151
|
Locatelli A, Lange CA. Met receptors induce Sam68-dependent cell migration by activation of alternate extracellular signal-regulated kinase family members. J Biol Chem 2011; 286:21062-72. [PMID: 21489997 DOI: 10.1074/jbc.m110.211409] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The hepatocyte growth factor (HGF)/Met receptor signaling pathway is deregulated in diverse human malignancies and plays a central role in oncogenesis, tumor progression, and invasive cancer growth. Similarly, altered expression and splicing (i.e. inclusion of variant exon 5, "v5") of the cell adhesion marker, CD44, is associated with advanced cancer phenotypes. We sought to further understand how HGF regulates CD44v5 expression. Immortalized nontumorigenic keratinocyte (HaCaT) cells abundantly express both Met receptors and CD44v5 transmembrane glycoproteins. HGF stimulated CD44v5 protein expression and HaCaT cell migration; these events required activation of the ERK1/2 MAPK module and Sam68, a protein involved in RNA processing, splicing, and v5 inclusion. Similar to HaCaT cells, highly migratory MDA-MB-231 breast cancer cells also required Sam68 expression for HGF-induced migration. However, MDA-MB-231 cell migration occurred independently of ERK1/2 and CD44v5 expression and instead required ERK5 signaling to Sam68. Phospho-mutant, but not WT-Sam68, blocked HGF-induced cell migration in both cell types; MDA-MB-435 cells behaved similarly. These results suggest that Sam68 acts as a convergence point for ERK signaling to cell migration; blockade of phospho-Sam68 may provide a new avenue for therapeutic inhibition of metastatic cancers.
Collapse
Affiliation(s)
- Alessia Locatelli
- Department of Medicine (Division of Hematology, Oncology, and Transplantation), Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | |
Collapse
|
152
|
Sunadome K, Yamamoto T, Ebisuya M, Kondoh K, Sehara-Fujisawa A, Nishida E. ERK5 regulates muscle cell fusion through Klf transcription factors. Dev Cell 2011; 20:192-205. [PMID: 21316587 DOI: 10.1016/j.devcel.2010.12.005] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Revised: 11/26/2010] [Accepted: 12/10/2010] [Indexed: 01/05/2023]
Abstract
In skeletal muscle differentiation, muscle-specific genes are regulated by two groups of transcription factors, the MyoD and MEF2 families, which work together to drive the differentiation process. Here, we show that ERK5 regulates muscle cell fusion through Klf transcription factors. The inhibition of ERK5 activity suppresses muscle cell fusion with minimal effects on the expression of MyoD, MEF2, and their target genes. Promoter analysis coupled to microarray assay reveals that Klf-binding motifs are highly enriched in the promoter regions of ERK5-dependent upregulated genes. Remarkably, Klf2 and Klf4 expression are also upregulated during differentiation in an ERK5-dependent manner, and knockdown of Klf2 or Klf4 specifically suppresses muscle cell fusion. Moreover, we show that Sp1 transcription factor links ERK5 to Klf2/4, and that nephronectin, a Klf transcriptional target, is involved in muscle cell fusion. Therefore, an ERK5/Sp1/Klf module plays a key role in the fusion process during skeletal muscle differentiation.
Collapse
Affiliation(s)
- Kazunori Sunadome
- Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan
| | | | | | | | | | | |
Collapse
|
153
|
Vertebrate paralogous MEF2 genes: origin, conservation, and evolution. PLoS One 2011; 6:e17334. [PMID: 21394201 PMCID: PMC3048864 DOI: 10.1371/journal.pone.0017334] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2010] [Accepted: 01/31/2011] [Indexed: 01/04/2023] Open
Abstract
Background The myocyte enhancer factor 2 (MEF2) gene family is broadly expressed during the development and maintenance of muscle cells. Although a great deal has been elucidated concerning MEF2 transcription factors' regulation of specific gene expression in diverse programs and adaptive responses, little is known about the origin and evolution of the four members of the MEF2 gene family in vertebrates. Methodology/Principal Findings By phylogenetic analyses, we investigated the origin, conservation, and evolution of the four MEF2 genes. First, among the four MEF2 paralogous branches, MEF2B is clearly distant from the other three branches in vertebrates, mainly because it lacks the HJURP_C (Holliday junction recognition protein C-terminal) region. Second, three duplication events might have occurred to produce the four MEF2 paralogous genes and the latest duplication event occurred near the origin of vertebrates producing MEF2A and MEF2C. Third, the ratio (Ka/Ks) of non-synonymous to synonymous nucleotide substitution rates showed that MEF2B evolves faster than the other three MEF2 proteins despite purifying selection on all of the four MEF2 branches. Moreover, a pair model of M0 versus M3 showed that variable selection exists among MEF2 proteins, and branch-site analysis presented that sites 53 and 64 along the MEF2B branch are under positive selection. Finally, and interestingly, substitution rates showed that type II MADS genes (i.e., MEF2-like genes) evolve as slowly as type I MADS genes (i.e., SRF-like genes) in animals, which is inconsistent with the fact that type II MADS genes evolve much slower than type I MADS genes in plants. Conclusion Our findings shed light on the relationship of MEF2A, B, C, and D with functional conservation and evolution in vertebrates. This study provides a rationale for future experimental design to investigate distinct but overlapping regulatory roles of the four MEF2 genes in various tissues.
Collapse
|
154
|
Wattenberg EV. Modulation of protein kinase signaling cascades by palytoxin. Toxicon 2011; 57:440-8. [PMID: 21070801 PMCID: PMC3072206 DOI: 10.1016/j.toxicon.2010.11.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 10/26/2010] [Accepted: 11/02/2010] [Indexed: 10/18/2022]
Abstract
Although known for its acutely toxic action, palytoxin has also been identified as a type of carcinogenic agent called a tumor promoter. In general tumor promoters do not damage DNA, but instead contribute to carcinogenesis by disrupting the regulation of cellular signaling. The identification of palytoxin as a tumor promoter, together with the recognition that the Na(+), K(+)-ATPase is its receptor, led to research on how palytoxin triggers the modulation of signal transduction pathways. This review focuses on mitogen activated protein (MAP) kinases as mediators of palytoxin-stimulated signaling. MAP kinases are a family of serine/threonine kinases that relay a variety of signals to the cellular machinery that regulates cell fate and function. The studies discussed in this review investigated how palytoxin stimulates MAP kinase activity and, in turn, how MAP kinases mediate the response of cells to palytoxin.
Collapse
Affiliation(s)
- Elizabeth V Wattenberg
- Division of Environmental Health Sciences, School of Public Health, University of Minnesota, Mayo Mail Code #807, 420 Delaware Street SE, Minneapolis, MN 55455, USA.
| |
Collapse
|
155
|
Ramsay AK, McCracken SRC, Soofi M, Fleming J, Yu AX, Ahmad I, Morland R, Machesky L, Nixon C, Edwards DR, Nuttall RK, Seywright M, Marquez R, Keller E, Leung HY. ERK5 signalling in prostate cancer promotes an invasive phenotype. Br J Cancer 2011; 104:664-72. [PMID: 21266977 PMCID: PMC3049582 DOI: 10.1038/sj.bjc.6606062] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 11/19/2010] [Accepted: 11/29/2010] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Aberrant mitogen/extracellular signal-regulated kinase 5 (MEK5)-extracellular signal-regulated protein kinase 5 (ERK5)-mediated signalling has been implicated in a number of tumour types including prostate cancer (PCa). The molecular basis of ERK5-driven carcinogenesis and its clinical relevance remain to be fully characterised. METHODS Modulation of ERK5 expression or function in human PCa PC3 and PC3-ERK5 (stably transfected with ERK5) cells was performed using siRNA-mediated knockdown or the MEK inhibitor PD18435 respectively. In vitro significance of ERK5 signalling was assessed by assays for proliferation, motility, invasion and invadopodia. Expression of matrix metalloproteinases/tissue inhibitors of metalloproteases was determined by Q-RT-PCR. Extracellular signal-regulated protein kinase 5 expression in primary and metastatic PCa was examined using immunohistochemistry. RESULTS Reduction of ERK5 expression or signalling significantly inhibited the motility and invasive capability of PC3 cells. Extracellular signal-regulated protein kinase 5-mediated signalling significantly promoted formation of in vivo metastasis in an orthotopic PCa model (P<0.05). Invadopodia formation was also enhanced by forced ERK5 expression in PC3 cells. Furthermore, in metastatic PCa, nuclear ERK5 immunoreactivity was significantly upregulated when compared with benign prostatic hyperplasia and primary PCa (P=0.013 and P<0.0001, respectively). CONCLUSION Our in vitro, in vivo and clinical data support an important role for the MEK5-ERK5 signalling pathway in invasive PCa, which represents a potential target for therapy in primary and metastatic PCa.
Collapse
Affiliation(s)
- A K Ramsay
- Institute for Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1BD, UK
- Urology Research, The Beatson Institute for Cancer Research, Glasgow Centre for Cancer Research, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - S R C McCracken
- Urology Research Group, Northern Institute for Cancer Research, Medical School, University of Newcastle Upon Tyne, Framlington Place, Newcastle Upon Tyne NE2 4HH, UK
| | - M Soofi
- Institute for Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1BD, UK
- Urology Research, The Beatson Institute for Cancer Research, Glasgow Centre for Cancer Research, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - J Fleming
- Urology Research, The Beatson Institute for Cancer Research, Glasgow Centre for Cancer Research, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - A X Yu
- Urology Research, The Beatson Institute for Cancer Research, Glasgow Centre for Cancer Research, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - I Ahmad
- Institute for Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1BD, UK
- Urology Research, The Beatson Institute for Cancer Research, Glasgow Centre for Cancer Research, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - R Morland
- Institute for Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1BD, UK
- Urology Research, The Beatson Institute for Cancer Research, Glasgow Centre for Cancer Research, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - L Machesky
- Urology Research, The Beatson Institute for Cancer Research, Glasgow Centre for Cancer Research, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - C Nixon
- Urology Research, The Beatson Institute for Cancer Research, Glasgow Centre for Cancer Research, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - D R Edwards
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - R K Nuttall
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - M Seywright
- Department of Pathology, Western Infirmary, NHS Greater Glasgow and Clyde, Glasgow G11 6NT, UK
| | - R Marquez
- WestCHEM, Department of Chemistry, University of Glasgow, Glasgow G12 8QQ, UK
| | - E Keller
- Departments of Urology and Pathology, Michigan University, 1500 East Medical Center Drive, 5304 CCGC, Ann Arbor, MI 48109, USA
| | - H Y Leung
- Institute for Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1BD, UK
- Urology Research, The Beatson Institute for Cancer Research, Glasgow Centre for Cancer Research, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| |
Collapse
|
156
|
Clark PR, Jensen T, Kluger MS, Morelock M, Hanidu A, Qi Z, Tatake RJ, Pober JS. MEK5 is activated by shear stress, activates ERK5 and induces KLF4 to modulate TNF responses in human dermal microvascular endothelial cells. Microcirculation 2011; 18:102-17. [PMID: 21166929 PMCID: PMC3075844 DOI: 10.1111/j.1549-8719.2010.00071.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
OBJECTIVE ECs lining arteries respond to LSS by suppressing pro-inflammatory changes, in part through the activation of MEK5, ERK5 and induction of KLF4. We examined if this anti-inflammatory pathway operates in human ECs lining microvessels, the principal site of inflammatory responses. METHODS We used immunofluorescence microscopy of human skin to assess ERK5 activation and KLF4 expression in HDMECs in situ. We applied LSS to or overexpressed MEK5/CA in cultured HDMECs and assessed gene expression by microarrays and qRT-PCR and protein expression by Western blotting. We assessed effects of MEK5/CA on TNF responses using qRT-PCR, FACS and measurements of HDMEC monolayer electrical resistance. We used siRNA knockdown to assess the role of ERK5 and KLF4 in these responses. RESULTS ERK5 phosphorylation and KLF4 expression is observed in HDMECs in situ. LSS activates ERK5 and induces KLF4 in cultured HDMECs. MEK5/CA-transduced HDMECs show activated ERK5 and increased KLF4, thrombomodulin, eNOS, and ICAM-1 expression. MEK5 induction of KLF4 is mediated by ERK5. MEK5/CA-transduced HDMECs are less responsive to TNF, an effect partly mediated by KLF4. CONCLUSIONS MEK5 activation by LSS inhibits inflammatory responses in microvascular ECs, in part through ERK5-dependent induction of KLF4.
Collapse
Affiliation(s)
- Paul R. Clark
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520-8089
| | - Todd Jensen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520-8089
| | - Martin S. Kluger
- Department of Dermatology, Yale University School of Medicine, New Haven, CT 06520-8089
| | | | - Adedayo Hanidu
- Boehringer-Ingelheim Pharmaceuticals Inc., Ridgefield, CT 06877
| | - Zhenhao Qi
- Boehringer-Ingelheim Pharmaceuticals Inc., Ridgefield, CT 06877
| | | | - Jordan S. Pober
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520-8089
- Department of Dermatology, Yale University School of Medicine, New Haven, CT 06520-8089
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520-8089
| |
Collapse
|
157
|
Nardozzi JD, Lott K, Cingolani G. Phosphorylation meets nuclear import: a review. Cell Commun Signal 2010; 8:32. [PMID: 21182795 PMCID: PMC3022542 DOI: 10.1186/1478-811x-8-32] [Citation(s) in RCA: 189] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Accepted: 12/23/2010] [Indexed: 12/18/2022] Open
Abstract
Phosphorylation is the most common and pleiotropic modification in biology, which plays a vital role in regulating and finely tuning a multitude of biological pathways. Transport across the nuclear envelope is also an essential cellular function and is intimately linked to many degeneration processes that lead to disease. It is therefore not surprising that phosphorylation of cargos trafficking between the cytoplasm and nucleus is emerging as an important step to regulate nuclear availability, which directly affects gene expression, cell growth and proliferation. However, the literature on phosphorylation of nucleocytoplasmic trafficking cargos is often confusing. Phosphorylation, and its mirror process dephosphorylation, has been shown to have opposite and often contradictory effects on the ability of cargos to be transported across the nuclear envelope. Without a clear connection between attachment of a phosphate moiety and biological response, it is difficult to fully understand and predict how phosphorylation regulates nucleocytoplasmic trafficking. In this review, we will recapitulate clue findings in the field and provide some general rules on how reversible phosphorylation can affect the nuclear-cytoplasmic localization of substrates. This is only now beginning to emerge as a key regulatory step in biology.
Collapse
Affiliation(s)
- Jonathan D Nardozzi
- Dept, of Biochemistry and Molecular Biology, Thomas Jefferson University, 233 South 10th Street, Philadelphia, PA 19107, USA.
| | | | | |
Collapse
|
158
|
Nakamura K, Johnson GL. Activity assays for extracellular signal-regulated kinase 5. Methods Mol Biol 2010; 661:91-106. [PMID: 20811978 DOI: 10.1007/978-1-60761-795-2_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Extracellular signal-regulated kinase 5 (ERK5) is also known as big MAPK (BMK1) or MAPK7. ERK5 is 115 kDa in mass and therefore larger than the other MAPKs such as ERK1/2, JNK, and p38. Like other MAPKs, ERK5 is ubiquitously expressed in mammalian cells and is part of a three kinase cascade involving a MAPK kinase (MEK5) and MAPK kinase kinase (primarily MEKK2 and MEKK3). ERK5 is important for proliferative responses to growth factors like epidermal growth factor and stress responses such as hyperosmolarity. Upon stimulation, ERK5 rapidly translocates to the nucleus for the control of transcription. ERK5 is also critical for maintenance of vascular integrity and endothelial cell survival. In this chapter, we define methods used to measure the activation of ERK5 using different biochemical and cell-based assays.
Collapse
Affiliation(s)
- Kazuhiro Nakamura
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | | |
Collapse
|
159
|
Keshet Y, Seger R. The MAP kinase signaling cascades: a system of hundreds of components regulates a diverse array of physiological functions. Methods Mol Biol 2010; 661:3-38. [PMID: 20811974 DOI: 10.1007/978-1-60761-795-2_1] [Citation(s) in RCA: 439] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Sequential activation of kinases within the mitogen-activated protein (MAP) kinase (MAPK) cascades is a common, and evolutionary-conserved mechanism of signal transduction. Four MAPK cascades have been identified in the last 20 years and those are usually named according to the MAPK components that are the central building blocks of each of the cascades. These are the extracellular signal-regulated kinase 1/2 (ERK1/2), c-Jun N-Terminal kinase (JNK), p38, and ERK5 cascades. Each of these cascades consists of a core module of three tiers of protein kinases termed MAPK, MAPKK, and MAP3K, and often two additional tiers, the upstream MAP4K and the downstream MAPKAPK, which can complete five tiers of each cascade in certain cell lines or stimulations. The transmission of the signal via each cascade is mediated by sequential phosphorylation and activation of the components in the sequential tiers. These cascades cooperate in transmitting various extracellular signals and thus control a large number of distinct and even opposing cellular processes such as proliferation, differentiation, survival, development, stress response, and apoptosis. One way by which the specificity of each cascade is regulated is through the existence of several distinct components in each tier of the different cascades. About 70 genes, which are each translated to several alternatively spliced isoforms, encode the entire MAPK system, and allow the wide array of cascade's functions. These components, their regulation, as well as their involvement together with other mechanisms in the determination of signaling specificity by the MAPK cascade is described in this review. Mis-regulation of the MAPKs signals usually leads to diseases such as cancer and diabetes; therefore, studying the mechanisms of specificity-determination may lead to better understanding of these signaling-related diseases.
Collapse
Affiliation(s)
- Yonat Keshet
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
160
|
Rose BA, Force T, Wang Y. Mitogen-activated protein kinase signaling in the heart: angels versus demons in a heart-breaking tale. Physiol Rev 2010; 90:1507-46. [PMID: 20959622 PMCID: PMC3808831 DOI: 10.1152/physrev.00054.2009] [Citation(s) in RCA: 574] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Among the myriad of intracellular signaling networks that govern the cardiac development and pathogenesis, mitogen-activated protein kinases (MAPKs) are prominent players that have been the focus of extensive investigations in the past decades. The four best characterized MAPK subfamilies, ERK1/2, JNK, p38, and ERK5, are the targets of pharmacological and genetic manipulations to uncover their roles in cardiac development, function, and diseases. However, information reported in the literature from these efforts has not yet resulted in a clear view about the roles of specific MAPK pathways in heart. Rather, controversies from contradictive results have led to a perception that MAPKs are ambiguous characters in heart with both protective and detrimental effects. The primary object of this review is to provide a comprehensive overview of the current progress, in an effort to highlight the areas where consensus is established verses the ones where controversy remains. MAPKs in cardiac development, cardiac hypertrophy, ischemia/reperfusion injury, and pathological remodeling are the main focuses of this review as these represent the most critical issues for evaluating MAPKs as viable targets of therapeutic development. The studies presented in this review will help to reveal the major challenges in the field and the limitations of current approaches and point to a critical need in future studies to gain better understanding of the fundamental mechanisms of MAPK function and regulation in the heart.
Collapse
Affiliation(s)
- Beth A Rose
- Departments of Anesthesiology, Physiology, and Medicine, David Geffen School of Medicine, Molecular Biology, Institute, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
161
|
Roberts OL, Holmes K, Müller J, Cross DAE, Cross MJ. ERK5 is required for VEGF-mediated survival and tubular morphogenesis of primary human microvascular endothelial cells. J Cell Sci 2010; 123:3189-200. [DOI: 10.1242/jcs.072801] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Extracellular signal-regulated kinase 5 (ERK5) is activated in response to environmental stress and growth factors. Gene ablation of Erk5 in mice is embryonically lethal as a result of disruption of cardiovascular development and vascular integrity. We investigated vascular endothelial growth factor (VEGF)-mediated ERK5 activation in primary human dermal microvascular endothelial cells (HDMECs) undergoing proliferation on a gelatin matrix, and tubular morphogenesis within a collagen gel matrix. VEGF induced sustained ERK5 activation on both matrices. However, manipulation of ERK5 activity by siRNA-mediated gene silencing disrupted tubular morphogenesis without impacting proliferation. Overexpression of constitutively active MEK5 and ERK5 stimulated tubular morphogenesis in the absence of VEGF. Analysis of intracellular signalling revealed that ERK5 regulated AKT phosphorylation. On a collagen gel, ERK5 regulated VEGF-mediated phosphorylation of the pro-apoptotic protein BAD and increased expression of the anti-apoptotic protein BCL2, resulting in decreased caspase-3 activity and apoptosis suppression. Our findings suggest that ERK5 is required for AKT phosphorylation and cell survival and is crucial for endothelial cell differentiation in response to VEGF.
Collapse
Affiliation(s)
- Owain Llŷr Roberts
- NWCRF Institute, School of Biological Sciences, College of Natural Sciences, Bangor University, Bangor, LL57 2UW, UK
| | - Katherine Holmes
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK
| | - Jürgen Müller
- Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - Darren A. E. Cross
- AstraZeneca, Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, UK
| | - Michael J. Cross
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK
| |
Collapse
|
162
|
DÍaz-RodrÍguez E, Pandiella A. Multisite phosphorylation of Erk5 in mitosis. J Cell Sci 2010; 123:3146-56. [DOI: 10.1242/jcs.070516] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The MAP kinase Erk5 plays important roles in cellular proliferation, and has recently been implicated in the regulation of mitosis. The classic pathway of Erk5 activation involves dual phosphorylation at its TEY microdomain by the upstream regulating kinase MEK5. Here we describe a second pathway that controls Erk5 phosphorylation. This pathway is activated in mitotic cells and involves kinase activities distinct from MEK5. Studies aimed at identifying these kinases suggested that CDK1 activity is required to sustain Erk5 phosphorylation in mitosis, as treatment with RO3306, a CDK1 inhibitor, reversed mitotic phosphorylation of Erk5. Moreover, CDK1 co-precipitated with Erk5 in mitotic cells. The mitotic phosphorylation of Erk5 occurs at multiple sites located at its unique C-terminal region, within an Erk5 subdomain that has formerly been implicated in the control of the subcellular location of Erk5. Furthermore, molecular studies indicated that phosphorylation at these sites may participate in the control of the transit of Erk5 between the cytosol and the nucleus, in addition to regulating its transcriptional activity. Together, our results demonstrate the existence of a second Erk5 phosphorylation pathway, that is activated in mitosis, and that may participate in the regulation of Erk5 functions.
Collapse
Affiliation(s)
- Elena DÍaz-RodrÍguez
- Instituto de BiologÍa Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Campus Miguel de Unamuno, 37007-Salamanca, Spain
| | - Atanasio Pandiella
- Instituto de BiologÍa Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Campus Miguel de Unamuno, 37007-Salamanca, Spain
| |
Collapse
|
163
|
Yang Q, Deng X, Lu B, Cameron M, Fearns C, Patricelli MP, Yates JR, Gray NS, Lee JD. Pharmacological inhibition of BMK1 suppresses tumor growth through promyelocytic leukemia protein. Cancer Cell 2010; 18:258-67. [PMID: 20832753 PMCID: PMC2939729 DOI: 10.1016/j.ccr.2010.08.008] [Citation(s) in RCA: 167] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Revised: 06/01/2010] [Accepted: 07/21/2010] [Indexed: 12/18/2022]
Abstract
BMK1 is activated by mitogens and oncogenic signals and, thus, is strongly implicated in tumorigenesis. We found that BMK1 interacted with promyelocytic leukemia protein (PML), and inhibited its tumor-suppressor function through phosphorylation. Furthermore, activated BMK1 notably inhibited PML-dependent activation of p21. To further investigate the BMK-mediated inhibition of the tumor suppressor activity of PML in tumor cells, we developed a small-molecule inhibitor of the kinase activity of BMK1, XMD8-92. Inhibition of BMK1 by XMD8-92 blocked tumor cell proliferation in vitro and significantly inhibited tumor growth in vivo by 95%, demonstrating the efficacy and tolerability of BMK1-targeted cancer treatment in animals.
Collapse
Affiliation(s)
- Qingkai Yang
- Department of Immunology and Microbial Science, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
164
|
Yao Z, Yoon S, Kalie E, Raviv Z, Seger R. Calcium regulation of EGF-induced ERK5 activation: role of Lad1-MEKK2 interaction. PLoS One 2010; 5:e12627. [PMID: 20830310 PMCID: PMC2935384 DOI: 10.1371/journal.pone.0012627] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Accepted: 08/13/2010] [Indexed: 12/17/2022] Open
Abstract
The ERK5 cascade is a MAPK pathway that transmits both mitogenic and stress signals, yet its mechanism of activation is not fully understood. Using intracellular calcium modifiers, we found that ERK5 activation by EGF is inhibited both by the depletion and elevation of intracellular calcium levels. This calcium effect was found to occur upstream of MEKK2, which is the MAP3K of the ERK5 cascade. Co-immunoprecipitation revealed that EGF increases MEKK2 binding to the adaptor protein Lad1, and this interaction was reduced by the intracellular calcium modifiers, indicating that a proper calcium concentration is required for the interactions and transmission of EGF signals to ERK5. In vitro binding assays revealed that the proper calcium concentration is required for a direct binding of MEKK2 to Lad1. The binding of these proteins is not affected by c-Src-mediated phosphorylation on Lad1, but slightly affects the Tyr phosphorylation of MEKK2, suggesting that the interaction with Lad1 is necessary for full Tyr phosphorylation of MEKK2. In addition, we found that changes in calcium levels affect the EGF-induced nuclear translocation of MEKK2 and thereby its effect on the nuclear ERK5 activity. Taken together, these findings suggest that calcium is required for EGF-induced ERK5 activation, and this effect is probably mediated by securing proper interaction of MEKK2 with the upstream adaptor protein Lad1.
Collapse
Affiliation(s)
- Zhong Yao
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Seunghee Yoon
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal Kalie
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Ziv Raviv
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Rony Seger
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
- * E-mail:
| |
Collapse
|
165
|
Expression of myocyte enhancer factor-2 and downstream genes in ground squirrel skeletal muscle during hibernation. Mol Cell Biochem 2010; 344:151-62. [PMID: 20617369 DOI: 10.1007/s11010-010-0538-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2010] [Accepted: 06/24/2010] [Indexed: 01/03/2023]
Abstract
Myocyte enhancer factor-2 (MEF2) transcription factors regulate the expression of a variety of genes encoding contractile proteins and other proteins associated with muscle performance. We proposed that changes in MEF2 levels and expression of selected downstream targets would aid the skeletal muscle of thirteen-lined ground squirrels (Spermophilus tridecemlineatus) in meeting metabolic challenges associated with winter hibernation; e.g., cycles of torpor-arousal, body temperature that can fall to near 0°C, long periods of inactivity that could lead to atrophy. MEF2A protein levels were significantly elevated when animals were in torpor (maximally 2.8-fold higher than in active squirrels) and the amount of phosphorylated active MEF2A Thr312 increased during entrance into torpor. MEF2C levels also rose significantly during entrance and torpor as did the amount of phosphorylated MEF2C Ser387. Furthermore, both MEF2 members showed elevated amounts in the nuclear fraction during torpor as well as enhanced binding to DNA indicating that MEF2-mediated gene expression was up-regulated in torpid animals. Indeed, the protein products of two MEF2 downstream gene targets increased in muscle during torpor (glucose transporter isoforms 4; GLUT4) or early arousal (myogenic differentiation; MyoD). Significant increases in Glut4 and MyoD mRNA transcript levels correlated with the rise in protein product levels and provided further support for the activation of MEF2-mediated gene expression in the hibernator. Transcript levels of Mef2a and Mef2c also showed time-dependent patterns with levels of both being highest during arousal from torpor. The data suggest a significant role for MEF2-mediated gene transcription in the selective adjustment of muscle protein complement over the course of torpor-arousal cycles.
Collapse
|
166
|
Ohnesorge N, Viemann D, Schmidt N, Czymai T, Spiering D, Schmolke M, Ludwig S, Roth J, Goebeler M, Schmidt M. Erk5 activation elicits a vasoprotective endothelial phenotype via induction of Kruppel-like factor 4 (KLF4). J Biol Chem 2010; 285:26199-210. [PMID: 20551324 DOI: 10.1074/jbc.m110.103127] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The MEK5/Erk5 MAPK cascade has recently been implicated in the regulation of endothelial integrity and represents a candidate pathway mediating the beneficial effects of laminar flow, a major factor preventing vascular dysfunction and disease. Here we expressed a constitutively active mutant of MEK5 (MEK5D) to study the transcriptional and functional responses to Erk5 activation in human primary endothelial cells. We provide evidence that constitutive Erk5 activation elicits an overall protective phenotype characterized by increased apoptosis resistance and a decreased angiogenic, migratory, and inflammatory potential. This is supported by bioinformatic microarray analysis, which uncovered a statistical overrepresentation of corresponding functional clusters as well as a significant induction of anti-thrombotic, hemostatic, and vasodilatory genes. We identify KLF4 as a novel Erk5 target and demonstrate a critical role of this transcription factor downstream of Erk5. We show that KLF4 expression largely reproduces the protective phenotype in endothelial cells, whereas KLF4 siRNA suppresses expression of various Erk5 targets. Additionally, we show that vasoprotective statins potently induce KLF4 and KLF4-dependent gene expression via activation of Erk5. Our data underscore a major protective function of the MEK5/Erk5/KLF4 module in ECs and implicate agonistic Erk5 activation as potential strategy for treatment of vascular diseases.
Collapse
Affiliation(s)
- Nils Ohnesorge
- Department of Dermatology, University Hospital Mannheim, University of Heidelberg, Mannheim 68167, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
el Azzouzi H, van Oort RJ, van der Nagel R, Sluiter W, Bergmann MW, De Windt LJ. MEF2 transcriptional activity maintains mitochondrial adaptation in cardiac pressure overload. Eur J Heart Fail 2010; 12:4-12. [PMID: 20023039 DOI: 10.1093/eurjhf/hfp165] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS The transcription factor MEF2 is a downstream target for several hypertrophic signalling pathways in the heart, suggesting that MEF2 may act as a valuable therapeutic target in the treatment of heart failure. METHODS AND RESULTS In this study, we investigated the potential benefits of overall MEF2 inhibition in a mouse model of chronic pressure overloading, by subjecting transgenic mice expressing a dominant negative form of MEF2 (DN-MEF2 Tg) in the heart, to transverse aortic constriction (TAC). Histological analysis revealed no major differences in cardiac remodelling between DN-MEF2 Tg and control mice after TAC. Surprisingly, echocardiographic analysis revealed that DN-MEF2 Tg mice had a decrease in cardiac function compared with control animals. Analysis of the mitochondrial respiratory chain showed that DN-MEF2 Tg mice displayed lower expression of NADH dehydrogenase subunit 6 (ND6), part of mitochondrial Complex I. The reduced expression of ND6 in DN-MEF2 Tg mice after pressure overload correlated with an increase in cell death secondary to overproduction of reactive oxygen species (ROS). CONCLUSION Our data suggest that MEF2 transcriptional activity is required for mitochondrial function and its inhibition predisposes the heart to impaired mitochondrial function, overproduction of ROS, enhanced cell death, and cardiac dysfunction, following pressure overload.
Collapse
Affiliation(s)
- Hamid el Azzouzi
- Department of Cardiology, Faculty of Health, Medicine and Life Sciences, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | | | | | | | | | | |
Collapse
|
168
|
Biyashev D, Veliceasa D, Kwiatek A, Sutanto MM, Cohen RN, Volpert OV. Natural angiogenesis inhibitor signals through Erk5 activation of peroxisome proliferator-activated receptor gamma (PPARgamma). J Biol Chem 2010; 285:13517-24. [PMID: 20185831 PMCID: PMC2859512 DOI: 10.1074/jbc.m110.117374] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Indexed: 01/30/2023] Open
Abstract
Erk-5, a member of the MAPK superfamily, has a catalytic domain similar to Erk1/2 and a unique C-terminal domain enabling binding with transcription factors. Aberrant vascularization in the Erk5-null mice suggested a link to angiogenesis. Ectopic expression of constitutively active Erk5 blocks endothelial cell morphogenesis and causes HIF1-alpha destabilization/degradation. However the mechanisms by which endogenous Erk5 regulates angiogenesis remain unknown. We show that Erk5 and its activating kinase MEK5 are the upstream mediators of the anti-angiogenic signal by the natural angiogenesis inhibitor, pigment epithelial-derived factor (PEDF). We demonstrate that Erk5 phosphorylation allows activation of PPARgamma transcription factor by displacement of SMRT co-repressor. PPARgamma, in turn is critical for NFkappaB activation, PEDF-dependent apoptosis, and anti-angiogenesis. The dominant negative MEK5 mutant and Erk5 shRNA diminished PEDF-dependent apoptosis, inhibition of the endothelial cell chemotaxis, and angiogenesis. This is the first evidence of Erk5-dependent transduction of signals by endogenous angiogenesis inhibitors.
Collapse
Affiliation(s)
- Dauren Biyashev
- From the Urology Department and RH Lurie Comprehensive Cancer Center and
| | - Dorina Veliceasa
- From the Urology Department and RH Lurie Comprehensive Cancer Center and
| | - Angela Kwiatek
- the Physiology Department, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611 and
| | | | - Ronald N. Cohen
- the Department of Medicine, Section of Endocrinology, Diabetes, and Metabolism, University of Chicago, Chicago, Illinois 60637
| | - Olga V. Volpert
- From the Urology Department and RH Lurie Comprehensive Cancer Center and
| |
Collapse
|
169
|
Whyte J, Bergin O, Bianchi A, McNally S, Martin F. Key signalling nodes in mammary gland development and cancer. Mitogen-activated protein kinase signalling in experimental models of breast cancer progression and in mammary gland development. Breast Cancer Res 2010; 11:209. [PMID: 19818165 PMCID: PMC2790844 DOI: 10.1186/bcr2361] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Seven classes of mitogen-activated protein kinase (MAPK) intracellular signalling cascades exist, four of which are implicated in breast disease and function in mammary epithelial cells. These are the extracellular regulated kinase (ERK)1/2 pathway, the ERK5 pathway, the p38 pathway and the c-Jun N-terminal kinase (JNK) pathway. In some forms of human breast cancer and in many experimental models of breast cancer progression, signalling through the ERK1/2 pathway, in particular, has been implicated as being important. We review the influence of ERK1/2 activity on the organised three-dimensional association of mammary epithelial cells, and in models of breast cancer cell invasion. We assess the importance of epidermal growth factor receptor family signalling through ERK1/2 in models of breast cancer progression and the influence of ERK1/2 on its substrate, the oestrogen receptor, in this context. In parallel, we consider the importance of these MAPK-centred signalling cascades during the cycle of mammary gland development. Although less extensively studied, we highlight the instances of signalling through the p38, JNK and ERK5 pathways involved in breast cancer progression and mammary gland development.
Collapse
Affiliation(s)
- Jacqueline Whyte
- Physiology and Medical Physics, Royal College of Surgeons in Ireland, St Stephens Green, Dublin 2, Ireland.
| | | | | | | | | |
Collapse
|
170
|
Asur R, Balasubramaniam M, Marples B, Thomas RA, Tucker JD. Bystander effects induced by chemicals and ionizing radiation: evaluation of changes in gene expression of downstream MAPK targets. Mutagenesis 2010; 25:271-9. [PMID: 20130020 DOI: 10.1093/mutage/geq003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Radiation-induced bystander effects have been evaluated extensively, including the involvement of the mitogen-activated protein kinase (MAPK) pathways. However, few studies have examined the ability of chemicals to induce bystander effects, and the molecular mechanisms involved in chemical bystander effects have not been investigated. We have previously demonstrated the ability of mitomycin C (MMC) and phleomycin (PHL) to induce bystander effects in normal human lymphoblastoid cells. Here, we demonstrate changes in the expression of MAPK target genes following bystander exposure to MMC or PHL or ionizing radiation. The expression changes of 18 genes, which code for proteins that are downstream targets of MAPK proteins, were evaluated at various time points following direct or bystander exposure to MMC, PHL and ionizing radiation. The 18 genes were analysed as groups belonging to one of the seven possible combinations of the three MAPK pathways. We observed statistically significant changes in expression of several genes following exposure to each agent. However, when the expression changes were analysed in the bystander cells alone, significant increases in expression of MAPK target genes were observed for MMC- and radiation-induced bystander effects but not for PHL. PHL is an acknowledged radiomimetic agent; however, in the present study, PHL responses did not resemble those of radiation. These results provide evidence for bystander-induced changes in MAPK proteins and downstream targets and suggest that the bystander effects are a part of a general stress response.
Collapse
Affiliation(s)
- Rajalakshmi Asur
- Department of Biological Sciences, Wayne State University, 5047 Gullen Mall, Suite 1370, Detroit, MI 48202-3917, USA
| | | | | | | | | |
Collapse
|
171
|
Wu Y, Dey R, Han A, Jayathilaka N, Philips M, Ye J, Chen L. Structure of the MADS-box/MEF2 domain of MEF2A bound to DNA and its implication for myocardin recruitment. J Mol Biol 2010; 397:520-33. [PMID: 20132824 DOI: 10.1016/j.jmb.2010.01.067] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Revised: 01/26/2010] [Accepted: 01/28/2010] [Indexed: 12/30/2022]
Abstract
Myocyte enhancer factor 2 (MEF2) regulates specific gene expression in diverse developmental programs and adaptive responses. MEF2 recognizes DNA and interacts with transcription cofactors through a highly conserved N-terminal domain referred to as the MADS-box/MEF2 domain. Here we present the crystal structure of the MADS-box/MEF2 domain of MEF2A bound to DNA. In contrast to previous structural studies showing that the MEF2 domain of MEF2A is partially unstructured, the present study reveals that the MEF2 domain participates with the MADS-box in both dimerization and DNA binding as a single domain. The sequence divergence at and immediately following the C-terminal end of the MEF2 domain may allow different MEF2 dimers to recognize different DNA sequences in the flanking regions. The current structure also suggests that the ligand-binding pocket previously observed in the Cabin1-MEF2B-DNA complex and the HDAC9 (histone deacetylase 9)-MEF2B-DNA complex is not induced by cofactor binding but rather preformed by intrinsic folding. However, the structure of the ligand-binding pocket does undergo subtle but significant conformational changes upon cofactor binding. On the basis of these observations, we generated a homology model of MEF2 bound to a myocardin family protein, MASTR, that acts as a potent coactivator of MEF2-dependent gene expression. The model shows excellent shape and chemical complementarity at the binding interface and is consistent with existing mutagenesis data. The apo structure presented here can also serve as a target for virtual screening and soaking studies of small molecules that can modulate the function of MEF2 as research tools and therapeutic leads.
Collapse
Affiliation(s)
- Yongqing Wu
- Division of Molecular and Computational Biology, Departments of Biological Sciences and Chemistry, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, RRI 204c, 1050 Childs Way, Los Angeles, CA 90089, USA
| | | | | | | | | | | | | |
Collapse
|
172
|
Abstract
ERK5 (extracellular-signal-regulated kinase 5), also termed BMK1 [big MAPK1 (mitogen-activated protein kinase 1)], is the most recently discovered member of the MAPK family. It is expressed in a variety of tissues and is activated by a range of growth factors, cytokines and cellular stresses. Targeted deletion of Erk5 in mice has revealed that the ERK5 signalling cascade is critical for normal cardiovascular development and vascular integrity. In vitro studies have revealed that in endothelial cells, ERK5 is required for preventing apoptosis, mediating shear-stress signalling, regulating hypoxia, tumour angiogenesis and cell migration. This review focuses on our current understanding of the role of ERK5 in regulating endothelial cell function.
Collapse
|
173
|
Fluid shear stress stimulates phosphorylation-dependent nuclear export of HDAC5 and mediates expression of KLF2 and eNOS. Blood 2009; 115:2971-9. [PMID: 20042720 DOI: 10.1182/blood-2009-05-224824] [Citation(s) in RCA: 137] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Fluid shear stress generated by steady laminar blood flow protects vessels from atherosclerosis. Krüppel-like factor 2 (KLF2) and endothelial nitric oxide synthase (eNOS) are fluid shear stress-responsive genes and key mediators in flow anti-inflammatory and antiatherosclerotic actions. However, the molecular mechanisms underlying flow induction of KLF2 and eNOS remain largely unknown. Here, we show a novel role of histone deacetylase 5 (HDAC5) in flow-mediated KLF2 and eNOS expression. We found for the first time that fluid shear stress stimulated HDAC5 phosphorylation and nuclear export in endothelial cells through a calcium/calmodulin-dependent pathway. Consequently, flow induced the dissociation of HDAC5 and myocyte enhancer factor-2 (MEF2) and enhanced MEF2 transcriptional activity, which leads to expression of KLF2 and eNOS. Adenoviral overexpression of a HDAC5 phosphorylation-defective mutant (Ser259/Ser498 were replaced by Ala259/Ala498, HDAC5-S/A), which shows resistance to flow-induced nuclear export, suppressed flow-mediated MEF2 transcriptional activity and expression of KLF2 and eNOS. Importantly, HDAC5-S/A attenuated the flow-inhibitory effect on monocyte adhesion to endothelial cells. Taken together, our results reveal that phosphorylation-dependent derepression of HDAC5 mediates flow-induced KLF2 and eNOS expression as well as flow anti-inflammation, and suggest that HDAC5 could be a potential therapeutic target for the prevention of atherosclerosis.
Collapse
|
174
|
Abstract
In the "canonical" view of transforming growth factor beta (TGF-beta) signaling, Smad7 plays an inhibitory role. While Smad7 represses Smad3 activation by TGF-beta, it does not reverse the inhibitory effect of TGF-beta on myogenesis, suggesting a different function in myogenic cells. We previously reported a promyogenic role of Smad7 mediated by an interaction with MyoD. Based on this association, we hypothesized a possible nuclear function of Smad7 independent of its role at the level of the receptor. We therefore engineered a chimera of Smad7 with a nuclear localization signal (NLS), which serves to prevent and therefore bypass binding to the TGF-beta receptor while concomitantly constitutively localizing Smad7 to the nucleus. This Smad7-NLS did not repress Smad3 activation by TGF-beta but did retain its ability to enhance myogenic gene activation and phenotypic myogenesis, indicating that the nuclear, receptor-independent function of Smad7 is sufficient to promote myogenesis. Furthermore, Smad7 physically interacts with MyoD and antagonizes the repressive effects of active MEK on MyoD. Reporter and myogenic conversion assays indicate a pivotal regulation of MyoD transcriptional properties by the balance between Smad7 and active MEK. Thus, Smad7 has a nuclear coactivator function that is independent of TGF-beta signaling and necessary to promote myogenic differentiation.
Collapse
|
175
|
Villarreal G, Zhang Y, Larman HB, Gracia-Sancho J, Koo A, García-Cardeña G. Defining the regulation of KLF4 expression and its downstream transcriptional targets in vascular endothelial cells. Biochem Biophys Res Commun 2009; 391:984-9. [PMID: 19968965 DOI: 10.1016/j.bbrc.2009.12.002] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Accepted: 12/02/2009] [Indexed: 11/25/2022]
Abstract
The Kruppel-like factor 2 (KLF2) and Kruppel-like factor 4 (KLF4) transcription factors have recently been shown to act as critical regulators of endothelial homeostasis. While several insights have been made into the signaling mechanisms orchestrating endothelial KLF2 expression, those governing the expression of KLF4 in the vascular endothelium remain largely unknown. Here, we show that diverse vasoprotective stimuli including an atheroprotective shear stress waveform, simvastatin, and resveratrol induce the expression of KLF4 in cultured human endothelial cells. We further demonstrate that the induction of KLF4 by resveratrol and atheroprotective shear stress occurs via a MEK5/MEF2-dependent signaling pathway. Since MEK5 activation is also critical for the expression of KLF2, we assessed the individual contribution of KLF4 and KLF2 to the global transcriptional activity triggered by MEK5 activation. Genome-wide transcriptional profiling of endothelial cells overexpressing KLF4, KLF2, or constitutively active MEK5 revealed that 59.2% of the genes regulated by the activation of MEK5 were similarly controlled by either KLF2 or KLF4. Collectively, our data identify a significant degree of mechanistic and functional conservation between KLF2 and KLF4, and importantly, provide further insights into the complex regulatory networks governing endothelial vasoprotection.
Collapse
Affiliation(s)
- Guadalupe Villarreal
- Laboratory for Systems Biology, Center for Excellence in Vascular Biology, Department of Pathology, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
176
|
Charlson AT, Zeliadt NA, Wattenberg EV. Extracellular signal regulated kinase 5 mediates signals triggered by the novel tumor promoter palytoxin. Toxicol Appl Pharmacol 2009; 241:143-53. [PMID: 19716839 PMCID: PMC2784154 DOI: 10.1016/j.taap.2009.08.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Revised: 08/14/2009] [Accepted: 08/17/2009] [Indexed: 10/20/2022]
Abstract
Palytoxin is classified as a non-12-O-tetradecanoylphorbol-13-acetate (TPA)-type skin tumor because it does not bind to or activate protein kinase C. Palytoxin is thus a novel tool for investigating alternative signaling pathways that may affect carcinogenesis. We previously showed that palytoxin activates three major members of the mitogen activated protein kinase (MAPK) family, extracellular signal regulated kinase 1 and 2 (ERK1/2), c-Jun N-terminal kinase (JNK), and p38. Here we report that palytoxin also activates another MAPK family member, called ERK5, in HeLa cells and in keratinocytes derived from initiated mouse skin (308 cells). By contrast, TPA does not activate ERK5 in these cell lines. The major cell surface receptor for palytoxin is the Na+,K+-ATPase. Accordingly, ouabain blocked the ability of palytoxin to activate ERK5. Ouabain alone did not activate ERK5. ERK5 thus represents a divergence in the signaling pathways activated by these two agents that bind to the Na+,K+-ATPase. Cycloheximide, okadaic acid, and sodium orthovanadate did not mimic the effect of palytoxin on ERK5. These results indicate that the stimulation of ERK5 by palytoxin is not simply due to inhibition of protein synthesis or inhibition of serine/threonine or tyrosine phosphatases. Therefore, the mechanism by which palytoxin activates ERK5 differs from that by which it activates ERK1/2, JNK, and p38. Finally, studies that used pharmacological inhibitors and shRNA to block ERK5 action indicate that ERK5 contributes to palytoxin-stimulated c-Fos gene expression. These results suggest that ERK5 can act as an alternative mediator for transmitting diverse tumor promoter-stimulated signals.
Collapse
Affiliation(s)
- Aaron T. Charlson
- Division of Environmental Health Sciences, School of Public Health, University of Minnesota, Minneapolis, MN 55455, USA
| | - Nicholette A. Zeliadt
- Division of Environmental Health Sciences, School of Public Health, University of Minnesota, Minneapolis, MN 55455, USA
| | - Elizabeth V. Wattenberg
- Division of Environmental Health Sciences, School of Public Health, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
177
|
Clapé C, Fritz V, Henriquet C, Apparailly F, Fernandez PL, Iborra F, Avancès C, Villalba M, Culine S, Fajas L. miR-143 interferes with ERK5 signaling, and abrogates prostate cancer progression in mice. PLoS One 2009; 4:e7542. [PMID: 19855844 PMCID: PMC2763222 DOI: 10.1371/journal.pone.0007542] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2009] [Accepted: 09/29/2009] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Micro RNAs are small, non-coding, single-stranded RNAs that negatively regulate gene expression at the post-transcriptional level. Since miR-143 was found to be down-regulated in prostate cancer cells, we wanted to analyze its expression in human prostate cancer, and test the ability of miR-43 to arrest prostate cancer cell growth in vitro and in vivo. RESULTS Expression of miR-143 was analyzed in human prostate cancers by quantitative PCR, and by in situ hybridization. miR-143 was introduced in cancer cells in vivo by electroporation. Bioinformatics analysis and luciferase-based assays were used to determine miR-143 targets. We show in this study that miR-143 levels are inversely correlated with advanced stages of prostate cancer. Rescue of miR-143 expression in cancer cells results in the arrest of cell proliferation and the abrogation of tumor growth in mice. Furthermore, we show that the effects of miR-143 are mediated, at least in part by the inhibition of extracellular signal-regulated kinase-5 (ERK5) activity. We show here that ERK5 is a miR-143 target in prostate cancer. CONCLUSIONS miR-143 is as a new target for prostate cancer treatment.
Collapse
Affiliation(s)
- Cyrielle Clapé
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM, U896, Montpellier, France
- Université de Montpellier1, Montpellier, France
- Centre Regional de Lutte contre le Cancer Val d'Aurelle Paul Lamarque, Montpellier, France
| | - Vanessa Fritz
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM, U896, Montpellier, France
- Université de Montpellier1, Montpellier, France
- Centre Regional de Lutte contre le Cancer Val d'Aurelle Paul Lamarque, Montpellier, France
| | - Corinne Henriquet
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM, U896, Montpellier, France
- Université de Montpellier1, Montpellier, France
- Centre Regional de Lutte contre le Cancer Val d'Aurelle Paul Lamarque, Montpellier, France
| | | | - Pedro Luis Fernandez
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Pathology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - François Iborra
- Service d'urologie, Centre Hospitalier Universitaire Lapeyronie, Montpellier, France
| | - Christophe Avancès
- Service d'Urologie, CHU Groupe Hospitalisation Carémeau, Nîmes, France
- Service d'Urologie, Polyclinique Kennedy, Nîmes, France
| | - Martin Villalba
- Institut de Génétique Moléculaire, Montpellier, France
- CNRS, UMR5535, Montpellier, France
- Université Montpellier2, Montpellier, France
| | - Stéphane Culine
- Department of Medical Oncology, CHU Henri Mondor, Creteil, France
| | - Lluis Fajas
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM, U896, Montpellier, France
- Université de Montpellier1, Montpellier, France
- Centre Regional de Lutte contre le Cancer Val d'Aurelle Paul Lamarque, Montpellier, France
- Service d'urologie, Centre Hospitalier Universitaire Lapeyronie, Montpellier, France
| |
Collapse
|
178
|
Urushihara M, Takamatsu M, Shimizu M, Kondo S, Kinoshita Y, Suga K, Kitamura A, Matsuura S, Yoshizumi M, Tamaki T, Kawachi H, Kagami S. ERK5 activation enhances mesangial cell viability and collagen matrix accumulation in rat progressive glomerulonephritis. Am J Physiol Renal Physiol 2009; 298:F167-76. [PMID: 19846573 DOI: 10.1152/ajprenal.00124.2009] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The mitogen-activated protein kinase (MAPK) cascade plays an important role in the regulation of various cellular functions in glomerulonephritis (GN). Here, we investigated whether extracellular signal-regulated kinase 5 (ERK5), a member of the MAPK family, is involved in the pathogenesis of chronic mesangioproliferative GN, using a rat model induced by uninephrectomy and anti-Thy-1 antibody injection. Immunostaining of kidneys obtained at different time points revealed that phospho-ERK5 was weakly expressed in control glomeruli but dramatically increased in a typical mesangial pattern after 28 and 56 days of GN. A semiquantitative assessment indicated that glomerular phospho-ERK5 expression closely paralleled the accumulation of extracellular matrix (ECM), collagen type I, as well as glomerular expression of reactive oxygen species (ROS) and ANG II. On the other hand, phospho-ERK1/2 expression increased on day 7 during the phase of enhanced mesangial cell (MC) proliferation and decreased thereafter. H(2)O(2) and ANG II each induced ERK5 phosphorylation by cultured rat MCs. Costimulation with both H(2)O(2) and ANG II synergistically increased ERK5 phosphorylation in MCs. Cultured MCs transfected with ERK5-specific small interference RNA showed a significant decrease in H(2)O(2) or ANG II-induced cell viability and soluble collagen secretion compared with control cells. Treatment of GN rats with an ANG II type 1 receptor blocker resulted in significant decreases in phospho-ERK5 expression and collagen accumulation accompanied by remarkable histological improvement. Taken together, these results suggest that MC ERK5 phosphorylation by ANG II or H(2)O(2) enhances cell viability and ECM accumulation in an experimental model of chronic GN.
Collapse
Affiliation(s)
- Maki Urushihara
- Department of Pediatrics, Institute of Health Bioscience, University of Tokushima Graduate School, Tokushima, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
179
|
Wang X, She H, Mao Z. Phosphorylation of neuronal survival factor MEF2D by glycogen synthase kinase 3beta in neuronal apoptosis. J Biol Chem 2009; 284:32619-26. [PMID: 19801631 DOI: 10.1074/jbc.m109.067785] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Glycogen synthase kinase 3beta (GSK3beta) has been identified to play important roles in neuronal death. Evidence from both in vitro and in vivo studies indicates that increased GSK3beta activity contributes to neurodegeneration and to the pathogenesis of Alzheimer disease. But the molecular mechanisms that underlie GSK3beta-mediated neurotoxicity remain poorly understood. We reported here that myocyte enhancer factor 2D (MEF2D), a nuclear transcription factor known to promote neuronal survival, is directly phosphorylated by GSK3beta. Our data showed that phosphorylation of MEF2D by GSK3beta at three specific residues in its transactivation domain inhibits MEF2D transcriptional activity. Withdrawal of neuronal activity in cerebellar granule neurons activated GSK3beta in the nucleus, leading to GSK3beta-dependent inhibition of MEF2 function. This inhibition contributed to GSK3beta-mediated neuronal toxicity. Overexpression of MEF2D mutant that is resistant to GSK3beta inhibition protected cerebellar granule neurons from either GSK3beta activation- or neuronal activity deprivation-induced toxicity. These results identify survival factor MEF2D as a novel downstream effector targeted by GSK3beta and define a molecular link between activation of GSK3beta and neuronal survival machinery which may underlie in part GSK3beta-mediated neurotoxicity.
Collapse
Affiliation(s)
- Xuemin Wang
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | |
Collapse
|
180
|
Casals-Casas C, Alvarez E, Serra M, de la Torre C, Farrera C, Sánchez-Tilló E, Caelles C, Lloberas J, Celada A. CREB and AP-1 activation regulates MKP-1 induction by LPS or M-CSF and their kinetics correlate with macrophage activation versus proliferation. Eur J Immunol 2009; 39:1902-13. [PMID: 19585511 DOI: 10.1002/eji.200839037] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
MAPK phosphatase-1 (MKP-1) is a protein phosphatase that plays a crucial role in innate immunity. This phosphatase inactivates ERK1/2, which are involved in two opposite functional activities of the macrophage, namely proliferation and activation. Here we found that although macrophage proliferation and activation induce MKP-1 with different kinetics, gene expression is mediated by the proximal promoter sequences localized between -380 and -180 bp. Mutagenesis experiments of the proximal element determined that CRE/AP-1 is required for LPS- or M-CSF-induced activation of the MKP-1 gene. Moreover, the results from gel shift analysis and chromatin immunoprecipitation indicated that c-Jun and CREB bind to the CRE/AP-1 box. The distinct kinetics shown by M-CSF and LPS correlates with the induction of JNK and c-jun, as well as the requirement for Raf-1. The signal transduction pathways that activate the induction of MKP-1 correlate kinetically with induction by M-CSF and LPS.
Collapse
|
181
|
Qi SH, Guan QH, Wang M, Zhang GY. Action of ERK5 in ischemic tolerance suggests its probable participation in the signaling mechanism. J Recept Signal Transduct Res 2009; 29:38-43. [PMID: 19519168 DOI: 10.1080/10799890802675767] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Here we examined the effects of ischemia preconditioning and ketamine, an NMDA receptor antagonist, on the activation and its nucleus translocation of ERK5 in hippocampal CA1 region. Our results showed ERK5 was not activated in rat hippocampus CA1 region. But in cytosol extracts preconditioned with 3 min of sublethal ischaemia, ERK5 activation was enhanced significantly, with two peaks occurring at 3 hr and 3 days, respectively. This activation returned to base level 3 days later. The results lead us to conclude that preconditioning increased the activations of ERK5 during reperfusion after lethal ischemia through NMDA receptor. Preconditioning increased the activation and nucleus translocation of ERK5 during reperfusion after lethal ischemia through the NMDA receptor. These findings might provide some clues to understanding the mechanism underlying ischemia tolerance and to finding clinical therapies for stroke using the endogenous neuroprotection.
Collapse
Affiliation(s)
- Su-Hua Qi
- School of Life Science and Biotechnology, China Pharmaceutical University, 24 Tongjia Street, Nanjing, PR China
| | | | | | | |
Collapse
|
182
|
Kumar A, Hoffman TA, Dericco J, Naqvi A, Jain MK, Irani K. Transcriptional repression of Kruppel like factor-2 by the adaptor protein p66shc. FASEB J 2009; 23:4344-52. [PMID: 19696221 DOI: 10.1096/fj.09-138743] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The adaptor protein p66shc promotes cellular oxidative stress and apoptosis. Here, we demonstrate a novel mechanistic relationship between p66shc and the kruppel like factor-2 (KLF2) transcription factor and show that this relationship has biological relevance to p66shc-regulated cellular oxidant level, as well as KLF2-induced target gene expression. Genetic knockout of p66shc in mouse embryonic fibroblasts (MEFs) stimulates activity of the core KLF2 promoter and increases KLF2 mRNA and protein expression. Similarly, shRNA-induced knockdown of p66shc increases KLF2-promoter activity in HeLa cells. The increase in KLF2-promoter activity in p66shc-knockout MEFs is dependent on a myocyte enhancing factor-2A (MEF2A)-binding sequence in the core KLF2 promoter. Short-hairpin RNA-induced knockdown of p66shc in endothelial cells also stimulates KLF2 mRNA and protein expression, as well as expression of the endothelial KLF2 target gene thrombomodulin. MEF2A protein and mRNA are more abundant in p66shc-knockout MEFs, resulting in greater occupancy of the KLF2 promoter by MEF2A. In endothelial cells, the increase in KLF2 and thrombomodulin protein by shRNA-induced decrease in p66shc expression is partly abrogated by knockdown of MEF2A. Finally, knockdown of KLF2 abolishes the decrease in the cellular reactive oxygen species hydrogen peroxide observed with knockdown of p66shc, and KLF2 overexpression suppresses cellular hydrogen peroxide levels, independent of p66shc expression. These findings illustrate a novel mechanism by which p66shc promotes cellular oxidative stress, through suppression of MEF2A expression and consequent repression of KLF2 transcription.
Collapse
Affiliation(s)
- Ajay Kumar
- Cardiovascular Institute, University of Pittsburgh Medical Center, 623S Scaife Hall, 200 Lothrop St., Pittsburgh, PA 15213, USA.
| | | | | | | | | | | |
Collapse
|
183
|
Geng J, Zhao Z, Kang W, Wang W, Liu G, Sun Y, Zhang Y, Ge Z. Hypertrophic response to angiotensin II is mediated by protein kinase D-extracellular signal-regulated kinase 5 pathway in human aortic smooth muscle cells. Biochem Biophys Res Commun 2009; 388:517-22. [PMID: 19666008 DOI: 10.1016/j.bbrc.2009.08.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2009] [Accepted: 08/04/2009] [Indexed: 11/17/2022]
Abstract
Angiotensin II plays a critical role in hypertrophy of vascular smooth muscle cells, however, the molecular underpinnings remain unclear. The present study indicated that AT1/PKC/PKD pathway was able to regulate downstream ERK5, affecting pro-hypertrophic responses to Ang II. Ang II-stimulated phosphorylation of ERK5 in a time- and dose-dependent manner in human aortic smooth muscle cells (HASMCs). The pharmacological inhibitors for AT1 and PKCs significantly inhibited Ang II-induced ERK5 activation, suggesting the involvement of the AT1/PKC pathway. In particular, PKD was critical for Ang II-induced ERK5 activation since silencing PKD by siRNA markedly inhibited Ang II-induced ERK5 activation. Consequently, we found that Losartan, Gö 6983 and PKD siRNA significantly attenuated ERK5 activated translocation and hypertrophy of HASMCs by Ang II. Taken together, we demonstrated for the first time that Ang II activates ERK5 via the AT1/PKC/PKD pathway and revealed a critical role of ERK5 in Ang II-induced HASMCs hypertrophy.
Collapse
Affiliation(s)
- Jing Geng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital, Jinan, Shandong 250012, PR China
| | | | | | | | | | | | | | | |
Collapse
|
184
|
Spiering D, Schmolke M, Ohnesorge N, Schmidt M, Goebeler M, Wegener J, Wixler V, Ludwig S. MEK5/ERK5 signaling modulates endothelial cell migration and focal contact turnover. J Biol Chem 2009; 284:24972-80. [PMID: 19605361 PMCID: PMC2757201 DOI: 10.1074/jbc.m109.042911] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The formation of new blood vessels from pre-existing ones requires highly coordinated restructuring of endothelial cells (EC) and the surrounding extracellular matrix. Directed EC migration is a central step in this process and depends on cellular signaling cascades that initiate and control the structural rearrangements. On the basis of earlier findings that ERK5 deficiency in mouse EC results in massive defects in vessel architecture, we focused on the impact of the MEK5/ERK5 signaling pathway on EC migration. Using a retroviral gene transfer approach, we found that constitutive activation of MEK5/ERK5 signaling strongly inhibits EC migration and results in massive morphological changes. The area covered by spread EC was dramatically enlarged, accompanied by an increase in focal contacts and altered organization of actin filaments. Consequently, cells were more rigid and show reduced motility. This phenotype was most likely based on decreased focal contact turnover caused by reduced expression of p130Cas, a key player in directed cell migration. We demonstrate for the first time that ERK5 signaling not only is involved in EC survival and stress response but also controls migration and morphology of EC.
Collapse
Affiliation(s)
- Désirée Spiering
- Institute of Molecular Virology (IMV), Center of Molecular Biology of Inflammation (ZMBE), and Interdisciplinary Center of Medical Research (IZKF), Universitaetsklinikum Muenster, 48149 Muenster, Germany
| | | | | | | | | | | | | | | |
Collapse
|
185
|
Obara Y, Yamauchi A, Takehara S, Nemoto W, Takahashi M, Stork PJS, Nakahata N. ERK5 activity is required for nerve growth factor-induced neurite outgrowth and stabilization of tyrosine hydroxylase in PC12 cells. J Biol Chem 2009; 284:23564-73. [PMID: 19581298 DOI: 10.1074/jbc.m109.027821] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Extracellular signal-regulated kinases (ERKs) play important physiological roles in proliferation, differentiation, and gene expression. ERK5 is approximately twice the size of ERK1/2, and its amino-terminal half contains the kinase domain that shares homology with ERK1/2 and TEY activation motif, whereas the carboxyl-terminal half is unique. In this study, we examined a physiological role of ERK5 in rat pheochromocytoma cells (PC12), comparing it with ERK1/2. Nerve growth factor (NGF) induced phosphorylation of both ERK5 and ERK1/2, whereas the cAMP analog dibutyryl cAMP (Bt(2)cAMP) caused only ERK1/2 phosphorylation. U0126, at 30 mum, that blocks ERK1/2 signaling selectively attenuated neurite outgrowth induced by NGF and Bt(2)cAMP, but BIX02188 and BIX02189, at 30 mum, that block ERK5 signaling and an ERK5 dominant-negative mutant suppressed only NGF-induced neurite outgrowth. Next, we examined the expression of tyrosine hydroxylase, a rate-limiting enzyme of catecholamine biosynthesis. Both NGF and Bt(2)cAMP increased tyrosine hydroxylase gene promoter activity in an ERK1/2-dependent manner but was ERK5-independent. However, when both ERK5 and ERK1/2 signalings were inhibited, tyrosine hydroxylase protein up-regulation by NGF and Bt(2)cAMP was abolished, because of the loss of stabilization of tyrosine hydroxylase protein by ERK5. Taking these results together, ERK5 is involved in neurite outgrowth and stabilization of tyrosine hydroxylase in PC12 cells, and ERK5, along with ERK1/2, plays essential roles in the neural differentiation process.
Collapse
Affiliation(s)
- Yutaro Obara
- Department of Cellular Signaling, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan.
| | | | | | | | | | | | | |
Collapse
|
186
|
Gilley R, March HN, Cook SJ. ERK1/2, but not ERK5, is necessary and sufficient for phosphorylation and activation of c-Fos. Cell Signal 2009; 21:969-77. [PMID: 19249353 DOI: 10.1016/j.cellsig.2009.02.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2009] [Accepted: 02/16/2009] [Indexed: 12/22/2022]
Abstract
Growth factor-stimulated expression and activation of c-Fos is regulated by the ERK1/2 pathway. However, recent reports have also suggested a prominent role for the closely related ERK5 pathway in regulating the expression, transcriptional activation and nuclear localization of c-Fos. Here we have compared the role of ERK1/2 and ERK5 in regulating c-Fos using a combination of conditional protein kinases, selective biochemical inhibitors and ERK5 null fibroblasts. We demonstrate that activation of the ERK1/2 pathway, but not ERK5, is sufficient for c-Fos phosphorylation and transcriptional activation. Furthermore, growth factor-dependent expression of c-Fos is blocked by low doses of PD184352 that selectively inhibit the ERK1/2 pathway but proceeds normally in ERK5-/- 3T9 cells; in addition, nuclear localization of c-Fos is normal in ERK5-/- cells. ERK5-/- cells are, however, defective for c-Jun expression but this is reversed by re-expression of ERK5. In addition to ERK5, neither the JNK nor p38 pathways can substitute for ERK1/2 in the regulation of c-Fos transcriptional activity. These results demonstrate that c-Fos transcriptional activity is not regulated by the ERK5 pathway; rather, of all the MAPKs and SAPKs, c-Fos activation appears to be predominantly linked to the ERK1/2 pathway.
Collapse
Affiliation(s)
- Rebecca Gilley
- Laboratory of Molecular Signalling, The Babraham Institute, Babraham Research Campus, Cambridge, UK.
| | | | | |
Collapse
|
187
|
Chen RQ, Yang QK, Chen YL, Oliveira VA, Dalton WS, Fearns C, Lee JD. Kinome siRNA screen identifies SMG-1 as a negative regulator of hypoxia-inducible factor-1alpha in hypoxia. J Biol Chem 2009; 284:16752-16758. [PMID: 19406746 PMCID: PMC2719310 DOI: 10.1074/jbc.m109.014316] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Hypoxia-inducible factor-1 (HIF-1) plays a central role in tumor progression by regulating genes involved in proliferation, glycolysis, angiogenesis, and metastasis. To improve our understanding of HIF-1 regulation by kinome, we screened a kinase-specific small interference RNA library using a hypoxia-response element (HRE) luciferase reporter assay under hypoxic conditions. This screen determined that depletion of cellular SMG-1 kinase most significantly modified cellular HIF-1 activity in hypoxia. SMG-1 is the newest and least studied member of the phosphoinositide 3-kinase-related kinase family, which consists of ATM, ATR, DNA-PKcs, mTOR, and SMG-1. We individually depleted members of the phosphoinositide 3-kinase-related kinase family, and only SMG-1 deficiency significantly augmented HIF-1 activity in hypoxia. We subsequently discovered that SMG-1 kinase activity was activated by hypoxia, and depletion of SMG-1 up-regulated MAPK activity under low oxygen. Suppressing cellular MAPK by silencing ERK1/2 or by treatment with U0126, a MAPK inhibitor, partially blocked the escalation of HIF-1 activity resulting from SMG-1 deficiency in hypoxic cells. Increased expression of SMG-1 but not kinase-dead SMG-1 effectively inhibited the activity of HIF-1alpha. In addition, cellular SMG-1 deficiency increased secretion of the HIF-1alpha-regulated angiogenic factor, vascular epidermal growth factor, and survival factor, carbonic anhydrase IX (CA9), as well as promoted the hypoxic cell motility. Taken together, we discovered that SMG-1 negatively regulated HIF-1alpha activity in hypoxia, in part through blocking MAPK activation.
Collapse
Affiliation(s)
- Run-Qiang Chen
- From the Departments of Immunology and Microbial Science, La Jolla, California 92037
| | - Qing-Kai Yang
- From the Departments of Immunology and Microbial Science, La Jolla, California 92037
| | - Yan-Ling Chen
- From the Departments of Immunology and Microbial Science, La Jolla, California 92037
| | - Vasco A Oliveira
- Chemistry, The Scripps Research Institute, La Jolla, California 92037
| | - William S Dalton
- Chemistry, The Scripps Research Institute, La Jolla, California 92037
| | - Colleen Fearns
- Department of Experimental Therapeutics and Interdisciplinary Oncology, H. Lee Moffitt Cancer Center & Research Institute, University of South Florida, Tampa, Florida 33612
| | - Jiing-Dwan Lee
- From the Departments of Immunology and Microbial Science, La Jolla, California 92037.
| |
Collapse
|
188
|
Direct interaction between myocyte enhancer factor 2 (MEF2) and protein phosphatase 1alpha represses MEF2-dependent gene expression. Mol Cell Biol 2009; 29:3355-66. [PMID: 19364819 DOI: 10.1128/mcb.00227-08] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The myocyte enhancer factor 2 (MEF2) transcription factors play important roles in neuronal, cardiac, and skeletal muscle tissues. MEF2 serves as a nuclear sensor, integrating signals from several signaling cascades through protein-protein interactions with kinases, chromatin remodeling factors, and other transcriptional regulators. Here, we report a novel interaction between the catalytic subunit of protein phosphatase 1alpha (PP1alpha) and MEF2. Interaction occurs within the nucleus, and binding of PP1alpha to MEF2 potently represses MEF2-dependent transcription. The interaction utilizes uncharacterized domains in both PP1alpha and MEF2, and PP1alpha phosphatase activity is not obligatory for MEF2 repression. Moreover, a MEF2-PP1alpha regulatory complex leads to nuclear retention and recruitment of histone deacetylase 4 to MEF2 transcription complexes. PP1alpha-mediated repression of MEF2 overrides the positive influence of calcineurin signaling, suggesting PP1alpha exerts a dominant level of control over MEF2 function. Indeed, PP1alpha-mediated repression of MEF2 function interferes with the prosurvival effect of MEF2 in primary hippocampal neurons. The PP1alpha-MEF2 interaction constitutes a potent locus of control for MEF2-dependent gene expression, having potentially important implications for neuronal cell survival, cardiac remodeling in disease, and terminal differentiation of vascular, cardiac, and skeletal muscle.
Collapse
|
189
|
Cundiff P, Liu L, Wang Y, Zou J, Pan YW, Abel G, Duan X, Ming GL, Englund C, Hevner R, Xia Z. ERK5 MAP kinase regulates neurogenin1 during cortical neurogenesis. PLoS One 2009; 4:e5204. [PMID: 19365559 PMCID: PMC2664926 DOI: 10.1371/journal.pone.0005204] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2009] [Accepted: 02/16/2009] [Indexed: 01/15/2023] Open
Abstract
The commitment of multi-potent cortical progenitors to a neuronal fate depends on the transient induction of the basic-helix-loop-helix (bHLH) family of transcription factors including Neurogenin 1 (Neurog1). Previous studies have focused on mechanisms that control the expression of these proteins while little is known about whether their pro-neural activities can be regulated by kinase signaling pathways. Using primary cultures and ex vivo slice cultures, here we report that both the transcriptional and pro-neural activities of Neurog1 are regulated by extracellular signal-regulated kinase (ERK) 5 signaling in cortical progenitors. Activation of ERK5 potentiated, while blocking ERK5 inhibited Neurog1-induced neurogenesis. Furthermore, endogenous ERK5 activity was required for Neurog1-initiated transcription. Interestingly, ERK5 activation was sufficient to induce Neurog1 phosphorylation and ERK5 directly phosphorylated Neurog1 in vitro. We identified S179/S208 as putative ERK5 phosphorylation sites in Neurog1. Mutations of S179/S208 to alanines inhibited the transcriptional and pro-neural activities of Neurog1. Our data identify ERK5 phosphorylation of Neurog1 as a novel mechanism regulating neuronal fate commitment of cortical progenitors.
Collapse
Affiliation(s)
- Paige Cundiff
- Department of Pharmacology, University of Washington, Seattle, Washington, United States of America
| | - Lidong Liu
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Yupeng Wang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Junhui Zou
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Yung-Wei Pan
- Graduate Program in Molecular and Cellular Biology, University of Washington, Seattle, Washington, United States of America
| | - Glen Abel
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Xin Duan
- Institute for Cell Engineering, Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Guo-li Ming
- Institute for Cell Engineering, Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Chris Englund
- Graduate Program in Neurobiology and Behavior, University of Washington, Seattle, Washington, United States of America
| | - Robert Hevner
- Graduate Program in Neurobiology and Behavior, University of Washington, Seattle, Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Zhengui Xia
- Department of Pharmacology, University of Washington, Seattle, Washington, United States of America
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
- Graduate Program in Molecular and Cellular Biology, University of Washington, Seattle, Washington, United States of America
- Graduate Program in Neurobiology and Behavior, University of Washington, Seattle, Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
190
|
Sawhney RS, Liu W, Brattain MG. A novel role of ERK5 in integrin-mediated cell adhesion and motility in cancer cells via Fak signaling. J Cell Physiol 2009; 219:152-61. [PMID: 19089993 PMCID: PMC7064882 DOI: 10.1002/jcp.21662] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In metastatic cancer, high expression levels of vitronectin (VN) receptors (integrins), FAK, and ERK5 are reported. We hypothesized that integrin-mediated ERK5 activation via FAK may play a pivotal role in cell adhesion, motility, and metastasis. ERK5 and FAK phosphorylation when metastatic MDA-MB-231 and PC-3 cells were plated on VN was enhanced. Further experiments showed co-immunoprecipitation of integrins beta1, alpha V beta 3, or alpha V beta 5 with ERK5 and FAK. To gain better insight into the mechanism of ERK5, FAK, and VN receptors in cell adhesion and motility, we performed loss-of-function experiments using integrin blocking antibodies, and specific mutants of FAK and ERK5. Ectopic expression of dominant negative ERK5/AEF decreased ERK5 and FAK (Y397) phosphorylation, cell adhesion, and haptotactic motility (micromotion) on VN. Additionally, DN FAK expression attenuated ERK5 phosphorylation, cell adhesion, and motility. This study documents the novel finding that in breast and prostate cancer cells, ERK5 is a critical target of FAK in cell adhesion signaling. Using different cancer cells, our experiments unveil a novel mechanism by which VN receptors and FAK could promote cancer metastasis via ERK5 activation.
Collapse
Affiliation(s)
- Rajinder S Sawhney
- Department of Pharmacology & Toxicology, SUNY at Buffalo, Buffalo, New York, USA.
| | | | | |
Collapse
|
191
|
della Gaspera B, Armand AS, Sequeira I, Lecolle S, Gallien CL, Charbonnier F, Chanoine C. The Xenopus MEF2 gene family: evidence of a role for XMEF2C in larval tendon development. Dev Biol 2009; 328:392-402. [PMID: 19389348 DOI: 10.1016/j.ydbio.2009.01.039] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Revised: 01/12/2009] [Accepted: 01/28/2009] [Indexed: 01/22/2023]
Abstract
MEF2 transcription factors are well-established regulators of muscle development. In this report, we describe the cloning of multiple splicing isoforms of the XMEF2A and XMEF2C encoding genes, differentially expressed during Xenopus development. Using whole-mount in situ hybridization, we found that the accumulation of XMEF2C mRNA in the tadpole stages was restricted to intersomitic regions and to the peripheral edges of hypaxial and cranial muscle masses in contrast to XMEF2A and XMEF2D, characterized by a continuous muscle cell expression. The XMEF2C positive cells express the bHLH transcription factor, Xscleraxis, known as a specific marker for tendons. Gain of function experiments revealed that the use of a hormone-inducible XMEF2C construct is able to induce Xscleraxis expression. Furthermore, XMEF2C specifically cooperates with Xscleraxis to induce tenascin C and betaig-h3, two genes preferentially expressed in Xenopus larval tendons. These findings 1) highlight a previously unappreciated and specific role for XMEF2C in tendon development and 2) identify a novel gene transactivation pathway where MEF2C cooperates with the bHLH protein, Xscleraxis, to activate specific gene expression.
Collapse
Affiliation(s)
- Bruno della Gaspera
- UMR 7060 CNRS, Equipe Biologie du Développement et de la Différenciation Neuromusculaire, Centre Universitaire des Saints-Pères, 45, rue des Saints-Pères, Université Paris Descartes, F-75270 Paris Cedex 06, France
| | | | | | | | | | | | | |
Collapse
|
192
|
Zen K, Yasui K, Nakajima T, Zen Y, Zen K, Gen Y, Mitsuyoshi H, Minami M, Mitsufuji S, Tanaka S, Itoh Y, Nakanuma Y, Taniwaki M, Arii S, Okanoue T, Yoshikawa T. ERK5 is a target for gene amplification at 17p11 and promotes cell growth in hepatocellular carcinoma by regulating mitotic entry. Genes Chromosomes Cancer 2009; 48:109-120. [PMID: 18973138 DOI: 10.1002/gcc.20624] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2025] Open
Abstract
Using high-density oligonucleotide microarrays, we investigated DNA copy-number aberrations in cell lines derived from hepatocellular carcinomas (HCCs) and detected a novel amplification at 17p11. To identify the target of amplification at 17p11, we defined the extent of the amplicon and examined HCC cell lines for expression of all seven genes in the 750-kb commonly amplified region. Mitogen-activated protein kinase (MAPK) 7, which encodes extracellular-regulated protein kinase (ERK) 5, was overexpressed in cell lines in which the gene was amplified. An increase in MAPK7 copy number was detected in 35 of 66 primary HCC tumors. Downregulation of MAPK7 by small interfering RNA suppressed the growth of SNU449 cells, the HCC cell line with the greatest amplification and overexpression of MAPK7. ERK5, phosphorylated during the G2/M phases of the cell cycle, regulated entry into mitosis in SNU449 cells. In conclusion, our results suggest that MAPK7 is likely the target of 17p11 amplification and that the ERK5 protein product of MAPK7 promotes the growth of HCC cells by regulating mitotic entry.
Collapse
MESH Headings
- Analysis of Variance
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Cycle/genetics
- Cell Line, Tumor
- Chi-Square Distribution
- Chromosomes, Human, Pair 17/genetics
- Chromosomes, Human, Pair 17/metabolism
- Down-Regulation/genetics
- Gene Dosage
- Gene Expression Regulation, Neoplastic
- Gene Order/genetics
- Humans
- Immunohistochemistry
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Mitogen-Activated Protein Kinase 7/genetics
- Mitogen-Activated Protein Kinase 7/metabolism
- Mitosis/genetics
- Mitotic Index
- Oligonucleotide Array Sequence Analysis
- Phosphorylation/genetics
- RNA, Small Interfering
- Signal Transduction/genetics
Collapse
Affiliation(s)
- Keika Zen
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
193
|
Sundaramurthy P, Gakkhar S, Sowdhamini R. Analysis of the impact of ERK5, JNK, and P38 kinase cascades on each other: a systems approach. Bioinformation 2009; 3:244-9. [PMID: 19255643 PMCID: PMC2646858 DOI: 10.6026/97320630003244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Accepted: 11/11/2008] [Indexed: 11/23/2022] Open
Abstract
The classical concept of linear pathways is being increasingly challenged by network representations, which emphasize the importance of interactions between components of a biological system, and motivates for adopting a system-level approach in biology. We have developed a dynamical system that integrates quantitative, dynamic and topological representation of network of ERK5 (Extracellular signal-regulated kinases 5), JNK(c-Jun N-terminal kinases) and P38 kinase cascades. We have observered that, the transient activation of ERK5, JNK1 and P38beta kinase, and the persistent activation of JNK2, JNK3 and P38 delta kinase does not get affected due to the cross-talks between ERK5, JNK and P38 kinase cascades. But it is due to the cross - talks, the transiently activated P38alpha kinase become inactivated, and the transiently activated P38gamma kinase become persistently activated. The impacts of one-way cross-talks between the cascades are insignificant and differ from the impact of two-way cross-talks. We generate a hypothesis that, signaling pathways should be studied as a system by considering the cross-talks between the two adjacent cascades.
Collapse
Affiliation(s)
- Pandurangan Sundaramurthy
- Department of Mathematics, Indian Institute of Technology Roorkee, Roorkee - 247667, Uttarakhand, India.
| | | | | |
Collapse
|
194
|
Andrographolide reduces IL-2 production in T-cells by interfering with NFAT and MAPK activation. Eur J Pharmacol 2009; 602:413-21. [DOI: 10.1016/j.ejphar.2008.11.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2008] [Revised: 10/07/2008] [Accepted: 11/03/2008] [Indexed: 12/30/2022]
|
195
|
Sako K, Fukuhara S, Minami T, Hamakubo T, Song H, Kodama T, Fukamizu A, Gutkind JS, Koh GY, Mochizuki N. Angiopoietin-1 induces Kruppel-like factor 2 expression through a phosphoinositide 3-kinase/AKT-dependent activation of myocyte enhancer factor 2. J Biol Chem 2008; 284:5592-601. [PMID: 19106103 DOI: 10.1074/jbc.m806928200] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Angiopoietin-1 (Ang1) regulates both vascular quiescence and angiogenesis through the receptor tyrosine kinase Tie2. We and another group have recently shown that Ang1 and Tie2 form distinct signaling complexes at cell-cell and cell-matrix contacts and further demonstrated that the former selectively induces expression of Krüppel-like factor 2 (KLF2), a transcription factor involved in vascular quiescence. Here, we investigated the mechanism of how Ang1/Tie2 signal induces KLF2 expression to clarify the role of KLF2 in Ang1/Tie2 signal-mediated vascular quiescence. Ang1 stimulated KLF2 promoter-driven reporter gene expression in endothelial cells, whereas it failed when a myocyte enhancer factor 2 (MEF2)-binding site of KLF2 promoter was mutated. Depletion of MEF2 by siRNAs abolished Ang1-induced KLF2 expression, indicating the requirement of MEF2 in KLF2 induction by Ang1. Constitutive active phosphoinositide 3-kinase (PI3K) and AKT increased the MEF2-dependent reporter gene expression by enhancing its transcriptional activity and stimulated the KLF2 promoter activity cooperatively with MEF2. Consistently, inhibition of either PI3K or AKT and depletion of AKT abrogated Ang1-induced KLF2 expression. In addition, we confirmed the dispensability of extracellular signal-regulated kinase 5 (ERK5) for Ang1-induced KLF2 expression. Furthermore, depletion of KLF2 resulted in the loss of the inhibitory effect of Ang1 on vascular endothelial growth factor (VEGF)-mediated expression of vascular cell adhesion molecule-1 in endothelial cells and VEGF-mediated monocyte adhesion to endothelial cells. Collectively, these findings indicate that Ang1/Tie2 signal stimulates transcriptional activity of MEF2 through a PI3K/AKT pathway to induce KLF2 expression, which may counteract VEGF-mediated inflammatory responses.
Collapse
Affiliation(s)
- Keisuke Sako
- Department of Structural Analysis, National Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
196
|
Walden TB, Timmons JA, Keller P, Nedergaard J, Cannon B. Distinct expression of muscle-specific microRNAs (myomirs) in brown adipocytes. J Cell Physiol 2008; 218:444-9. [PMID: 18937285 DOI: 10.1002/jcp.21621] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
MicroRNAs, a novel class of post-transcriptional gene regulators, have been demonstrated to be involved in several cellular processes regulating the expression of protein-coding genes. Here we examine murine white and brown primary cell cultures for differential expression of miRNAs. The adipogenesis-related miRNA miR-143 was highly expressed in mature white adipocytes but was low in mature brown adipocytes. Three classical "myogenic" miRNAs miR-1, miR-133a and miR-206 were absent from white adipocytes but were specifically expressed both in brown pre- and mature adipocytes, reinforcing the concept that brown adipocytes and myocytes derive from a common cell lineage that specifies energy-dissipating cells. Augmentation of adipocyte differentiation status with norepinephrine or rosiglitazone did not affect the expression of the above miRNAs, the expression levels of which were thus innately regulated. However, expression of the miRNA miR-455 was enhanced during brown adipocyte differentiation, similarly to the expression pattern of the brown adipocyte differentiation marker UCP1. In conclusion, miRNAs are differentially expressed in white and brown adipocytes and may be important in defining the common precursor cell for myocytes and brown adipocytes and thus have distinct roles in energy-storing versus energy-dissipating cells.
Collapse
Affiliation(s)
- Tomas B Walden
- The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
197
|
Wang RM, Zhang QG, Li J, Yang LC, Yang F, Brann DW. The ERK5-MEF2C transcription factor pathway contributes to anti-apoptotic effect of cerebral ischemia preconditioning in the hippocampal CA1 region of rats. Brain Res 2008; 1255:32-41. [PMID: 19103177 DOI: 10.1016/j.brainres.2008.12.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Revised: 11/25/2008] [Accepted: 12/01/2008] [Indexed: 11/18/2022]
Abstract
The purpose of the present study was to investigate the role of myocyte enhancer binding factor 2C (MEF2C), a common substrate of p38 kinase and extracellular signal-regulated kinase 5 (ERK5) in the hippocampal CA1 region following cerebral ischemia preconditioning (CIP) and without CIP. In animals that did not undergo preconditioning, MEF2C was significantly activated with an early peak at 30 min of reperfusion, which was followed by a pronounced decrease of MEF2C protein levels in the late phase of reperfusion (3-5 d). Co-immunoprecipitation studies failed to show an interaction between ERK5 and MEF2C, and ERK5-antisense oligonucleotide (ERK5-AS) had no effect on MEF2C activation, suggesting that the MEF2C activation is mediated by a kinase other than ERK5. Following preconditioning (3 min ischemia), MEF2C was strongly activated during the late stage of reperfusion (6 h-5 d). Co-immunoprecipitation studies showed that the interaction of ERK5 and MEF2C significantly increased at 3 d of reperfusion, and this increase was markedly inhibited by ERK5-AS. Inhibition of the ERK5-MEF2C pathway resulted in a significant increase in the number of TUNEL-positive apoptotic cells compared with CIP groups in the hippocampal CA1 region, and abolished the neuroprotective effect induced by CIP. Taken together, these results demonstrate that ERK5-MEF2C signaling is significantly enhanced in the hippocampus CA1 following CIP, and that ERK5-MEF2C signaling plays a critical role in the mediation of the anti-apoptotic and neuroprotective actions of ischemic preconditioning.
Collapse
Affiliation(s)
- Rui-Min Wang
- Research Center for Molecular Biology, North China Coal Medical University, Tangshan 063000, China.
| | | | | | | | | | | |
Collapse
|
198
|
Tatake RJ, O’Neill MM, Kennedy CA, Wayne AL, Jakes S, Wu D, Kugler SZ, Kashem MA, Kaplita P, Snow RJ. Identification of pharmacological inhibitors of the MEK5/ERK5 pathway. Biochem Biophys Res Commun 2008; 377:120-5. [DOI: 10.1016/j.bbrc.2008.09.087] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2008] [Accepted: 09/19/2008] [Indexed: 11/27/2022]
|
199
|
Abstract
c-Abl is a non-receptor tyrosine kinase which is localized both in the nucleus and cytoplasm, and is involved in the regulation of cell growth, survival and morphogenesis. Although c-Abl nuclear function has been extensively studied, recent data also indicate an important role in cytoplasmic signalling through mitogenic and adhesive receptors. Here, we review the mechanisms by which growth factors promote cytoplasmic c-Abl activation and signalling and its function in the induction of DNA synthesis, changes in cell morphology and receptor endocytosis. The importance of de-regulated c-Abl cytoplasmic signalling in solid tumours is also discussed.
Collapse
|
200
|
Activation of ERK5 in angiotensin II-induced hypertrophy of human aortic smooth muscle cells. Mol Cell Biochem 2008; 322:171-8. [PMID: 19011954 DOI: 10.1007/s11010-008-9954-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2008] [Accepted: 10/28/2008] [Indexed: 12/17/2022]
Abstract
Extracellular signal-regulated kinase 5 (ERK5), a recently discovered mitogen-activated protein kinase (MAPK), plays a key role in the development and pathogenesis of cardiovascular disease. In order to clarify the pathophysiological significance of ERK5 in vascular remodeling, we investigated ERK5 phosphorylation in hypertrophy of human aortic smooth muscle cells (HASMCs) induced by angiotensin II (Ang II). The AT1 receptor was involved in Ang II-induced ERK5 activity. Hypertrophy was detected by the measurement of protein synthesis with [(3)H]-Leu incorporation in cultured HASMCs. Ang II rapidly induced phosphorylation of ERK5 at Thr218/Tyr220 residues in a time- and dose-dependent manner. Activation of myocyte enhancer factor-2C (MEF2C) by ERK5 was inhibited by PD98059. Transfecting HASMCs with small interfering RNA (siRNA) to silence ERK5 inhibited Ang II-induced cell hypertrophy. Thus, ERK5 phosphorylation contributes to MEF2C activation and subsequent HASMC hypertrophy induced by Ang II, for a novel molecular mechanism in cardiovascular diseases induced by Ang II.
Collapse
|