensitivity to growth factor deprivation. Consistent with the role of CRIF1 as a tumor suppressor, the CRIF1-knockdown Jcam cells were more resistant to starvation-induced cell death (Fig. 5B).

Since Lck binds to CRIF1 in the nucleus (Fig. 4), we hypothesized that Lck interaction with CRIF1 may interfere with CRIF1’s function as a tumor suppressor. Indeed, similar to Jcam cells with CRIF1 knockdown (Fig. 5B), Jurkat cells were more resistant to starvation-induced cell death as compared to Jcam cells (Fig. 5D). To verify the contribution of Lck, wild-type Lck was reconstituted in Jcam cells and was capable of binding to CRIF1 (Fig. 5C, lane 3). The exogenous Lck confers protection from starvation-induced cell death to a level similar to endogenous Lck in Jurkat (Fig. 5D). These results support the role of Lck in promoting cell survival by its interaction with CRIF1 and, potentially, inhibition of CRIF1 activity.

Discussion

In summary, our present study revealed a novel function of nuclear Lck in promoting cell survival through interaction with a tumor suppressor. However, we do not exclude the possibility that nuclear Lck may exert additional functions. For example, our previous study demonstrated the role of Lck as a nuclear transcription factor in regulating genes important in oncogenesis (19). Other PTKs, such as EGFR, also exhibit multiple functions in the nucleus. Nuclear EGFR binds to the promoter regions of distinct target genes, upregulates gene expression, and promotes cell cycle progression (6,30). Interaction of EGFR with DNA protein kinase in the nucleus, on the other hand, enhances DNA damage repair and augments breast cancer cell’s resistance to cisplatin and ionizing radiation treatment (31). Additionally, modulation of PCNA and Cdc2 stability and activity through their association with nuclear EGFR contributes to uncontrolled proliferation and DNA repair in breast cancer cells (7,32). Our data further expand the biological significance of nuclear PTKs from receptor PTK to non-receptor PTK as well as from solid tumors to blood cancer.

Our results also support a positive effect of Lck kinase activity on its nuclear translocation. Consistent with our findings, kinase activity of EGFR is also important in its nuclear translocation in breast cancer cells (33). It should be noted, however, that a small amount of kinase-dead Lck (Fig. 2B) and inactive Lck (Fig. 3B) can still be detected in the nuclear compartment. It is possible that other kinase-independent mechanisms also contribute to Lck nuclear translocation. While a nuclear localization signal (NLS) has been identified in EGFR (34), there is no discernible NLS in Lck. It remains to be determined whether and how a non-classical NLS may mediate nuclear trafficking of Lck.

Our mass spectrometry analysis identified CRIF1 as a novel Lck-interacting partner. Consistent with its role as a tumor suppressor, CRIF1 interacts with and inhibits CDK2 to induce cell cycle arrest in leukemia cells (28). CRIF1-induced cell cycle arrest can also be mediated by its interaction with other nuclear proteins, including GADD45 and Nur77 (29,35). Our CRIF1 knockdown data in Jcam cells further support the role of CRIF1 in sensitizing leukemic cells to cell death, another important cancer hallmark (36). CRIF1 interaction with nuclear proteins may also contribute to its role in cell death. More importantly, our data suggest that Lck can bind to CRIF1 and inhibit its function as a tumor suppressor (Fig. 5). It remains to be determined whether Lck competes with other nuclear proteins in binding to CRIF1.

Other than an important nuclear regulator, CRIF also plays a critical role in the mitochondria (37). CRIF1 associates with the inner membrane of mitochondria and participates in the synthesis of oxidative phosphorylation polypeptides encoded by the mitochondrial circular genome. CRIF1 also facilitates their insertion into the inner membrane for the assembly of functional electron transport chain complex. As expected, CRIF1 deficiency in the mitochondrial compartment results in mitochondrial dysfunction. In brain-specific CRIF1-knockout mice, fatal neurodegeneration is associated with abnormal mitochondrial morphology (37). A recent study on an Alzheimer’s disease animal model further suggests a protective role of mitochondrial CRIF1 in neuronal cells against apoptosis (38). While the functions of nuclear CRIF1 are not fully addressed in neuronal cells, these studies do suggest that CRIF1 can promote the survival of distinct cell types, such as neuronal cells. In our T cell model, CRIF1 knockdown in Jcam cells also resulted in slightly more dead cells before serum deprivation (data not shown). However, under stress condition, CRIF1 promoted Jcam cell death (Fig. 5B). The overall effect of CRIF1 on cell survival, therefore, may depend on the cell type and the experimental condition.

Consistent with the dual functions of CRIF1 in different organelles, we also observed mitochondrial localization of CRIF1 in Jurkat leukemia (unpublished data). Similarly, we confirmed the presence of mitochondrial Lck in Jurkat T cells. The concomitant localization of Lck in the cytoplasm, mitochondria and nucleus further highlights the complexity of crosstalk between different subcellular compartments. Other receptor and non-receptor PTKs have been reported to exhibit diverse functions in the cytoplasm, mitochondria and nucleus (3941). These findings of non-canonical signaling represent an important paradigm shift of how PTKs coordinately regulate cellular activities. They also provide critical insights in identifying novel molecular targets in interfering with the oncogenic signal transduction network.

Acknowledgments

This study was supported in part by funds from the National Institute of Health R01 CA107210 (to C.-L.Y.) and the RFUMS-H.M. Bligh Cancer Research Fund (to B.C.). The Midwest Proteome Center (RFUMS) was supported in part by NIH S10 OD010662-01. We sincerely thank Xinli Yang, Patricia Loomis and Virginie Bottero for the technical assistance concerning mass spectrometry, confocal microscopy, and lentiviral transduction, respectively. We also thank Srividya Venkitachalam for her input in initiating this project.

Abbreviations:

PTK

protein tyrosine kinase

EGFR

epidermal growth factor receptor

SFK

Src family kinase

CRIF1

CR6-interacting factor 1

Eps15

epidermal growth factor receptor substrate 15

GAPDH

glyceraldehyde-3-phosphate dehydrogenase

PLA

proximity ligation assay

CDK2

cyclin-dependent kinase 2

NLS

nuclear localization signal

Lck

lymphocyte-specific protein tyrosine kinase

References

1 

Krause DS and Van Etten RA: Tyrosine kinases as targets for cancer therapy. N Engl J Med. 353:172–187. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Lo HW and Hung MC: Nuclear EGFR signalling network in cancers: Linking EGFR pathway to cell cycle progression, nitric oxide pathway and patient survival. Br J Cancer. 94:184–188. 2006. View Article : Google Scholar : PubMed/NCBI

3 

Lin SY, Makino K, Xia W, Matin A, Wen Y, Kwong KY, Bourguignon L and Hung MC: Nuclear localization of EGF receptor and its potential new role as a transcription factor. Nat Cell Biol. 3:802–808. 2001. View Article : Google Scholar : PubMed/NCBI

4 

Chou RH, Wang YN, Hsieh YH, Li LY, Xia W, Chang WC, Chang LC, Cheng CC, Lai CC, Hsu JL, et al: EGFR modulates DNA synthesis and repair through Tyr phosphorylation of histone H4. Dev Cell. 30:224–237. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Marti U, Burwen SJ, Wells A, Barker ME, Huling S, Feren AM and Jones AL: Localization of epidermal growth factor receptor in hepatocyte nuclei. Hepatology. 13:15–20. 1991. View Article : Google Scholar : PubMed/NCBI

6 

Hadzisejdić I, Mustać E, Jonjić N, Petković M and Grahovac B: Nuclear EGFR in ductal invasive breast cancer: Correlation with cyclin-D1 and prognosis. Mod Pathol. 23:392–403. 2010. View Article : Google Scholar

7 

Tan M, Jing T, Lan KH, Neal CL, Li P, Lee S, Fang D, Nagata Y, Liu J, Arlinghaus R, et al: Phosphorylation on tyrosine-15 of p34(Cdc2) by ErbB2 inhibits p34(Cdc2) activation and is involved in resistance to taxol-induced apoptosis. Mol Cell. 9:993–1004. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Domingues I, Rino J, Demmers JA, de Lanerolle P and Santos SC: VEGFR2 translocates to the nucleus to regulate its own transcription. PLoS One. 6:e256682011. View Article : Google Scholar : PubMed/NCBI

9 

Takahashi A, Obata Y, Fukumoto Y, Nakayama Y, Kasahara K, Kuga T, Higashiyama Y, Saito T, Yokoyama KK and Yamaguchi N: Nuclear localization of Src-family tyrosine kinases is required for growth factor-induced euchromatinization. Exp Cell Res. 315:1117–1141. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Van Laethem F, Tikhonova AN, Pobezinsky LA, Tai X, Kimura MY, Le Saout C, Guinter TI, Adams A, Sharrow SO, Bernhardt G, et al: Lck availability during thymic selection determines the recognition specificity of the T cell repertoire. Cell. 154:1326–1341. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Palacios EH and Weiss A: Function of the Src-family kinases, Lck and Fyn, in T-cell development and activation. Oncogene. 23:7990–8000. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Burnett RC, David JC, Harden AM, Le Beau MM, Rowley JD and Diaz MO: The LCK gene is involved in the t(1;7)(p34;q34) in the T-cell acute lymphoblastic leukemia derived cell line, HSB-2. Genes Chromosomes Cancer. 3:461–467. 1991. View Article : Google Scholar : PubMed/NCBI

13 

Majolini MB, Boncristiano M and Baldari CT: Dysregulation of the protein tyrosine kinase LCK in lymphoproliferative disorders and in other neoplasias. Leuk Lymphoma. 35:245–254. 1999. View Article : Google Scholar

14 

Accordi B, Espina V, Giordan M, VanMeter A, Milani G, Galla L, Ruzzene M, Sciro M, Trentin L, De Maria R, et al: Functional protein network activation mapping reveals new potential molecular drug targets for poor prognosis pediatric BCP-ALL. PLoS One. 5:e135522010. View Article : Google Scholar : PubMed/NCBI

15 

Veillette A, Foss FM, Sausville EA, Bolen JB and Rosen N: Expression of the lck tyrosine kinase gene in human colon carcinoma and other non-lymphoid human tumor cell lines. Oncogene Res. 1:357–374. 1987.PubMed/NCBI

16 

Robinson D, He F, Pretlow T and Kung HJ: A tyrosine kinase profile of prostate carcinoma. Proc Natl Acad Sci USA. 93:5958–5962. 1996. View Article : Google Scholar : PubMed/NCBI

17 

Chueh FY and Yu CL: Engagement of T-cell antigen receptor and CD4/CD8 co-receptors induces prolonged STAT activation through autocrine/paracrine stimulation in human primary T cells. Biochem Biophys Res Commun. 426:242–246. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Elsberger B, Fullerton R, Zino S, Jordan F, Mitchell TJ, Brunton VG, Mallon EA, Shiels PG and Edwards J: Breast cancer patients’ clinical outcome measures are associated with Src kinase family member expression. Br J Cancer. 103:899–909. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Venkitachalam S, Chueh FY and Yu CL: Nuclear localization of lymphocyte-specific protein tyrosine kinase (Lck) and its role in regulating LIM domain only 2 (Lmo2) gene. Biochem Biophys Res Commun. 417:1058–1062. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Chueh FY, Cronk RJ, Alsuwaidan AN, Mallers TM, Jaiswal MK, Beaman KD and Yu CL: Mouse LSTRA leukemia as a model of human natural killer T cell and highly aggressive lymphoid malignancies. Leuk Lymphoma. 55:706–708. 2014. View Article : Google Scholar

21 

Chueh FY, Leong KF and Yu CL: Mitochondrial translocation of signal transducer and activator of transcription 5 (STAT5) in leukemic T cells and cytokine-stimulated cells. Biochem. Biophys Res Commun. 402:778–783. 2010. View Article : Google Scholar

22 

Cooper JC, Shi M, Chueh FY, Venkitachalam S and Yu CL: Enforced SOCS1 and SOCS3 expression attenuates Lck-mediated cellular transformation. Int J Oncol. 36:1201–1208. 2010.PubMed/NCBI

23 

Venkitachalam S, Chueh FY, Leong KF, Pabich S and Yu CL: Suppressor of cytokine signaling 1 interacts with oncogenic lymphocyte-specific protein tyrosine kinase. Oncol Rep. 25:677–683. 2011.PubMed/NCBI

24 

Yu CL, Jove R and Burakoff SJ: Constitutive activation of the Janus kinase-STAT pathway in T lymphoma overexpressing the Lck protein tyrosine kinase. J Immunol. 159:5206–5210. 1997.

25 

Abraham RT and Weiss A: Jurkat T cells and development of the T-cell receptor signalling paradigm. Nat Rev Immunol. 4:301–308. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Shi M, Cooper JC and Yu CL: A constitutively active Lck kinase promotes cell proliferation and resistance to apoptosis through signal transducer and activator of transcription 5b activation. Mol Cancer Res. 4:39–45. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Straus DB and Weiss A: Genetic evidence for the involvement of the lck tyrosine kinase in signal transduction through the T cell antigen receptor. Cell. 70:585–593. 1992. View Article : Google Scholar : PubMed/NCBI

28 

Ran Q, Hao P, Xiao Y, Xiang L, Ye X, Deng X, Zhao J and Li Z: CRIF1 interacting with CDK2 regulates bone marrow microenvironment-induced G0/G1 arrest of leukemia cells. PLoS One. 9:e853282014. View Article : Google Scholar : PubMed/NCBI

29 

Park KC, Song KH, Chung HK, Kim H, Kim DW, Song JH, Hwang ES, Jung HS, Park SH, Bae I, et al: CR6-interacting factor 1 interacts with orphan nuclear receptor Nur77 and inhibits its transactivation. Mol Endocrinol. 19:12–24. 2005. View Article : Google Scholar

30 

Hanada N, Lo HW, Day CP, Pan Y, Nakajima Y and Hung MC: Co-regulation of B-Myb expression by E2F1 and EGF receptor. Mol Carcinog. 45:10–17. 2006. View Article : Google Scholar

31 

Liccardi G, Hartley JA and Hochhauser D: EGFR nuclear trans-location modulates DNA repair following cisplatin and ionizing radiation treatment. Cancer Res. 71:1103–1114. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Yu YL, Chou RH, Liang JH, Chang WJ, Su KJ, Tseng YJ, Huang WC, Wang SC and Hung MC: Targeting the EGFR/PCNA signaling suppresses tumor growth of triple-negative breast cancer cells with cell-penetrating PCNA peptides. PLoS One. 8:e613622013. View Article : Google Scholar : PubMed/NCBI

33 

Brand TM, Iida M, Li C and Wheeler DL: The nuclear epidermal growth factor receptor signaling network and its role in cancer. Discov Med. 12:419–432. 2011.PubMed/NCBI

34 

Hsu SC and Hung MC: Characterization of a novel tripartite nuclear localization sequence in the EGFR family. J Biol Chem. 282:10432–10440. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Chung HK, Yi YW, Jung NC, Kim D, Suh JM, Kim H, Park KC, Song JH, Kim DW, Hwang ES, et al: CR6-interacting factor 1 interacts with Gadd45 family proteins and modulates the cell cycle. J Biol Chem. 278:28079–28088. 2003. View Article : Google Scholar : PubMed/NCBI

36 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Kim SJ, Kwon MC, Ryu MJ, Chung HK, Tadi S, Kim YK, Kim JM, Lee SH, Park JH, Kweon GR, et al: CRIF1 is essential for the synthesis and insertion of oxidative phosphorylation polypeptides in the mammalian mitochondrial membrane. Cell Metab. 16:274–283. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Byun J, Son SM, Cha MY, Shong M, Hwang YJ, Kim Y, Ryu H, Moon M, Kim KS and Mook-Jung I: CR6-interacting factor 1 is a key regulator in Aβ-induced mitochondrial disruption and pathogenesis of Alzheimer’s disease. Cell Death Differ. Nov 7–2014.Epub ahead of print. View Article : Google Scholar

39 

Demory ML, Boerner JL, Davidson R, Faust W, Miyake T, Lee I, Hüttemann M, Douglas R, Haddad G and Parsons SJ: Epidermal growth factor receptor translocation to the mitochondria: Regulation and effect. J Biol Chem. 284:36592–36604. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Tibaldi E, Brunati AM, Massimino ML, Stringaro A, Colone M, Agostinelli E, Arancia G and Toninello A: Src-tyrosine kinases are major agents in mitochondrial tyrosine phosphorylation. J Cell Biochem. 104:840–849. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Ding Y, Liu Z, Desai S, Zhao Y, Liu H, Pannell LK, Yi H, Wright ER, Owen LB, Dean-Colomb W, et al: Receptor tyrosine kinase ErbB2 translocates into mitochondria and regulates cellular metabolism. Nat Commun. 3:12712012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2015
Volume 34 Issue 1

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Vahedi S, Chueh F, Dutta S, Chandran B and Yu C: Nuclear lymphocyte-specific protein tyrosine kinase and its interaction with CR6-interacting factor 1 promote the survival of human leukemic T cells. Oncol Rep 34: 43-50, 2015
APA
Vahedi, S., Chueh, F., Dutta, S., Chandran, B., & Yu, C. (2015). Nuclear lymphocyte-specific protein tyrosine kinase and its interaction with CR6-interacting factor 1 promote the survival of human leukemic T cells. Oncology Reports, 34, 43-50. https://doi.org/10.3892/or.2015.3990
MLA
Vahedi, S., Chueh, F., Dutta, S., Chandran, B., Yu, C."Nuclear lymphocyte-specific protein tyrosine kinase and its interaction with CR6-interacting factor 1 promote the survival of human leukemic T cells". Oncology Reports 34.1 (2015): 43-50.
Chicago
Vahedi, S., Chueh, F., Dutta, S., Chandran, B., Yu, C."Nuclear lymphocyte-specific protein tyrosine kinase and its interaction with CR6-interacting factor 1 promote the survival of human leukemic T cells". Oncology Reports 34, no. 1 (2015): 43-50. https://doi.org/10.3892/or.2015.3990