201
|
Taylor TR, Whalen MM. Ziram activates mitogen-activated protein kinases and decreases cytolytic protein levels in human natural killer cells. Toxicol Mech Methods 2011; 21:577-84. [PMID: 21859362 DOI: 10.3109/15376516.2011.578170] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human natural killer (NK) cells are central in immune defense with their ability to lyse tumor cells and virally infected cells. Tumor formation and viral infection may increase if NK cytotoxic function is disrupted. Ziram (zinc dithiocarbamate) is used as an accelerating agent in the production of latex and to protect various fruits and vegetables from fungal infection. Previously, we have shown that exposure to ziram inhibits NK lytic function. Butyltin environmental contaminants, which also inhibit NK lytic function, cause rapid activations of mitogen-activated protein kinases (MAPKs) and decreases in expression of the cytolytic proteins granzyme B and perforin (after 24 h) in exposed NK cells. MAPKs are important regulators of the lytic response of NK cells, and spurious activation of these enzymes by contaminants would leave the NK cells unable to respond to appropriate targets. This study examined the effects of ziram exposures on MAPKs (p44/42, p38, and c-jun-N-terminal kinase) and on levels of cytolytic proteins. Ten-minute to 6-h exposures of NK cells to ziram caused activation of MAPKs, p44/42, and p38. Exposure to ziram for 24 h caused a decrease in granzyme B and perforin levels. MAPK inhibitors were able to prevent these ziram-induced decreases in granzyme B and perforin. These results suggest that ziram-induced MAPK activation is at least in part responsible for decreased cytolytic function in ziram-exposed NK cells. Furthermore, the results indicate that these changes are in common with other environmental contaminants that have been shown to decrease NK lytic function.
Collapse
Affiliation(s)
- Thyneice R Taylor
- Department of Biological Sciences, Tennessee State University , Nashville, TN 37209 , USA
| | | |
Collapse
|
202
|
Tang CW, Feng WM, Du HM, Bao Y, Zhu M. Delayed administration of D-Ala2-D-Leu5-enkephalin, a delta-opioid receptor agonist, improves survival in a rat model of sepsis. TOHOKU J EXP MED 2011; 224:69-76. [PMID: 21551984 DOI: 10.1620/tjem.224.69] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Sepsis is the major cause of death in intensive care units, despite enormous efforts in the development of antimicrobial therapies. Sepsis is mediated by early [e.g., tumor necrosis factor (TNF)-α and interleukin (IL)-1β] and late [e.g., high-mobility group box 1 protein (HMGB1)] proinflammatory cytokines. HMGB1, which is secreted into extracellular milieu by activated macrophages or passively released by destroyed macrophages, stimulates intensive inflammatory responses. D-Ala2-D-Leu5-enkephalin (DADLE), a synthetic δ-opioid receptor agonist, has been shown to protect rats from sepsis. Here we elucidated the mechanism for protective effect of DADLE against sepsis. Sepsis was established in Sprague-Dawley rats by means of cecal ligation and puncture (CLP). In this model, the serum levels of TNF-α and IL-1β were increased after 2-3 h, while those of HMGB1 were increased after 18 h. Administration of DADLE (5 mg/kg) concurrently with CLP improved survival, which was associated with the decreases in the serum levels of TNF-α, IL-1β and HMGB1. Importantly, DADLE administrated 4 h after CLP showed comparable protective effect as the concurrent administration, with decreased serum HMGB1 levels. Moreover, peritoneal macrophages isolated from rats were challenged with lipopolysaccharide (LPS). Concurrent or delayed DADLE administration at 10(-6) M suppressed the LPS-induced cell death. DADLE also suppressed the release of HMGB1 from macrophages that was induced by LPS, TNF-α or interferon-γ. In conclusion, DADLE protects rats from sepsis probably by decreasing the serum level of HMGB1. We propose DADLE as a candidate for septic shock therapy, even if it is administered after the onset of sepsis.
Collapse
Affiliation(s)
- Cheng Wu Tang
- Department of Hepatobiliary Surgery and Molecular Surgery, First People's Hospital affiliated to Huzhou University Medical College, PR China
| | | | | | | | | |
Collapse
|
203
|
Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases. Microbiol Mol Biol Rev 2011; 75:50-83. [PMID: 21372320 DOI: 10.1128/mmbr.00031-10] [Citation(s) in RCA: 2186] [Impact Index Per Article: 168.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The mitogen-activated protein kinases (MAPKs) regulate diverse cellular programs by relaying extracellular signals to intracellular responses. In mammals, there are more than a dozen MAPK enzymes that coordinately regulate cell proliferation, differentiation, motility, and survival. The best known are the conventional MAPKs, which include the extracellular signal-regulated kinases 1 and 2 (ERK1/2), c-Jun amino-terminal kinases 1 to 3 (JNK1 to -3), p38 (α, β, γ, and δ), and ERK5 families. There are additional, atypical MAPK enzymes, including ERK3/4, ERK7/8, and Nemo-like kinase (NLK), which have distinct regulation and functions. Together, the MAPKs regulate a large number of substrates, including members of a family of protein Ser/Thr kinases termed MAPK-activated protein kinases (MAPKAPKs). The MAPKAPKs are related enzymes that respond to extracellular stimulation through direct MAPK-dependent activation loop phosphorylation and kinase activation. There are five MAPKAPK subfamilies: the p90 ribosomal S6 kinase (RSK), the mitogen- and stress-activated kinase (MSK), the MAPK-interacting kinase (MNK), the MAPK-activated protein kinase 2/3 (MK2/3), and MK5 (also known as p38-regulated/activated protein kinase [PRAK]). These enzymes have diverse biological functions, including regulation of nucleosome and gene expression, mRNA stability and translation, and cell proliferation and survival. Here we review the mechanisms of MAPKAPK activation by the different MAPKs and discuss their physiological roles based on established substrates and recent discoveries.
Collapse
|
204
|
Runchel C, Matsuzawa A, Ichijo H. Mitogen-activated protein kinases in mammalian oxidative stress responses. Antioxid Redox Signal 2011; 15:205-18. [PMID: 21050144 DOI: 10.1089/ars.2010.3733] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
All aerobic organisms are exposed to oxidative stress during their lifetime and are required to respond appropriately for maintenance of their survival and homeostasis. Sustained exposure to oxidative stress has devastating effects in organisms, and, not surprisingly, oxidative stress has been implicated in numerous human diseases. Therefore, an understanding of how mammals respond to oxidative stress is crucial both biologically and clinically. Intracellular signaling pathways, which are activated in response to excessive oxygen radicals, play essential roles in overcoming oxidative stress. The mitogen-activated protein kinase (MAPK) signaling pathways are involved in diverse physiological processes, and are critical for induction of oxidative stress responses. In this review, we will discuss the physiological roles of MAPKs in oxidative stress, the upstream signaling pathways leading to MAPK activation, their regulation, and the MAPK downstream substrates, with a focus on mammalian systems.
Collapse
Affiliation(s)
- Christopher Runchel
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| | | | | |
Collapse
|
205
|
Adachi S, Yasuda I, Nakashima M, Yamauchi T, Kawaguchi J, Shimizu M, Itani M, Nakamura M, Nishii Y, Yoshioka T, Hirose Y, Okano Y, Moriwaki H, Kozawa O. Ultraviolet irradiation can induce evasion of colon cancer cells from stimulation of epidermal growth factor. J Biol Chem 2011; 286:26178-87. [PMID: 21646361 DOI: 10.1074/jbc.m111.240630] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Receptor down-regulation is the most prominent regulatory system of EGF receptor (EGFR) signal attenuation and a critical target for therapy against colon cancer, which is highly dependent on the function of the EGFR. In this study, we investigated the effect of ultraviolet-C (UV-C) on down-regulation of EGFR in human colon cancer cells (SW480, HT29, and DLD-1). UV-C caused inhibition of cell survival and proliferation, concurrently inducing the decrease in cell surface EGFR and subsequently its degradation. UV-C, as well as EGFR kinase inhibitors, decreased the expression level of cyclin D1 and the phosphorylated level of retinoblastoma, indicating that EGFR down-regulation is correlated to cell cycle arrest. Although UV-C caused a marked phosphorylation of EGFR at Ser-1046/1047, UV-C also induced activation of p38 MAPK, a stress-inducible kinase believed to negatively regulate tumorigenesis, and the inhibition of p38 MAPK canceled EGFR phosphorylation at Ser-1046/1047, as well as subsequent internalization and degradation, suggesting that p38 MAPK mediates EGFR down-regulation by UV-C. In addition, phosphorylation of p38 MAPK induced by UV-C was mediated through transforming growth factor-β-activated kinase-1. Moreover, pretreatment of the cells with UV-C suppressed EGF-induced phosphorylation of EGFR at tyrosine residues in addition to cell survival signal, Akt. Together, these results suggest that UV-C irradiation induces the removal of EGFRs from the cell surface that can protect colon cancer cells from oncogenic stimulation of EGF, resulting in cell cycle arrest. Hence, UV-C might be applied for clinical strategy against human colon cancers.
Collapse
Affiliation(s)
- Seiji Adachi
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
206
|
Neveu WA, Bernardo E, Allard JL, Nagaleekar V, Wargo MJ, Davis RJ, Iwakura Y, Whittaker LA, Rincon M. Fungal allergen β-glucans trigger p38 mitogen-activated protein kinase-mediated IL-6 translation in lung epithelial cells. Am J Respir Cell Mol Biol 2011; 45:1133-41. [PMID: 21642586 DOI: 10.1165/rcmb.2011-0054oc] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In addition to immune cells, airway epithelial cells can contribute to and shape the immune response in the lung by secreting specific cytokines. IL-6 is a key factor in determining the effector fate of CD4(+) T cells. Here we show that under basal conditions, the IL-6 gene is already highly expressed in lung epithelial cells, but not in immune cells resident in the lung. However, upon exposure of the lungs to fungal allergens, the direct contact of β-glucans present in the fungus cell wall with lung epithelial cells is sufficient to trigger the rapid synthesis and secretion of IL-6 protein. This posttranscriptional regulation of IL-6 in response to fungal extracts is mediated by the p38 mitogen-activated protein kinase pathway. The inhalation of β-glucans with a nonallergenic antigen is sufficient to provide an adjuvant effect that leads to mucous hyperplasia in the airways. Thus, β-glucans may constitute a common determinant of the fungal and plant-derived allergens responsible for some of the pathological features in allergic asthma.
Collapse
Affiliation(s)
- Wendy A Neveu
- Division of Immunobiology, Department of Medicine, Vermont Lung Center, University of Vermont College of Medicine, Burlington, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
207
|
Novel screening cascade identifies MKK4 as key kinase regulating Tau phosphorylation at Ser422. Mol Cell Biochem 2011; 357:199-207. [PMID: 21638028 DOI: 10.1007/s11010-011-0890-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 05/17/2011] [Indexed: 10/18/2022]
Abstract
Phosphorylation of Tau at serine 422 promotes Tau aggregation. The kinase that is responsible for this key phosphorylation event has so far not been identified but could be a potential drug target for Alzheimer's disease. We describe here an assay strategy to identify this kinase. Using a combination of screening a library of 65'000 kinase inhibitors and in vitro inhibitor target profiling of the screening hits using the Ambit kinase platform, MKK4 was identified as playing a key role in Tau-S422 phosphorylation in human neuroblastoma cells.
Collapse
|
208
|
The bottleneck of JNK signaling: Molecular and functional characteristics of MKK4 and MKK7. Eur J Cell Biol 2011; 90:536-44. [DOI: 10.1016/j.ejcb.2010.11.008] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 11/23/2010] [Accepted: 11/26/2010] [Indexed: 12/18/2022] Open
|
209
|
Davis SJ, Choong DYH, Ramakrishna M, Ryland GL, Campbell IG, Gorringe KL. Analysis of the mitogen-activated protein kinase kinase 4 (MAP2K4) tumor suppressor gene in ovarian cancer. BMC Cancer 2011; 11:173. [PMID: 21575258 PMCID: PMC3115913 DOI: 10.1186/1471-2407-11-173] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 05/17/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND MAP2K4 is a putative tumor and metastasis suppressor gene frequently found to be deleted in various cancer types. We aimed to conduct a comprehensive analysis of this gene to assess its involvement in ovarian cancer. METHODS We screened for mutations in MAP2K4 using High Resolution Melt analysis of 149 primary ovarian tumors and methylation at the promoter using Methylation-Specific Single-Stranded Conformation Polymorphism analysis of 39 tumors. We also considered the clinical impact of changes in MAP2K4 using publicly available expression and copy number array data. Finally, we used siRNA to measure the effect of reducing MAP2K4 expression in cell lines. RESULTS In addition to 4 previously detected homozygous deletions, we identified a homozygous 16 bp truncating deletion and a heterozygous 4 bp deletion, each in one ovarian tumor. No promoter methylation was detected. The frequency of MAP2K4 homozygous inactivation was 5.6% overall, and 9.8% in high-grade serous cases. Hemizygous deletion of MAP2K4 was observed in 38% of samples. There were significant correlations of copy number and expression in three microarray data sets. There was a significant correlation between MAP2K4 expression and overall survival in one expression array data set, but this was not confirmed in an independent set. Treatment of JAM and HOSE6.3 cell lines with MAP2K4 siRNA showed some reduction in proliferation. CONCLUSIONS MAP2K4 is targeted by genetic inactivation in ovarian cancer and restricted to high grade serous and endometrioid carcinomas in our cohort.
Collapse
Affiliation(s)
- Sally J Davis
- VBCRC Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
210
|
Koivisto E, Kaikkonen L, Tokola H, Pikkarainen S, Aro J, Pennanen H, Karvonen T, Rysä J, Kerkelä R, Ruskoaho H. Distinct regulation of B-type natriuretic peptide transcription by p38 MAPK isoforms. Mol Cell Endocrinol 2011; 338:18-27. [PMID: 21354263 DOI: 10.1016/j.mce.2011.02.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 01/27/2011] [Accepted: 02/17/2011] [Indexed: 11/17/2022]
Abstract
Persistent controversy underlies the functional roles of specific p38 MAPK isoforms in cardiac biology and regulation of hypertrophy-associated genes. Here we show that adenoviral gene transfer of p38β but not p38α increased B-type natriuretic peptide (BNP) mRNA levels in vitro as well as atrial natriuretic peptide mRNA levels both in vitro and in vivo. Overexpression of p38α, in turn, augmented the expression fibrosis-related genes connective tissue growth factor, basic fibroblast growth factor and matrix metalloproteinase-9 both in vitro and in vivo. p38β-induced BNP transcription was diminished by mutation of GATA-4 binding site, whereas overexpression of MKK6b, an upstream regulator of p38α and p38β, activated BNP transcription through both GATA-4 and AP-1. Overexpression of MKK3, upstream regulator of p38α, induced BNP transcription independently from AP-1 and GATA-4. These data provide new evidence for diversity in downstream targets and functional roles of p38 pathway kinases in regulation of hypertrophy-associated cardiac genes.
Collapse
Affiliation(s)
- Elina Koivisto
- Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
211
|
Ray RM, Jin S, Bavaria MN, Johnson LR. Regulation of JNK activity in the apoptotic response of intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2011; 300:G761-70. [PMID: 21350193 PMCID: PMC3094148 DOI: 10.1152/ajpgi.00405.2010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We have studied apoptosis of gastrointestinal epithelial cells by examining the receptor-mediated and DNA damage-induced pathways using TNF-α and camptothecin (CPT), respectively. TNF-α requires inhibition of antiapoptotic protein synthesis by cycloheximide (CHX). CHX also results in high levels of active JNK, which are necessary for TNF-induced apoptosis. While CPT induces apoptosis, the increase in JNK activity was not proportional to the degree of apoptosis. Thus the mechanism of activation of JNK and its role in apoptosis are unclear. We examined the course of JNK activation in response to a combination of TNF-α and CPT (TNF + CPT), which resulted in a three- to fourfold increase in apoptosis compared with CPT alone, indicating an amplification of apoptotic signaling pathways. TNF + CPT caused apoptosis by activating JNK, p38, and caspases-8, -9, and -3. TNF-α stimulated a transient phosphorylation of JNK1/2 and ERK1/2 at 15 min, which returned to basal by 60 min and remained low for 4 h. CPT increased JNK1/2 activity between 3 and 4 h. TNF + CPT caused a sustained and robust JNK1/2 and ERK1/2 phosphorylation by 2 h, which remained high at 4 h, suggesting involvement of MEKK4/7 and MEK1, respectively. When administered with TNF + CPT, SP-600125, a specific inhibitor of MEKK4/7, completely inhibited JNK1/2 and decreased apoptosis. However, administration of SP-600125 at 1 h after TNF + CPT failed to prevent JNK1/2 phosphorylation, and the protective effect of SP-600125 on apoptosis was abolished. These results indicate that the persistent activation of JNK might be due to inhibition of JNK-specific MAPK phosphatase 1 (MKP1). Small interfering RNA-mediated knockdown of MKP1 enhanced TNF + CPT-induced activity of JNK1/2 and caspases-9 and -3. Taken together, these results suggest that MKP1 activity determines the duration of JNK1/2 and p38 activation and, thereby, apoptosis in response to TNF + CPT.
Collapse
Affiliation(s)
- Ramesh M. Ray
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Shi Jin
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Mitulkumar N. Bavaria
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Leonard R. Johnson
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
212
|
Lee DE, Lee KW, Byun S, Jung SK, Song N, Lim SH, Heo YS, Kim JE, Kang NJ, Kim BY, Bowden GT, Bode AM, Lee HJ, Dong Z. 7,3',4'-Trihydroxyisoflavone, a metabolite of the soy isoflavone daidzein, suppresses ultraviolet B-induced skin cancer by targeting Cot and MKK4. J Biol Chem 2011; 286:14246-56. [PMID: 21378167 PMCID: PMC3077626 DOI: 10.1074/jbc.m110.147348] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Revised: 02/28/2011] [Indexed: 01/05/2023] Open
Abstract
Nonmelanoma skin cancer is one of the most frequently occurring cancers in the United States. Chronic exposure to UVB irradiation is a major cause of this cancer. Daidzein, along with genistein, is a major isoflavone found in soybeans; however, little is known about the chemopreventive effects of daidzein and its metabolites in UVB-induced skin cancer. Here, we found that 7,3',4'-trihydroxyisoflavone (THIF), a major metabolite of daidzein, effectively inhibits UVB-induced cyclooxygenase 2 (COX-2) expression through the inhibition of NF-κB transcription activity in mouse skin epidermal JB6 P+ cells. In contrast, daidzein had no effect on COX-2 expression levels. Data from Western blot and kinase assays showed that 7,3',4'-THIF inhibited Cot and MKK4 activity, thereby suppressing UVB-induced phosphorylation of mitogen-activated protein kinases. Pull-down assays indicated that 7,3',4'-THIF competed with ATP to inhibit Cot or MKK4 activity. Topical application of 7,3',4'-THIF clearly suppressed the incidence and multiplicity of UVB-induced tumors in hairless mouse skin. Hairless mouse skin results also showed that 7,3',4'-THIF inhibits Cot or MKK4 kinase activity directly, resulting in suppressed UVB-induced COX-2 expression. A docking study revealed that 7,3',4'-THIF, but not daidzein, easily docked to the ATP binding site of Cot and MKK4, which is located between the N- and C-lobes of the kinase domain. Collectively, these results provide insight into the biological actions of 7,3',4'-THIF, a potential skin cancer chemopreventive agent.
Collapse
Affiliation(s)
- Dong Eun Lee
- From the World Class University, Biomodulation Major, Department of Agricultural Biotechnology
- Food Science and Biotechnology Program, Seoul National University, Seoul 151-921, Republic of Korea
- The Hormel Institute, University of Minnesota, Minnesota 55912
| | - Ki Won Lee
- Food Science and Biotechnology Program, Seoul National University, Seoul 151-921, Republic of Korea
| | - Sanguine Byun
- From the World Class University, Biomodulation Major, Department of Agricultural Biotechnology
- Food Science and Biotechnology Program, Seoul National University, Seoul 151-921, Republic of Korea
- The Hormel Institute, University of Minnesota, Minnesota 55912
| | - Sung Keun Jung
- From the World Class University, Biomodulation Major, Department of Agricultural Biotechnology
- Food Science and Biotechnology Program, Seoul National University, Seoul 151-921, Republic of Korea
- The Hormel Institute, University of Minnesota, Minnesota 55912
| | - Nury Song
- From the World Class University, Biomodulation Major, Department of Agricultural Biotechnology
- Food Science and Biotechnology Program, Seoul National University, Seoul 151-921, Republic of Korea
- The Hormel Institute, University of Minnesota, Minnesota 55912
| | - Sung Hwan Lim
- From the World Class University, Biomodulation Major, Department of Agricultural Biotechnology
| | - Yong-Seok Heo
- the Department of Chemistry, Konkuk University, Seoul 143-701, Republic of Korea
| | - Jong Eun Kim
- From the World Class University, Biomodulation Major, Department of Agricultural Biotechnology
- Food Science and Biotechnology Program, Seoul National University, Seoul 151-921, Republic of Korea
- The Hormel Institute, University of Minnesota, Minnesota 55912
| | - Nam Joo Kang
- the School of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Bo Yeon Kim
- the Korea Research Institute of Bioscience and Biotechnology, Choongbuk 363-883, Republic of Korea, and
| | - G. Tim Bowden
- the University of Arizona Cancer Center, Tucson, Arizona 85724
| | - Ann M. Bode
- Food Science and Biotechnology Program, Seoul National University, Seoul 151-921, Republic of Korea
| | - Hyong Joo Lee
- From the World Class University, Biomodulation Major, Department of Agricultural Biotechnology
| | - Zigang Dong
- Food Science and Biotechnology Program, Seoul National University, Seoul 151-921, Republic of Korea
| |
Collapse
|
213
|
Abstract
The Ras/Raf/mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway is often implicated in sensitivity and resistance to leukemia therapy. Dysregulated signaling through the Ras/Raf/MEK/ERK pathway is often the result of genetic alterations in critical components in this pathway as well as mutations at upstream growth factor receptors. Unrestricted leukemia proliferation and decreased sensitivity to apoptotic-inducing agents and chemoresistance are typically associated with activation of pro-survival pathways. Mutations in this pathway and upstream signaling molecules can alter sensitivity to small molecule inhibitors targeting components of this cascade as well as to inhibitors targeting other key pathways (for example, phosphatidylinositol 3 kinase (PI3K)/phosphatase and tensin homologue deleted on chromosome 10 (PTEN)/Akt/mammalian target of rapamycin (mTOR)) activated in leukemia. Similarly, PI3K mutations can result in resistance to inhibitors targeting the Ras/Raf/MEK/ERK pathway, indicating important interaction points between the pathways (cross-talk). Furthermore, the Ras/Raf/MEK/ERK pathway can be activated by chemotherapeutic drugs commonly used in leukemia therapy. This review discusses the mechanisms by which abnormal expression of the Ras/Raf/MEK/ERK pathway can contribute to drug resistance as well as resistance to targeted leukemia therapy. Controlling the expression of this pathway could improve leukemia therapy and ameliorate human health.
Collapse
|
214
|
Crittenden PL, Filipov NM. Manganese modulation of MAPK pathways: effects on upstream mitogen activated protein kinase kinases and mitogen activated kinase phosphatase-1 in microglial cells. J Appl Toxicol 2011; 31:1-10. [PMID: 20589745 DOI: 10.1002/jat.1552] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Multiple studies demonstrate that manganese (Mn) exposure potentiates inflammatory mediator output from activated glia; this increased output is associated with enhanced mitogen activated protein kinase (MAPK: p38, ERK and JNK) activity. We hypothesized that Mn activates MAPK by activating the kinases upstream of MAPK, i.e. MKK-3/6, MKK-1/2 and MKK-4 (responsible for activation of p38, ERK, and JNK, respectively), and/or by inhibiting a major phosphatase responsible for MAPK inactivation, MKP-1. Exposure of N9 microglia to Mn (250 µm), LPS (100 ng ml⁻¹) or Mn + LPS increased MKK-3/6 and MKK-4 activity at 1 h; the effect of Mn + LPS on MKK-4 activation was greater than the rest. At 4 h, Mn, LPS, and Mn + LPS increased MKK-3/6 and MKK-1/2 phosphorylation, whereas MKK-4 was activated only by Mn and Mn + LPS. Besides activating MKK-4 via Ser257/Thr261 phosphorylation, Mn (4 h) prevented MKK-4's phosphorylation on Ser80, which negatively regulates MKK-4 activity. Exposure to Mn or Mn + LPS (1 h) decreased both mRNA and protein expression of MKP-1, the negative MAPK regulator. In addition, we observed that at 4 h, but not at 1 h, a time point coinciding with increased MAPK activity, Mn + LPS markedly increased TNF-α, IL-6 and Cox-2 mRNA, suggesting a delayed effect. The fact that all three major groups of MKKs, MKK-1/2, MKK-3/6 and MKK-4, are activated by Mn suggests that Mn-induced activation of MAPK occurs via traditional mechanisms, which perhaps involve the MAPKs furthest upstream, MKKKs (MAP3Ks). In addition, for all MKKs, Mn-induced activation was persistent at least for 4 h, indicating a long-term effect.
Collapse
Affiliation(s)
- Patrick L Crittenden
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, MS 39762, USA
| | | |
Collapse
|
215
|
Role of mitogen-activated protein kinases in peptidoglycan-induced expression of inducible nitric oxide synthase and nitric oxide in mouse peritoneal macrophages: extracellular signal-related kinase, a negative regulator. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 18:994-1001. [PMID: 21450974 DOI: 10.1128/cvi.00541-10] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The expression of inducible nitric oxide synthase (iNOS) and the production of nitric oxide (NO) are important host defense mechanisms against pathogens in mononuclear phagocytes. The objectives of this study were to examine the roles of mitogen-activated protein kinases (MAPKs) and transcription factors (nuclear factor-κB [NF-κB] and activating protein 1 [AP-1]) in peptidoglycan (PGN)-induced iNOS expression and NO production in macrophages. PGN is a cell wall component of Gram-positive bacteria that stimulates inflammatory responses both ex vivo and in vivo. PGN stimulates the activation of all three classes of MAPKs, extracellular signal-related kinase (ERK), c-Jun N-terminal kinase (JNK), and p38(mapk) in macrophages, albeit with differential activation kinetics. Using a selective inhibitor of JNK (SP600125) and JNK1/2 small interfering RNA (siRNA) knocked-down macrophages, it was observed that PGN-induced iNOS and NO expression is significantly inhibited. This suggested that JNK MAPK plays an essential role in PGN-induced iNOS expression and NO production. In contrast, inhibition of the ERK pathway using PD98059 dose dependently enhanced PGN-induced iNOS expression and NO production. PGN-induced ERK activation was attenuated in ERK1/2 siRNA knocked-down macrophages; however, NO and iNOS expression were significantly enhanced. An electrophoretic mobility shift assay showed that SP600125 inhibited PGN-induced NF-κB and AP-1 activation, whereas inhibition of the ERK pathway enhanced NF-κB activation, but with no effect on AP-1. These results indicate that the JNK MAPK positively regulate PGN-induced iNOS and NO expression by activating NF-κB and AP-1 transcription factors, whereas the ERK pathway plays a negative regulatory role via affecting NF-κB activity.
Collapse
|
216
|
Haeusgen W, Herdegen T, Waetzig V. MKK7γ1 reverses nerve growth factor signals: proliferation and cell death instead of neuritogenesis and protection. Cell Signal 2011; 23:1281-90. [PMID: 21406225 DOI: 10.1016/j.cellsig.2011.03.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Accepted: 03/03/2011] [Indexed: 12/31/2022]
Abstract
c-Jun N-terminal kinases (JNKs) are the exclusive downstream substrates of mitogen-activated protein kinase kinase 7 (MKK7). Recently, we have shown that a single MKK7 splice variant, MKK7γ1, substantially changes the functions of JNKs in naïve PC12 cells. Here we provide evidence that MKK7γ1 blocks NGF-mediated differentiation and sustains proliferation by interfering with the NGF-triggered differentiation programme at several levels: (i) down-regulation of the NGF receptors TrkA and p75; (ii) attenuation of the differentiation-promoting pathways ERK1/2 and AKT; (iii) increase of JNK1 and JNK2, especially the JNK2 54kDa splice variants; (iv) repression of the cyclin-dependent kinase inhibitor p21(WAF1/CIP1), which normally supports NGF-mediated cell cycle arrest; (v) strong induction of the cell cycle promoter CyclinD1, and (vi) profound changes of p53 functions. Moreover, MKK7γ1 substantially changes the responsiveness to stress. Whereas NGF differentiation protects PC12 cells against taxol-induced apoptosis, MKK7γ1 triggers an escape from cell cycle arrest and renders transfected cells sensitive to taxol-induced death. This stress response completely differs from naïve PC12 cells, where MKK7γ1 protects against taxol-induced cell death. These novel aspects on the regulation of JNK signalling emphasise the importance of MKK7γ1 in its ability to reverse basic cellular programmes by simply using JNKs as effectors. Furthermore, our results highlight the necessity for the cells to balance the expression of JNK activators to ensure precise intracellular processes.
Collapse
Affiliation(s)
- Wiebke Haeusgen
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Hospitalstrasse 4, 24105 Kiel, Germany
| | | | | |
Collapse
|
217
|
Cargnello M, Roux PP. Activation and Function of the MAPKs and Their Substrates, the MAPK-Activated Protein Kinases. Microbiol Mol Biol Rev 2011. [DOI: 78495111110.1128/mmbr.00031-10' target='_blank'>'"<>78495111110.1128/mmbr.00031-10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [78495111110.1128/mmbr.00031-10','', '10.1126/science.7839144')">Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
78495111110.1128/mmbr.00031-10" />
Abstract
SUMMARYThe mitogen-activated protein kinases (MAPKs) regulate diverse cellular programs by relaying extracellular signals to intracellular responses. In mammals, there are more than a dozen MAPK enzymes that coordinately regulate cell proliferation, differentiation, motility, and survival. The best known are the conventional MAPKs, which include the extracellular signal-regulated kinases 1 and 2 (ERK1/2), c-Jun amino-terminal kinases 1 to 3 (JNK1 to -3), p38 (α, β, γ, and δ), and ERK5 families. There are additional, atypical MAPK enzymes, including ERK3/4, ERK7/8, and Nemo-like kinase (NLK), which have distinct regulation and functions. Together, the MAPKs regulate a large number of substrates, including members of a family of protein Ser/Thr kinases termed MAPK-activated protein kinases (MAPKAPKs). The MAPKAPKs are related enzymes that respond to extracellular stimulation through direct MAPK-dependent activation loop phosphorylation and kinase activation. There are five MAPKAPK subfamilies: the p90 ribosomal S6 kinase (RSK), the mitogen- and stress-activated kinase (MSK), the MAPK-interacting kinase (MNK), the MAPK-activated protein kinase 2/3 (MK2/3), and MK5 (also known as p38-regulated/activated protein kinase [PRAK]). These enzymes have diverse biological functions, including regulation of nucleosome and gene expression, mRNA stability and translation, and cell proliferation and survival. Here we review the mechanisms of MAPKAPK activation by the different MAPKs and discuss their physiological roles based on established substrates and recent discoveries.
Collapse
Affiliation(s)
- Marie Cargnello
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, Quebec, Canada
- Molecular Biology Program, Université de Montréal, Montreal, Quebec, Canada
| | - Philippe P. Roux
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, Quebec, Canada
- Molecular Biology Program, Université de Montréal, Montreal, Quebec, Canada
- Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
218
|
Klinger M, Sacks S, Cervero F. A role for extracellular signal-regulated kinases 1 and 2 in the maintenance of persistent mechanical hyperalgesia in ovariectomized mice. Neuroscience 2011; 172:483-93. [DOI: 10.1016/j.neuroscience.2010.10.043] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 10/14/2010] [Accepted: 10/15/2010] [Indexed: 12/13/2022]
|
219
|
Ramgolam VS, DeGregorio SD, Rao GK, Collinge M, Subaran SS, Markovic-Plese S, Pardi R, Bender JR. T cell LFA-1 engagement induces HuR-dependent cytokine mRNA stabilization through a Vav-1, Rac1/2, p38MAPK and MKK3 signaling cascade. PLoS One 2010; 5:e14450. [PMID: 21206905 PMCID: PMC3012057 DOI: 10.1371/journal.pone.0014450] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Accepted: 12/06/2010] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Engagement of the β2 integrin, lymphocyte function-associated antigen-1 (LFA-1), results in stabilization of T cell mRNA transcripts containing AU-rich elements (AREs) by inducing rapid nuclear-to-cytosolic translocation of the RNA-stabilizing protein, HuR. However, little is known regarding integrin-induced signaling cascades that affect mRNA catabolism. This study examines the role of the GTPases, Rac 1 and Rac 2, and their downstream effectors, in the LFA-1-induced effects on mRNA. METHODOLOGY/PRINCIPAL FINDINGS Engagement of LFA-1 to its ligand, ICAM-1, in human peripheral T cells resulted in rapid activation of Rac1 and Rac2. siRNA-mediated knockdown of either Rac1 or Rac2 prevented LFA-1-stimulated stabilization of the labile transcripts encoding IFN-γ and TNF-α, and integrin mediated IFN-γ mRNA stabilization was absent in T cells obtained from Rac2 gene-deleted mice. LFA-1 engagement-induced translocation of HuR and stabilization of TNF- α mRNA was lost in Jurkat cells deficient in the Rac guanine nucleotide exchange factor Vav-1 (J.Vav1). The transfection of J.Vav1 cells with constitutively active Rac1 or Rac2 stabilized a labile β-globin reporter mRNA, in a HuR-dependent manner. Furthermore, LFA-1-mediated mRNA stabilization and HuR translocation in mouse splenic T cells was dependent on the phosphorylation of the mitogen-activated protein kinase kinase, MKK3, and its target MAP kinase p38MAPK, and lost in T cells obtained from MKK3 gene-deleted mice. CONCLUSIONS/SIGNIFICANCE Collectively, these results demonstrate that LFA-1-induced stabilization of ARE-containing mRNAs in T cells is dependent on HuR, and occurs through the Vav-1, Rac1/2, MKK3 and p38MAPK signaling cascade. This pathway constitutes a molecular switch that enhances immune and pro-inflammatory gene expression in T cells undergoing adhesion at sites of activation and effector function.
Collapse
Affiliation(s)
- Vinod S. Ramgolam
- Raymond and Beverly Sackler Foundation Cardiovascular Laboratory, Departments of Medicine (Cardiovascular Medicine) and Immunobiology, Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Scott D. DeGregorio
- Raymond and Beverly Sackler Foundation Cardiovascular Laboratory, Departments of Medicine (Cardiovascular Medicine) and Immunobiology, Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Gautham K. Rao
- Raymond and Beverly Sackler Foundation Cardiovascular Laboratory, Departments of Medicine (Cardiovascular Medicine) and Immunobiology, Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Mark Collinge
- Raymond and Beverly Sackler Foundation Cardiovascular Laboratory, Departments of Medicine (Cardiovascular Medicine) and Immunobiology, Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Sharmila S. Subaran
- Raymond and Beverly Sackler Foundation Cardiovascular Laboratory, Departments of Medicine (Cardiovascular Medicine) and Immunobiology, Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Silva Markovic-Plese
- Department of Neurology and of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Ruggero Pardi
- Department of Molecular Pathology, Universitá Vita-Salute School of Medicine, San Raffaele Scientific Institute, Milan, Italy
| | - Jeffrey R. Bender
- Raymond and Beverly Sackler Foundation Cardiovascular Laboratory, Departments of Medicine (Cardiovascular Medicine) and Immunobiology, Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
220
|
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: 426] [Impact Index Per Article: 30.4] [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
|
221
|
Ewen K, Jackson A, Wilhelm D, Koopman P. A Male-Specific Role for p38 Mitogen-Activated Protein Kinase in Germ Cell Sex Differentiation in Mice1. Biol Reprod 2010; 83:1005-14. [DOI: 10.1095/biolreprod.110.086801] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
|
222
|
Lin HH, Lai SC, Chau LY. Heme oxygenase-1/carbon monoxide induces vascular endothelial growth factor expression via p38 kinase-dependent activation of Sp1. J Biol Chem 2010; 286:3829-38. [PMID: 21115498 DOI: 10.1074/jbc.m110.168831] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is a stress-inducible enzyme catalyzing the oxidative degradation of heme to free iron, CO, and biliverdin. Previous studies demonstrated that HO-1 overexpression promoted VEGF expression and angiogenesis in the ischemic heart. However, the underlying mechanism remained elusive. Here we show that adenovirus-mediated HO-1 transduction of rat primary cardiomyocytes and H9C2 myocytes resulted in significant induction of VEGF expression, and a similar effect was seen in cells directly exposed to CO gas or a CO-releasing compound, tricarbonyldichlororuthenium (II) dimer. HO-1/CO-induced VEGF expression was significantly suppressed by pharmacological inhibition of p38 kinase, but not of AKT, activation. VEGF promoter-luciferase reporter assays, electrophoretic mobility shift assays, supershift assay, and chromatin immunoprecipitation showed that CO-induced VEGF promoter activation requires the binding of the Sp1 transcriptional factor to a cis-regulatory sequence located at the VEGF promoter. Western blot analysis and immunostaining experiments demonstrated that HO-1/CO induced p38-dependent phosphorylation of Sp1 at Thr-453 and Thr-739 both in vitro and in vivo. Overexpression of Sp1 protein with an alanine mutation at Thr-453 or Thr-739 suppressed CO-induced Sp1 binding to the VEGF promoter and its transcriptional activation. Collectively, these data suggest that p38-dependent phosphorylation of Sp1 at Thr-453/Thr-739 is crucial for HO-1/CO-induced VEGF expression in myocytes.
Collapse
Affiliation(s)
- Heng-Huei Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | | | | |
Collapse
|
223
|
Oxidative stress induces reactivation of Kaposi's sarcoma-associated herpesvirus and death of primary effusion lymphoma cells. J Virol 2010; 85:715-24. [PMID: 21068240 DOI: 10.1128/jvi.01742-10] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Kaposi's sarcoma (KS) and primary effusion lymphoma (PEL) cells are predominantly infected with latent Kaposi's sarcoma-associated herpesvirus (KSHV), presenting a barrier to the destruction of tumor cells. Latent KSHV can be reactivated to undergo lytic replication. Here we report that in PEL cells, oxidative stress induced by upregulated reactive oxygen species (ROS) can lead to KSHV reactivation or cell death. ROS are upregulated by NF-κB inhibition and are required for subsequent KSHV reactivation. Disruption of the intracellular redox balance through depletion of the antioxidant glutathione or inhibition of the antioxidant enzyme catalase also induces KSHV reactivation, suggesting that hydrogen peroxide induces reactivation. In addition, p38 signaling is required for KSHV reactivation induced by ROS. Furthermore, treatment of PEL cells with a higher concentration of the NF-κB inhibitor than that used for inducing KSHV reactivation further upregulates ROS and induces massive cell death. ROS, but not p38 signaling, are required for PEL cell death induced by NF-κB inhibition as well as by glutathione depletion. Importantly, anticancer drugs, such as cisplatin and arsenic trioxide, also induce KSHV reactivation and PEL cell death in a ROS-dependent manner. Our study thus establishes a critical role for ROS and oxidative stress in the regulation of KSHV reactivation and PEL cell death. Disrupting the cellular redox balance may be a potential strategy for treating KSHV-associated lymphoma.
Collapse
|
224
|
Wattenberg EV. Modulation of protein kinase signaling cascades by palytoxin. Toxicon 2010; 57:440-8. [PMID: 21070801 DOI: 10.1016/j.toxicon.2010.11.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [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
|
225
|
Marber MS, Rose B, Wang Y. The p38 mitogen-activated protein kinase pathway--a potential target for intervention in infarction, hypertrophy, and heart failure. J Mol Cell Cardiol 2010; 51:485-90. [PMID: 21062627 DOI: 10.1016/j.yjmcc.2010.10.021] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2010] [Revised: 10/19/2010] [Accepted: 10/19/2010] [Indexed: 12/20/2022]
Abstract
The p38 mitogen-activated protein kinases (p38s) are stress-activated Ser/Thr kinases. Their activation has been associated with various pathological stressors in the heart. Activated p38 is implicated in a wide spectrum of cardiac pathologies, including hypertrophy, myocardial infarction, as well as systolic and diastolic heart failure. In this review, the specific contribution of different isoforms of p38 kinases to cardiac diseases as well as TAB-1-mediated non-canonical activation pathway are discussed as a rationale for inhibiting p38 activity to treat cardiac hypertrophy, ischemic injury, and heart failure. Finally, a summary of current clinical trials targeting p38 kinases in cardiovascular diseases is provided to highlight the potential promise as well as existing challenges of this therapeutic approach. This article is part of a special issue entitled "Key Signaling Molecules in Hypertrophy and Heart Failure."
Collapse
Affiliation(s)
- Michael S Marber
- King's College London BHF Centre, Cardiovascular Division, The Rayne Institute, St. Thomas' Hospital, London SE1 7EH, United Kingdom.
| | | | | |
Collapse
|
226
|
Abraha AB, Rana K, Whalen MM. Role of protein kinase C in TBT-induced inhibition of lytic function and MAPK activation in human natural killer cells. ARCHIVES OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2010; 59:661-669. [PMID: 20390410 PMCID: PMC2909453 DOI: 10.1007/s00244-010-9520-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Accepted: 03/29/2010] [Indexed: 05/29/2023]
Abstract
Human natural killer (NK) cells are lymphocytes that destroy tumor and virally infected cells. Previous studies have shown that exposure of NK cells to tributyltin (TBT) greatly diminishes their ability to destroy tumor cells (lytic function) while activating mitogen-activated protein kinases (MAPK) (p44/42, p38, and JNK) in NK cells. The signaling pathway that regulates NK lytic function appears to include activation of protein kinase C(PKC) as well as MAPK activity. TBT-induced activation of MAPKs would trigger a portion of the NK lytic signaling pathway, which would then leave the NK cell unable to trigger this pathway in response to a subsequent encounter with a target cell. In the present study we evaluated the involvement of PKC in inhibition of NK lysis of tumor cells and activation of MAPKs caused by TBT exposure. TBT caused a 2–3-fold activation of PKC at concentrations ranging from 50 to 300 nM (16–98 ng/ml),indicating that activation of PKC occurs in response to TBT exposure. This would then leave the NK cell unable to respond to targets. Treatment with the PKC inhibitor, bisindolylmaleimide I, caused an 85% decrease in the ability of NK cells to lyse tumor cells, validating the involvement of PKC in the lytic signaling pathway. The role of PKC in the activation of MAPKs by TBT was also investigated using bisindolylmaleimide I. The results indicated that, in NK cells where PKC activation was blocked, there was no activation of the MAPK, p44/42 in response to TBT.However, TBT-induced activation of the MAPKs, p38 and JNK did not require PKC activation. These results indicate the pivotal role of PKC in the TBT-induced loss of NK lytic function including activation of p44/42 by TBT in NK cells.
Collapse
Affiliation(s)
- Abraham B. Abraha
- Department of Biological Sciences, Tennessee State University, Nashville, TN 37209, USA
| | - Krupa Rana
- Department of Biological Sciences, Tennessee State University, Nashville, TN 37209, USA
| | - Margaret M. Whalen
- Department of Chemistry, Tennessee State University, Nashville, TN 37209, USA
| |
Collapse
|
227
|
Verotoxin-1 treatment or manipulation of its receptor globotriaosylceramide (gb3) for reversal of multidrug resistance to cancer chemotherapy. Toxins (Basel) 2010; 2:2467-77. [PMID: 22069561 PMCID: PMC3153170 DOI: 10.3390/toxins2102467] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2010] [Revised: 10/15/2010] [Accepted: 10/19/2010] [Indexed: 01/08/2023] Open
Abstract
A major problem with anti-cancer drug treatment is the development of acquired multidrug resistance (MDR) of the tumor cells. Verotoxin-1 (VT-1) exerts its cytotoxicity by targeting the globotriaosylceramide membrane receptor (Gb3), a glycolipid associated with multidrug resistance. Gb3 is overexpressed in many human tumors and tumor cell lines with inherent or acquired MDR. Gb3 is co-expressed and interplays with the membrane efflux transporter P-gp encoded by the MDR1 gene. P-gp could act as a lipid flippase and stimulate Gb3 induction when tumor cells are exposed to cancer chemotherapy. Recent work has shown that apoptosis and inherent or acquired multidrug resistance in Gb3-expressing tumors could be affected by VT-1 holotoxin, a sub-toxic concentration of the holotoxin concomitant with chemotherapy or its Gb3-binding B-subunit coupled to cytotoxic or immunomodulatory drug, as well as chemical manipulation of Gb3 expression. The interplay between Gb3 and P-gp thus gives a possible physiological approach to augment the chemotherapeutic effect in multidrug resistant tumors.
Collapse
|
228
|
Polter AM, Li X. 5-HT1A receptor-regulated signal transduction pathways in brain. Cell Signal 2010; 22:1406-12. [PMID: 20363322 PMCID: PMC2903656 DOI: 10.1016/j.cellsig.2010.03.019] [Citation(s) in RCA: 162] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Accepted: 03/25/2010] [Indexed: 10/19/2022]
Abstract
Serotonin is an influential monoamine neurotransmitter that signals through a number of receptors to modulate brain function. Among different serotonin receptors, the serotonin 1A (5-HT1A) receptors have been tied to a variety of physiological and pathological processes, notably in anxiety, mood, and cognition. 5-HT1A receptors couple not only to the classical inhibitory G protein-regulated signaling pathway, but also to signaling pathways traditionally regulated by growth factors. Despite the importance of 5-HT1A receptors in brain function, little is known about how these signaling mechanisms link 5-HT1A receptors to regulation of brain physiology and behavior. Following a brief summary of the known physiological and behavioral effects of 5-HT1A receptors, this article will review the signaling pathways regulated by 5-HT1A receptors, and discuss the potential implication of these signaling pathways in 5-HT1A receptor-regulated physiological processes and behaviors.
Collapse
Affiliation(s)
- Abigail M. Polter
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Xiaohua Li
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
229
|
Odman-Ghazi SO, Abraha A, Isom ET, Whalen MM. Dibutyltin activates MAP kinases in human natural killer cells, in vitro. Cell Biol Toxicol 2010; 26:469-79. [PMID: 20333459 PMCID: PMC2892640 DOI: 10.1007/s10565-010-9157-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 02/25/2010] [Indexed: 11/28/2022]
Abstract
Previous studies have shown that dibutyltin (DBT) interferes with the function of human natural killer (NK) cells, diminishing their capacity to destroy tumor cells, in vitro. DBT is a widespread environmental contaminant and has been found in human blood. As NK cells are our primary immune defense against tumor cells, it is important to understand the mechanism by which DBT interferes with their function. The current study examines the effects of DBT exposures on key enzymes in the signaling pathway that regulates NK responsiveness to tumor cells. These include several protein tyrosine kinases (PTKs), mitogen-activated protein kinases (MAPKs), and mitogen-activated protein kinase kinases (MAP2Ks). The results showed that in vitro exposures of NK cells to DBT had no effect on PTKs. However, exposures to DBT for as little as 10 min were able to increase the phosphorylation (activation) of the MAPKs. The DBT-induced activations of these MAPKs appear to be due to DBT-induced activations of the immediate upstream activators of the MAPKs, MAP2Ks. The results suggest that DBT-interference with the MAPK signaling pathway is a consequence of DBT exposures, which could account for DBT-induced decreases in NK function.
Collapse
Affiliation(s)
- Sabah O. Odman-Ghazi
- Department of Biological Sciences, Tennessee State University, Nashville, TN 37209
| | - Abraham Abraha
- Department of Biological Sciences, Tennessee State University, Nashville, TN 37209
| | - Erica Taylor Isom
- Department of Biological Sciences, Tennessee State University, Nashville, TN 37209
| | - Margaret M. Whalen
- Department of Chemistry, Tennessee State University, Nashville, TN 37209
| |
Collapse
|
230
|
Bhattacharjee A, Mulya A, Pal S, Roy B, Feldman GM, Cathcart MK. Monocyte 15-lipoxygenase gene expression requires ERK1/2 MAPK activity. THE JOURNAL OF IMMUNOLOGY 2010; 185:5211-24. [PMID: 20861348 DOI: 10.4049/jimmunol.1000514] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
IL-13 induces profound expression of 15-lipoxygenase (15-LO) in primary human monocytes. Our studies have defined the functional IL-13R complex, association of Jaks with the receptor components, and the tyrosine phosphorylation of several Stat molecules in response to IL-13. Furthermore, we identified both p38MAPK and protein kinase Cδ as critical regulators of 15-LO expression. In this study, we report an ERK1/2-dependent signaling cascade that regulates IL-13-mediated 15-LO gene expression. We show the rapid phosphorylation/activation of ERK1/2 upon IL-13 exposure. Our results indicate that Tyk2 kinase is required for the activation of ERK1/2, which is independent of the Jak2, p38MAPK, and protein kinase Cδ pathways, suggesting bifurcating parallel regulatory pathways downstream of the receptor. To investigate the signaling mechanisms associated with the ERK1/2-dependent expression of 15-LO, we explored the involvement of transcription factors, with predicted binding sites in the 15-LO promoter, in this process including Elk1, early growth response-1 (Egr-1), and CREB. Our findings indicate that IL-13 induces Egr-1 nuclear accumulation and CREB serine phosphorylation and that both are markedly attenuated by inhibition of ERK1/2 activity. We further show that ERK1/2 activity is required for both Egr-1 and CREB DNA binding to their cognate sequences identified within the 15-LO promoter. Furthermore, by transfecting monocytes with the decoy oligodeoxyribonucleotides specific for Egr-1 and CREB, we discovered that Egr-1 and CREB are directly involved in regulating 15-LO gene expression. These studies characterize an important regulatory role for ERK1/2 in mediating IL-13-induced monocyte 15-LO expression via the transcription factors Egr-1 and CREB.
Collapse
Affiliation(s)
- Ashish Bhattacharjee
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | | | | | | | | | | |
Collapse
|
231
|
Matsumoto T, Kinoshita T, Kirii Y, Yokota K, Hamada K, Tada T. Crystal structures of MKK4 kinase domain reveal that substrate peptide binds to an allosteric site and induces an auto-inhibition state. Biochem Biophys Res Commun 2010; 400:369-73. [PMID: 20732303 DOI: 10.1016/j.bbrc.2010.08.071] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Accepted: 08/17/2010] [Indexed: 11/19/2022]
Abstract
MKK4 activates both JNKs and p38s. We determined the crystal structures of human non-phosphorylated MKK4 kinase domain (npMKK4) complexed with AMP-PNP (npMKK4/AMP) and a ternary complex of npMKK4, AMP-PNP and p38α peptide (npMKK4/AMP/p38). These crystal structures revealed that the p38α peptide-bound npMKK4 at the allosteric site rather than at the putative substrate binding site and induced an auto-inhibition state. While the activation loop of the npMKK4/AMP complex was disordered, in the npMKK4/AMP/p38 complex it configured a long α-helix, which prevented substrate access to the active site and αC-helix movement to the active configuration of MKK4.
Collapse
Affiliation(s)
- Takashi Matsumoto
- PharmAxess, Inc., 3-9-12 Matsubara-cho, Akishima, Tokyo 196-8666, Japan
| | | | | | | | | | | |
Collapse
|
232
|
Nader N, Ng SSM, Lambrou GI, Pervanidou P, Wang Y, Chrousos GP, Kino T. AMPK regulates metabolic actions of glucocorticoids by phosphorylating the glucocorticoid receptor through p38 MAPK. Mol Endocrinol 2010; 24:1748-64. [PMID: 20660302 DOI: 10.1210/me.2010-0192] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Glucocorticoids play central roles in the regulation of energy metabolism by shifting it toward catabolism, whereas AMP-activated protein kinase (AMPK) is the master regulator of energy homeostasis, sensing energy depletion and stimulating pathways of increasing fuel uptake and saving on peripheral supplies. We showed here that AMPK regulates glucocorticoid actions on carbohydrate metabolism by targeting the glucocorticoid receptor (GR) and modifying transcription of glucocorticoid-responsive genes in a tissue- and promoter-specific fashion. Activation of AMPK in rats reversed glucocorticoid-induced hepatic steatosis and suppressed glucocorticoid-mediated stimulation of glucose metabolism. Transcriptomic analysis in the liver suggested marked overlaps between the AMPK and glucocorticoid signaling pathways directed mostly from AMPK to glucocorticoid actions. AMPK accomplishes this by phosphorylating serine 211 of the human GR indirectly through phosphorylation and consequent activation of p38 MAPK and by altering attraction of transcriptional coregulators to DNA-bound GR. In human peripheral mononuclear cells, AMPK mRNA expression positively correlated with that of glucocorticoid-responsive glucocorticoid-inducible leucine zipper protein, which correlated also positively with the body mass index of subjects. These results indicate that the AMPK-mediated energy control system modulates glucocorticoid action at target tissues. Because increased action of glucocorticoids is associated with the development of metabolic disorders, activation of AMPK could be a promising target for developing pharmacological interventions to these pathologies.
Collapse
Affiliation(s)
- Nancy Nader
- Unit on Molecular Hormone Action, Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Building 10, Clinical Research Center, Room 1-3140, 10 Center Drive MSC 1109, Bethesda, MD 20892-1109, USA
| | | | | | | | | | | | | |
Collapse
|
233
|
Moens U, Shiryaev A, Dumitriu G. Comment on "Increased MKK4 abundance with replicative senescence is linked to the joint reduction of multiple microRNAs". Sci Signal 2010; 3:lc1; author reply lc2. [PMID: 20647589 DOI: 10.1126/scisignal.3131lc1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Marasa et al. (Research Article, 27 October 2009, DOI: 10.1126/scisignal.2000442) reported that the human kinase p38-regulated/activated protein kinase (PRAK) was phosphorylated on residue Ser(93) in senescent cells. We have been unable to detect phosphorylation at this site with the antibody that they used, and the commercial supplier of this antibody has discontinued its availability, which casts doubt on whether this residue of PRAK is phosphorylated.
Collapse
|
234
|
Specific regulation of JNK signalling by the novel rat MKK7gamma1 isoform. Cell Signal 2010; 22:1761-72. [PMID: 20633641 DOI: 10.1016/j.cellsig.2010.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Revised: 07/02/2010] [Accepted: 07/05/2010] [Indexed: 01/20/2023]
Abstract
The c-Jun N-terminal kinases (JNKs) mediate a diversity of physiological and pathophysiological effects. Apart from isoform-specific JNK activation, upstream kinases are supposed to be the relevant regulators, which are involved in the context- and signalosome-depending functions. In the present study we report the cloning and characterization of the novel rat MKK7gamma1, a splice variant of MKK7 with an additional exon in the N-terminal region, in the neuronal pheochromocytoma cell line PC12. Transfected MKK7gamma1 increased basal JNK activity, in particular phosphorylation of JNK2. Consequently, JNK signalling was changed in mRNA-, protein- and activation-levels of JNK targets, such as transcription factors (c-Jun, p53, c-Myc), cell cycle regulators (p21, CyclinD1) and apoptotic proteins (Fas, Bim, Bcl-2, Bcl-xl). These alterations promote the sensitivity of MKK7gamma1-transfected cells towards cell death and repress cell proliferation under normal cell growth conditions. Complexes of JIP-1, MKK7 and JNK2 were the major JNK signalosomes under basal conditions. After stimulation with taxol (5muM) and tunicamycin (1.4mug/ml), MKK7gamma1- but not MKK7beta1-transfection, reduced cell death and even increased cell proliferation. Cellular stress also led to an increased phosphorylation of JNK1 and the almost complete abrogation of complexes of JIP-1, MKK7 and JNK2 in MKK7gamma1-transfected PC12 cells. Summarizing, MKK7gamma1 affects the function and activity of individual JNK isoforms and the formation of their signalosomes. This study demonstrates for the first time that one splice-variant of MKK7 tightly controls JNK signalling and effectively adapts JNK functions to the cellular context.
Collapse
|
235
|
Porzionato A, Macchi V, Parenti A, De Caro R. Extracellular signal-regulated kinase and phosphatidylinositol-3-kinase/AKT signalling pathways in the human carotid body and peripheral ganglia. Acta Histochem 2010; 112:305-16. [PMID: 19232686 DOI: 10.1016/j.acthis.2008.09.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2007] [Revised: 09/02/2008] [Accepted: 09/02/2008] [Indexed: 12/20/2022]
Abstract
Extracellular signal-regulated kinase (ERK) and phosphatidylinositol-3-kinase (PI3K)/AKT signalling pathways are involved in various cell functions, but their developmental regulation in the carotid body and peripheral ganglia has not yet been fully investigated. ERK and AKT immunolocalisation and activation were studied by anti-ERK, -pERK, -AKT and -pAKT immunohistochemistry in carotid bodies and peripheral (sympathetic and sensory) ganglia, sampled at autopsy from 4 foetuses (mean gestational age 177 days), 8 infants (mean age 10 months), 8 young adults (mean age 38 years) and 6 aged adults (mean age 72.4 years). ERK and AKT immunopositivity and activation were demonstrated in both glomic type I cells and peripheral ganglionic cells and are ascribed to local action by neuromodulators or neurotrophic factors. Mean percentages of ERK- and pERK-immunopositive glomic type I cells were lower in foetuses than in infants and young adults, and those of AKT-immunopositive glomic type I cells were lower in foetuses than in young and old adults, suggesting incomplete maturation of these two signalling pathways in foetal life. Both pERK and pAKT immunoreactions were detected only in post-natal sympathetic and sensory ganglia, demonstrating that, also in peripheral ganglia, these pathways are not yet fully operative during the foetal stage.
Collapse
|
236
|
Affiliation(s)
- P Lenormand
- Centre de Biochimie-CNRS UMR 134, Université de Nice, Parc Valrose, 06108 Nice Cedex 2, France
| |
Collapse
|
237
|
Langereis JD, Raaijmakers HAJA, Ulfman LH, Koenderman L. Abrogation of NF-κB signaling in human neutrophils induces neutrophil survival through sustained p38-MAPK activation. J Leukoc Biol 2010; 88:655-64. [PMID: 20573801 DOI: 10.1189/jlb.0809544] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
NF-κB, an important transcription factor in the regulation of cellular inflammation, is one of the prime targets for novel anti-inflammatory therapeutics. Nowadays, anti-inflammatory therapies rely mostly on steroids, which among other effects, inhibit NF-κB activity. However, steroids have only limited efficacy in the treatment on neutrophil-driven diseases, such as COPD. Human neutrophils play an important role in the pathogenesis of COPD, and clearance of these cells by apoptosis is an effective pathway for resolution of inflammation. In this study, we tested the hypothesis that modulation of the NF-κB pathway in human neutrophils affects survival. Importantly, the pharmacological NF-κB inhibitor Bay 11-7082 inhibited NF-κB signaling in human neutrophils as expected. However, we found that complete inhibition of NF-κB activity with 10 μM Bay 11-7082 prolonged neutrophil survival significantly, which was not observed with inhibitors for other signaling pathways. Bay 11-7082-induced neutrophil survival was dependent on p38-MAPK kinase activity, as the p38 kinase activity inhibitor SB203580 abrogated this response completely. Bay 11-7082 induced rapid and sustained p38 activation that correlated with inhibited NF-κB signaling and prolonged neutrophil survival. The precise role of NF-κB in regulation of p38-MAPK activation remains to be established. Under these conditions of survival, the stability of Bcl-xL but not Mcl-1 was enhanced. Although inhibition of NF-κB leads to down-regulation of inflammatory genes in many cell types, our results illustrate that interference with basal NF-κB signaling in neutrophils as a drug target should be used with caution.
Collapse
Affiliation(s)
- Jeroen D Langereis
- Department of Respiratory Medicine, University Medical Center, Utrecht, The Netherlands
| | | | | | | |
Collapse
|
238
|
Harding SJ, Browne GJ, Miller BW, Prigent SA, Dickens M. Activation of ASK1, downstream MAPKK and MAPK isoforms during cardiac ischaemia. Biochim Biophys Acta Mol Basis Dis 2010; 1802:733-40. [PMID: 20550965 PMCID: PMC2954285 DOI: 10.1016/j.bbadis.2010.06.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Revised: 06/08/2010] [Accepted: 06/09/2010] [Indexed: 11/28/2022]
Abstract
p38 MAPK is activated potently during cardiac ischaemia, although the precise mechanism by which it is activated is unclear. We used the isolated perfused rat heart to investigate the signalling pathways activated upstream of p38 during global cardiac ischaemia. Ischaemia strongly activated p38α but not the JNK pathway. The MAPKKs, MKK3, MKK4 and MKK6 have previously been identified as potential upstream activators of p38; however, in the ischaemic perfused heart, we saw activation of MKK3 and MKK6 but not MKK4. MKK3 and MKK6 showed different temporal patterns of activity, indicating distinct modes of activation and physiological function. Consistent with a lack of JNK activation, we saw no activation of MKK4 or MKK7 at any time point during ischaemia. A lack of MKK4 activation indicates, at least in the ischaemic heart, that MKK4 is not a physiologically relevant activator of p38. The MAPKKK, ASK1, was strongly activated late during ischaemia, with a similar time course to that of MKK6 and in ischaemic neonatal cardiac myocytes ASK1 expression preferentially activated MKK6 rather than MKK3. These observations suggest that during ischaemia ASK1 is coupled to p38 activation primarily via MKK6. Potent activation of ASK1 during ischaemia without JNK activation shows that during cardiac ischaemia, ASK1 preferentially activates the p38 pathway. These results demonstrate a specificity of responses seldom seen in previous studies and illustrate the benefits of using direct assays in intact tissues responding to physiologically relevant stimuli to unravel the complexities of MAPK signalling.
Collapse
Affiliation(s)
- Stephen J Harding
- Department of Biochemistry, Henry Wellcome Building, University of Leicester, Leicester, UK
| | | | | | | | | |
Collapse
|
239
|
Gurtner A, Starace G, Norelli G, Piaggio G, Sacchi A, Bossi G. Mutant p53-induced up-regulation of mitogen-activated protein kinase kinase 3 contributes to gain of function. J Biol Chem 2010; 285:14160-9. [PMID: 20223820 PMCID: PMC2863202 DOI: 10.1074/jbc.m109.094813] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Revised: 03/09/2010] [Indexed: 01/02/2023] Open
Abstract
Mitogen-activated protein kinase kinase 3 (MAP2K3) is a member of the dual specificity kinase group. Growing evidence links MAP2K3 to invasion and tumor progression. Here, we identify MAP2K3 as a transcriptional target of endogenous gain-of-function p53 mutants R273H, R175H, and R280K. We show that MAP2K3 modulation occurred at the mRNA and protein levels and that endogenous mutant p53 proteins are capable of binding to and activate the MAP2K3 promoter. In addition, we found that the studied p53 mutants regulate MAP2K3 gene expression through the involvement of the transcriptional cofactors NF-Y and NF-kappaB. Finally, functional studies showed that endogenous MAP2K3 knockdown inhibits proliferation and survival of human tumor cells, whereas the ectopic expression of MAP2K3 can rescue the proliferative defect induced by mutant p53 knockdown. Taken together, our findings define a novel player through which mutant p53 exerts its gain-of-function activity in cancer cells.
Collapse
Affiliation(s)
- Aymone Gurtner
- From the Molecular Oncogenesis Laboratory, Department of Experimental Oncology, Regina Elena Cancer Institute, Via delle Messi D'Oro 156, 00158 Rome, Italy and
| | - Giuseppe Starace
- the National Council of Research, Istituto di Neurobiologia e Medicina Molecolare, 00133 Rome, Italy
| | - Giuseppe Norelli
- From the Molecular Oncogenesis Laboratory, Department of Experimental Oncology, Regina Elena Cancer Institute, Via delle Messi D'Oro 156, 00158 Rome, Italy and
| | - Giulia Piaggio
- From the Molecular Oncogenesis Laboratory, Department of Experimental Oncology, Regina Elena Cancer Institute, Via delle Messi D'Oro 156, 00158 Rome, Italy and
| | - Ada Sacchi
- From the Molecular Oncogenesis Laboratory, Department of Experimental Oncology, Regina Elena Cancer Institute, Via delle Messi D'Oro 156, 00158 Rome, Italy and
| | - Gianluca Bossi
- From the Molecular Oncogenesis Laboratory, Department of Experimental Oncology, Regina Elena Cancer Institute, Via delle Messi D'Oro 156, 00158 Rome, Italy and
| |
Collapse
|
240
|
Schonhoff CM, Webster CRL, Anwer MS. Cyclic AMP stimulates Mrp2 translocation by activating p38{alpha} MAPK in hepatic cells. Am J Physiol Gastrointest Liver Physiol 2010; 298:G667-74. [PMID: 20203059 PMCID: PMC2867428 DOI: 10.1152/ajpgi.00506.2009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Cyclic AMP (cAMP) induces translocation of multidrug resistant protein 2 (Mrp2) to the canalicular membrane and activates p38 MAPK in rat hepatocytes. In this study, we tested the hypothesis that cAMP-induced Mrp2 translocation may be mediated via p38 MAPK. Studies were conducted in rat hepatocytes and in a human hepatoma cell line, HuH-7. In rat hepatocytes, cAMP increased Mrp2 translocation and p38 MAPK activity. These effects of cAMP were inhibited by SB203580, an inhibitor of p38 MAPK. Wortmannin, a specific inhibitor of phosphoinositide-3-kinase (PI3K), did not inhibit cAMP induced activation of p38 MAPK, indicating PI3K-independent activation of p38 MAPK by cAMP. To further define the role of p38 MAPK, molecular approaches were used to up- or downregulate p38 MAPK activity in HuH-7 cells using constitutively active (CA) and dominant-negative (DN) MAPK kinase 3 and 6 (MKK3/6). MKK3/6 are upstream kinases responsible for the activation of p38 MAPK. Cells transfected with CAMKK6 showed increased p38 MAPK activity and MRP2 translocation compared with empty vector. cAMP-induced activation of p38 MAPK was inhibited in cells transfected with DNMKK3/6 and DNMKK3, but not with DNMKK6. DNMKK3/6 and DNMKK3 also inhibited cAMP-induced MRP2 translocation. cAMP selectively activated p38alpha MAPK in HuH-7 cells. Knockdown of p38alpha MAPK by short heterodimer RNA resulted in decreased level of p38 MAPK and failure of cAMP to stimulate MRP2 translocation. Taken together, these results suggest that cAMP-induced MRP2 translocation in hepatic cells is mediated via PI3K-independent and MKK3-mediated activation of p38alpha MAPK.
Collapse
Affiliation(s)
| | - Cynthia R. L. Webster
- 2Clinical Sciences, Cummings School of Veterinary Medicine at Tufts University, North Grafton, Massachusetts
| | | |
Collapse
|
241
|
Thyroid cancer: current molecular perspectives. JOURNAL OF ONCOLOGY 2010; 2010:351679. [PMID: 20369062 PMCID: PMC2847382 DOI: 10.1155/2010/351679] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2009] [Accepted: 01/21/2010] [Indexed: 11/28/2022]
Abstract
The thyroid cancer is a rare oncological entity, representing no more than 1% of all human malignant neoplasms. Recently, it has been demonstrated a sharp increase in incidence of differentiated thyroid carcinoma, equally occurring in both sexes. So far, multiple genetic alterations have been identified in differentiated thyroid carcinoma, leading to investigate the clinical utility of genetic studies. In particular, molecular genetic approaches searching for gene mutations in the material collected by fine needle ago-biopsy may have a particular utility in small nodules and in those specimens with an indeterminate cytology. The expansion of knowledge about genetic mutations occurring in different thyroid tumors has characterized recent years, allowing the identification of a correlation between specific mutations and phenotypic characteristics of thyroid cancers, essential for their prognosis. This review will briefly report on the histological features and the new entity represented by thyroid microcarcinoma and will focus on both environmental and genetic aspects associated with the occurrence of thyroid cancer.
Collapse
|
242
|
Schwer CI, Mutschler M, Stoll P, Goebel U, Humar M, Hoetzel A, Schmidt R. Carbon monoxide releasing molecule-2 inhibits pancreatic stellate cell proliferation by activating p38 mitogen-activated protein kinase/heme oxygenase-1 signaling. Mol Pharmacol 2010; 77:660-9. [PMID: 20053955 DOI: 10.1124/mol.109.059519] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Proliferation of pancreatic stellate cells (PSCs) plays a cardinal role during fibrosis development. Therefore, the suppression of PSC growth represents a therapeutic option for the treatment of pancreatic fibrosis. It has been shown that up-regulation of the enzyme heme oxygenase-1 (HO-1) could exert antiproliferative effects on PSCs, but no information is available on the possible role of carbon monoxide (CO), a catalytic byproduct of the HO metabolism, in this process. In the present study, we have examined the effect of CO releasing molecule-2 (CORM-2) liberated CO on PSC proliferation and have elucidated the mechanisms involved. Using primary rat PSCs, we found that CORM-2 inhibited PSC proliferation at nontoxic concentrations by arresting cells at the G(0)/G(1) phase of the cell cycle. This effect was associated with activation of p38 mitogen-activated protein kinase (MAPK) signaling, induction of HO-1 protein, and up-regulation of the cell cycle inhibitor p21(Waf1/Cip1). The p38 MAPK inhibitor 4-(4-flurophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)imidazole (SB203580) abolished the inhibitory effect of CORM-2 on PSC proliferation and prevented both CORM-2-induced HO-1 and p21(Waf1/Cip1) up-regulation. Treatment with tin protoporphyrin IX, an HO inhibitor, or transfection of HO-1 small interfering RNA abolished the inductive effect of CORM-2 on p21(Waf1/Cip1) and reversed the suppressive effect of CORM-2 on PSC growth. The ability of CORM-2 to induce cell cycle arrest was abrogated in p21(Waf1/Cip1)-silenced cells. Taken together, our results suggest that CORM-2 inhibits PSC proliferation by activation of the p38/HO-1 pathway. These findings may indicate a therapeutic potential of CO carriers in the treatment of pancreatic fibrosis.
Collapse
Affiliation(s)
- Christian I Schwer
- Department of Anesthesiology, University Medical Center, Hugstetter Strasse 55, D-79106 Freiburg, Germany
| | | | | | | | | | | | | |
Collapse
|
243
|
Clavel S, Siffroi-Fernandez S, Coldefy AS, Boulukos K, Pisani DF, Dérijard B. Regulation of the intracellular localization of Foxo3a by stress-activated protein kinase signaling pathways in skeletal muscle cells. Mol Cell Biol 2010; 30:470-80. [PMID: 19917721 PMCID: PMC2798458 DOI: 10.1128/mcb.00666-09] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Revised: 06/27/2009] [Accepted: 10/21/2009] [Indexed: 12/13/2022] Open
Abstract
Muscle atrophy is a debilitating process associated with many chronic wasting diseases, like cancer, diabetes, sepsis, and renal failure. Rapid loss of muscle mass occurs mainly through the activation of protein breakdown by the ubiquitin proteasome pathway. Foxo3a transcription factor is critical for muscle atrophy, since it activates the expression of ubiquitin ligase Atrogin-1. In several models of atrophy, inhibition of the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway induces nuclear import of Foxo3a through an Akt-dependent process. This study aimed to identify signaling pathways involved in the control of Foxo3a nuclear translocation in muscle cells. We observed that after nuclear import of Foxo3a by PI3K/Akt pathway inhibition, activation of stress-activated protein kinase (SAPK) pathways induced nuclear export of Foxo3a through CRM1. This mechanism involved the c-Jun NH(2)-terminal kinase (JNK) signaling pathway and was independent of Akt. Likewise, we showed that inhibition of p38 induced a massive nuclear relocalization of Foxo3a. Our results thus suggest that SAPKs are involved in the control of Foxo3a nucleocytoplasmic translocation in C2C12 cells. Moreover, activation of SAPKs decreases the expression of Atrogin-1, and stable C2C12 myotubes, in which the p38 pathway is constitutively activated, present partial protection against atrophy.
Collapse
Affiliation(s)
- Stephan Clavel
- LBPSI, Université de Nice-Sophia Antipolis/CNRS, FRE3094, Nice, France, Université de Nice-Sophia Antipolis EA 4319/INSERM ERI-21, Nice, France, Institute of Developmental Biology and Cancer, Université de Nice-Sophia Antipolis, CNRS, UMR6543, Nice, France
| | - Sandrine Siffroi-Fernandez
- LBPSI, Université de Nice-Sophia Antipolis/CNRS, FRE3094, Nice, France, Université de Nice-Sophia Antipolis EA 4319/INSERM ERI-21, Nice, France, Institute of Developmental Biology and Cancer, Université de Nice-Sophia Antipolis, CNRS, UMR6543, Nice, France
| | - Anne Sophie Coldefy
- LBPSI, Université de Nice-Sophia Antipolis/CNRS, FRE3094, Nice, France, Université de Nice-Sophia Antipolis EA 4319/INSERM ERI-21, Nice, France, Institute of Developmental Biology and Cancer, Université de Nice-Sophia Antipolis, CNRS, UMR6543, Nice, France
| | - Kim Boulukos
- LBPSI, Université de Nice-Sophia Antipolis/CNRS, FRE3094, Nice, France, Université de Nice-Sophia Antipolis EA 4319/INSERM ERI-21, Nice, France, Institute of Developmental Biology and Cancer, Université de Nice-Sophia Antipolis, CNRS, UMR6543, Nice, France
| | - Didier F. Pisani
- LBPSI, Université de Nice-Sophia Antipolis/CNRS, FRE3094, Nice, France, Université de Nice-Sophia Antipolis EA 4319/INSERM ERI-21, Nice, France, Institute of Developmental Biology and Cancer, Université de Nice-Sophia Antipolis, CNRS, UMR6543, Nice, France
| | - Benoît Dérijard
- LBPSI, Université de Nice-Sophia Antipolis/CNRS, FRE3094, Nice, France, Université de Nice-Sophia Antipolis EA 4319/INSERM ERI-21, Nice, France, Institute of Developmental Biology and Cancer, Université de Nice-Sophia Antipolis, CNRS, UMR6543, Nice, France
| |
Collapse
|
244
|
Procaccia S, Kraus S, Seger R. Determination of ERK activity: anti-phospho-ERK antibodies and in vitro phosphorylation. Methods Mol Biol 2010; 661:39-58. [PMID: 20811975 DOI: 10.1007/978-1-60761-795-2_2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The ERK signaling cascade is composed of several protein kinases that sequentially activate each other by phosphorylation. This pathway is a central component of a complex signaling network that regulates important cellular processes including proliferation, differentiation, and survival. In most of these cases, the ERK cascade is activated downstream of the small GTPase Ras that, upon activation, recruits and activates the first tier in the cascade, which contains the Raf kinases. Afterward the signal is further transmitted by MEKs, ERKs, and often RSKs in the MAPKK, MAPK, and MAPKAPKs tiers of the cascade, respectively. ERKs and RSKs can further disseminate the signal by phosphorylating and modulating the activity of a large number of regulatory proteins including transcription factors and chromatin modifying enzymes. Understanding the mechanisms of activation and the regulation of the various components of this cascade will enhance our insight into the regulation of the ERK-dependent cellular processes in normal cells or of their malfunctioning in various diseases, including cancer. In this chapter, we describe methods used to determine the activity of ERKs, which upon slight modifications can also be used for the study of other signaling kinases, either within the cascade or in other pathways. These methods have been successfully applied to study the ERK signaling cascades in a variety of tissue-cultured cell lines, homo-genized animal organs, and lower organisms. As such, the use of these methods should expand our knowledge on the regulation of many distinct systems and upon induction of various stimulations.
Collapse
Affiliation(s)
- Shiri Procaccia
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel
| | | | | |
Collapse
|
245
|
Leitao B, Jones MC, Fusi L, Higham J, Lee Y, Takano M, Goto T, Christian M, Lam EWF, Brosens JJ. Silencing of the JNK pathway maintains progesterone receptor activity in decidualizing human endometrial stromal cells exposed to oxidative stress signals. FASEB J 2009; 24:1541-51. [PMID: 20026682 PMCID: PMC2857868 DOI: 10.1096/fj.09-149153] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Survival of the conceptus is dependent on continuous progesterone signaling in the maternal decidua but how this is achieved under conditions of oxidative stress that characterize early pregnancy is unknown. Using primary cultures, we show that modest levels of reactive oxygen species (ROS) increase sumoylation in human endometrial stromal cells (HESCs), leading to enhanced modification and transcriptional inhibition of the progesterone receptor (PR). The ability of ROS to induce a sustained hypersumoylation response, or interfere with PR activity, was lost upon differentiation of HESCs into decidual cells. Hypersumoylation in response to modest levels of ROS requires activation of the JNK pathway. Although ROS-dependent JNK signaling is disabled on decidualization, the cells continue to mount a transcriptional response, albeit distinct from that observed in undifferentiated HESCs. We further show that attenuated JNK signaling in decidual cells is a direct consequence of altered expression of key pathway modulators, including induction of MAP kinase phosphatase 1 (MKP1). Overexpression of MKP1 dampens JNK signaling, prevents hypersumoylation, and maintains PR activity in undifferentiated HESCs exposed to ROS. Thus, JNK silencing uncouples ROS signaling from the SUMO conjugation pathway and maintains progesterone responses and cellular homeostasis in decidual cells under oxidative stress conditions imposed by pregnancy.—Leitao, B., Jones, M. C., Fusi, L., Higham, J., Lee, Y. Takano, M., Goto, T., Christian, M., Lam, E. W.-F., Brosens, J. J. Silencing of the Jnk pathway maintains progesterone receptor activity in decidualizing human endometrial stromal cells exposed to oxidative stress signals.
Collapse
Affiliation(s)
- Beatriz Leitao
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Campus, Du Cane Rd., London W12 0NN
| | | | | | | | | | | | | | | | | | | |
Collapse
|
246
|
Johansson D, Andersson C, Moharer J, Johansson A, Behnam-Motlagh P. Cisplatin-induced expression of Gb3 enables verotoxin-1 treatment of cisplatin resistance in malignant pleural mesothelioma cells. Br J Cancer 2009; 102:383-91. [PMID: 20010943 PMCID: PMC2816648 DOI: 10.1038/sj.bjc.6605467] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Background: A major problem with cisplatin treatment is the development of acquired-drug resistance of the tumour cells. Verotoxin-1 (VT-1) exerts its cytotoxicity by targeting the membrane glycolipid globotriasosylceramide (Gb3), a molecule associated with drug resistance. Cisplatin- and VT-1-induced apoptosis involves mitogen-activated protein kinase (MAPK) activation, and deactivation of MAPKs is associated with cisplatin resistance. This study aimed to investigate whether a sub-toxic concentration of VT-1 could enhance cisplatin-induced apoptosis and overcome acquired-cisplatin resistance in cultured cancer cell lines. Method: P31 and H1299 cells with corresponding cisplatin-resistant sub-lines (P31res/H1299res) were incubated with VT-1 and/or cisplatin followed by determination of Gb3 expression, cell viability, apoptosis, and signalling pathways. Results: Cells from the resistant sub-lines had elevated Gb3 expression compared with the parental cell lines, and cisplatin further increased Gb3 expression, whereas VT-1 reduced the percentage of Gb3-expressing cells. Combination of cisplatin and sub-toxic concentrations of VT-1 led to a super-additive increase of cytotoxicity and TUNEL staining, especially in the cisplatin-resistant sub-lines. Blockade of Gb3 synthesis by a Gb3 synthesis inhibitor not only led to eradicated TUNEL staining of P31 cells, but also sensitised P31res cells to the induction of apoptosis by cisplatin alone. Cisplatin- and VT-1-induced apoptosis involved the MAPK pathways with increased C-Jun N-terminal kinase and MAPK kinase-3 and -6 phosphorylation. Conclusions: We show the presence of Gb3 in acquired-cisplatin resistance in P31res and H1299res cells. Cisplatin up-regulated Gb3 expression in all cells and thus sensitised the cells to VT-1-induced cytotoxicity. A strong super-additive effect of combined cisplatin and a sub-toxic concentration of VT-1 in cisplatin-resistant malignant pleural mesothelioma cells were observed, indicating a new potential clinical-treatment approach.
Collapse
Affiliation(s)
- D Johansson
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå S-901 85, Sweden
| | | | | | | | | |
Collapse
|
247
|
Geest CR, Buitenhuis M, Laarhoven AG, Bierings MB, Bruin MCA, Vellenga E, Coffer PJ. p38 MAP kinase inhibits neutrophil development through phosphorylation of C/EBPalpha on serine 21. Stem Cells 2009; 27:2271-82. [PMID: 19544470 DOI: 10.1002/stem.152] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Many extracellular stimuli regulate growth, survival, and differentiation responses through activation of the dual specificity mitogen activated protein kinase (MAPK) kinase three (MKK3) and its downstream effector p38 MAPK. Using CD34+ hematopoietic progenitor cells, here we describe a novel role for MKK3-p38MAPK in the regulation of myelopoiesis. Inhibition of p38MAPK utilizing the pharmacological inhibitor SB203580, enhanced neutrophil development ex vivo, but conversely reduced eosinophil differentiation. In contrast, constitutive activation of MKK3 dramatically inhibited neutrophil differentiation. Transplantation of beta2-microglobulin(-/-) nonobese diabetic/severe combined immune deficient (NOD/SCID) mice with CD34+ cells ectopically expressing constitutively active MKK3 resulted in reduced neutrophil differentiation in vivo, whereas eosinophil development was enhanced. Inhibitory phosphorylation of CCAAT/enhancer binding protein alpha (C/EBPalpha) on serine 21 was induced upon activation of p38MAPK. Moreover, ectopic expression of a non-phosphorylatable C/EBPalpha mutant was sufficient to abrogate MKK3-induced inhibition of neutrophil development. Furthermore, treatment of CD34+ progenitors from patients with severe congenital neutropenia with SB203580 restored neutrophil development. These results establish a novel role for MKK3-p38MAPK in the regulation of lineage choices during myelopoiesis through modulation of C/EBPalpha activity. This signaling module may thus provide an important therapeutic target in the treatment of bone marrow failure.
Collapse
Affiliation(s)
- Christian R Geest
- Molecular Immunology Lab, Department of Immunology, University Medical Center, Utrecht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
248
|
Burk DR, Senechal-Willis P, Lopez LC, Hogue BG, Daskalova SM. Suppression of lipopolysaccharide-induced inflammatory responses in RAW 264.7 murine macrophages by aqueous extract of Clinopodium vulgare L. (Lamiaceae). JOURNAL OF ETHNOPHARMACOLOGY 2009; 126:397-405. [PMID: 19770031 DOI: 10.1016/j.jep.2009.09.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2009] [Revised: 08/24/2009] [Accepted: 09/14/2009] [Indexed: 05/28/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The wild basil Clinopodium vulgare L. is commonly used in Bulgarian folk medicine for treatment of irritated skin, mastitis- and prostatitis-related swelling, as well as for some disorders accompanied with significant degree of inflammation (e.g. gastric ulcers, diabetes, and cancer). AIM OF STUDY To determine the effect of aqueous extract of Clinopodium vulgare L. on LPS-induced inflammatory responses of murine RAW 264.7 macrophages. MATERIALS AND METHODS Cell cytotoxicity was evaluated by MTT assay. Protein expression levels were monitored by Western blot analysis. Production of NO and PGE(2) was measured by the Griess colorimetric method and enzyme immunoassay, respectively. Activation of MMP-9 was visualized by gelatin zymography. Cytokine levels were determined by BioPlex assay. Intracellular ROS and free radical scavenging potential were measured by DCFH-DA and DPPH method, respectively. Xanthine oxidase activity was evaluated spectrophotometrically. RESULTS The extract suppresses NF-kappaB activation by preventing I kappa-B phosphorylation and inhibits the phosphorylation of p38 and SAPK/JNK MAPKs. It down-regulates iNOS expression which manifests as a drastic decrease of NO production, inhibits MMP-9 activation, but does not affect COX-2 protein levels and reduces only slightly the released PGE(2). Secretion of IL-1 beta and Il-10 is greatly reduced, whereas suppression of TNF-alpha and GM-CSF production is less dramatic. The extract has strong free radical scavenging properties and exerts inhibitory effect on xanthine oxidase activity, which lowers the levels of intracellular ROS. CONCLUSION The study provides evidence for the anti-inflammatory potential of Clinopodium vulgare L. aqueous extract.
Collapse
Affiliation(s)
- David R Burk
- Center for Infectious Diseases and Vaccinology, Arizona State University, 1001 S. McAllister Avenue, Tempe, AZ 85287, USA.
| | | | | | | | | |
Collapse
|
249
|
Patil SS, Schlick F, Höger H, Lubec G. Involvement of individual hippocampal signaling protein levels in spatial memory formation is strain-dependent. Amino Acids 2009; 39:75-87. [PMID: 19890699 DOI: 10.1007/s00726-009-0379-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2009] [Accepted: 10/14/2009] [Indexed: 12/30/2022]
Abstract
Although a series of signaling cascades involved in spatial memory have been identified, their link to spatial memory and strain-dependent expression has not been reported so far. Hippocampal levels of the abovementioned signaling proteins were determined in laboratory inbred strain C57BL/6J, the wild-derived inbred strain PWD/PhJ and the wild caught mouse Apodemus sylvaticus (AS) by immunoblotting. The resulting hippocampal protein levels were correlated with results from MWM. Hippocampal signaling protein (hSP) levels were tested also in yoked controls. Within-strain comparison between trained and yoked controls revealed significant differences between levels of Phospho-CaMKII (alpha), Phospho-CREB, Egr-1, c-Src, Phospho-ERK5, Phospho-MEK5 and NOS1 in all of the three strains tested. In addition, the three strains revealed different involvement of individual hSP levels clearly indicating that individual mouse strains were linked to individual hSPs in spatial memory. Phospho-ERK5 levels were not detectable in hippocampi of yoked controls of each strain. We learn from this study that a series of hSPs are associated with spatial memory and that different hSPs are linked to spatial memory in different strains that show different outcome in the MWM. Even correlational patterns in the individual hSPs differed between mouse strains. This is of importance for the interpretation of previous studies on the abovementioned signaling cascades as well as for the design of future studies on these hippocampal proteins. It is intriguing that individual mouse strains, laboratory or wild caught, may use different signaling pathways for spatial memory in the Morris water maze.
Collapse
Affiliation(s)
- Sudarshan S Patil
- Division of Pediatric Neuroscience, Department of Pediatrics, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | | | | | | |
Collapse
|
250
|
McCubrey JA, Abrams SL, Stadelman K, Chappell WH, Lahair M, Ferland RA, Steelman LS. Targeting signal transduction pathways to eliminate chemotherapeutic drug resistance and cancer stem cells. ADVANCES IN ENZYME REGULATION 2009; 50:285-307. [PMID: 19895837 PMCID: PMC2862855 DOI: 10.1016/j.advenzreg.2009.10.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|