101
|
Deibler KK, Mishra RK, Clutter MR, Antanasijevic A, Bergan R, Caffrey M, Scheidt KA. A Chemical Probe Strategy for Interrogating Inhibitor Selectivity Across the MEK Kinase Family. ACS Chem Biol 2017; 12:1245-1256. [PMID: 28263556 PMCID: PMC5652073 DOI: 10.1021/acschembio.6b01060] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
MEK4 is an upstream kinase in MAPK signaling pathways where it phosphorylates p38 MAPK and JNK in response to mitogenic and cellular stress queues. MEK4 is overexpressed and induces metastasis in advanced prostate cancer lesions. However, the value of MEK4 as an oncology target has not been pharmacologically validated because selective chemical probes targeting MEK4 have not been developed. Despite a high level of sequence homology in the ATP-binding site, most reported MEK inhibitors are selective for MEK1/2 and display reduced potency toward other MEKs. Here, we present the first functional and binding selectivity-profiling platform of the MEK family. We applied the platform to profile a set of known kinase inhibitors and used the results to develop an in silico approach for small molecule docking against MEK proteins. The docking studies identified molecular features of the ligands and corresponding amino acids in MEK proteins responsible for high affinity binding versus those driving selectivity. WaterLOGSY and saturation transfer difference (STD) NMR spectroscopy techniques were utilized to understand the binding modes of active compounds. Further minor synthetic manipulations provide a proof of concept by showing how information gained through this platform can be utilized to perturb selectivity across the MEK family. This inhibitor-based approach pinpoints key features governing MEK family selectivity and clarifies empirical selectivity profiles for a set of kinase inhibitors. Going forward, the platform provides a rationale for facilitating the development of MEK-selective inhibitors, particularly MEK4 selective inhibitors, and repurposing of kinase inhibitors for probing the structural selectivity of isoforms.
Collapse
Affiliation(s)
- Kristine K. Deibler
- Department of Chemistry, Northwestern University, Evanston, 60208, Illinois, United States
| | - Rama K. Mishra
- Center for Molecular Innovation and Drug Discovery, Northwestern University, 2145 Sheridan Rd, Evanston, Illinois 60208, United States
| | - Matthew R. Clutter
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Rd, Evanston, Illinois 60208, United States
| | - Aleksandar Antanasijevic
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, 900 S Ashland Ave, Chicago, Illinois 60607, United States
| | - Raymond Bergan
- Knight Cancer Institute, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Rd., Portland, Oregon 97239, United States
| | - Michael Caffrey
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, 900 S Ashland Ave, Chicago, Illinois 60607, United States
| | - Karl A. Scheidt
- Department of Chemistry, Northwestern University, Evanston, 60208, Illinois, United States
- Center for Molecular Innovation and Drug Discovery, Northwestern University, 2145 Sheridan Rd, Evanston, Illinois 60208, United States
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Rd, Evanston, Illinois 60208, United States
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| |
Collapse
|
102
|
Involvement of the MEK-ERK/p38-CREB/c-fos signaling pathway in Kir channel inhibition-induced rat retinal Müller cell gliosis. Sci Rep 2017; 7:1480. [PMID: 28469203 PMCID: PMC5431154 DOI: 10.1038/s41598-017-01557-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/28/2017] [Indexed: 11/07/2022] Open
Abstract
Our previous studies have demonstrated that activation of group I metabotropic glutamate receptors downregulated Kir channels in chronic ocular hypertension (COH) rats, thus contributing to Müller cell gliosis, characterized by upregulated expression of glial fibrillary acidic protein (GFAP). In the present study, we explored possible signaling pathways linking Kir channel inhibition and GFAP upregulation. In normal retinas, intravitreal injection of BaCl2 significantly increased GFAP expression in Müller cells, which was eliminated by co-injecting mitogen-activated protein kinase (MAPK) inhibitor U0126. The protein levels of phosphorylated extracellular signal-regulated protein kinase1/2 (p-ERK1/2) and its upstream regulator, p-MEK, were significantly increased, while the levels of phosphorylated c-Jun N-terminal kinase (p-JNK) and p38 kinase (p-p38) remained unchanged. Furthermore, the protein levels of phosphorylated cAMP response element binding protein (p-CREB) and c-fos were also increased, which were blocked by co-injecting ERK inhibitor FR180204. In purified cultured rat Müller cells, BaCl2 treatment induced similar changes in these protein levels apart from p-p38 levels and the p-p38:p38 ratio showing significant upregulation. Moreover, intravitreal injection of U0126 eliminated the upregulated GFAP expression in COH retinas. Together, these results suggest that Kir channel inhibition-induced Müller cell gliosis is mediated by the MEK-ERK/p38-CREB/c-fos signaling pathway.
Collapse
|
103
|
Natriuretic peptide receptor guanylyl cyclase-A pathway counteracts glomerular injury evoked by aldosterone through p38 mitogen-activated protein kinase inhibition. Sci Rep 2017; 7:46624. [PMID: 28429785 PMCID: PMC5399490 DOI: 10.1038/srep46624] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 03/22/2017] [Indexed: 12/11/2022] Open
Abstract
Guanylyl cyclase-A (GC-A) signaling, a natriuretic peptide receptor, exerts renoprotective effects by stimulating natriuresis and reducing blood pressure. Previously we demonstrated massive albuminuria with hypertension in uninephrectomized, aldosterone-infused, and high salt-fed (ALDO) systemic GC-A KO mice with enhanced phosphorylation of p38 mitogen-activated protein kinase (MAPK) in podocytes. In the present study, we examined the interaction between p38 MAPK and GC-A signaling. The administration of FR167653, p38 MAPK inhibitor, reduced systolic blood pressure (SBP), urinary albumin excretion, segmental sclerosis, podocyte injury, and apoptosis. To further investigate the local action of natriuretic peptide and p38 MAPK in podocytes, we generated podocyte-specific (pod) GC-A conditional KO (cKO) mice. ALDO pod GC-A cKO mice demonstrated increased urinary albumin excretion with marked mesangial expansion, podocyte injury and apoptosis, but without blood pressure elevation. FR167653 also suppressed urinary albumin excretion without reducing SBP. Finally, we revealed that atrial natriuretic peptide increased phosphorylation of MAPK phosphatase-1 (MKP-1) concomitant with inhibited phosphorylation of p38 MAPK in response to MAPK kinase 3 activation, thereby resulting in decreased mRNA expression of the apoptosis-related gene, Bax, and Bax/Bcl2 ratio in cultured podocytes. These results indicate that natriuretic peptide exerts a renoprotective effect via inhibiting phosphorylation of p38 MAPK in podocytes.
Collapse
|
104
|
Abstract
The ADP-ribosyltransferase C3 exoenzyme from C. botulinum selectively inactivates Rho and is therefore often used as an inhibitor for investigations on Rho signaling. Previous studies of our group revealed that C3 inhibited cell proliferation in HT22 cells accompanied by increased transcriptional activities of Sp1 and c-Jun and reduced levels of cyclin D1, p21 and phosphorylated p38. By use of a p38α-deficient and a p38α-expressing control cell line, the impact of p38 on C3-mediated inhibition of cell proliferation and alterations on MAPK signaling was studied by growth kinetic experiments and Western blot analyses. The cell growth of p38α-expressing cells was impaired by C3, while the p38α-deficient cells did not exhibit any C3-induced effect. The activity of the MKK3/6-p38 MAPK signaling cascade as well as the phosphorylation of c-Jun and JNK was reduced by C3 exclusively in the presence of p38α. Moreover, the activity of upstream MAPKKK TAK1 was lowered in the p38α-expressing cells. These results indicated a resistance of p38α-deficient cells to C3-mediated inhibition of cell growth. This anti-proliferative effect was highly associated with the decreased activity of c-Jun and upstream p38 and JNK MAPK signaling as a consequence of the absence of p38α in these cells.
Collapse
|
105
|
Qian F, Deng J, Wang G, Ye RD, Christman JW. Pivotal Role of Mitogen-Activated Protein Kinase-Activated Protein Kinase 2 in Inflammatory Pulmonary Diseases. Curr Protein Pept Sci 2016; 17:332-42. [PMID: 26119506 DOI: 10.2174/1389203716666150629121324] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 06/25/2015] [Accepted: 06/26/2015] [Indexed: 01/11/2023]
Abstract
Mitogen-activated protein kinase (MAPK)-activated protein kinase (MK2) is exclusively regulated by p38 MAPK in vivo. Upon activation of p38 MAPK, MK2 binds with p38 MAPK, leading to phosphorylation of TTP, Hsp27, Akt, and Cdc25 that are involved in regulation of various essential cellular functions. In this review, we discuss current knowledge about molecular mechanisms of MK2 in regulation of TNF-α production, NADPH oxidase activation, neutrophil migration, and DNA-damage-induced cell cycle arrest which are involved in the molecular pathogenesis of acute lung injury, pulmonary fibrosis, and non-small-cell lung cancer. Collectively current and emerging new information indicate that developing MK2 inhibitors and blocking MK2-mediated signal pathways are potential therapeutic strategies for treatment of inflammatory and fibrotic lung diseases and lung cancer.
Collapse
Affiliation(s)
- Feng Qian
- Department of Internal Medicine, The Ohio State University, 201 Davis Heart and Lung Research Institute, 473 West 12th Avenue, Columbus, OH 43210, USA.
| | | | | | | | - John W Christman
- Department of Internal Medicine, The Ohio State University, 201 Davis Heart and Lung Research Institute, 473 West 12th Avenue, Columbus, OH 43210, USA.
| |
Collapse
|
106
|
Affiliation(s)
- Gianluca Bossi
- Experimental Oncology Laboratories, and Laboratory of Medical Physics and Expert Systems, Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
107
|
Weng Q, Liu Z, Li B, Liu K, Wu W, Liu H. Oxidative Stress Induces Mouse Follicular Granulosa Cells Apoptosis via JNK/FoxO1 Pathway. PLoS One 2016; 11:e0167869. [PMID: 27936150 PMCID: PMC5148000 DOI: 10.1371/journal.pone.0167869] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 11/21/2016] [Indexed: 01/03/2023] Open
Abstract
The c-Jun N-terminal protein kinase (JNK) plays an important role in the regulation of cell apoptosis. Forkhead box O (FoxO) transcription factors are involved in diverse biological processes, including cellular metabolism, cell apoptosis, and cell cycle. However, the JNK/FoxO1 pathway involved in the process of apoptosis induced by oxidative stress remains to be elucidated. Here, we demonstrated that the JNK activity significantly increased in response to oxidative stress in mouse follicular granulosa cells (MGCs). SP600125, a selective JNK inhibitor, attenuated the oxidative stress-induced MGCs apoptosis. Oxidative stress enhanced the FoxO1 nuclear translocation by activating the JNK activity. Moreover, JNK mediated the dissociation of FoxO1 from 14-3-3 proteins in MGCs after the treatment with H2O2. Finally, oxidative stress up-regulated the expression of FoxO1 via JNK mediation of FoxO1 self-regulation in MGCs. Taken together, our findings suggest that JNK/FoxO1 is involved in the regulation of oxidative stress-induced cell apoptosis in MGCs.
Collapse
Affiliation(s)
- Qiannan Weng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zequn Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- Liaoning Province of Animal Product Safety Supervision, Shenyang, China
| | - Bojiang Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Kaiqing Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Wangjun Wu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Honglin Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- * E-mail:
| |
Collapse
|
108
|
Lin B, Xu D, Leaman DW. X-linked inhibitor of apoptosis-associated factor 1 regulates TNF receptor 1 complex stability. FEBS Lett 2016; 590:4381-4392. [PMID: 27768232 DOI: 10.1002/1873-3468.12467] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 08/17/2016] [Accepted: 08/30/2016] [Indexed: 11/06/2022]
Abstract
X-linked inhibitor of apoptosis (XIAP)-associated factor 1 (XAF1) is a cytokine-regulated, tumor necrosis factor (TNF) receptor-associated factor (TRAF) domain-containing protein that has a poorly defined cellular function. Here, we show that ectopically expressed XAF1 inhibits TNF-ɑ-induced NF-κB activation, whereas shRNA silencing of endogenous XAF1 augments it. Our data suggest that XAF1 may inhibit TNF-ɑ-induced NF-κB activation by disrupting the assembly of the TRADD/TRAF2/RIP1 complex (complex I) downstream of TNF receptor activation. XAF1 interacts with TRAF2 and inhibits TRAF2-dependent NF-κB activation, in part, by blocking TRAF2 polyubiquitination. Our findings also indicate that although XAF1 does not directly inhibit RIP1-dependent NF-κB activation, it binds RIP1 and disrupts RIP1/TRADD association. Our data suggest that XAF1 acts as a feedback regulator of the TNF receptor signaling pathway to suppress NF-κB activation.
Collapse
Affiliation(s)
- Boren Lin
- Department of Biological Sciences, The University of Toledo, OH, USA
| | - Da Xu
- Department of Biological Sciences, The University of Toledo, OH, USA
| | - Douglas W Leaman
- Department of Biological Sciences, The University of Toledo, OH, USA
| |
Collapse
|
109
|
Qu F, Xiang Z, Zhang Y, Li J, Xiao S, Zhang Y, Mao F, Ma H, Yu Z. A novel p38 MAPK indentified from Crassostrea hongkongensis and its involvement in host response to immune challenges. Mol Immunol 2016; 79:113-124. [PMID: 27768933 DOI: 10.1016/j.molimm.2016.10.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 10/08/2016] [Accepted: 10/11/2016] [Indexed: 01/06/2023]
Abstract
p38 mitogen-activated protein kinases (MAPKs) are conserved serine/threonine-specific kinases that are activated by various extracellular stimuli and play crucial regulatory roles in immunity, development and homeostasis. However, the function of p38s in mollusks, the second most diverse group of animals, is still poorly understood. In this study, a novel molluscan p38 (designated Chp38) was cloned and characterized from the Hong Kong oyster Crassostrea hongkongensis. Its full-length cDNA encoded a putative protein of 353 amino acids with a calculated molecular weight of approximately 40.3kDa. Similar to other reported p38 family proteins, the deduced Chp38 sequence contained a conserved dual phosphorylation TGY motif and a substrate binding site of ATRW. Phylogenetic analysis revealed that Chp38 was closest to its homolog from the Pacific oyster and belonged to the mollusk cluster. Quantitative real-time PCR analysis showed that Chp38 was constitutively expressed in all examined oyster tissues and developmental stages and that its expression in hemocytes was significantly up-regulated after pathogen (Vibrio alginolyticus and Staphylococcus haemolyticus) and PAMP (lipopolysaccharide and peptidoglycan) infections. Moreover, overexpression analysis revealed that Chp38 was localized in both the cytoplasm and nucleus of HEK293T cells and that it could significantly enhance AP-1 reporter gene activation in a dose-dependent manner. Altogether, these results provide the first experimental evidence of a functional p38 in oysters and suggest its involvement in the innate immunity of C. hongkongensis.
Collapse
Affiliation(s)
- Fufa Qu
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301,China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou 510275, China; Department of Biological and Environmental Engineering, Changsha University, Changsha 410022, China
| | - Zhiming Xiang
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301,China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou 510275, China.
| | - Yang Zhang
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301,China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou 510275, China
| | - Jun Li
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301,China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou 510275, China
| | - Shu Xiao
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301,China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou 510275, China
| | - Yuehuan Zhang
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301,China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou 510275, China
| | - Fan Mao
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301,China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou 510275, China
| | - Haitao Ma
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301,China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou 510275, China
| | - Ziniu Yu
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301,China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou 510275, China.
| |
Collapse
|
110
|
Yu L, Ham K, Gao X, Castro L, Yan Y, Kissling GE, Tucker CJ, Flagler N, Dong R, Archer TK, Dixon D. Epigenetic regulation of transcription factor promoter regions by low-dose genistein through mitogen-activated protein kinase and mitogen-and-stress activated kinase 1 nongenomic signaling. Cell Commun Signal 2016; 14:18. [PMID: 27582276 PMCID: PMC5007815 DOI: 10.1186/s12964-016-0141-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 08/17/2016] [Indexed: 02/06/2023] Open
Abstract
Background The phytoestrogen, genistein at low doses nongenomically activates mitogen-activated protein kinase p44/42 (MAPKp44/42) via estrogen receptor alpha (ERα) leading to proliferation of human uterine leiomyoma cells. In this study, we evaluated if MAPKp44/42 could activate downstream effectors such as mitogen- and stress-activated protein kinase 1 (MSK1), which could then epigenetically modify histone H3 by phosphorylation following a low dose (1 μg/ml) of genistein. Results Using hormone-responsive immortalized human uterine leiomyoma (ht-UtLM) cells, we found that genistein activated MAPKp44/42 and MSK1, and also increased phosphorylation of histone H3 at serine10 (H3S10ph) in ht-UtLM cells. Colocalization of phosphorylated MSK1 and H3S10ph was evident by confocal microscopy in ht-UtLM cells (r = 0.8533). Phosphorylation of both MSK1and H3S10ph was abrogated by PD98059 (PD), a MEK1 kinase inhibitor, thereby supporting genistein’s activation of MSK1 and Histone H3 was downstream of MAPKp44/42. In proliferative (estrogenic) phase human uterine fibroid tissues, phosphorylated MSK1 and H3S10ph showed increased immunoexpression compared to normal myometrial tissues, similar to results observed in in vitro studies following low-dose genistein administration. Real-time RT-PCR arrays showed induction of growth-related transcription factor genes, EGR1, Elk1, ID1, and MYB (cMyb) with confirmation by western blot, downstream of MAPK in response to low-dose genistein in ht-UtLM cells. Additionally, genistein induced associations of promoter regions of the above transcription factors with H3S10ph as evidenced by Chromatin Immunoprecipitation (ChIP) assays, which were inhibited by PD. Therefore, genistein epigenetically modified histone H3 by phosphorylation of serine 10, which was regulated by MSK1 and MAPK activation. Conclusion Histone H3 phosphorylation possibly represents a mechanism whereby increased transcriptional activation occurs following low-dose genistein exposure. Electronic supplementary material The online version of this article (doi:10.1186/s12964-016-0141-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Linda Yu
- Molecular Pathogenesis Group, National Toxicology Program (NTP) Laboratory, Division of the NTP (DNTP), National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), U.S. Department of Health and Human Services (HHS), Research Triangle Park, North Carolina, 27709, USA
| | - Kyle Ham
- Molecular Pathogenesis Group, National Toxicology Program (NTP) Laboratory, Division of the NTP (DNTP), National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), U.S. Department of Health and Human Services (HHS), Research Triangle Park, North Carolina, 27709, USA
| | - Xiaohua Gao
- Molecular Pathogenesis Group, National Toxicology Program (NTP) Laboratory, Division of the NTP (DNTP), National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), U.S. Department of Health and Human Services (HHS), Research Triangle Park, North Carolina, 27709, USA
| | - Lysandra Castro
- Molecular Pathogenesis Group, National Toxicology Program (NTP) Laboratory, Division of the NTP (DNTP), National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), U.S. Department of Health and Human Services (HHS), Research Triangle Park, North Carolina, 27709, USA
| | - Yitang Yan
- Molecular Pathogenesis Group, National Toxicology Program (NTP) Laboratory, Division of the NTP (DNTP), National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), U.S. Department of Health and Human Services (HHS), Research Triangle Park, North Carolina, 27709, USA
| | - Grace E Kissling
- Biostatistics and Computational Biology Branch, Division of Intramural Research (DIR), NIEHS, NIH, HHS, Research Triangle Park, North Carolina, 27709, USA
| | - Charles J Tucker
- Signal Transduction Laboratory, DIR, NIEHS, NIH, HHS, Research Triangle Park, North Carolina, 27709, USA
| | - Norris Flagler
- Cellular and Molecular Pathology Branch, DNTP, NIEHS, NIH, HHS, Research Triangle Park, North Carolina, 27709, USA
| | - Ray Dong
- Molecular Pathogenesis Group, National Toxicology Program (NTP) Laboratory, Division of the NTP (DNTP), National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), U.S. Department of Health and Human Services (HHS), Research Triangle Park, North Carolina, 27709, USA
| | - Trevor K Archer
- Chromatin and Gene Expression Group, Epigenetics and Stem Cell Biology Laboratory, DIR, NIEHS, NIH, HHS, Research Triangle Park, North Carolina, 27709, USA
| | - Darlene Dixon
- Molecular Pathogenesis Group, National Toxicology Program (NTP) Laboratory, Division of the NTP (DNTP), National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), U.S. Department of Health and Human Services (HHS), Research Triangle Park, North Carolina, 27709, USA.
| |
Collapse
|
111
|
Meng H, Gao Y, Kang YF, Zhao YP, Yang GJ, Wang Y, Cao Y, Gan YH, Xie QF. Molecular Changes Involving MEK3-p38 MAPK Activation in Chronic Masticatory Myalgia. J Dent Res 2016; 95:1169-75. [PMID: 27418173 DOI: 10.1177/0022034516659441] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The exact mechanism underlying chronic masticatory myalgia (CMM), a conspicuous symptom in temporomandibular disorders, remains unclear. This investigation compared gene expression profiles between CMM patients and healthy subjects. Peripheral blood leukocytes were collected in 8 cases and 8 controls and subjected to whole genome microarray analyses. Data were analyzed with Gene Ontology and interactive pathways analyses. According to Gene Ontology analysis, categories such as ion transport, response to stimuli, and metabolic process were upregulated. The pathway analysis suggested overexpression of the mitogen-activated protein kinase (MAPK) pathway in CMM patients and to a higher degree in a pathway network. Overexpression of representative members of the MAPK pathway-including MAPK kinase 3 (MEK3), calcium voltage-gated channel auxiliary subunit gamma 2 (CACNG2), and growth arrest and DNA damage-inducible gamma (GADD45G)-was validated with real-time polymerase chain reaction. The upregulation of MEK3 was negatively correlated with the age of the CMM group. In the next step, the authors focused on MEK3, the gene that exhibited the greatest degree of differential expression, and its downstream target protein p38 MAPK. The results revealed upregulation of MEK3, as well as phosphorylated MEK3 and phosphorylated p38 MAPK, in CMM patients. These results provide a "fingerprint" for mechanistic studies of CMM in the future and highlight the importance of MEK3-p38 MAPK activation in CMM.
Collapse
Affiliation(s)
- H Meng
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China Center for Oral Functional Diagnosis, Treatment, and Research, Peking University School and Hospital of Stomatology, Beijing, China Department of Prosthodontics, North China University of Science and Technology School of Stomatology, Hebei, China
| | - Y Gao
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China Center for Oral Functional Diagnosis, Treatment, and Research, Peking University School and Hospital of Stomatology, Beijing, China
| | - Y F Kang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China Center for Oral Functional Diagnosis, Treatment, and Research, Peking University School and Hospital of Stomatology, Beijing, China
| | - Y P Zhao
- Center for TMD and Orofacial Pain, Peking University School and Hospital of Stomatology, Beijing, China
| | - G J Yang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China Center for Oral Functional Diagnosis, Treatment, and Research, Peking University School and Hospital of Stomatology, Beijing, China
| | - Y Wang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China Center for Oral Functional Diagnosis, Treatment, and Research, Peking University School and Hospital of Stomatology, Beijing, China
| | - Y Cao
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China Center for Oral Functional Diagnosis, Treatment, and Research, Peking University School and Hospital of Stomatology, Beijing, China
| | - Y H Gan
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Q F Xie
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China Center for Oral Functional Diagnosis, Treatment, and Research, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
112
|
|
113
|
TLR and TNF-R1 activation of the MKK3/MKK6-p38α axis in macrophages is mediated by TPL-2 kinase. Biochem J 2016; 473:2845-61. [PMID: 27402796 PMCID: PMC5095906 DOI: 10.1042/bcj20160502] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 07/11/2016] [Indexed: 01/08/2023]
Abstract
Previous studies suggested that Toll-like receptor (TLR) stimulation of the p38α MAP kinase (MAPK) is mediated by transforming growth factor-β-activated kinase 1 (TAK1) activation of MAPK kinases, MKK3, MKK4 and MKK6. We used quantitative mass spectrometry to monitor tumour progression locus 2 (TPL-2)-dependent protein phosphorylation following TLR4 stimulation with lipopolysaccharide, comparing macrophages from wild-type mice and Map3k8(D270A/D270A) mice expressing catalytically inactive TPL-2 (MAP3K8). In addition to the established TPL-2 substrates MKK1/2, TPL-2 kinase activity was required to phosphorylate the activation loops of MKK3/6, but not of MKK4. MKK3/6 activation required IκB kinase (IKK) phosphorylation of the TPL-2 binding partner nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB1) p105, similar to MKK1/2 activation. Tumour necrosis factor (TNF) stimulation of MKK3/6 phosphorylation was similarly dependent on TPL-2 catalytic activity and IKK phosphorylation of NF-κB1 p105. Owing to redundancy of MKK3/6 with MKK4, Map3k8(D270A) mutation only fractionally decreased lipopolysaccharide activation of p38α. TNF activation of p38α, which is mediated predominantly via MKK3/6, was substantially reduced. TPL-2 catalytic activity was also required for MKK3/6 and p38α activation following macrophage stimulation with Mycobacterium tuberculosis and Listeria monocytogenes Our experiments demonstrate that the IKK/NF-κB1 p105/TPL-2 signalling pathway, downstream of TAK1, regulates MKK3/6 and p38α activation in macrophages in inflammation.
Collapse
|
114
|
Fung TS, Liao Y, Liu DX. Regulation of Stress Responses and Translational Control by Coronavirus. Viruses 2016; 8:v8070184. [PMID: 27384577 PMCID: PMC4974519 DOI: 10.3390/v8070184] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 06/21/2016] [Accepted: 06/28/2016] [Indexed: 01/02/2023] Open
Abstract
Similar to other viruses, coronavirus infection triggers cellular stress responses in infected host cells. The close association of coronavirus replication with the endoplasmic reticulum (ER) results in the ER stress responses, which impose a challenge to the viruses. Viruses, in turn, have come up with various mechanisms to block or subvert these responses. One of the ER stress responses is inhibition of the global protein synthesis to reduce the amount of unfolded proteins inside the ER lumen. Viruses have evolved the capacity to overcome the protein translation shutoff to ensure viral protein production. Here, we review the strategies exploited by coronavirus to modulate cellular stress response pathways. The involvement of coronavirus-induced stress responses and translational control in viral pathogenesis will also be briefly discussed.
Collapse
Affiliation(s)
- To Sing Fung
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| | - Ying Liao
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Ziyue Road 518, Shanghai 200241, China.
| | - Ding Xiang Liu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| |
Collapse
|
115
|
Whalen MM, Odman-Ghazi SO. Effects of adenylyl cyclase and protein kinase A inhibition on signaling enzymes in natural killer cells: comparison to tributyltin. Hum Exp Toxicol 2016; 25:333-40. [PMID: 16866191 DOI: 10.1191/0960327106ht630oa] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Natural killer (NK) cells are lymphocytes capable of destroying tumor cells and virally-infected cells without prior sensitization. In a previous study, we found that inhibition of adenylyl cyclase (AC) or cAMP-dependent protein kinase (PKA) decreased the ability of NK cells to destroy tumor cells. We also found that the environmental contaminant tributyltin (TBT), at concentrations of 300 500 nM, decreased tumor-cell lysis by NK cells, as well as their intracellular levels of cAMP. This suggested that the decreases in cAMP associated with TBT (300 500 nM) may, in part, be responsible for loss of cytotoxic function. Here, we investigated the effects of inhibition of AC or PKA on enzymes that are required in the NK tumorolytic process and compared them to those of TBT exposure. The enzymes studied were: the protein tyrosine kinase (PTK), syk; phospholipase C gamma1 (PLCg1); and the mitogen activated protein kinase (MAPK), p44/42. Exposure of NK cells to the AC inhibitor 2?,5?-dideoxyadenosine (DDA) significantly increased the total level of PLCg1 by 67% after 60 min and the level of p44/42 by about 30%. Exposure to the PKA inhibitor H-89 significantly increased the levels of the phosphorylated (activated) p44/42 (90%) after 60 min. Exposure to TBT increased the levels of PLCg1 by about 50%. Previously, we found that exposure to TBT increased the phosphorylation of p44/42 within 5 min. These results indicate that AC inhibition caused alterations of the levels of key enzymes, while decreased PKA activity caused an increase in p44/42 activation. They also suggest that the effects of decreased levels of cAMP on these key cytotoxic signaling proteins may overlap, to a very limited extent, with those of TBT.
Collapse
Affiliation(s)
- M M Whalen
- Department of Chemistry, Tennessee State University, Nashville, TN 37209, USA.
| | | |
Collapse
|
116
|
Fujikawa R, Higuchi S, Nakatsuji M, Yasui M, Ikedo T, Nagata M, Yokode M, Minami M. EP4 Receptor-Associated Protein in Microglia Promotes Inflammation in the Brain. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1982-1988. [PMID: 27315781 DOI: 10.1016/j.ajpath.2016.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 03/09/2016] [Accepted: 04/04/2016] [Indexed: 10/21/2022]
Abstract
Microglial cells play a key role in neuronal damage in neurodegenerative disorders. Overactivated microglia induce detrimental neurotoxic effects through the excess production of proinflammatory cytokines. However, the mechanisms of microglial activation are poorly understood. We focused on prostaglandin E2 type 4 receptor-associated protein (EPRAP), which suppresses macrophage activation. We demonstrated that EPRAP exists in microglia in the brain. Furthermore, EPRAP-deficient mice displayed less microglial accumulation, and intraperitoneal administration of lipopolysaccharide (LPS) led to reduced expression of tumor necrosis factor-α and monocyte chemoattractant protein-1 mRNA in the brains of EPRAP-deficient mice. Consistently, EPRAP-deficient microglia showed a marked decrease in the production of tumor necrosis factor-α and monocyte chemoattractant protein-1 induced by LPS treatment compared with wild-type controls. In addition, EPRAP deficiency decreased microglial activation and neuronal cell death induced by intraventricular injection of kainic acid. EPRAP deficiency impaired the LPS-induced phosphorylation of c-jun N-terminal kinase and p38 mitogen-activated protein kinase in microglia. The phosphorylation levels of mitogen-activated protein kinase kinase 4-which phosphorylates c-jun N-terminal kinase and p38 mitogen-activated protein kinase-were also decreased in EPRAP-deficient microglia after LPS stimulation. Although EPRAP in macrophages plays a role in the attenuation of inflammation, EPRAP promotes proinflammatory activation of microglia through mitogen-activated protein kinase kinase 4-mediated signaling and may be key to the deteriorating neuronal damage brought on by brain inflammation.
Collapse
Affiliation(s)
- Risako Fujikawa
- Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Japan Society for the Promotion of Science, Kyoto, Japan
| | - Sei Higuchi
- Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masato Nakatsuji
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mika Yasui
- Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Taichi Ikedo
- Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Manabu Nagata
- Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masayuki Yokode
- Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Manabu Minami
- Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| |
Collapse
|
117
|
Schonhoff CM, Park SW, Webster CR, Anwer MS. p38 MAPK α and β isoforms differentially regulate plasma membrane localization of MRP2. Am J Physiol Gastrointest Liver Physiol 2016; 310:G999-G1005. [PMID: 27012769 PMCID: PMC4935486 DOI: 10.1152/ajpgi.00005.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/14/2016] [Indexed: 01/31/2023]
Abstract
In hepatocytes, cAMP both activates p38 mitogen-activated protein kinase (MAPK) and increases the amount of multidrug resistance-associated protein-2 (MRP2) in the plasma membrane (PM-MRP2). Paradoxically, taurolithocholate (TLC) activates p38 MAPK but decreases PM-MRP2 in hepatocytes. These opposing effects of cAMP and TLC could be mediated via different p38 MAPK isoforms (α and β) that are activated differentially by upstream kinases (MKK3, MKK4, and MKK6). Thus we tested the hypothesis that p38α MAPK and p38β MAPK mediate increases and decreases in PM-MRP2 by cAMP and TLC, respectively. Studies were conducted in hepatocytes isolated from C57BL/6 wild-type (WT) and MKK3-knockout (MKK3(-/-)) mice and in a hepatoma cell line (HuH7) that overexpresses sodium-taurocholate cotransporting polypeptide (NTCP) (HuH-NTCP). Cyclic AMP activated MKK3, p38 MAPK, and p38α MAPK and increased PM-MRP2 in WT hepatocytes, but failed to activate p38α MAPK or increase PM-MRP2 in MKK3(-/-) hepatocytes. In contrast to cAMP, TLC activated total p38 MAPK but decreased PM-MRP2, and did not activate MKK3 or p38α MAPK in WT hepatocytes. In MKK3(-/-) hepatocytes, TLC still decreased PM-MRP2 and activated p38 MAPK, indicating that these effects are not MKK3-dependent. Additionally, TLC activated MKK6 in MKK3(-/-) hepatocytes, and small interfering RNA knockdown of p38β MAPK abrogated TLC-mediated decreases in PM-MRP2 in HuH-NTCP cells. Taken together, these results suggest that p38α MAPK facilitates plasma membrane insertion of MRP2 by cAMP, whereas p38β MAPK mediates retrieval of PM-MRP2 by TLC.
Collapse
Affiliation(s)
- Christopher M. Schonhoff
- 1Department of Biomedical Sciences, Cummings School of Veterinary Medicine at Tufts University, North Grafton, Massachusetts; and
| | - Se Won Park
- 1Department of Biomedical Sciences, Cummings School of Veterinary Medicine at Tufts University, North Grafton, Massachusetts; and
| | - Cynthia R.L. Webster
- 2Department of Clinical Sciences, Cummings School of Veterinary Medicine at Tufts University, North Grafton, Massachusetts
| | - M. Sawkat Anwer
- 1Department of Biomedical Sciences, Cummings School of Veterinary Medicine at Tufts University, North Grafton, Massachusetts; and
| |
Collapse
|
118
|
Huth HW, Albarnaz JD, Torres AA, Bonjardim CA, Ropert C. MEK2 controls the activation of MKK3/MKK6-p38 axis involved in the MDA-MB-231 breast cancer cell survival: Correlation with cyclin D1 expression. Cell Signal 2016; 28:1283-1291. [PMID: 27181679 DOI: 10.1016/j.cellsig.2016.05.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 05/10/2016] [Accepted: 05/11/2016] [Indexed: 10/21/2022]
Abstract
The Ras-Raf-MEK-ERK1/2 signaling pathway regulates fundamental processes in malignant cells. However, the exact contributions of MEK1 and MEK2 to the development of cancer remain to be established. We studied the effects of MEK small-molecule inhibitors (PD98059 and U0126) and MEK1 and MEK2 knock-down on cell proliferation, apoptosis and MAPK activation. We showed a diminution of cell viability that was associated with a downregulation of cyclin D1 expression and an increase of apoptosis marker in MEK2 silenced cells; by contrast, a slight increase of cell survival was observed in the absence of MEK1 that correlated with an augment of cyclin D1 expression. These data indicate that MEK2 but not MEK1 is essential for MDA-MB-231 cell survival. Importantly, the role of MEK2 in cell survival appeared independent on ERK1/2 phosphorylation since its absence did not alter the level of activated ERK1/2. Indeed, we have reported an unrevealed link between MEK2 and MKK3/MKK6-p38 MAPK axis where MEK2 was essential for the phosphorylation of MKK3/MKK6 and p38 MAPK that directly impacted on cyclin D1 expression. Importantly, the MEK1 inhibitor PD98059, like MEK1 silencing, induced an augment of cyclin D1 expression that correlated with an increase of MDA-MB-231 cell proliferation suggesting that MEK1 may play a regulatory role in these cells. In sum, the crucial role of MEK2 in MDA-MB-231 cell viability and the unknown relationship between MEK2 and MKK3/MKK6-p38 axis here revealed may open new therapeutic strategies for aggressive breast cancer.
Collapse
Affiliation(s)
- Hugo W Huth
- Departamento de Morfologia, Universidade Federal de Minas Gerais, 31270-910 Belo Horizonte, Minas Gerais, Brazil
| | - Jonas D Albarnaz
- Departamento de Microbiologia, Universidade Federal de Minas Gerais, 31270-910 Belo Horizonte, Minas Gerais, Brazil; Department of Pathology, University of Cambridge, Cambridge, UK
| | - Alice A Torres
- Departamento de Microbiologia, Universidade Federal de Minas Gerais, 31270-910 Belo Horizonte, Minas Gerais, Brazil
| | - Claudio A Bonjardim
- Departamento de Microbiologia, Universidade Federal de Minas Gerais, 31270-910 Belo Horizonte, Minas Gerais, Brazil
| | - Catherine Ropert
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, 31270-910 Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
119
|
Qin Y, Sundaram S, Essaid L, Chen X, Miller SM, Yan F, Darr DB, Galanko JA, Montgomery SA, Major MB, Johnson GL, Troester MA, Makowski L. Weight loss reduces basal-like breast cancer through kinome reprogramming. Cancer Cell Int 2016; 16:26. [PMID: 27042159 PMCID: PMC4818517 DOI: 10.1186/s12935-016-0300-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 03/22/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Obesity is associated with an aggressive subtype of breast cancer called basal-like breast cancer (BBC). BBC has no targeted therapies, making the need for mechanistic insight urgent. Reducing adiposity in adulthood can lower incidence of BBC in humans. Thus, this study investigated whether a dietary intervention to reduce adiposity prior to tumor onset would reverse HFD-induced BBC. METHODS Adult C3(1)-Tag mice were fed a low or high fat diet (LFD, HFD), and an obese group initially exposed to HFD was then switched to LFD to induce weight loss. A subset of mice was sacrificed prior to average tumor latency to examine unaffected mammary gland. Latency, tumor burden and progression was evaluated for effect of diet exposure. Physiologic, histology and proteomic analysis was undertaken to determine mechanisms regulating obesity and weight loss in BBC risk. Statistical analysis included Kaplan-Meier and log rank analysis to investigate latency. Student's t tests or ANOVA compared variables. RESULTS Mice that lost weight displayed significantly delayed latency compared to mice fed HFD, with latency matching those on LFD. Plasma leptin concentrations significantly increased with adiposity, were reduced to control levels with weight loss, and negatively correlated with tumor latency. HFD increased atypical ductal hyperplasia and ductal carcinoma in situ in mammary gland isolated prior to mean latency-a phenomenon that was lost in mice induced to lose weight. Importantly, kinome analysis revealed that weight loss reversed HFD-upregulated activity of PKC-α, PKD1, PKA, and MEK3 and increased AMPKα activity in unaffected mammary glands isolated prior to tumor latency. CONCLUSIONS Weight loss prior to tumor onset protected against the effects of HFD on latency and pre-neoplastic lesions including atypical ductal hyperplasia and DCIS. Using innovative kinomics, multiple kinases upstream of MAPK/P38α were demonstrated to be activated by HFD-induced weight gain and reversed with weight loss, providing novel targets in obesity-associated BBC. Thus, the HFD-exposed microenvironment that promoted early tumor onset was reprogrammed by weight loss and the restoration of a lean phenotype. Our work contributes to an understanding of underlying mechanisms associated with tumor and normal mammary changes that occur with weight loss.
Collapse
Affiliation(s)
- Yuanyuan Qin
- />CB 7461, Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 2203 McGavran Greenberg Hall, Chapel Hill, NC 27599-7461 USA
| | - Sneha Sundaram
- />CB 7461, Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 2203 McGavran Greenberg Hall, Chapel Hill, NC 27599-7461 USA
| | - Luma Essaid
- />CB 7461, Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 2203 McGavran Greenberg Hall, Chapel Hill, NC 27599-7461 USA
| | - Xin Chen
- />Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Samantha M. Miller
- />Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Feng Yan
- />Department of Cell and Developmental Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - David B. Darr
- />Mouse Phase I Unit, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Joseph A. Galanko
- />Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Stephanie A. Montgomery
- />Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Michael B. Major
- />Department of Cell and Developmental Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Gary L. Johnson
- />Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
- />Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Melissa A. Troester
- />Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
- />Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Liza Makowski
- />CB 7461, Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 2203 McGavran Greenberg Hall, Chapel Hill, NC 27599-7461 USA
- />Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| |
Collapse
|
120
|
Campbell J, Ryan CJ, Brough R, Bajrami I, Pemberton HN, Chong IY, Costa-Cabral S, Frankum J, Gulati A, Holme H, Miller R, Postel-Vinay S, Rafiq R, Wei W, Williamson CT, Quigley DA, Tym J, Al-Lazikani B, Fenton T, Natrajan R, Strauss SJ, Ashworth A, Lord CJ. Large-Scale Profiling of Kinase Dependencies in Cancer Cell Lines. Cell Rep 2016; 14:2490-501. [PMID: 26947069 PMCID: PMC4802229 DOI: 10.1016/j.celrep.2016.02.023] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 11/07/2015] [Accepted: 02/01/2016] [Indexed: 12/27/2022] Open
Abstract
One approach to identifying cancer-specific vulnerabilities and therapeutic targets is to profile genetic dependencies in cancer cell lines. Here, we describe data from a series of siRNA screens that identify the kinase genetic dependencies in 117 cancer cell lines from ten cancer types. By integrating the siRNA screen data with molecular profiling data, including exome sequencing data, we show how vulnerabilities/genetic dependencies that are associated with mutations in specific cancer driver genes can be identified. By integrating additional data sets into this analysis, including protein-protein interaction data, we also demonstrate that the genetic dependencies associated with many cancer driver genes form dense connections on functional interaction networks. We demonstrate the utility of this resource by using it to predict the drug sensitivity of genetically or histologically defined subsets of tumor cell lines, including an increased sensitivity of osteosarcoma cell lines to FGFR inhibitors and SMAD4 mutant tumor cells to mitotic inhibitors.
Collapse
MESH Headings
- Cell Line, Tumor
- Gene Expression Profiling
- Humans
- Mutation
- Neoplasms/enzymology
- Neoplasms/genetics
- Neoplasms/pathology
- Protein Kinases/chemistry
- Protein Kinases/genetics
- Protein Kinases/metabolism
- RNA Interference
- RNA, Small Interfering/metabolism
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Smad4 Protein/antagonists & inhibitors
- Smad4 Protein/genetics
- Smad4 Protein/metabolism
Collapse
Affiliation(s)
- James Campbell
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Colm J Ryan
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland
| | - Rachel Brough
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Ilirjana Bajrami
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Helen N Pemberton
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Irene Y Chong
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK; Royal Marsden Hospital, London SW3 6JJ, UK
| | - Sara Costa-Cabral
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Jessica Frankum
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Aditi Gulati
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Harriet Holme
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK; UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Rowan Miller
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK; UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Sophie Postel-Vinay
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK; Gustave Roussy Cancer Campus, 94805 Villejuif, France
| | - Rumana Rafiq
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Wenbin Wei
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Chris T Williamson
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - David A Quigley
- UCSF Helen Diller Family Comprehensive Cancer Centre, San Francisco, CA 94158, USA
| | - Joe Tym
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, Sutton SM2 5NG, UK
| | - Bissan Al-Lazikani
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, Sutton SM2 5NG, UK
| | - Timothy Fenton
- UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Rachael Natrajan
- Functional Genomics Laboratory, The Breast Cancer Now Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Sandra J Strauss
- UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Alan Ashworth
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK.
| | - Christopher J Lord
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK.
| |
Collapse
|
121
|
Chen J, Zhou A, Xie S, Wang C, Lv Z, Zou J. Comparative Proteomic Identification of Mature and Immature Sperm in the Catfish Cranoglanis bouderius. PLoS One 2016; 11:e0151254. [PMID: 26964044 PMCID: PMC4786320 DOI: 10.1371/journal.pone.0151254] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/25/2016] [Indexed: 02/06/2023] Open
Abstract
To understand the molecular responses of mature and immature sperm in the catfish Cranoglanis bouderius, we used the iTRAQ proteomics approach to perform proteomic profiling of spermatogenesis in C. bouderius. As a result, 1,941 proteins were identified, including 361 differentially expressed proteins, 157 upregulated proteins and 204 downregulated proteins in mature sperm relative to immature sperm. All of the identified proteins were categorized into seven types of subcellular localizations and three molecular functions and were found to be involved in nine biological processes. All of the differential proteins were involved in 235 different pathways. Moreover, we found that the tricarboxylic acid (TCA) pathway played an important role in the energy metabolism of sperm and that the EABB pathway was involved in the mechanism of spermatogenesis. Our study is the first to use the iTRAQ-based proteomic approach to analyze the catfish sperm proteome, and the results we obtained using this approach are valuable for understanding the molecular mechanisms of fish spermatogenesis.
Collapse
Affiliation(s)
- Jintao Chen
- College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Aiguo Zhou
- College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Shaolin Xie
- College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Chao Wang
- College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Zijun Lv
- College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
- Qingyuan North River Fishery Science Institute, Qingyuan, Guangdong, China
| | - Jixing Zou
- College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
- * E-mail:
| |
Collapse
|
122
|
刘 春, 江 庆, 林 丹, 陈 炜, 肖 艳, 林 莉, 邓 渊, 蒋 惠, 郭 遂. [Coexpression of MAP2K4 and vimentin proteins in human endometrial carcinoma and its clinicopathological significance]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2016; 37:157-164. [PMID: 28219857 PMCID: PMC6779670 DOI: 10.3969/j.issn.1673-4254.2017.02.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Indexed: 06/06/2023]
Abstract
OBJECTIVE To analyze the expression of MAP2K4 and vimentin in human endometrial carcinoma (EC) and their association with the clinicopathological features and prognosis of the patients. METHODS MAP2K4 and vimentin expressions were detected immunohistochemically in paraffin-embedded tissue sections from 128 patients with EC, and the correlation of MAP2K4 and vimentin expressions with the clinicopathological factors of the patients was analyzed. RESULTS MAP2K4 and vimentin proteins were positively expressed in 49 (38.3%) and 83 (64.8%) of the patients, respectively. A positive expression of MAP2K4 was negatively correlated with FIGO stage of the tumor (P=0.010) and lymph node status (P=0.016); a positive expression of vimentin was positively correlated with FIGO stage of the tumor (P=0.025), histological grades (P=0.017), depth of myometrial invasion (P=0.044) and lymph node status (P=0.032). MAP2K4 was inversely associated with vimentin expression in EC(r=-0.598, P<0.001). Patients positive for MAP2K4 tended to have a higher overall survival rate (P=0.002), and those positive for vimentin tended to have a lower overall survival rate (P=0.007); patients positive for MAP2K4 but negative for vimentin had the longest survival time, while those negative for MAP2K4 and positive for vimentin had lowest survival rate (P=0.004). CONCLUSION Detection of MAP2K4 and vimentin might help in early diagnosis and prognostic evaluation of patients with EC.
Collapse
Affiliation(s)
- 春花 刘
- 南方医科大学第三附属医院妇产科, 广东 广州 510630Department of Obstetrics and Gynecology, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - 庆萍 江
- 广州医科大学第三附属医院病理科, 广东 广州 510150Department of Pathology, Third Affiliated Hospital of Guangzhou Medical College, Guangzhou 510150, China
| | - 丹 林
- 南方医科大学第三附属医院妇产科, 广东 广州 510630Department of Obstetrics and Gynecology, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - 炜 陈
- 南方医科大学第三附属医院妇产科, 广东 广州 510630Department of Obstetrics and Gynecology, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - 艳怡 肖
- 南方医科大学第三附属医院妇产科, 广东 广州 510630Department of Obstetrics and Gynecology, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - 莉 林
- 南方医科大学第三附属医院妇产科, 广东 广州 510630Department of Obstetrics and Gynecology, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - 渊润 邓
- 南方医科大学第三附属医院妇产科, 广东 广州 510630Department of Obstetrics and Gynecology, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - 惠萍 蒋
- 南方医科大学第三附属医院妇产科, 广东 广州 510630Department of Obstetrics and Gynecology, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - 遂群 郭
- 南方医科大学第三附属医院妇产科, 广东 广州 510630Department of Obstetrics and Gynecology, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| |
Collapse
|
123
|
Sharma V, Young L, Allison AB, Owen K. Registered report: Diverse somatic mutation patterns and pathway alterations in human cancers. eLife 2016; 5. [PMID: 26894955 PMCID: PMC4769161 DOI: 10.7554/elife.11566] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 01/08/2016] [Indexed: 11/13/2022] Open
Abstract
The Reproducibility Project: Cancer Biology seeks to address growing concerns about reproducibility in scientific research by conducting replications of selected experiments from a number of high-profile papers in the field of cancer biology. The papers, which were published between 2010 and 2012, were selected on the basis of citations and Altmetric scores (Errington et al., 2014). This Registered Report describes the proposed replication plan of key experiments from "Diverse somatic mutation patterns and pathway alterations in human cancers" by Kan and colleagues published in Nature in 2010 (Kan et al., 2010). The experiments to be replicated are those reported in Figures 3D-F and 4C-F. Kan and colleagues utilized mismatch repair detection (MRD) technology to identify somatic mutations in primary human tumor samples and identified a previously uncharacterized arginine 243 to histidine (R243H) mutation in the G-protein α subunit GNAO1 in breast carcinoma tissue. In Figures 3D-F, Kan and colleagues demonstrated that stable expression of mutant GNAO1(R243D) conferred a significant growth advantage in human mammary epithelial cells, confirming the oncogenic potential of this mutation. Similarly, expression of variants with somatic mutations in MAP2K4, a JNK pathway kinase (shown in Figures 4C-E) resulted in a significant increase in anchorage-independent growth. Interestingly, these mutants exhibited reduced kinase activity compared to wild type MAP2K4, indicating these mutations impose a dominant-negative influence to promote growth (Figure 4F). The Reproducibility Project: Cancer Biology is a collaboration between the Center for Open Science and Science Exchange and the results of the replications will be published in eLife.
Collapse
Affiliation(s)
| | - Lisa Young
- Applied Biological Materials, Richmond, Canada
| | - Anne B Allison
- Piedmond Virginia Community College, Charlottesville, United States
| | - Kate Owen
- University of Virginia, Charlottesville, United States
| | | | | |
Collapse
|
124
|
Rodriguez-Canales J, Parra-Cuentas E, Wistuba II. Diagnosis and Molecular Classification of Lung Cancer. Cancer Treat Res 2016; 170:25-46. [PMID: 27535388 DOI: 10.1007/978-3-319-40389-2_2] [Citation(s) in RCA: 165] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Lung cancer is a complex disease composed of diverse histological and molecular types with clinical relevance. The advent of large-scale molecular profiling has been helpful to identify novel molecular targets that can be applied to the treatment of particular lung cancer patients and has helped to reshape the pathological classification of lung cancer. Novel directions include the immunotherapy revolution, which has opened the door for new opportunities for cancer therapy and is also redefining the classification of multiple tumors, including lung cancer. In the present chapter, we will review the main current basis of the pathological diagnosis and classification of lung cancer incorporating the histopathological and molecular dimensions of the disease.
Collapse
Affiliation(s)
- Jaime Rodriguez-Canales
- Department of Translational Molecular Pathology, Unit 951, The University of Texas MD Anderson Cancer Center, 2130 Holcombe Blvd., Houston, TX, 77030, USA
| | - Edwin Parra-Cuentas
- Department of Translational Molecular Pathology, Unit 951, The University of Texas MD Anderson Cancer Center, 2130 Holcombe Blvd., Houston, TX, 77030, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, Unit 951, The University of Texas MD Anderson Cancer Center, 2130 Holcombe Blvd., Houston, TX, 77030, USA.
| |
Collapse
|
125
|
Park SI, Park SJ, Lee J, Kim HE, Park SJ, Sohn JW, Park YG. Inhibition of cyclic AMP response element-directed transcription by decoy oligonucleotides enhances tumor-specific radiosensitivity. Biochem Biophys Res Commun 2015; 469:363-9. [PMID: 26655813 DOI: 10.1016/j.bbrc.2015.11.122] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 11/26/2015] [Indexed: 10/22/2022]
Abstract
The radiation stress induces cytotoxic responses of cell death as well as cytoprotective responses of cell survival. Understanding exact cellular mechanism and signal transduction pathways is important in improving cancer radiotherapy. Increasing evidence suggests that cyclic AMP response element binding protein (CREB)/activating transcription factor (ATF) family proteins act as a survival factor and a signaling molecule in response to stress. We postulated that CREB inhibition via CRE decoy oligonucleotide increases tumor cell sensitization to γ-irradiation-induced cytotoxic stress. In the present study, we demonstrate that CREB phosphorylation and CREB DNA-protein complex formation increased in time- and radiation dose-dependent manners, while there was no significant change in total protein level of CREB. In addition, CREB was phosphorylated in response to γ-irradiation through p38 MAPK pathway. Further investigation revealed that CREB blockade by decoy oligonucleotides functionally inhibited transactivation of CREB, and significantly increased radiosensitivity of multiple human cancer cell lines including TP53- and/or RB-mutated cells with minimal effects on normal cells. We also demonstrate that tumor cells ectopically expressing dominant negative mutant CREB (KCREB) and the cells treated with p38 MAPK inhibitors were more sensitive to γ-irradiation than wild type parental cells or control-treated cells. Taken together, we conclude that CREB protects tumor cells from γ-irradiation, and combination of CREB inhibition plus ionizing radiation will be a promising radiotherapeutic approach.
Collapse
Affiliation(s)
- Serk In Park
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea; The BK21 Plus Program for Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea; Department of Medicine and Center for Bone Biology, Vanderbilt University School of Medicine, Nashville, TN, USA.
| | - Sung-Jun Park
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea; Laboratory of Obesity and Aging Research, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Junghan Lee
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hye Eun Kim
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Su Jin Park
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jeong-Won Sohn
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yun Gyu Park
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
126
|
Chun KS, Shim M. EP2 Induces p38 Phosphorylation via the Activation of Src in HEK 293 Cells. Biomol Ther (Seoul) 2015; 23:539-48. [PMID: 26535079 PMCID: PMC4624070 DOI: 10.4062/biomolther.2015.043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 07/14/2015] [Accepted: 08/17/2015] [Indexed: 12/14/2022] Open
Abstract
Prostaglandin E2 (PGE2), a major product of cyclooxygenase, binds to four different prostaglandin E2 receptors (EP1, EP2, EP3, and EP4) which are G-protein coupled transmembrane receptors (GPCRs). Although GPCRs including EP receptors have been shown to be associated with their specific G proteins, recent evidences suggest that GPCRs can regulate MAPK signaling via non-G protein coupled pathways including Src. EP2 is differentially expressed in various tissues and the expression of EP2 is induced by extracellular stimuli. We hypothesized that an increased level of EP2 expression may affect MAPK signaling. The overexpression of EP2 in HEK 293 cells resulted in significant increase in intracellular cAMP levels response to treatment with butaprost, a specific EP2 agonist, while overexpression of EP2 alone did not increase intracellular cAMP levels. However, EP2 overexpression in the absence of PGE2 induced an increase in the level of p38 phosphorylation as well as the kinase activity of p38, suggesting that up-regulation of EP2 may promote p38 activation via non-G protein coupled pathway. Inhibition of Src completely blocked EP2-induced p38 phosphorylation and overexpression of Src increased the level of p38 phosphorylation, indicating that Src is upstream kinase for EP2-induced p38 phosphorylation. EP2 overexpression also increased the Src activity and EP2 protein was co-immunoprecipitated with Src. Furthermore, sequential co-immunoprecipitation studies showed that EP2, Src, and β-arrestin can form a complex. Our study found a novel pathway in which EP2 is associated with Src, regulating p38 pathway.
Collapse
Affiliation(s)
- Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Minsub Shim
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| |
Collapse
|
127
|
Protective Mechanisms of Flavonoids in Parkinson's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:314560. [PMID: 26576219 PMCID: PMC4630416 DOI: 10.1155/2015/314560] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 01/29/2015] [Indexed: 12/11/2022]
Abstract
Parkinson's disease is a chronic, debilitating neurodegenerative movement disorder characterized by progressive degeneration of dopaminergic neurons in the substantia nigra pars compacta region in human midbrain. To date, oxidative stress is the well accepted concept in the etiology and progression of Parkinson's disease. Hence, the therapeutic agent is targeted against suppressing and alleviating the oxidative stress-induced cellular damage. Within the past decades, an explosion of research discoveries has reported on the protective mechanisms of flavonoids, which are plant-based polyphenols, in the treatment of neurodegenerative disease using both in vitro and in vivo models. In this paper, we have reviewed the literature on the neuroprotective mechanisms of flavonoids in protecting the dopaminergic neurons hence reducing the symptoms of this movement disorder. The mechanism reviewed includes effect of flavonoids in activation of endogenous antioxidant enzymes, suppressing the lipid peroxidation, inhibition of inflammatory mediators, flavonoids as a mitochondrial target therapy, and modulation of gene expression in neuronal cells.
Collapse
|
128
|
Bardwell AJ, Bardwell L. Two hydrophobic residues can determine the specificity of mitogen-activated protein kinase docking interactions. J Biol Chem 2015; 290:26661-74. [PMID: 26370088 DOI: 10.1074/jbc.m115.691436] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Indexed: 11/06/2022] Open
Abstract
MAPKs bind to many of their upstream regulators and downstream substrates via a short docking motif (the D-site) on their binding partner. MAPKs that are in different families (e.g. ERK, JNK, and p38) can bind selectively to D-sites in their authentic substrates and regulators while discriminating against D-sites in other pathways. Here we demonstrate that the short hydrophobic region at the distal end of the D-site plays a critical role in determining the high selectivity of JNK MAPKs for docking sites in their cognate MAPK kinases. Changing just 1 or 2 key hydrophobic residues in this submotif is sufficient to turn a weak JNK-binding D-site into a strong one, or vice versa. These specificity-determining differences are also found in the D-sites of the ETS family transcription factors Elk-1 and Net. Moreover, swapping two hydrophobic residues between these D-sites switches the relative efficiency of Elk-1 and Net as substrates for ERK versus JNK, as predicted. These results provide new insights into docking specificity and suggest that this specificity can evolve rapidly by changes to just 1 or 2 amino acids.
Collapse
Affiliation(s)
- A Jane Bardwell
- From the Department of Developmental and Cell Biology, Center for Complex Biological Systems, University of California, Irvine, California 92697
| | - Lee Bardwell
- From the Department of Developmental and Cell Biology, Center for Complex Biological Systems, University of California, Irvine, California 92697
| |
Collapse
|
129
|
Tian T, Harding A. How MAP kinase modules function as robust, yet adaptable, circuits. Cell Cycle 2015; 13:2379-90. [PMID: 25483189 DOI: 10.4161/cc.29349] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Genetic and biochemical studies have revealed that the diversity of cell types and developmental patterns evident within the animal kingdom is generated by a handful of conserved, core modules. Core biological modules must be robust, able to maintain functionality despite perturbations, and yet sufficiently adaptable for random mutations to generate phenotypic variation during evolution. Understanding how robust, adaptable modules have influenced the evolution of eukaryotes will inform both evolutionary and synthetic biology. One such system is the MAP kinase module, which consists of a 3-tiered kinase circuit configuration that has been evolutionarily conserved from yeast to man. MAP kinase signal transduction pathways are used across eukaryotic phyla to drive biological functions that are crucial for life. Here we ask the fundamental question, why do MAPK modules follow a conserved 3-tiered topology rather than some other number? Using computational simulations, we identify a fundamental 2-tiered circuit topology that can be readily reconfigured by feedback loops and scaffolds to generate diverse signal outputs. When this 2-kinase circuit is connected to proximal input kinases, a 3-tiered modular configuration is created that is both robust and adaptable, providing a biological circuit that can regulate multiple phenotypes and maintain functionality in an uncertain world. We propose that the 3-tiered signal transduction module has been conserved through positive selection, because it facilitated the generation of phenotypic variation during eukaryotic evolution.
Collapse
Affiliation(s)
- Tianhai Tian
- a School of Mathematical Science; Monash University; Victoria, Australia
| | | |
Collapse
|
130
|
Jin LG, Chu JJ, Pang QF, Zhang FZ, Wu G, Zhou LY, Zhang XJ, Xing CG. Caffeic acid phenethyl ester attenuates ionize radiation-induced intestinal injury through modulation of oxidative stress, apoptosis and p38MAPK in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 40:156-163. [PMID: 26122083 DOI: 10.1016/j.etap.2015.05.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Revised: 05/26/2015] [Accepted: 05/26/2015] [Indexed: 06/04/2023]
Abstract
Caffeic acid phenyl ester (CAPE) is a potent anti-inflammatory agent and it can eliminate the free radicals. This study aimed to investigate the radioprotective effects of CAPE on X-ray irradiation induced intestinal injury in rats. Rats were intragastrically administered with 10 μmol/kg/d CAPE for 7 consecutive days before exposing them to a single dose of X-ray irradiation (9Gy) to abdomen. Rats were sacrificed 72 h after exposure to radiation. We found that pretreatment with CAPE effectively attenuated intestinal pathology changes, apoptosis, oxidative stress, bacterial translocation, the content of nitric oxide and myeloperoxidase as well as the concentration of plasma tumor necrosis factor-α. Pretreatment with CAPE also reversed the activation of p38MAPK and the increased expression of intercellular cell adhesion molecule-1 induced by radiation in intestinal mucosa. Taken together, these results suggest that pretreatment with CAPE could be a promising candidate for treating radiation-induced intestinal injury.
Collapse
Affiliation(s)
- Liu-Gen Jin
- Department of Surgery, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, 215004 Suzhou, China
| | - Jian-Jun Chu
- Department of Oncology, The Affiliated Hospital of Jiangnan University, 200 Huihe Road, 214122 Wuxi, China
| | - Qing-Feng Pang
- Wuxi Medical School, Jiangnan University, 1800 Lihu Road, 214122 Wuxi, China
| | - Fu-Zheng Zhang
- Department of Oncology, The Affiliated Hospital of Jiangnan University, 200 Huihe Road, 214122 Wuxi, China
| | - Gang Wu
- Department of Oncology, The Affiliated Hospital of Jiangnan University, 200 Huihe Road, 214122 Wuxi, China
| | - Le-Yuan Zhou
- Department of Oncology, The Affiliated Hospital of Jiangnan University, 200 Huihe Road, 214122 Wuxi, China
| | - Xiao-Jun Zhang
- Department of Oncology, The Affiliated Hospital of Jiangnan University, 200 Huihe Road, 214122 Wuxi, China
| | - Chun-Gen Xing
- Department of Surgery, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, 215004 Suzhou, China.
| |
Collapse
|
131
|
Martin ED, Bassi R, Marber MS. p38 MAPK in cardioprotection - are we there yet? Br J Pharmacol 2015; 172:2101-13. [PMID: 25204838 PMCID: PMC4386984 DOI: 10.1111/bph.12901] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 08/22/2014] [Accepted: 08/28/2014] [Indexed: 12/14/2022] Open
Abstract
PKs transfer a phosphate from ATP to the side-chain hydroxyl group of a serine, threonine or tyrosine residue of a substrate protein. This in turn can alter that protein's function; modulating fundamental cellular processes including, metabolism, transcription, growth, division, differentiation, motility and survival. PKs are subdivided into families based on homology. One such group are the stress-activated kinases, which as the name suggests, are activated in response to cellular stresses such as toxins, cytokines, mechanical deformation and osmotic stress. Members include the p38 MAPK family, which is composed of α, β, γ and δ, isoforms which are encoded by separate genes. These kinases transduce extracellular signals and coordinate the cellular responses needed for adaptation and survival. However, in cardiovascular and other disease states, these same systems can trigger maladaptive responses that aggravate, rather than alleviate, the disease. This situation is analogous to adrenergic, angiotensin and aldosterone signalling in heart failure, where inhibition is beneficial despite the importance of these hormones to homeostasis. The question is whether similar benefits could accrue from p38 inhibition? In this review, we will discuss the structure and function of p38, the history of p38 inhibitors and their use in preclinical studies. Finally, we will summarize the results of recent cardiovascular clinical trials with p38 inhibitors.
Collapse
Affiliation(s)
- E D Martin
- King's College London BHF Centre of Research Excellence, Cardiovascular Division, The Rayne Institute, St Thomas' HospitalLondon, UK
| | - R Bassi
- King's College London BHF Centre of Research Excellence, Cardiovascular Division, The Rayne Institute, St Thomas' HospitalLondon, UK
| | - M S Marber
- King's College London BHF Centre of Research Excellence, Cardiovascular Division, The Rayne Institute, St Thomas' HospitalLondon, UK
| |
Collapse
|
132
|
Lee JH, Kim JE, Jang YJ, Lee CC, Lim TG, Jung SK, Lee E, Lim SS, Heo YS, Seo SG, Son JE, Kim JR, Lee CY, Lee HJ, Lee KW. Dehydroglyasperin C suppresses TPA-induced cell transformation through direct inhibition of MKK4 and PI3K. Mol Carcinog 2015; 55:552-62. [DOI: 10.1002/mc.22302] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 12/26/2014] [Accepted: 01/21/2015] [Indexed: 01/08/2023]
Affiliation(s)
- Ji Hoon Lee
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence; Seoul National University; Seoul Republic of Korea
- Advanced Institutes of Convergence Technology; Seoul National University; Suwon Republic of Korea
| | - Jong-Eun Kim
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence; Seoul National University; Seoul Republic of Korea
- Advanced Institutes of Convergence Technology; Seoul National University; Suwon Republic of Korea
- Research Institute of Bio Food Industry, Institute of Green Bio Science and Technology; Seoul National University; Pyeongchang Republic of Korea
| | - Young Jin Jang
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence; Seoul National University; Seoul Republic of Korea
- Division of Creative Food Science for Health; Korea Food Research Institute; Seongnam Republic of Korea
| | - Charles C. Lee
- Department of Food Science and Technology; Cornell University; Ithaca NY 14456 USA
| | - Tae-Gyu Lim
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence; Seoul National University; Seoul Republic of Korea
- Advanced Institutes of Convergence Technology; Seoul National University; Suwon Republic of Korea
| | - Sung Keun Jung
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence; Seoul National University; Seoul Republic of Korea
- Division of Creative Food Science for Health; Korea Food Research Institute; Seongnam Republic of Korea
| | - Eunjung Lee
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence; Seoul National University; Seoul Republic of Korea
- Traditional Alcoholic Beverage Research Team; Korea Food Research Institute; Seongnam Republic of Korea
| | - Soon Sung Lim
- Department of Food Science and Nutrition; Hallym University; Chuncheon Republic of Korea
| | - Yong Seok Heo
- Department of Chemistry; Konkuk University; Seoul Republic of Korea
| | - Sang Gwon Seo
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence; Seoul National University; Seoul Republic of Korea
- Advanced Institutes of Convergence Technology; Seoul National University; Suwon Republic of Korea
| | - Joe Eun Son
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence; Seoul National University; Seoul Republic of Korea
- Advanced Institutes of Convergence Technology; Seoul National University; Suwon Republic of Korea
| | - Jong Rhan Kim
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence; Seoul National University; Seoul Republic of Korea
- Advanced Institutes of Convergence Technology; Seoul National University; Suwon Republic of Korea
| | - Chang Yong Lee
- Department of Food Science and Technology; Cornell University; Ithaca NY 14456 USA
- Department of Biochemistry; King Abdulaziz University; Jeddah SA
| | - Hyong Joo Lee
- Research Institute of Bio Food Industry, Institute of Green Bio Science and Technology; Seoul National University; Pyeongchang Republic of Korea
| | - Ki Won Lee
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence; Seoul National University; Seoul Republic of Korea
- Advanced Institutes of Convergence Technology; Seoul National University; Suwon Republic of Korea
- Research Institute of Bio Food Industry, Institute of Green Bio Science and Technology; Seoul National University; Pyeongchang Republic of Korea
- Institute on Aging; Seoul National University; Seoul Republic of Korea
| |
Collapse
|
133
|
Antileishmanial effect of 18β-glycyrrhetinic acid is mediated by Toll-like receptor-dependent canonical and noncanonical p38 activation. Antimicrob Agents Chemother 2015; 59:2531-9. [PMID: 25691644 DOI: 10.1128/aac.03997-14] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 02/05/2015] [Indexed: 01/08/2023] Open
Abstract
18β-Glycyrrhetinic acid (GRA), a natural immunomodulator, greatly reduced the parasite load in experimental visceral leishmaniasis through nitric oxide (NO) upregulation, proinflammatory cytokine expression, and NF-κB activation. For the GRA-mediated effect, the primary kinase responsible was found to be p38, and analysis of phosphorylation kinetics as well as studies with dominant-negative (DN) constructs revealed mitogen-activated protein kinase kinase 3 (MKK3) and MKK6 as the immediate upstream regulators of p38. However, detection of remnant p38 kinase activity in the presence of both DN MKK3 and MKK6 suggested alternative pathways of p38 activation. That residual p38 activity was attributed to an autophosphorylation event ensured by the transforming growth factor β-activated kinase 1 (TAK1)-binding protein 1 (TAB1)-p38 interaction and was completely abolished upon pretreatment with SB203580 in DN MKK3/6 double-transfected macrophage cells. Further upstream signaling evaluation by way of phosphorylation kinetics and transfection studies with DN constructs identified TAK1, myeloid differentiation factor 88 (MyD88), interleukin 1 receptor (IL-1R)-activated kinase 1 (IRAK1), and tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6) as important contributors to GRA-mediated macrophage activation. Finally, gene knockdown studies revealed Toll-like receptor 2 (TLR2) and TLR4 as the membrane receptors associated with GRA-mediated antileishmanial activity. Together, the results of this study brought mechanistic insight into the antileishmanial activity of GRA, which is dependent on the TLR2/4-MyD88 signaling axis, leading to MKK3/6-mediated canonical and TAB1-mediated noncanonical p38 activation.
Collapse
|
134
|
Targeting MKK3 as a novel anticancer strategy: molecular mechanisms and therapeutical implications. Cell Death Dis 2015; 6:e1621. [PMID: 25633290 PMCID: PMC4669782 DOI: 10.1038/cddis.2014.591] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 12/11/2014] [Accepted: 12/15/2014] [Indexed: 01/10/2023]
Abstract
Mitogen-activated protein kinase kinase 3 (MAP2K3, MKK3) is a member of the dual specificity protein kinase group that belongs to the MAP kinase kinase family. This kinase is activated by mitogenic or stress-inducing stimuli and participates in the MAP kinase-mediated signaling cascade, leading to cell proliferation and survival. Several studies highlighted a critical role for MKK3 in tumor progression and invasion, and we previously identified MKK3 as transcriptional target of mutant (mut) p53 to sustain cell proliferation and survival, thus rendering MKK3 a promising target for anticancer therapies. Here, we found that targeting MKK3 with RNA interference, in both wild-type (wt) and mutp53-carrying cells, induced endoplasmic reticulum stress and autophagy that, respectively, contributed to stabilize wtp53 and degrade mutp53. MKK3 depletion reduced cancer cell proliferation and viability, whereas no significant effects were observed in normal cellular context. Noteworthy, MKK3 depletion in combination with chemotherapeutic agents increased tumor cell response to the drugs, in both wtp53 and mutp53 cancer cells, as demonstrated by enhanced poly (ADP-ribose) polymerase cleavage and reduced clonogenic ability in vitro. In addition, MKK3 depletion reduced tumor growth and improved biological response to chemotherapeutic in vivo. The overall results indicate MKK3 as a novel promising molecular target for the development of more efficient anticancer treatments in both wtp53- and mutp53-carrying tumors.
Collapse
|
135
|
Suhail TV, Singh P, Manna TK. Suppression of centrosome protein TACC3 induces G1 arrest and cell death through activation of p38-p53-p21 stress signaling pathway. Eur J Cell Biol 2015; 94:90-100. [PMID: 25613365 DOI: 10.1016/j.ejcb.2014.12.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 11/23/2014] [Accepted: 12/08/2014] [Indexed: 11/28/2022] Open
Abstract
The centrosome regulates diverse cellular processes, including cell proliferation and differentiation. TACC3, a member of the human transforming acidic coiled-coil protein family, is a key centrosomal protein that is up-regulated in many cancers. Previous studies have demonstrated that TACC3 is essential for the survival of vertebrates and is involved in cell cycle regulation in human cells. However, the details of the underlying mechanisms in its cell cycle regulatory activity remain poorly understood. In this study, we showed that suppression of TACC3 expression induced G1 cell cycle arrest and triggered cell death in human cells. TACC3 depletion-induced G1 arrest and cell death were significantly reduced in cells either lacking p53 or with pharmacologically-inhibited p38, indicating that G1 arrest and cell death induction both require p53 and p38. TACC3 depletion up-regulated the levels of p53 and p21 and induced the accumulation of p53 both in the nucleus and at the centrosome. Interestingly, TACC3 depletion led to the activation of p38 and stimulated the recruitment of activated p38 to the centrosome. Depletion of TACC3 up-regulated the phosphorylation of p53 at Serine 33, a site known to be phosphorylated by p38 under cellular stress and further induced the accumulation of phosphorylated p53 to the centrosome. Loss of TACC3 affected centrosome integrity by disrupting the localization of components of the γ-tubulin ring complex at the centrosome. The results demonstrate that TACC3 depletion induces G1 arrest and cell death by activating p38-p53-p21 signaling and triggering a centrosome-mediated cellular stress response.
Collapse
Affiliation(s)
- Thazhath V Suhail
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, CET Campus, Trivandrum 695016, Kerala, India
| | - Puja Singh
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, CET Campus, Trivandrum 695016, Kerala, India
| | - Tapas K Manna
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, CET Campus, Trivandrum 695016, Kerala, India.
| |
Collapse
|
136
|
Zhang L, Zhou M, Wang Y, Huang W, Qin G, Weintraub NL, Tang Y. miR-92a inhibits vascular smooth muscle cell apoptosis: role of the MKK4-JNK pathway. Apoptosis 2014; 19:975-83. [PMID: 24705900 DOI: 10.1007/s10495-014-0987-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Vascular smooth muscle cell (VSMC) apoptosis plays an important role in vascular remodeling and atherosclerotic plaque instability. Oxidative stress in diseased vessels promotes VSMC apoptosis in part by activating the c-Jun N-terminal kinase (JNK) pathway, which has been identified as a molecular target of miR-92a in macrophages. Here, we examined the expression and biological activity of miR-92a in VSMC. Quiescent VSMC exhibited a low basal expression of miR-92a, which was positively regulated by serum stimulation and negatively regulated by H2O2. Overexpression of miR-92a decreased H2O2-induced VSMC apoptosis as indicated by TUNEL assay and cleaved caspase-3 protein levels. Using 3'UTR-reporter assay, we found that miR-92a overexpression led to suppression of both mitogen-activated protein kinase kinase 4 (MKK4)- and JNK1-dependent luciferase activity. We also found that 10 mer seed match between miRNA:mRNA pair is more efficient than 8 mer seed match for us to identify authentic miRNA target. Protein levels of active phospho-JNK and phospho-c-Jun, downstream targets of the MKK4-JNK1 pathway, were also decreased by overexpressing miR-92a in VSMC under oxidative stress. Consistent with these findings, overexpression of MKK4 reversed the anti-apoptotic effects of miR-92a in oxidatively stressed VSMC. In conclusion, miR-92a overexpression inhibits H2O2-induced VSMC apoptosis by directly targeting the MKK4-JNK1 pathway.
Collapse
Affiliation(s)
- Lan Zhang
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | | | | | | | | | | | | |
Collapse
|
137
|
Widespread genetic epistasis among cancer genes. Nat Commun 2014; 5:4828. [PMID: 25407795 DOI: 10.1038/ncomms5828] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 07/28/2014] [Indexed: 12/17/2022] Open
Abstract
Quantitative genetic epistasis has been hypothesized to be an important factor in the development and progression of complex diseases. Cancers in particular are driven by the accumulation of mutations that may act epistatically during the course of the disease. However, as cancer mutations are uncovered at an unprecedented rate, determining which combinations of genetic alterations interact to produce cancer phenotypes remains a challenge. Here we show that by using combinatorial RNAi screening in cell culture, dense and often previously undetermined interactions among cancer genes were revealed by assessing gene pairs that are frequently co-altered in primary breast cancers. These interacting gene pairs are significantly associated with survival time when co-altered in patients, indicating that genetic interaction mapping may be leveraged to improve risk assessment. As many of these interacting gene pairs involve known drug targets, personalized treatment regimens may be improved by overlaying genetic interactions with mutational profiling.
Collapse
|
138
|
Ben-Ami Shor D, Blank M, Reuter S, Matthias T, Beiglass I, Volkov A, Barshack I, Shoenfeld Y. Anti-ribosomal-P antibodies accelerate lupus glomerulonephritis and induce lupus nephritis in naïve mice. J Autoimmun 2014; 54:118-26. [DOI: 10.1016/j.jaut.2014.02.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 02/18/2014] [Accepted: 02/26/2014] [Indexed: 11/24/2022]
|
139
|
Arcila ME, Drilon A, Sylvester BE, Lovly CM, Borsu L, Reva B, Kris MG, Solit DB, Ladanyi M. MAP2K1 (MEK1) Mutations Define a Distinct Subset of Lung Adenocarcinoma Associated with Smoking. Clin Cancer Res 2014; 21:1935-43. [PMID: 25351745 DOI: 10.1158/1078-0432.ccr-14-2124] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 10/09/2014] [Indexed: 01/26/2023]
Abstract
PURPOSE Genetic alterations affecting the MAPK/ERK pathway are common in lung adenocarcinoma (LAD). Early steps of the signaling pathway are most often affected with EGFR, KRAS, and BRAF mutations encompassing more than 70% of all alterations. Somatic mutations in MEK1, located downstream of BRAF, are rare and remain poorly defined as a distinct molecular subset. EXPERIMENTAL DESIGN Tumors harboring MEK1 mutations were identified through targeted screening of a large LAD cohort concurrently interrogated for recurrent mutations in MEK1, EGFR, KRAS, BRAF, ERBB2/HER2, NRAS, PIK3CA, and AKT. Additional cases were identified through a search of publically available cancer genomic datasets. Mutations were correlated with patient characteristics and treatment outcomes. Overall survival was compared with stage-matched patients with KRAS- and EGFR-mutant LADs. RESULTS We identified 36 MEK1-mutated cases among 6,024 LAD (0.6%; 95% confidence interval, 0.42-0.85). The majority of patients were smokers (97%, n = 35/36). There was no association with age, sex, race, or stage. The most common mutations were K57N (64%, 23/36) followed by Q56P (19%, 7/36), all mutually exclusive with other driver mutations in the targeted panel. Transversions G:C>T:A were predominant (89%, 31/35), in keeping with smoking-associated DNA damage. Additional less common somatic mutations were identified in the kinase domain, all of which are predicted to converge into a single interaction area based on in silico 3D modeling. CONCLUSIONS MEK1 mutations define a distinct subset of lung cancers (∼1%) with potential sensitivity to MEK inhibitors. Mutations are predominantly transversions, in keeping with a strong association with smoking.
Collapse
Affiliation(s)
- Maria E Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Alexander Drilon
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, New York
| | - Brooke E Sylvester
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christine M Lovly
- Department of Medicine, Division of Hematology-Oncology, Vanderbilt University School of Medicine, Vanderbilt Ingram Cancer Center, Nashville, Tennessee
| | - Laetitia Borsu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Boris Reva
- Computational Biology Center, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mark G Kris
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, New York
| | - David B Solit
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York. Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
140
|
Cheung LWT, Yu S, Zhang D, Li J, Ng PKS, Panupinthu N, Mitra S, Ju Z, Yu Q, Liang H, Hawke DH, Lu Y, Broaddus RR, Mills GB. Naturally occurring neomorphic PIK3R1 mutations activate the MAPK pathway, dictating therapeutic response to MAPK pathway inhibitors. Cancer Cell 2014; 26:479-94. [PMID: 25284480 PMCID: PMC4198486 DOI: 10.1016/j.ccell.2014.08.017] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 06/05/2014] [Accepted: 08/26/2014] [Indexed: 12/17/2022]
Abstract
PIK3R1 (p85α regulatory subunit of PI3K) is frequently mutated across cancer lineages. Herein, we demonstrate that the most common recurrent PIK3R1 mutation PIK3R1(R348∗) and a nearby mutation PIK3R1(L370fs), in contrast to wild-type and mutations in other regions of PIK3R1, confers an unexpected sensitivity to MEK and JNK inhibitors in vitro and in vivo. Consistent with the response to inhibitors, PIK3R1(R348∗) and PIK3R1(L370fs) unexpectedly increase JNK and ERK phosphorylation. Surprisingly, p85α R348(∗) and L370fs localize to the nucleus where the mutants provide a scaffold for multiple JNK pathway components facilitating nuclear JNK pathway activation. Our findings uncover an unexpected neomorphic role for PIK3R1(R348∗) and neighboring truncation mutations in cellular signaling, providing a rationale for therapeutic targeting of these mutant tumors.
Collapse
Affiliation(s)
- Lydia W T Cheung
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Shuangxing Yu
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dong Zhang
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jie Li
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Patrick K S Ng
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nattapon Panupinthu
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Shreya Mitra
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhenlin Ju
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Qinghua Yu
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Han Liang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - David H Hawke
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yiling Lu
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Russell R Broaddus
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gordon B Mills
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
141
|
Darling NJ, Cook SJ. The role of MAPK signalling pathways in the response to endoplasmic reticulum stress. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2150-63. [DOI: 10.1016/j.bbamcr.2014.01.009] [Citation(s) in RCA: 222] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 12/20/2013] [Accepted: 01/09/2014] [Indexed: 12/30/2022]
|
142
|
Wu RCC, Cho WL. Cloning and characterization of microbial activated Aedes aegypti MEK4 (AaMEK4): influences of noncatalytic domains on enzymatic activity. INSECT MOLECULAR BIOLOGY 2014; 23:644-655. [PMID: 25039995 DOI: 10.1111/imb.12116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Protein kinases are known to be involved in a number of signal transduction cascades. Both the stress-activated Jun N-terminal kinase (JNK) and mitogen-activated protein kinase (MAPK) p38 pathways have been shown to correlate with the insect immune response to microbial infection. MAP kinase kinase 4 (MEK4) is an upstream kinase of JNK and p38 kinase. The cDNA of AaMEK4 was cloned and characterized. AaMEK4 was activated by microbial lysates of Gram-positive, Gram-negative bacteria and yeast. The conserved lysine (K112 ) and the putative phosphorylation sites (S238 and T242 ) were shown to be important for kinase activity by site-directed mutagenesis. A common MAPK docking site (MAPK_dsA) was found and in addition, a new nearby docking site, MAPK_dsB, was identified in the N-terminal noncatalytic domain of AaMEK4. MAPK_dsB was shown to be a unique element in the MEK4 family. In this study, both MAPK_dsA and _dsB were demonstrated to be important to AaMEK4 enzymatic activity for the downstream protein kinase, Aap38.
Collapse
Affiliation(s)
- R C-C Wu
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei City, Taiwan
| | | |
Collapse
|
143
|
Bracken CP, Li X, Wright JA, Lawrence DM, Pillman KA, Salmanidis M, Anderson MA, Dredge BK, Gregory PA, Tsykin A, Neilsen C, Thomson DW, Bert AG, Leerberg JM, Yap AS, Jensen KB, Khew-Goodall Y, Goodall GJ. Genome-wide identification of miR-200 targets reveals a regulatory network controlling cell invasion. EMBO J 2014; 33:2040-56. [PMID: 25069772 PMCID: PMC4195771 DOI: 10.15252/embj.201488641] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 06/06/2014] [Accepted: 06/12/2014] [Indexed: 12/14/2022] Open
Abstract
The microRNAs of the miR-200 family maintain the central characteristics of epithelia and inhibit tumor cell motility and invasiveness. Using the Ago-HITS-CLIP technology for transcriptome-wide identification of direct microRNA targets in living cells, along with extensive validation to verify the reliability of the approach, we have identified hundreds of miR-200a and miR-200b targets, providing insights into general features of miRNA target site selection. Gene ontology analysis revealed a predominant effect of miR-200 targets in widespread coordinate control of actin cytoskeleton dynamics. Functional characterization of the miR-200 targets indicates that they constitute subnetworks that underlie the ability of cancer cells to migrate and invade, including coordinate effects on Rho-ROCK signaling, invadopodia formation, MMP activity, and focal adhesions. Thus, the miR-200 family maintains the central characteristics of the epithelial phenotype by acting on numerous targets at multiple levels, encompassing both cytoskeletal effectors that control actin filament organization and dynamics, and upstream signals that locally regulate the cytoskeleton to maintain cell morphology and prevent cell migration.
Collapse
Affiliation(s)
- Cameron P Bracken
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia Discipline of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Xiaochun Li
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Josephine A Wright
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - David M Lawrence
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Katherine A Pillman
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Marika Salmanidis
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Matthew A Anderson
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - B Kate Dredge
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, Australia
| | - Philip A Gregory
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia Discipline of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Anna Tsykin
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Corine Neilsen
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Daniel W Thomson
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Andrew G Bert
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Joanne M Leerberg
- Division of Molecular Cell Biology, Institute for Molecular Bioscience University of Queensland, St Lucia, Brisbane, Qld, Australia
| | - Alpha S Yap
- Division of Molecular Cell Biology, Institute for Molecular Bioscience University of Queensland, St Lucia, Brisbane, Qld, Australia
| | - Kirk B Jensen
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, Australia
| | - Yeesim Khew-Goodall
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia School of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, Australia
| | - Gregory J Goodall
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia Discipline of Medicine, University of Adelaide, Adelaide, SA, Australia School of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
144
|
Yabu T, Shiba H, Shibasaki Y, Nakanishi T, Imamura S, Touhata K, Yamashita M. Stress-induced ceramide generation and apoptosis via the phosphorylation and activation of nSMase1 by JNK signaling. Cell Death Differ 2014; 22:258-73. [PMID: 25168245 PMCID: PMC4291487 DOI: 10.1038/cdd.2014.128] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 07/14/2014] [Accepted: 07/15/2014] [Indexed: 12/20/2022] Open
Abstract
Neutral sphingomyelinase (nSMase) activation in response to environmental stress or inflammatory cytokine stimuli generates the second messenger ceramide, which mediates the stress-induced apoptosis. However, the signaling pathways and activation mechanism underlying this process have yet to be elucidated. Here we show that the phosphorylation of nSMase1 (sphingomyelin phosphodiesterase 2, SMPD2) by c-Jun N-terminal kinase (JNK) signaling stimulates ceramide generation and apoptosis and provide evidence for a signaling mechanism that integrates stress- and cytokine-activated apoptosis in vertebrate cells. An nSMase1 was identified as a JNK substrate, and the phosphorylation site responsible for its effects on stress and cytokine induction was Ser-270. In zebrafish cells, the substitution of Ser-270 for alanine blocked the phosphorylation and activation of nSMase1, whereas the substitution of Ser-270 for negatively charged glutamic acid mimicked the effect of phosphorylation. The JNK inhibitor SP600125 blocked the phosphorylation and activation of nSMase1, which in turn blocked ceramide signaling and apoptosis. A variety of stress conditions, including heat shock, UV exposure, hydrogen peroxide treatment, and anti-Fas antibody stimulation, led to the phosphorylation of nSMase1, activated nSMase1, and induced ceramide generation and apoptosis in zebrafish embryonic ZE and human Jurkat T cells. In addition, the depletion of MAPK8/9 or SMPD2 by RNAi knockdown decreased ceramide generation and stress- and cytokine-induced apoptosis in Jurkat cells. Therefore the phosphorylation of nSMase1 is a pivotal step in JNK signaling, which leads to ceramide generation and apoptosis under stress conditions and in response to cytokine stimulation. nSMase1 has a common central role in ceramide signaling during the stress and cytokine responses and apoptosis.
Collapse
Affiliation(s)
- T Yabu
- Nihon University, College of Bioresource Sciences, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - H Shiba
- Nihon University, College of Bioresource Sciences, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - Y Shibasaki
- Nihon University, College of Bioresource Sciences, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - T Nakanishi
- Nihon University, College of Bioresource Sciences, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - S Imamura
- Food Safety Assessment Research Group, National Research Institute of Fisheries Science, 12-4 Fukuura 2, Kanazawa-ku, Yokohama, Kanagawa 236-8648, Japan
| | - K Touhata
- Food Safety Assessment Research Group, National Research Institute of Fisheries Science, 12-4 Fukuura 2, Kanazawa-ku, Yokohama, Kanagawa 236-8648, Japan
| | - M Yamashita
- Food Safety Assessment Research Group, National Research Institute of Fisheries Science, 12-4 Fukuura 2, Kanazawa-ku, Yokohama, Kanagawa 236-8648, Japan
| |
Collapse
|
145
|
Pahl S, Tapken D, Haering SC, Hollmann M. Trafficking of kainate receptors. MEMBRANES 2014; 4:565-95. [PMID: 25141211 PMCID: PMC4194049 DOI: 10.3390/membranes4030565] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Revised: 08/04/2014] [Accepted: 08/12/2014] [Indexed: 11/17/2022]
Abstract
Ionotropic glutamate receptors (iGluRs) mediate the vast majority of excitatory neurotransmission in the central nervous system of vertebrates. In the protein family of iGluRs, kainate receptors (KARs) comprise the probably least well understood receptor class. Although KARs act as key players in the regulation of synaptic network activity, many properties and functions of these proteins remain elusive until now. Especially the precise pre-, extra-, and postsynaptic localization of KARs plays a critical role for neuronal function, as an unbalanced localization of KARs would ultimately lead to dysregulated neuronal excitability. Recently, important advances in the understanding of the regulation of surface expression, function, and agonist-dependent endocytosis of KARs have been achieved. Post-translational modifications like PKC-mediated phosphorylation and SUMOylation have been reported to critically influence surface expression and endocytosis, while newly discovered auxiliary proteins were shown to shape the functional properties of KARs.
Collapse
Affiliation(s)
- Steffen Pahl
- Department of Biochemistry I, Ruhr University Bochum, Universitätsstr. 150, 44780 Bochum, Germany.
| | - Daniel Tapken
- Department of Biochemistry I, Ruhr University Bochum, Universitätsstr. 150, 44780 Bochum, Germany.
| | - Simon C Haering
- Department of Biochemistry I, Ruhr University Bochum, Universitätsstr. 150, 44780 Bochum, Germany.
| | - Michael Hollmann
- Department of Biochemistry I, Ruhr University Bochum, Universitätsstr. 150, 44780 Bochum, Germany.
| |
Collapse
|
146
|
Abstract
In a recent Cell paper, Kitambi and colleagues identify a small molecule (Vacquinol-1) that has beneficial effects on a glioblastoma multiforme mouse model by oral administration. In glioblastoma cells, Vacquinol-1 targets macropinocytosis, a cellular process that will not lead to cell death in normal cells.
Collapse
Affiliation(s)
- Yufeng Shi
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - S Kyun Lim
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Luis F Parada
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
147
|
Jarvis I, Bergvall C, Morales D, Kummrow F, Umbuzeiro G, Westerholm R, Stenius U, Dreij K. Nanomolar levels of PAHs in extracts from urban air induce MAPK signaling in HepG2 cells. Toxicol Lett 2014; 229:25-32. [DOI: 10.1016/j.toxlet.2014.06.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 06/04/2014] [Accepted: 06/05/2014] [Indexed: 12/13/2022]
|
148
|
Zaidi SK, Shen WJ, Bittner S, Bittner A, McLean MP, Han J, Davis RJ, Kraemer FB, Azhar S. p38 MAPK regulates steroidogenesis through transcriptional repression of STAR gene. J Mol Endocrinol 2014; 53:1-16. [PMID: 24780837 PMCID: PMC4077990 DOI: 10.1530/jme-13-0287] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
STAR/StarD1, part of a protein complex, mediates the transport of cholesterol from the outer to inner mitochondrial membrane, which is the rate-limiting step for steroidogenesis, and where steroid hormone synthesis begins. Herein, we examined the role of oxidant-sensitive p38 MAPKs in the regulation of STAR gene transcription, using model steroidogenic cell lines. Our data indicate that oxidant activation of p38 MAPK exhibits a negative regulatory role in the induction of functional expression of STAR, as evidenced by enhanced induction of STAR (mRNA/protein) expression and increased steroidogenesis during pharmacological inhibition of p38 MAPK or in cells with increased transient overexpression of a dominant-negative (dn) form of p38 MAPKα or p38 MAPKβ. Studies with rat Star-promoter demonstrated that overexpression of p38 MAPKα-wt, -β, or -γ significantly reduced both basal and cAMP-sensitive promoter activity. In contrast, overexpression of p38 MAPKα-dn, -β, or -γ enhanced the Star promoter activity under basal conditions and in response to cAMP stimulation. Use of various constitutively active and dn constructs and designer knock-out cell lines demonstrated that MKK3 and MKK6, the upstream activators of p38 MAPKs, play a role in p38 MAPKα-mediated inhibition of Star promoter activity. In addition, our studies raised the possibility of CREB being a potential target of the p38 MAPK inhibitory effect on Star promoter activity. Collectively, these data provide novel mechanistic information about how oxidant-sensitive p38 MAPKs, particularly p38 MAPKα, contribute to the negative regulation of Star gene expression and inhibit steroidogenesis.
Collapse
Affiliation(s)
- Syed Kashif Zaidi
- Geriatric ResearchEducation and Clinical Center (GRECC-182B), VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, California 94304, USADivision of EndocrinologyDivision of Gastroenterology and HepatologyStanford University, Stanford, California 94305, USADepartment of Obstetrics and GynecologyUniversity of South Florida College of Medicine, Tampa, Florida 33612, USAState Key Laboratory of Cellular Stress BiologySchool of Life Sciences, Xiamen University, Xiamen, Fujian 361005, ChinaProgram in Molecular MedicineUniversity of Massachusetts Medical School, Worcester, Massachusetts 01605, USAGeriatric ResearchEducation and Clinical Center (GRECC-182B), VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, California 94304, USADivision of EndocrinologyDivision of Gastroenterology and HepatologyStanford University, Stanford, California 94305, USADepartment of Obstetrics and GynecologyUniversity of South Florida College of Medicine, Tampa, Florida 33612, USAState Key Laboratory of Cellular Stress BiologySchool of Life Sciences, Xiamen University, Xiamen, Fujian 361005, ChinaProgram in Molecular MedicineUniversity of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Wen-Jun Shen
- Geriatric ResearchEducation and Clinical Center (GRECC-182B), VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, California 94304, USADivision of EndocrinologyDivision of Gastroenterology and HepatologyStanford University, Stanford, California 94305, USADepartment of Obstetrics and GynecologyUniversity of South Florida College of Medicine, Tampa, Florida 33612, USAState Key Laboratory of Cellular Stress BiologySchool of Life Sciences, Xiamen University, Xiamen, Fujian 361005, ChinaProgram in Molecular MedicineUniversity of Massachusetts Medical School, Worcester, Massachusetts 01605, USAGeriatric ResearchEducation and Clinical Center (GRECC-182B), VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, California 94304, USADivision of EndocrinologyDivision of Gastroenterology and HepatologyStanford University, Stanford, California 94305, USADepartment of Obstetrics and GynecologyUniversity of South Florida College of Medicine, Tampa, Florida 33612, USAState Key Laboratory of Cellular Stress BiologySchool of Life Sciences, Xiamen University, Xiamen, Fujian 361005, ChinaProgram in Molecular MedicineUniversity of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Stefanie Bittner
- Geriatric ResearchEducation and Clinical Center (GRECC-182B), VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, California 94304, USADivision of EndocrinologyDivision of Gastroenterology and HepatologyStanford University, Stanford, California 94305, USADepartment of Obstetrics and GynecologyUniversity of South Florida College of Medicine, Tampa, Florida 33612, USAState Key Laboratory of Cellular Stress BiologySchool of Life Sciences, Xiamen University, Xiamen, Fujian 361005, ChinaProgram in Molecular MedicineUniversity of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Alex Bittner
- Geriatric ResearchEducation and Clinical Center (GRECC-182B), VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, California 94304, USADivision of EndocrinologyDivision of Gastroenterology and HepatologyStanford University, Stanford, California 94305, USADepartment of Obstetrics and GynecologyUniversity of South Florida College of Medicine, Tampa, Florida 33612, USAState Key Laboratory of Cellular Stress BiologySchool of Life Sciences, Xiamen University, Xiamen, Fujian 361005, ChinaProgram in Molecular MedicineUniversity of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Mark P McLean
- Geriatric ResearchEducation and Clinical Center (GRECC-182B), VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, California 94304, USADivision of EndocrinologyDivision of Gastroenterology and HepatologyStanford University, Stanford, California 94305, USADepartment of Obstetrics and GynecologyUniversity of South Florida College of Medicine, Tampa, Florida 33612, USAState Key Laboratory of Cellular Stress BiologySchool of Life Sciences, Xiamen University, Xiamen, Fujian 361005, ChinaProgram in Molecular MedicineUniversity of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Jiahuai Han
- Geriatric ResearchEducation and Clinical Center (GRECC-182B), VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, California 94304, USADivision of EndocrinologyDivision of Gastroenterology and HepatologyStanford University, Stanford, California 94305, USADepartment of Obstetrics and GynecologyUniversity of South Florida College of Medicine, Tampa, Florida 33612, USAState Key Laboratory of Cellular Stress BiologySchool of Life Sciences, Xiamen University, Xiamen, Fujian 361005, ChinaProgram in Molecular MedicineUniversity of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Roger J Davis
- Geriatric ResearchEducation and Clinical Center (GRECC-182B), VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, California 94304, USADivision of EndocrinologyDivision of Gastroenterology and HepatologyStanford University, Stanford, California 94305, USADepartment of Obstetrics and GynecologyUniversity of South Florida College of Medicine, Tampa, Florida 33612, USAState Key Laboratory of Cellular Stress BiologySchool of Life Sciences, Xiamen University, Xiamen, Fujian 361005, ChinaProgram in Molecular MedicineUniversity of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Fredric B Kraemer
- Geriatric ResearchEducation and Clinical Center (GRECC-182B), VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, California 94304, USADivision of EndocrinologyDivision of Gastroenterology and HepatologyStanford University, Stanford, California 94305, USADepartment of Obstetrics and GynecologyUniversity of South Florida College of Medicine, Tampa, Florida 33612, USAState Key Laboratory of Cellular Stress BiologySchool of Life Sciences, Xiamen University, Xiamen, Fujian 361005, ChinaProgram in Molecular MedicineUniversity of Massachusetts Medical School, Worcester, Massachusetts 01605, USAGeriatric ResearchEducation and Clinical Center (GRECC-182B), VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, California 94304, USADivision of EndocrinologyDivision of Gastroenterology and HepatologyStanford University, Stanford, California 94305, USADepartment of Obstetrics and GynecologyUniversity of South Florida College of Medicine, Tampa, Florida 33612, USAState Key Laboratory of Cellular Stress BiologySchool of Life Sciences, Xiamen University, Xiamen, Fujian 361005, ChinaProgram in Molecular MedicineUniversity of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Salman Azhar
- Geriatric ResearchEducation and Clinical Center (GRECC-182B), VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, California 94304, USADivision of EndocrinologyDivision of Gastroenterology and HepatologyStanford University, Stanford, California 94305, USADepartment of Obstetrics and GynecologyUniversity of South Florida College of Medicine, Tampa, Florida 33612, USAState Key Laboratory of Cellular Stress BiologySchool of Life Sciences, Xiamen University, Xiamen, Fujian 361005, ChinaProgram in Molecular MedicineUniversity of Massachusetts Medical School, Worcester, Massachusetts 01605, USAGeriatric ResearchEducation and Clinical Center (GRECC-182B), VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, California 94304, USADivision of EndocrinologyDivision of Gastroenterology and HepatologyStanford University, Stanford, California 94305, USADepartment of Obstetrics and GynecologyUniversity of South Florida College of Medicine, Tampa, Florida 33612, USAState Key Laboratory of Cellular Stress BiologySchool of Life Sciences, Xiamen University, Xiamen, Fujian 361005, ChinaProgram in Molecular MedicineUniversity of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| |
Collapse
|
149
|
Chaudhari N, Talwar P, Parimisetty A, Lefebvre d'Hellencourt C, Ravanan P. A molecular web: endoplasmic reticulum stress, inflammation, and oxidative stress. Front Cell Neurosci 2014; 8:213. [PMID: 25120434 PMCID: PMC4114208 DOI: 10.3389/fncel.2014.00213] [Citation(s) in RCA: 460] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 07/15/2014] [Indexed: 12/13/2022] Open
Abstract
Execution of fundamental cellular functions demands regulated protein folding homeostasis. Endoplasmic reticulum (ER) is an active organelle existing to implement this function by folding and modifying secretory and membrane proteins. Loss of protein folding homeostasis is central to various diseases and budding evidences suggest ER stress as being a major contributor in the development or pathology of a diseased state besides other cellular stresses. The trigger for diseases may be diverse but, inflammation and/or ER stress may be basic mechanisms increasing the severity or complicating the condition of the disease. Chronic ER stress and activation of the unfolded-protein response (UPR) through endogenous or exogenous insults may result in impaired calcium and redox homeostasis, oxidative stress via protein overload thereby also influencing vital mitochondrial functions. Calcium released from the ER augments the production of mitochondrial Reactive Oxygen Species (ROS). Toxic accumulation of ROS within ER and mitochondria disturbs fundamental organelle functions. Sustained ER stress is known to potentially elicit inflammatory responses via UPR pathways. Additionally, ROS generated through inflammation or mitochondrial dysfunction could accelerate ER malfunction. Dysfunctional UPR pathways have been associated with a wide range of diseases including several neurodegenerative diseases, stroke, metabolic disorders, cancer, inflammatory disease, diabetes mellitus, cardiovascular disease, and others. In this review, we have discussed the UPR signaling pathways, and networking between ER stress-induced inflammatory pathways, oxidative stress, and mitochondrial signaling events, which further induce or exacerbate ER stress.
Collapse
Affiliation(s)
- Namrata Chaudhari
- Apoptosis and Cell Death Research Lab, School of Biosciences and Technology, Vellore Institute of Technology University , Vellore , India
| | - Priti Talwar
- Apoptosis and Cell Death Research Lab, School of Biosciences and Technology, Vellore Institute of Technology University , Vellore , India
| | - Avinash Parimisetty
- Groupe d'Etude sur l'Inflammation Chronique et l'Obésité, EA 41516, Plateforme CYROI, Université de La Réunion , Saint Denis de La Réunion , France
| | - Christian Lefebvre d'Hellencourt
- Groupe d'Etude sur l'Inflammation Chronique et l'Obésité, EA 41516, Plateforme CYROI, Université de La Réunion , Saint Denis de La Réunion , France
| | - Palaniyandi Ravanan
- Apoptosis and Cell Death Research Lab, School of Biosciences and Technology, Vellore Institute of Technology University , Vellore , India
| |
Collapse
|
150
|
Abstract
c-Jun NH2-terminal kinase (JNK) was discovered almost 20 years ago as the protein kinase responsible for phosphorylating c-Jun at Ser-63 and Ser-73. These sites had previously been demonstrated to be essential for the stimulation of c-Jun activity and for cooperation with Ha-ras in oncogenic transformation. This led to the idea that JNK was a positive regulator of cellular transformation. However, the analysis of jnk gene deletion in various mouse models of cancer has produced conflicting findings, with some studies supporting the pro-oncogenic function of JNK and others providing evidence that JNK acts as a tumor suppressor. This review will discuss how these unexpected findings have increased our understanding of the role of JNK signaling in cancer and have provided a source of new working hypotheses.
Collapse
|