Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters











Publication year range
1.
J Cell Biochem ; 122(8): 835-850, 2021 08.
Article in English | MEDLINE | ID: mdl-33876843

ABSTRACT

Triple-negative breast cancer (TNBC) presents a clinical challenge due to the aggressive nature of the disease and a lack of targeted therapies. Constitutive activation of the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway has been linked to chemoresistance and metastatic progression through distinct mechanisms, including activation of epithelial-to-mesenchymal transition (EMT) when cells adopt a motile and invasive phenotype through loss of epithelial markers (CDH1), and acquisition of mesenchymal markers (VIM, CDH2). Although MAPK/ERK1/2 kinase inhibitors (MEKi) are useful antitumor agents in a clinical setting, including the Food and Drug Administration (FDA)-approved MEK1,2 dual inhibitors cobimetinib and trametinib, there are limitations to their clinical utility, primarily adaptation of the BRAF pathway and ocular toxicities. The MEK5 (HGNC: MAP2K5) pathway has important roles in metastatic progression of various cancer types, including those of the prostate, colon, bone and breast, and elevated levels of ERK5 expression in breast carcinomas are linked to a worse prognoses in TNBC patients. The purpose of this study is to explore MEK5 regulation of the EMT axis and to evaluate a novel pan-MEK inhibitor on clinically aggressive TNBC cells. Our results show a distinction between the MEK1/2 and MEK5 cascades in maintenance of the mesenchymal phenotype, suggesting that the MEK5 pathway may be necessary and sufficient in EMT regulation while MEK1/2 signaling further sustains the mesenchymal state of TNBC cells. Furthermore, additive effects on MET induction are evident through the inhibition of both MEK1/2 and MEK5. Taken together, these data demonstrate the need for a better understanding of the individual roles of MEK1/2 and MEK5 signaling in breast cancer and provide a rationale for the combined targeting of these pathways to circumvent compensatory signaling and subsequent therapeutic resistance.


Subject(s)
Cell Movement , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , MAP Kinase Kinase 5/metabolism , MAP Kinase Signaling System , Proto-Oncogene Proteins c-fos/biosynthesis , Triple Negative Breast Neoplasms/metabolism , Female , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/genetics , MAP Kinase Kinase 5/antagonists & inhibitors , MAP Kinase Kinase 5/genetics , MCF-7 Cells , Proto-Oncogene Proteins c-fos/genetics , Triple Negative Breast Neoplasms/genetics
2.
Bioorg Med Chem Lett ; 30(23): 127552, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32946922

ABSTRACT

The importance of ERK5 kinase signaling in tumorigenicity, metastasis, and drug resistance of cancer stem cells (CSCs) has been recognized recently, and we report a unique dual inhibitor that blocks binding of the ERK5 activator and ERK5 autophosphorylation simultaneously. The conventional ATP-binding site inhibitors have not yet yielded expected level of anti-cancer effects, due to complexities in converting ERK5 activation into CSC biological effects. We designed the first ERK5-targeted anti-CSC dual active hetero-bivalent inhibitor that blocks the regulatory peptide interaction involved in ERK5 kinase activation and that simultaneously inhibits the conventional ATP-binding pocket as well. We utilized two assay systems to independently prove disruption of these two ERK5 activities via a single compound. We also showed that this compound inhibited CSC activities, such as colony formation, cell proliferation, and migration.


Subject(s)
Antineoplastic Agents/pharmacology , MAP Kinase Kinase 5/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Neoplastic Stem Cells/drug effects , Peptides/pharmacology , Protein Kinase Inhibitors/pharmacology , Amino Acid Sequence , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Design , Drug Screening Assays, Antitumor , Humans , MAP Kinase Kinase 5/chemistry , Mitogen-Activated Protein Kinase 7/chemistry , Mitogen-Activated Protein Kinase 7/metabolism , Molecular Docking Simulation , Peptides/chemical synthesis , Peptides/metabolism , Phosphorylation/drug effects , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Protein Multimerization/drug effects , Signal Transduction/drug effects
3.
Oncogene ; 39(12): 2467-2477, 2020 03.
Article in English | MEDLINE | ID: mdl-31980741

ABSTRACT

Radiotherapy is commonly used to treat a variety of solid human tumors, including localized prostate cancer. However, treatment failure often ensues due to tumor intrinsic or acquired radioresistance. Here we find that the MEK5/ERK5 signaling pathway is associated with resistance to genotoxic stress in aggressive prostate cancer cells. MEK5 knockdown by RNA interference sensitizes prostate cancer cells to ionizing radiation (IR) and etoposide treatment, as assessed by clonogenic survival and short-term proliferation assays. Mechanistically, MEK5 downregulation impairs phosphorylation of the catalytic subunit of DNA-PK at serine 2056 in response to IR or etoposide treatment. Although MEK5 knockdown does not influence the initial appearance of radiation- and etoposide-induced γH2AX and 53BP1 foci, it markedly delays their resolution, indicating a DNA repair defect. A cell-based assay shows that nonhomologous end joining (NHEJ) is compromised in cells with ablated MEK5 protein expression. Finally, MEK5 silencing combined with focal irradiation causes strong inhibition of tumor growth in mouse xenografts, compared with MEK5 depletion or radiation alone. These findings reveal a convergence between MEK5 signaling and DNA repair by NHEJ in conferring resistance to genotoxic stress in advanced prostate cancer and suggest targeting MEK5 as an effective therapeutic intervention in the management of this disease.


Subject(s)
Antineoplastic Agents/pharmacology , DNA End-Joining Repair , DNA, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , MAP Kinase Kinase 5/genetics , Mutagens/pharmacology , Prostatic Neoplasms/drug therapy , Animals , Cell Cycle/radiation effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , DNA End-Joining Repair/drug effects , Drug Delivery Systems , Gene Knockdown Techniques , Humans , MAP Kinase Kinase 5/antagonists & inhibitors , MAP Kinase Kinase 5/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/radiation effects , Male , Mice , Mitogen-Activated Protein Kinase 7/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/radiotherapy , Xenograft Model Antitumor Assays
4.
J Cell Biochem ; 121(2): 1156-1168, 2020 02.
Article in English | MEDLINE | ID: mdl-31464004

ABSTRACT

Triple-negative breast cancers (TNBCs) represent 15% to 20% of all breast cancers and are often associated with poor prognosis. The lack of targeted therapies for TNBCs contributes to higher mortality rates. Aberrations in the phosphoinositide-3-kinase (PI3K) and mitogen-activated protein kinase pathways have been linked to increased breast cancer proliferation and survival. It has been proposed that these survival characteristics are enhanced through compensatory signaling and crosstalk mechanisms. While the crosstalk between PI3K and extracellular signal-regulated kinase 1/2 (ERK1/2) pathways has been characterized in several systems, new evidence suggests that MEK5/ERK5 signaling is a key component in the proliferation and survival of several aggressive cancers. In this study, we examined the effects of dual inhibition of PI3K/protein kinase B (Akt) and MEK5/ERK5 in the MDA-MB-231, BT-549, and MDA-MB-468 TNBC cell lines. We used the Akt inhibitor ipatasertib, ERK5 inhibitors XMD8-92 and AX15836, and the novel MEK5 inhibitor SC-1-181 to investigate the effects of dual inhibition. Our results indicated that dual inhibition of PI3K/Akt and MEK5/ERK5 signaling was more effective at reducing the proliferation and survival of TNBCs than single inhibition of either pathway alone. In particular, a loss of Bad phosphorylation at two distinct sites was observed with dual inhibition. Furthermore, the inhibition of both pathways led to p21 restoration, decreased cell proliferation, and induced apoptosis. In addition, the dual inhibition strategy was determined to be synergistic in MDA-MB-231 and BT-549 cells and was relatively nontoxic in the nonneoplastic MCF-10 cell line. In summary, the results from this study provide a unique prospective into the utility of a novel dual inhibition strategy for targeting TNBCs.


Subject(s)
Cell Survival/drug effects , MAP Kinase Kinase 5/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/metabolism , Apoptosis/drug effects , Benzodiazepinones/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Synergism , Female , Humans , MAP Kinase Kinase 5/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyridones/pharmacology , Pyrimidines/pharmacology , Pyrimidinones/pharmacology
5.
Biochem Biophys Res Commun ; 513(4): 925-932, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31005259

ABSTRACT

The most prominent hallmarks of many neurodegenerative diseases are the accumulation of misfolded protein aggregates and the death of certain neuronal populations. Autophagy is the major intracellular mechanism that degrades protein aggregates and damaged cellular components. Many studies have reported that the dysfunction of autophagy is associated with several neurodegenerative diseases, such as Alzheimer's disease, amyotrophic lateral sclerosis (ALS), and Parkinson's disease. Here, we identified a novel mechanism of autophagy regulation. Inhibition of MEK5 reduced the level of p62 and increased the ratio of LC3-II to LC3-I, which is a marker for the activation of the autophagy-lysosome pathway (ALP). One of the most well-known regulators of the ALP is mTOR, and previous studies have reported that the major substrate of MEK5 is ERK5. However, we found that MEK5 modulates the autophagy-lysosome pathway in an mTOR- and ERK5-independent manner. Moreover, MEK5 inhibition alleviated the mislocalization of TDP-43 (an ALS-associated protein) and cell death in TDP-43-GFP-expressing neuronal cells. Taken together, these findings suggest that MEK5 is a novel autophagy modulator and that this kinase could be a therapeutic target for neurodegenerative diseases such as amyotrophic lateral sclerosis.


Subject(s)
Autophagy , DNA-Binding Proteins/toxicity , Lysosomes/metabolism , MAP Kinase Kinase 5/antagonists & inhibitors , Metabolic Networks and Pathways/physiology , Neurons/cytology , Animals , Humans , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , RNA-Binding Proteins/metabolism , TOR Serine-Threonine Kinases/physiology
7.
J Mol Neurosci ; 61(4): 607-616, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28265860

ABSTRACT

Neuregulin1ß (NRG1ß), a member of the excitomotor of tyrosine kinase receptor (erbB) family, was recently shown to play a neuroprotective role in cerebral ischemia-reperfusion injury. The present study analyzed the effects and its possible signaling pathway of NRG1ß on brain tissues after cerebral ischemia-reperfusion injury. A focal cerebral ischemic model was established by inserting a monofilament thread to achieve middle cerebral artery occlusion, followed by an NRG1ß injection via the internal carotid artery. NRG1ß injection resulted in significantly improved neurobehavioral activity according to the modified neurological severity score test. Tetrazolium chloridestaining revealed a smaller cerebral infarction volume; hematoxylin-eosin staining and transmission electron microscopy showed significantly alleviated neurodegeneration in the middle cerebral artery occlusion rats. Moreover, expression of phosphorylated MEK5, phosphorylated ERK5, and phosphorylated MEK2C increased after NRG1ß treatment, and the neuroprotective effect of NRG1ß was attenuated by an injection of the MEK5 inhibitor, BIX02189. Results from the present study demonstrate that NRG1ß provides neuroprotection following cerebral ischemia-reperfusion injury via the ERK5-dependent MAPK pathway.


Subject(s)
Infarction, Middle Cerebral Artery/drug therapy , MAP Kinase Signaling System , Neuregulin-1/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Brain/drug effects , Brain/metabolism , Infarction, Middle Cerebral Artery/metabolism , MAP Kinase Kinase 5/antagonists & inhibitors , MAP Kinase Kinase 5/metabolism , Male , Mitogen-Activated Protein Kinase 7/metabolism , Neuregulin-1/administration & dosage , Neuregulin-1/pharmacology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar
8.
Cancer Lett ; 392: 51-59, 2017 04 28.
Article in English | MEDLINE | ID: mdl-28153789

ABSTRACT

Mitogen-activated protein kinases (MAPKs) regulate diverse cellular processes including proliferation, cell survival, differentiation, and apoptosis. While conventional MAPK constituents have well-defined roles in oncogenesis, the MEK5 pathway has only recently emerged in cancer research. In this review, we consider the MEK5 signaling cascade, focusing specifically on its involvement in drug resistance and regulation of aggressive cancer phenotypes. Moreover, we explore the role of MEK5/ERK5 in tumorigenesis and metastatic progression, discussing the discrepancies in preclinical studies and assessing its viability as a therapeutic target for anti-cancer agents.


Subject(s)
MAP Kinase Kinase 5/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Animals , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Differentiation , Cell Proliferation , Drug Resistance, Neoplasm , Humans , MAP Kinase Kinase 5/antagonists & inhibitors , MAP Kinase Kinase 5/genetics , Molecular Targeted Therapy , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins/genetics , Signal Transduction/drug effects
9.
Oncol Rep ; 37(1): 83-90, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27878304

ABSTRACT

Mitogen extracellular-signal-regulated kinase kinase 5 (MEK5) plays an important role in promoting cell proliferation and tumorigenesis. The aberrant expression of MEK5 has been reported in various malignant diseases including cancers of breast, prostate, lung, colorectal and brain. However, the function and regulation of MEK5 signaling pathway are ambiguous and remain elusive with respect to its oncogenic roles in various cancers, especially in the regulation of the initiation and progression of cancer invasion and metastasis. Ectopic expression of MEK5 or knockdown of MEK5 by shRNA with in vitro cell based models demonstrated the role of MEK5 in regulation of epithelial mesenchymal transition (EMT) and breast cancer invasion and metastasis. Here, we show that MEK5 upregulated by Stat3 promotes breast cancer cell invasion through EMT. Further study demonstrated that Stat3 could bind to promoter region of MEK5 and enhanced MEK5 transcription and expression. In addition, the phosphorylation of MEK5 significantly increased in breast cancer cells corresponding to metastatic capability of breast cancer cells. The depletion of MEK5 by shRNA significantly decreased breast cancer invasion. Ectopic expression of MEK5 could confer non-invasive breast cancer cells to become invasion capable cells. Moreover, the phosphorylation of Erk5, a MEK5-regulated downstream kinase, was also upregulated consistent with the increased level of active MEK5. Our studies provide insights into a molecular mechanism by which MEK5 transcriptionally upregulated by Stat3 augments breast cancer cell EMT, which subsequently enhances cancer cell invasion and metastasis. This finding may suggest that Stat3 and MEK5/Erk5 pathways could be an effective therapeutic target for inhibition of breast cancer invasion and metastasis.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Movement/genetics , MAP Kinase Kinase 5/genetics , STAT3 Transcription Factor/physiology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MAP Kinase Kinase 5/antagonists & inhibitors , MAP Kinase Kinase 5/metabolism , MCF-7 Cells , Neoplasm Invasiveness , Neoplasm Metastasis , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Up-Regulation/drug effects
10.
Cancer Lett ; 381(2): 314-22, 2016 10 28.
Article in English | MEDLINE | ID: mdl-27543359

ABSTRACT

Transforming growth factor-ß1 (TGF-ß1) promotes tumor metastasis by inducing an epithelial-to-mesenchymal transition (EMT) in cancer cells. In this study, we investigated the effects of BIX02189 and XMD8-92, pharmacologic inhibitors of the MEK5 [mitogen-activated protein kinase/extracellular-signal-regulated kinase (ERK)5] signaling pathway, on the EMT and migration of cancer cells induced by TGF-ß1. In human A549 lung cancer cells, TGF-ß1-induced EMT, cell motility, and expression of matrix metalloproteinase-2 were completely inhibited by BIX02189, but not by XMD8-92 or small interference RNAs specific to MEK5 and ERK5. Interestingly, BIX02189 strongly blocked the activation of TGF-ß1 signaling components, and this inhibitory effect was not reproduced by MEK5 inhibition. Molecular docking simulation and kinase assays revealed that BIX02189 binds directly to the ATP-binding site of the TGF-ß receptor type I (TßRI) and suppresses its kinase activity. Finally, the anti-metastatic effect of BIX02189 was validated in a TßRI-derived A549 xenograft mouse model. Collectively, these findings newly characterize BIX02189 as a potent inhibitor of TßRI that can block the tumor metastatic activity of TGF-ß1.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Movement/drug effects , Indoles/pharmacology , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta1/pharmacology , A549 Cells , Adenosine Triphosphate/metabolism , Aniline Compounds/metabolism , Animals , Antineoplastic Agents/metabolism , Binding Sites , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/secondary , Dose-Response Relationship, Drug , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Indoles/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MAP Kinase Kinase 5/antagonists & inhibitors , MAP Kinase Kinase 5/genetics , MAP Kinase Kinase 5/metabolism , Mice, Inbred BALB C , Mice, Nude , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/genetics , Mitogen-Activated Protein Kinase 7/metabolism , Molecular Docking Simulation , Neoplasm Invasiveness , Protein Binding , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Transfection , Xenograft Model Antitumor Assays
11.
Oncotarget ; 7(23): 34322-40, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27144434

ABSTRACT

The MEK5/ERK5 signaling pathway is emerging as an important contributor to colon cancer onset, progression and metastasis; however, its relevance to chemotherapy resistance remains unknown. Here, we evaluated the impact of the MEK5/ERK5 cascade in colon cancer cell sensitivity to 5-fluorouracil (5-FU). Increased ERK5 expression was correlated with poor overall survival in colon cancer patients. In colon cancer cells, 5-FU exposure impaired endogenous KRAS/MEK5/ERK5 expression and/or activation. In turn, MEK5 constitutive activation reduced 5-FU-induced cytotoxicity. Using genetic and pharmacological approaches, we showed that ERK5 inhibition increased caspase-3/7 activity and apoptosis following 5-FU exposure. Mechanistically, this was further associated with increased p53 transcriptional activation of p21 and PUMA. In addition, ERK5 inhibition increased the response of HCT116 p53+/+ cells to 5-FU, but failed to sensitize HCT116 p53-/- cells to the cytotoxic effects of this chemotherapeutic agent, suggesting a p53-dependent axis mediating 5-FU sensitization. Finally, ERK5 inhibition using XMD8-92 was shown to increase the antitumor effects of 5-FU in a murine subcutaneous xenograft model, enhancing apoptosis while markedly reducing tumor growth. Collectively, our results suggest that ERK5-targeted inhibition provides a promising therapeutic approach to overcome resistance to 5-FU-based chemotherapy and improve colon cancer treatment.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Colonic Neoplasms/drug therapy , Fluorouracil/pharmacology , MAP Kinase Kinase 5/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Benzodiazepinones/pharmacology , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/genetics , Enzyme Activation/drug effects , HCT116 Cells , Humans , MAP Kinase Kinase 5/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred BALB C , Mice, SCID , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
12.
Biochem Biophys Res Commun ; 463(3): 241-7, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-25998381

ABSTRACT

Extracellular signal-regulated kinase 5 (ERK5) is a member of the mitogen-activated protein kinase (MAPK) family and is activated by its upstream kinase, MAPK kinase 5 (MEK5), which is a member of the MEK family. Although the role of MEK5 has been investigated in several fields, little is known about its role in osteoblastic differentiation. In this study, we have demonstrated the role of MEK5 in osteoblastic differentiation in mouse preosteoblastic MC3T3-E1 cells and bone marrow stromal ST2 cells. We found that treatment with BIX02189, an inhibitor of MEK5, increased alkaline phosphatase (ALP) activity and the gene expression of ALP, osteocalcin (OCN) and osterix, as well as it enhanced the calcification of the extracellular matrix. Moreover, osteoblastic cell proliferation decreased at a concentration of greater than 0.5 µM. In addition, knockdown of MEK5 using siRNA induced an increase in ALP activity and in the gene expression of ALP, OCN, and osterix. In contrast, overexpression of wild-type MEK5 decreased ALP activity and attenuated osteoblastic differentiation markers including ALP, OCN and osterix, but promoted cell proliferation. In summary, our results indicated that MEK5 suppressed the osteoblastic differentiation, but promoted osteoblastic cell proliferation. These results implied that MEK5 may play a pivotal role in cell signaling to modulate the differentiation and proliferation of osteoblasts. Thus, inhibition of MEK5 signaling in osteoblasts may be of potential use in the treatment of osteoporosis.


Subject(s)
Cell Differentiation , MAP Kinase Kinase 5/metabolism , Osteoblasts/cytology , Aniline Compounds/pharmacology , Animals , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Indoles/pharmacology , MAP Kinase Kinase 5/antagonists & inhibitors , MAP Kinase Kinase 5/genetics , Mice , Mitogen-Activated Protein Kinase 7/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Phosphorylation/drug effects , RNA Interference , Up-Regulation
13.
PLoS One ; 10(4): e0125054, 2015.
Article in English | MEDLINE | ID: mdl-25885811

ABSTRACT

The MEK/ERK pathways are critical for controlling cell proliferation and differentiation. In this study, we show that the MEK5/ERK5 pathway participates in osteoclast differentiation. ERK5 was activated by M-CSF, which is one of the essential factors in osteoclast differentiation. Inhibition of MEK5 by BIX02189 or inhibition of ERK5 by XMD 8-92 blocked osteoclast differentiation. MEK5 knockdown inhibited osteoclast differentiation. RAW264.7D clone cells, which are monocytic cells, differentiate into osteoclasts after stimulation with sRANKL. ERK5 was activated without any stimulation in these cells. Inhibition of the MEK5/ERK5 pathway by the inhibitors also blocked the differentiation of RAW264.7D cells into osteoclasts. Moreover, expression of the transcription factor c-Fos, which is indispensable for osteoclast differentiation, was inhibited by treatment with MEK5 or ERK5 inhibitors. Therefore, activation of ERK5 is required for the induction of c-Fos. These events were confirmed in experiments using M-CSF-dependent bone marrow macrophages. Taken together, the present results show that activation of the MEK5/ERK5 pathway with M-CSF is required for osteoclast differentiation, which may induce differentiation through the induction of c-Fos.


Subject(s)
Mitogen-Activated Protein Kinase 7/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Acid Phosphatase/metabolism , Aniline Compounds/pharmacology , Animals , Benzodiazepinones/pharmacology , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cell Line/drug effects , Enzyme Activation/drug effects , Humans , Indoles/pharmacology , Isoenzymes/metabolism , MAP Kinase Kinase 5/antagonists & inhibitors , MAP Kinase Kinase 5/genetics , MAP Kinase Kinase 5/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Male , Mice, Inbred Strains , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/genetics , Osteoclasts/drug effects , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , RANK Ligand/metabolism , RANK Ligand/pharmacology , Tartrate-Resistant Acid Phosphatase
14.
PLoS One ; 8(10): e76505, 2013.
Article in English | MEDLINE | ID: mdl-24124570

ABSTRACT

Wallerian degeneration is delayed when sufficient levels of proteins with NMNAT activity are maintained within axons after injury. This has been proposed to form the basis of 'slow Wallerian degeneration' (Wld (S)), a neuroprotective phenotype conferred by an aberrant fusion protein, Wld(S). Proteasome inhibition also delays Wallerian degeneration, although much less robustly, with stabilization of NMNAT2 likely to play a key role in this mechanism. The pan-MEK inhibitor U0126 has previously been shown to reverse the axon-protective effects of proteasome inhibition, suggesting that MEK-ERK signaling plays a role in delayed Wallerian degeneration, in addition to its established role in promoting neuronal survival. Here we show that whilst U0126 can also reverse Wld(S)-mediated axon protection, more specific inhibitors of MEK1/2 and MEK5, PD184352 and BIX02189, have no significant effect on the delay to Wallerian degeneration in either situation, whether used alone or in combination. This suggests that an off-target effect of U0126 is responsible for reversion of the axon protective effects of Wld(S) expression or proteasome inhibition, rather than inhibition of MEK1/2-ERK1/2 or MEK5-ERK5 signaling. Importantly, this off-target effect does not appear to result in alterations in the stabilities of either Wld(S) or NMNAT2.


Subject(s)
Axons/drug effects , Axons/metabolism , Butadienes/pharmacology , MAP Kinase Signaling System/drug effects , Nitriles/pharmacology , Protein Kinase Inhibitors/pharmacology , Wallerian Degeneration/metabolism , Animals , Cell Line , Enzyme Stability/drug effects , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 5/antagonists & inhibitors , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Phenotype , Rats , Superior Cervical Ganglion/drug effects , Superior Cervical Ganglion/metabolism , Wallerian Degeneration/drug therapy
15.
Biochim Biophys Acta ; 1825(1): 37-48, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22020294

ABSTRACT

While conventional MAP kinase pathways are one of the most highly studied signal transduction molecules, less is known about the MEK5 signaling pathway. This pathway has been shown to play a role in normal cell growth cycles, survival and differentiation. The MEK5 pathway is also believed to mediate the effects of a number of oncogenes. MEK5 is the upstream activator of ERK5 in many epithelial cells. Activation of the MEK-MAPK pathway is a frequent event in malignant tumor formation and contributes to chemoresistance and anti-apoptotic signaling. This pathway may be involved in a number of more aggressive, metastatic varieties of cancer due to its role in cell survival, proliferation and EMT transitioning. Further study of this pathway may lead to new prognostic factors and new drug targets to combat more aggressive forms of cancer.


Subject(s)
MAP Kinase Kinase 5/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 7/metabolism , Neoplasms/metabolism , Cell Differentiation , Cell Survival , Epithelial-Mesenchymal Transition , Humans , MAP Kinase Kinase 5/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors
16.
Biochem Biophys Res Commun ; 412(2): 307-12, 2011 Aug 26.
Article in English | MEDLINE | ID: mdl-21820407

ABSTRACT

Fms-like tyrosine kinase-3 (FLT3) is a growth factor receptor normally expressed on hematopoietic progenitor cells. Approximately one third of all patients with AML carry an activating mutation in FLT3 that drives proliferation and survival of the leukemic cells. The most common activating mutation is the so-called internal tandem duplication (ITD), which involves an in-frame duplication of a segment of varying length in the region of the FLT3 gene that encodes the juxtamembrane domain. The pathways downstream of FLT3-ITD are partially known but further knowledge regarding the downstream signal transduction molecules is important in order to develop alternative strategies for pharmacological intervention. In this paper we have studied the role of MEK/ERK5 in FLT3-ITD mediated transformation. We have found that both wild-type FLT3 and FLT3-ITD activate MEK5 leading to the activation of ERK5. By use of the selective inhibitor of MEK5, BIX02188, we have shown that activation of AKT downstream of FLT3 is partially dependent on ERK5. Furthermore, inhibition of MEK5/ERK5 induces apoptosis of both FLT3-ITD transfected Ba/F3 cells as well as the FLT3-ITD carrying leukemic cell lines MV4-11 and MOLM-13. These results suggest that MEK5/ERK5 is important for FLT3-ITD induced hematopoietic transformation and may thus represent an alternative therapeutic target in the treatment of FLT3-ITD positive leukemia.


Subject(s)
Apoptosis , MAP Kinase Kinase 5/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Oncogene Proteins/biosynthesis , fms-Like Tyrosine Kinase 3/biosynthesis , Aniline Compounds/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Indoles/pharmacology , Leukemia/enzymology , Ligands , MAP Kinase Kinase 5/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oncogene Proteins/genetics , Phosphorylation/drug effects , fms-Like Tyrosine Kinase 3/genetics
17.
Chem Biol ; 18(6): 699-710, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21700206

ABSTRACT

Protein kinases are intensely studied mediators of cellular signaling, yet important questions remain regarding their regulation and in vivo properties. Here, we use a probe-based chemoprotemics platform to profile several well studied kinase inhibitors against >200 kinases in native cell proteomes and reveal biological targets for some of these inhibitors. Several striking differences were identified between native and recombinant kinase inhibitory profiles, in particular, for the Raf kinases. The native kinase binding profiles presented here closely mirror the cellular activity of these inhibitors, even when the inhibition profiles differ dramatically from recombinant assay results. Additionally, Raf activation events could be detected on live cell treatment with inhibitors. These studies highlight the complexities of protein kinase behavior in the cellular context and demonstrate that profiling with only recombinant/purified enzymes can be misleading.


Subject(s)
Protein Kinases/chemistry , Adenosine Triphosphate/chemistry , Cell Line, Tumor , Dasatinib , Humans , MAP Kinase Kinase 5/antagonists & inhibitors , MAP Kinase Kinase 5/metabolism , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinases/genetics , Protein Kinases/metabolism , Pyrimidines/chemistry , Pyrimidines/pharmacology , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thiazoles/chemistry , Thiazoles/pharmacology , raf Kinases/antagonists & inhibitors , raf Kinases/genetics , raf Kinases/metabolism
18.
Microcirculation ; 18(2): 102-17, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21166929

ABSTRACT

OBJECTIVE: ECs lining arteries respond to LSS by suppressing pro-inflammatory changes, in part through the activation of MEK5, ERK5 and induction of KLF4. We examined if this anti-inflammatory pathway operates in human ECs lining microvessels, the principal site of inflammatory responses. METHODS: We used immunofluorescence microscopy of human skin to assess ERK5 activation and KLF4 expression in HDMECs in situ. We applied LSS to or overexpressed MEK5/CA in cultured HDMECs and assessed gene expression by microarrays and qRT-PCR and protein expression by Western blotting. We assessed effects of MEK5/CA on TNF responses using qRT-PCR, FACS and measurements of HDMEC monolayer electrical resistance. We used siRNA knockdown to assess the role of ERK5 and KLF4 in these responses. RESULTS: ERK5 phosphorylation and KLF4 expression is observed in HDMECs in situ. LSS activates ERK5 and induces KLF4 in cultured HDMECs. MEK5/CA-transduced HDMECs show activated ERK5 and increased KLF4, thrombomodulin, eNOS, and ICAM-1 expression. MEK5 induction of KLF4 is mediated by ERK5. MEK5/CA-transduced HDMECs are less responsive to TNF, an effect partly mediated by KLF4. CONCLUSIONS: MEK5 activation by LSS inhibits inflammatory responses in microvascular ECs, in part through ERK5-dependent induction of KLF4.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/metabolism , Kruppel-Like Transcription Factors/biosynthesis , MAP Kinase Kinase 5/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Base Sequence , Cells, Cultured , Enzyme Activation , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/genetics , MAP Kinase Kinase 5/antagonists & inhibitors , MAP Kinase Kinase 5/genetics , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Shear Strength , Stress, Mechanical
19.
Bioorg Med Chem Lett ; 20(9): 2892-6, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20382528

ABSTRACT

The MEK-signaling pathways are complex but critical signaling cascades that correlate an extracellular signaling event with internal cell processes. To date at least seven MEK isozymes have been identified. MEK5, in particular, is upregulated in multiple forms of tumors. Analysis of the EGF-induced MEK5 signaling cascade in cultured HEK cells has identified compounds that can inhibit MEK5 phosphorylation of ERK5; observed biological activity is dependent on chemical variation.


Subject(s)
Benzimidazoles/chemistry , MAP Kinase Kinase 5/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Signal Transduction/drug effects , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacology , Cell Line , Epidermal Growth Factor/pharmacology , Humans , MAP Kinase Kinase 5/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Phosphorylation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology
20.
Biochem Biophys Res Commun ; 377(1): 120-5, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-18834865

ABSTRACT

We have identified two novel MEK5 inhibitors, BIX02188 and BIX02189, which inhibited catalytic function of purified, MEK5 enzyme. The MEK5 inhibitors blocked phosphorylation of ERK5, without affecting phosphorylation of ERK1/2 in sorbitol-stimulated HeLa cells. The compounds also inhibited transcriptional activation of MEF2C, a downstream substrate of the MEK5/ERK5 signaling cascade, in a cellular trans-reporter assay system. These inhibitors offer novel pharmacological tools to better characterize the role of the MEK5/ERK5 pathway in various biological systems.


Subject(s)
Aniline Compounds/pharmacology , Indoles/pharmacology , MAP Kinase Kinase 5/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Aniline Compounds/isolation & purification , HeLa Cells , Humans , Indoles/isolation & purification , MADS Domain Proteins/antagonists & inhibitors , MADS Domain Proteins/genetics , MAP Kinase Kinase 5/metabolism , MEF2 Transcription Factors , Mitogen-Activated Protein Kinase 7/metabolism , Myogenic Regulatory Factors/antagonists & inhibitors , Myogenic Regulatory Factors/genetics , Phosphorylation/drug effects , Protein Kinase Inhibitors/isolation & purification , Sorbitol/pharmacology , Transcriptional Activation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL