51
|
Egom EEA, Bae JS, Capel R, Richards M, Ke Y, Pharithi RB, Maher V, Kruzliak P, Lei M. Effect of sphingosine-1-phosphate on L-type calcium current and Ca2+ transient in rat ventricular myocytes. Mol Cell Biochem 2016; 419:83-92. [DOI: 10.1007/s11010-016-2752-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 06/21/2016] [Indexed: 01/05/2023]
|
52
|
Zhao LJ, Yuan HM, Guo WD, Yang CP. Digital Gene Expression Analysis of Populus simonii × P. nigra Pollen Germination and Tube Growth. FRONTIERS IN PLANT SCIENCE 2016; 7:825. [PMID: 27379121 PMCID: PMC4908133 DOI: 10.3389/fpls.2016.00825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 05/26/2016] [Indexed: 05/27/2023]
Abstract
Pollen tubes are an ideal model for the study of cell growth and morphogenesis because of their extreme elongation without cell division; however, the genetic basis of pollen germination and tube growth remains largely unknown. Using the Illumina/Solexa digital gene expression system, we identified 13,017 genes (representing 28.3% of the unigenes on the reference genes) at three stages, including mature pollen, hydrated pollen, and pollen tubes of Populus simonii × P. nigra. Comprehensive analysis of P. simonii × P. nigra pollen revealed dynamic changes in the transcriptome during pollen germination and pollen tube growth (PTG). Gene ontology analysis of differentially expressed genes showed that genes involved in functional categories such as catalytic activity, binding, transporter activity, and enzyme regulator activity were overrepresented during pollen germination and PTG. Some highly dynamic genes involved in pollen germination and PTG were detected by clustering analysis. Genes related to some key pathways such as the mitogen-activated protein kinase signaling pathway, regulation of the actin cytoskeleton, calcium signaling, and ubiquitin-mediated proteolysis were significantly changed during pollen germination and PTG. These data provide comprehensive molecular information toward further understanding molecular mechanisms underlying pollen germination and PTG.
Collapse
Affiliation(s)
- Li-Juan Zhao
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry UniversityHarbin, China
- Department of Crop Molecular Breeding, Crop Breeding Institute, Heilongjiang Academy of Agricultural SciencesHarbin, China
| | - Hong-Mei Yuan
- Medical Plant Research Center, Economic Crop Institute, Heilongjiang Academy of Agricultural SciencesHarbin, China
| | - Wen-Dong Guo
- Biotechnology Research Center, Institute of Natural Resources and Ecology, Heilongjiang Academy of SciencesHarbin, China
| | - Chuan-Ping Yang
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry UniversityHarbin, China
| |
Collapse
|
53
|
Parvathy M, Sreeja S, Kumar R, Pillai MR. Potential role of p21 Activated Kinase 1 (PAK1) in the invasion and motility of oral cancer cells. BMC Cancer 2016; 16 Suppl 1:293. [PMID: 27229476 PMCID: PMC4896241 DOI: 10.1186/s12885-016-2263-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Oral cancer malignancy consists of uncontrolled division of cells primarily in and around the floor of the oral cavity, gingiva, oropharynx, lower lip and base of the tongue. According to GLOBOCAN 2012 report, oral cancer is one of the most common cancers among males and females in India. Even though significant advancements have been made in the field of oral cancer treatment modalities, the overall prognosis for the patients has not improved in the past few decades and hence, this demands a new thrust for the identification of novel therapeutic targets in oral cancer. p21 Activated Kinases (PAKs) are potential therapeutic targets that are involved in numerous physiological functions. PAKs are serine-threonine kinases and they serve as important regulators of cytoskeletal dynamics and cell motility, transcription through MAP kinase cascades, death and survival signalling, and cell-cycle progression. Although PAKs are known to play crucial roles in cancer progression, the role and clinical significance of PAKs in oral cancer remains poorly understood. RESULTS Our results suggest that PAK1 is over-expressed in oral cancer cell lines. Stimulation of Oral Squamous Cell Carcinoma (OSCC) cells with serum growth factors leads to PAK1 re-localization and might cause a profound cytoskeletal remodelling. PAK1 was also found to be involved in the invasion, migration and cytoskeletal remodelling of OSCC cells. CONCLUSIONS Our study revealed that PAK1 may play a crucial role in the progression of OSCC. Studying the role of PAK1 and its substrates is likely to enhance our understanding of oral carcinogenesis and potential therapeutic value of PAKs in oral cancer.
Collapse
Affiliation(s)
- Muraleedharan Parvathy
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Sreeharshan Sreeja
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Rakesh Kumar
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Department of Biochemistry and Molecular Medicine, George Washington University, Washington DC, USA
| | | |
Collapse
|
54
|
Regulation of the Postsynaptic Compartment of Excitatory Synapses by the Actin Cytoskeleton in Health and Its Disruption in Disease. Neural Plast 2016; 2016:2371970. [PMID: 27127658 PMCID: PMC4835652 DOI: 10.1155/2016/2371970] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 03/09/2016] [Indexed: 02/07/2023] Open
Abstract
Disruption of synaptic function at excitatory synapses is one of the earliest pathological changes seen in wide range of neurological diseases. The proper control of the segregation of neurotransmitter receptors at these synapses is directly correlated with the intact regulation of the postsynaptic cytoskeleton. In this review, we are discussing key factors that regulate the structure and dynamics of the actin cytoskeleton, the major cytoskeletal building block that supports the postsynaptic compartment. Special attention is given to the complex interplay of actin-associated proteins that are found in the synaptic specialization. We then discuss our current understanding of how disruption of these cytoskeletal elements may contribute to the pathological events observed in the nervous system under disease conditions with a particular focus on Alzheimer's disease pathology.
Collapse
|
55
|
Uribe RA, Buzzi AL, Bronner ME, Strobl-Mazzulla PH. Histone demethylase KDM4B regulates otic vesicle invagination via epigenetic control of Dlx3 expression. J Cell Biol 2016; 211:815-27. [PMID: 26598618 PMCID: PMC4657164 DOI: 10.1083/jcb.201503071] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
In vertebrate embryos, the histone demethylase KDM4B affects otic placode proliferation, intercellular adhesion, and invagination by directly regulating Dlx3 expression. In vertebrates, the inner ear arises from the otic placode, a thickened swathe of ectoderm that invaginates to form the otic vesicle. We report that histone demethylase KDM4B is dynamically expressed during early stages of chick inner ear formation. A loss of KDM4B results in defective invagination and striking morphological changes in the otic epithelium, characterized by abnormal localization of adhesion and cytoskeletal molecules and reduced expression of several inner ear markers, including Dlx3. In vivo chromatin immunoprecipitation reveals direct and dynamic occupancy of KDM4B and its target, H3K9me3, at regulatory regions of the Dlx3 locus. Accordingly, coelectroporations of DLX3 or KDM4B encoding constructs, but not a catalytically dead mutant of KDM4B, rescue the ear invagination phenotype caused by KDM4B knockdown. Moreover, a loss of DLX3 phenocopies a loss of KDM4B. Collectively, our findings suggest that KDM4B play a critical role during inner ear invagination via modulating histone methylation of the direct target Dlx3.
Collapse
Affiliation(s)
- Rosa A Uribe
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Ailín L Buzzi
- Laboratory of Developmental Biology, Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús (CONICET-UNSAM), 7130 Chascomús, Argentina
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Pablo H Strobl-Mazzulla
- Laboratory of Developmental Biology, Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús (CONICET-UNSAM), 7130 Chascomús, Argentina
| |
Collapse
|
56
|
p21-activated kinase 2 regulates HSPC cytoskeleton, migration, and homing via CDC42 activation and interaction with β-Pix. Blood 2016; 127:1967-75. [PMID: 26932803 DOI: 10.1182/blood-2016-01-693572] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 02/19/2016] [Indexed: 12/13/2022] Open
Abstract
Cytoskeletal remodeling of hematopoietic stem and progenitor cells (HSPCs) is essential for homing to the bone marrow (BM). The Ras-related C3 botulinum toxin substrate (Rac)/cell division control protein 42 homolog (CDC42) effector p21-activated kinase (Pak2) has been implicated in HSPC homing and engraftment. However, the molecular pathways mediating Pak2 functions in HSPCs are unknown. Here, we demonstrate that both Pak2 kinase activity and its interaction with the PAK-interacting exchange factor-β (β-Pix) are required to reconstitute defective ITALIC! Pak2 (ITALIC! Δ/Δ)HSPC homing to the BM. Pak2 serine/threonine kinase activity is required for stromal-derived factor-1 (SDF1α) chemokine-induced HSPC directional migration, whereas Pak2 interaction with β-Pix is required to regulate the velocity of HSPC migration and precise F-actin assembly. Lack of SDF1α-induced filopodia and associated abnormal cell protrusions seen in ITALIC! Pak2 (ITALIC! Δ/Δ)HSPCs were rescued by wild-type (WT) Pak2 but not by a Pak2-kinase dead mutant (KD). Expression of a β-Pix interaction-defective mutant of Pak2 rescued filopodia formation but led to abnormal F-actin bundles. Although CDC42 has previously been considered an upstream regulator of Pak2, we found a paradoxical decrease in baseline activation of CDC42 in ITALIC! Pak2 (ITALIC! Δ/Δ)HSPCs, which was rescued by expression of Pak2-WT but not by Pak2-KD; defective homing of ITALIC! Pak2-deleted HSPCs was rescued by constitutive active CDC42. These data demonstrate that both Pak2 kinase activity and its interaction with β-Pix are essential for HSPC filopodia formation, cytoskeletal integrity, and homing via activation of CDC42. Taken together, we provide mechanistic insights into the role of Pak2 in HSPC migration and homing.
Collapse
|
57
|
Nohata N, Uchida Y, Stratman AN, Adams RH, Zheng Y, Weinstein BM, Mukouyama YS, Gutkind JS. Temporal-specific roles of Rac1 during vascular development and retinal angiogenesis. Dev Biol 2016; 411:183-194. [PMID: 26872874 DOI: 10.1016/j.ydbio.2016.02.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/07/2016] [Accepted: 02/07/2016] [Indexed: 01/04/2023]
Abstract
Angiogenesis, the formation of new blood vessels by remodeling and growth of pre-existing vessels, is a highly orchestrated process that requires a tight balance between pro-angiogenic and anti-angiogenic factors and the integration of their corresponding signaling networks. The family of Rho GTPases, including RhoA, Rac1, and Cdc42, play a central role in many cell biological processes that involve cytoskeletal changes and cell movement. Specifically for Rac1, we have shown that excision of Rac1 using a Tie2-Cre animal line results in embryonic lethality in midgestation (embryonic day (E) 9.5), with multiple vascular defects. However, Tie2-Cre can be also expressed during vasculogenesis, prior to angiogenesis, and is active in some hematopoietic precursors that can affect vessel formation. To circumvent these limitations, we have now conditionally deleted Rac1 in a temporally controlled and endothelial-restricted fashion using Cdh5(PAC)-iCreERT2 transgenic mice. In this highly controlled experimental in vivo system, we now show that Rac1 is required for embryonic vascular integrity and angiogenesis, and for the formation of superficial and deep vascular networks in the post-natal developing retina, the latter involving a novel specific function for Rac1 in vertical blood vessel sprouting. Aligned with these findings, we show that RAC1 is spatially involved in endothelial cell migration, invasion, and radial sprouting activities in 3D collagen matrix in vitro models. Hence, Rac1 and its downstream molecules may represent potential anti-angiogeneic therapeutic targets for the treatment of many human diseases that involve aberrant neovascularization and blood vessel overgrowth.
Collapse
Affiliation(s)
- Nijiro Nohata
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, United States
| | - Yutaka Uchida
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20814, United States
| | - Amber N Stratman
- Section on Vertebrate Development, Program in the Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, United States
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine and Faculty of Medicine, University of Münster, D-48149 Münster, Germany
| | - Yi Zheng
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital, University of Cincinnati College of Medicine, Cincinnati, OH 45229, United States
| | - Brant M Weinstein
- Section on Vertebrate Development, Program in the Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, United States
| | - Yoh-Suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20814, United States
| | - J Silvio Gutkind
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, United States; Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, United States.
| |
Collapse
|
58
|
Group I Paks as therapeutic targets in NF2-deficient meningioma. Oncotarget 2015; 6:1981-94. [PMID: 25596744 PMCID: PMC4385830 DOI: 10.18632/oncotarget.2810] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 11/24/2014] [Indexed: 11/25/2022] Open
Abstract
Neurofibromatosis type 2 (NF2) is an autosomal dominant disorder characterized by the development of multiple tumors in the central nervous system, most notably schwannomas and meningiomas. Mutational inactivation of NF2 is found in 40-60% of sporadic meningiomas, but the molecular mechanisms underlying malignant changes of meningioma cells remain unclear. Because group I p21-activated kinases (Paks) bind to and are inhibited by the NF2-encoded protein Merlin, we assessed the signaling and anti-tumor effects of three group-I specific Pak inhibitors - Frax597, 716 and 1036 - in NF2-/- meningiomas in vitro and in an orthotopic mouse model. We found that these Pak inhibitors suppressed the proliferation and motility of both benign (Ben-Men1) and malignant (KT21-MG1) meningiomas cells. In addition, we found a strong reduction in phosphorylation of Mek and S6, and decreased cyclin D1 expression in both cell lines after treatment with Pak inhibitors. Using intracranial xenografts of luciferase-expressing KT21-MG1 cells, we found that treated mice showed significant tumor suppression for all three Pak inhibitors. Similar effects were observed in Ben-Men1 cells. Tumors dissected from treated animals exhibited an increase in apoptosis without notable change in proliferation. Collectively, these results suggest that Pak inhibitors might be useful agents in treating NF2-deficient meningiomas.
Collapse
|
59
|
Flores-Benitez D, Knust E. Crumbs is an essential regulator of cytoskeletal dynamics and cell-cell adhesion during dorsal closure in Drosophila. eLife 2015; 4. [PMID: 26544546 PMCID: PMC4718732 DOI: 10.7554/elife.07398] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 11/06/2015] [Indexed: 12/12/2022] Open
Abstract
The evolutionarily conserved Crumbs protein is required for epithelial polarity and morphogenesis. Here we identify a novel role of Crumbs as a negative regulator of actomyosin dynamics during dorsal closure in the Drosophila embryo. Embryos carrying a mutation in the FERM (protein 4.1/ezrin/radixin/moesin) domain-binding motif of Crumbs die due to an overactive actomyosin network associated with disrupted adherens junctions. This phenotype is restricted to the amnioserosa and does not affect other embryonic epithelia. This function of Crumbs requires DMoesin, the Rho1-GTPase, class-I p21-activated kinases and the Arp2/3 complex. Data presented here point to a critical role of Crumbs in regulating actomyosin dynamics, cell junctions and morphogenesis. DOI:http://dx.doi.org/10.7554/eLife.07398.001 A layer of epithelial cells covers the body surface of animals. Epithelial cells have a property known as polarity; this means that they have two different poles, one of which is in contact with the environment. Midway through embryonic development, the Drosophila embryo is covered by two kinds of epithelial sheets; the epidermis on the front, the belly and the sides of the embryo, and the amnioserosa on the back. In the second half of embryonic development, the amnioserosa is brought into the embryo in a process called dorsal closure, while the epidermis expands around the back of the embryo to encompass it. One of the major activities driving dorsal closure is the contraction of amnioserosa cells. This contraction depends on the highly dynamic activity of the protein network that helps give cells their shape, known as the actomyosin cytoskeleton. One major question in the field is how changes in the actomyosin cytoskeleton are controlled as tissues take shape (a process known as “morphogenesis”) and how the integrity of epithelial tissues is maintained during these processes. A key regulator of epidermal and amnioserosa polarity is an evolutionarily conserved protein called Crumbs. The epithelial tissues of mutant embryos that do not produce Crumbs lose polarity and integrity, and the embryos fail to develop properly. Flores-Benitez and Knust have now studied the role of Crumbs in the morphogenesis of the amnioserosa during dorsal closure. This revealed that fly embryos that produce a mutant Crumbs protein that cannot interact with a protein called Moesin (which links the cell membrane and the actomyosin cytoskeleton) are unable to complete dorsal closure. Detailed analyses showed that this failure of dorsal closure is due to the over-activity of the actomyosin cytoskeleton in the amnioserosa. This results in increased and uncoordinated contractions of the cells, and is accompanied by defects in cell-cell adhesion that ultimately cause the amnioserosa to lose integrity. Flores-Benitez and Knust’s genetic analyses further showed that several different signalling systems participate in this process. Flores-Benitez and Knust’s results reveal an unexpected role of Crumbs in coordinating polarity, actomyosin activity and cell-cell adhesion. Further work is now needed to understand the molecular mechanisms and interactions that enable Crumbs to coordinate these processes; in particular, to unravel how Crumbs influences the periodic contractions that drive changes in cell shape. It will also be important to investigate whether Crumbs is involved in similar mechanisms that operate in other developmental events in which actomyosin oscillations have been linked to tissue morphogenesis. DOI:http://dx.doi.org/10.7554/eLife.07398.002
Collapse
Affiliation(s)
| | - Elisabeth Knust
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
60
|
Davis RT, Simon JN, Utter M, Mungai P, Alvarez MG, Chowdhury SAK, Heydemann A, Ke Y, Wolska BM, Solaro RJ. Knockout of p21-activated kinase-1 attenuates exercise-induced cardiac remodelling through altered calcineurin signalling. Cardiovasc Res 2015; 108:335-47. [PMID: 26464331 DOI: 10.1093/cvr/cvv234] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 10/03/2015] [Indexed: 01/14/2023] Open
Abstract
AIMS Despite its known cardiovascular benefits, the intracellular signalling mechanisms underlying physiological cardiac growth remain poorly understood. Therefore, the purpose of this study was to investigate a novel role of p21-activated kinase-1 (Pak1) in the regulation of exercise-induced cardiac hypertrophy. METHODS AND RESULTS Wild-type (WT) and Pak1 KO mice were subjected to 6 weeks of treadmill endurance exercise training (ex-training). Cardiac function was assessed via echocardiography, in situ haemodynamics, and the pCa-force relations in skinned fibre preparations at baseline and at the end of the training regimen. Post-translational modifications to the sarcomeric proteins and expression levels of calcium-regulating proteins were also assessed following ex-training. Heart weight/tibia length and echocardiography data revealed that there was marked hypertrophy following ex-training in the WT mice, which was not evident in the KO mice. Additionally, following ex-training, WT mice demonstrated an increase in cardiac contractility, myofilament calcium sensitivity, and phosphorylation of cardiac myosin-binding protein C, cardiac TnT, and tropomyosin compared with KO mice. With ex-training in WT mice, there were also increased protein levels of calcineurin and increased phosphorylation of phospholamban. CONCLUSIONS Our data suggest that Pak1 is essential for adaptive physiological cardiac remodelling and support previous evidence that demonstrates Pak1 signalling is important for cardiac growth and survival.
Collapse
Affiliation(s)
- Robert T Davis
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Jillian N Simon
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Megan Utter
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Paul Mungai
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Manuel G Alvarez
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Shamim A K Chowdhury
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Ahlke Heydemann
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Yunbo Ke
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Beata M Wolska
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA Department of Medicine, Section of Cardiology, Center for Cardiovascular Research, University of Illinois, Chicago, IL 60612, USA
| | - R John Solaro
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| |
Collapse
|
61
|
p21-Activated Kinase 2 Regulates Endothelial Development and Function through the Bmk1/Erk5 Pathway. Mol Cell Biol 2015; 35:3990-4005. [PMID: 26391956 DOI: 10.1128/mcb.00630-15] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 09/08/2015] [Indexed: 02/03/2023] Open
Abstract
p21-activated kinases (Paks) have been shown to regulate cytoskeleton rearrangements, cell proliferation, attachment, and migration in a variety of cellular contexts, including endothelial cells. However, the role of endothelial Pak in embryo development has not been reported, and currently, there is no consensus on the endothelial function of individual Pak isoforms, in particular p21-activated kinase 2 (Pak2), the main Pak isoform expressed in endothelial cells. In this work, we employ genetic and molecular studies that show that Pak2, but not Pak1, is a critical mediator of development and maintenance of endothelial cell function. Endothelial depletion of Pak2 leads to early embryo lethality due to flawed blood vessel formation in the embryo body and yolk sac. In adult endothelial cells, Pak2 depletion leads to severe apoptosis and acute angiogenesis defects, and in adult mice, endothelial Pak2 deletion leads to increased vascular permeability. Furthermore, ubiquitous Pak2 deletion is lethal in adult mice. We show that many of these defects are mediated through a newly unveiled Pak2/Bmk1 pathway. Our results demonstrate that endothelial Pak2 is essential during embryogenesis and also for adult blood vessel maintenance, and they also pinpoint the Bmk1/Erk5 pathway as a critical mediator of endothelial Pak2 signaling.
Collapse
|
62
|
Fehér A, Lajkó DB. Signals fly when kinases meet Rho-of-plants (ROP) small G-proteins. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2015; 237:93-107. [PMID: 26089155 DOI: 10.1016/j.plantsci.2015.05.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Revised: 05/11/2015] [Accepted: 05/12/2015] [Indexed: 05/11/2023]
Abstract
Rho-type small GTP-binding plant proteins function as two-state molecular switches in cellular signalling. There is accumulating evidence that Rho-of-plants (ROP) signalling is positively controlled by plant receptor kinases, through the ROP guanine nucleotide exchange factor proteins. These signalling modules regulate cell polarity, cell shape, hormone responses, and pathogen defence, among other things. Other ROP-regulatory proteins might also be subjected to protein phosphorylation by cellular kinases (e.g., mitogen-activated protein kinases or calcium-dependent protein kinases), in order to integrate various cellular signalling pathways with ROP GTPase-dependent processes. In contrast to the role of kinases in upstream ROP regulation, much less is known about the potential link between ROP GTPases and downstream kinase signalling. In other eukaryotes, Rho-type G-protein-activated kinases are widespread and have a key role in many cellular processes. Recent data indicate the existence of structurally different ROP-activated kinases in plants, but their ROP-dependent biological functions still need to be validated. In addition to these direct interactions, ROPs may also indirectly control the activity of mitogen-activated protein kinases or calcium-dependent protein kinases. These kinases may therefore function as upstream as well as downstream kinases in ROP-mediated signalling pathways, such as the phosphatidylinositol monophosphate kinases involved in cell polarity establishment.
Collapse
Affiliation(s)
- Attila Fehér
- Institute of Plant Biology, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary.
| | - Dézi Bianka Lajkó
- Institute of Plant Biology, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary
| |
Collapse
|
63
|
Singh NK, Kotla S, Dyukova E, Traylor JG, Orr AW, Chernoff J, Marion TN, Rao GN. Disruption of p21-activated kinase 1 gene diminishes atherosclerosis in apolipoprotein E-deficient mice. Nat Commun 2015; 6:7450. [PMID: 26104863 PMCID: PMC4480433 DOI: 10.1038/ncomms8450] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 05/09/2015] [Indexed: 12/26/2022] Open
Abstract
Pak1 plays an important role in various cellular processes, including cell motility, polarity, survival and proliferation. To date, its role in atherogenesis has not been explored. Here we report the effect of Pak1 on atherogenesis using atherosclerosis-prone apolipoprotein E-deficient (ApoE−/−) mice as a model. Disruption of Pak1 in ApoE−/− mice results in reduced plaque burden, significantly attenuates circulating IL-6 and MCP-1 levels, limits the expression of adhesion molecules and diminishes the macrophage content in the aortic root of ApoE−/− mice. We also observed reduced oxidized LDL uptake and increased cholesterol efflux by macrophages and smooth muscle cells of ApoE−/−:Pak1−/− mice as compared with ApoE−/− mice. In addition, we detect increased Pak1 phosphorylation in human atherosclerotic arteries, suggesting its role in human atherogenesis. Altogether, these results identify Pak1 as an important factor in the initiation and progression of atherogenesis. Atherogenesis involves coordinated action of different cell types and factors. Here the authors show that the kinase Pak1 represents a key pro-atherogenic factor affecting the function of macrophages and vascular smooth muscle cells, including their production of proinflammatory cytokine IL-6 and chemokine MCP-1, and retention of cholesterol.
Collapse
Affiliation(s)
- Nikhlesh K Singh
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Memphis, Tennessee 38163, USA
| | - Sivareddy Kotla
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Memphis, Tennessee 38163, USA
| | - Elena Dyukova
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Memphis, Tennessee 38163, USA
| | - James G Traylor
- Department of Pathology, LSU Health Sciences Center, Shreveport, Louisiana 71103, USA
| | - A Wayne Orr
- Department of Pathology, LSU Health Sciences Center, Shreveport, Louisiana 71103, USA
| | - Jonathan Chernoff
- Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, Pennsylvania 19111, USA
| | - Tony N Marion
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Gadiparthi N Rao
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Memphis, Tennessee 38163, USA
| |
Collapse
|
64
|
Whalley HJ, Porter AP, Diamantopoulou Z, White GRM, Castañeda-Saucedo E, Malliri A. Cdk1 phosphorylates the Rac activator Tiam1 to activate centrosomal Pak and promote mitotic spindle formation. Nat Commun 2015; 6:7437. [PMID: 26078008 PMCID: PMC4490568 DOI: 10.1038/ncomms8437] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 05/08/2015] [Indexed: 01/13/2023] Open
Abstract
Centrosome separation is critical for bipolar spindle formation and the accurate segregation of chromosomes during mammalian cell mitosis. Kinesin-5 (Eg5) is a microtubule motor essential for centrosome separation, and Tiam1 and its substrate Rac antagonize Eg5-dependent centrosome separation in early mitosis promoting efficient chromosome congression. Here we identify S1466 of Tiam1 as a novel Cdk1 site whose phosphorylation is required for the mitotic function of Tiam1. We find that this phosphorylation of Tiam1 is required for the activation of group I p21-activated kinases (Paks) on centrosomes in prophase. Further, we show that both Pak1 and Pak2 counteract centrosome separation in a kinase-dependent manner and demonstrate that they act downstream of Tiam1. We also show that depletion of Pak1/2 allows cells to escape monopolar arrest by Eg5 inhibition, highlighting the potential importance of this signalling pathway for the development of Eg5 inhibitors as cancer therapeutics.
Collapse
Affiliation(s)
- Helen J. Whalley
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Andrew P. Porter
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Zoi Diamantopoulou
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Gavin R. M. White
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Eduardo Castañeda-Saucedo
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Angeliki Malliri
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| |
Collapse
|
65
|
Ha BH, Morse EM, Turk BE, Boggon TJ. Signaling, Regulation, and Specificity of the Type II p21-activated Kinases. J Biol Chem 2015; 290:12975-83. [PMID: 25855792 DOI: 10.1074/jbc.r115.650416] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The p21-activated kinases (PAKs) are a family of six serine/threonine kinases that act as key effectors of RHO family GTPases in mammalian cells. PAKs are subdivided into two groups: type I PAKs (PAK1, PAK2, and PAK3) and type II PAKs (PAK4, PAK5, and PAK6). Although these groups are involved in common signaling pathways, recent work indicates that the two groups have distinct modes of regulation and have both unique and common substrates. Here, we review recent insights into the molecular level details that govern regulation of type II PAK signaling. We also consider mechanisms by which signal transduction is regulated at the level of substrate specificity. Finally, we discuss the implications of these studies for clinical targeting of these kinases.
Collapse
Affiliation(s)
| | - Elizabeth M Morse
- Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06520
| | | | | |
Collapse
|
66
|
Wang Y, Tsui H, Bolton EL, Wang X, Huang CLH, Solaro RJ, Ke Y, Lei M. Novel insights into mechanisms for Pak1-mediated regulation of cardiac Ca(2+) homeostasis. Front Physiol 2015; 6:76. [PMID: 25852566 PMCID: PMC4362298 DOI: 10.3389/fphys.2015.00076] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 02/25/2015] [Indexed: 11/13/2022] Open
Abstract
A series of recent studies report novel roles for Pak1, a key member of the highly conserved family of serine-threonine protein kinases regulated by Ras-related small G-proteins, Cdc42/Rac1, in cardiac physiology and cardioprotection. Previous studies had identified Pak1 in the regulation of hypertrophic remodeling that could potentially lead to heart failure. This article provides a review of more recent findings on the roles of Pak1 in cardiac Ca(2+) homeostasis. These findings identified crucial roles for Pak1 in cardiomyocyte Ca(2+) handling and demonstrated that it functions through unique mechanisms involving regulation of the post-transcriptional activity of key Ca(2+)-handling proteins, including the expression of Ca(2+)-ATPase SERCA2a, along with the speculative possibility of an involvement in the maintenance of transverse (T)-tubular structure. They highlight important regulatory functions of Pak1 in Ca(2+) homeostasis in cardiac cells, and identify novel potential therapeutic strategies directed at manipulation of Pak1 signaling for the management of cardiac disease, particularly heart failure.
Collapse
Affiliation(s)
- Yanwen Wang
- Department of Pharmacology, University of Oxford Oxford, UK
| | - Hoyee Tsui
- Faculty of Life Science, University of Manchester Manchester, UK
| | - Emma L Bolton
- Department of Pharmacology, University of Oxford Oxford, UK
| | - Xin Wang
- Faculty of Life Science, University of Manchester Manchester, UK
| | | | - R John Solaro
- Department of Physiology and Biophysics, University of Illinois Chicago, IL, USA
| | - Yunbo Ke
- Department of Physiology and Biophysics, University of Illinois Chicago, IL, USA
| | - Ming Lei
- Department of Pharmacology, University of Oxford Oxford, UK
| |
Collapse
|
67
|
Nie F, Wang XF, Zhao SY, Bu L, Liu XH. Gene silencing of Rac1 with RNA interference mediated by ultrasound and microbubbles in human LoVo cells: evaluation of cell invasion inhibition and metastatic. J Drug Target 2015; 23:380-6. [PMID: 25673262 DOI: 10.3109/1061186x.2014.1002500] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE The objective of this study was to assess the change of cytoskeleton and cell cycle in LoVo (human colorectal cancer) cell via gene silencing of Rac1 with RNA interference mediated by microbubble (SonoVue) and ultrasound (US). METHODS The compound of plasmid Rac1-shRNA, LoVo cells, and SonoVue was exposed to US (1 MHz, 2 W/cm(2), 5 min). The expression of Rac1 mRNA and Rac1 protein was detected by RT-PCR and Western blot. Cytoskeleton was taken by confocal microscope in a random fashion. Cell invasion was assayed using modified Boyden chambers, and cell cycle and apoptosis were analyzed by flow cytometry in LoVo cells. RESULTS Rac1 gene is overexpressed in human colorectal cancer cells, gene silencing of Rac1 with RNA interference mediated by microbubble and US strongly inhibited lamellipodia formation, cell invasion, and delayed cell cycle, as well as enhanced cell apoptosis of LoVo cells in vitro. CONCLUSION Silencing Rac1 gene mediated by microbubble and US may become a new treatment option for the inhibition of the invasion and metastasis of colorectal cancer cells.
Collapse
Affiliation(s)
- Fang Nie
- Department of Medical Ultrasonics, Lanzhou University Second Hospital , Lanzhou, Gansu , China and
| | | | | | | | | |
Collapse
|
68
|
Kim Y, Lee SE, Park J, Kim M, Lee B, Hwang D, Chang S. ADP-ribosylation factor 6 (ARF6) bidirectionally regulates dendritic spine formation depending on neuronal maturation and activity. J Biol Chem 2015; 290:7323-35. [PMID: 25605715 DOI: 10.1074/jbc.m114.634527] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Recent studies have reported conflicting results regarding the role of ARF6 in dendritic spine development, but no clear answer for the controversy has been suggested. We found that ADP-ribosylation factor 6 (ARF6) either positively or negatively regulates dendritic spine formation depending on neuronal maturation and activity. ARF6 activation increased the spine formation in developing neurons, whereas it decreased spine density in mature neurons. Genome-wide microarray analysis revealed that ARF6 activation in each stage leads to opposite patterns of expression of a subset of genes that are involved in neuronal morphology. ARF6-mediated Rac1 activation via the phospholipase D pathway is the coincident factor in both stages, but the antagonistic RhoA pathway becomes involved in the mature stage. Furthermore, blocking neuronal activity in developing neurons using tetrodotoxin or enhancing the activity in mature neurons using picrotoxin or chemical long term potentiation reversed the effect of ARF6 on each stage. Thus, activity-dependent dynamic changes in ARF6-mediated spine structures may play a role in structural plasticity of mature neurons.
Collapse
Affiliation(s)
- Yoonju Kim
- From the Department of Physiology and Biomedical Sciences, Neuroscience Research Institute, Medical Research Center, Biomembrane Plasticity Research Center, and
| | - Sang-Eun Lee
- From the Department of Physiology and Biomedical Sciences, Biomembrane Plasticity Research Center, and
| | - Joohyun Park
- From the Department of Physiology and Biomedical Sciences, Neuroscience Research Institute, Medical Research Center, Biomembrane Plasticity Research Center, and
| | - Minhyung Kim
- School of Interdisciplinary Bioscience and Bioengineering and Department of Chemical Engineering, Pohang University of Science and Technology, Pohang, Kyungbook 790-784, South Korea, and
| | - Boyoon Lee
- Interdisciplinary Program in Neuroscience, Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Daehee Hwang
- School of Interdisciplinary Bioscience and Bioengineering and Department of Chemical Engineering, Pohang University of Science and Technology, Pohang, Kyungbook 790-784, South Korea, and Center for Systems Biology of Plant Senescence and Life History, Institute for Basic Science, Daegu Gyeongbuk Institute of Science and Technology, Daegu, 711-873, South Korea
| | - Sunghoe Chang
- From the Department of Physiology and Biomedical Sciences, Neuroscience Research Institute, Medical Research Center, Biomembrane Plasticity Research Center, and Interdisciplinary Program in Neuroscience, Seoul National University College of Medicine, Seoul 110-799, South Korea,
| |
Collapse
|
69
|
Conroy BD, Herek TA, Shew TD, Latner M, Larson JJ, Allen L, Davis PH, Helikar T, Cutucache CE. Design, Assessment, and in vivo Evaluation of a Computational Model Illustrating the Role of CAV1 in CD4(+) T-lymphocytes. Front Immunol 2014; 5:599. [PMID: 25538703 PMCID: PMC4257089 DOI: 10.3389/fimmu.2014.00599] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 11/07/2014] [Indexed: 01/08/2023] Open
Abstract
Caveolin-1 (CAV1) is a vital scaffold protein heterogeneously expressed in both healthy and malignant tissue. We focus on the role of CAV1 when overexpressed in T-cell leukemia. Previously, we have shown that CAV1 is involved in cell-to-cell communication, cellular proliferation, and immune synapse formation; however, the molecular mechanisms have not been elucidated. We hypothesize that the role of CAV1 in immune synapse formation contributes to immune regulation during leukemic progression, thereby warranting studies of the role of CAV1 in CD4+ T-cells in relation to antigen-presenting cells. To address this need, we developed a computational model of a CD4+ immune effector T-cell to mimic cellular dynamics and molecular signaling under healthy and immunocompromised conditions (i.e., leukemic conditions). Using the Cell Collective computational modeling software, the CD4+ T-cell model was constructed and simulated under CAV1+/+, CAV1+/−, and CAV1−/− conditions to produce a hypothetical immune response. This model allowed us to predict and examine the heterogeneous effects and mechanisms of CAV1 in silico. Experimental results indicate a signature of molecules involved in cellular proliferation, cell survival, and cytoskeletal rearrangement that were highly affected by CAV1 knock out. With this comprehensive model of a CD4+ T-cell, we then validated in vivo protein expression levels. Based on this study, we modeled a CD4+ T-cell, manipulated gene expression in immunocompromised versus competent settings, validated these manipulations in an in vivo murine model, and corroborated acute T-cell leukemia gene expression profiles in human beings. Moreover, we can model an immunocompetent versus an immunocompromised microenvironment to better understand how signaling is regulated in patients with leukemia.
Collapse
Affiliation(s)
- Brittany D Conroy
- Department of Biology, University of Nebraska at Omaha , Omaha, NE , USA
| | - Tyler A Herek
- Department of Biology, University of Nebraska at Omaha , Omaha, NE , USA
| | - Timothy D Shew
- Department of Biology, University of Nebraska at Omaha , Omaha, NE , USA
| | - Matthew Latner
- Department of Biology, University of Nebraska at Omaha , Omaha, NE , USA
| | - Joshua J Larson
- Department of Biology, University of Nebraska at Omaha , Omaha, NE , USA
| | - Laura Allen
- Department of Biology, University of Nebraska at Omaha , Omaha, NE , USA
| | - Paul H Davis
- Department of Biology, University of Nebraska at Omaha , Omaha, NE , USA
| | - Tomáš Helikar
- Department of Biochemistry, University of Nebraska at Lincoln , Lincoln, NE , USA
| | | |
Collapse
|
70
|
Ke Y, Wang X, Jin XY, Solaro RJ, Lei M. PAK1 is a novel cardiac protective signaling molecule. Front Med 2014; 8:399-403. [PMID: 25416031 DOI: 10.1007/s11684-014-0380-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Accepted: 10/15/2014] [Indexed: 12/19/2022]
Abstract
We review here the novel cardiac protective effects of the multifunctional enzyme, p21-activated kinase 1 (PAK1), a member of a serine/threonine protein kinase family. Despite the large body of evidence from studies in noncardiac tissue indicating that PAK1 activity is key in the regulation of a number of cellular functions, the role of PAK1 in the heart has only been revealed over the past few years. In this review, we assemble an overview of the recent findings on PAK1 signaling in the heart, particularly its cardiac protective effects. We present a model for PAK1 signaling that provides a mechanism for specifically affecting cardiac cellular processes in which regulation of protein phosphorylation states by protein phosphatase 2A (PP2A) predominates.We discuss the anti-adrenergic and antihypertrophic cardiac protective effects of PAK1, as well as its role in maintaining ventricular Ca(2+) homeostasis and electrophysiological stability under physiological, β-adrenergic and hypertrophic stress conditions.
Collapse
Affiliation(s)
- Yunbo Ke
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | | | | | | | | |
Collapse
|
71
|
Elsherif L, Ozler M, Zayed MA, Shen JH, Chernoff J, Faber JE, Parise LV. Potential compensation among group I PAK members in hindlimb ischemia and wound healing. PLoS One 2014; 9:e112239. [PMID: 25379771 PMCID: PMC4224450 DOI: 10.1371/journal.pone.0112239] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 10/14/2014] [Indexed: 12/04/2022] Open
Abstract
PAKs are serine/threonine kinases that regulate cytoskeletal dynamics and cell migration. PAK1 is activated by binding to the small EF hand protein, CIB1, or to the Rho GTPases Rac1 or Cdc42. The role of PAK1 in angiogenesis was established based only on in vitro studies and its role in angiogenesis in vivo has never been examined. Here we tested the hypothesis that PAK1 is an essential regulator of ischemic neovascularization (arteriogenesis and angiogenesis) and wound healing using a global PAK1 knockout mouse. Neovascularization was assessed using unilateral hindlimb ischemia. We found that plantar perfusion, limb use and appearance were not significantly different between 6-8 week old PAK1-/- and PAK1+/+ mice throughout the 21-day period following hindlimb ischemia; however a slightly delayed healing was observed in 16 week old PAK1-/- mice. In addition, the wound healing rate, as assessed with an ear punch assay, was unchanged in PAK1-/- mice. Surprisingly, however, we observed a notable increase in PAK2 expression and phosphorylation in ischemic gastrocnemius tissue from PAK1-/- but not PAK1+/+ mice. Furthermore, we observed higher levels of activated ERK2, but not AKT, in ischemic and non-ischemic muscle of PAK1-/- mice upon hindlimb ischemic injury. A group I PAK inhibitor, IPA3, significantly inhibited endothelial cell sprouting from aortic rings in both PAK1-/- and PAK1+/+ mice, implying that PAK2 is a potential contributor to this process. Taken together, our data indicate that while PAK1 has the potential to contribute to neovascularization and wound healing, PAK2 may functionally compensate when PAK1 is deficient.
Collapse
Affiliation(s)
- Laila Elsherif
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Mehmet Ozler
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Mohamed A. Zayed
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Jessica H. Shen
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Jonathan Chernoff
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, United States of America
| | - James E. Faber
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Leslie V. Parise
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| |
Collapse
|
72
|
Verma NK, Kelleher D. Adaptor regulation of LFA-1 signaling in T lymphocyte migration: Potential druggable targets for immunotherapies? Eur J Immunol 2014; 44:3484-99. [PMID: 25251823 DOI: 10.1002/eji.201344428] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 09/16/2014] [Accepted: 09/22/2014] [Indexed: 01/24/2023]
Abstract
The integrin lymphocyte function associated antigen-1 (LFA-1) plays a key role in leukocyte trafficking and in adaptive immune responses through interactions with adhesive ligands, such as ICAM-1. Specific blockade of these interactions has validated LFA-1 as a therapeutic target in many chronic inflammatory diseases, however LFA-1 antagonists have not been clinically successful due to the development of a general immunosuppression, causing fatal side effects. Growing evidence has now established that LFA-1 mediates an array of intracellular signaling pathways by triggering a number of downstream molecules. In this context, a class of multimodular domain-containing proteins capable of recruiting two or more effector molecules, collectively known as "adaptor proteins," has emerged as important mediators in LFA-1 signal transduction. Here, we provide an overview of the adaptor proteins involved in the intracellular signaling cascades by which LFA-1 regulates T-cell motility and immune responses. The complexity of the LFA-1-associated signaling delineated in this review suggests that it may be an important and challenging focus for future research, enabling the identification of "tunable" targets for the development of immunotherapies.
Collapse
Affiliation(s)
- Navin K Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore; Singapore Eye Research Institute, Singapore, Singapore
| | | |
Collapse
|
73
|
Bisson N, Wedlich D, Moss T. The p21-activated kinase Pak1 regulates induction and migration of the neural crest in Xenopus. Cell Cycle 2014; 11:1316-24. [DOI: 10.4161/cc.19685] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
74
|
Abstract
p21-activated kinases are a family of highly conserved protein serine/threonine kinases that are increasingly recognized as playing essential roles in a variety of key signaling processes. Genetic analyses in mice, using constitutive or regulated gene disruption, have provided important new insights into PAK function. In this paper, we review the genetic analysis of all six PAK genes in mice. These data address the singular and redundant functions of the various PAK genes and suggest therapeutic possibilities for small molecule PAK inhibitors or activators.
Collapse
Affiliation(s)
- Mollie L Kelly
- Cancer Biology Program; Fox Chase Cancer Center; Philadelphia, PA USA
| | | |
Collapse
|
75
|
Ribeiro SA, D'Ambrosio MV, Vale RD. Induction of focal adhesions and motility in Drosophila S2 cells. Mol Biol Cell 2014; 25:3861-9. [PMID: 25273555 PMCID: PMC4244196 DOI: 10.1091/mbc.e14-04-0863] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In this study, normally immotile S2 cells are engineered to induce the formation of focal adhesions and cell motility by the transfection of a single gene encoding an integrin subunit. It is demonstrated that the focal adhesions recruit expected components and exhibit mechanosensitive behavior on integrin-ligand substrates of different stiffnesses. Focal adhesions are dynamic structures that interact with the extracellular matrix on the cell exterior and actin filaments on the cell interior, enabling cells to adhere and crawl along surfaces. We describe a system for inducing the formation of focal adhesions in normally non–ECM-adherent, nonmotile Drosophila S2 cells. These focal adhesions contain the expected molecular markers such as talin, vinculin, and p130Cas, and they require talin for their formation. The S2 cells with induced focal adhesions also display a nonpolarized form of motility on vitronectin-coated substrates. Consistent with findings in mammalian cells, the degree of motility can be tuned by changing the stiffness of the substrate and was increased after the depletion of PAK3, a p21-activated kinase. A subset of nonmotile, nonpolarized cells also exhibited focal adhesions that rapidly assembled and disassembled around the cell perimeter. Such cooperative and dynamic fluctuations of focal adhesions were decreased by RNA interference (RNAi) depletion of myosin II and focal adhesion kinase, suggesting that this behavior requires force and focal adhesion maturation. These results demonstrate that S2 cells, a cell line that is well studied for cytoskeletal dynamics and readily amenable to protein manipulation by RNAi, can be used to study the assembly and dynamics of focal adhesions and mechanosensitive cell motility.
Collapse
Affiliation(s)
- Susana A Ribeiro
- Howard Hughes Medical Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158
| | - Michael V D'Ambrosio
- Howard Hughes Medical Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158
| | - Ronald D Vale
- Howard Hughes Medical Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158
| |
Collapse
|
76
|
Evren S, Wen JWH, Luu O, Damm EW, Nagel M, Winklbauer R. EphA4-dependent Brachyury expression is required for dorsal mesoderm involution in the Xenopus gastrula. Development 2014; 141:3649-61. [PMID: 25209247 DOI: 10.1242/dev.111880] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Xenopus provides a well-studied model of vertebrate gastrulation, but a central feature, the movement of the mesoderm to the interior of the embryo, has received little attention. Here, we analyze mesoderm involution at the Xenopus dorsal blastopore lip. We show that a phase of rapid involution - peak involution - is intimately linked to an early stage of convergent extension, which involves differential cell migration in the prechordal mesoderm and a new movement of the chordamesoderm, radial convergence. The latter process depends on Xenopus Brachyury, the expression of which at the time of peak involution is controlled by signaling through the ephrin receptor, EphA4, its ligand ephrinB2 and its downstream effector p21-activated kinase. Our findings support a conserved role for Brachyury in blastopore morphogenesis.
Collapse
Affiliation(s)
- Sevan Evren
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, Ontario, Canada M5S 3G5
| | - Jason W H Wen
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, Ontario, Canada M5S 3G5
| | - Olivia Luu
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, Ontario, Canada M5S 3G5
| | - Erich W Damm
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, Ontario, Canada M5S 3G5
| | - Martina Nagel
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, Ontario, Canada M5S 3G5
| | - Rudolf Winklbauer
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, Ontario, Canada M5S 3G5
| |
Collapse
|
77
|
Koth AP, Oliveira BR, Parfitt GM, Buonocore JDQ, Barros DM. Participation of group I p21-activated kinases in neuroplasticity. ACTA ACUST UNITED AC 2014; 108:270-7. [PMID: 25174326 DOI: 10.1016/j.jphysparis.2014.08.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 04/25/2014] [Accepted: 08/20/2014] [Indexed: 12/15/2022]
Abstract
PAKs are a family of serine/threonine protein kinases activated by small GTPases of the Rho family, including Rac and Cdc42, and are categorized into group I (isoforms 1, 2 and 3) and group II (isoforms 4, 5 and 6). PAK1 and PAK3 are critically involved in biological mechanisms associated with neurodevelopment, neuroplasticity and maturation of the nervous system, and changes in their activity have been detected in pathological disorders, such as Alzheimer's disease, Huntington's disease and mental retardation. The group I PAKs have been associated with neurological processes due to their involvement in intracellular mechanisms that result in molecular and cellular morphological alterations that promote cytoskeletal outgrowth, increasing the efficiency of synaptic transmission. Their substrates in these processes include other intracellular signaling molecules, such as Raf, Mek and LIMK, as well as other components of the cytoskeleton, such as MLC and FLNa. In this review, we describe the characteristics of group I PAKs, such as their molecular structure, mechanisms of activation and importance in the neurobiological processes involved in synaptic plasticity.
Collapse
Affiliation(s)
- André P Koth
- Instituto de Ciências Biológicas (ICB), Programa de Pós-graduação em Ciências Fisiológicas, Fisiologia Animal Comparada, Laboratório de Neurociências, Universidade Federal do Rio Grande (FURG), Av Itália, Km 8, Rio Grande, RS 96203-900, Brazil.
| | - Bruno R Oliveira
- Instituto de Ciências Biológicas (ICB), Programa de Pós-graduação em Ciências Fisiológicas, Fisiologia Animal Comparada, Laboratório de Biologia Molecular, Universidade Federal do Rio Grande (FURG), Av Itália, Km 8, Rio Grande, RS 96203-900, Brazil.
| | - Gustavo M Parfitt
- Instituto de Ciências Biológicas (ICB), Programa de Pós-graduação em Ciências Fisiológicas, Fisiologia Animal Comparada, Laboratório de Neurociências, Universidade Federal do Rio Grande (FURG), Av Itália, Km 8, Rio Grande, RS 96203-900, Brazil.
| | - Juliana de Quadros Buonocore
- Instituto de Ciências Biológicas (ICB), Programa de Pós-graduação em Ciências Fisiológicas, Fisiologia Animal Comparada, Laboratório de Neurociências, Universidade Federal do Rio Grande (FURG), Av Itália, Km 8, Rio Grande, RS 96203-900, Brazil.
| | - Daniela M Barros
- Instituto de Ciências Biológicas (ICB), Programa de Pós-graduação em Ciências Fisiológicas, Fisiologia Animal Comparada, Laboratório de Neurociências, Universidade Federal do Rio Grande (FURG), Av Itália, Km 8, Rio Grande, RS 96203-900, Brazil.
| |
Collapse
|
78
|
Phee H, Au-Yeung BB, Pryshchep O, O'Hagan KL, Fairbairn SG, Radu M, Kosoff R, Mollenauer M, Cheng D, Chernoff J, Weiss A. Pak2 is required for actin cytoskeleton remodeling, TCR signaling, and normal thymocyte development and maturation. eLife 2014; 3:e02270. [PMID: 24843022 PMCID: PMC4017645 DOI: 10.7554/elife.02270] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The molecular mechanisms that govern thymocyte development and maturation are incompletely understood. The P21-activated kinase 2 (Pak2) is an effector for the Rho family GTPases Rac and Cdc42 that regulate actin cytoskeletal remodeling, but its role in the immune system remains poorly understood. In this study, we show that T-cell specific deletion of Pak2 gene in mice resulted in severe T cell lymphopenia accompanied by marked defects in development, maturation, and egress of thymocytes. Pak2 was required for pre-TCR β-selection and positive selection. Surprisingly, Pak2 deficiency in CD4 single positive thymocytes prevented functional maturation and reduced expression of S1P1 and KLF2. Mechanistically, Pak2 is required for actin cytoskeletal remodeling triggered by TCR. Failure to induce proper actin cytoskeletal remodeling impaired PLCγ1 and Erk1/2 signaling in the absence of Pak2, uncovering the critical function of Pak2 as an essential regulator that governs the actin cytoskeleton-dependent signaling to ensure normal thymocyte development and maturation. DOI:http://dx.doi.org/10.7554/eLife.02270.001 T cells are a key element of the immune system. There are many different types of T cells, and they all have their origins in hematopoietic stem cells that are found in the bone marrow. These stem cells leave the bone marrow and circulate in the body until they reach an organ called the thymus, where they become early thymic progenitor cells. These progenitor cells then undergo a process called differentiation to become specific types of T cells, which mature in the thymus before moving to the blood. Although various molecules and mechanisms are known to be involved in the development of T cells, many details of this process are not understood. One group of molecules that has been implicated in the differentiation of T cells is the p21-activated kinases. Kinases are proteins that activate or deactivate other proteins by adding phosphate groups to specific amino acids. Pak2 adds phosphorylate groups to various proteins that are involved in the reorganization of an important structure inside the cell called the cytoskeleton. A kinase called Pak2 has an important role in the reorganization of the cytoskeleton, and since this reorganization is involved in almost all aspects of T cell biology, it seems plausible that Pak2 is also involved in the development of T cells. However, it has not been possible to test this idea because deleting the gene for Pak2 in mice results in their death. Now, Phee et al. have overcome this problem by performing experiments in which the gene for Pak2 was only deleted in T cells. These mice had significantly fewer mature T cells than healthy mice. In particular, the absence of Pak2 in thymocytes (the cells that become T cells) prevented them from maturing into T cells, and also prevented them from producing a receptor protein that is needed for mature T cells to leave the thymus. This work implies that disruption of the Pak2-mediated signaling pathway that regulates the cytoskeleton may weaken the immune system in humans. DOI:http://dx.doi.org/10.7554/eLife.02270.002
Collapse
Affiliation(s)
- Hyewon Phee
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, United States
| | - Byron B Au-Yeung
- Department of Medicine, Division of Rheumatology, University of California, San Francisco, San Francisco, United States
| | - Olga Pryshchep
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, United States
| | - Kyle Leonard O'Hagan
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, United States
| | - Stephanie Grace Fairbairn
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, United States
| | - Maria Radu
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, United States
| | - Rachelle Kosoff
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, United States
| | - Marianne Mollenauer
- Department of Medicine, Division of Rheumatology, University of California, San Francisco, San Francisco, United States
| | - Debra Cheng
- Department of Medicine, Division of Rheumatology, University of California, San Francisco, San Francisco, United States
| | - Jonathan Chernoff
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, United States
| | - Arthur Weiss
- Department of Medicine, Division of Rheumatology, University of California, San Francisco, San Francisco, United States Rosalind Russell Medical Research Center for Arthritis, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
79
|
Cole J, Waurich B, Wensch-Dorendorf M, Bickhart D, Swalve H. A genome-wide association study of calf birth weight in Holstein cattle using single nucleotide polymorphisms and phenotypes predicted from auxiliary traits. J Dairy Sci 2014; 97:3156-72. [DOI: 10.3168/jds.2013-7409] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 01/28/2014] [Indexed: 02/04/2023]
|
80
|
Totaro A, Astro V, Tonoli D, de Curtis I. Identification of two tyrosine residues required for the intramolecular mechanism implicated in GIT1 activation. PLoS One 2014; 9:e93199. [PMID: 24699139 PMCID: PMC3974724 DOI: 10.1371/journal.pone.0093199] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Accepted: 03/03/2014] [Indexed: 11/21/2022] Open
Abstract
GIT1 is an ArfGAP and scaffolding protein regulating cell adhesion and migration. The multidomain structure of GIT1 allows the interaction with several partners. Binding of GIT1 to some of its partners requires activation of the GIT1 polypeptide. Our previous studies indicated that binding of paxillin to GIT1 is enhanced by release of an intramolecular interaction between the amino-terminal and carboxy-terminal portions that keeps the protein in a binding-incompetent state. Here we have addressed the mechanism mediating this intramolecular inhibitory mechanism by testing the effects of the mutation of several formerly identified GIT1 phosphorylation sites on the binding to paxillin. We have identified two tyrosines at positions 246 and 293 of the human GIT1 polypeptide that are needed to keep the protein in the inactive conformation. Interestingly, mutation of these residues to phenylalanine did not affect binding to paxillin, while mutation to either alanine or glutamic acid enhanced binding to paxillin, without affecting the constitutive binding to the Rac/Cdc42 exchange factor βPIX. The involvement of the two tyrosine residues in the intramolecular interaction was supported by reconstitution experiments showing that these residues are important for the binding between the amino-terminal fragment and carboxy-terminal portions of GIT1. Either GIT1 or GIT1-N tyrosine phosphorylation by Src and pervanadate treatment to inhibit protein tyrosine phosphatases did not affect the intramolecular binding between the amino- and carboxy-terminal fragments, nor the binding of GIT1 to paxillin. Mutations increasing the binding of GIT1 to paxillin positively affected cell motility, measured both by transwell migration and wound healing assays. Altogether these results show that tyrosines 246 and 293 of GIT1 are required for the intramolecular inhibitory mechanism that prevents the binding of GIT1 to paxillin. The data also suggest that tyrosine phosphorylation may not be sufficient to release the intramolecular interaction that keeps GIT1 in the inactive conformation.
Collapse
Affiliation(s)
- Antonio Totaro
- Cell Adhesion Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milano, Italy
| | - Veronica Astro
- Cell Adhesion Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milano, Italy
| | - Diletta Tonoli
- Cell Adhesion Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milano, Italy
| | - Ivan de Curtis
- Cell Adhesion Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| |
Collapse
|
81
|
Abstract
p21-Activated protein kinases (PAKs) are centrally involved in a plethora of cellular processes and functions. Their function as effectors of small GTPases Rac1 and Cdc42 has been extensively studied during the past two decades, particularly in the realms of cell proliferation, apoptosis, and hence tumorigenesis, as well as cytoskeletal remodeling and related cellular events in health and disease. In recent years, a large number of studies have shed light onto the fundamental role of group I PAKs, most notably PAK1, in metabolic homeostasis. In skeletal muscle, PAK1 was shown to mediate the function of insulin on stimulating GLUT4 translocation and glucose uptake, while in pancreatic β-cells, PAK1 participates in insulin granule localization and vesicle release. Furthermore, we demonstrated that PAK1 mediates the cross talk between insulin and Wnt/β-catenin signaling pathways and hence regulates gut proglucagon gene expression and the production of the incretin hormone glucagon-like peptide-1 (GLP-1). The utilization of chemical inhibitors of PAK and the characterization of Pak1(-/-) mice enabled us to gain mechanistic insights as well as to assess the overall contribution of PAKs in metabolic homeostasis. This review summarizes our current understanding of PAKs, with an emphasis on the emerging roles of PAK1 in glucose homeostasis.
Collapse
|
82
|
Chen H, Miao J, Li H, Wang C, Li J, Zhu Y, Wang J, Wu X, Qiao H. Expression and prognostic significance of p21-activated kinase 6 in hepatocellular carcinoma. J Surg Res 2014; 189:81-8. [PMID: 24576777 DOI: 10.1016/j.jss.2014.01.049] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Revised: 01/15/2014] [Accepted: 01/24/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND p21-activated protein kinase (PAK) 6 is a serine-threonine kinase belonging to the PAK family. Previous studies have indicated that abnormal expressions of PAK1, PAK2, and PAK5 played critical roles in hepatocellular carcinoma (HCC). Recent studies suggested that deregulation of PAK6 expression played an important role in oncogenesis. To explore the potential roles of PAK6 in HCC, expression of PAK6 was detected in human HCC specimens. METHODS Immunohistochemistry and Western blot analysis were performed for PAK6 in 121 HCC samples. The data were correlated with clinicopathologic features. The univariate and multivariate survival analyses were also performed to determine their clinical prognostic significance. RESULTS PAK6 was overexpressed in HCC as compared with the adjacent noncancerous liver tissues. High expression of PAK6 was associated with Edmondson-Steiner grade (P = 0.006) and number of tumor nodules (P < 0.001), and PAK6 was positively correlated with proliferation marker Ki-67 (P < 0.01). Univariate analysis suggested that PAK6 expression was associated with poor prognosis (P < 0.001). Multivariate analysis indicated that PAK6 and Ki-67 protein expressions were independent prognostic markers for HCC (P = 0.0245 and 0.0331, respectively). CONCLUSIONS Our results suggest that PAK6 overexpression is involved in the pathogenesis of HCC; it may be an independent poor prognostic factor for HCC.
Collapse
Affiliation(s)
- Hongwei Chen
- Department of Clinical Laboratory, The First Hospital of Qinhuangdao, Qinhuangdao, China.
| | - Jinlin Miao
- Department of Magnetic Resonance Imaging, The Fourth People's Hospital of Taizhou, Taizhou, China
| | - Hongchen Li
- Department of Clinical Laboratory, The First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Chunhua Wang
- Department of Clinical Laboratory, The First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Junliang Li
- Department of Clinical Laboratory, The First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Yong Zhu
- Department of Clinical Laboratory, The First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Jianxin Wang
- Department of Clinical Laboratory, The First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Xia Wu
- Department of Clinical Laboratory, The First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Hongying Qiao
- Department of Clinical Laboratory, The First Hospital of Qinhuangdao, Qinhuangdao, China
| |
Collapse
|
83
|
Abstract
p21-Activated kinases (PAKs) are positioned at the nexus of several oncogenic signalling pathways. Overexpression or mutational activation of PAK isoforms frequently occurs in various human tumours, and recent data suggest that excessive PAK activity drives many of the cellular processes that are the hallmarks of cancer. In this Review, we discuss the mechanisms of PAK activation in cancer, the key substrates that mediate the developmental and oncogenic effects of this family of kinases, and how small-molecule inhibitors of these enzymes might be best developed and deployed for the treatment of cancer.
Collapse
Affiliation(s)
- Maria Radu
- Cancer Biology Program; Fox Chase Cancer Center; Philadelphia, PA, USA
| | - Galina Semenova
- Cancer Biology Program; Fox Chase Cancer Center; Philadelphia, PA, USA
| | - Rachelle Kosoff
- Cancer Biology Program; Fox Chase Cancer Center; Philadelphia, PA, USA
- Cancer Biology program, University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan Chernoff
- Cancer Biology Program; Fox Chase Cancer Center; Philadelphia, PA, USA
- To whom correspondence should be addressed: Jonathan Chernoff, Cancer Biology Program, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA 19111, USA, Tel.: (215) 728 5319; Fax: (215) 728 3616;
| |
Collapse
|
84
|
Gadepalli R, Kotla S, Heckle MR, Verma SK, Singh NK, Rao GN. Novel role for p21-activated kinase 2 in thrombin-induced monocyte migration. J Biol Chem 2013; 288:30815-31. [PMID: 24025335 DOI: 10.1074/jbc.m113.463414] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
To understand the role of thrombin in inflammation, we tested its effects on migration of THP-1 cells, a human monocytic cell line. Thrombin induced THP-1 cell migration in a dose-dependent manner. Thrombin induced tyrosine phosphorylation of Pyk2, Gab1, and p115 RhoGEF, leading to Rac1- and RhoA-dependent Pak2 activation. Downstream to Pyk2, Gab1 formed a complex with p115 RhoGEF involving their pleckstrin homology domains. Furthermore, inhibition or depletion of Pyk2, Gab1, p115 RhoGEF, Rac1, RhoA, or Pak2 levels substantially attenuated thrombin-induced THP-1 cell F-actin cytoskeletal remodeling and migration. Inhibition or depletion of PAR1 also blocked thrombin-induced activation of Pyk2, Gab1, p115 RhoGEF, Rac1, RhoA, and Pak2, resulting in diminished THP-1 cell F-actin cytoskeletal remodeling and migration. Similarly, depletion of Gα12 negated thrombin-induced Pyk2, Gab1, p115 RhoGEF, Rac1, RhoA, and Pak2 activation, leading to attenuation of THP-1 cell F-actin cytoskeletal remodeling and migration. These novel observations reveal that thrombin induces monocyte/macrophage migration via PAR1-Gα12-dependent Pyk2-mediated Gab1 and p115 RhoGEF interactions, leading to Rac1- and RhoA-targeted Pak2 activation. Thus, these findings provide mechanistic evidence for the role of thrombin and its receptor PAR1 in inflammation.
Collapse
Affiliation(s)
- Ravisekhar Gadepalli
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | | | | | | | | | | |
Collapse
|
85
|
Structure-based functional site recognition for p21-activated kinase 4. Arch Pharm Res 2013; 36:1494-9. [DOI: 10.1007/s12272-013-0165-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Accepted: 07/25/2011] [Indexed: 01/15/2023]
|
86
|
Porcu G, Parsons AB, Di Giandomenico D, Lucisano G, Mosca MG, Boone C, Ragnini-Wilson A. Combined p21-activated kinase and farnesyltransferase inhibitor treatment exhibits enhanced anti-proliferative activity on melanoma, colon and lung cancer cell lines. Mol Cancer 2013; 12:88. [PMID: 23915247 PMCID: PMC3765434 DOI: 10.1186/1476-4598-12-88] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 07/26/2013] [Indexed: 01/05/2023] Open
Abstract
Background Farnesyltransferase inhibitors (FTIs) are anticancer agents with a spectrum of activity in Ras-dependent and independent tumor cellular and xenograph models. How inhibition of protein farnesylation by FTIs results in reduced cancer cell proliferation is poorly understood due to the multiplicity of potential FTase targets. The low toxicity and oral availability of FTIs led to their introduction into clinical trials for the treatment of breast cancer, hematopoietic malignancy, advanced solid tumor and pancreatic cancer treatment, and Hutchinson-Gilford Progeria Syndrome. Although their efficacy in combinatorial therapies with conventional anticancer treatment for myeloid malignancy and solid tumors is promising, the overall results of clinical tests are far below expectations. Further exploitation of FTIs in the clinic will strongly rely on understanding how these drugs affect global cellular activity. Methods Using FTase inhibitor I and genome-wide chemical profiling of the yeast barcoded deletion strain collection, we identified genes whose inactivation increases the antiproliferative action of this FTI peptidomimetic. The main findings were validated in a panel of cancer cell lines using FTI-277 in proliferation and biochemical assays paralleled by multiparametric image-based analyses. Results ABC transporter Pdr10 or p-21 activated kinase (PAK) gene deletion increases the antiproliferative action of FTase inhibitor I in yeast cells. Consistent with this, enhanced inhibition of cell proliferation by combining group I PAK inhibition, using IPA3, with FTI-277 was observed in melanoma (A375MM), lung (A549) and colon (HT29), but not in epithelial (HeLa) or breast (MCF7), cancer cell lines. Both HeLa and A375MM cells show changes in the nuclear localization of group 1 PAKs in response to FTI-277, but up-regulation of PAK protein levels is observed only in HeLa cells. Conclusions Our data support the view that group I PAKs are part of a pro-survival pathway activated by FTI treatment, and group I PAK inactivation potentiates the anti-proliferative action of FTIs in yeast as well as in cancer cells. These findings open new perspectives for the use of FTIs in combinatorial strategies with PAK inhibitors in melanoma, lung and colon malignancy.
Collapse
Affiliation(s)
- Giampiero Porcu
- Department of Translational Pharmacology, Consorzio Mario Negri Sud, S, Maria Imbaro, Italy
| | | | | | | | | | | | | |
Collapse
|
87
|
Ross AP, Mansilla MA, Choe Y, Helminski S, Sturm R, Maute RL, May SR, Hozyasz KK, Wójcicki P, Mostowska A, Davidson B, Adamopoulos IE, Pleasure SJ, Murray JC, Zarbalis KS. A mutation in mouse Pak1ip1 causes orofacial clefting while human PAK1IP1 maps to 6p24 translocation breaking points associated with orofacial clefting. PLoS One 2013; 8:e69333. [PMID: 23935987 PMCID: PMC3723895 DOI: 10.1371/journal.pone.0069333] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 06/08/2013] [Indexed: 01/05/2023] Open
Abstract
Orofacial clefts are among the most common birth defects and result in an improper formation of the mouth or the roof of the mouth. Monosomy of the distal aspect of human chromosome 6p has been recognized as causative in congenital malformations affecting the brain and cranial skeleton including orofacial clefts. Among the genes located in this region is PAK1IP1, which encodes a nucleolar factor involved in ribosomal stress response. Here, we report the identification of a novel mouse line that carries a point mutation in the Pak1ip1 gene. Homozygous mutants show severe developmental defects of the brain and craniofacial skeleton, including a median orofacial cleft. We recovered this line of mice in a forward genetic screen and named the allele manta-ray (mray). Our findings prompted us to examine human cases of orofacial clefting for mutations in the PAK1IP1 gene or association with the locus. No deleterious variants in the PAK1IP1 gene coding region were recognized, however, we identified a borderline association effect for SNP rs494723 suggesting a possible role for the PAK1IP1 gene in human orofacial clefting.
Collapse
Affiliation(s)
- Adam P. Ross
- Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, California, United States of America
| | - M. Adela Mansilla
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, United States of America
| | - Youngshik Choe
- Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
| | - Simon Helminski
- Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, California, United States of America
| | - Richard Sturm
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Roy L. Maute
- Institute for Cancer Genetics, Columbia University, New York City, New York, United States of America
| | - Scott R. May
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Kamil K. Hozyasz
- Department of Pediatrics, Institute of Mother and Child, Warsaw, Poland
| | - Piotr Wójcicki
- Department of Plastic Surgery, Wrocław Medical University, Polanica Zdroj, Poland
- Department of Plastic Surgery, Medical Centre, Polanica-Zdrój, Poland
| | - Adrianna Mostowska
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, Poznan, Poland
| | - Beth Davidson
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, United States of America
| | - Iannis E. Adamopoulos
- Department of Internal Medicine, University of California Davis, Sacramento, California, United States of America
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, California, United States of America
| | - Samuel J. Pleasure
- Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
| | - Jeffrey C. Murray
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, United States of America
| | - Konstantinos S. Zarbalis
- Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, California, United States of America
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, California, United States of America
| |
Collapse
|
88
|
Bassani S, Zapata J, Gerosa L, Moretto E, Murru L, Passafaro M. The neurobiology of X-linked intellectual disability. Neuroscientist 2013; 19:541-52. [PMID: 23820068 DOI: 10.1177/1073858413493972] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
X-linked intellectual disability (XLID) affects 1% to 3% of the population. XLID subsumes several heterogeneous conditions, all of which are marked by cognitive impairment and reduced adaptive skills. XLID arises from mutations on the X chromosome; to date, 102 XLID genes have been identified. The proteins encoded by XLID genes are involved in higher brain functions, such as cognition, learning and memory, and their molecular role is the subject of intense investigation. Here, we review recent findings concerning a representative group of XLID proteins: the fragile X mental retardation protein; methyl-CpG-binding protein 2 and cyclin-dependent kinase-like 5 proteins, which are involved in Rett syndrome; the intracellular signaling molecules of the Rho guanosine triphosphatases family; and the class of cell adhesion molecules. We discuss how XLID gene mutations affect the structure and function of synapses.
Collapse
Affiliation(s)
- Silvia Bassani
- CNR Institute of Neuroscience, Department BIOMETRA, University of Milan, Milan, Italy
| | | | | | | | | | | |
Collapse
|
89
|
Radu M, Rawat SJ, Beeser A, Iliuk A, Tao WA, Chernoff J. ArhGAP15, a Rac-specific GTPase-activating protein, plays a dual role in inhibiting small GTPase signaling. J Biol Chem 2013; 288:21117-21125. [PMID: 23760270 DOI: 10.1074/jbc.m113.459719] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Signaling from small GTPases is a tightly regulated process. In this work we used a protein microarray screen to identify the Rac-specific GAP, ArhGAP15, as a substrate of the Rac effectors Pak1 and Pak2. In addition to serving as a substrate of Pak1/2, we found that ArhGAP15, via its PH domain, bound to these kinases. The association of ArhGAP15 to Pak1/2 resulted in mutual inhibition of GAP and kinase catalytic activity, respectively. Knock-down of ArhGAP15 resulted in activation of Pak1/2, both indirectly, as a result of Rac activation, and directly, as a result of disruption of the ArhGAP15/Pak complex. Our data suggest that ArhGAP15 plays a dual negative role in regulating small GTPase signaling, by acting at the level of the GTPase itself, as well interacting with its effector, Pak kinase.
Collapse
Affiliation(s)
- Maria Radu
- From the Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111
| | - Sonali J Rawat
- the Department of Biochemistry and Molecular Biology, Drexel University, Philadelphia, Pennsylvania 19102, and
| | - Alexander Beeser
- From the Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111
| | - Anton Iliuk
- the Department of Biochemistry, Purdue University, West Lafayette, Indiana 47907
| | - Weiguo Andy Tao
- the Department of Biochemistry, Purdue University, West Lafayette, Indiana 47907
| | - Jonathan Chernoff
- From the Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111,.
| |
Collapse
|
90
|
Kundumani-Sridharan V, Singh NK, Kumar S, Gadepalli R, Rao GN. Nuclear factor of activated T cells c1 mediates p21-activated kinase 1 activation in the modulation of chemokine-induced human aortic smooth muscle cell F-actin stress fiber formation, migration, and proliferation and injury-induced vascular wall remodeling. J Biol Chem 2013; 288:22150-62. [PMID: 23737530 DOI: 10.1074/jbc.m113.454082] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Recent literature suggests that cyclin-dependent kinases (CDKs) mediate cell migration. However, the mechanisms were not known. Therefore, the objective of this study is to test whether cyclin/CDKs activate Pak1, an effector of Rac1, whose involvement in the modulation of cell migration and proliferation is well established. Monocyte chemotactic protein 1 (MCP1) induced Pak1 phosphorylation/activation in human aortic smooth muscle cells (HASMCs) in a delayed time-dependent manner. MCP1 also stimulated F-actin stress fiber formation in a delayed manner in HASMCs, as well as the migration and proliferation of these cells. Inhibition of Pak1 suppressed MCP1-induced HASMC F-actin stress fiber formation, migration, and proliferation. MCP1 induced cyclin D1 expression as well as CDK6 and CDK4 activities, and these effects were dependent on activation of NFATc1. Depletion of NFATc1, cyclin D1, CDK6, or CDK4 levels attenuated MCP1-induced Pak1 phosphorylation/activation and resulted in decreased HASMC F-actin stress fiber formation, migration, and proliferation. CDK4, which appeared to be activated downstream of CDK6, formed a complex with Pak1 in response to MCP1. MCP1 also activated Rac1 in a time-dependent manner, and depletion/inhibition of its levels/activation abrogated MCP1-induced NFATc1-cyclin D1-CDK6-CDK4-Pak1 signaling and, thereby, decreased HASMC F-actin stress fiber formation, migration, and proliferation. In addition, smooth muscle-specific deletion of NFATc1 led to decreased cyclin D1 expression and CDK6, CDK4, and Pak1 activities, resulting in reduced neointima formation in response to injury. Thus, these observations reveal that Pak1 is a downstream effector of CDK4 and Rac1-dependent, NFATc1-mediated cyclin D1 expression and CDK6 activity mediate this effect. In addition, smooth muscle-specific deletion of NFATc1 prevented the capacity of vascular smooth muscle cells for MCP-1-induced activation of the cyclin D1-CDK6-CDK4-Pak1 signaling axis, affecting their migration and proliferation in vitro and injury-induced neointima formation in vivo.
Collapse
|
91
|
Arias-Romero LE, Villamar-Cruz O, Huang M, Hoeflich KP, Chernoff J. Pak1 kinase links ErbB2 to β-catenin in transformation of breast epithelial cells. Cancer Res 2013; 73:3671-82. [PMID: 23576562 DOI: 10.1158/0008-5472.can-12-4453] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
p21-Activated kinase-1 (Pak1) is frequently upregulated in human breast cancer and is required for transformation of mammary epithelial cells by ErbB2. Here, we show that loss of Pak1, but not the closely related Pak2, leads to diminished expression of β-catenin and its target genes. In MMTV-ErbB2 transgenic mice, loss of Pak1 prolonged survival, and mammary tissues of such mice showed loss of β-catenin. Expression of a β-catenin mutant bearing a phospho-mimetic mutation at Ser 675, a specific Pak1 phosphorylation site, restored transformation to ErbB2-positive, Pak1-deficient mammary epithelial cells. Mice bearing xenografts of ErbB2-positive breast cancer cells showed tumor regression when treated with small-molecule inhibitors of Pak or β-catenin, and combined inhibition by both agents was synergistic. These data delineate a signaling pathway from ErbB2 to Pak to β-catenin that is required for efficient transformation of mammary epithelial cells, and suggest new therapeutic strategies in ErbB2-positive breast cancer.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Apoptosis/genetics
- Blotting, Western
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/prevention & control
- Cell Line
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Cell Survival/genetics
- Cell Transformation, Neoplastic/drug effects
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Dose-Response Relationship, Drug
- Epithelial Cells/drug effects
- Epithelial Cells/metabolism
- Humans
- Mice
- Mice, Inbred ICR
- Mice, Knockout
- Mice, SCID
- Mice, Transgenic
- Pyrazoles/pharmacology
- Pyrroles/pharmacology
- RNA Interference
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Xenograft Model Antitumor Assays
- beta Catenin/genetics
- beta Catenin/metabolism
- p21-Activated Kinases/genetics
- p21-Activated Kinases/metabolism
Collapse
Affiliation(s)
- Luis E Arias-Romero
- Cancer Biology Program and Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| | | | | | | | | |
Collapse
|
92
|
Wang B, Ma W, Xu X, Wang C, Zhu Y, An N, An L, Wu Z, Tian J. Phosphorylation of histone H3 on Ser10 by auto-phosphorylated PAK1 is not essential for chromatin condensation and meiotic progression in porcine oocytes. J Anim Sci Biotechnol 2013; 4:13. [PMID: 23521812 PMCID: PMC3639857 DOI: 10.1186/2049-1891-4-13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 03/01/2013] [Indexed: 11/10/2022] Open
Abstract
Background The p21-activated kinase 1 (PAK1) is essential for mitosis and plays an important role in the regulation of microtubule assembly during oocyte meiotic maturation in mice; however, little is known about its role in porcine oocytes. Result Total p21-activated kinase 1 (PAK1) and phosphorylated PAK1 at Thr423 (PAK1Thr423) were consistently expressed in porcine oocytes from the germinal vesicle (GV) to the second metaphase (MII) stages, but phosphorylation of histone H3 at Ser10 (H3Ser10) was only expressed after the GV stage. Immunofluorescence analysis revealed that PAK1Thr423 and H3Ser10 colocalized on chromosomes after the GV stage. Blocking of endogenous PAK1Thr423 by injecting a specific antibody decreased the phosphorylation level of H3Ser10; however, it had no impact on chromatin condensation, meiotic progression, cleavage rate of blastomeres or the rate of blastocyst formation. Conclusion Phosphorylation of PAK1Thr423 is a spontaneous activation process and the activated PAK1Thr423 can promote the phosphorylation of H3Ser10; however, this pathway is not required for meiotic maturation of porcine oocytes or early embryonic development.
Collapse
Affiliation(s)
- Bingyuan Wang
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
| | - Wei Ma
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China.,Department of Histology and Embryology; School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, P. R. China
| | - Xiaoling Xu
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China.,Institute of Animal Husbandry and Veterinary Medicine, Beijing Municipal Academy of Agriculture and Forestry Sciences, Beijing, 100097, P. R. China
| | - Chao Wang
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
| | - Yubo Zhu
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
| | - Na An
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
| | - Lei An
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
| | - Zhonghong Wu
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
| | - Jianhui Tian
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
| |
Collapse
|
93
|
Peters EC, Gossett AJ, Goldstein B, Der CJ, Reiner DJ. Redundant canonical and noncanonical Caenorhabditis elegans p21-activated kinase signaling governs distal tip cell migrations. G3 (BETHESDA, MD.) 2013; 3:181-95. [PMID: 23390595 PMCID: PMC3564979 DOI: 10.1534/g3.112.004416] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 11/27/2012] [Indexed: 11/18/2022]
Abstract
p21-activated kinases (Paks) are prominent mediators of Rac/Cdc42-dependent and -independent signaling and regulate signal transduction and cytoskeletal-based cell movements. We used the reproducible migrations of the Caenorhabditis elegans gonadal distal tip cells to show that two of the three nematode Pak proteins, MAX-2 and PAK-1, function redundantly in regulation of cell migration but are regulated by very different mechanisms. First, we suggest that MAX-2 requires CED-10/Rac function and thus functions canonically. Second, PIX-1 and GIT-1 function in the same role as PAK-1, and PAK-1 interaction with PIX-1 is required for PAK-1 activity; thus, PAK-1 functions noncanonically. The human Pak-Pix-Git complex is central to noncanonical Pak signaling and requires only modest Rac/CDC-42 input. Unlike the human complex, our results suggest that the C. elegans Pak-Pix-Git complex requires PAK-1 kinase domain activity. This study delineates signaling network relationships in this cell migration model, thus providing potential further mechanistic insights and an assessment of total Pak contribution to cell migration events.
Collapse
Affiliation(s)
- Eldon C. Peters
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Andrea J. Gossett
- Department of Biology, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Bob Goldstein
- Department of Biology, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Channing J. Der
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina 27599
| | - David J. Reiner
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina 27599
| |
Collapse
|
94
|
The Rac GTPase effector p21-activated kinase is essential for hematopoietic stem/progenitor cell migration and engraftment. Blood 2013; 121:2474-82. [PMID: 23335370 DOI: 10.1182/blood-2012-10-460709] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The p21-activated kinases (Paks) are serine/threonine kinases that are major effectors of the Rho guanosine 5'\x{2011}triphosphatase, Rac, and Cdc42. Rac and Cdc42 are known regulators of hematopoietic stem and progenitor cell (HSPC) function, however, a direct role for Paks in HSPCs has yet to be elucidated. Lin(-)Sca1(+)c-kit(+) (LSK) cells from wild-type mice were transduced with retrovirus expressing Pak inhibitory domain (PID), a well-characterized inhibitor of Pak activation. Defects in marrow homing and in vitro cell migration, assembly of the actin cytoskeleton, proliferation, and survival were associated with engraftment failure of PID-LSK. The PID-LSK demonstrated decreased phosphorylation of extracellular signal-regulated kinase (ERK), whereas constitutive activation of ERK in these cells led to rescue of hematopoietic progenitor cell proliferation in vitro and partial rescue of Pak-deficient HSPC homing and engraftment in vivo. Using conditional knock-out mice, we demonstrate that among group A Paks, Pak2(-/-) HSPC show reduced homing to the bone marrow and altered cell shape similar to PID-LSK cells in vitro and are completely defective in HSPC engraftment. These data demonstrate that Pak proteins are key components of multiple engraftment-associated HSPC functions and play a direct role in activation of ERK in HSPCs, and that Pak2 is specifically essential for HSPC engraftment.
Collapse
|
95
|
Chiang YA, Shao W, Xu XX, Chernoff J, Jin T. P21-activated protein kinase 1 (Pak1) mediates the cross talk between insulin and β-catenin on proglucagon gene expression and its ablation affects glucose homeostasis in male C57BL/6 mice. Endocrinology 2013. [PMID: 23183186 DOI: 10.1210/en.2012-1781] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In gut endocrine L cells, the Wnt signaling pathway effector β-catenin (β-cat)/transcription factor 7-like 2 mediates the stimulatory effect of insulin on proglucagon (gcg) expression and glucagon-like peptide-1 (GLP-1) production. In several other cell lineages, insulin is able to stimulate p21-activated protein kinase 1 (Pak1). Here we determined the role of Pak1 in gcg expression and the effect of Pak1 deletion on glucose homeostasis. Insulin stimulated Pak1 activation through increasing its Thr423 phosphorylation in gut gcg-expressing cell lines, associated with increased gcg mRNA levels. This stimulation was attenuated by the Pak inhibitor 2,2'-dihydroxy-1,1'-dinaphthyldisulfide (IPA3) or dominant-negative Pak1. Both insulin and cAMP-promoting agents activated β-cat Ser675 phosphorylation, which was attenuated by IPA3 or protein kinase A inhibition, respectively. Gut gcg levels were reduced in male Pak1(-/-) mice, associated with impaired glucose tolerance after an ip or oral glucose challenge. These mice had lower circulating active GLP-1 levels after a glucose challenge as well as reduced distal ileum GLP-1 content after insulin treatment. Finally, the Pak1(-/-) mice exhibited reduced brainstem gcg level and abolished β-cat Ser675 phosphorylation in brain neurons after insulin treatment. We suggest that Pak1 mediates the cross talk between insulin and Wnt signaling pathways on gut and brain gcg expression, and its ablation impairs glucose homeostasis.
Collapse
Affiliation(s)
- Yuting Alex Chiang
- Departments of Physiology, University of Toronto, Ontario, Canada M5S 1A8
| | | | | | | | | |
Collapse
|
96
|
Kosoff R, Chow HY, Radu M, Chernoff J. Pak2 kinase restrains mast cell FcεRI receptor signaling through modulation of Rho protein guanine nucleotide exchange factor (GEF) activity. J Biol Chem 2012. [PMID: 23204526 DOI: 10.1074/jbc.m112.422295] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
p21-activated kinase-1 (Pak1) is a serine/threonine kinase that plays a key role in mediating antigen-stimulated extracellular calcium influx and degranulation in mast cells. Another isoform in this kinase family, Pak2, is expressed at very high levels in mast cells, but its function is unknown. Here we show that Pak2 loss in murine bone marrow-derived mast cells, unlike loss of Pak1, induces increased antigen-mediated adhesion, degranulation, and cytokine secretion without changes to extracellular calcium influx. This phenotype is associated with an increase in RhoA-GTPase signaling activity to downstream effectors, including myosin light chain and p38(MAPK), and is reversed upon treatment with a Rho-specific inhibitor. Pak2, but not Pak1, negatively regulates RhoA via phosphorylation of the guanine nucleotide exchange factor GEF-H1 at an inhibitory site, leading to increased GEF-H1 microtubule binding and loss of RhoA stimulation. These data suggest that Pak2 plays a unique inhibitory role in mast cell degranulation by down-regulating RhoA via GEF-H1.
Collapse
Affiliation(s)
- Rachelle Kosoff
- Cancer Biology Program, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | |
Collapse
|
97
|
Staser K, Shew MA, Michels EG, Mwanthi MM, Yang FC, Clapp DW, Park SJ. A Pak1-PP2A-ERM signaling axis mediates F-actin rearrangement and degranulation in mast cells. Exp Hematol 2012; 41:56-66.e2. [PMID: 23063725 DOI: 10.1016/j.exphem.2012.10.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 10/01/2012] [Accepted: 10/06/2012] [Indexed: 01/12/2023]
Abstract
Mast cells coordinate allergy and allergic asthma and are crucial cellular targets in therapeutic approaches to inflammatory disease. Allergens cross-link immunoglobulin E bound at high-affinity receptors on the mast cell's surface, causing release of preformed cytoplasmic granules containing inflammatory molecules, including histamine, a principal effector of fatal septic shock. Both p21 activated kinase 1 (Pak1) and protein phosphatase 2A (PP2A) modulate mast cell degranulation, but the molecular mechanisms underpinning these observations and their potential interactions in common or disparate pathways are unknown. In this study, we use genetic and other approaches to show that Pak1's kinase-dependent interaction with PP2A potentiates PP2A's subunit assembly and activation. PP2A then dephosphorylates threonine 567 of Ezrin/Radixin/Moesin (ERM) molecules that have been shown to couple F-actin to the plasma membrane in other cell systems. In our study, the activity of this Pak1-PP2A-ERM axis correlates with impaired systemic histamine release in Pak1(-/-) mice and defective F-actin rearrangement and impaired degranulation in Ezrin disrupted (Mx1Cre(+)Ezrin(flox/flox)) primary mast cells. This heretofore unknown mechanism of mast cell degranulation provides novel therapeutic targets in allergy and asthma and may inform studies of kinase regulation of cytoskeletal dynamics in other cell lineages.
Collapse
Affiliation(s)
- Karl Staser
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | | | | | | | |
Collapse
|
98
|
Watari A. [Nature of cancer explored from the perspective of the functional evolution of proto-oncogenes]. YAKUGAKU ZASSHI 2012; 132:1165-70. [PMID: 23037702 DOI: 10.1248/yakushi.12-00204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The products of proto-oncogene play critical roles in the development or maintenance of multicellular societies in animals via strict regulatory systems. When these regulatory systems are disrupted, proto-oncogenes can become oncogenes, and thereby induce cell transformation and carcinogenesis. To understand the molecular basis for development of the regulatory system of proto-oncogenes during evolution, we screened for ancestral proto-oncogenes from the unicellular choanoflagellate Monosiga ovata (M. ovata) by monitoring their transforming ability in mammalian cells; consequently, we isolated a Pak gene ortholog, which encodes a serine/threonine kinase as a 'primitive oncogene'. We also cloned Pak orthologs from fungi and the multicellular sponge Ephydatia fluviatilis, and compared their regulatory features with that of M. ovata Pak (MoPak). MoPak is constitutively active and induces cell transformation in mammalian cells. In contrast, Pak orthologs from multicellular animals are strictly regulated. Analyses of Pak mutants revealed that structural alterations in the auto-inhibitory domain (AID) are responsible for the enhanced kinase activity and the oncogenic activity of MoPak. Furthermore, we show that Rho family GTPases-mediated regulatory system of Pak kinase is conserved throughout the evolution from unicellular to multicellular animals, but the MoPak is more sensitive to the Rho family GTPases-mediated activation than multicellular Pak. These results show that maturation of AID function was required for the development of the strict regulatory system of the Pak proto-oncogene, and support the potential link between the development of the regulatory system of proto-oncogenes and the evolution of multicellularity. Further analysis of oncogenic functions of proto-oncogene orthologs in the unicellular genes would provide some insights into the mechanisms of the destruction of multicellular society in cancer.
Collapse
Affiliation(s)
- Akihiro Watari
- Department of Bio-Functional Molecular Chemistry, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, Japan.
| |
Collapse
|
99
|
Chow HY, Jubb AM, Koch JN, Jaffer ZM, Stepanova D, Campbell DA, Duron SG, O'Farrell M, Cai KQ, Klein-Szanto AJP, Gutkind JS, Hoeflich KP, Chernoff J. p21-Activated kinase 1 is required for efficient tumor formation and progression in a Ras-mediated skin cancer model. Cancer Res 2012; 72:5966-75. [PMID: 22983922 DOI: 10.1158/0008-5472.can-12-2246] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The RAS genes are the most commonly mutated oncogenes in human cancer and present a particular therapeutic dilemma, as direct targeting of Ras proteins by small molecules has proved difficult. Signaling pathways downstream of Ras, in particular Raf/Mek/Erk and PI3K/Akt/mTOR, are dominated by lipid and protein kinases that provide attractive alternate targets in Ras-driven tumors. As p21-activated kinase 1 (Pak1) has been shown to regulate both these signaling pathways and is itself upregulated in many human cancers, we assessed the role of Pak1 in Ras-driven skin cancer. In human squamous cell carcinoma (SCC), we found a strong positive correlation between advanced stage and grade and PAK1 expression. Using a mouse model of Kras-driven SCC, we showed that deletion of the mouse Pak1 gene led to markedly decreased tumorigenesis and progression, accompanied by near total loss of Erk and Akt activity. Treatment of Kras(G12D) mice with either of two distinct small molecule Pak inhibitors (PF3758309 and FRAX597) caused tumor regression and loss of Erk and Akt activity. Tumor regression was also seen in mice treated with a specific Mek inhibitor, but not with an Akt inhibitor. These findings establish Pak1 as a new target in KRAS-driven tumors and suggest a mechanism of action through the Erk, but not the Akt, signaling pathway.
Collapse
Affiliation(s)
- Hoi Yee Chow
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Hough C, Radu M, Doré JJE. Tgf-beta induced Erk phosphorylation of smad linker region regulates smad signaling. PLoS One 2012; 7:e42513. [PMID: 22880011 PMCID: PMC3412844 DOI: 10.1371/journal.pone.0042513] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 07/09/2012] [Indexed: 01/17/2023] Open
Abstract
The Transforming Growth Factor-Beta (TGF-β) family is involved in regulating a variety of cellular processes such as apoptosis, differentiation, and proliferation. TGF-β binding to a Serine/Threonine kinase receptor complex causes the recruitment and subsequent activation of transcription factors known as smad2 and smad3. These proteins subsequently translocate into the nucleus to negatively or positively regulate gene expression. In this study, we define a second signaling pathway leading to TGF-β receptor activation of Extracellular Signal Regulated Kinase (Erk) in a cell-type dependent manner. TGF-β induced Erk activation was found in phenotypically normal mesenchymal cells, but not normal epithelial cells. By activating phosphotidylinositol 3-kinase (PI3K), TGF-β stimulates p21-activated kinase2 (Pak2) to phosphorylate c-Raf, ultimately resulting in Erk activation. Activation of Erk was necessary for TGF-β induced fibroblast replication. In addition, Erk phosphorylated the linker region of nuclear localized smads, resulting in increased half-life of C-terminal phospho-smad 2 and 3 and increased duration of smad target gene transcription. Together, these data show that in mesenchymal cell types the TGF-β/PI3K/Pak2/Raf/MEK/Erk pathway regulates smad signaling, is critical for TGF-β-induced growth and is part of an integrated signaling web containing multiple interacting pathways rather than discrete smad/non-smad pathways.
Collapse
Affiliation(s)
- Chris Hough
- BioMedical Sciences, Memorial University, St. John's, Newfoundland, Canada
| | - Maria Radu
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Jules J. E. Doré
- BioMedical Sciences, Memorial University, St. John's, Newfoundland, Canada
- * E-mail:
| |
Collapse
|