1
|
Li Y, Yu P, Zou Y, Cai W, Sun W, Han N. KRas-ERK signalling promotes the onset and maintenance of uveal melanoma through regulating JMJD6-mediated H2A.X phosphorylation at tyrosine 39. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 47:4257-4265. [PMID: 31736361 DOI: 10.1080/21691401.2019.1673764] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Since DNA damage is a first incident occurred during a tumour attack, it is rational that histone H2A.X phosphorylation on tyrosine 39 (H2A.XY39ph) may act as a tumour-relevant factor. This study was aimed to test the authenticity of the hypothesis. Uveal melanoma MP65 cells were transfected for expression of KRas mutated. H2A.X phosphorylation and ERK1/2 was measured, and transwell experiment was performed to examine the consequents of H2A.XY39ph on MP65 cells developing and migration. Regulatory relationship between H2A.XY39ph and ERK1/2 downstream genes were measured. Moreover, whether JMJD6 and MDM2 are involved in H2A.X phosphorylation was studied. Mutation of Ras activated ERK1/2 signalling and inhibited H2A.X phosphorylation at Y39. Silence of H2A.XY39ph contributed to the regulation of MP65 cells growth, migration and transcription of ERK1/2 downstream genes, including CYR61, IGFBP3, WNT16B, NT5E, GDF15 and CARD16. The repressed H2A.X phosphorylation through Ras-ERK1/2 signalling might be through MDM2-mediated JMJD6 degradation. Our study suggested that Ras-ERK1/2 signalling inhibited H2A.X phosphorylation at Y39, which led to the uncontrolled developing and migration of uveal melanoma cells. In addition, H2A.X phosphorylation was mediated possibly through JMJD6 which could be degraded by MDM2.
Collapse
Affiliation(s)
- Yaping Li
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, PR China
| | - Peng Yu
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, PR China
| | - Ying Zou
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, PR China
| | - Wenrui Cai
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, PR China
| | - Weixuan Sun
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, PR China
| | - Ning Han
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, PR China
| |
Collapse
|
2
|
Tian P, Zhu Y, Zhang C, Guo X, Zhang P, Xue H. Ras-ERK1/2 signaling contributes to the development of colorectal cancer via regulating H3K9ac. BMC Cancer 2018; 18:1286. [PMID: 30577849 PMCID: PMC6303919 DOI: 10.1186/s12885-018-5199-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 12/09/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUNDS/AIMS Ras is a control switch of ERK1/2 pathway, and hyperactivation of Ras-ERK1/2 signaling appears frequently in human cancers. However, the molecular regulation following by Ras-ERK1/2 activation is still unclear. This work aimed to reveal whether Ras-ERK1/2 promoted the development of colorectal cancer via regulating H3K9ac. METHODS A vector for expression of K-Ras mutated at G12 V and T35S was transfected into SW48 cells, and the acetylation of H3K9 was measured by Western blot analysis. MTT assay, colony formation assay, transwell assay, chromatin immunoprecipitation and RT-qPCR were performed to detect whether H3K9ac was contributed to K-Ras-mediated cell growth and migration. Furthermore, whether HDAC2 and PCAF involved in modification of H3K9ac following Ras-ERK1/2 activation were studied. RESULTS K-Ras mutated at G12 V and T35S induced a significant activation of ERK1/2 signaling and a significant down-regulation of H3K9ac. Recovering H3K9 acetylation by using a mimicked H3K9ac expression vector attenuated the promoting effects of Ras-ERK1/2 on tumor cells growth and migration. Besides, H3K9ac can be deacetylated by HDAC2 and MDM2-depedent degradation of PCAF. CONCLUSION H3K9ac was a specific target for Ras-ERK1/2 signaling pathway. H3K9 acetylation can be modulated by HDAC2 and MDM2-depedent degradation of PCAF. The revealed regulation provides a better understanding of Ras-ERK1/2 signaling in tumorigenesis.
Collapse
Affiliation(s)
- Peng Tian
- Department of Gastrointestinal Surgery, Zhengzhou University People's Hospital (Henan Provincial People's Hospital), Zhengzhou, 450003, China
| | - Yanfei Zhu
- Department of General Surgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214023, China
| | - Chao Zhang
- Department of Gastrointestinal Surgery, Zhengzhou University People's Hospital (Henan Provincial People's Hospital), Zhengzhou, 450003, China
| | - Xinyu Guo
- Department of Gastrointestinal Surgery, Zhengzhou University People's Hospital (Henan Provincial People's Hospital), Zhengzhou, 450003, China
| | - Peng Zhang
- Department of Gastrointestinal Surgery, Zhengzhou University People's Hospital (Henan Provincial People's Hospital), Zhengzhou, 450003, China
| | - Huanzhou Xue
- Department of General Surgery, Zhengzhou University People's Hospital (Henan Provincial People's Hospital), No.7, Weiwu Road, Zhengzhou, 450003, Henan, China.
| |
Collapse
|
3
|
The Ras-ERK pathway modulates cytoskeleton organization, cell motility and lung metastasis signature genes in MDA-MB-231 LM2. Oncogene 2013; 33:3668-76. [PMID: 23995792 DOI: 10.1038/onc.2013.341] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 06/24/2013] [Accepted: 07/01/2013] [Indexed: 02/07/2023]
Abstract
MDA-MB-231 LM2 (herein referred to as LM2) is a derivative of MDA-MB-231 cells that was selected for its ability to metastasize to lung tissue in vivo. We investigated cellular properties of LM2 including actin cytoskeleton organization, motility and signaling pathways that drive the expression of genes associated with the lung metastasis signature. Parental cells exhibit well-developed stress fibers, whereas LM2 had poorly organized stress fibers. LM2 exhibited higher levels of K-Ras protein and corresponding higher levels of phosphorylated ERK compared with parental cells. The Ras-ERK pathway was responsible for the disruption of stress fibers because inhibition of MEK with UO126 or small interfering RNA (siRNA) against K-Ras or ERK1/2 resulted in restoration of stress fibers and focal adhesions. We observed that the K-Ras-ERK pathway is important for the expression of genes associated with the lung metastasis signature. Paradoxically, inhibition of the Ras-ERK pathway did not result in inhibition of cell motility but was accompanied by activation of the phosphatidylinositol 3-kinase (PI3K) pathway. Inhibition of both ERK and PI3K pathways was required to inhibit motility of LM2 cells. These results suggest that both ERK and PI3K pathways drive motile functions of metastatic LM2 cells and genes associated with the lung metastasis signature.
Collapse
|
4
|
From the Cover: Neutralization of terminal differentiation in gliomagenesis. Proc Natl Acad Sci U S A 2013; 110:14520-7. [PMID: 23918370 DOI: 10.1073/pnas.1308610110] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
An immature state of cellular differentiation--characterized by stem cell-like tendencies and impaired differentiation--is a hallmark of cancer. Using glioblastoma multiforme (GBM) as a model system, we sought to determine whether molecular determinants that drive cells toward terminal differentiation are also genetically targeted in carcinogenesis and whether neutralizing such genes also plays an active role to reinforce the impaired differentiation state and promote malignancy. To that end, we screened 71 genes with known roles in promoting nervous system development that also sustain copy number loss in GBM through antineoplastic assay and identified A2BP1 (ataxin 2 binding protein 1, Rbfox1), an RNA-binding and splicing regulator that is deleted in 10% of GBM cases. Integrated in silico analysis of GBM profiles to elucidate the A2BP1 pathway and its role in glioma identified myelin transcription factor 1-like (Myt1L) as a direct transcriptional regulator of A2BP1. Reintroduction of A2BP1 or Myt1L in GBM cell lines and glioma stem cells profoundly inhibited tumorigenesis in multiple assays, and conversely, shRNA-mediated knockdown of A2BP1 or Myt1L in premalignant neural stem cells compromised neuronal lineage differentiation and promoted orthotopic tumor formation. On the mechanistic level, with the top-represented downstream target TPM1 as an illustrative example, we demonstrated that, among its multiple functions, A2BP1 serves to regulate TPM1's alternative splicing to promote cytoskeletal organization and terminal differentiation and suppress malignancy. Thus, in addition to the activation of self-renewal pathways, the neutralization of genetic programs that drive cells toward terminal differentiation may also promote immature and highly plastic developmental states that contribute to the aggressive malignant properties of GBM.
Collapse
|
5
|
Choi C, Kim D, Kim S, Jeong S, Song E, Helfman DM. From skeletal muscle to cancer: insights learned elucidating the function of tropomyosin. J Struct Biol 2011; 177:63-9. [PMID: 22119848 DOI: 10.1016/j.jsb.2011.11.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 11/08/2011] [Accepted: 11/09/2011] [Indexed: 12/17/2022]
Abstract
The tropomyosins (Tms) are a family of actin filament binding proteins that possess a simple dimeric α-helical coiled-coil structure along their entire length. Our knowledge of Tm structure and function has greatly expanded since they were first discovered in skeletal muscle almost 65 years ago. In multicellular organisms they exhibit extensive cell type specific isoform diversity. In this essay we discuss the genetic mechanisms by which this diversity is generated and its significance to actin-based cellular functions.
Collapse
Affiliation(s)
- Cheolwon Choi
- Department of Biological Sciences, Korean Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | | | | | | | | | | |
Collapse
|
6
|
Zare M, Jazii FR, Soheili ZS, Moghanibashi MM. Downregulation of tropomyosin-1 in squamous cell carcinoma of esophagus, the role of Ras signaling and methylation. Mol Carcinog 2011; 51:796-806. [DOI: 10.1002/mc.20847] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Revised: 07/08/2011] [Accepted: 07/27/2011] [Indexed: 12/16/2022]
|
7
|
Helfman DM, Flynn P, Khan P, Saeed A. Tropomyosin as a regulator of cancer cell transformation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 644:124-31. [PMID: 19209818 DOI: 10.1007/978-0-387-85766-4_10] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Tropomyosins (Tms) are among the most studied structural proteins of the actin cytoskeleton that are implicated in neoplastic-specific alterations in actin filament organization. Decreased expression of specific nonmuscle Tm isoforms is commonly associated with the transformed phenotype. These changes in Tm expression appear to contribute to the rearrangement of microfilament bundles and morphological alterations, increased cell motility and oncogenic signaling properties of transformed cells. Below we review aspects of Tm biology as it specifically relates to transformation and cancer including its expression in culture models of transformed cells and human tumors, mechanisms that regulate Tm expression and the role of Tm in oncogenic signaling.
Collapse
Affiliation(s)
- David M Helfman
- Department of Cell Biology and Anatomy, Sylvester Comprehensive Cancer Center, Leonard M. Miller School of Medicine, Papanicolaou Building, Room 317, 1550 NW 10th Avenue (M-877), Miami, Florida 33136, USA.
| | | | | | | |
Collapse
|
8
|
Gunning P, O'Neill G, Hardeman E. Tropomyosin-based regulation of the actin cytoskeleton in time and space. Physiol Rev 2008; 88:1-35. [PMID: 18195081 DOI: 10.1152/physrev.00001.2007] [Citation(s) in RCA: 368] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Tropomyosins are rodlike coiled coil dimers that form continuous polymers along the major groove of most actin filaments. In striated muscle, tropomyosin regulates the actin-myosin interaction and, hence, contraction of muscle. Tropomyosin also contributes to most, if not all, functions of the actin cytoskeleton, and its role is essential for the viability of a wide range of organisms. The ability of tropomyosin to contribute to the many functions of the actin cytoskeleton is related to the temporal and spatial regulation of expression of tropomyosin isoforms. Qualitative and quantitative changes in tropomyosin isoform expression accompany morphogenesis in a range of cell types. The isoforms are segregated to different intracellular pools of actin filaments and confer different properties to these filaments. Mutations in tropomyosins are directly involved in cardiac and skeletal muscle diseases. Alterations in tropomyosin expression directly contribute to the growth and spread of cancer. The functional specificity of tropomyosins is related to the collaborative interactions of the isoforms with different actin binding proteins such as cofilin, gelsolin, Arp 2/3, myosin, caldesmon, and tropomodulin. It is proposed that local changes in signaling activity may be sufficient to drive the assembly of isoform-specific complexes at different intracellular sites.
Collapse
Affiliation(s)
- Peter Gunning
- Oncology Research Unit, The Children's Hospital at Westmead, and Muscle Development Unit, Children's Medical Research Institute, Westmead; New South Wales, Australia.
| | | | | |
Collapse
|
9
|
Tropomyosin Gene Expression in Vivo and in Vitro. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008. [DOI: 10.1007/978-0-387-85766-4_4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
10
|
Tropomyosins as interpreters of the signalling environment to regulate the local cytoskeleton. Semin Cancer Biol 2007; 18:35-44. [PMID: 17942320 DOI: 10.1016/j.semcancer.2007.08.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2007] [Accepted: 08/28/2007] [Indexed: 01/11/2023]
Abstract
A key regulator of cell morphology is the actin cytoskeleton and it has long been appreciated that the cytoskeleton is characteristically altered in cancer. Actin is organized into polymeric structures with distinct dynamics which in turn participate in a wide variety of cell processes including adhesion, migration, cell division and apoptosis. Despite displaying an altered actin cytoskeleton, transformed cells retain--and in many cases increase--their ability to adhere, move, divide and respond to apoptotic stimuli. Thus cancer cells maintain responsive actin cytoskeletons. Actin dynamics are regulated by numerous actin-binding proteins and chief among these are the tropomyosins which are core components of the microfilament. Recent advances in genomic and proteomic profiling confirm that Tm expression profiles are profoundly changed in transformed cells. It is therefore timely to review the role of Tms in the regulation of actin dynamics that pertain to crucial phenotypic changes in cancer. In this review we discuss how actin filaments containing different Tm isoforms respond to the activation of cell signalling pathways and consider the implications of this for cancer progression and therapy.
Collapse
|
11
|
McMichael BK, Kotadiya P, Singh T, Holliday LS, Lee BS. Tropomyosin isoforms localize to distinct microfilament populations in osteoclasts. Bone 2006; 39:694-705. [PMID: 16765662 DOI: 10.1016/j.bone.2006.04.031] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2005] [Revised: 03/15/2006] [Accepted: 04/04/2006] [Indexed: 12/26/2022]
Abstract
Osteoclasts resorb bone through transient rearrangement of their cytoskeletons to create a polarized phenotype in which an apical ruffled membrane is surrounded by a ring of F-actin that creates a tight seal against bone substrate. This process, coupled with the capacity for rapid motility, necessitates the presence of a dynamic, multi-functional actin cytoskeleton. Tropomyosins are a large class of actin-binding proteins that can regulate microfilament stability and organization by recruiting other regulatory proteins to actin, or alternately, by inhibiting their binding. Tropomyosins are expressed from four distinct genes (alpha, beta, gamma, and delta) that are alternately spliced to produce over forty isoforms. In recent years, it has become clear that nonmuscle isoforms of tropomyosin may be differentially distributed among intracellular pools of F-actin possessing different functions. Here we have used Western analysis and immunocytochemistry coupled with confocal microscopy to identify the isoforms of tropomyosin expressed by osteoclasts, as well as their distributions within cells. Osteoclasts express at least seven isoforms with markedly different distributions. The products of the alpha gene (Tm-2, -3, and -5a/5b) are up-regulated during osteoclastogenesis, indicating potential cell-specific functions. Some isoforms (Tm-5a/5b, Tm-4) are specifically enriched within and around osteoclast attachment structures, the sealing zone and podosomes, whereas others are more abundant in internal regions of the cell. This compartmentalization of tropomyosins to specific actin structures within osteoclasts is likely to play a critical role in determining the dynamic properties of the actin cytoskeleton and thus osteoclast activity.
Collapse
Affiliation(s)
- Brooke K McMichael
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | |
Collapse
|
12
|
Abstract
Hypertrophied myocardium is associated with reductions in the transient outward K(+) current (Ito) and expression of pore-forming Kv4.2/4.3 and auxiliary KChIP2 subunits. Here we show that KChIP2 mRNA and protein levels are dramatically decreased to 10% to 30% of control levels in the left ventricle of aorta-constricted rats in vivo and phenylephrine (PE)-treated myocytes in vitro. PE also markedly decreases Ito density. Inhibition of protein kinase Cs (PKCs) does not affect the PE-induced reduction in KChIP2 mRNA level, whereas activation of PKC with phorbol ester (phorbol myristate [PMA]) causes a marked reduction in KChIP2 mRNA level. Pharmacological inhibition of MEKs or overexpression of a dominant-negative MEK1 increases the basal KChIP2 mRNA expression and blocks the PMA-induced decrease in auxiliary subunit mRNA level. In addition, a constitutively active MEK1 decreases the basal KChIP2 mRNA level, and PMA causes no further reduction in auxiliary subunit mRNA level in active MEK1-expressing cells. Furthermore, pharmacological inhibition of JNKs or overexpression of a dominant-negative JNK1 prevents the PE-induced, but not PMA-induced, reduction in KChIP2 mRNA expression. These results suggest that downregulation of KChIP2 expression significantly contributes to the hypertrophy-associated reduction in Ito density. They also indicate that the expression of KChIP2 mRNA is controlled by the 2 branches of mitogen-activated protein kinase pathways: JNKs play a predominant role in mediating the PE-induced reduction, whereas the MEK-ERK pathway influences the basal expression and mediates the PKC-mediated downregulation.
Collapse
Affiliation(s)
| | - Koichi Takimoto
- Correspondence to Koichi Takimoto, Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, 3343 Forbes Ave, Pittsburgh, PA 15260. E-mail
| |
Collapse
|
13
|
Sukezane T, Oneyama C, Kakumoto K, Shibutani K, Hanafusa H, Akagi T. Human diploid fibroblasts are resistant to MEK/ERK-mediated disruption of the actin cytoskeleton and invasiveness stimulated by Ras. Oncogene 2005; 24:5648-55. [PMID: 16007212 DOI: 10.1038/sj.onc.1208724] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Ras-induced transformation is characterized not only by uncontrolled proliferation but also by drastic morphological changes accompanied by the disruption of the actin cytoskeleton. Previously, we reported that human fibroblasts are more resistant than rodent fibroblasts to Ras-induced transformation. To explore the molecular basis for the difference in susceptibility to Ras-induced transformation, we investigated the effect of activated H-Ras on the actin cytoskeleton in human diploid fibroblasts and in rat embryo fibroblasts, both of which are immortalized by SV40 early region. We demonstrate here that Ras-induced morphological changes, decreased expression of tropomyosin isoforms, and suppression of the ROCK/LIMK/Cofilin pathway observed in the rat fibroblasts were not detected in the human fibroblasts even with high expression levels of Ras. We also show that activation of the MEK/ERK pathway sufficed to induce all of these alterations in the rat fibroblasts, whereas the human fibroblasts were refractory to these MEK/ERK-mediated changes. In addition to morphological changes, we demonstrated that the expression of activated Ras induced an invasive phenotype in the rat, but not in the human fibroblasts. These studies provide evidence for the existence of human-specific mechanisms that resist Ras/MEK/ERK-mediated transformation.
Collapse
Affiliation(s)
- Taiko Sukezane
- Laboratory of Molecular Oncology, Osaka Bioscience Institute, 6-2-4 Furuedai, Suita, Osaka 565-0874, Japan
| | | | | | | | | | | |
Collapse
|
14
|
Akula SM, Ford PW, Whitman AG, Hamden KE, Shelton JG, McCubrey JA. Raf promotes human herpesvirus-8 (HHV-8/KSHV) infection. Oncogene 2004; 23:5227-41. [PMID: 15122343 DOI: 10.1038/sj.onc.1207643] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Human herpesvirus-8 (HHV-8/KSHV) is etiologically associated with Kaposi's sarcoma (KS) and other tumors. Constitutive activation of the mitogen-activated protein kinase (MAPK) signaling pathway has been associated with a variety of tumors, including AIDS-related KS. The oncoprotein Raf is situated at a pivotal position in regulating the MAPK pathway. Hence, we analysed the effect of oncoprotein Raf on HHV-8 infectious entry into target cells. Here we report Raf expression to significantly enhance HHV-8 infection of target cells. These findings implicate a role for Raf not only in the infectious entry of HHV-8 but also in modulating KS pathogenesis.
Collapse
Affiliation(s)
- Shaw M Akula
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA.
| | | | | | | | | | | |
Collapse
|
15
|
Akimov SS, Belkin AM. Opposing roles of Ras/Raf oncogenes and the MEK1/ERK signaling module in regulation of expression and adhesive function of surface transglutaminase. J Biol Chem 2003; 278:35609-19. [PMID: 12832399 DOI: 10.1074/jbc.m303488200] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Tissue transglutaminase (tTG) serves as a potent and ubiquitous integrin-associated adhesion co-receptor for fibronectin on the cell surface and affects several key integrin functions. Here we report that in fibroblasts, activated H-Ras and Raf-1 oncogenes decrease biosynthesis, association with beta1 integrins, and surface expression of tTG because of down-regulation of tTG mRNA. In turn, the reduction of surface tTG inhibits adhesion of H-Ras- and Raf-1-transformed cells on fibronectin and, in particular, on its tTG-binding fragment I(6)II(1,2)I(7-9), which does not interact directly with integrins. Analysis of Ras/Raf downstream signaling with specific pharmacological inhibitors reveals that the decrease in tTG expression is mediated by the p38 MAPK, c-Jun NH2-terminal kinase, and phosphatidylinositol 3-kinase pathways. In contrast, increased activation of the ERK pathway by constitutively active MEK1 stimulates tTG mRNA expression, biosynthesis, and surface expression of tTG, whereas MEK inhibitors or dominant negative MEK1 exert an opposite effect. This modulation of surface tTG by ERK signaling alters adhesion of cells on fibronectin and its fragment that binds tTG. Furthermore, transient stimulation of ERK signaling in untransformed fibroblasts by adhesion on fibronectin or growth factors elevates tTG biosynthesis, increases complex formation with beta1 integrins, and raises surface expression of tTG. Finally, ERK activation is required for growth factor-induced redistribution of tTG on the surface of adherent fibroblasts and co-clustering of beta1 integrins and tTG at cell-matrix adhesion contacts. Together, our data indicate that down-regulation of surface tTG by Ras and Raf oncogenes contributes to adhesive deficiency of transformed fibroblasts, whereas stimulation of biosynthesis and surface expression of tTG by the MEK1/ERK module promotes and sustains cell-matrix adhesion of untransformed cells. Contrasting effects of Ras/Raf oncogenes and their immediate downstream signaling module, MEK1/ERK, on tTG expression are consistent with adhesive function of surface tTG.
Collapse
Affiliation(s)
- Sergey S Akimov
- Department of Biochemistry, The Holland Laboratory, American Red Cross, Rockville, Maryland 20855, USA
| | | |
Collapse
|
16
|
Shelton JG, Moye PW, Steelman LS, Blalock WL, Lee JT, Franklin RA, McMahon M, McCubrey JA. Differential effects of kinase cascade inhibitors on neoplastic and cytokine-mediated cell proliferation. Leukemia 2003; 17:1765-82. [PMID: 12970777 DOI: 10.1038/sj.leu.2403052] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The Raf/MEK/ERK and PI3K/Akt pathways regulate proliferation and prevent apoptosis, and their altered expression is commonly observed in human cancer due to the high mutation frequency of upstream regulators. In this study, the effects of Raf, MEK, and PI3K inhibitors on conditionally transformed hematopoietic cells were examined to determine if they would display cytotoxic differences between cytokine- and oncogene-mediated proliferation, and whether inhibition of both pathways was a more effective means to induce apoptosis. In the hematopoietic model system employed, proliferation was conditional and occurred when either interleukin-3 (IL-3) or the estrogen receptor antagonist 4-hydroxytamoxifen (4HT), which activates the conditional oncoprotein (DeltaRaf:ER), were provided. Thus, upon the addition of the signal transduction inhibitors and either IL-3 or 4HT, the effects of these drugs were examined in the same cell under 'cytokine-' and 'oncoprotein' -mediated growth conditions avoiding genetic and differentiation stage heterogeneity. At drug concentrations around the reported IC(50) for the Raf inhibitor L-779,450, it suppressed DNA synthesis and induced apoptosis in hematopoietic FDC-P1 cells transformed to grow in response to either Raf-1 or A-Raf (FD/DeltaRaf-1:ER and FD/DeltaA-Raf:ER), but it displayed less effects on DNA synthesis and apoptosis when the cells were cultured in IL-3. This Raf inhibitor was less effective on B-Raf- or MEK1-responsive cells, demonstrating the specificity of this drug. MEK inhibitors also suppressed DNA synthesis and induced apoptosis in Raf-responsive cells and the effects were more significant on Raf-responsive compared to cytokine-mediated growth. The PI3K inhibitor LY294002 suppressed Raf-mediated growth, indicating that part of the long-term proliferative effects mediated by Raf are PI3K dependent. Simultaneous inhibition of both Raf/MEK/ERK and PI3K/Akt pathways proved a more efficient means to suppress DNA synthesis and induce apoptosis at lower drug concentrations.
Collapse
Affiliation(s)
- J G Shelton
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville 27858, USA
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Carystinos GD, Kandouz M, Alaoui-Jamali MA, Batist G. Unexpected induction of the human connexin 43 promoter by the ras signaling pathway is mediated by a novel putative promoter sequence. Mol Pharmacol 2003; 63:821-31. [PMID: 12644583 DOI: 10.1124/mol.63.4.821] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Connexin 43 (Cx43) is essential for survival and is tightly regulated at the transcriptional and post-transcriptional levels. A number of previous studies have demonstrated altered expression in malignant tissues, and in the presence of carcinogenic factors. We examined the effect of protooncogenes of Cx43 expression, and found no effect on Cx43 promoter activity in cells transformed with Src or erbB2. On the other hand, we identified and characterized a novel sequence that mediates Cx43 promoter regulation in cell lines engineered to overexpress H-Ras. Compared with wild-type NIH3T3 cells, both Cx43 mRNA and protein levels are increased in NIH3T3-Ras cells. The H-Ras+ cells also have enhanced Cx43 promoter activation, which is inhibited by the MEK1 inhibitor 2'-amino-3'-methoxyflavone (PD98059), suggesting that Ras-mediated Cx43 overexpression is via the mitogen activated protein kinase kinase/extracellular signal-regulated pathway. Deletion analysis of the Cx43 promoter revealed a 200-bp region downstream of the Cx43 transcription start site as the minimal sequence essential for the Ras-mediated Cx43 up-regulation. Using this 200-base pair fragment in electrophoretic mobility shift assays, we identified one main protein complex that binds efficiently and is more abundant in nuclear extracts from NIH3T3-Ras and MCF7-Ras cells compared with their matched controls. This complex selectively recognizes a consensus sequence, AGTTCAATCA, located at positions +149 to +158 of the Cx43 promoter. Supershift assays identified the 90-kDa heat shock protein (HSP90) and c-Myc as constituents of this DNA-binding complex. Treatment of cells with the HSP90 inhibitor geldanamycin resulted in repression of the Cx43 promoter activity, and inhibits binding of the complex to the Cx43 promoter. Coimmunoprecipitation studies confirmed the interaction between endogenous HSP90 and c-Myc. This study provides evidence that the transcriptional up-regulation of Cx43 by Ras-Raf-MAPK is mediated via the interaction of a novel Cx43 promoter element with a protein complex that contains both HSP90 and c-Myc.
Collapse
Affiliation(s)
- George D Carystinos
- Department of Pharmacology & Therapeutics and the Montreal Centre for Experimental Therapeutics in Cancer, Lady Davis Institute of the Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, Canada
| | | | | | | |
Collapse
|
18
|
Janssen RAJ, Kim PN, Mier JW, Morrison DK. Overexpression of kinase suppressor of Ras upregulates the high-molecular-weight tropomyosin isoforms in ras-transformed NIH 3T3 fibroblasts. Mol Cell Biol 2003; 23:1786-97. [PMID: 12588996 PMCID: PMC151698 DOI: 10.1128/mcb.23.5.1786-1797.2003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2002] [Revised: 10/10/2002] [Accepted: 12/12/2002] [Indexed: 01/08/2023] Open
Abstract
The down-regulation of the high-molecular-weight isoforms of tropomyosin (TM) is considered to be an essential event in cellular transformation. In ras-transformed fibroblasts, the suppression of TM is dependent on the activity of the Raf-1 kinase; however, the requirement for other downstream effectors of Ras, such as MEK and ERK, is less clear. In this study, we have utilized the mitogen-activated protein kinase scaffolding protein Kinase Suppressor of Ras (KSR) to further investigate the regulation of TM and to clarify the importance of MEK/ERK signaling in this process. Here, we report that overexpression of wild-type KSR1 in ras-transformed fibroblasts restores TM expression and induces cell flattening and stress fiber formation. Moreover, we find that the transcriptional activity of a TM-alpha promoter is decreased in ras-transformed cells and that the restoration of TM by KSR1 coincides with increased transcription from this promoter. Although ERK activity was suppressed in cells overexpressing KSR1, ERK inhibition alone was insufficient to upregulate TM expression. The KSR1-mediated effects on stress fiber formation and TM transcription required the activity of the ROCK kinase, because these effects could be suppressed by the ROCK inhibitor, Y27632. Overexpression of KSR1 did not directly regulate ROCK activity, but did permit the recoupling of ROCK to the actin polymerization machinery. Finally, all of the KSR1-induced effects were mediated by the C-terminal domain of KSR1 and were dependent on the KSR-MEK interaction.
Collapse
Affiliation(s)
- Richard A J Janssen
- Regulation of Cell Growth Laboratory, National Cancer Institute-Frederick, Frederick, Maryland 21702, USA.
| | | | | | | |
Collapse
|
19
|
Feldker DEM, Datson NA, Veenema AH, Meulmeester E, de Kloet ER, Vreugdenhil E. Serial analysis of gene expression predicts structural differences in hippocampus of long attack latency and short attack latency mice. Eur J Neurosci 2003; 17:379-87. [PMID: 12542675 DOI: 10.1046/j.1460-9568.2003.02440.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The genetically selected long attack latency (LAL) and short attack latency (SAL) mice differ in a wide variety of behavioural traits and display differences in the serotonergic system and the hypothalamus-pituitary-adrenocortical (HPA)-axis. Serial analysis of gene expression (SAGE) was used to generate a hippocampal expression profile of almost 30 000 genes in LAL and SAL mice. Using SAGE, we found differential expression of 191 genes. Among these were genes involved in growth, signal transduction, and cell metabolism. The SAGE study was supported by GeneChip analysis (Affymetrix). Strikingly, both SAGE and GeneChips showed a higher expression of numerous cytoskeleton genes, such as cofilin and several tubulin isotypes in LAL mice. LAL mice also showed a higher expression of several calmodulin-related genes and genes encoding components of a MAPK cascade, namely raf-related oncogene and ERK2. The findings were confirmed by in situ hybridization. Our results of differential expression of cytoskeleton and signal transduction genes therefore suggest differential regulation of the raf/ERK pathway that may be related to structural differences in the hippocampus of LAL and SAL mice. As stress-related disorders, such as depression, are also linked to differential regulation of the HPA-axis and the serotonergic system and are associated with altered hippocampal morphology, differential regulation of these genes may be involved in the pathogenesis of these diseases.
Collapse
Affiliation(s)
- Dorine E M Feldker
- Division of Medical Pharmacology, Leiden/Amsterdam Center for Drug Research, Leiden University Medical Centre, PO Box 9502, 2300 RA Leiden, The Netherlands.
| | | | | | | | | | | |
Collapse
|
20
|
Bharadwaj S, Prasad GL. Tropomyosin-1, a novel suppressor of cellular transformation is downregulated by promoter methylation in cancer cells. Cancer Lett 2002; 183:205-13. [PMID: 12065096 DOI: 10.1016/s0304-3835(02)00119-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Tropomyosins (TMs) are a family of microfilament binding proteins, which are suppressed in the transformed cells. We have investigated the mechanism of suppression of TMs, in particular that of tropomyosin-1 (TM1), in breast cancer cells. Inhibition of DNA methyl transferase with 5-aza-2'-deoxycytidine (AZA) alone did not induce TM1 expression. However, combined treatment of trichostatin A (TSA) and AZA resulted in readily detectable expression of TM1, but not that of other TM isoforms. Upregulation of TM1 expression paralleled with the reemergence of TM1 containing microfilaments, and in abolition of anchorage-independent growth. The synergistic action of AZA and TSA in reactivation of TM1 gene was also evident in ras-transformed fibroblasts. These data, for the first time, show that hypermethylation of TM1 gene and chromatin remodeling are the predominant mechanisms by which TM1 expression is downregulated in breast cancer cells.
Collapse
Affiliation(s)
- Shantaram Bharadwaj
- Department of General Surgery, Medical Center Boulevard, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | |
Collapse
|
21
|
Pawlak G, Helfman DM. Post-transcriptional down-regulation of ROCKI/Rho-kinase through an MEK-dependent pathway leads to cytoskeleton disruption in Ras-transformed fibroblasts. Mol Biol Cell 2002. [PMID: 11809843 DOI: 10.1091/mbc.01-02-0302] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Transformation by oncogenic Ras profoundly alters actin cytoskeleton organization. We investigated Ras-dependent signaling pathways involved in cytoskeleton disruption by transfecting normal rat kidney (NRK) cells with different Ras mutants. RasV12S35, a mutant known to activate specifically the Raf/MAPK pathway, led to stress fiber and focal contact disruption, whereas the adherens junctions remained intact. Next, we found that pharmacological inhibition of MEK was sufficient to restore the cytoskeletal defects of ras-transformed NRK cells, including assembly of stress fibers and focal contacts, but it did not induce reorganization of the cell-cell junctions. Investigating the mechanism underlying this phenotypic reversion, we found that the sustained MAPK signaling resulting from Ras-transformation down-regulated the expression of ROCKI and Rho-kinase, two-Rho effectors required for stress fiber formation, at the post-transcriptional level. On MEK inhibition, ROCKI/Rho-kinase expression and cofilin phosphorylation were increased, demonstrating that the Rho-kinase/LIM-kinase/cofilin pathway was functionally restored. Finally, using dominant negative or constitutively active mutants, we demonstrated that expression of ROCKI/Rho-kinase was both necessary and sufficient to promote cytoskeleton reorganization in NRK/ras cells. These findings further establish the Ras/MAPK pathway as the critical pathway involved in cytoskeleton disruption during Ras-transformation, and they suggest a new mechanism, involving alteration in ROCKI/Rho-kinase expression, by which oncogenic Ras can specifically target the actin-based cytoskeleton and achieve morphological transformation of the cells.
Collapse
Affiliation(s)
- Geraldine Pawlak
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | |
Collapse
|
22
|
Shields JM, Mehta H, Pruitt K, Der CJ. Opposing roles of the extracellular signal-regulated kinase and p38 mitogen-activated protein kinase cascades in Ras-mediated downregulation of tropomyosin. Mol Cell Biol 2002; 22:2304-17. [PMID: 11884615 PMCID: PMC133695 DOI: 10.1128/mcb.22.7.2304-2317.2002] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
We showed previously that activated Ras, but not Raf, causes transformation of RIE-1 epithelial cells, demonstrating the importance of Raf-independent pathways in mediating Ras transformation. To assess the mechanism by which Raf-independent effector signaling pathways contribute to Ras-mediated transformation, we recently utilized representational difference analysis to identify genes expressed in a deregulated fashion by activated Ras but not Raf. One gene identified in these analyses encodes for alpha-tropomyosin. Therefore, we evaluated the mechanism by which Ras causes the downregulation of tropomyosin expression. By using RIE-1 cells that harbor inducible expression of activated H-Ras(12V), we determined that the downregulation of tropomyosin expression correlated with the onset of morphological transformation. We found that the reversal of Ras transformation caused by inhibition of extracellular signal-regulated kinase activation corresponded to a restoration of tropomyosin expression. Inhibition of p38 activity in Raf-expressing RIE-1 cells caused both morphological transformation and loss of tropomyosin expression. Thus, a reduction in tropomyosin expression correlated strictly with morphological transformation of RIE-1 cells. However, forced overexpression of tropomyosin in Ras-transformed cells did not reverse morphological or growth transformation, a finding consistent with the possibility that multiple changes in gene expression contribute to Ras transformation. We also determined that tropomyosin expression was low in two human tumor cell lines, DLD-1 and HT1080, that harbor endogenous mutated alleles of ras, but high in transformation-impaired, derivative cell lines in which the mutant ras allele has been genetically deleted. Finally, treatment with azadeoxycytidine restored tropomyosin expression in Ras-transformed RIE-1, HT1080, and DLD-1 cells, suggesting a role for DNA methylation in downregulating tropomyosin expression.
Collapse
Affiliation(s)
- Janiel M Shields
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 27599, USA.
| | | | | | | |
Collapse
|
23
|
Shields JM, Rogers-Graham K, Der CJ. Loss of transgelin in breast and colon tumors and in RIE-1 cells by Ras deregulation of gene expression through Raf-independent pathways. J Biol Chem 2002; 277:9790-9. [PMID: 11773051 DOI: 10.1074/jbc.m110086200] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Activated Ras but not Raf can transform RIE-1 and other epithelial cells, indicating the critical importance of Raf-independent effector function in Ras transformation of epithelial cells. To elucidate the nature of these Raf-independent activities, we utilized representational difference analysis to identify genes aberrantly expressed by Ras through Raf-independent mechanisms in RIE-1 cells. We identified a total of 22 genes, both known and novel, whose expression was either activated or abolished by Ras but not Raf. The genes up-regulated encode proteins involved in protein or DNA synthesis, regulation of protease activity, or ligand binding, whereas those genes down-regulated encode actin cytoskeletal-, extracellular matrix-, and gap junction-associated proteins, and transmembrane receptor- or cytokine-like proteins. These results suggest that a key function of Raf-independent signaling involves deregulation of gene expression. We further characterized transgelin as a gene whose expression was abolished by Ras. Transgelin was identified previously as a protein whose expression was lost in virally transformed cell lines. We show that this loss is regulated at the level of gene expression and that both Raf-dependent and Raf-independent pathways are required to cause Ras down-regulation of transgelin in RIE-1 cells, whereas Raf alone is sufficient to cause its loss in NIH 3T3 fibroblasts. We also found that Ras-dependent and Ras-independent mechanisms can cause the down-regulation of transgelin in human breast and colon carcinoma cells lines and patient-derived tumor samples. We conclude that loss of transgelin gene expression may be an important early event in tumor progression and a diagnostic marker for breast and colon cancer development.
Collapse
Affiliation(s)
- Janiel M Shields
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599-7295, USA.
| | | | | |
Collapse
|
24
|
Schumann D, Chen CJ, Kaplan B, Shively JE. Carcinoembryonic antigen cell adhesion molecule 1 directly associates with cytoskeleton proteins actin and tropomyosin. J Biol Chem 2001; 276:47421-33. [PMID: 11595750 DOI: 10.1074/jbc.m109110200] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
CEA cell adhesion molecule 1 (CEACAM1), a type 1 transmembrane and homotypic cell adhesion protein belonging to the carcinoembryonic antigen (CEA) gene family and expressed on epithelial cells, is alternatively spliced to produce four major isoforms with three or four Ig-like ectodomains and either long (CEACAM1-L) or short (CEACAM1-S) cytoplasmic domains. When murine MC38 (methylcholanthrene-induced adenocarcinoma 38) cells were transfected with human CEACAM1-L and stimulated with sodium pervanadate, actin was found to co-localize with CEACAM1-L at cell-cell boundaries but not in untreated cells. When CEACAM1-L was immunoprecipitated from pervanadate-treated MC38/CEACAM1-L cells and the associated proteins were analyzed by two-dimensional gel analysis and mass spectrometry, actin and tropomyosin, among other proteins, were identified. Whereas a glutathione S-transferase (GST) fusion protein containing the l-isoform (GST-Cyto-L) bound poorly to F-actin in a co-sedimentation assay, the S-isoform fusion protein (GST-Cyto-S) co-sedimented with F-actin, especially when incubated with G-actin during polymerization (K(D) = 7.0 microm). Both GST-Cyto-S and GST-Cyto-L fusion proteins bind G-actin and tropomyosin by surface plasmon resonance studies with binding constants of 0.7 x 10(-8) and 1.0 x 10(-7) m for GST-Cyto-L to G-actin and tropomyosin, respectively, and 3.1 x 10(-8) and 1.3 x 10(-7) m for GST-Cyto-S to G-actin and tropomyosin, respectively. Calmodulin or EDTA inhibited binding of the GST-Cyto-L fusion protein to G-actin, whereas calmodulin and G-actin, but not EDTA, stimulated binding to tropomyosin. A biotinylated 14-amino acid peptide derived from the juxtamembrane portion of the cytoplasmic domain of CEACAM1-L associated with both G-actin and tropomyosin with K(D) values of 1.3 x 10(-5) and 1.8 x 10(-5) m, respectively. These studies demonstrate the direct interaction of CEACAM1 isoforms with G-actin and tropomyosin and the direct interaction of CEACAM1-S with F-actin.
Collapse
MESH Headings
- Actins/chemistry
- Actins/metabolism
- Amino Acids/chemistry
- Animals
- Antigens, CD/chemistry
- Antigens, CD/metabolism
- Antigens, Differentiation/chemistry
- Antigens, Differentiation/metabolism
- Biotinylation
- Carcinoembryonic Antigen
- Cell Adhesion
- Cell Adhesion Molecules
- Cytoplasm/metabolism
- Cytoskeleton/metabolism
- Dose-Response Relationship, Drug
- Electrophoresis, Gel, Two-Dimensional
- Electrophoresis, Polyacrylamide Gel
- Enzyme Inhibitors/pharmacology
- Epithelial Cells/metabolism
- Glutathione Transferase/metabolism
- Humans
- Kinetics
- Mass Spectrometry
- Mice
- Microscopy, Confocal
- Peptides/chemistry
- Precipitin Tests
- Protein Binding
- Protein Isoforms
- Protein Structure, Tertiary
- Recombinant Fusion Proteins/metabolism
- Recombinant Proteins/metabolism
- Surface Plasmon Resonance
- Time Factors
- Transfection
- Tropomyosin/chemistry
- Tropomyosin/metabolism
- Tumor Cells, Cultured
- Vanadates/pharmacology
Collapse
Affiliation(s)
- D Schumann
- Division of Immunology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | | | | | | |
Collapse
|
25
|
Sahai E, Olson MF, Marshall C. Cross-talk between Ras and Rho signalling pathways in transformation favours proliferation and increased motility. EMBO J 2001; 20:755-66. [PMID: 11179220 PMCID: PMC145410 DOI: 10.1093/emboj/20.4.755] [Citation(s) in RCA: 308] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Transformation by oncogenic Ras requires the function of the Rho family GTPases. We find that Ras-transformed cells have elevated levels of RhoA-GTP, which functions to inhibit the expression of the cell cycle inhibitor p21/Waf1. These high levels of Rho-GTP are not a direct consequence of Ras signalling but are selected for in response to sustained ERK-MAP kinase signalling. While the elevated levels of Rho-GTP control the level of p21/Waf, they no longer regulate the formation of actin stress fibres in transformed cells. We show that the sustained ERK-MAP kinase signalling resulting from transformation by oncogenic Ras down-regulates ROCK1 and Rho-kinase, two Rho effectors required for actin stress fibre formation. The repression of Rho- dependent stress fibre formation by ERK-MAP kinase signalling contributes to the increased motility of Ras-transformed fibroblasts. Overexpression of the ROCK target LIM kinase restores actin stress fibres and inhibits the motility of Ras-transformed fibroblasts. We propose a model in which Ras and Rho signalling pathways cross-talk to promote signalling pathways favouring transformation.
Collapse
Affiliation(s)
| | | | - C.J. Marshall
- CRC Centre for Cell and Molecular Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
Corresponding author e-mail:
| |
Collapse
|
26
|
Abstract
Research during the past couple of years has provided important new information as to how the actin cytoskeleton contributes to growth control in both normal and transformed cells. The cytoskeleton can no longer be viewed as simply a structural framework playing a role in cell shape and motile events such as cell movement, intracellular transport, contractile-ring formation and chromosome movement. More recent experiments show that the cytoskeleton plays a critical role in the regulation of various cellular processes linked to transformation including proliferation, contact inhibition, anchorage-independent cell growth, and apoptosis.
Collapse
Affiliation(s)
- G Pawlak
- Cold Spring Harbor Laboratory, PO Box 100, Cold Spring Harbor, New York 11724, USA.
| | | |
Collapse
|
27
|
Pearson G, Bumeister R, Henry DO, Cobb MH, White MA. Uncoupling Raf1 from MEK1/2 impairs only a subset of cellular responses to Raf activation. J Biol Chem 2000; 275:37303-6. [PMID: 11018021 DOI: 10.1074/jbc.c000570200] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Raf family of serine/threonine protein kinases is intimately involved in the transmission of cell regulatory signals controlling proliferation and differentiation. The best characterized Raf substrates are MEK1 and MEK2. The activation of MEK1/2 by Raf is required to mediate many of the cellular responses to Raf activation, suggesting that MEK1/2 are the dominant Raf effector proteins. However, accumulating evidence suggests that there are additional Raf substrates and that subsets of Raf-induced regulatory events are mediated independently of Raf activation of MEK1/2. To examine the possibility that there is bifurcation at the level of Raf in activation of MEK1/2-dependent and MEK1/2-independent cell regulatory events, we engineered a kinase-active Raf1 variant (RafBXB(T481A)) with an amino acid substitution that disrupts MEK1 binding. We find that disruption of MEK1/2 association uncouples Raf from activation of ERK1/2, induction of serum-response element-dependent gene expression, and induction of growth and morphological transformation. However, activation of NF-kappaB-dependent gene expression and induction of neurite differentiation were unimpaired. In addition, Raf-dependent activation of p90 ribosomal S6 kinase was only slightly impaired. These results support the hypothesis that Raf kinases utilize multiple downstream effectors to regulate distinct cellular activities.
Collapse
Affiliation(s)
- G Pearson
- Departments of Cell Biology and Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | | | | | | | |
Collapse
|
28
|
Lawrence B, Perez-Atayde A, Hibbard MK, Rubin BP, Dal Cin P, Pinkus JL, Pinkus GS, Xiao S, Yi ES, Fletcher CDM, Fletcher JA. TPM3-ALK and TPM4-ALK oncogenes in inflammatory myofibroblastic tumors. THE AMERICAN JOURNAL OF PATHOLOGY 2000; 157:377-84. [PMID: 10934142 PMCID: PMC1850130 DOI: 10.1016/s0002-9440(10)64550-6] [Citation(s) in RCA: 487] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Inflammatory myofibroblastic tumors (IMTs) are neoplastic mesenchymal proliferations featuring an inflammatory infiltrate composed primarily of lymphocytes and plasma cells. The myofibroblastic cells in some IMTs contain chromosomal rearrangements involving the ALK receptor tyrosine-kinase locus region (chromosome band 2p23). ALK-which is normally restricted in its expression to neural tissues-is expressed strikingly in the IMT cells with 2p23 rearrangements. We now report a recurrent oncogenic mechanism, in IMTs, in which tropomyosin (TPM) N-terminal coiled-coil domains are fused to the ALK C-terminal kinase domain. We have cloned two ALK fusion genes, TPM4-ALK and TPM3-ALK, which encode approximately 95-kd fusion oncoproteins characterized by constitutive kinase activity and tyrosylphosphorylation. Immunohistochemical and molecular correlations, in other IMTs, implicate non-TPM ALK oncoproteins that are predominantly cytoplasmic or pre- dominantly nuclear, presumably depending on the subcellular localization of the ALK fusion partner. Notably, a TPM3-ALK oncogene was reported recently in anaplastic lymphoma, and TPM3-ALK is thereby the first known fusion oncogene that transforms, in vivo, both mesenchymal and lymphoid human cell lineages.
Collapse
Affiliation(s)
- Brandon Lawrence
- Brigham and Women’s Hospital, Boston, Massachusetts; the Department of Pathology,†
| | - Antonio Perez-Atayde
- Children’s Hospital Boston, Massachusetts; and the Department of Pediatric Oncology,§
| | - Michele K. Hibbard
- Brigham and Women’s Hospital, Boston, Massachusetts; the Department of Pathology,†
| | - Brian P. Rubin
- Brigham and Women’s Hospital, Boston, Massachusetts; the Department of Pathology,†
| | - Paola Dal Cin
- Brigham and Women’s Hospital, Boston, Massachusetts; the Department of Pathology,†
| | - Jack L. Pinkus
- Brigham and Women’s Hospital, Boston, Massachusetts; the Department of Pathology,†
| | - Geraldine S. Pinkus
- Brigham and Women’s Hospital, Boston, Massachusetts; the Department of Pathology,†
| | - Sheng Xiao
- Brigham and Women’s Hospital, Boston, Massachusetts; the Department of Pathology,†
| | - Eunhee S. Yi
- University of California, San Diego Medical Center, San Diego, California
| | | | - Jonathan A. Fletcher
- Brigham and Women’s Hospital, Boston, Massachusetts; the Department of Pathology,†
| |
Collapse
|
29
|
Seddighzadeh M, Linder S, Shoshan MC, Auer G, Alaiya AA. Inhibition of extracellular signal-regulated kinase 1/2 activity of the breast cancer cell line MDA-MB-231 leads to major alterations in the pattern of protein expression. Electrophoresis 2000; 21:2737-43. [PMID: 10949153 DOI: 10.1002/1522-2683(20000701)21:13<2737::aid-elps2737>3.0.co;2-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Biochemical and genetic strategies have implied that aberrant signaling in the extracellular signal-regulated kinase (ERK)/mitogen-activated protein (MAP) kinase pathway contributes significantly to transformed phenotypes. Using PD98059, an inhibitor of the ERK-kinase MEK1, we have here assessed the effects of ERK inhibition on the pattern of protein expression in the metastatic human breast cancer cell line MDA-MB-231. At a concentration of inhibitor which did not significantly affect cell growth, PD98059 induced large changes in the expression of MDA-MB-231 polypeptides. The majority of these changes were due to decreased expression of low-abundance proteins. Decreases of more abundant proteins such as glutathione-S-transferase pi, hsp80 and hsp100 were also recorded. The levels of a few proteins increased, among them cytokeratin 8. We conclude that PD98059 treatment of MDA-MB-231 cells induces large changes in protein expression.
Collapse
Affiliation(s)
- M Seddighzadeh
- Radiumhemmet's Research Laboratory, Department of Oncology and Pathology, Karolinska Institute and Hospital, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
30
|
Abe J, Okuda M, Huang Q, Yoshizumi M, Berk BC. Reactive oxygen species activate p90 ribosomal S6 kinase via Fyn and Ras. J Biol Chem 2000; 275:1739-48. [PMID: 10636870 DOI: 10.1074/jbc.275.3.1739] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Reactive oxygen species and growth factors stimulate similar intracellular signal transduction events including activation of Src kinase family members and extracellular signal-regulated kinases (ERK1/2). A potentially important downstream effector of Src and ERK1/2 is p90 ribosomal S6 kinase (p90RSK), which plays an important role in cell growth by activating several transcription factors as well as the Na(+)/H(+) exchanger. In the present study, we determined whether H(2)O(2) activates p90RSK to gain insight into signal transduction mechanisms activated by reactive oxygen species. H(2)O(2) (200 microM) stimulated ERK1/2 and p90RSK activity in lymphocytes, endothelial cells, and fibroblasts. The MEK-1 inhibitor, PD98059 (30 microM), inhibited H(2)O(2)-mediated activation of ERK1/2 but not of p90RSK. An essential role for Fyn and Ras in p90RSK activation was suggested by five findings. 1) The tyrosine kinase inhibitor, herbimycin A, and the specific Src kinase family inhibitor, PP1, blocked p90RSK activation by H(2)O(2) in a concentration-dependent manner. 2) p90RSK activation by H(2)O(2) was significantly reduced in fibroblasts derived from transgenic mice deficient in Fyn, but not c-Src. 3) H(2)O(2) rapidly activated Ras (peak at 2-5 min), which preceded p90RSK activation (peak at 20 min). 4) Dominant negative Ras completely blocked H(2)O(2)-induced activation of p90RSK. 5) In Fyn-/- fibroblasts, activation of Ras by H(2)O(2) was significantly attenuated. These results show essential roles for Fyn and Ras in H(2)O(2)-mediated activation of p90RSK and establish redox-sensitive regulation of Ras and p90RSK as a new function for Fyn.
Collapse
Affiliation(s)
- J Abe
- Center for Cardiovascular Research, University of Rochester, Rochester, New York 14642, USA
| | | | | | | | | |
Collapse
|