Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
Int J Mol Sci ; 23(3)2022 Feb 02.
Article in English | MEDLINE | ID: mdl-35163650

ABSTRACT

Focal adhesion kinase (FAK), a non-receptor tyrosine kinase, is overexpressed and activated in many cancer types. FAK regulates diverse cellular processes, including growth factor signaling, cell cycle progression, cell survival, cell motility, angiogenesis, and the establishment of immunosuppressive tumor microenvironments through kinase-dependent and kinase-independent scaffolding functions in the cytoplasm and nucleus. Mounting evidence has indicated that targeting FAK, either alone or in combination with other agents, may represent a promising therapeutic strategy for various cancers. In this review, we summarize the mechanisms underlying FAK-mediated signaling networks during tumor development. We also summarize the recent progress of FAK-targeted small-molecule compounds for anticancer activity from preclinical and clinical evidence.


Subject(s)
Focal Adhesion Kinase 1/physiology , Neoplasms , Animals , Antineoplastic Agents/pharmacology , Humans , Neoplasms/metabolism , Neoplasms/therapy , Tumor Microenvironment
2.
Sci Rep ; 11(1): 23285, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34857846

ABSTRACT

Machine learning approaches have shown great promise in biology and medicine discovering hidden information to further understand complex biological and pathological processes. In this study, we developed a deep learning-based machine learning algorithm to meaningfully process image data and facilitate studies in vascular biology and pathology. Vascular injury and atherosclerosis are characterized by neointima formation caused by the aberrant accumulation and proliferation of vascular smooth muscle cells (VSMCs) within the vessel wall. Understanding how to control VSMC behaviors would promote the development of therapeutic targets to treat vascular diseases. However, the response to drug treatments among VSMCs with the same diseased vascular condition is often heterogeneous. Here, to identify the heterogeneous responses of drug treatments, we created an in vitro experimental model system using VSMC spheroids and developed a machine learning-based computational method called HETEROID (heterogeneous spheroid). First, we established a VSMC spheroid model that mimics neointima-like formation and the structure of arteries. Then, to identify the morphological subpopulations of drug-treated VSMC spheroids, we used a machine learning framework that combines deep learning-based spheroid segmentation and morphological clustering analysis. Our machine learning approach successfully showed that FAK, Rac, Rho, and Cdc42 inhibitors differentially affect spheroid morphology, suggesting that multiple drug responses of VSMC spheroid formation exist. Overall, our HETEROID pipeline enables detailed quantitative drug characterization of morphological changes in neointima formation, that occurs in vivo, by single-spheroid analysis.


Subject(s)
Machine Learning , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Atherosclerosis/pathology , Cells, Cultured , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/physiology , Humans , Neointima/pathology , Spheroids, Cellular/physiology , Vascular System Injuries/pathology , cdc42 GTP-Binding Protein/antagonists & inhibitors , cdc42 GTP-Binding Protein/physiology , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/physiology
3.
Theranostics ; 11(13): 6154-6172, 2021.
Article in English | MEDLINE | ID: mdl-33995651

ABSTRACT

SH2 domain containing tyrosine phosphatase 2 (Shp2; PTPN11) regulates several intracellular pathways downstream of multiple growth factor receptors. Our studies implicate that Shp2 interacts with Caveolin-1 (Cav-1) protein in retinal ganglion cells (RGCs) and negatively regulates BDNF/TrkB signaling. This study aimed to investigate the mechanisms underlying the protective effects of shp2 silencing in the RGCs in glaucomatous conditions. Methods: Shp2 was silenced in the Cav-1 deficient mice and the age matched wildtype littermates using adeno-associated viral (AAV) constructs. Shp2 expression modulation was performed in an acute and a chronic mouse model of experimental glaucoma. AAV2 expressing Shp2 eGFP-shRNA under a strong synthetic CAG promoter was administered intravitreally in the animals' eyes. The contralateral eye received AAV-eGFP-scramble-shRNA as control. Animals with Shp2 downregulation were subjected to either microbead injections or acute ocular hypertension experimental paradigm. Changes in inner retinal function were evaluated by measuring positive scotopic threshold response (pSTR) while structural and biochemical alterations were evaluated through H&E staining, western blotting and immunohistochemical analysis of the retinal tissues. Results: A greater loss of pSTR amplitudes was observed in the WT mice compared to Cav-1-/- retinas in both the models. Silencing of Shp2 phosphatase imparted protection against inner retinal function loss in chronic glaucoma model in WT mice. The functional rescue also translated to structural preservation of ganglion cell layer in the chronic glaucoma condition in WT mice which was not evident in Cav-1-/- mice retinas. Conclusions: This study indicates that protective effects of Shp2 ablation under chronic experimental glaucoma conditions are dependent on Cav-1 in the retina, suggesting in vivo interactions between the two proteins.


Subject(s)
Caveolin 1/physiology , Genetic Therapy , Genetic Vectors/therapeutic use , Glaucoma/therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Retina/pathology , Alpha-Globulins/genetics , Animals , Apoptosis , Brain-Derived Neurotrophic Factor/physiology , Caveolin 1/deficiency , Caveolin 1/genetics , DNA, Complementary/genetics , Dependovirus/genetics , Focal Adhesion Kinase 1/physiology , Gene Knockdown Techniques , Genes, Reporter , Genes, Synthetic , Glaucoma/metabolism , Glaucoma/pathology , Integrin beta1/physiology , Intraocular Pressure , Intravitreal Injections , Membrane Glycoproteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , Protein Tyrosine Phosphatase, Non-Receptor Type 11/biosynthesis , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein-Tyrosine Kinases/physiology , Up-Regulation
4.
Oncogene ; 40(4): 806-820, 2021 01.
Article in English | MEDLINE | ID: mdl-33262460

ABSTRACT

Uveal melanoma (UM) is a currently untreatable form of melanoma with a 50% mortality rate. Characterization of the essential signaling pathways driving this cancer is critical to develop target therapies. Activating mutations in the Gαq signaling pathway at the level of GNAQ, GNA11, or rarely CYSLTR2 or PLCß4 are considered alterations driving proliferation in UM and several other neoplastic disorders. Here, we systematically examined the oncogenic signaling output of various mutations recurrently identified in human tumors. We demonstrate that CYSLTR2 → GNAQ/11 → PLCß act in a linear signaling cascade that, via protein kinase C (PKC), activates in parallel the MAP-kinase and FAK/Yes-associated protein pathways. Using genetic ablation and pharmacological inhibition, we show that the PKC/RasGRP3/MAPK signaling branch is the essential component that drives the proliferation of UM. Only inhibition of the MAPK branch but not the FAK branch synergizes with inhibition of the proximal cascade, providing a blueprint for combination therapy. All oncogenic signaling could be extinguished by the novel GNAQ/11 inhibitor YM-254890, in all UM cells with driver mutation in the Gαq subunit or the upstream receptor. Our findings highlight the GNAQ/11 → PLCß â†’ PKC → MAPK pathway as the central signaling axis to be suppressed pharmacologically to treat for neoplastic disorders with Gαq pathway mutations.


Subject(s)
Melanoma/genetics , Oncogenes/physiology , Uveal Neoplasms/genetics , Animals , Cell Line, Tumor , Focal Adhesion Kinase 1/physiology , GTP-Binding Protein alpha Subunits/physiology , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Humans , MAP Kinase Signaling System , Melanoma/pathology , Mice , Mutation , Phospholipase C beta/physiology , Protein Kinase C/physiology , Receptors, Leukotriene/physiology , Signal Transduction/physiology , Uveal Neoplasms/pathology
5.
Int J Mol Sci ; 21(22)2020 Nov 10.
Article in English | MEDLINE | ID: mdl-33182556

ABSTRACT

Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma of children and adolescents. The fusion-positive (FP)-RMS variant expressing chimeric oncoproteins such as PAX3-FOXO1 and PAX7-FOXO1 is at high risk. The fusion negative subgroup, FN-RMS, has a good prognosis when non-metastatic. Despite a multimodal therapeutic approach, FP-RMS and metastatic FN-RMS often show a dismal prognosis with 5-year survival of less than 30%. Therefore, novel targets need to be discovered to develop therapies that halt tumor progression, reducing long-term side effects in young patients. Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that regulates focal contacts at the cellular edges. It plays a role in cell motility, survival, and proliferation in response to integrin and growth factor receptors' activation. FAK is often dysregulated in cancer, being upregulated and/or overactivated in several adult and pediatric tumor types. In RMS, both in vitro and preclinical studies point to a role of FAK in tumor cell motility/invasion and proliferation, which is inhibited by FAK inhibitors. In this review, we summarize the data on FAK expression and modulation in RMS. Moreover, we give an overview of the approaches to inhibit FAK in both preclinical and clinical cancer settings.


Subject(s)
Focal Adhesion Kinase 1/physiology , Rhabdomyosarcoma/physiopathology , Soft Tissue Neoplasms/physiopathology , Animals , Carcinogenesis , Child , Clinical Trials as Topic , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation, Neoplastic , Humans , Models, Biological , Molecular Targeted Therapy , Muscle Development , Neoplasm Invasiveness , Neoplasm Metastasis , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/physiology , Protein Kinase Inhibitors/pharmacology , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/therapy , Signal Transduction , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/therapy
6.
Endocrinology ; 161(10)2020 10 01.
Article in English | MEDLINE | ID: mdl-32761085

ABSTRACT

The blood-testis barrier (BTB) in the testis is an important ultrastructure to support spermatogenesis. This blood-tissue barrier undergoes remodeling at late stage VII to early stage IX of the epithelial cycle to support the transport of preleptotene spermatocytes across the BTB to prepare for meiosis I/II at the apical compartment through a mechanism that remains to be delineated. Studies have shown that NC1-peptide-derived collagen α3 (IV) chain in the basement membrane is a bioactive peptide that induces BTB remodeling. It also promotes the release of fully developed spermatids into the tubule lumen. Thus, this endogenously produced peptide coordinates these 2 cellular events across the seminiferous epithelium. Using an NC1-peptide complementary deoxyribonucleic acid (cDNA) construct to transfect adult rat testes for overexpression, NC1-peptide was found to effectively induce germ cell exfoliation and BTB remodeling, which was associated with a surge and activation of p-rpS6, the downstream signaling protein of mTORC1 and the concomitant downregulation of p-FAK-Y407 in the testis. In order to define the functional relationship between p-rpS6 and p-FAK-Y407 signaling to confer the ability of NC1-peptide to regulate testis function, a phosphomimetic (and thus constitutively active) mutant of p-FAK-Y407 (p-FAK-Y407E-MT) was used for its co-transfection, utilizing Sertoli cells cultured in vitro with a functional tight junction (TJ) barrier that mimicked the BTB in vivo. Overexpression of p-FAK-Y407E-MT blocked the effects of NC1-peptide to perturb Sertoli cell BTB function by promoting F-actin and microtubule cytoskeleton function, and downregulated the NC1-peptide-mediated induction of p-rpS6 activation. In brief, NC1-peptide is an important endogenously produced biomolecule that regulates BTB dynamics.


Subject(s)
Basement Membrane/metabolism , Collagen Type IV/physiology , Focal Adhesion Kinase 1/physiology , Peptide Fragments/physiology , Spermatogenesis/physiology , Testis/metabolism , Animals , Basement Membrane/chemistry , Blood-Testis Barrier/metabolism , Collagen Type IV/chemistry , Collagen Type IV/genetics , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mutation , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6/metabolism , Signal Transduction/physiology
7.
Br J Cancer ; 123(7): 1154-1163, 2020 09.
Article in English | MEDLINE | ID: mdl-32632148

ABSTRACT

BACKGROUND: Hepatitis B virus (HBV) has a crucial role in the progression of hepatocellular carcinoma (HCC). Tumour cells must develop anoikis resistance in order to survive before metastasis. This study aimed to investigate the mechanism of IQGAP1 in HBV-mediated anoikis evasion and metastasis in HCC cells. METHODS: IQGAP1 expression was detected by immunohistochemistry, real-time PCR and immunoblot analysis. Lentiviral-mediated stable upregulation or knockdown of IGAQP1, immunoprecipitation, etc. were used in function and mechanism study. RESULTS: IQGAP1 was markedly upregulated in HBV-positive compared with HBV-negative HCC cells and tissues. IQGAP1 was positively correlated to poor prognosis of HBV-associated HCC patients. IQGAP1 overexpression significantly enhanced the anchorage-independent growth and metastasis, whereas IQGAP1-deficient HCC cells are more sensitive to anoikis. Mechanistically, we found that HBV-induced ROS enhanced the association of IQGAP1 and Rac1 that activated Rac1, leading to phosphorylation of Src/FAK pathway. Antioxidants efficiently inhibited IQGAP1-mediated anoikis resistance and metastasis. CONCLUSIONS: Our study indicated an important mechanism by which upregulated IQGAP1 by HBV promoted anoikis resistance, migration and invasion of HCC cells through Rac1-dependent ROS accumulation and activation of Src/FAK signalling, suggesting IQGAP1 as a prognostic indicator and a novel therapeutic target in HCC patients with HBV infection.


Subject(s)
Carcinoma, Hepatocellular/pathology , Focal Adhesion Kinase 1/physiology , Liver Neoplasms/pathology , Reactive Oxygen Species/metabolism , rac1 GTP-Binding Protein/physiology , ras GTPase-Activating Proteins/physiology , src-Family Kinases/physiology , Animals , Anoikis , Cell Line, Tumor , Female , Hepatitis B/complications , Humans , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Signal Transduction/physiology
8.
Sci Rep ; 8(1): 2550, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29416084

ABSTRACT

Focal adhesion kinase (FAK) is essential in embryonic angiogenesis by regulating endothelial cell (EC) survival and barrier functions through its kinase-independent and -dependent activities. Here, we generated EC-specific tamoxifen-inducible FAK knockout and FAK kinase-defective (KD) mutant knockin mice to investigate the role of FAK and its kinase activity in angiogenesis of adult animals. Unlike previous observations of their differential defects in embryonic vascular development, both FAK ablation and inactivation of its kinase activity resulted in deficient angiogenesis in wound-healing as well as retinal angiogenesis models. Consistent with these phenotypes, loss of FAK or its kinase activity decreased EC proliferation and migration to similar extents, suggesting FAK primarily acts as a kinase for the regulation of adult EC-mediated angiogenesis. Further mechanistic analyses were carried out using an established mouse EC line MS1 cells. Interestingly, we found that FAK regulated the expression of VEGFR2, a central mediator of various EC functions and angiogenesis, which requires both FAK kinase activity and its translocation into the nucleus. Moreover, nuclear FAK was detected in the RNA polymerase II complex associated with VEGFR2 promoter, suggesting its direct participation in the transcriptional regulation of VEGFR2. Together, our results provide significant insights into the signaling mechanisms of FAK in angiogenesis that may contribute to future design of more effective angiogenesis related therapy.


Subject(s)
Endothelial Cells/metabolism , Focal Adhesion Kinase 1/metabolism , Retinal Neovascularization/genetics , Transcription, Genetic , Vascular Endothelial Growth Factor Receptor-2/genetics , Animals , Cell Movement , Cell Nucleus/metabolism , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Female , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/physiology , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Signal Transduction
9.
Rheumatology (Oxford) ; 57(3): 572-577, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29228301

ABSTRACT

Objectives: The aim was to explore the function of the T-cell cytokine IFNγ for mesenchymal tissue remodelling in RA and to determine whether IFNγ signalling controls the invasive potential of fibroblast-like synoviocytes (FLS). Methods: To assess architectural responses, FLS were cultured in three-dimensional micromasses. FLS motility was analysed in migration and invasion assays. Signalling events relevant to cellular motility were defined by western blots. Baricitinib and small interfering RNA pools were used to suppress Janus kinase (JAK) functions. Results: Histological analyses of micromasses revealed unique effects of IFNγ on FLS shape and tissue organization. This was consistent with accelerated migration upon IFNγ stimulation. Given that cell shape and cell motility are under the control of the focal adhesion kinase (FAK), we next analysed its activity. Indeed, IFNγ stimulation induced the phosphorylation of FAK-Y925, a phosphosite implicated in FAK-mediated cell migration. Small interfering RNA knockdown of JAK2, but not JAK1, substantially abrogated FAK activation by IFNγ. Correspondingly, IFNγ-induced FAK activation and invasion of FLS was abrogated by the JAK inhibitor, baricitinib. Conclusion: Our study contributes insight into the synovial response to IFNγ and reveals JAK2 as a potential therapeutic target for FLS-mediated joint destruction in arthritis, especially in RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , Fibroblasts/metabolism , Interferon-gamma/physiology , Janus Kinase 2/antagonists & inhibitors , Synoviocytes/metabolism , Adult , Arthritis, Rheumatoid/drug therapy , Azetidines/pharmacology , Cell Culture Techniques , Cell Movement/physiology , Cells, Cultured , Female , Focal Adhesion Kinase 1/physiology , Humans , Janus Kinase Inhibitors/pharmacology , Male , Middle Aged , Purines , Pyrazoles , RNA, Small Interfering/pharmacology , Sulfonamides/pharmacology
10.
Brain Res Bull ; 135: 149-156, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29031858

ABSTRACT

Treatment of neuropathic pain (NP) continues to be a clinical challenge and the underlying mechanisms of NP remain elusive. More evidence suggests that glial cell line-derived neurotrophic factor (GDNF) has potent anti-nociceptive effects on NP, but the underlying mechanisms are still largely unknown. Recent data have shown that integrin ß1 plays an important part in NP induction, and that the activity of integrin ß1 signaling is associated with the phosphorylation of the conserved threonines in the cytoplasmic domain and recruitment of focal adhesion kinase (FAK) to the integrin ß1 tail and phosphorylation. We assessed the effect of GDNF on integrinß1/FAK signaling in NP states. Immunostaining results showed that integrin ß1 was mainly observed in the superficial dorsal horn in the spinal cord of rats, and was mostly expressed in intrinsic neurons. Expression of p-integrin ß1 and the phosphorylation of integrin ß1-associated FAK, but not integrin ß1 itself, was up-regulated after chronic constriction injury (CCI), which could be reversed by GDNF, and the effect of GDNF on integrin ß1/FAK signaling was inhibited by pre-treatment with RET function-blocking antibody (RET Ab). Moreover, pre-treatment with RET Ab could antagonize the effect of GDNF on inhibiting the NP induced by CCI. These data suggest that GDNF can regulate integrin ß1 activity via a RET-related mechanism.


Subject(s)
Focal Adhesion Kinase 1/metabolism , Integrin beta1/physiology , Neuralgia/drug therapy , Neuralgia/metabolism , Analgesics/pharmacology , Animals , Focal Adhesion Kinase 1/physiology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor/physiology , Integrin beta1/metabolism , Integrins/metabolism , Integrins/physiology , Male , Nerve Growth Factors/metabolism , Nerve Growth Factors/pharmacology , Nerve Growth Factors/physiology , Neuralgia/physiopathology , Neuroglia/metabolism , Neuroglia/physiology , Phosphorylation , Proto-Oncogene Proteins c-ret/metabolism , Rats , Rats, Sprague-Dawley , Sciatic Nerve/injuries , Signal Transduction/drug effects , Spinal Cord Dorsal Horn/metabolism , Up-Regulation/drug effects
11.
Acta Biochim Biophys Sin (Shanghai) ; 49(7): 581-587, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28475688

ABSTRACT

The growth arrest-specific gene 7 (GAS7), a member of the growth-arrest-specific family, encodes three protein isoforms (GAS7A, GAS7B, and GAS7C) and plays a potential role in lung cancer as a tumor suppressor gene. In the present study, we found low endogenous expressions of GAS7C mRNA and protein in hepatocellular carcinoma (HCC) cell lines compared with normal liver cells, and that there was a distinct increase of GAS7C expression in HCC cells treated with oxaliplatin. CCK8, apoptosis, and Transwell migration assays showed that cell proliferation and motility of HepG2 and MHCC-97 H cells were inhibited by oxaliplatin, while apoptosis was increased. Interestingly, western blot analysis showed that treatment with oxaliplatin increased GAS7C and N-WASP protein levels and decreased the levels of proteins involved in the fibronectin/integrin/FAK pathway, such as FAK, in both HCC cell lines. In addition, ectopically overexpressed GAS7C obviously inhibited cell proliferation and cell motility. Flow cytometry results showed that overexpression of GAS7C induced apoptosis of HepG2 and MHCC-97 H cells. We further confirmed the correlation between GAS7C and the N-WASP/FAK/F-actin pathway by q-PCR and western blot analysis of in GAS7C-overexpressing HepG2 and MHCC-97 H cells. Inhibition of GAS7C substantially reversed the anti-cancer effect of oxaliplatin and blocked the activity of the N-WASP/FAK/F-actin pathway. Taken together, our results showed that oxaliplatin inhibits HCC cell proliferation and migration ability by up-regulating GAS7C and activating the N-WASP/FAK/F-actin pathway.


Subject(s)
Actins/physiology , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Focal Adhesion Kinase 1/physiology , Liver Neoplasms/drug therapy , Nerve Tissue Proteins/physiology , Organoplatinum Compounds/pharmacology , Signal Transduction/drug effects , Wiskott-Aldrich Syndrome Protein, Neuronal/physiology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Liver Neoplasms/pathology , Oxaliplatin
12.
Oncotarget ; 7(43): 70336-70352, 2016 Oct 25.
Article in English | MEDLINE | ID: mdl-27611942

ABSTRACT

A key hallmark of cancer cells is the loss of positional control over growth and survival. Focal adhesion kinase (FAK) is a tyrosine kinase localised at sites of integrin-mediated cell adhesion to the extracellular matrix. FAK controls a number of adhesion-dependent cellular functions, including migration, proliferation and survival. Although FAK is overexpressed and activated in metastatic tumours, where it promotes invasion, it can also be elevated in cancers that have yet to become invasive. The contribution of FAK to the early stages of tumourigenesis is not known. We have examined the effect of activating FAK in non-transformed mammary epithelial cells (MECs) to understand its role in tumour initiation. In agreement with previous studies, we find FAK activation in 2D-culture promotes proliferation, migration, and epithelial-to-mesenchymal transition. However in 3D-cultures that better resemble normal tissue morphology, mammary cells largely respond to FAK activation via suppression of apoptosis, promoting aberrant acinar morphogenesis. This is an acquired function of FAK, because endogenous FAK signalling is not required for normal morphogenesis in 3D-culture or in vivo. Thus, FAK activation may facilitate tumour initiation by causing resistance to apoptosis. We suggest that aberrant FAK activation in breast epithelia is dependent upon the tissue context in which it occurs.


Subject(s)
Apoptosis , Breast Neoplasms/etiology , Focal Adhesion Kinase 1/physiology , Animals , Breast/pathology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic , Epithelial-Mesenchymal Transition , Female , Focal Adhesion Kinase 1/antagonists & inhibitors , Humans , Hyperplasia , Mice
13.
Clin Exp Pharmacol Physiol ; 43(10): 939-50, 2016 10.
Article in English | MEDLINE | ID: mdl-27297262

ABSTRACT

Coumarins, identified as plant secondary metabolites possess diverse biological activities including anti-angiogenic properties. Daphnetin (DAP), a plant derived dihydroxylated derivative of coumarin has shown significant pharmacological properties such as anticancer, anti-arthritic and anti-inflammatory. The present study was performed to investigate the anti-angiogenic potential of DAP, focusing on the mechanism of action. The in vivo anti-angiogenic potential of DAP was evaluated by vascular endothelial growth factor (VEGF)-induced rat aortic ring (RAR) assay and chick chorioallantoic membrane (CAM) assay. For in vitro evaluation, wounding migration, transwell invasion, tube formation and apoptosis assays were performed on VEGF (8 ng/mL)-induced human umbilical vein endothelial cells (HUVECs). The cellular mechanism of DAP was examined on TNFα (10 ng/mL) and VEGF-induced HUVECs by extracting the mRNA and protein levels using RT-qPCR and western blotting. Our data demonstrated that DAP inhibited the in vivo angiogenesis in the RAR and CAM assay. DAP also inhibited the different steps of angiogenesis, such as migration, invasion, and tube formation in HUVECs. DAP inhibited nuclear factor-κB signalling together including TNF-α induced IκBα degradation; phosphorylation of IκB kinase (IKKα/ß) and translocation of the NF-κB-p65 protein. Furthermore, western blotting revealed that DAP significantly down-regulated the VEGF-induced signalling such as c-Src, FAK, ERK1/2 and the related phosphorylation of protein kinase B (Akt) and VEGFR2 expressions. DAP reduced the elevated mRNA expression of iNOS, MMP2 and also, induced apoptosis in VEGF-stimulated HUVECs by the caspase-3 dependent pathway. Taken together, this study reveals that DAP may have novel prospective as a new multi-targeted medication for the anti-angiogenesis and cancer therapy.


Subject(s)
MAP Kinase Signaling System/drug effects , NF-kappa B/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Umbelliferones/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Angiogenesis Inducing Agents/pharmacology , Animals , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Chick Embryo , Chorioallantoic Membrane/drug effects , Chorioallantoic Membrane/physiology , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/physiology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/physiology , Humans , MAP Kinase Signaling System/physiology , Male , NF-kappa B/physiology , Organ Culture Techniques , Proto-Oncogene Proteins c-akt/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/pharmacology , Vascular Endothelial Growth Factor A/pharmacology
14.
Rheumatology (Oxford) ; 55(5): 928-38, 2016 May.
Article in English | MEDLINE | ID: mdl-26715774

ABSTRACT

OBJECTIVE: The objective of this study was to investigate the roles of dickkopf-1 (DKK-1) and integrin-related focal adhesion kinase (FAK) by TNF-α on the migration of fibroblast-like synoviocytes (FLSs) in RA. METHODS: Wound scratch assays were performed to assess FLS migration. Western blotting was used to measure the levels of DKK-1, Wnt signalling molecules and FAK signalling molecules. Quantitative real-time PCR was used to measure the expression levels of DKK-1, integrin αv, laminin, fibronectin, E-cadherin, MMP-8 and MMP-13. The concentrations of DKK-1, TNF-α and GSK-3ß were measured by ELISA. Genetic silencing of TNF-α was achieved by the transfection of small interfering RNA into cells. RESULTS: Migrating RA FLSs exhibited higher levels of DKK-1 and TNF-α expression compared with those in OA FLSs and/or stationary RA FLSs. Moreover, migrating FLSs exhibited significantly higher levels of FAK, p-JNK, paxillin and cdc42 expression, whereas the level of cytosolic ß-catenin was lower. WAY-262611, Wnt pathway agonist via inhibition of DKK-1, markedly inhibited cell migration of RA FLSs through the accumulation of cytosolic ß-catenin and suppression of FAK-related signalling pathways. TNF-α treatment to RA FLSs up-regulated expression of DKK-1, integrin αv, fibronectin, laminin and MMP-13. TNF-α stimulation also suppressed cytosolic ß-catenin and E-cadherin expression in a time-dependent manner. Moreover, TNF-α small interfering RNA-transfected migrating FLSs exhibited decreased activation of integrin-related FAK, paxillin, p-JNK and cdc42 signalling pathways. CONCLUSION: This study demonstrates that the activation of DKK-1 and the integrin-related FAK signalling pathway stimulated by TNF-α induces the dissociation of ß-catenin/E-cadherin, thus promoting RA FLS migration.


Subject(s)
Arthritis, Rheumatoid/pathology , Focal Adhesion Kinase 1/physiology , Intercellular Signaling Peptides and Proteins/physiology , Synovial Membrane/pathology , Tumor Necrosis Factor-alpha/physiology , Aged , Arthritis, Rheumatoid/metabolism , Cadherins/metabolism , Cell Movement/physiology , Cells, Cultured , Female , Fibroblasts/physiology , Focal Adhesion Kinase 1/genetics , Gene Silencing , Humans , Intercellular Signaling Peptides and Proteins/genetics , Middle Aged , Osteoarthritis/metabolism , Osteoarthritis/pathology , RNA, Small Interfering/genetics , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/genetics , beta Catenin/metabolism
15.
Oncogene ; 35(15): 1926-42, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-26119934

ABSTRACT

Cancer cells often gains a growth advantage by taking up glucose at a high rate and undergoing aerobic glycolysis through intrinsic cellular factors that reprogram glucose metabolism. Focal adhesion kinase (FAK), a key transmitter of growth factor and anchorage stimulation, is aberrantly overexpressed or activated in most solid tumors, including pancreatic ductal adenocarcinomas (PDACs). We determined whether FAK can act as an intrinsic driver to promote aerobic glycolysis and tumorigenesis. FAK inhibition decreases and overexpression increases intracellular glucose levels during unfavorable conditions, including growth factor deficiency and cell detachment. Amplex glucose assay, fluorescence and carbon-13 tracing studies demonstrate that FAK promotes glucose consumption and glucose-to-lactate conversion. Extracellular flux analysis indicates that FAK enhances glycolysis and decreases mitochondrial respiration. FAK increases key glycolytic proteins, including enolase, pyruvate kinase M2 (PKM2), lactate dehydrogenase and monocarboxylate transporter. Furthermore, active/tyrosine-phosphorylated FAK directly binds to PKM2 and promotes PKM2-mediated glycolysis. On the other hand, FAK-decreased levels of mitochondrial complex I can result in reduced oxidative phosphorylation (OXPHOS). Attenuation of FAK-enhanced glycolysis re-sensitizes cancer cells to growth factor withdrawal, decreases cell viability and reduces growth of tumor xenografts. These observations, for the first time, establish a vital role of FAK in cancer glucose metabolism through alterations in the OXPHOS-to-glycolysis balance. Broadly targeting the common phenotype of aerobic glycolysis and more specifically FAK-reprogrammed glucose metabolism will disrupt the bioenergetic and biosynthetic supply for uncontrolled growth of tumors, particularly glycolytic PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Focal Adhesion Kinase 1/physiology , Glycolysis , Mitochondria/physiology , Neoplasm Proteins/physiology , Pancreatic Neoplasms/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Adhesion , Cell Line, Tumor , Cells, Cultured , Electron Transport Complex I/metabolism , Epithelial Cells/metabolism , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Glucose Transporter Type 1/biosynthesis , Glucose Transporter Type 1/genetics , Humans , Lactates/metabolism , Neoplasm Proteins/genetics , Oxidative Phosphorylation , Pancreatic Ducts/cytology , Pancreatic Ducts/metabolism , Pancreatic Neoplasms/pathology , Phosphorylation , Phosphotyrosine/metabolism , Protein Processing, Post-Translational , Protein Structure, Tertiary , RNA Interference , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/metabolism , Transfection
16.
J Clin Invest ; 125(7): 2690-701, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26053662

ABSTRACT

Current stem cell-based strategies for tissue regeneration involve ex vivo manipulation of these cells to confer features of the desired progenitor population. Recently, the concept that endogenous stem/progenitor cells could be used for regenerating tissues has emerged as a promising approach that potentially overcomes the obstacles related to cell transplantation. Here we applied this strategy for the regeneration of injured tendons in a rat model. First, we identified a rare fraction of tendon cells that was positive for the known tendon stem cell marker CD146 and exhibited clonogenic capacity, as well as multilineage differentiation ability. These tendon-resident CD146+ stem/progenitor cells were selectively enriched by connective tissue growth factor delivery (CTGF delivery) in the early phase of tendon healing, followed by tenogenic differentiation in the later phase. The time-controlled proliferation and differentiation of CD146+ stem/progenitor cells by CTGF delivery successfully led to tendon regeneration with densely aligned collagen fibers, normal level of cellularity, and functional restoration. Using siRNA knockdown to evaluate factors involved in tendon generation, we demonstrated that the FAK/ERK1/2 signaling pathway regulates CTGF-induced proliferation and differentiation of CD146+ stem/progenitor cells. Together, our findings support the use of endogenous stem/progenitor cells as a strategy for tendon regeneration without cell transplantation and suggest this approach warrants exploration in other tissues.


Subject(s)
Adult Stem Cells/physiology , Regeneration/physiology , Tendon Injuries/therapy , Tendons/cytology , Tendons/physiology , Adult Stem Cells/cytology , Adult Stem Cells/transplantation , Animals , CD146 Antigen/metabolism , Cell Differentiation , Cell Proliferation , Colony-Forming Units Assay , Connective Tissue Growth Factor/administration & dosage , Focal Adhesion Kinase 1/physiology , MAP Kinase Signaling System , Multipotent Stem Cells/cytology , Multipotent Stem Cells/physiology , Multipotent Stem Cells/transplantation , Rats , Rats, Sprague-Dawley , Tendon Injuries/pathology , Tendon Injuries/physiopathology , Tissue Engineering/methods , Wound Healing
17.
Exp Dermatol ; 24(8): 579-84, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25877039

ABSTRACT

One of the key features of keloid is its fibroblasts migrating beyond the original wound border. During migration, cells not only undergo molecular changes but also mechanical modulation. This process is led by actin filaments serving as the backbone of intra-cellular force and transduces external mechanical signal via focal adhesion complex into the cell. Here, we focus on determining the mechanical changes of actin filaments and the spatial distribution of forces in response to changing chemical stimulations and during cell migration. Atomic force microscopy and micropost array detector are used to determine and compare the magnitude and distribution of filament elasticity and force generation in fibroblasts and keloid fibroblasts. We found both filament elasticity and force generation show spatial distribution in a polarized and migrating cell. Such spatial distribution is disrupted when mechano-signalling is perturbed by focal adhesion kinase inhibitor and in keloid fibroblasts. The demonstration of keloid pathology at the nanoscale highlights the coupling of cytoskeletal function with physical characters at the subcellular level and provides new research directions for migration-related disease such as keloid.


Subject(s)
Cytoskeleton/physiology , Fibroblasts/physiology , Keloid/pathology , Actin Cytoskeleton/physiology , Animals , Cell Movement , Cell Polarity , Elasticity , Fibroblasts/ultrastructure , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/physiology , Focal Adhesions/physiology , Humans , Mice , Microscopy, Atomic Force , NIH 3T3 Cells , Quinolones/pharmacology , Stress, Mechanical , Sulfones/pharmacology , Wound Healing
18.
Wound Repair Regen ; 23(3): 394-402, 2015.
Article in English | MEDLINE | ID: mdl-25847391

ABSTRACT

Tendon regeneration and healing requires tenocytes to move to the repair site followed by proliferation and synthesis of the extracellular matrix. A novel synthetic growth factor, mechano-growth factor (MGF), has been discovered to have positive roles in tissue repair through the improvement of cell proliferation and migration and the protection of cells against injury-induced apoptosis. However, it remains unclear whether MGF has the potential to accelerate tendon repair. In this study, using a transwell system, we found that MGF-C25E (a synthetic mechano-growth factor E peptide) significantly promotes tenocyte invasion, which was accompanied by the increased phosphorylation of focal adhesion kinase (FAK) and extracellular signal regulated kinase1/2 (ERK1/2) as well as the increased activity of matrix metalloproteinases-2 (MMP-2). The MMP-2 inhibitor OA-Hy blocked MGF-C25E-promoted tenocyte invasion. Inhibitors of FAK or ERK1/2 blocked MGF-C25E-promoted tenocyte invasion and MMP-2 activity as well. These results indicate that MGF-C25E promotes tenocyte invasion by increasing MMP-2 activity via the FAK-ERK1/2 signaling pathway. Taken together, our findings provide the first evidence that MGF-C25E enhances tenocyte invasion and indicate that it may serve as a potential repair material for promoting the healing and regeneration of injured tendons.


Subject(s)
Achilles Tendon/pathology , Focal Adhesion Kinase 1/metabolism , Insulin-Like Growth Factor I/metabolism , MAP Kinase Signaling System/physiology , Matrix Metalloproteinase 2/metabolism , Wound Healing , Achilles Tendon/cytology , Achilles Tendon/injuries , Animals , Cell Proliferation , Disease Models, Animal , Focal Adhesion Kinase 1/physiology , Male , Rats , Rats, Sprague-Dawley , Regeneration
19.
Circ Heart Fail ; 8(2): 342-51, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25654972

ABSTRACT

BACKGROUND: Myocardial infarction (MI) is one of the major causes of death worldwide. Chronic heart failure is a serious complication of MI that leads to poor prognosis. We recently found that neuron-derived neurotrophic factor (NDNF) is a proangiogenic secretory protein that is upregulated in ischemic skeletal muscle. Here, we examined whether NDNF modulates cardiac remodeling in response to chronic ischemia. METHODS AND RESULTS: C57BL/6J wild-type mice were subjected to the permanent ligation of the left anterior descending coronary artery to create MI. Adenoviral vectors expressing NDNF or ß-galactosidase (control) were intramuscularly injected into mice 3 days before permanent left anterior descending coronary artery ligation. Intramuscular administration of adenoviral vectors expressing NDNF to mice resulted in increased levels of circulating NDNF. Adenoviral vectors expressing NDNF administration improved left ventricular systolic dysfunction and dilatation after MI surgery. Moreover, adenoviral vectors expressing NDNF enhanced capillary formation and reduced cardiomyocyte apoptosis and hypertrophy in the post-MI hearts. Treatment of cultured cardiomyocytes with recombinant NDNF protein led to reduced apoptosis under conditions of hypoxia. NDNF also promoted the phosphorylation of Akt and focal adhesion kinase in cardiomyocytes. Blockade of focal adhesion kinase activation blocked the stimulatory effects of NDNF on cardiomyocyte survival and Akt phosphorylation. Similarly, treatment of cultured endothelial cells with NDNF protein led to enhancement of network formation and Akt phosphorylation, which was diminished by focal adhesion kinase inhibition. CONCLUSIONS: These data suggest that NDNF ameliorates adverse myocardial remodeling after MI by its abilities to enhance myocyte survival and angiogenesis in the heart through focal adhesion kinase/Akt-dependent mechanisms.


Subject(s)
Myocardial Infarction/physiopathology , Myocytes, Cardiac/physiology , Nerve Growth Factors/physiology , Animals , Apoptosis/physiology , Cell Survival/physiology , Disease Models, Animal , Focal Adhesion Kinase 1/physiology , Injections, Intramuscular , Male , Mice, Inbred C57BL , Neovascularization, Physiologic/physiology , Nerve Growth Factors/administration & dosage , Nerve Growth Factors/metabolism , Phosphorylation/physiology , Proto-Oncogene Proteins c-akt/physiology
20.
Exp Dermatol ; 23(12): 936-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25308203

ABSTRACT

The lymphatic system in skin plays important roles in drainage of wastes and in the afferent phase of immune response. We previously showed that activation of vascular endothelial growth factor receptor (VEGFR), specifically the VEGFC/VEGFR-3 pathway, attenuates oedema and inflammation by promoting lymphangiogenesis, suggesting a protective role of lymphatic vessels against skin inflammation. However, it remains unknown how physical stimuli promote lymphatic function. Here, we show that lymphatic endothelial cells (LECs) are activated by direct-current (DC) electrical stimulation, which induced extension of actin filaments of LECs, increased calcium influx into LECs, and increased phosphorylation of p38 mitogen-activated protein kinase (MAPK). An inhibitor of focal adhesion kinase, which plays a role in cellular adhesion and motility, diminished the DC-induced extension of F-actin and abrogated p38 phosphorylation. Time-lapse imaging revealed that pulsed-DC stimulation promoted proliferation and migration of LECs. Overall, these results indicate that electro-stimulation activates lymphatic function by activating p38 MAPK.


Subject(s)
Endothelial Cells/physiology , Calcium Signaling , Cell Movement , Cell Proliferation , Cells, Cultured , Cytoskeleton/physiology , Electric Stimulation , Endothelial Cells/cytology , Focal Adhesion Kinase 1/physiology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...