Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Head Neck ; 45(5): 1255-1271, 2023 05.
Article in English | MEDLINE | ID: mdl-36939040

ABSTRACT

BACKGROUND: The tyrosine kinase receptors Axl and MerTK are highly overexpressed in head and neck cancer (HNC) cells, where they are critical drivers of survival, proliferation, metastasis, and therapeutic resistance. METHODS: We investigated the role of Axl and MerTK in creating an immunologically "cold" tumor immune microenvironment (TIME) by targeting both receptors simultaneously with a small molecule inhibitor of Axl and MerTK (INCB081776). Effects of INCB081776 and/or anti-PDL1 on mouse oral cancer (MOC) cell growth and on the TIME were evaluated. RESULTS: Targeting Axl and MerTK can reduce M2 and induce M1 macrophage polarization. In vivo, INCB081776 treatment alone or with anti-PDL1 appears to slow MOC tumor growth, increase proinflammatory immune infiltration, and decrease anti-inflammatory immune infiltration. CONCLUSIONS: This data indicates that simultaneous targeting of Axl and MerTK with INCB081776, either alone or in combination with anti-PDL1, slows tumor growth and creates a proinflammatory TIME in mouse models of HNC.


Subject(s)
Head and Neck Neoplasms , Proto-Oncogene Proteins , Animals , Mice , c-Mer Tyrosine Kinase , Cell Line, Tumor , Protein Kinase Inhibitors/pharmacology , Tumor Microenvironment
2.
Mol Cancer Res ; 18(8): 1189-1201, 2020 08.
Article in English | MEDLINE | ID: mdl-32321766

ABSTRACT

The Crk adaptor protein, a critical modifier of multiple signaling pathways, is overexpressed in many cancers where it contributes to tumor progression and metastasis. Recently, we have shown that Crk interacts with the peptidyl prolyl cis-trans isomerase, Cyclophilin A (CypA; PP1A) via a G219P220Y221 (GPY) motif in the carboxyl-terminal linker region of Crk, thereby delaying pY221 phosphorylation and preventing downregulation of Crk signaling. Here, we investigate the physiologic significance of the CypA/Crk interaction and query whether CypA inhibition affects Crk signaling in vitro and in vivo. We show that CypA, when induced under conditions of hypoxia, regulates Crk pY221 phosphorylation and signaling in cancer cell lines. Using nuclear magnetic resonance spectroscopy, we show that CypA binds to the Crk GPY motif via the catalytic PPII domain of CypA, and small-molecule nonimmunosuppressive inhibitors of CypA (Debio-025) disrupt the CypA-CrkII interaction and restores phosphorylation of Crk Y221. In cultured cell lines, Debio-025 suppresses cell migration, and when administered in vivo in an orthotopic model of triple-negative breast cancer, Debio-025 showed antitumor efficacy either alone or in combination with anti-PD-1 mAb, reducing both tumor volume and metastatic lung dispersion. Furthermore, when analyzed by NanoString immune profiling, treatment of Debio-025 with anti-PD-1 mAb increased both T-cell signaling and innate immune signaling in tumor microenvironment. IMPLICATIONS: These data suggest that pharmacologic inhibition of CypA may provide a promising and unanticipated consequence in cancer biology, in part by targeting the CypA/CrkII axis that regulates cell migration, tumor metastasis, and host antitumor immune evasion.


Subject(s)
Breast Neoplasms/drug therapy , Cyclosporine/administration & dosage , Immune Checkpoint Inhibitors/administration & dosage , Peptidylprolyl Isomerase/metabolism , Proto-Oncogene Proteins c-crk/metabolism , Animals , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/pharmacology , Breast Neoplasms/metabolism , Cell Hypoxia , Cell Line, Tumor , Cell Movement/drug effects , Cyclosporine/pharmacology , Drug Synergism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immune Checkpoint Inhibitors/pharmacology , Mice , Models, Molecular , Neoplasm Metastasis , Peptidylprolyl Isomerase/chemistry , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Domains , Proto-Oncogene Proteins c-crk/chemistry , Sequence Analysis, RNA , Tumor Microenvironment/drug effects
3.
Front Immunol ; 8: 1521, 2017.
Article in English | MEDLINE | ID: mdl-29176978

ABSTRACT

The Tyro3, Axl, and Mertk (TAM) receptors are homologous type I receptor tyrosine kinases that have critical functions in the clearance of apoptotic cells in multicellular organisms. TAMs are activated by their endogenous ligands, growth arrest-specific 6 (Gas6), and protein S (Pros1), that function as bridging molecules between externalized phosphatidylserine (PS) on apoptotic cells and the TAM ectodomains. However, the molecular mechanisms by which Gas6/Pros1 promote TAM activation remains elusive. Using TAM/IFNγR1 reporter cell lines to monitor functional TAM activity, we found that Gas6 activity was exquisitely dependent on vitamin K-mediated γ-carboxylation, whereby replacing vitamin K with anticoagulant warfarin, or by substituting glutamic acid residues involved in PS binding, completely abrogated Gas6 activity as a TAM ligand. Furthermore, using domain and point mutagenesis, Gas6 activity also required both an intact Gla domain and intact EGF-like domains, suggesting these domains function cooperatively in order to achieve TAM activation. Despite the requirement of γ-carboxylation and the functional Gla domain, non-γ-carboxylated Gas6 and Gla deletion/EGF-like domain deletion mutants still retained their ability to bind TAMs and acted as blocking decoy ligands. Finally, we found that distinct sources of PS-positive cells/vesicles (including apoptotic cells, calcium-induced stressed cells, and exosomes) bound Gas6 and acted as cell-derived or exosome-derived ligands to activate TAMs. Taken together, our findings indicate that PS is indispensable for TAM activation by Gas6, and by inference, provides new perspectives on how PS, regulates TAM receptors and efferocytosis.

4.
Sci Rep ; 7: 43908, 2017 03 08.
Article in English | MEDLINE | ID: mdl-28272423

ABSTRACT

TAM receptors (Tyro-3, Axl, and Mertk) are a family of three homologous type I receptor tyrosine kinases that are implicated in several human malignancies. Overexpression of TAMs and their major ligand Growth arrest-specific factor 6 (Gas6) is associated with more aggressive staging of cancers, poorer predicted patient survival, acquired drug resistance and metastasis. Here we describe small molecule inhibitors (RU-301 and RU-302) that target the extracellular domain of Axl at the interface of the Ig-1 ectodomain of Axl and the Lg-1 of Gas6. These inhibitors effectively block Gas6-inducible Axl receptor activation with low micromolar IC50s in cell-based reporter assays, inhibit Gas6-inducible motility in Axl-expressing cell lines, and suppress H1299 lung cancer tumor growth in a mouse xenograft NOD-SCIDγ model. Furthermore, using homology models and biochemical verifications, we show that RU301 and 302 also inhibit Gas6 inducible activation of Mertk and Tyro3 suggesting they can act as pan-TAM inhibitors that block the interface between the TAM Ig1 ectodomain and the Gas6 Lg domain. Together, these observations establish that small molecules that bind to the interface between TAM Ig1 domain and Gas6 Lg1 domain can inhibit TAM activation, and support the further development of small molecule Gas6-TAM interaction inhibitors as a novel class of cancer therapeutics.


Subject(s)
Carcinogenesis/drug effects , Intercellular Signaling Peptides and Proteins/metabolism , Isoxazoles/pharmacology , Piperazines/pharmacology , Protein Kinase Inhibitors/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , c-Mer Tyrosine Kinase/metabolism , Animals , Binding Sites , Body Weight/drug effects , Cell Line, Tumor , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins/chemistry , Receptor Protein-Tyrosine Kinases/chemistry , Transplantation, Heterologous , c-Mer Tyrosine Kinase/chemistry , Axl Receptor Tyrosine Kinase
5.
Mol Cancer Res ; 15(6): 753-764, 2017 06.
Article in English | MEDLINE | ID: mdl-28184013

ABSTRACT

Tyro3, Axl, and Mertk (collectively TAM receptors) are three homologous receptor tyrosine kinases that bind vitamin K-dependent endogenous ligands, Protein S (ProS), and growth arrest-specific factor 6 (Gas6), and act as bridging molecules to promote phosphatidylserine (PS)-mediated clearance of apoptotic cells (efferocytosis). TAM receptors are overexpressed in a vast array of tumor types, whereby the level of expression correlates with the tumor grade and the emergence of chemo- and radioresistance to targeted therapeutics, but also have been implicated as inhibitory receptors on infiltrating myeloid-derived cells in the tumor microenvironment that can suppress host antitumor immunity. In the present study, we utilized TAM-IFNγR1 reporter lines and expressed TAM receptors in a variety of epithelial cell model systems to show that each TAM receptor has a unique pattern of activation by Gas6 or ProS, as well as unique dependency for PS on apoptotic cells and PS liposomes for activity. In addition, we leveraged this system to engineer epithelial cells that express wild-type TAM receptors and show that although each receptor can promote PS-mediated efferocytosis, AKT-mediated chemoresistance, as well as upregulate the immune checkpoint molecule PD-L1 on tumor cells, Mertk is most dominant in the aforementioned pathways. Functionally, TAM receptor-mediated efferocytosis could be partially blocked by PS-targeting antibody 11.31 and Annexin V, demonstrating the existence of a PS/PS receptor (i.e., TAM receptor)/PD-L1 axis that operates in epithelial cells to foster immune escape. These data provide a rationale that PS-targeting, anti-TAM receptor, and anti-PD-L1-based therapeutics will have merit as combinatorial checkpoint inhibitors.Implications: Many tumor cells are known to upregulate the immune checkpoint inhibitor PD-L1. This study demonstrates a role for PS and TAM receptors in the regulation of PD-L1 on cancer cells. Mol Cancer Res; 15(6); 753-64. ©2017 AACR.


Subject(s)
B7-H1 Antigen/metabolism , Drug Resistance, Neoplasm/physiology , Phosphatidylserines/metabolism , Proto-Oncogene Proteins c-akt/metabolism , c-Mer Tyrosine Kinase/metabolism , B7-H1 Antigen/genetics , Cell Line, Tumor , Epithelial Cells/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Liposomes , Protein Domains , Protein S/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , c-Mer Tyrosine Kinase/genetics , Axl Receptor Tyrosine Kinase , Interferon gamma Receptor
6.
Cancers (Basel) ; 8(12)2016 Nov 29.
Article in English | MEDLINE | ID: mdl-27916840

ABSTRACT

The TAM family of receptors (i.e., Tyro3, Axl, and Mertk), and their ligands Growth arrest specific factor 6 (Gas6) and Protein S (Pros1) contribute to several oncogenic processes, such as cell survival, invasion, migration, chemo-resistance, and metastasis, whereby expression often correlates with poor clinical outcomes. In recent years, there has been great interest in the study of TAM receptors in cancer, stemming both from their roles as oncogenic signaling receptors, as well as their roles in tumor immunology. As a result, several classes of TAM inhibitors that include small molecule tyrosine kinase inhibitors, monoclonal antibodies, decoy receptors, as well as novel strategies to target TAM ligands are being developed. This paper will review the biology of TAM receptors and their ligands with a focus on cancer, as well as evidence-based data for the continued pursuit of TAM/Gas6 inhibitors in clinical practice.

7.
Cell Commun Signal ; 14(1): 19, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27595981

ABSTRACT

BACKGROUND: Tyro3, Axl, and Mertk (TAMs) are a family of three conserved receptor tyrosine kinases that have pleiotropic roles in innate immunity and homeostasis and when overexpressed in cancer cells can drive tumorigenesis. METHODS: In the present study, we engineered EGFR/TAM chimeric receptors (EGFR/Tyro3, EGFR/Axl, and EGF/Mertk) with the goals to interrogate post-receptor functions of TAMs, and query whether TAMs have unique or overlapping post-receptor activation profiles. Stable expression of EGFR/TAMs in EGFR-deficient CHO cells afforded robust EGF inducible TAM receptor phosphorylation and activation of downstream signaling. RESULTS: Using a series of unbiased screening approaches, that include kinome-view analysis, phosphor-arrays, RNAseq/GSEA analysis, as well as cell biological and in vivo readouts, we provide evidence that each TAM has unique post-receptor signaling platforms and identify an intrinsic role for Axl that impinges on cell motility and invasion compared to Tyro3 and Mertk. CONCLUSION: These studies demonstrate that TAM show unique post-receptor signatures that impinge on distinct gene expression profiles and tumorigenic outcomes.


Subject(s)
ErbB Receptors/metabolism , Mammary Neoplasms, Experimental/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Animals , CHO Cells , Cell Movement , Cricetinae , Cricetulus , ErbB Receptors/genetics , Female , Humans , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , c-Mer Tyrosine Kinase , Axl Receptor Tyrosine Kinase
8.
Front Immunol ; 5: 566, 2014.
Article in English | MEDLINE | ID: mdl-25426118

ABSTRACT

The rapid and efficient clearance of apoptotic cells results in the elimination of auto-antigens and provides a strong anti-inflammatory and immunosuppressive signal to prevent autoimmunity. While professional and non-professional phagocytes utilize a wide array of surface receptors to recognize apoptotic cells, the recognition of phosphatidylserine (PS) on apoptotic cells by PS receptors on phagocytes is the emblematic signal for efferocytosis in metazoans. PS-dependent efferocytosis is associated with the production of anti-inflammatory factors such as IL-10 and TGF-ß that function, in part, to maintain tolerance to auto-antigens. In contrast, when apoptotic cells fail to be recognized and processed for degradation, auto-antigens persist, such as self-nucleic acids, which can trigger immune activation leading to autoantibody production and autoimmunity. Despite the fact that genetic mouse models clearly demonstrate that loss of PS receptors can lead to age-dependent auto-immune diseases reminiscent of systemic lupus erythematosus (SLE), the link between PS and defective clearance in chronic inflammation and human autoimmunity is not well delineated. In this perspective, we review emerging questions developing in the field that may be of relevance to SLE and human autoimmunity.

9.
J Biol Chem ; 289(37): 25737-49, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25074939

ABSTRACT

MERTK, a member of the TAM (TYRO3, AXL, and MERTK) receptor tyrosine kinases, has complex and diverse roles in cell biology. On the one hand, knock-out of MERTK results in age-dependent autoimmunity characterized by failure of apoptotic cell clearance, while on the other, MERTK overexpression in cancer drives classical oncogene pathways leading to cell transformation. To better understand the interplay between cell transformation and efferocytosis, we stably expressed MERTK in human MCF10A cells, a non-tumorigenic breast epithelial cell line devoid of endogenous MERTK. While stable expression of MERTK in MCF10A resulted in enhanced motility and AKT-mediated chemoprotection, MERTK-10A cells did not form stable colonies in soft agar, or enhance proliferation compared with parental MCF10A cells. Concomitant to chemoresistance, MERTK also stimulated efferocytosis in a gain-of-function capacity. However, unlike AXL, MERTK activation was highly dependent on apoptotic cells, suggesting MERTK may preferentially interface with phosphatidylserine. Consistent with this idea, knockdown of MERTK in breast cancer cells MDA-MB 231 reduced efferocytosis, while transient or stable expression of MERTK stimulated apoptotic cell clearance in all cell lines tested. Moreover, human breast cancer cells with higher endogenous MERTK showed higher levels of efferocytosis that could be blocked by soluble TAM receptors. Finally, through MERTK, apoptotic cells induced PD-L1 expression, an immune checkpoint blockade, suggesting that cancer cells may adopt MERTK-driven efferocytosis as an immune suppression mechanism for their advantage. These data collectively identify MERTK as a significant link between cancer progression and efferocytosis, and a potentially unrealized tumor-promoting event when MERTK is overexpressed in epithelial cells.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Epithelial Cells/metabolism , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Apoptosis/genetics , Breast Neoplasms/pathology , Cell Movement/genetics , Epithelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Phagocytosis/genetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , c-Mer Tyrosine Kinase , Axl Receptor Tyrosine Kinase
10.
Photochem Photobiol ; 89(1): 139-49, 2013.
Article in English | MEDLINE | ID: mdl-22803957

ABSTRACT

Phthalocyanine photosensitizers are effective in anticancer photodynamic therapy (PDT) but suffer from limited solubility, limited cellular uptake and limited selectivity for cancer cells. To improve these characteristics, we synthesized isopropylidene-protected and partially deprotected tetra ß-glycosylated zinc (II) phthalocyanines and compared their uptake and accumulation kinetics, subcellular localization, in vitro photocytotoxicity and reactive oxygen species generation with those of disulfonated aluminum phthalocyanine. In MCF-7 cancer cells, one of the compounds, zinc phthalocyanine {4}, demonstrated 10-fold higher uptake, 5-fold greater PDT-induced cellular reactive oxygen species concentration and 2-fold greater phototoxicity than equimolar (9 µm) disulfonated aluminum phthalocyanine. Thus, isopropylidene-protected ß-glycosylation of phthalocyanines provides a simple method of improving the efficacy of PDT.


Subject(s)
Antineoplastic Agents/pharmacology , Indoles/pharmacology , Organometallic Compounds/pharmacology , Photosensitizing Agents/pharmacology , Alkenes/chemistry , Antineoplastic Agents/chemical synthesis , Biological Transport , Cell Survival/drug effects , Cell Survival/radiation effects , Glycosylation , Humans , Indoles/chemical synthesis , Isoindoles , Kinetics , Light , MCF-7 Cells , Organometallic Compounds/chemical synthesis , Photochemotherapy , Photosensitizing Agents/chemical synthesis , Reactive Oxygen Species/metabolism , Zinc Compounds
11.
J Org Chem ; 77(23): 10638-47, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23126407

ABSTRACT

PEGylated chlorin e(6) photosensitizers were synthesized with tri(ethylene glycol) attached at the ester bond(s) for a 1:1 conjugate at the 17(3)-position, a 2:1 conjugate at the 15(2)- and 17(3)-positions, and a 3:1 conjugate at the 13(1)-, 15(2)-, and 17(3)-positions. These chlorin sensitizers were studied for hydrolytic stability and solubility, as well as ovarian OVCAR-5 cancer cell uptake, localization, and phototoxicity. Increasing numbers of the PEG groups in the mono-, di-, and tri-PEG chlorin conjugates increased the water solubility and sensitivity to hydrolysis and uptake into the ovarian cancer cells. The PEG chlorin conjugates accumulated in the cytoplasm and mitrochondria, but not in lysosomes. Higher phototoxicity was roughly correlated with higher numbers of PEG groups, with the tri-PEG chlorin conjugate showing the best overall ovarian cancer cell photokilling of the series. Singlet oxygen lifetimes, solvent deuteration, and the effects of additives azide ion and d-mannitol were examined to help clarify the photokilling mechanisms. A Type-II (singlet oxygen) photosensitized mechanism is suggested for the di- and tri-PEG chlorin conjugates; however, a more complicated process based in part on a Type-I (radicals or radical ions) mechanism is suggested for the parent chlorin e(6) and the mono-PEG chlorin conjugate.


Subject(s)
Ovarian Neoplasms/drug therapy , Photochemotherapy/methods , Polyethylene Glycols/pharmacology , Polyethylene Glycols/therapeutic use , Porphyrins/pharmacology , Porphyrins/therapeutic use , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/therapeutic use , Chlorophyllides , Female , Humans
12.
Photochem Photobiol ; 88(1): 175-87, 2012.
Article in English | MEDLINE | ID: mdl-22044030

ABSTRACT

Photodynamic therapy (PDT) is an increasingly popular anticancer treatment that uses photosensitizer, light and tissue oxygen to generate cytotoxic reactive oxygen species (ROS) within illuminated cells. Acting to counteract ROS-mediated damage are various cellular antioxidant pathways. In this study, we combined PDT with specific antioxidant inhibitors to potentiate PDT cytotoxicity in MCF-7 cancer cells. We used disulphonated aluminium phthalocyanine photosensitizer plus various combinations of the antioxidant inhibitors: diethyl-dithiocarbamate (DDC, a Cu/Zn-SOD inhibitor), 2-methoxyestradiol (2-ME, a Mn-SOD inhibitor), l-buthionine sulfoximine (BSO, a glutathione synthesis inhibitor) and 3-amino-1,2,4-triazole (3-AT, a catalase inhibitor). BSO, singly or in combination with other antioxidant inhibitors, significantly potentiated PDT cytotoxicity, corresponding with increased ROS levels and apoptosis. The greatest potentiation of cell death over PDT alone was seen when cells were preincubated for 24 h with 300 µM BSO plus 10 mM 3-AT (1.62-fold potentiation) or 300 µM BSO plus 1 µM 2-ME (1.52-fold), or with a combination of all four inhibitors (300 µM BSO, 10 mM 3-AT, 1 µM 2-ME and 10 µM DDC: 1.4-fold). As many of these inhibitors have already been clinically tested, this work facilitates future in vivo studies.


Subject(s)
Antioxidants , Photochemotherapy , Cell Line, Tumor , Humans
13.
Exp Cell Res ; 315(10): 1745-58, 2009 Jun 10.
Article in English | MEDLINE | ID: mdl-19233168

ABSTRACT

Prostaglandin E(1) (PGE(1)) lowers dermal interstitial fluid pressure (IFP) in vivo and inhibits fibroblast-mediated collagen gel contraction in vitro. PDGF-BB, in contrast, stimulates contraction and normalizes IFP lowered as a result of anaphylaxis. Human diploid AG1518 fibroblasts expressed EP2, EP3 and IP prostaglandin receptors. The inhibitory effect of PGE(1) on contraction depended on cAMP. Short-term stimulation with PDGF-BB transiently induced formation of actin-containing membrane and circular ruffles and breakdown of stress fibers. PGE(1) had no effect on stress fibers nor did it modulate the effects of PDGF-BB. PGE(1) alone or in combination with PDGF-BB inhibited initial adhesion and spreading to collagen. PDGF-BB had no effect on adhesion but stimulated cell spreading. Two-dimensional gel electrophoresis and MALDI TOF analyses of SDS/Triton X-100-soluble proteins revealed changes in migration pattern of actin-binding proteins. Interestingly, PDGF-BB and PGE(1) affected both similar and different sets of actin-binding proteins. PDGF-BB and PGE(1) did not trans-modulate their respective effects on actin-binding proteins, cytoskeletal organization or initial adhesion. Our data show that PDGF-BB stimulates actin cytoskeleton dynamics, whereas PGE(1) inhibits processes dependent on cytoskeletal motor functions. We suggest that these different activities may partly explain the contrasting effects of PGE(1) and PDGF-BB on contraction and IFP.


Subject(s)
Alprostadil/pharmacology , Cell Movement/drug effects , Microfilament Proteins/metabolism , Platelet-Derived Growth Factor/pharmacology , Actins/metabolism , Animals , Becaplermin , Calmodulin-Binding Proteins/metabolism , Cattle , Cell Adhesion/drug effects , Collagen/metabolism , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Electrophoresis, Gel, Two-Dimensional , Extracellular Signal-Regulated MAP Kinases/metabolism , Gels , Gene Expression Profiling , Humans , Myosin-Light-Chain Kinase/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism , Proto-Oncogene Proteins c-sis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Prostaglandin E/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...