1
|
Modeling invasion patterns in the glioblastoma battlefield. PLoS Comput Biol 2021; 17:e1008632. [PMID: 33513131 PMCID: PMC7875342 DOI: 10.1371/journal.pcbi.1008632] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 02/10/2021] [Accepted: 12/14/2020] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma is the most aggressive tumor of the central nervous system, due to its great infiltration capacity. Understanding the mechanisms that regulate the Glioblastoma invasion front is a major challenge with preeminent potential clinical relevances. In the infiltration front, the key features of tumor dynamics relate to biochemical and biomechanical aspects, which result in the extension of cellular protrusions known as tumor microtubes. The coordination of metalloproteases expression, extracellular matrix degradation, and integrin activity emerges as a leading mechanism that facilitates Glioblastoma expansion and infiltration in uncontaminated brain regions. We propose a novel multidisciplinary approach, based on in vivo experiments in Drosophila and mathematical models, that describes the dynamics of active and inactive integrins in relation to matrix metalloprotease concentration and tumor density at the Glioblastoma invasion front. The mathematical model is based on a non-linear system of evolution equations in which the mechanisms leading chemotaxis, haptotaxis, and front dynamics compete with the movement induced by the saturated flux in porous media. This approach is able to capture the relative influences of the involved agents and reproduce the formation of patterns, which drive tumor front evolution. These patterns have the value of providing biomarker information that is related to the direction of the dynamical evolution of the front and based on static measures of proteins in several tumor samples. Furthermore, we consider in our model biomechanical elements, like the tissue porosity, as indicators of the healthy tissue resistance to tumor progression. Glioblastoma (GB) is a type of brain cancer that originated from glial cells. The infiltrative nature of GB cells is a key feature for understanding its aggressiveness and resistance to current treatments. Cellular protrusions, named as Tumor Microtubes (TMs) in GB, mediate the interaction between tumor and healthy tissue and the processes leading GB invasion. These protrusions are also responsible for several cell communication pathways (e.g. Hedgehog or WNT). We have developed a multidisciplinary approach, which combined biological biomarker measurements performed in Drosophila GB with a novel mathematical model, to determine the interactions between proteases, integrins, and TM dynamics. The resulting model is able to predict the formation and infiltration of GB fronts, and, therefore, the directionality, aggressiveness, and progression of the tumor.
Collapse
|
2
|
Hung IC, Chen TM, Lin JP, Tai YL, Shen TL, Lee SJ. Wnt5b integrates Fak1a to mediate gastrulation cell movements via Rac1 and Cdc42. Open Biol 2020; 10:190273. [PMID: 32097584 PMCID: PMC7058935 DOI: 10.1098/rsob.190273] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Focal adhesion kinase (FAK) mediates vital cellular pathways during development. Despite its necessity, how FAK regulates and integrates with other signals during early embryogenesis remains poorly understood. We found that the loss of Fak1a impaired epiboly, convergent extension and hypoblast cell migration in zebrafish embryos. We also observed a clear disturbance in cortical actin at the blastoderm margin and distribution of yolk syncytial nuclei. In addition, we investigated a possible link between Fak1a and a well-known gastrulation regulator, Wnt5b, and revealed that the overexpression of fak1a or wnt5b could cross-rescue convergence defects induced by a wnt5b or fak1a antisense morpholino (MO), respectively. Wnt5b and Fak1a were shown to converge in regulating Rac1 and Cdc42, which could synergistically rescue wnt5b and fak1a morphant phenotypes. Furthermore, we generated several alleles of fak1a mutants using CRISPR/Cas9, but those mutants only revealed mild gastrulation defects. However, injection of a subthreshold level of the wnt5b MO induced severe gastrulation defects in fak1a mutants, which suggested that the upregulated expression of wnt5b might complement the loss of Fak1a. Collectively, we demonstrated that a functional interaction between Wnt and FAK signalling mediates gastrulation cell movements via the possible regulation of Rac1 and Cdc42 and subsequent actin dynamics.
Collapse
Affiliation(s)
- I-Chen Hung
- Department of Life Science, National Taiwan University, No. 1, Roosevelt Road, Section 4, Taipei 10617, Taiwan
| | - Tsung-Ming Chen
- Department of Life Science, National Taiwan University, No. 1, Roosevelt Road, Section 4, Taipei 10617, Taiwan.,Department of Plant Pathology and Microbiology, National Taiwan University, No. 1, Roosevelt Road, Section 4, Taipei 10617, Taiwan.,Department and Graduate Institute of Aquaculture, National Kaohsiung Marine University, Kaohsiung, Taiwan
| | - Jing-Ping Lin
- Department of Plant Pathology and Microbiology, National Taiwan University, No. 1, Roosevelt Road, Section 4, Taipei 10617, Taiwan
| | - Yu-Ling Tai
- Department of Plant Pathology and Microbiology, National Taiwan University, No. 1, Roosevelt Road, Section 4, Taipei 10617, Taiwan
| | - Tang-Long Shen
- Department of Plant Pathology and Microbiology, National Taiwan University, No. 1, Roosevelt Road, Section 4, Taipei 10617, Taiwan.,Center for Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Shyh-Jye Lee
- Department of Life Science, National Taiwan University, No. 1, Roosevelt Road, Section 4, Taipei 10617, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.,Center for Biotechnology, National Taiwan University, Taipei, Taiwan.,Center for Systems Biology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
3
|
Macabenta F, Stathopoulos A. Migrating cells control morphogenesis of substratum serving as track to promote directional movement of the collective. Development 2019; 146:dev.177295. [PMID: 31239242 DOI: 10.1242/dev.177295] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 06/17/2019] [Indexed: 11/20/2022]
Abstract
In Drosophila embryos, caudal visceral mesoderm (CVM) cells undergo bilateral migration along the trunk visceral mesoderm (TVM) in order to form midgut muscles. Mutation of FGF receptor Heartless (Htl) has been shown to cause CVM migration defects, particularly midline crossing of the bilateral groups. Here, we show that htl mutants also exhibit TVM defects including contralateral merging. Both CVM mismigration and TVM contralateral merging are attenuated by restoring FGF signaling specifically in the CVM, suggesting that migrating CVM cells influence TVM morphogenesis; however, the inverse, supplying FGF to the TVM, does not rescue CVM mismigration. In addition, we show that FGF regulates integrin expression in both tissues, but only providing a source of integrin specifically to the TVM attenuates the contralateral merging phenotype. Finally, we demonstrate that the CVM influences cell shape in the TVM, and a loss of CVM results in TVM morphological defects. In summary, this study provides insight into how a migrating collective of cells can influence their tissue substrate and supports the view that morphogenesis of tissues during development is interdependent.
Collapse
Affiliation(s)
- Frank Macabenta
- California Institute of Technology, Division of Biology and Biological Engineering, 1200 East California Boulevard, Pasadena, CA 91125, USA
| | - Angelike Stathopoulos
- California Institute of Technology, Division of Biology and Biological Engineering, 1200 East California Boulevard, Pasadena, CA 91125, USA
| |
Collapse
|
4
|
Mapping Heart Development in Flies: Src42A Acts Non-Autonomously to Promote Heart Tube Formation in Drosophila. Vet Sci 2017; 4:vetsci4020023. [PMID: 29056682 PMCID: PMC5606601 DOI: 10.3390/vetsci4020023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 11/27/2016] [Accepted: 12/07/2016] [Indexed: 12/26/2022] Open
Abstract
Congenital heart defects, clinically identified in both small and large animals, are multifactorial and complex. Although heritable factors are known to have a role in cardiovascular disease, the full genetic aetiology remains unclear. Model organism research has proven valuable in providing a deeper understanding of the essential factors in heart development. For example, mouse knock-out studies reveal a role for the Integrin adhesion receptor in cardiac tissue. Recent research in Drosophila melanogaster (the fruit fly), a powerful experimental model, has demonstrated that the link between the extracellular matrix and the cell, mediated by Integrins, is required for multiple aspects of cardiogenesis. Here we test the hypothesis that Integrins signal to the heart cells through Src42A kinase. Using the powerful genetics and cell biology analysis possible in Drosophila, we demonstrate that Src42A acts in early events of heart tube development. Careful examination of mutant heart tissue and genetic interaction data suggests that Src42A’s role is independent of Integrin and the Integrin-related Focal Adhesion Kinase. Rather, Src42A acts non-autonomously by promoting programmed cell death of the amnioserosa, a transient tissue that neighbors the developing heart.
Collapse
|
5
|
Anllo L, Schüpbach T. Signaling through the G-protein-coupled receptor Rickets is important for polarity, detachment, and migration of the border cells in Drosophila. Dev Biol 2016; 414:193-206. [PMID: 27130192 PMCID: PMC4887387 DOI: 10.1016/j.ydbio.2016.04.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 04/08/2016] [Accepted: 04/24/2016] [Indexed: 01/25/2023]
Abstract
Cell migration plays crucial roles during development. An excellent model to study coordinated cell movements is provided by the migration of border cell clusters within a developing Drosophila egg chamber. In a mutagenesis screen, we isolated two alleles of the gene rickets (rk) encoding a G-protein-coupled receptor. The rk alleles result in border cell migration defects in a significant fraction of egg chambers. In rk mutants, border cells are properly specified and express the marker Slbo. Yet, analysis of both fixed as well as live samples revealed that some single border cells lag behind the main border cell cluster during migration, or, in other cases, the entire border cell cluster can remain tethered to the anterior epithelium as it migrates. These defects are observed significantly more often in mosaic border cell clusters, than in full mutant clusters. Reduction of the Rk ligand, Bursicon, in the border cell cluster also resulted in migration defects, strongly suggesting that Rk signaling is utilized for communication within the border cell cluster itself. The mutant border cell clusters show defects in localization of the adhesion protein E-cadherin, and apical polarity proteins during migration. E-cadherin mislocalization occurs in mosaic clusters, but not in full mutant clusters, correlating well with the rk border cell migration phenotype. Our work has identified a receptor with a previously unknown role in border cell migration that appears to regulate detachment and polarity of the border cell cluster coordinating processes within the cells of the cluster themselves.
Collapse
Affiliation(s)
- Lauren Anllo
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Trudi Schüpbach
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
6
|
Cordero JB, Ridgway RA, Valeri N, Nixon C, Frame MC, Muller WJ, Vidal M, Sansom OJ. c-Src drives intestinal regeneration and transformation. EMBO J 2014; 33:1474-91. [PMID: 24788409 PMCID: PMC4194090 DOI: 10.1002/embj.201387454] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 03/11/2014] [Accepted: 04/08/2014] [Indexed: 12/11/2022] Open
Abstract
The non-receptor tyrosine kinase c-Src, hereafter referred to as Src, is overexpressed or activated in multiple human malignancies. There has been much speculation about the functional role of Src in colorectal cancer (CRC), with Src amplification and potential activating mutations in up to 20% of the human tumours, although this has never been addressed due to multiple redundant family members. Here, we have used the adult Drosophila and mouse intestinal epithelium as paradigms to define a role for Src during tissue homeostasis, damage-induced regeneration and hyperplasia. Through genetic gain and loss of function experiments, we demonstrate that Src is necessary and sufficient to drive intestinal stem cell (ISC) proliferation during tissue self-renewal, regeneration and tumourigenesis. Surprisingly, Src plays a non-redundant role in the mouse intestine, which cannot be substituted by the other family kinases Fyn and Yes. Mechanistically, we show that Src drives ISC proliferation through upregulation of EGFR and activation of Ras/MAPK and Stat3 signalling. Therefore, we demonstrate a novel essential role for Src in intestinal stem/progenitor cell proliferation and tumourigenesis initiation in vivo.
Collapse
Affiliation(s)
- Julia B Cordero
- The Beatson Institute for Cancer Research, Bearsden Glasgow, UK
| | | | | | - Colin Nixon
- The Beatson Institute for Cancer Research, Bearsden Glasgow, UK
| | - Margaret C Frame
- Edinburgh Cancer Research Centre Institute of Genetics & Molecular Medicine University of Edinburgh, Edinburgh, UK
| | - William J Muller
- Goodman Cancer Research Center McGill University, Montreal, QC, Canada
| | - Marcos Vidal
- The Beatson Institute for Cancer Research, Bearsden Glasgow, UK
| | - Owen J Sansom
- The Beatson Institute for Cancer Research, Bearsden Glasgow, UK
| |
Collapse
|
7
|
Zhang J, Ren R, Luo X, Fan P, Liu X, Liang S, Ma L, Yu P, Bai H. A small physiological electric field mediated responses of extravillous trophoblasts derived from HTR8/SVneo cells: involvement of activation of focal adhesion kinase signaling. PLoS One 2014; 9:e92252. [PMID: 24643246 PMCID: PMC3958492 DOI: 10.1371/journal.pone.0092252] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 02/18/2014] [Indexed: 11/18/2022] Open
Abstract
Moderate invasion of trophoblast cells into endometrium is essential for the placental development and normal pregnancy. Electric field (EF)-induced effects on cellular behaviors have been observed in many cell types. This study was to investigate the effect of physiological direct current EF (dc EF) on cellular responses such as elongation, orientation and motility of trophoblast cells. Immortalized first trimester extravillous trophoblast cells (HTR-8/SVneo) were exposed to the dc EF at physiological magnitude. Cell images were recorded and analyzed by image analyzer. Cell lysates were used to detect protein expression by Western blot. Cultured in the dc EFs the cells showed elongation, orientation and enhanced migration rate compared with non-EF stimulated cells at field strengths of 100 mV/mm to 200 mV/mm. EF exposure increased focal adhesion kinase (FAK) phosphorylation in a time-dependent manner and increased expression levels of MMP-2. Pharmacological inhibition of FAK impaired the EF-induced responses including motility and abrogated the elevation of MMP-2 expression. However, the expression levels of integrins like integrin α1, α5, αV and β1 were not affected by EF stimulation. Our results demonstrate the importance of FAK activation in migration/motility of trophobalst cells driven by EFs. In addition, it raises the feasibility of using applied EFs to promote placentation through effects on trophoblast cells.
Collapse
Affiliation(s)
- Juan Zhang
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Rongmei Ren
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Xuefeng Luo
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Ping Fan
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Xinghui Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Shanshan Liang
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Lei Ma
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Ping Yu
- Laboratory of Cell and Gene Therapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Huai Bai
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P. R. China
- * E-mail:
| |
Collapse
|
8
|
Macagno JP, Diaz Vera J, Yu Y, MacPherson I, Sandilands E, Palmer R, Norman JC, Frame M, Vidal M. FAK acts as a suppressor of RTK-MAP kinase signalling in Drosophila melanogaster epithelia and human cancer cells. PLoS Genet 2014; 10:e1004262. [PMID: 24676055 PMCID: PMC3967952 DOI: 10.1371/journal.pgen.1004262] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Accepted: 02/10/2014] [Indexed: 11/18/2022] Open
Abstract
Receptor Tyrosine Kinases (RTKs) and Focal Adhesion Kinase (FAK) regulate multiple signalling pathways, including mitogen-activated protein (MAP) kinase pathway. FAK interacts with several RTKs but little is known about how FAK regulates their downstream signalling. Here we investigated how FAK regulates signalling resulting from the overexpression of the RTKs RET and EGFR. FAK suppressed RTKs signalling in Drosophila melanogaster epithelia by impairing MAPK pathway. This regulation was also observed in MDA-MB-231 human breast cancer cells, suggesting it is a conserved phenomenon in humans. Mechanistically, FAK reduced receptor recycling into the plasma membrane, which resulted in lower MAPK activation. Conversely, increasing the membrane pool of the receptor increased MAPK pathway signalling. FAK is widely considered as a therapeutic target in cancer biology; however, it also has tumour suppressor properties in some contexts. Therefore, the FAK-mediated negative regulation of RTK/MAPK signalling described here may have potential implications in the designing of therapy strategies for RTK-driven tumours.
Collapse
Affiliation(s)
- Juan Pablo Macagno
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Jesica Diaz Vera
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Yachuan Yu
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Iain MacPherson
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Emma Sandilands
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | - Ruth Palmer
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Jim C. Norman
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Margaret Frame
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | - Marcos Vidal
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| |
Collapse
|
9
|
Golubovskaya VM. Targeting FAK in human cancer: from finding to first clinical trials. Front Biosci (Landmark Ed) 2014; 19:687-706. [PMID: 24389213 DOI: 10.2741/4236] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
It is twenty years since Focal Adhesion Kinase (FAK) was found to be overexpressed in many types of human cancer. FAK plays an important role in adhesion, spreading, motility, invasion, metastasis, survival, angiogenesis, and recently has been found to play an important role as well in epithelial to mesenchymal transition (EMT), cancer stem cells and tumor microenvironment. FAK has kinase-dependent and kinase independent scaffolding, cytoplasmic and nuclear functions. Several years ago FAK was proposed as a potential therapeutic target; the first clinical trials were just reported, and they supported further studies of FAK as a promising therapeutic target. This review discusses the main functions of FAK in cancer, and specifically focuses on recent novel findings on the role of FAK in cancer stem cells, microenvironment, epithelial-to-mesenchymal transition, invasion, metastasis, and also highlight new approaches of targeting FAK and critically discuss challenges that lie ahead for its targeted therapeutics. The review provides a summary of translational approaches of FAK-targeted and combination therapies and outline perspectives and future directions of FAK research.
Collapse
|
10
|
Fernandez-Gonzalez R, Zallen JA. Wounded cells drive rapid epidermal repair in the early Drosophila embryo. Mol Biol Cell 2013; 24:3227-37. [PMID: 23985320 PMCID: PMC3806660 DOI: 10.1091/mbc.e13-05-0228] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Epithelial tissues are protective barriers that display a remarkable ability to repair wounds. Wound repair is often associated with an accumulation of actin and nonmuscle myosin II around the wound, forming a purse string. The role of actomyosin networks in generating mechanical force during wound repair is not well understood. Here we investigate the mechanisms of force generation during wound repair in the epidermis of early and late Drosophila embryos. We find that wound closure is faster in early embryos, where, in addition to a purse string around the wound, actomyosin networks at the medial cortex of the wounded cells contribute to rapid wound repair. Laser ablation demonstrates that both medial and purse-string actomyosin networks generate contractile force. Quantitative analysis of protein localization dynamics during wound closure indicates that the rapid contraction of medial actomyosin structures during wound repair in early embryos involves disassembly of the actomyosin network. By contrast, actomyosin purse strings in late embryos contract more slowly in a mechanism that involves network condensation. We propose that the combined action of two force-generating structures--a medial actomyosin network and an actomyosin purse string--contributes to the increased efficiency of wound repair in the early embryo.
Collapse
Affiliation(s)
- Rodrigo Fernandez-Gonzalez
- Institute of Biomaterials and Biomedical Engineering and Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G9, Canada Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada Howard Hughes Medical Institute, Developmental Biology Program, Sloan-Kettering Institute, New York, NY 10065
| | | |
Collapse
|
11
|
Top3β is an RNA topoisomerase that works with fragile X syndrome protein to promote synapse formation. Nat Neurosci 2013; 16:1238-47. [PMID: 23912945 PMCID: PMC3853347 DOI: 10.1038/nn.3479] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 06/21/2013] [Indexed: 12/11/2022]
Abstract
Topoisomerases are crucial to solve DNA topological problems, but they have not been linked to RNA metabolism. Here we show that human topoisomerase 3β (Top3β) is an RNA topoisomerase that biochemically and genetically interacts with FMRP, a protein deficient in Fragile X syndrome and known to regulate translation of mRNAs important for neuronal function and autism. Notably, the FMRP-Top3β interaction is abolished by a disease-associated FMRP mutation, suggesting that Top3β may contribute to pathogenesis of mental disorders. Top3β binds multiple mRNAs encoded by genes with neuronal functions related to schizophrenia and autism. Expression of one such gene, ptk2/FAK, is reduced in neuromuscular junctions of Top3β mutant flies. Synapse formation is defective in Top3β mutant flies and mice, as observed in FMRP mutant animals. Our findings suggest that Top3β acts as an RNA topoisomerase and works with FMRP to promote expression of mRNAs critical for neurodevelopment and mental health.
Collapse
|
12
|
Nagaosa K, Okada R, Nonaka S, Takeuchi K, Fujita Y, Miyasaka T, Manaka J, Ando I, Nakanishi Y. Integrin βν-mediated phagocytosis of apoptotic cells in Drosophila embryos. J Biol Chem 2011; 286:25770-7. [PMID: 21592968 DOI: 10.1074/jbc.m110.204503] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To identify molecules that play roles in the clearance of apoptotic cells by Drosophila phagocytes, we examined a series of monoclonal antibodies raised against larval hemocytes for effects on phagocytosis in vitro. One antibody that inhibited phagocytosis recognized terribly reduced optic lobes (Trol), a core protein of the perlecan-type proteoglycan, and the level of phagocytosis in embryos of a Trol-lacking fly line was lower than in a control line. The treatment of a hemocyte cell line with a recombinant Trol protein containing the amino acid sequence RGD augmented the phosphorylation of focal adhesion kinase, a hallmark of integrin activation. A loss of integrin βν, one of the two β subunits of Drosophila integrin, brought about a reduction in the level of apoptotic cell clearance in embryos. The presence of integrin βν at the surface of embryonic hemocytes was confirmed, and forced expression of integrin βν in hemocytes of an integrin βν-lacking fly line recovered the defective phenotype of phagocytosis. Finally, the level of phagocytosis in a fly line that lacks both integrin βν and Draper, another receptor required for the phagocytosis of apoptotic cells, was lower than that in a fly line lacking either protein. We suggest that integrin βν serves as a phagocytosis receptor responsible for the clearance of apoptotic cells in Drosophila, independent of Draper.
Collapse
Affiliation(s)
- Kaz Nagaosa
- Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Zhang M, Wang H, Li D, Xu X. A novel focal adhesion kinase from Marsupenaeus japonicus and its response to WSSV infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2009; 33:533-539. [PMID: 19013481 DOI: 10.1016/j.dci.2008.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Revised: 10/17/2008] [Accepted: 10/18/2008] [Indexed: 05/27/2023]
Abstract
Focal adhesion kinase (FAK) is a cytoplasmic protein-tyrosine kinase involved in integrin-mediated signal transduction which regulates multiple cell functions in mammalian cells. In contrast to the well document of FAK in mammalian cells, the properties of FAK in crustacean have not been reported yet and even none of their gene or protein sequences is known to date. Here, we report for the first time the cloning of FAK from Marsupenaeus japonicus (designated as MjFAK) and the identification of its involvement in the virus infection and host defense. Sequence analysis displayed that MjFAK shared strong similarity to FAK family protein-tyrosine kinase, including conserved tyrosine phosphorylation sites, PTK domain and FAT domain. Immunofluorescence staining analysis showed that MjFAK was located prominently at the cell periphery and partly in cytoplasm and nucleus. Notably, considerable high content of MjFAK and MjFAK (pY399) were found in shrimp, which differs greatly from the low level of endogenous FAK and pFAK in the mammalian cells. It implies that pMjFAK may play a significant role in shrimp. Moreover, pMjFAK increased at the early infection stage, and the hemocyte adhesion activity of fibronectin also increased significantly accompanying with its phosphorylation. These results suggest that pMjFAK may not only promote the WSSV infection, but also participate in the defense mechanism via the enhancement of the immune-cell adhesion. Our data provide a clue to recognize the FAK-mediated signaling connection in the control of immunity and virus infection in crustaceans, which will be helpful to shrimp viral disease control.
Collapse
Affiliation(s)
- Mingchang Zhang
- School of Life Science, Xiamen University, Xiamen 361005, PR China
| | | | | | | |
Collapse
|
14
|
Tsai PI, Kao HH, Grabbe C, Lee YT, Ghose A, Lai TT, Peng KP, Van Vactor D, Palmer RH, Chen RH, Yeh SR, Chien CT. Fak56 functions downstream of integrin alphaPS3betanu and suppresses MAPK activation in neuromuscular junction growth. Neural Dev 2008; 3:26. [PMID: 18925939 PMCID: PMC2576229 DOI: 10.1186/1749-8104-3-26] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2008] [Accepted: 10/16/2008] [Indexed: 12/11/2022] Open
Abstract
Background Focal adhesion kinase (FAK) functions in cell migration and signaling through activation of the mitogen-activated protein kinase (MAPK) signaling cascade. Neuronal function of FAK has been suggested to control axonal branching; however, the underlying mechanism in this process is not clear. Results We have generated mutants for the Drosophila FAK gene, Fak56. Null Fak56 mutants display overgrowth of larval neuromuscular junctions (NMJs). Localization of phospho-FAK and rescue experiments suggest that Fak56 is required in presynapses to restrict NMJ growth. Genetic analyses imply that FAK mediates the signaling pathway of the integrin αPS3βν heterodimer and functions redundantly with Src. At NMJs, Fak56 downregulates ERK activity, as shown by diphospho-ERK accumulation in Fak56 mutants, and suppression of Fak56 mutant NMJ phenotypes by reducing ERK activity. Conclusion We conclude that Fak56 is required to restrict NMJ growth during NMJ development. Fak56 mediates an extracellular signal through the integrin receptor. Unlike its conventional role in activating MAPK/ERK, Fak56 suppresses ERK activation in this process. These results suggest that Fak56 mediates a specific neuronal signaling pathway distinct from that in other cellular processes.
Collapse
Affiliation(s)
- Pei-I Tsai
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Ueda A, Grabbe C, Lee J, Lee J, Palmer RH, Wu CF. Mutation of Drosophila focal adhesion kinase induces bang-sensitive behavior and disrupts glial function, axonal conduction and synaptic transmission. Eur J Neurosci 2008; 27:2860-70. [PMID: 18540882 DOI: 10.1111/j.1460-9568.2008.06252.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The role of the conserved focal adhesion kinase (FAK) family of protein tyrosine kinases in the development and physiological functions of the CNS has long been an area of interest among neuroscientists. In this report, we observe that Drosophila mutants lacking Fak56 exhibit a decreased lifespan, accompanied by a bang-sensitive phenotype, which is characterized by sensitivity to mechanical and high-frequency electrical stimulation. Fak56 mutant animals display lower thresholds and higher rates of seizures in response to electroconvulsive stimuli. Direct measurements of action potential conduction in larval segmental nerves demonstrate a slowed propagation speed and failure during high-frequency nerve stimulation. In addition, neuromuscular junctions in Fak56 mutant animals display transmission blockade during high-frequency activity as a result of action potential failure. Endogenous Fak56 protein is abundant in glial cells ensheathing the axon bundles, and structural alterations of segmental nerve bundles can be observed in mutants. Manipulation of Fak56 function specifically in glial cells also disrupts action potential conduction and neurotransmission, suggesting a glial component in the Fak56 bang-sensitive phenotype. Furthermore, we show that increased intracellular calcium levels result in the dephosphorylation of endogenous Fak56 protein in Drosophila cell lines, in parallel with our observations of highly variable synaptic potentials at a higher Ca2+ level in Fak56 mutant larvae. Together these findings suggest that modulation of Fak56 function is important for action potential propagation and Ca2+-regulated neuromuscular transmission in vivo.
Collapse
Affiliation(s)
- Atsushi Ueda
- Department of Biological Sciences, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | | | | | |
Collapse
|
16
|
Drosophila follicle cells: morphogenesis in an eggshell. Semin Cell Dev Biol 2008; 19:271-82. [PMID: 18304845 DOI: 10.1016/j.semcdb.2008.01.004] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2007] [Accepted: 01/16/2008] [Indexed: 01/15/2023]
Abstract
Epithelial morphogenesis is important for organogenesis and pivotal for carcinogenesis, but mechanisms that control it are poorly understood. The Drosophila follicular epithelium is a genetically tractable model to understand these mechanisms in vivo. This epithelium of follicle cells encases germline cells to create an egg. In this review, we summarize progress toward understanding mechanisms that maintain the epithelium or permit migrations essential for oogenesis. Cell-cell communication is important, but the same signals are used repeatedly to control distinct events. Understanding intrinsic mechanisms that alter responses to developmental signals will be important to understand regulation of cell shape and organization.
Collapse
|
17
|
Roles of focal adhesion kinase (FAK) in megakaryopoiesis and platelet function: studies using a megakaryocyte lineage specific FAK knockout. Blood 2007; 111:596-604. [PMID: 17925492 DOI: 10.1182/blood-2007-05-089680] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Focal adhesion kinase (FAK) plays a key role in mediating signaling downstream of integrins and growth factor receptors. In this study, we determined the roles of FAK in vivo by generating a megakaryocyte lineage-specific FAK-null mouse (Pf4-Cre/FAK-floxed). Megakaryocyte and platelet FAK expression was ablated in Pf4-Cre/FAK-floxed mice without affecting expression of the FAK homologue PYK2, although PYK2 phosphorylation was increased in FAK-/- megakaryocytes in response to fibrinogen. Megakaryopoiesis is greatly enhanced in Pf4-Cre/FAK-floxed mice, with significant increases in megakaryocytic progenitors (CFU-MK), mature megakaryocytes, megakaryocyte ploidy, and moderate increases in resting platelet number and platelet recovery following a thrombocytopenic stress. Thrombopoietin (Tpo)-mediated activation of Lyn kinase, a negative regulator of megakaryopoiesis, is severely attenuated in FAK-null megakaryocytes compared with wild-type controls. In contrast, Tpo-mediated activation of positive megakaryopoiesis regulators such as ERK1/2 and AKT is increased in FAK-null megakaryocytes, providing a plausible explanation for the observed increases in megakaryopoiesis in these mice. In Pf4-Cre/FAK-floxed mice, rebleeding times are significantly increased, and FAK-null platelets exhibit diminished spreading on immobilized fibrinogen. These studies establish clear roles for FAK in megakaryocyte growth and platelet function, setting the stage for manipulation of this component of the Tpo signaling apparatus for therapeutic benefit.
Collapse
|
18
|
Mamali I, Tatari MN, Micheva I, Lampropoulou M, Marmaras VJ. Apoptosis in medfly hemocytes is regulated during pupariation through FAK, Src, ERK, PI-3K p85a, and Akt survival signaling. J Cell Biochem 2007; 101:331-47. [PMID: 17177294 DOI: 10.1002/jcb.21175] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Focal adhesion kinase (FAK) and its downstream signaling targets are implicated in the process of apoptosis induced by external stimuli, in several mammalian systems. In this report, we demonstrate, that medfly (Ceratitis capitata) hemocytes do undergo apoptosis during larval development. In particular, we show using Western blot, ELISA and flow cytometry analysis, that FAK expression silencing in transfected by FAK double-stranded RNA (dsRNA) hemocytes, enhances twofold hemocyte apoptosis, by signaling through Src, MEK/ERK, and PI-3K/Akt signaling pathways. FAK expression silencing, in response to FAK dsRNA treatment, blocks partially the phosphorylation of its downstream targets. Pre-incubation of hemocytes, with specific inhibitors of FAK downstream signaling molecules, demonstrated that all these inhibitors reduced hemocyte viability and enhanced the magnitude of apoptosis about threefold. This data suggest that these pathways contribute to hemocyte survival and/or death during development. The expression and phosphorylation of FAK, Src, PI-3K p85a, Akt, and ERK signaling molecules appear to be dependent upon developmental stages. The expression and phosphorylation of the above signaling molecules, in annexin-positive and annexin-negative hemocytes is also distinct. The maximum expression and phosphorylation of FAK, Src, PI-3K p85a, Akt, and ERK appeared in annexin-positive hemocytes, in both early and late apoptotic hemocytes. The novel aspect of this report is based on the fact that hemocytes attempt to suppress apoptosis, by increasing the expression/phosphorylation of FAK and, hence its downstream targets signaling molecules Src, ERK, PI-3K p85a, and Akt. Evidently, the basic survival pathways among insects and mammals appear to remain unchanged, during evolution.
Collapse
Affiliation(s)
- Irene Mamali
- Department of Biology, University of Patras, Patras, Greece
| | | | | | | | | |
Collapse
|
19
|
Murakami S, Umetsu D, Maeyama Y, Sato M, Yoshida S, Tabata T. Focal adhesion kinase controls morphogenesis of the Drosophilaoptic stalk. Development 2007; 134:1539-48. [PMID: 17360775 DOI: 10.1242/dev.001529] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Photoreceptor cell axons (R axons) innervate optic ganglia in the Drosophila brain through the tubular optic stalk. This structure consists of surface glia (SG) and forms independently of R axon projection. In a screen for genes involved in optic stalk formation, we identified Fak56D encoding a Drosophila homolog of mammalian focal adhesion kinase (FAK). FAK is a main component of the focal adhesion signaling that regulates various cellular events, including cell migration and morphology. We show that Fak56D mutation causes severe disruption of the optic stalk structure. These phenotypes were completely rescued by Fak56D transgene expression in the SG cells but not in photoreceptor cells. Moreover, Fak56D genetically interacts with myospheroid, which encodes an integrin β subunit. In addition,we found that CdGAPr is also required for optic stalk formation and genetically interacts with Fak56D. CdGAPr encodes a GTPase-activating domain that is homologous to that of mammalian CdGAP, which functions in focal adhesion signaling. Hence the optic stalk is a simple monolayered structure that can serve as an ideal system for studying glial cell morphogenesis and the developmental role(s) of focal adhesion signaling.
Collapse
Affiliation(s)
- Satoshi Murakami
- Laboratory of Morphogenesis, Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-Ku, Tokyo 113-0032, Japan
| | | | | | | | | | | |
Collapse
|
20
|
Lamprou I, Mamali I, Dallas K, Fertakis V, Lampropoulou M, Marmaras VJ. Distinct signalling pathways promote phagocytosis of bacteria, latex beads and lipopolysaccharide in medfly haemocytes. Immunology 2007; 121:314-27. [PMID: 17376199 PMCID: PMC2265961 DOI: 10.1111/j.1365-2567.2007.02576.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
In insects, phagocytosis is an important innate immune response against pathogens and parasites, and several signal transduction pathways regulate this process. The focal adhesion kinase (FAK)/Src and mitogen activated protein kinase (MAPK) pathways are of central importance because their activation upon pathogen challenge regulates phagocytosis via haemocyte secretion and activation of the prophenoloxidase (proPO) cascade. The goal of this study was to explore further the mechanisms underlying the process of phagocytosis. In particular, in this report, we used flow cytometry, RNA interference, enzyme-linked immunosorbent assay, Western blot and immunoprecipitation analysis to demonstrate that (1) phagocytosis of bacteria (both Gram-negative and Gram-positive) is dependent on RGD-binding receptors, FAK/Src and MAPKs, (2) latex bead phagocytosis is RGD-binding-receptor-independent and dependent on FAK/Src and MAPKs, (3) lipopolysaccharide internalization is RGD-binding-receptor-independent and FAK/Src-independent but MAPK-dependent and (4) in unchallenged haemocytes in suspension, FAK, Src and extracellular signal-regulated kinase (ERK) signalling molecules participating in phagocytosis show both a functional and a physical association. Overall, this study has furthered knowledge of FAK/Src and MAPK signalling pathways in insect haemocyte immunity and has demonstrated that distinct signalling pathways regulate the phagocytic activity of biotic and abiotic components in insect haemocytes. Evidently, the basic phagocytic signalling pathways among insects and mammals appear to have remained unchanged during evolution.
Collapse
Affiliation(s)
- Irene Lamprou
- Department of Biology, University of Patras, Patras, Greece
| | | | | | | | | | | |
Collapse
|
21
|
Plows LD, Cook RT, Davies AJ, Walker AJ. Integrin engagement modulates the phosphorylation of focal adhesion kinase, phagocytosis, and cell spreading in molluscan defence cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2006; 1763:779-86. [PMID: 16766054 DOI: 10.1016/j.bbamcr.2006.04.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2005] [Revised: 04/07/2006] [Accepted: 04/07/2006] [Indexed: 12/21/2022]
Abstract
Integrins play a key role in cellular immune responses in a variety of organisms; however, knowledge of integrins and their effects on cell signalling and functional responses in molluscan defence reactions is poor. Using integrin-mediated cell adhesion kits, alphaVbeta3 and beta1 integrin-like subunits were identified on the surface of Lymnaea stagnalis haemocytes. Haemocyte binding via these integrins was found to be dependent on Ca2+/Mg2+. Western blotting with an anti-phospho (anti-active) focal adhesion kinase (FAK) antibody revealed a 120-125 kDa FAK-like protein in these cells; this protein was transiently phosphorylated upon haemocyte adhesion over 90 min, with maximal phosphorylation occurring after 30 min binding. Also, integrin engagement with the tetrapeptide Arg-Gly-Asp-Ser (RGDS) resulted in a rapid increase in phosphorylation of the FAK-like protein; however, RGDS did not affect the phosphorylation of extracellular signal-regulated kinase. Treatment of haemocytes with RGDS (2 mM) inhibited phagocytosis of E. coli bioparticles by 88%. Moreover, at this concentration, RGDS reduced cell spreading by 61%; stress fiber formation was also impaired. Taken together, these results demonstrate a role for integrins in L. stagnalis haemocyte adhesion and defence reactions and, for the first time, link integrin engagement to FAK activation in molluscs.
Collapse
Affiliation(s)
- Louise D Plows
- School of Life Sciences, Kingston University, Penrhyn Road, Kingston-upon-Thames, Surrey, KT1 2EE, UK
| | | | | | | |
Collapse
|
22
|
Corsi JM, Rouer E, Girault JA, Enslen H. Organization and post-transcriptional processing of focal adhesion kinase gene. BMC Genomics 2006; 7:198. [PMID: 16889663 PMCID: PMC1570463 DOI: 10.1186/1471-2164-7-198] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2006] [Accepted: 08/04/2006] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase critical for processes ranging from embryo development to cancer progression. Although isoforms with specific molecular and functional properties have been characterized in rodents and chicken, the organization of FAK gene throughout phylogeny and its potential to generate multiple isoforms are not well understood. Here, we study the phylogeny of FAK, the organization of its gene, and its post-transcriptional processing in rodents and human. RESULTS A single orthologue of FAK and the related PYK2 was found in non-vertebrate species. Gene duplication probably occurred in deuterostomes after the echinoderma embranchment, leading to the evolution of PYK2 with distinct properties. The amino acid sequence of FAK and PYK2 is conserved in their functional domains but not in their linker regions, with the absence of autophosphorylation site in C. elegans. Comparison of mouse and human FAK genes revealed the existence of multiple combinations of conserved and non-conserved 5'-untranslated exons in FAK transcripts suggesting a complex regulation of their expression. Four alternatively spliced coding exons (13, 14, 16, and 31), previously described in rodents, are highly conserved in vertebrates. Cis-regulatory elements known to regulate alternative splicing were found in conserved alternative exons of FAK or in the flanking introns. In contrast, other reported human variant exons were restricted to Homo sapiens, and, in some cases, other primates. Several of these non-conserved exons may correspond to transposable elements. The inclusion of conserved alternative exons was examined by RT-PCR in mouse and human brain during development. Inclusion of exons 14 and 16 peaked at the end of embryonic life, whereas inclusion of exon 13 increased steadily until adulthood. Study of various tissues showed that inclusion of these exons also occurred, independently from each other, in a tissue-specific fashion. CONCLUSION The alternative coding exons 13, 14, 16, and 31 are highly conserved in vertebrates and their inclusion in mRNA is tightly but independently regulated. These exons may therefore be crucial for FAK function in specific tissues or during development. Conversely pathological disturbance of the expression of FAK and of its isoforms could lead to abnormal cellular regulation.
Collapse
Affiliation(s)
- Jean-Marc Corsi
- Unité Mixte de Recherche-Santé (UMR-S) 536, Institut National de la Santé et de la Recherche Médicale (INSERM) F-75005, Paris, France; Université Pierre et Marie Curie-Paris 6, F-75005, Paris, France; Institut du Fer-à-Moulin, F-75005, Paris, France
| | - Evelyne Rouer
- Unité Mixte de Recherche-Santé (UMR-S) 536, Institut National de la Santé et de la Recherche Médicale (INSERM) F-75005, Paris, France; Université Pierre et Marie Curie-Paris 6, F-75005, Paris, France; Institut du Fer-à-Moulin, F-75005, Paris, France
| | - Jean-Antoine Girault
- Unité Mixte de Recherche-Santé (UMR-S) 536, Institut National de la Santé et de la Recherche Médicale (INSERM) F-75005, Paris, France; Université Pierre et Marie Curie-Paris 6, F-75005, Paris, France; Institut du Fer-à-Moulin, F-75005, Paris, France
| | - Hervé Enslen
- Unité Mixte de Recherche-Santé (UMR-S) 536, Institut National de la Santé et de la Recherche Médicale (INSERM) F-75005, Paris, France; Université Pierre et Marie Curie-Paris 6, F-75005, Paris, France; Institut du Fer-à-Moulin, F-75005, Paris, France
| |
Collapse
|
23
|
Zalewska T, Makarewicz D, Janik B, Ziemka-Nałecz M. Neonatal cerebral hypoxia-ischemia: involvement of FAK-dependent pathway. Int J Dev Neurosci 2005; 23:657-62. [PMID: 16095866 DOI: 10.1016/j.ijdevneu.2005.05.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2005] [Revised: 05/19/2005] [Accepted: 05/24/2005] [Indexed: 01/24/2023] Open
Abstract
Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase thought to play a major role in transducing extracellular matrix (ECM)-derived survival signals into cells. Thus, modulation of FAK activity may affect the linkage between ECM and signaling cascade to which it is connected and may participate in a variety of pathological settings. In the present study, we investigated the effect of neonatal cerebral hypoxia-ischemia (HI) on levels and tyrosine phosphorylation of focal adhesion kinase and the interaction of this enzyme with Src protein tyrosine kinase and adapter protein p130Cas, involved in FAK-mediated signaling pathway. The total amount of focal adhesion kinase as well as its phosphorylated form declined substantially to about 50% of the control between 24 and 48 h after the insult. Concomitantly a decreased association of FAK with its investigated molecular partners, Src kinase and p130Cas protein has been observed. This early response to brain hypoxia-ischemia was attenuated during prolonged recovery with almost complete return to control values at 7 days. These data are indicative of an involvement of FAK-dependent signaling pathway in the evolution of HI-induced neuronal degeneration.
Collapse
Affiliation(s)
- Teresa Zalewska
- NeuroRepair Department, Medical Research Institute, Polish Academy of Sciences, 5 Pawinskiego Str., 02-106 Warsaw, Poland.
| | | | | | | |
Collapse
|
24
|
Chen GC, Turano B, Ruest PJ, Hagel M, Settleman J, Thomas SM. Regulation of Rho and Rac signaling to the actin cytoskeleton by paxillin during Drosophila development. Mol Cell Biol 2005; 25:979-87. [PMID: 15657426 PMCID: PMC544021 DOI: 10.1128/mcb.25.3.979-987.2005] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Paxillin is a prominent focal adhesion docking protein that regulates cell adhesion and migration. Although numerous paxillin-binding proteins have been identified and paxillin is required for normal embryogenesis, the precise mechanism by which paxillin functions in vivo has not yet been determined. We identified an ortholog of mammalian paxillin in Drosophila (Dpax) and have undertaken a genetic analysis of paxillin function during development. Overexpression of Dpax disrupted leg and wing development, suggesting a role for paxillin in imaginal disc morphogenesis. These defects may reflect a function for paxillin in regulation of Rho family GTPase signaling as paxillin interacts genetically with Rac and Rho in the developing eye. Moreover, a gain-of-function suppressor screen identified a genetic interaction between Dpax and cdi in wing development. cdi belongs to the cofilin kinase family, which includes the downstream Rho target, LIM kinase (LIMK). Significantly, strong genetic interactions were detected between Dpax and Dlimk, as well as downstream effectors of Dlimk. Supporting these genetic data, biochemical studies indicate that paxillin regulates Rac and Rho activity, positively regulating Rac and negatively regulating Rho. Taken together, these data indicate the importance of paxillin modulation of Rho family GTPases during development and identify the LIMK pathway as a critical target of paxillin-mediated Rho regulation.
Collapse
Affiliation(s)
- Guang-Chao Chen
- Massachusetts General Hospital Cancer Center and Harvard Medical School, 149 13th Street, Charlestown, MA 02129, USA
| | | | | | | | | | | |
Collapse
|
25
|
Grabbe C, Zervas CG, Hunter T, Brown NH, Palmer RH. Focal adhesion kinase is not required for integrin function or viability inDrosophila. Development 2004; 131:5795-805. [PMID: 15525665 DOI: 10.1242/dev.01462] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The mammalian focal adhesion kinase (FAK) family of non-receptor protein-tyrosine kinases has been implicated in controlling a multitude of cellular responses to the engagement of cell-surface integrins and G-protein-coupled receptors. The high level of sequence conservation between the mammalian proteins and the Drosophila homologue of FAK, Fak56,suggested that it would have similar functions. However, we show here that Drosophila Fak56 is not essential for integrin functions in adhesion,migration or signaling in vivo. Furthermore, animals lacking Fak56 are viable and fertile, demonstrating that Fak56 is not essential for other developmental or physiological functions. Despite this, overexpressed Fak56 is a potent inhibitor of integrins binding to the extracellular matrix, suggesting that Fak56 may play a subtle role in the negative regulation of integrin adhesion.
Collapse
Affiliation(s)
- Caroline Grabbe
- Umeå Center for Molecular Pathogenesis, Building 6L, Umeå University, Umeå, 901 87, Sweden
| | | | | | | | | |
Collapse
|
26
|
García MG, Toney SJ, Hille MB. Focal adhesion kinase (FAK) expression and phosphorylation in sea urchin embryos. Gene Expr Patterns 2004; 4:223-34. [PMID: 15161103 DOI: 10.1016/j.modgep.2003.08.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2003] [Revised: 08/13/2003] [Accepted: 08/19/2003] [Indexed: 11/20/2022]
Abstract
We have cloned three cDNA isoforms of focal adhesion kinase (FAK) from the sea urchin, Lytechinus variegatus. The sea urchin FAK is more closely related to FAK from other deuterostomes than from invertebrate protostomes or to cell adhesion kinase beta (CAKbeta/Pyk2/FAK2). FAK is expressed in all cells of sea urchin embryos by the 120-cell stage and strongly in blastulae. Phospho-FAK concentrates on basal surfaces of epithelial cells in early blastulae and occurs in syncytial cables of primary mesenchyme cells (PMC). Inhibition of FAK by constructs of FAK-related non-kinase delays blastocoel expansion and early PMC ingression. These results suggest that FAK has roles in cell adhesion and in the shape and integrity of the epithelial cells in sea urchin embryos.
Collapse
Affiliation(s)
- María Guadalupe García
- Department of Biology, University of Washington, Box 351800, Seattle, WA 98195-1800, USA
| | | | | |
Collapse
|
27
|
Schlaepfer DD, Mitra SK, Ilic D. Control of motile and invasive cell phenotypes by focal adhesion kinase. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2004; 1692:77-102. [PMID: 15246681 DOI: 10.1016/j.bbamcr.2004.04.008] [Citation(s) in RCA: 350] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2004] [Accepted: 04/08/2004] [Indexed: 01/09/2023]
Abstract
Cell motility is stimulated by extracellular stimuli and initiated by intracellular signaling proteins that localize to sites of cell contact with the extracellular matrix termed focal contacts. Focal adhesion kinase (FAK) is an intracellular protein-tyrosine kinase (PTK) that acts to regulate the cycle of focal contact formation and disassembly required for efficient cell movement. FAK is activated by a variety of cell surface receptors and transmits signals to a range of targets. Thus, FAK acts as an integrator of cell motility-associated signaling events. We will review the stimulatory and regulatory mechanisms of FAK activation, the different signaling connections of FAK that are mediated by a growing number of FAK-interacting proteins, and the modulation of FAK function by tyrosine and serine phosphorylation. We will also summarize findings with regard to FAK function in vertebrate and invertebrate development as well as recent insights into the mechanistic role(s) of FAK in promoting cell migration. As increased FAK expression and tyrosine phosphorylation have been correlated with the progression to an invasive cell phenotype, there is growing interest in elucidating the important FAK-related signaling connections promoting invasive tumor cell movement. To this end, we will discuss the effects of FAK inhibition via the dominant-negative expression of the FAK C-terminal domain termed FAK-related non-kinase (FRNK) and how these studies have uncovered a distinct role for FAK in promoting cell invasion that may differ from its role in promoting cell motility.
Collapse
Affiliation(s)
- David D Schlaepfer
- Department of Immunology, Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | | | |
Collapse
|
28
|
Abstract
The protein tyrosine kinase focal adhesion kinase (FAK) plays a prominent role in integrin signaling. FAK activation, demonstrated by an increase in phosphorylation of Tyr397 as well as other sites in the protein, is best understood in the context of the engagement of integrins at the cell surface. Activation of FAK results in recruitment of a number of SH2-domain- and SH3-domain-containing proteins, which mediate signaling to several downstream pathways. FAK-dependent activation of these pathways has been implicated in a diverse array of cellular processes, including cell migration, growth factor signaling, cell cycle progression and cell survival.
Collapse
Affiliation(s)
- J Thomas Parsons
- Department of Microbiology, University of Virginia Health System, Charlottesville, VA 22908, USA.
| |
Collapse
|
29
|
Santoni G, Lucciarini R, Amantini C, Jacobelli J, Spreghini E, Ballarini P, Piccoli M, Gismondi A. Candida albicans expresses a focal adhesion kinase-like protein that undergoes increased tyrosine phosphorylation upon yeast cell adhesion to vitronectin and the EA.hy 926 human endothelial cell line. Infect Immun 2002; 70:3804-15. [PMID: 12065524 PMCID: PMC128043 DOI: 10.1128/iai.70.7.3804-3815.2002] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The signaling pathways triggered by adherence of Candida albicans to the host cells or extracellular matrix are poorly understood. We provide here evidence in C. albicans yeasts of a p105 focal adhesion kinase (Fak)-like protein (that we termed CaFak), antigenically related to the vertebrate p125Fak, and its involvement in integrin-like-mediated fungus adhesion to vitronectin (VN) and EA.hy 926 human endothelial cell line. Biochemical analysis with different anti-chicken Fak antibodies identified CaFak as a 105-kDa protein and immunofluorescence and cytofluorimetric analysis on permeabilized cells specifically stain C. albicans yeasts; moreover, confocal microscopy evidences CaFak as a cytosolic protein that colocalizes on the membrane with the integrin-like VN receptors upon yeast adhesion to VN. The protein tyrosine kinase (PTK) inhibitors genistein and herbimycin A strongly inhibited C. albicans yeast adhesion to VN and EA.hy 926 endothelial cells. Moreover, engagement of alpha v beta 3 and alpha v beta 5 integrin-like on C. albicans either by specific monoclonal antibodies or upon adhesion to VN or EA.hy 926 endothelial cells stimulates CaFak tyrosine phosphorylation that is blocked by PTK inhibitor. A role for CaFak in C. albicans yeast adhesion was also supported by the failure of VN to stimulate its tyrosine phosphorylation in a C. albicans mutant showing normal levels of CaFak and VNR-like integrins but displaying reduced adhesiveness to VN and EA.hy 926 endothelial cells. Our results suggest that C. albicans Fak-like protein is involved in the control of yeast cell adhesion to VN and endothelial cells.
Collapse
Affiliation(s)
- Giorgio Santoni
- Department of Pharmacological Sciences and Experimental Medicine, University of Camerino, 62032 Camerino, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Silver DL, Montell DJ. Paracrine signaling through the JAK/STAT pathway activates invasive behavior of ovarian epithelial cells in Drosophila. Cell 2001; 107:831-41. [PMID: 11779460 DOI: 10.1016/s0092-8674(01)00607-9] [Citation(s) in RCA: 243] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The JAK/STAT signaling pathway, renowned for its effects on cell proliferation and survival, is constitutively active in various human cancers, including ovarian. We have found that JAK and STAT are required to convert the border cells in the Drosophila ovary from stationary, epithelial cells to migratory, invasive cells. The ligand for this pathway, Unpaired (UPD), is expressed by two central cells within the migratory cell cluster. Mutations in upd or jak cause defects in migration and a reduction in the number of cells recruited to the cluster. Ectopic expression of either UPD or JAK is sufficient to induce extra epithelial cells to migrate. Thus, a localized signal activates the JAK/STAT pathway in neighboring epithelial cells, causing them to become invasive.
Collapse
Affiliation(s)
- D L Silver
- Department of Biological Chemistry, Johns Hopkins School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | | |
Collapse
|
31
|
Abstract
Paxillin is a focal adhesion-associated, phosphotyrosine-containing protein that may play a role in several signaling pathways. Paxillin contains a number of motifs that mediate protein-protein interactions, including LD motifs, LIM domains, an SH3 domain-binding site and SH2 domain-binding sites. These motifs serve as docking sites for cytoskeletal proteins, tyrosine kinases, serine/threonine kinases, GTPase activating proteins and other adaptor proteins that recruit additional enzymes into complex with paxillin. Thus paxillin itself serves as a docking protein to recruit signaling molecules to a specific cellular compartment, the focal adhesions, and/or to recruit specific combinations of signaling molecules into a complex to coordinate downstream signaling. The biological function of paxillin coordinated signaling is likely to regulate cell spreading and motility.
Collapse
Affiliation(s)
- M D Schaller
- Department of Cell and Developmental Biology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, NC 27599, USA.
| |
Collapse
|
32
|
Humphries JE, Elizondo L, Yoshino TP. Protein kinase C regulation of cell spreading in the molluscan Biomphalaria glabrata embryonic (Bge) cell line. BIOCHIMICA ET BIOPHYSICA ACTA 2001; 1540:243-52. [PMID: 11583819 DOI: 10.1016/s0167-4889(01)00136-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Cellular adhesion and spreading are critical components involved in the processes of cell and tissue development, and immune responses in molluscs, but at present, little is known regarding the signaling pathways involved in these basic cellular functions. In the present study, the molluscan Biomphalaria glabrata embryonic (Bge) cell line was used as an in vitro model to study the signal transduction pathways regulating molluscan cell adhesion and spreading behavior. Western blot analysis using antibodies specific to mitogen-activated protein kinase (MAPK) revealed the presence of an MAPK-like immunoreactive protein in Bge cells, that was phosphorylated upon exposure to phorbol myristate acetate (PMA). Moreover, Bge cell treatment with inhibitors of protein kinase C (PKC), Ras and MAPK kinase (Mek) suppressed PMA-induced expression of activated MAPK, suggesting that PKC-, Ras- and Mek-like molecules may be acting upstream of MAPK. Similarly, in vitro Bge cell-spreading assays were performed in conjunction with the same panel of inhibitors to determine the potential involvement of PKC, Ras and Mek in cellular adhesion/spreading. Results revealed a similar pattern of inhibition of cell-spreading behavior strongly implying that the Bge cell spreading also may be regulated through a MAPK-associated signal transduction pathway(s) involving proteins similar to PKC, Ras and Mek.
Collapse
Affiliation(s)
- J E Humphries
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin - Madison, 53706, USA.
| | | | | |
Collapse
|
33
|
Schaller MD. Biochemical signals and biological responses elicited by the focal adhesion kinase. BIOCHIMICA ET BIOPHYSICA ACTA 2001; 1540:1-21. [PMID: 11476890 DOI: 10.1016/s0167-4889(01)00123-9] [Citation(s) in RCA: 419] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The focal adhesion kinase, FAK, is an important component of an integrin-dependent signaling pathway, which functions to transmit signals from the extracellular matrix into the cytoplasm. FAK is an essential gene product, since the fak-/- mouse exhibits embryonic lethality. A number of important biological processes, including cell motility and cell survival, are controlled by integrin-dependent signals and FAK has been implicated in regulating these processes. This review will focus upon recent findings providing insight into the mechanisms by which FAK transmits biochemical signals and elicits biological effects.
Collapse
Affiliation(s)
- M D Schaller
- Department of Cell and Developmental Biology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill 27599, USA.
| |
Collapse
|
34
|
Ilić D, Genbacev O, Jin F, Caceres E, Almeida EA, Bellingard-Dubouchaud V, Schaefer EM, Damsky CH, Fisher SJ. Plasma membrane-associated pY397FAK is a marker of cytotrophoblast invasion in vivo and in vitro. THE AMERICAN JOURNAL OF PATHOLOGY 2001; 159:93-108. [PMID: 11438458 PMCID: PMC1850415 DOI: 10.1016/s0002-9440(10)61677-x] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
During human pregnancy specialized placental cells of fetal origin, termed cytotrophoblasts, invade the uterus and its blood vessels. This tumor-like process anchors the conceptus to the mother and diverts the flow of uterine blood to the placenta. Previously, we showed that the expression of molecules with important functional roles, including a number of extracellular matrix integrin receptors, is precisely modulated during cytotrophoblast invasion in situ. Here we exploited this observation to study the role of the focal adhesion kinase (FAK), which transduces signals from the extracellular matrix and recruits additional signaling proteins to focal adhesions. Immunolocalization studies on tissue sections showed that FAK is expressed by cytotrophoblasts in all stages of differentiation. Because extracellular matrix-induced integrin clustering results in FAK (auto)phosphorylation on tyrosine 397 (Y397FAK), we also localized this form of the molecule. Immunolocalization experiments detected Y397FAK in a subset of cytotrophoblasts near the surface of the uterine wall. To assess the functional relevance of this observation, we used an adenovirus strategy to inhibit cytotrophoblast expression of FAK as the cells differentiated along the invasive pathway in vitro. Compared to control cells transduced with a wild-type virus, cytotrophoblasts that expressed antisense FAK exhibited a striking reduction in their ability to invade an extracellular matrix substrate. When cytotrophoblast differentiation was compromised (hypoxia in vitro, preeclampsia in vivo), Y397FAK levels associated with the plasma membrane were strikingly lower, although total FAK levels did not change. Together our results suggest that (auto)phosphorylation of Y397 on FAK is a critical component of the signaling pathway that mediates cytotrophoblast migration/invasion.
Collapse
Affiliation(s)
- D Ilić
- Department of Stomatology, University of California San Francisco, 94143-0512, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
The invasiveness of cancer cells resembles the normal behavior of cells that migrate into surrounding tissues during development. For example, the border cells in the Drosophila ovary undergo a partial epithelial to mesenchymal transition and invade the neighboring cluster of germline cells, migrating to the oocyte border. Once there, they provide patterning information to the oocyte and produce an eggshell specialization known as the micropyle. Border cell migration has been subjected to extensive genetic analyses using a variety of screening approaches. Recent findings demonstrate that conversion of the border cells from a stationary group of epithelial cells to invasive cells requires integration of the activities of at least two transcriptional regulatory pathways. One such pathway requires the slbo gene, which encodes Drosophila C/EBP, a basic region/leucine zipper transcriptional activator that is required for elevated expression of a number of downstream targets, including DE-cadherin and focal adhesion kinase (FAK). An independent pathway requires the activity of the ecdysone receptor and a recently identified co-activator for the ecdysone receptor known as Taiman (abbreviated TAI, pronounced ti-maan', meaning too slow). Ecdysone is produced in the Drosophila ovary in response to adequate nutrition and is required for progression of oogenesis through stage 9, when border cell migration occurs. Border cells mutant for tai accumulate abnormally high levels of adhesion complexes at their surfaces, which may account for their inability to migrate. Thus border cell migration requires a differentiation program mediated by the C/EBP pathway, which is required for elevated expression of a number of proteins required for motility. In addition, migration requires a hormonal signal that relays information regarding nutritional status and appears to be required for regulation of the proper localization of some of the C/EBP targets. These findings suggest that steroid hormones can regulate cell motility relatively directly, independent of the effects on proliferation. This may contribute to the metastatic effects of steroid hormones on certain cancers and the inhibition of metastasis by steroid hormone antagonists such as tamoxifen.
Collapse
Affiliation(s)
- D J Montell
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, 725 North Wolfe Street, 21205-2185, Baltimore, MD, USA.
| |
Collapse
|
36
|
Metheniti A, Paraskevopoulou N, Lambropoulou M, Marmaras VJ. Involvement of FAK/Src complex in the processes of Escherichia coli phagocytosis by insect hemocytes. FEBS Lett 2001; 496:55-9. [PMID: 11343706 DOI: 10.1016/s0014-5793(01)02405-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Recently we demonstrated that lipopolysaccharide promotes activation of the Ras/mitogen-activated protein cascade in hemocytes and that phagocytosis of Escherichia coli by insect hemocytes is mediated by an integrin-dependent process [Foukas et al. (1998) J. Biol. Chem. 273, 14813--14818]. Here we report data concerning the focal adhesion kinase (FAK) tyrosine phosphorylation status in hemocytes in response to E. coli. We demonstrate that E. coli-triggering stimulates a significant increase in tyrosine phosphorylation of FAK in hemocytes. Furthermore, immunoblotting analysis using anti-Y397 demonstrated intense FAK activity at the Y397/SH2-binding site in hemocytes treated with E. coli. In addition, antibody-mediated inhibition of FAK and Src-kinase has been shown to abolish FAK phosphorylation and E. coli phagocytosis, indicating a specific role for the FAK/Src complex in the processes of promoting cell phagocytosis. These findings expand the known signaling functions of FAK and provide insight into signal transduction events associated with hemocyte phagocytosis in response to E. coli.
Collapse
Affiliation(s)
- A Metheniti
- Department of Biology, University of Patras, Patras 265 00, Greece
| | | | | | | |
Collapse
|
37
|
Slack JK, Adams RB, Rovin JD, Bissonette EA, Stoker CE, Parsons JT. Alterations in the focal adhesion kinase/Src signal transduction pathway correlate with increased migratory capacity of prostate carcinoma cells. Oncogene 2001; 20:1152-63. [PMID: 11313859 DOI: 10.1038/sj.onc.1204208] [Citation(s) in RCA: 166] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2000] [Revised: 12/28/2000] [Accepted: 01/03/2001] [Indexed: 01/29/2023]
Abstract
Focal adhesion kinase (FAK) has been implicated in the regulation of cell migration. In addition, FAK expression is increased in a number of highly metastatic tumor cell lines. Therefore, we investigated the role of FAK in regulating migration of prostate carcinoma cell lines with increasing metastatic potential. We show that highly tumorigenic PC3 and DU145 cells exhibit intrinsic migratory capacity, while poorly tumorigenic LNCaP cells require a stimulus to migrate. Increased metastatic potential of PC3 and DU145 cells correlates with increased FAK expression, overall tyrosine phosphorylation and activity, as measured by autophosphorylation of tyrosine 397. However, in PC3 and DU145 cells, FAK autophosphorylation is adhesion dependent whereas a second site of tyrosine phosphorylation, tyrosine 861, a Src specific site, is uncoupled from adhesion-dependent signaling events. Finally, inhibiting the FAK/Src signal transduction pathway by over expressing FRNK (Focal adhesion kinase-Related Non-Kinase), an inhibitor of FAK activation, or treatment with PP2, a Src family kinase inhibitor, significantly inhibited migration of prostate carcinoma cell lines, demonstrating that tumor cell migration continues to be dependent on signals emanating from this pathway.
Collapse
Affiliation(s)
- J K Slack
- Department of Microbiology, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | | | | |
Collapse
|
38
|
Wheeler GN, Hynes RO. The cloning, genomic organization and expression of the focal contact protein paxillin in Drosophila. Gene 2001; 262:291-9. [PMID: 11179695 DOI: 10.1016/s0378-1119(00)00512-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Paxillin is a focal adhesion scaffolding protein, which has been proposed to play a role in focal adhesion dynamics. We have isolated a cDNA clone of the Drosophila homologue of paxillin. Comparison of the Drosophila paxillin sequence with those of vertebrate paxillins shows strong conservation of the LIM domains and LD repeats. Using the Drosophila genomic sequence we have identified two partial curated transcripts and deduced the structure of the paxillin gene. No homologues of other members of the paxillin family such as HIC-5 or leupaxin are to be found in the Drosophila genome. Surprisingly paxillin mRNA is expressed in a restricted pattern during embryogenesis. In particular it is strongly expressed in cells and tissues undergoing cell shape changes or cell migration. Many of the sites of expression are also known to be sites of integrin function or FAK expression. The data support a role for paxillin as an adapter and/or signaling protein during developmental processes involving integrin-mediated adhesion.
Collapse
Affiliation(s)
- G N Wheeler
- Howard Hughes Medical Institute and Center for Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | |
Collapse
|
39
|
Bai J, Uehara Y, Montell DJ. Regulation of invasive cell behavior by taiman, a Drosophila protein related to AIB1, a steroid receptor coactivator amplified in breast cancer. Cell 2000; 103:1047-58. [PMID: 11163181 DOI: 10.1016/s0092-8674(00)00208-7] [Citation(s) in RCA: 223] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Steroid hormones are key regulators of numerous physiological and developmental processes, including metastasis of breast and ovarian cancer. Here we report the identification of a Drosophila gene, named taiman, which encodes a steroid hormone receptor coactivator related to AIB1. Mutations in tai caused defects in the migration of specific follicle cells, the border cells, in the Drosophila ovary. Mutant cells exhibited abnormal accumulation of E-cadherin, beta-catenin, and focal adhesion kinase. TAI protein colocalized with the ecdysone receptor in vivo and augmented transcriptional activation by the ecdysone receptor in cultured cells. The finding of this type of coactivator required for cell motility suggests a novel role for steroid hormones, in stimulating invasive cell behavior, independent of effects on proliferation.
Collapse
Affiliation(s)
- J Bai
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
40
|
Abstract
Members of the integrin family of cell adhesion receptors influence several important aspects of cancer cell behavior, including motility and invasiveness, cell growth, and cell survival. Engagement of integrins with extracellular matrix (ECM) proteins can activate members of the Rho-family of small GTPases; conversely, Rho- and Ras-family proteins can influence the ability of integrins to bind their ligands. These events impinge on the control of cell motility, and ultimately on invasive and metastatic behavior. Integrin engagement with ECM also has important effects on cell survival, particularly for cells of epithelial origin. In some cases, specific integrins have selective effects on the efficiency of signal transduction in cell survival pathways.
Collapse
Affiliation(s)
- L V Parise
- Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | |
Collapse
|
41
|
Parsons JT, Martin KH, Slack JK, Taylor JM, Weed SA. Focal adhesion kinase: a regulator of focal adhesion dynamics and cell movement. Oncogene 2000; 19:5606-13. [PMID: 11114741 DOI: 10.1038/sj.onc.1203877] [Citation(s) in RCA: 507] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Engagement of integrin receptors with extracellular ligands gives rise to the formation of complex multiprotein structures that link the ECM to the cytoplasmic actin cytoskeleton. These adhesive complexes are dynamic, often heterogeneous structures, varying in size and organization. In motile cells, sites of adhesion within filopodia and lamellipodia are relatively small and transient and are referred to as 'focal complexes,' whereas adhesions underlying the body of the cell and localized to the ends of actin stress fibers are referred to as 'focal adhesions'. Signal transduction through focal complexes and focal adhesions has been implicated in the regulation of a number of key cellular processes, including growth factor induced mitogenic signals, cell survival and cell locomotion. The formation and remodeling of focal contacts is a dynamic process under the regulation of protein tyrosine kinases and small GTPases of the Rho family. In this review, we consider the role of the focal complex associated protein tyrosine kinase, Focal Adhesion Kinase (FAK), in the regulation of cell movement with the emphasis on how FAK regulates the flow of signals from the ECM to the actin cytoskeleton.
Collapse
Affiliation(s)
- J T Parsons
- Department of Microbiology, Health Science Center, University of Virginia, Charlottesville, Virginia, VA 22908, USA
| | | | | | | | | |
Collapse
|
42
|
Vial D, Okazaki H, Siraganian RP. The NH2-terminal region of focal adhesion kinase reconstitutes high affinity IgE receptor-induced secretion in mast cells. J Biol Chem 2000; 275:28269-75. [PMID: 10862765 DOI: 10.1074/jbc.m002842200] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Focal adhesion kinase (FAK) is tyrosine-phosphorylated by adherence of cells and also by FcepsilonRI aggregation in RBL-2H3 mast cells. Using phosphorylation site-specific antibodies, we observed that FcepsilonRI activation in these cells led to an increase in FAK phosphorylation at the same tyrosine residues that are phosphorylated by integrin-induced activation. Previous studies in the 3B6 line, a FAK-deficient variant of the RBL-2H3 cells, suggest that FAK plays a role in FcepsilonRI-induced secretion. Stable cell lines expressing either full-length or truncated forms of FAK were isolated after transfection of the FAK-deficient 3B6 variant cells. The NH(2) domain of FAK, which lacks the enzymatic and the COOH-terminal regions, was sufficient to reconstitute secretion. The different truncated forms of FAK were still tyrosine-phosphorylated after FcepsilonRI aggregation. Therefore, the kinase domain and the COOH-terminal region are not essential for FcepsilonRI-induced tyrosine phosphorylation of FAK or for secretion. Taken together, our data demonstrate that the reconstitution of secretion is dissociated from FAK activation and that the NH(2)-terminal region of FAK is the only critical element that may play a role in FcepsilonRI-induced secretion by acting as an adaptor or linker molecule.
Collapse
Affiliation(s)
- D Vial
- Receptors and Signal Transduction Section, OIIB, NIDCR, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | |
Collapse
|
43
|
Palmer RH, Fessler LI, Edeen PT, Madigan SJ, McKeown M, Hunter T. DFak56 is a novel Drosophila melanogaster focal adhesion kinase. J Biol Chem 1999; 274:35621-9. [PMID: 10585440 PMCID: PMC1975812 DOI: 10.1074/jbc.274.50.35621] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The mammalian focal adhesion kinase (FAK) family of nonreceptor protein-tyrosine kinases have been implicated in controlling a multitude of cellular responses to the engagement of cell surface integrins and G protein-coupled receptors. We describe here a Drosophila melanogaster FAK homologue, DFak56, which maps to band 56D on the right arm of the second chromosome. Full-length DFak56 cDNA encodes a phosphoprotein of 140 kDa, which shares strong sequence similarity not only with mammalian p125(FAK) but also with the more recently described mammalian Pyk2 (also known as CAKbeta, RAFTK, FAK2, and CADTK) FAK family member. DFak56 has intrinsic tyrosine kinase activity and is phosphorylated on tyrosine in vivo. As is the case for FAK, tyrosine phosphorylation of DFak56 is increased upon plating Drosophila embryo cells on extracellular matrix proteins. In situ hybridization and immunofluorescence staining analysis showed that DFak56 is ubiquitously expressed with particularly high levels within the developing central nervous system. We utilized the UAS-GAL4 expression system to express DFak56 and analyze its function in vivo. Overexpression of DFak56 in the wing imaginal disc results in wing blistering in adults, a phenotype also observed with both position-specific integrin loss of function and position-specific integrin overexpression. Our results imply a role for DFak56 in adhesion-dependent signaling pathways in vivo during D. melanogaster development.
Collapse
Affiliation(s)
- R H Palmer
- Salk Institute, Molecular Biology and Virology Laboratory, La Jolla, California 92037-1099, USA
| | | | | | | | | | | |
Collapse
|