Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
MicroPubl Biol ; 20242024.
Article in English | MEDLINE | ID: mdl-38660565

ABSTRACT

PAK1 and prolactin (PRL) regulate breast cancer. Prolactin-activated JAK2 tyrosyl phosphorylates PAK1 (pTyr-PAK1). We demonstrate here that pTyr-PAK1 regulates epithelial-mesenchymal transition (EMT) in breast cancer cells. PRL treatment of T47D PAK1 WT cells leads to downregulation of E-cadherin surface expression and "ectodomain shedding" (extracellular cleavage of E-cadherin). pTyr-PAK1 increases mRNA levels of Snail, Slug, and Twist2, transcriptional factors implicated in E-cadherin repression. pTyr-PAK1 also significantly increases PRL-dependent Slug activity leading to expression of vimentin, a hallmark of EMT. Thus, our current data on pTyr-PAK1 regulation of EMT bring insight into the role of PAK1 and PRL in human breast cancer.

2.
PLoS One ; 17(1): e0261098, 2022.
Article in English | MEDLINE | ID: mdl-35089929

ABSTRACT

JAK2 is cytokine-activated non-receptor tyrosine kinase. Although JAK2 is mainly localized at the plasma membrane, it is also present on the centrosome. In this study, we demonstrated that JAK2 localization to the centrosome depends on the SH2 domain and intact kinase activity. We created JAK2 mutants deficient in centrosomal localization ΔSH2, K882E and (ΔSH2, K882E). We showed that JAK2 WT clone strongly enhances cell proliferation as compared to control cells while JAK2 clones ΔSH2, K882E and (ΔSH2, K882E) proliferate slower than JAK2 WT cells. These mutant clones also progress much slower through the cell cycle as compared to JAK2 WT clone and the enhanced proliferation of JAK2 WT cells is accompanied by increased S -> G2 progression. Both the SH2 domain and the kinase activity of JAK2 play a role in prolactin-dependent activation of JAK2 substrate STAT5. We showed that JAK2 is an important regulator of centrosome function as the SH2 domain of JAK2 regulates centrosome amplification. The cells overexpressing ΔSH2 and (ΔSH2, K-E) JAK2 have almost three-fold the amplified centrosomes of WT cells. In contrast, the kinase activity of JAK2 is dispensable for centrosome amplification. Our observations provide novel insight into the role of SH2 domain and kinase activity of JAK2 in centrosome localization of JAK2 and in the regulation of cell growth and centrosome biogenesis.


Subject(s)
Cell Proliferation , Centrosome/metabolism , Janus Kinase 2/metabolism , src Homology Domains/genetics , Animals , COS Cells , Cell Cycle Checkpoints , Cell Line , Chlorocebus aethiops , Humans , Interferon-gamma/pharmacology , Janus Kinase 2/chemistry , Janus Kinase 2/genetics , Mutagenesis, Site-Directed , Protein Transport/drug effects , STAT5 Transcription Factor/metabolism
3.
BMC Cell Biol ; 17(1): 31, 2016 08 20.
Article in English | MEDLINE | ID: mdl-27542844

ABSTRACT

BACKGROUND: The serine/threonine kinase PAK1 is an important regulator of cell motility. Both PAK1 and the hormone/cytokine prolactin (PRL) have been implicated in breast cancer cell motility, however, the exact mechanisms guiding PRL/PAK1 signaling in breast cancer cells have not been fully elucidated. Our lab has previously demonstrated that PRL-activated tyrosine kinase JAK2 phosphorylates PAK1 on tyrosines 153, 201, and 285, and that tyrosyl phosphorylated PAK1 (pTyr-PAK1) augments migration and invasion of breast cancer cells. RESULTS: Here we further investigate the mechanisms by which pTyr-PAK1 enhances breast cancer cell motility in response to PRL. We demonstrate a distinct reduction in PRL-induced FAK auto-phosphorylation in T47D and TMX2-28 breast cancer cells overexpressing wild-type PAK1 (PAK1 WT) when compared to cells overexpressing either GFP or phospho-tyrosine-deficient mutant PAK1 (PAK1 Y3F). Furthermore, pTyr-PAK1 phosphorylates MEK1 on Ser298 resulting in subsequent ERK1/2 activation. PRL-induced FAK auto-phosphorylation is rescued in PAK1 WT cells by inhibiting tyrosine phosphatases and tyrosine phosphatase inhibition abrogates cell motility and invasion in response to PRL. siRNA-mediated knockdown of the tyrosine phosphatase PTP-PEST rescues FAK auto-phosphorylation in PAK1 WT cells and reduces both cell motility and invasion. Finally, we provide evidence that PRL-induced pTyr-PAK1 stimulates tumor cell metastasis in vivo. CONCLUSION: These data provide insight into the mechanisms guiding PRL-mediated breast cancer cell motility and invasion and highlight a significant role for pTyr-PAK1 in breast cancer metastasis.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Movement/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Phosphotyrosine/metabolism , Prolactin/pharmacology , p21-Activated Kinases/metabolism , Animals , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Silencing/drug effects , Humans , Mice , Models, Biological , Neoplasm Invasiveness , Neoplasm Metastasis , Phosphorylation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 12/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 12/metabolism
4.
Biochem Biophys Res Commun ; 473(1): 206-211, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-27003261

ABSTRACT

Tyrosyl phosphorylation of the p21-activated serine-threonine kinase 1 (PAK1) has an essential role in regulating PAK1 functions in breast cancer cells. We previously demonstrated that PAK1 serves as a common node for estrogen (E2)- and prolactin (PRL)-dependent pathways. We hypothesize herein that intracellular localization of PAK1 is affected by PRL and E2 treatments differently. We demonstrate by immunocytochemical analysis that PAK1 nuclear translocation is ligand-dependent: only PRL but not E2 stimulated PAK1 nuclear translocation. Tyrosyl phosphorylation of PAK1 is essential for this nuclear translocation because phospho-tyrosyl-deficient PAK1 Y3F mutant is retained in the cytoplasm in response to PRL. We confirmed these data by Western blot analysis of subcellular fractions. In 30 min of PRL treatment, only 48% of pTyr-PAK1 is retained in the cytoplasm of PAK1 WT clone while 52% re-distributes into the nucleus and pTyr-PAK1 shuttles back to the cytoplasm by 60 min of PRL treatment. In contrast, PAK1 Y3F is retained in the cytoplasm. E2 treatment causes nuclear translocation of neither PAK1 WT nor PAK1 Y3F. Finally, we show by an in vitro kinase assay that PRL but not E2 stimulates PAK1 kinase activity in the nuclear fraction. Thus, PAK1 nuclear translocation is ligand-dependent: PRL activates PAK1 and induces translocation of activated pTyr-PAK1 into nucleus while E2 activates pTyr-PAK1 only in the cytoplasm.


Subject(s)
Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Estradiol/pharmacology , Prolactin/pharmacology , p21-Activated Kinases/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Cytoplasm/metabolism , Female , Humans , Immunohistochemistry , MCF-7 Cells , Microscopy, Fluorescence , Phosphorylation
5.
Cancer Res ; 76(9): 2600-11, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26944939

ABSTRACT

Serine/threonine kinase PAK1 is activated by estrogen and plays an important role in breast cancer. However, the integration of PAK1 into the estrogen response is not fully understood. In this study, we investigated the mechanisms underlying the hormone-induced activation of estrogen receptor (ERα, ESR1). We show that estrogen activated PAK1 through both the ERα and GPER1 membrane receptors. Estrogen-dependent activation of PAK1 required the phosphorylation of tyrosine residues by Etk/Bmx and protein kinase A (PKA) within an assembled signaling complex comprising pTyr-PAK1, Etk/Bmx, the heterotrimer G-protein subunits Gß1, Gγ2, and/or Gγ5, PAK-associated guanine nucleotide exchange factor (ßPIX, ARHGEF7), and PKA. Moreover, the PKA RIIß subunit is a direct target of PAK1, and thus in response to estrogen, the activated pTyr-PAK1 complex reciprocally potentiated PKA activity, suggesting a positive feedback mechanism. We also demonstrate that PKA phosphorylated Ser305-ERα in response to estrogen, but pTyr-PAK1 phosphorylated Ser305-ERα in response to prolactin (PRL), implying that maximal ERα phosphorylation is achieved when cells are exposed to both PRL and estrogen. Furthermore, S305-ERα activation led to enhanced phosphorylation of Ser118-ERα and promoted cell proliferation and tumor growth. Together, these data strongly support a critical interplay between PRL and estrogen via PAK1 and suggest that ligand-independent activation of ERα through PRL/PAK1 may impart resistance to anti-estrogen therapies. Cancer Res; 76(9); 2600-11. ©2016 AACR.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Prolactin/metabolism , p21-Activated Kinases/metabolism , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Heterografts , Humans , Immunoprecipitation , Mice , Mice, Inbred NOD , Mice, SCID , Phosphorylation
6.
Int J Cancer Clin Res ; 2(3): 1-6, 2015.
Article in English | MEDLINE | ID: mdl-26688830

ABSTRACT

Differentiation-inducing factors 1-3 (DIFs 1-3), chlorinated alkylphenones identified in the cellular slime mold Dictyostelium discoideum, are considered anti-tumor agents because they inhibit proliferation of a variety of mammalian tumor cells in vitro. Although the anti-proliferative effects of DIF-1 and DIF-3 are well-documented, the precise molecular mechanisms underlying the actions of DIFs have not been fully elucidated. In this study, we examined the effects of DIFs and their derivatives on PAK1, a key serine-threonine kinase, which is activated by multiple ligands and regulates cell proliferation. We examined the effect of DIF derivatives on PAK1 kinase activity in cells. We also examined the effect of DIF-3(+1) derivative on PAK1 kinase activity in vitro, cyclin D1 promoter activity and breast cancer cell proliferation. It was found that some derivatives strongly inhibited PAK1 kinase activity in human breast cancer MCF-7 cells stably over expressing PAK1. Among the derivatives, DIF-3(+1) was most potent, which directly inhibited kinase activity of recombinant purified PAK1 in an in vitro kinase assay. Furthermore, DIF-3(+1) strongly inhibited both cyclin D1 promoter activity and proliferation of MCF-7 and T47D breast cancer cells stably over expressing PAK1 in response to prolactin, estrogen, epidermal growth factor and heregulin. In the present study we propose PAK1 as DIF-3(+1) target mediating its anti-proliferative effect.

7.
Biochem Biophys Res Commun ; 463(4): 644-9, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26043691

ABSTRACT

The hormone/cytokine prolactin (PRL) is implicated in breast cancer cell invasion and metastasis. PRL-induced pathways are mediated by two non-receptor tyrosine kinases, JAK2 and Src. We previously demonstrated that prolactin stimulates invasion of breast cancer cells TMX2-28 through JAK2 and its target serine/threonine kinase PAK1. We hypothesize herein that the actin-binding protein cortactin, a protein involved in invadopodia formation and cell invasion, is activated by PRL. We demonstrate that TMX2-28 cells are more invasive than T47D breast cancer cells in response to PRL. We determine that cortactin is tyrosyl phosphorylated in response to PRL in a time and dose-dependent manner in TMX2-28 cells, but not in T47D cells. Furthermore, we show that PRL mediates cortactin tyrosyl phosphorylation via Src, but not JAK2. Finally, we demonstrate that maximal PRL-mediated TMX2-28 cell invasion requires both Src and JAK2 kinase activity, while T47D cell invasion is JAK2- but not Src-dependent. Thus PRL may induce cell invasion via two pathways: through a JAK2/PAK1 mediated pathway that we have previously demonstrated, and Src-dependent activation and tyrosyl phosphorylation of cortactin.


Subject(s)
Cortactin/metabolism , Prolactin/physiology , Tyrosine/metabolism , src-Family Kinases/metabolism , Humans , MCF-7 Cells , Phosphorylation
8.
Adv Exp Med Biol ; 846: 97-137, 2015.
Article in English | MEDLINE | ID: mdl-25472536

ABSTRACT

Despite efforts to discover the cellular pathways regulating breast cancer metastasis, little is known as to how prolactin (PRL) cooperates with extracellular environment and cytoskeletal proteins to regulate breast cancer cell motility and invasion. We implicated serine-threonine kinase p21-activated kinase 1 (PAK1) as a novel target for PRL-activated Janus-kinase 2 (JAK2). JAK2-dependent PAK1 tyrosyl phosphorylation plays a critical role in regulation of both PAK1 kinase activity and scaffolding properties of PAK1. Tyrosyl phosphorylated PAK1 facilitates PRL-dependent motility via at least two mechanisms: formation of paxillin/GIT1/ßPIX/pTyr-PAK1 complexes resulting in increased adhesion turnover and phosphorylation of actin-binding protein filamin A. Increased adhesion turnover is the basis for cell migration and phosphorylated filamin A stimulates the kinase activity of PAK1 and increases actin-regulating activity to facilitate cell motility. Tyrosyl phosphorylated PAK1 also stimulates invasion of breast cancer cells in response to PRL and three-dimensional (3D) collagen IV via transcription and secretion of MMP-1 and MMP-3 in a MAPK-dependent manner. These data illustrate the complex interaction between PRL and the cell microenvironment in breast cancer cells and suggest a pivotal role for PRL/PAK1 signaling in breast cancer metastasis.


Subject(s)
Breast Neoplasms/pathology , Cell Movement , Prolactin/pharmacology , p21-Activated Kinases/physiology , Animals , Breast Neoplasms/genetics , Cell Movement/drug effects , Cell Movement/genetics , Female , Humans , Neoplasm Invasiveness , Neoplasm Metastasis
9.
Mol Cell Biol ; 35(1): 111-31, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25332239

ABSTRACT

JAK2 is a cytoplasmic tyrosine kinase critical for cytokine signaling. In this study, we have identified a novel centrosome-associated complex containing ninein and JAK2. We have found that active JAK2 localizes around the mother centrioles, where it partly colocalizes with ninein, a protein involved in microtubule (MT) nucleation and anchoring. We demonstrated that JAK2 is an important regulator of centrosome function. Depletion of JAK2 or use of JAK2-null cells causes defects in MT anchoring and increased numbers of cells with mitotic defects; however, MT nucleation is unaffected. We showed that JAK2 directly phosphorylates the N terminus of ninein while the C terminus of ninein inhibits JAK2 kinase activity in vitro. Overexpressed wild-type (WT) or C-terminal (amino acids 1179 to 1931) ninein inhibits JAK2. This ninein-dependent inhibition of JAK2 significantly decreases prolactin- and interferon gamma (IFN-γ)-induced tyrosyl phosphorylation of STAT1 and STAT5. Downregulation of ninein enhances JAK2 activation. These results indicate that JAK2 is a novel member of centrosome-associated complex and that this localization regulates both centrosomal function and JAK2 kinase activity, thus controlling cytokine-activated molecular pathways.


Subject(s)
Centrosome/ultrastructure , Cytoskeletal Proteins/metabolism , Gene Expression Regulation, Enzymologic , Janus Kinase 2/metabolism , Nuclear Proteins/metabolism , Animals , COS Cells , Centrioles/ultrastructure , Centrosome/metabolism , Chlorocebus aethiops , Gene Silencing , HEK293 Cells , HeLa Cells , Humans , Interferon-gamma/metabolism , Microtubules/ultrastructure , Mitosis , Phosphorylation , Prolactin/metabolism , Protein Structure, Tertiary
10.
FASEB J ; 29(3): 943-59, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25466889

ABSTRACT

The p21-activated serine-threonine kinase (PAK1) regulates cell motility and adhesion. We have previously shown that the prolactin (PRL)-activated tyrosine kinase JAK2 phosphorylates PAK1 in vivo and in vitro and identified tyrosines 153, 201, and 285 in PAK1 as sites of JAK2 tyrosyl phosphorylation. Here, we further investigate the role of the tyrosyl phosphorylated PAK1 (pTyr-PAK1) in regulation of cell adhesion. We use human breast cancer T47D cell lines that stably overexpress PAK1 wild type or PAK1 Y3F mutant in which these 3 JAK2 phosphorylation sites were mutated to phenylalanine. We demonstrate that PRL/JAK2-dependent phosphorylation of these tyrosines promotes a motile phenotype in the cells upon adhesion, participates in regulation of cell adhesion on collagen IV, and is required for maximal PAK1 kinase activity. Down-regulation of PAK1 abolishes the effect of PAK1 on cell adhesion. We show that the tyrosyl phosphorylation of PAK1 promotes PAK1 binding to ß-PAK1-interacting guanine-nucleotide exchange factor (ßPIX) and G protein-coupled receptor kinase-interacting target 1 (GIT1), phosphorylation of paxillin on Ser273, and formation and distribution of adhesion complexes. Using phosphospecific antibodies (Abs) directed to single phosphorylated tyrosines on PAK1, we identified Tyr285 as a site of PRL-dependent phosphorylation of PAK1 by JAK2. Furthermore, using PAK1 Y285F mutant, we provide evidence for a role of pTyr285 in cell adhesion, enhanced ßPIX/GIT1 binding, and adhesion turnover. Our immunohistochemistry analysis demonstrates that pTyr285- PAK1 may modulate PAK1 signaling during tumor progression.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/metabolism , Cell Adhesion/physiology , Cell Cycle Proteins/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Tyrosine/metabolism , p21-Activated Kinases/metabolism , Blotting, Western , Breast Neoplasms/pathology , Cell Movement , Cell Proliferation , Female , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Immunoprecipitation , Paxillin/metabolism , Phosphorylation , Protein Binding , Signal Transduction , Tumor Cells, Cultured
11.
Mol Endocrinol ; 27(7): 1048-64, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23744893

ABSTRACT

p21-Activated serine-threonine kinase (PAK1) is implicated in breast cancer. We have shown previously that PAK1 is tyrosyl phosphorylated by prolactin (PRL)-activated Janus tyrosine kinase (JAK2). Although a role for both PRL and PAK1 in breast cancer is widely acknowledged, the mechanism remains poorly understood. In the present study, PRL-activated PAK1 stimulates the invasion of TMX2-28 human breast cancer cells through Matrigel. Three-dimensional (3D) collagen IV stimulates the secretion of the matrix proteases, metalloproteinase (MMP)-1 and -3 that is further enhanced by the PRL-dependent tyrosyl phosphorylation of PAK1. 3D collagen IV also stimulates the expression and secretion of MMP-2, but in contrast to MMP-1 and -3, PRL/PAK1 signaling down-regulates MMP-2 expression and secretion. In contrast, MMP-9 expression and secretion are stimulated by 3D collagen I, not collagen IV, and are not affected by PRL but are down-regulated by PAK1. MMP-1 and -3 are required and MMP-2 contributes to PRL-dependent invasion. ERK1/2 signaling appears to be required for the enhanced expression and secretion of MMP-1 and -3 and enhanced PRL-dependent invasion. p38 MAPK and c-Jun N-terminal kinase 1/2 pathways participate in production of MMP-1 and -3 as well as in PRL/PAK1-dependent cell invasion. Together, these data illustrate the complex interaction between the substratum and PRL/PAK1 signaling in human breast cancer cells and suggest a pivotal role for PRL-dependent PAK1 tyrosyl phosphorylation in MMP secretion.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Collagen Type IV/pharmacology , Matrix Metalloproteinases/metabolism , Prolactin/pharmacology , p21-Activated Kinases/metabolism , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Enzyme Activation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Matrix Metalloproteinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Mutant Proteins/metabolism , NF-kappa B/metabolism , Neoplasm Invasiveness , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Rats , Transcription, Genetic/drug effects
12.
Mol Endocrinol ; 27(3): 455-65, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23340249

ABSTRACT

The p21-activated serine-threonine kinase (PAK1) is activated by small GTPase-dependent and -independent mechanisms and regulates cell motility. Both PAK1 and the hormone prolactin (PRL) have been implicated in breast cancer by numerous studies. We have previously shown that the PRL-activated tyrosine kinase JAK2 (Janus tyrosine kinase 2) phosphorylates PAK1 in vivo and identified tyrosines (Tyr) 153, 201, and 285 in the PAK1 molecule as sites of JAK2 tyrosyl phosphorylation. Here, we have used human breast cancer T47D cells stably overexpressing PAK1 wild type or PAK1 Y3F mutant in which Tyr(s) 153, 201, and 285 were mutated to phenylalanines to demonstrate that phosphorylation of these three tyrosines are required for maximal PRL-dependent ruffling. In addition, phosphorylation of these three tyrosines is required for increased migration of T47D cells in response to PRL as assessed by two independent motility assays. Finally, we show that PAK1 phosphorylates serine (Ser) 2152 of the actin-binding protein filamin A to a greater extent when PAK1 is tyrosyl phosphorylated by JAK2. Down-regulation of PAK1 or filamin A abolishes the effect of PRL on cell migration. Thus, our data presented here bring some insight into the mechanism of PRL-stimulated motility of breast cancer cells.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Movement/drug effects , Contractile Proteins/metabolism , Microfilament Proteins/metabolism , Phosphotyrosine/metabolism , Prolactin/pharmacology , p21-Activated Kinases/metabolism , Cell Line, Tumor , Clone Cells , Female , Filamins , Green Fluorescent Proteins/metabolism , Humans , Janus Kinase 2/metabolism , Models, Biological , Mutant Proteins/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism
13.
Mol Endocrinol ; 25(9): 1565-78, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21719533

ABSTRACT

Prolactin (PRL) is critical for alveolar proliferation and differentiation in normal mammary development and is also implicated in breast cancer. PRL influences cell proliferation and growth by altering the expression of cyclin D1. Cyclin D1 expression is directly regulated by PRL through the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 5-mediated transcriptional activation of the cyclin D1 promoter. A p21-activated serine-threonine kinase (PAK)1 has also been implicated in the regulation of cyclin D1 gene expression. We have previously demonstrated that JAK2 directly phosphorylates PAK1 and extend these data here to demonstrate that PAK1 activates the cyclin D1 promoter in response to PRL. We show that mutation of PAK1 Tyr 153, 201, and 285 (sites of JAK2 phosphorylation; PAK1 Y3F) decreases both PAK1 nuclear translocation in response to PRL and PRL-induced cyclin D1 promoter activity by 55%. Mutation of the PAK1 nuclear localization signals decreases PRL-induced cyclin D1 promoter activity by 46%. A PAK1 Y3F mutant lacking functional nuclear localization signals decreases PRL-induced cyclin D1 activity by 68%, suggesting that there is another PAK1-dependent mechanism to activate the cyclin D1 promoter. We have found that adapter protein Nck sequesters PAK1 in the cytoplasm and that coexpression of both PAK1 and Nck inhibits the amplifying effect of PRL-induced PAK1 on cyclin D1 promoter activity (95% inhibition). This inhibition is partially abolished by disruption of PAK1-Nck binding. We propose two PAK1-dependent mechanisms to activate cyclin D1 promoter activity in response to PRL: via nuclear translocation of tyrosyl-phosphorylated PAK1 and via formation of a Nck-PAK1 complex that sequesters PAK1 in the cytoplasm.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cyclin D1/genetics , Oncogene Proteins/metabolism , Prolactin/pharmacology , Promoter Regions, Genetic , p21-Activated Kinases/metabolism , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Humans , Models, Biological , Nuclear Localization Signals/metabolism , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Binding/drug effects , Protein Transport/drug effects
14.
Mol Endocrinol ; 25(7): 1231-43, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21566085

ABSTRACT

Prolactin (PRL) regulates cytoskeletal rearrangement and cell motility. PRL-activated Janus tyrosine kinase 2 (JAK2) phosphorylates the p21-activated serine-threonine kinase (PAK)1 and the Src homology 2 (SH2) domain-containing adapter protein SH2B1ß. SH2B1ß is an actin-binding protein that cross-links actin filaments, whereas PAK1 regulates the actin cytoskeleton by different mechanisms, including direct phosphorylation of the actin-binding protein filamin A (FLNa). Here, we have used a FLNa-deficient human melanoma cell line (M2) and its derivative line (A7) that stably expresses FLNa to demonstrate that SH2B1ß and FLNa are required for maximal PRL-dependent cell ruffling. We have found that in addition to two actin-binding domains, SH2B1ß has a FLNa-binding domain (amino acids 200-260) that binds directly to repeats 17-23 of FLNa. The SH2B1ß-FLNa interaction participates in PRL-dependent actin rearrangement. We also show that phosphorylation of the three tyrosines of PAK1 by JAK2, as well as the presence of FLNa, play a role in PRL-dependent cell ruffling. Finally, we show that the actin- and FLNa-binding-deficient mutant of SH2B1ß (SH2B1ß 3Δ) abolished PRL-dependent ruffling and PRL-dependent cell migration when expressed along with PAK1 Y3F (JAK2 tyrosyl-phosphorylation-deficient mutant). Together, these data provide insight into a novel mechanism of PRL-stimulated regulation of the actin cytoskeleton and cell motility via JAK2 signaling through FLNa, PAK1, and SH2B1ß. We propose a model for PRL-dependent regulation of the actin cytoskeleton that integrates our findings with previous studies.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Movement , Contractile Proteins/metabolism , Cytoskeleton/metabolism , Microfilament Proteins/metabolism , Prolactin/pharmacology , Actins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Amino Acid Motifs , Binding Sites , Cell Line, Tumor , Cell Surface Extensions/metabolism , Filamins , Humans , Janus Kinase 2/metabolism , Phosphorylation , Prolactin/physiology , Protein Binding , Protein Isoforms/metabolism , Protein Transport , p21-Activated Kinases/metabolism
15.
Mol Endocrinol ; 23(7): 1065-76, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19342444

ABSTRACT

The Src homology 2 (SH2) domain-containing adapter protein SH2B1beta plays a role in severe obesity, leptin and insulin resistance, and infertility. SH2B1beta was initially identified as a Janus tyrosine kinase 2 (JAK2) substrate, and it has been implicated in cell motility and regulation of the actin rearrangement in response to GH and platelet-derived growth factor. SH2B1beta is also required for maximal actin-based motility of Listeria. Here we have used a low-speed pelleting assay and electron microscopy to demonstrate that SH2B1beta has two actin-binding sites and that it cross-links actin filaments in vitro. Wild-type SH2B1beta localized to cell ruffles and along filopodia, but deletion of amino acids 150-200 (the first actin-binding site) led to mislocalization of the protein to filopodia tip complexes where it colocalized with vasodilator-stimulated phosphoprotein (VASP). Based on studies performed in VASP-deficient MVD7(-/-) cells, with or without green fluorescent protein-VASP reconstitution, we concluded that the proper intracellular localization of native SH2B1beta required the presence of the first SH2B1beta actin-binding site and VASP. Finally, we found that both SH2B1beta actin-binding domains were required for maximal GH- and prolactin-induced cell ruffling. Together, these results suggest that SH2B1beta functions as an adapter protein that cross-links actin filaments, leading to modulation of cellular responses in response to JAK2 activation.


Subject(s)
Actins/metabolism , Adaptor Proteins, Signal Transducing/physiology , Cytoskeleton/metabolism , Protein Multimerization/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/physiology , Cell Movement/physiology , Cells, Cultured , Humans , Janus Kinase 2/metabolism , Mice , Mice, Knockout , Microfilament Proteins/metabolism , Microfilament Proteins/physiology , Microvilli/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutant Proteins/physiology , Phosphoproteins/metabolism , Phosphoproteins/physiology , Protein Binding/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology , Pseudopodia/metabolism
16.
J Cell Biochem ; 106(1): 33-41, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19009561

ABSTRACT

Activating ras mutations are frequently found in malignant tumors of the pancreas, colon, lung and other tissues. RAS activates a number of downstream pathways that ultimately cause cellular transformation. Several recent studies suggested that one of those pathways involves Aurora kinases. Overexpression of Aurora-B kinase can augment transformation by oncogenic RAS, however the mechanism was not determined. The cooperative effect of high levels of Aurora kinase is important since this kinase is frequently overexpressed in human tumors. We have used two Aurora kinase inhibitors to test their effect on RAS signaling. We find that these inhibitors have no effect on the phosphorylation of MEK1/2 or MAPK in response to RAS. Furthermore, inhibiting Aurora kinases in human cancer cells with or without activated RAS did not change the length of the cell cycle nor induce apoptosis suggesting that these kinases do not play a direct role in these key cellular responses to activated RAS. Overexpression of Aurora B can cause cells to become polyploid. Also, inducing polyploidy with cytochalasin D was reported to induce neoplastic transformation, suggesting that Aurora overexpression may cooperate with RAS indirectly by inducing polyploidy. We find that inducing polyploidy with cytochalasin D or blebbistatin does not enhance transformation by oncogenic RAS. Our observations argue against a direct role for Aurora kinases in the RAS-MAPK pathway, and suggest that the polyploid state does not enhance transformation by RAS.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Signal Transduction , ras Proteins/metabolism , Animals , Aurora Kinase B , Aurora Kinases , Benzamides/pharmacology , Cell Cycle , Cell Line, Tumor , Fibroblasts/metabolism , Histones/metabolism , Humans , MAP Kinase Kinase Kinases/metabolism , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , NIH 3T3 Cells , Phosphorylation , Polyploidy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Quinazolines/pharmacology , Rats , raf Kinases/metabolism
17.
J Biol Chem ; 282(42): 30985-96, 2007 Oct 19.
Article in English | MEDLINE | ID: mdl-17726028

ABSTRACT

The serine-threonine kinase PAK1 is activated by small GTPase-dependent and -independent mechanisms and promotes cell survival. However, the role of tyrosyl phosphorylation in the regulation of PAK1 function is poorly understood. In this study, we have shown that the prolactin-activated tyrosine kinase JAK2 phosphorylates PAK1 in vivo. Wild type, but not kinase-dead, JAK2 directly phosphorylates PAK1 in cells and in an in vitro kinase assay. PAK1 tyrosines 153, 201, and 285 were identified as sites of JAK2 tyrosyl phosphorylation by mass spectrometry and two-dimensional peptide mapping. Mutation of PAK1 tyrosines 153, 201, and 285 to phenylalanines individually or in combination implicated these PAK1 tyrosines in the regulation of PAK1 kinase activity. Tyrosyl phosphorylation by JAK2 significantly increases PAK1 kinase activity, whereas similar phosphorylation of the PAK1 Y153F,Y201F,Y285F mutant has no effect on PAK1 activity. Tyrosyl phosphorylation of wild type PAK1 decreases apoptosis induced by serum deprivation and staurosporine treatment and increases cell motility. In contrast, these parameters are unaltered in the PAK1 Y153F,Y201F,Y285F mutant. Our findings indicate that JAK2 phosphorylates PAK1 at these specific tyrosines and that this phosphorylation plays an important role in cell survival and motility.


Subject(s)
Apoptosis/physiology , Cell Movement/physiology , Janus Kinase 2/metabolism , Protein Processing, Post-Translational/physiology , p21-Activated Kinases/metabolism , Amino Acid Substitution , Animals , Apoptosis/drug effects , COS Cells , Cell Movement/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Chlorocebus aethiops , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Humans , Janus Kinase 2/genetics , Mutation, Missense , Peptide Mapping , Phosphorylation/drug effects , Prolactin/pharmacology , Protein Processing, Post-Translational/drug effects , Rats , Staurosporine/pharmacology , p21-Activated Kinases/genetics
18.
Infect Immun ; 75(7): 3581-93, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17452473

ABSTRACT

SH2-Bbeta (Src homology 2 Bbeta) is an adapter protein that is required for maximal growth hormone-dependent actin reorganization in membrane ruffling and cell motility. Here we show that SH2-Bbeta is also required for maximal actin-based motility of Listeria monocytogenes. SH2-Bbeta localizes to Listeria-induced actin tails and increases the rate of bacterial propulsion in infected cells and in cell extracts. Furthermore, Listeria motility is decreased in mouse embryo fibroblasts from SH2-B(-/-) mice. Both recruitment of SH2-Bbeta to Listeria and SH2-Bbeta stimulation of actin-based propulsion require the vasodilator-stimulated phosphoprotein (VASP), which binds ActA at the surfaces of Listeria cells and enhances bacterial actin-based motility. SH2-Bbeta enhances actin-based movement of ActA-coated beads in a biomimetic actin-based motility assay, provided that VASP is present. In vitro binding assays show that SH2-Bbeta binds ActA but not VASP; however, binding to ActA is greater in the presence of VASP. Because VASP also plays an essential regulatory role in actin-based processes in eukaryotic cells, the present results provide mechanistic insight into the functions of both SH2-Bbeta and VASP in motility and also increase our understanding of the fundamental mechanism by which Listeria spreads.


Subject(s)
Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion Molecules/metabolism , Listeria monocytogenes/physiology , Microfilament Proteins/metabolism , Movement/physiology , Phosphoproteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , COS Cells , Cell Adhesion Molecules/genetics , Chlorocebus aethiops , Fibroblasts/microbiology , Gene Expression Regulation, Bacterial , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Macrophages/microbiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Microfilament Proteins/genetics , Oocytes/microbiology , Phosphoproteins/genetics , Xenopus laevis/growth & development , Xenopus laevis/microbiology
19.
Endocrinology ; 148(5): 2045-55, 2007 May.
Article in English | MEDLINE | ID: mdl-17272398

ABSTRACT

GH excess in both the human and transgenic animal models is characterized by significant changes in blood pressure and renal function. The GH/GH receptor (GHR) axis is also implicated in the development of diabetic nephropathy. However, it is not clear whether GH's actions on renal function are due to indirect actions mediated via changes in blood pressure and vascular tone or due to direct action of GH on the kidney. We hypothesized that functional GHRs are expressed on the glomerular podocyte enabling direct actions of GH on glomerular function. Real-time PCR, immunohistochemistry, and Western blot analysis of murine podocyte cells (MPC-5) and kidney glomeruli demonstrated expression of GHR mRNA and protein. Exposure of both murine and human podocytes to GH (50-500 ng/ml) resulted in an increase in abundance of phosphorylated signal transducer and activator of transcription-5, Janus kinase-2, and ERK1/2 proteins. Exposure of podocytes to GH also caused changes in the intracellular distribution of the Janus kinase-2 adapter protein Src homology 2-Bbeta, stimulation of focal adhesion kinase, increase in reactive oxygen species, and GH-dependent changes in the actin cytoskeleton. We conclude that glomerular podocytes express functional GHRs and that GH increases levels of reactive oxygen species and induces reorganization of the actin cytoskeleton in these cells. These results provide a novel mechanistic link between GH's actions and glomerular dysfunction in disorders such as acromegaly and diabetic glomerulosclerosis.


Subject(s)
Growth Hormone/pharmacology , Growth Hormone/physiology , Podocytes/drug effects , Podocytes/physiology , Acromegaly/pathology , Acromegaly/physiopathology , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Animals , Cell Line, Transformed , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Gene Expression , Humans , Male , Mice , Mice, Inbred C57BL , Podocytes/cytology , Polymers , Reactive Oxygen Species/metabolism , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism
20.
J Biol Chem ; 278(14): 11970-8, 2003 Apr 04.
Article in English | MEDLINE | ID: mdl-12551917

ABSTRACT

SH2-Bbeta binds to the activated form of JAK2 and various receptor tyrosine kinases. It is a potent stimulator of JAK2, is required for growth hormone (GH)-induced membrane ruffling, and increases mitogenesis stimulated by platelet-derived growth factor (PDGF) and insulin-like growth factor I. Its domain structure suggests that SH2-Bbeta may act as an adapter protein to recruit downstream signaling proteins to kinase.SH2-Bbeta complexes. SH2-Bbeta is tyrosyl-phosphorylated in response to GH and interferon-gamma, stimulators of JAK2, as well as in response to PDGF and nerve growth factor. To begin to elucidate the role of tyrosyl phosphorylation in the function of SH2-Bbeta, we used phosphopeptide mapping, mutagenesis, and a phosphotyrosine-specific antibody to identify Tyr-439 and Tyr-494 in SH2-Bbeta as targets of JAK2 both in vitro and in intact cells. SH2-Bbeta lacking Tyr-439 and Tyr-494 inhibits GH-induced membrane ruffling but still activates JAK2. We provide evidence that JAK1, like JAK2, phosphorylates Tyr-439 and Tyr-494 in SH2-Bbeta and that PDGF receptor phosphorylates SH2-Bbeta on Tyr-439. Therefore, phosphorylated Tyr-439 and/or Tyr-494 in SH2-Bbeta may provide a binding site for one or more proteins linking cytokine receptor.JAK2 complexes and/or receptor tyrosine kinases to the actin cytoskeleton.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cytoskeleton/physiology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins , Receptors, Platelet-Derived Growth Factor/metabolism , 3T3 Cells , Actins/metabolism , Animals , COS Cells , Cell Membrane/drug effects , Cell Membrane/physiology , Cricetinae , Intracellular Signaling Peptides and Proteins , Janus Kinase 1 , Janus Kinase 2 , Mice , Mutagenesis/physiology , Phosphorylation , Platelet-Derived Growth Factor/pharmacology , Rats , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...