Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 522
Filter
1.
Stem Cell Reports ; 19(9): 1320-1335, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39151429

ABSTRACT

The homeostasis of human pluripotent stem cells (hPSCs) requires the signaling balance of extracellular factors. Exogenous regulators from cell culture medium have been widely reported, but little attention has been paid to the autocrine factor from hPSCs themselves. In this report, we demonstrate that extracellular signal-related kinase 5 (ERK5) regulates endogenous autocrine factors essential for pluripotency and differentiation. ERK5 inhibition leads to erroneous cell fate specification in all lineages even under lineage-specific induction. hPSCs can self-renew under ERK5 inhibition in the presence of fibroblast growth factor 2 (FGF2) and transforming growth factor ß (TGF-ß), although NANOG expression is partially suppressed. Further analysis demonstrates that ERK5 promotes the expression of autocrine factors such as NODAL, FGF8, and WNT3. The addition of NODAL protein rescues NANOG expression and differentiation phenotypes under ERK5 inhibition. We demonstrate that constitutively active ERK5 pathway allows self-renewal even without essential growth factors FGF2 and TGF-ß. This study highlights the essential contribution of autocrine pathways to proper maintenance and differentiation.


Subject(s)
Autocrine Communication , Mitogen-Activated Protein Kinase 7 , Nanog Homeobox Protein , Pluripotent Stem Cells , Humans , Cell Differentiation , Cell Line , Cell Lineage , Cell Proliferation , Cell Self Renewal , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Mitogen-Activated Protein Kinase 7/metabolism , Mitogen-Activated Protein Kinase 7/genetics , Nanog Homeobox Protein/metabolism , Nanog Homeobox Protein/genetics , Nodal Protein/metabolism , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Signal Transduction , Transforming Growth Factor beta/metabolism
2.
Biochim Biophys Acta Mol Cell Res ; 1871(7): 119810, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39128596

ABSTRACT

Lung cancer is the leading cause of cancer-related deaths worldwide, with non-small cell lung cancer (NSCLC) constituting approximately 84 % of all lung cancer cases. The role of inflammation in the initiation and progression of NSCLC tumors has been the focus of extensive research. Among the various inflammatory mediators, prostaglandin E2 (PGE2) plays a pivotal role in promoting the aggressiveness of epithelial tumors through multiple mechanisms, including the stimulation of growth, evasion of apoptosis, invasion, and induction of angiogenesis. The Extracellular signal-Regulated Kinase 5 (ERK5), the last discovered member among conventional mitogen-activated protein kinases (MAPK), is implicated in cancer-associated inflammation. In this study, we explored whether ERK5 is involved in the process of tumorigenesis induced by PGE2. Using A549 and PC9 NSCLC cell lines, we found that PGE2 triggers the activation of ERK5 via the EP1 receptor. Moreover, both genetic and pharmacological inhibition of ERK5 reduced PGE2-induced proliferation, migration, invasion and stemness of A549 and PC9 cells, indicating that ERK5 plays a critical role in PGE2-induced tumorigenesis. In summary, our study underscores the pivotal role of the PGE2/EP1/ERK5 axis in driving the malignancy of NSCLC cells in vitro. Targeting this axis holds promise as a potential avenue for developing novel therapeutic strategies aimed at controlling the advancement of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Cell Movement , Cell Proliferation , Dinoprostone , Lung Neoplasms , Mitogen-Activated Protein Kinase 7 , Humans , Dinoprostone/metabolism , Dinoprostone/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Mitogen-Activated Protein Kinase 7/metabolism , Mitogen-Activated Protein Kinase 7/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Cell Movement/drug effects , A549 Cells , Cell Line, Tumor , Carcinogenesis/genetics , Carcinogenesis/metabolism , Phenotype
3.
Bioorg Med Chem Lett ; 112: 129914, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39111728

ABSTRACT

Mitogen-activated protein kinase kinases (MAP2Ks) 1, 4, and 7 are potential targets for treating various diseases. Here, we solved the crystal structures of MAP2K1 and MAP2K4 complexed with covalent inhibitor 5Z-7-oxozeaenol (5Z7O). The elucidated structures showed that 5Z7O was non-covalently bound to the ATP binding site of MAP2K4, while it covalently attached to cysteine at the DFG-1 position of the deep ATP site of MAP2K1. In contrast, we previously showed that 5Z7O covalently binds to MAP2K7 via another cysteine on the solvent-accessible edge of the ATP site. Structural analyses and molecular dynamics calculations indicated that the configuration and mobility of conserved gatekeeper methionine located at the central ATP site regulated the binding and access of 5Z7O to the ATP site of MAP2Ks. These structural features provide clues for developing highly potent and selective inhibitors against MAP2Ks. Abbreviations: ATP, adenosine triphosphate; FDA, Food and Drug Administration; MAP2Ks, mitogen-activated protein kinase kinases; MD, molecular dynamics; NSCLC, non-small cell lung cancer; 5Z7O, 5Z-7-oxozeaenol; PDB, protein data bank; RMSD, root-mean-square deviation.


Subject(s)
Adenosine Triphosphate , Methionine , Protein Kinase Inhibitors , Zearalenone , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/chemistry , Humans , Methionine/chemistry , Methionine/metabolism , Binding Sites , Zearalenone/analogs & derivatives , Zearalenone/chemistry , Zearalenone/pharmacology , Zearalenone/metabolism , Zearalenone/administration & dosage , Mitogen-Activated Protein Kinase 7/metabolism , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/chemistry , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 7/metabolism , MAP Kinase Kinase 7/antagonists & inhibitors , MAP Kinase Kinase 7/chemistry , Structure-Activity Relationship , Molecular Dynamics Simulation , Crystallography, X-Ray , Molecular Structure , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Lactones , Resorcinols , MAP Kinase Kinase 4
4.
Theriogenology ; 226: 335-342, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38959844

ABSTRACT

Extracellular signal-regulated protein kinase 5 (Erk5), a member of the mitogen-activated protein kinase (MAPK) family, is ubiquitously expressed in all eukaryotic cells and is implicated in the various mitotic processes such as cell survival, proliferation, migration, and differentiation. However, the potential functional roles of Erk5 in oocyte meiosis have not been fully determined. In this study, we document that ERK5 participates in the meiotic maturation of mouse oocytes by regulating the spindle assembly to ensure the meiotic progression. We unexpectedly found that phosphorylated ERK5 was localized in the spindle pole region at metaphase I and II stages by immunostaining analysis. Inhibition of ERK5 activity using its specific inhibitor XMD8-92 dramatically reduced the incidence of first polar body extrusion. In addition, inhibition of ERK5 evoked the spindle assembly checkpoint to arrest oocytes at metaphase I stage by impairing the spindle assembly, chromosome alignment and kinetochore-microtubule attachment. Mechanically, over-strengthened microtubule stability was shown to disrupt the microtubule dynamics and thus compromise the spindle assembly in ERK5-inhibited oocytes. Conversely, overexpression of ERK5 caused decreased level of acetylated α-tubulin and spindle defects. Collectively, we conclude that ERK5 plays an important role in the oocyte meiotic maturation by regulating microtubule dynamics and spindle assembly.


Subject(s)
Meiosis , Mitogen-Activated Protein Kinase 7 , Oocytes , Spindle Apparatus , Animals , Oocytes/physiology , Meiosis/physiology , Mice , Spindle Apparatus/physiology , Mitogen-Activated Protein Kinase 7/metabolism , Mitogen-Activated Protein Kinase 7/genetics , Female
5.
FEBS Lett ; 598(16): 2011-2027, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38977937

ABSTRACT

Malignant melanoma, an aggressive skin cancer with a poor prognosis, frequently features BRAFV600E mutation resulting in activation of the MAPK pathway and melanocyte proliferation and survival. BRAFV600E inhibitors like vemurafenib and dabrafenib have enhanced patient survival, yet drug resistance remains a significant challenge. We investigated the role of the ERK5 pathway in BRAFV600E melanoma cells and cells with acquired resistance to PLX4720 (vemurafenib) and dabrafenib. In BRAFV600E melanoma, ERK5 inhibition minimally affected viability compared to ERK1/2 inhibition. In vemurafenib-resistant cells, ERK5 inhibition alone didn't impact viability or restore drug sensitivity to vemurafenib. However, in dabrafenib-resistant cells, ERK5 inhibition reduced viability and enhanced the anti-proliferative effect of MEK1/2 inhibition. Targeting the ERK5 pathway may represent a therapeutic opportunity in dabrafenib-resistant melanoma.


Subject(s)
Drug Resistance, Neoplasm , Imidazoles , Melanoma , Mitogen-Activated Protein Kinase 7 , Oximes , Proto-Oncogene Proteins B-raf , Vemurafenib , Oximes/pharmacology , Humans , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Melanoma/metabolism , Imidazoles/pharmacology , Vemurafenib/pharmacology , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/drug effects , Cell Line, Tumor , Mitogen-Activated Protein Kinase 7/metabolism , Mitogen-Activated Protein Kinase 7/genetics , Indoles/pharmacology , Sulfonamides/pharmacology , Cell Proliferation/drug effects , MAP Kinase Signaling System/drug effects , Cell Survival/drug effects , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Mutation
6.
Biomolecules ; 14(5)2024 May 04.
Article in English | MEDLINE | ID: mdl-38785963

ABSTRACT

Bones are vital for anchoring muscles, tendons, and ligaments, serving as a fundamental element of the human skeletal structure. However, our understanding of bone development mechanisms and the maintenance of bone homeostasis is still limited. Extracellular signal-related kinase 5 (ERK5), a recently identified member of the mitogen-activated protein kinase (MAPK) family, plays a critical role in the pathogenesis and progression of various diseases, especially neoplasms. Recent studies have highlighted ERK5's significant role in both bone development and bone-associated pathologies. This review offers a detailed examination of the latest research on ERK5 in different tissues and diseases, with a particular focus on its implications for bone health. It also examines therapeutic strategies and future research avenues targeting ERK5.


Subject(s)
Bone and Bones , Mitogen-Activated Protein Kinase 7 , Humans , Mitogen-Activated Protein Kinase 7/metabolism , Bone and Bones/metabolism , Bone and Bones/enzymology , Animals
7.
Anticancer Res ; 44(6): 2511-2515, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38821590

ABSTRACT

BACKGROUND/AIM: Thymic carcinoma is a rare cancer type with limited treatment options. Our previous study demonstrated that statins, which inhibit 3-hydroxy-3-methylglutaryl coenzyme A reductase, can prevent thymic carcinoma. However, the mechanisms through which statins affect intracellular events in cancer cells are not well understood. The aim of the study was to determine how thymic carcinoma modulates the intracellular signals in response to statin administration. MATERIALS AND METHODS: We analyzed statin-induced protein phosphorylation in Ty82 human thymic carcinoma cells, which were cultured with fluvastatin, and protein phosphorylation was examined using western blotting. RESULTS: Treating Ty82 with fluvastatin led to ERK5 phosphorylation via protein prenylation attenuation. The antitumor effects of fluvastatin on thymic carcinoma were enhanced when combined with an ERK5 inhibitor. CONCLUSION: Statin therapy in combination with ERK5 inhibition may be a promising therapeutic approach for treating thymic carcinoma.


Subject(s)
Fatty Acids, Monounsaturated , Fluvastatin , Indoles , Mitogen-Activated Protein Kinase 7 , Thymus Neoplasms , Fluvastatin/pharmacology , Humans , Thymus Neoplasms/drug therapy , Thymus Neoplasms/pathology , Thymus Neoplasms/metabolism , Cell Line, Tumor , Mitogen-Activated Protein Kinase 7/metabolism , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Phosphorylation/drug effects , Indoles/pharmacology , Fatty Acids, Monounsaturated/pharmacology , Thymoma/drug therapy , Thymoma/pathology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Animals
8.
Am J Pathol ; 194(8): 1581-1591, 2024 08.
Article in English | MEDLINE | ID: mdl-38705382

ABSTRACT

Melanoma is the deadliest skin cancer, with a poor prognosis in advanced stages. While available treatments have improved survival, long-term benefits are still unsatisfactory. The mitogen-activated protein kinase extracellular signal-regulated kinase 5 (ERK5) promotes melanoma growth, and ERK5 inhibition determines cellular senescence and the senescence-associated secretory phenotype. Here, latent-transforming growth factor ß-binding protein 1 (LTBP1) mRNA was found to be up-regulated in A375 and SK-Mel-5 BRAF V600E melanoma cells after ERK5 inhibition. In keeping with a key role of LTBP1 in regulating transforming growth factor ß (TGF-ß), TGF-ß1 protein levels were increased in lysates and conditioned media of ERK5-knockdown (KD) cells, and were reduced upon LTBP1 KD. Both LTBP1 and TGF-ß1 proteins were increased in melanoma xenografts in mice treated with the ERK5 inhibitor XMD8-92. Moreover, treatment with conditioned media from ERK5-KD melanoma cells reduced cell proliferation and invasiveness, and TGF-ß1-neutralizing antibodies impaired these effects. In silico data sets revealed that higher expression levels of both LTBP1 and TGF-ß1 mRNA were associated with better overall survival of melanoma patients. Increased LTBP1 or TGF-ß1 expression played a beneficial role in patients treated with anti-PD1 immunotherapy, making a possible immunosuppressive role of LTBP1/TGF-ß1 unlikely upon ERK5 inhibition. This study, therefore, identifies additional desirable effects of ERK5 targeting, providing evidence of an ERK5-dependent tumor-suppressive role of TGF-ß in melanoma.


Subject(s)
Cell Proliferation , Latent TGF-beta Binding Proteins , Melanoma , Mitogen-Activated Protein Kinase 7 , Transforming Growth Factor beta1 , Melanoma/metabolism , Melanoma/pathology , Melanoma/genetics , Melanoma/drug therapy , Humans , Latent TGF-beta Binding Proteins/metabolism , Latent TGF-beta Binding Proteins/genetics , Animals , Mitogen-Activated Protein Kinase 7/metabolism , Mitogen-Activated Protein Kinase 7/genetics , Mice , Transforming Growth Factor beta1/metabolism , Skin Neoplasms/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/genetics , Cell Line, Tumor , Xenograft Model Antitumor Assays
9.
Mol Biol Rep ; 51(1): 313, 2024 Feb 19.
Article in English | MEDLINE | ID: mdl-38374452

ABSTRACT

BACKGROUND: Glucagon-like peptide-1 (GLP-1) (7-36) amide, an endogenous active form of GLP-1, has been shown to modulate oxidative stress and neuronal cell survival in various neurological diseases. OBJECTIVE: This study investigated the potential effects of GLP-1(7-36) on oxidative stress and apoptosis in neuronal cells following traumatic brain injury (TBI) and explored the underlying mechanisms. METHODS: Traumatic brain injury (TBI) models were established in male SD rats for in vivo experiments. The extent of cerebral oedema was assessed using wet-to-dry weight ratios following GLP-1(7-36) intervention. Neurological dysfunction and cognitive impairment were evaluated through behavioural experiments. Histopathological changes in the brain were observed using haematoxylin and eosin staining. Oxidative stress levels in hippocampal tissues were measured. TUNEL staining and Western blotting were employed to examine cell apoptosis. In vitro experiments evaluated the extent of oxidative stress and neural apoptosis following ERK5 phosphorylation activation. Immunofluorescence colocalization of p-ERK5 and NeuN was analysed using immunofluorescence cytochemistry. RESULTS: Rats with TBI exhibited neurological deterioration, increased oxidative stress, and enhanced apoptosis, which were ameliorated by GLP-1(7-36) treatment. Notably, GLP-1(7-36) induced ERK5 phosphorylation in TBI rats. However, upon ERK5 inhibition, oxidative stress and neuronal apoptosis levels were elevated, even in the presence of GLP-1(7-36). CONCLUSION: In summary, this study suggested that GLP-1(7-36) suppressed oxidative damage and neuronal apoptosis after TBI by activating ERK5/CREB.


Subject(s)
Brain Injuries, Traumatic , Glucagon-Like Peptide 1 , Neuroprotective Agents , Animals , Male , Rats , Apoptosis , Brain Injuries, Traumatic/drug therapy , Disease Models, Animal , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide 1/therapeutic use , Hippocampus , Neuroprotective Agents/pharmacology , Oxidative Stress , Rats, Sprague-Dawley , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Mitogen-Activated Protein Kinase 7/drug effects , Mitogen-Activated Protein Kinase 7/metabolism , Cyclic AMP Response Element-Binding Protein/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism
10.
J Mol Recognit ; 37(1): e3067, 2024 01.
Article in English | MEDLINE | ID: mdl-37956676

ABSTRACT

Mitogen-activated protein kinase 7 (MAPK7) is a serine/threonine protein kinase that belongs to the MAPK family and plays a vital role in various cellular processes such as cell proliferation, differentiation, gene transcription, apoptosis, metabolism, and cell survival. The elevated expression of MAPK7 has been associated with the onset and progression of multiple aggressive tumors in humans, underscoring the potential of targeting MAPK7 pathways in therapeutic research. This pursuit holds promise for the advancement of anticancer drug development by developing potential MAPK7 inhibitors. To look for potential MAPK7 inhibitors, we exploited structure-based virtual screening of natural products from the ZINC database. First, the Lipinski rule of five criteria was used to filter a large library of ~90,000 natural compounds, followed by ADMET and pan-assay interference compounds (PAINS) filters. Then, top hits were chosen based on their strong binding affinity as determined by molecular docking. Further, interaction analysis was performed to find effective and specific compounds that can precisely bind to the binding pocket of MAPK7. Consequently, two compounds, ZINC12296700 and ZINC02123081, exhibited significant binding affinity and demonstrated excellent drug-like properties. All-atom molecular dynamics simulations for 200 ns confirmed the stability of MAPK7-ZINC12296700 and MAPK7-ZINC02123081 docked complexes. According to the molecular mechanics Poisson-Boltzmann surface area investigation, the binding affinities of both complexes were considerable. Overall, the result suggests that ZINC12296700 and ZINC02123081 might be used as promising leads to develop novel MAPK7 inhibitors. Since these compounds would interfere with the kinase activity of MAPK7, therefore, may be implemented to control cell growth and proliferation in cancer after required validations.


Subject(s)
Biological Products , Humans , Biological Products/pharmacology , Biological Products/chemistry , Mitogen-Activated Protein Kinase 7/genetics , Mitogen-Activated Protein Kinase 7/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Serine-Threonine Kinases/chemistry , Protein Kinase Inhibitors/chemistry
11.
Bioorg Med Chem ; 95: 117503, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37862935

ABSTRACT

The extracellular signal-regulated kinase 5 (Erk5) signaling plays a crucial role in cancer, and regulating its activity may have potential in cancer chemotherapy. In this study, a series of novel 7-azaindole derivatives (4a-5o) were designed and synthesized. Their antitumor activities on human lung cancer A549 cells was evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, 4',6-diamidino-2-phenylindole (DAPI) staining and colony formation assay. Among them, compounds 4a, 4 h, 5d and 5j exhibited good anti-proliferative activity with the IC50 values of 6.23 µg/mL, 8.52 µg/mL, 7.33 µg/mL and 4.56 µg/mL, respectively, equivalent to Erk5 positive control XMD8-92 (IC50 = 5.36 µg/mL). The results of structure-activity relationships (SAR) showed that double bond on the piperidine ring and N atoms at the N7 position of 7-azaindole was essential for their antiproliferative activity. Furthermore, compounds 4a and 5j exhibited good inhibition on Erk5 kinase through Western blot analysis and possible action site of compounds with Erk5 kinase was elucidated by molecular docking.


Subject(s)
Antineoplastic Agents , Mitogen-Activated Protein Kinase 7 , Humans , Mitogen-Activated Protein Kinase 7/metabolism , Molecular Docking Simulation , Cell Proliferation , Structure-Activity Relationship , Antineoplastic Agents/chemistry , Drug Screening Assays, Antitumor , Cell Line, Tumor , Molecular Structure
12.
Sci Rep ; 13(1): 11829, 2023 07 22.
Article in English | MEDLINE | ID: mdl-37481602

ABSTRACT

Newts have the extraordinary ability to fully regenerate lost or damaged cardiac, neural and retinal tissues, and even amputated limbs. In contrast, mammals lack these broad regenerative capabilities. While the molecular basis of newts' regenerative ability is the subject of active study, the underlying paracrine signaling factors involved remain largely uncharacterized. Extracellular vesicles (EVs) play an important role in cell-to-cell communication via EV cargo-mediated regulation of gene expression patterns within the recipient cells. Here, we report that newt myogenic precursor (A1) cells secrete EVs (A1EVs) that contain messenger RNAs associated with early embryonic development, neuronal differentiation, and cell survival. Exposure of rat primary superior cervical ganglion (SCG) neurons to A1EVs increased neurite outgrowth, facilitated by increases in mitochondrial respiration. Canonical pathway analysis pinpointed activation of NGF/ERK5 signaling in SCG neurons exposed to A1EV, which was validated experimentally. Thus, newt EVs drive neurite growth and complexity in mammalian primary neurons.


Subject(s)
Extracellular Vesicles , Neurons , Animals , Rats , Cells, Cultured , Neurons/cytology , Neurons/metabolism , Neurites/metabolism , Nerve Growth Factor/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Signal Transduction
13.
Circ Res ; 133(1): 25-44, 2023 06 23.
Article in English | MEDLINE | ID: mdl-37264926

ABSTRACT

BACKGROUND: ERK5 (extracellular signal-regulated kinase 5) is a dual kinase transcription factor containing an N-terminal kinase domain and a C-terminal transcriptional activation domain. Many ERK5 kinase inhibitors have been developed and tested to treat cancer and inflammatory diseases. However, recent data have raised questions about the role of the catalytic activity of ERK5 in proliferation and inflammation. We aimed to investigate how ERK5 reprograms myeloid cells to the proinflammatory senescent phenotype, subsequently leading to atherosclerosis. METHODS: A ERK5 S496A (dephosphorylation mimic) knock in (KI) mouse model was generated using CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeat-associated 9), and atherosclerosis was characterized by hypercholesterolemia induction. The plaque phenotyping in homozygous ERK5 S496A KI and wild type (WT) mice was studied using imaging mass cytometry. Bone marrow-derived macrophages were isolated from hypercholesterolemic mice and characterized using RNA sequencing and functional in vitro approaches, including senescence, mitochondria reactive oxygen species, and inflammation assays, as well as by metabolic extracellular flux analysis. RESULTS: We show that atherosclerosis was inhibited in ERK5 S496A KI mice. Furthermore, ERK5 S496 phosphorylation mediates both senescence-associated secretory phenotype and senescence-associated stemness by upregulating AHR (aryl hydrocarbon receptor) in plaque and bone marrow-derived macrophages isolated from hypercholesterolemic mice. We also discovered that ERK5 S496 phosphorylation could induce NRF2 (NFE2-related factor 2) SUMOylation at a novel K518 site to inhibit NRF2 transcriptional activity without altering ERK5 catalytic activity and mediates oxidized LDL (low-density lipoprotein)-induced senescence-associated secretory phenotype. Specific ERK5 kinase inhibitors (AX15836 and XMD8-92) also inhibited ERK5 S496 phosphorylation, suggesting the involvement of ERK5 S496 phosphorylation in the anti-inflammatory effects of these ERK5 kinase inhibitors. CONCLUSIONS: We discovered a novel mechanism by which the macrophage ERK5-NRF2 axis develops a unique senescence-associated secretory phenotype/stemness phenotype by upregulating AHR to engender atherogenesis. The finding of senescence-associated stemness phenotype provides a molecular explanation to resolve the paradox of senescence in proliferative plaque by permitting myeloid cells to escape the senescence-induced cell cycle arrest during atherosclerosis formation.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Animals , Mice , Atherosclerosis/metabolism , Inflammation , Mitogen-Activated Protein Kinase 7/genetics , Mitogen-Activated Protein Kinase 7/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism
14.
Cells ; 12(8)2023 04 13.
Article in English | MEDLINE | ID: mdl-37190064

ABSTRACT

Oxidative stress regulates many physiological and pathological processes. Indeed, a low increase in the basal level of reactive oxygen species (ROS) is essential for various cellular functions, including signal transduction, gene expression, cell survival or death, as well as antioxidant capacity. However, if the amount of generated ROS overcomes the antioxidant capacity, excessive ROS results in cellular dysfunctions as a consequence of damage to cellular components, including DNA, lipids and proteins, and may eventually lead to cell death or carcinogenesis. Both in vitro and in vivo investigations have shown that activation of the mitogen-activated protein kinase kinase 5/extracellular signal-regulated kinase 5 (MEK5/ERK5) pathway is frequently involved in oxidative stress-elicited effects. In particular, accumulating evidence identified a prominent role of this pathway in the anti-oxidative response. In this respect, activation of krüppel-like factor 2/4 and nuclear factor erythroid 2-related factor 2 emerged among the most frequent events in ERK5-mediated response to oxidative stress. This review summarizes what is known about the role of the MEK5/ERK5 pathway in the response to oxidative stress in pathophysiological contexts within the cardiovascular, respiratory, lymphohematopoietic, urinary and central nervous systems. The possible beneficial or detrimental effects exerted by the MEK5/ERK5 pathway in the above systems are also discussed.


Subject(s)
Antioxidants , Mitogen-Activated Protein Kinase 7 , Antioxidants/metabolism , MAP Kinase Kinase 5/genetics , MAP Kinase Kinase 5/metabolism , Mitogen-Activated Protein Kinase 7/genetics , Mitogen-Activated Protein Kinase 7/metabolism , Oxidative Stress , Reactive Oxygen Species , Humans , Animals
15.
Chem Biol Interact ; 380: 110562, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37224993

ABSTRACT

Regulation of the interplay between autophagy and oxidative stress is vital in maintaining neuronal homeostasis during neurotoxicity. The interesting involvement of NK1 receptor (NK1R) in neurodegeneration has highlighted the value of investigating the neuroprotective effect of aprepitant (Aprep), an NK1R antagonist in Parkinson's disease (PD). This study was conducted to disclose Aprep's ability to modulate extracellular signal-regulated kinase 5/Krüppel-like factor 4 (ERK5/KLF4) cue as molecular signaling implicated in regulating autophagy and redox signaling in response to rotenone neurotoxicity. Rotenone (1.5 mg/kg) was administered on alternate days, and rats were given Aprep simultaneously with or without PD98059, an ERK inhibitor, for 21 days. Aprep ameliorated motor deficits as verified by restored histological features, and intact neurons count in SN and striata along with tyrosine hydroxylase immunoreactivity in SN. The molecular signaling of Aprep was illustrated by the expression of KLF4 following the phosphorylation of its upstream target, ERK5. Nuclear factor erythroid 2-related factor 2 (Nrf2) was up-regulated, shifting the oxidant/antioxidant balance towards the antioxidant side, as evidenced by elevated GSH and suppressed MDA levels. In parallel, Aprep noticeably reduced phosphorylated α-synuclein aggregates due to autophagy induction as emphasized by marked LC3II/LC3I elevation and p62 level reduction. These effects were diminished upon PD98059 pre-administration. In conclusion, Aprep showed neuroprotective effects against rotenone-induced PD, which may be partially attributed to the activation of the ERK5/KLF4 signaling pathway. It modulated p62-mediated autophagy and Nrf2 axis which act cooperatively to counter rotenone-associated neurotoxicity pointing to Aprep's prospect as a curious candidate in PD research.


Subject(s)
Neuroprotective Agents , Parkinson Disease , Parkinsonian Disorders , Rats , Animals , Rotenone/toxicity , NF-E2-Related Factor 2/metabolism , Aprepitant/adverse effects , Antioxidants/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Receptors, Neurokinin-1/metabolism , Kruppel-Like Factor 4 , Parkinson Disease/metabolism , Signal Transduction , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress
16.
Cells ; 12(6)2023 03 22.
Article in English | MEDLINE | ID: mdl-36980305

ABSTRACT

Extracellular-signal-regulated kinase 5 (ERK5) is critical for normal cardiovascular development. Previous studies have defined a canonical pathway for ERK5 activation, showing that ligand stimulation leads to MEK5 activation resulting in dual phosphorylation of ERK5 on Thr218/Tyr220 residues within the activation loop. ERK5 then undergoes a conformational change, facilitating phosphorylation on residues in the C-terminal domain and translocation to the nucleus where it regulates MEF2 transcriptional activity. Our previous research into the importance of ERK5 in endothelial cells highlighted its role in VEGF-mediated tubular morphogenesis and cell survival, suggesting that ERK5 played a unique role in endothelial cells. Our current data show that in contrast to EGF-stimulated HeLa cells, VEGF-mediated ERK5 activation in human dermal microvascular endothelial cells (HDMECs) does not result in C-terminal phosphorylation of ERK5 and translocation to the nucleus, but instead to a more plasma membrane/cytoplasmic localisation. Furthermore, the use of small-molecule inhibitors to MEK5 and ERK5 shows that instead of regulating MEF2 activity, VEGF-mediated ERK5 is important for regulating AKT activity. Our data define a novel pathway for ERK5 activation in endothelial cells leading to cell survival.


Subject(s)
Mitogen-Activated Protein Kinase 7 , Proto-Oncogene Proteins c-akt , Vascular Endothelial Growth Factor A , Humans , Endothelial Cells/metabolism , HeLa Cells , Mitogen-Activated Protein Kinase 7/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Vascular Endothelial Growth Factor A/metabolism
17.
Cell Death Dis ; 14(1): 32, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36650140

ABSTRACT

YES-associated protein (YAP) is a transcriptional cofactor with a key role in the regulation of several physio-pathological cellular processes, by integrating multiple cell autonomous and microenvironmental cues. YAP is the main downstream effector of the Hippo pathway, a tumor-suppressive signaling able to transduce several extracellular signals. The Hippo pathway acts restraining YAP activity, since its activation induces YAP phosphorylation and cytoplasmic sequestration. However, recent observations indicate that YAP activity can be also modulated by Hippo independent/integrating pathways, still largely unexplored. In this study, we demonstrated the role of the extracellular signal-regulated kinase 5 (ERK5)/mitogen-activated protein kinase in the regulation of YAP activity. By means of ERK5 inhibition/silencing and overexpression experiments, and by using as model liver stem cells, hepatocytes, and hepatocellular carcinoma (HCC) cell lines, we provided evidence that ERK5 is required for YAP-dependent gene expression. Mechanistically, ERK5 controls the recruitment of YAP on promoters of target genes and its physical interaction with the transcriptional partner TEAD; moreover, it mediates the YAP activation occurring in cell adhesion, migration, and TGFß-induced EMT of liver cells. Furthermore, we demonstrated that ERK5 signaling modulates YAP activity in a LATS1/2-independent manner. Therefore, our observations identify ERK5 as a novel upstream Hippo-independent regulator of YAP activity, thus unveiling a new target for therapeutic approaches aimed at interfering with its function.


Subject(s)
Hepatocytes , Mitogen-Activated Protein Kinase 7 , YAP-Signaling Proteins , Humans , Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Liver Neoplasms/pathology , Mitogen-Activated Protein Kinase 7/genetics , Mitogen-Activated Protein Kinase 7/metabolism , Phosphoproteins/metabolism , YAP-Signaling Proteins/genetics , YAP-Signaling Proteins/metabolism , Hepatocytes/metabolism , Stem Cells
18.
J Mol Biol ; 435(2): 167890, 2023 01 30.
Article in English | MEDLINE | ID: mdl-36402225

ABSTRACT

14-3-3s are abundant proteins that regulate essentially all aspects of cell biology, including cell cycle, motility, metabolism, and cell death. 14-3-3s work by docking to phosphorylated Ser/Thr residues on a large network of client proteins and modulating client protein function in a variety of ways. In recent years, aided by improvements in proteomics, the discovery of 14-3-3 client proteins has far outpaced our ability to understand the biological impact of individual 14-3-3 interactions. The rate-limiting step in this process is often the identification of the individual phospho-serines/threonines that mediate 14-3-3 binding, which are difficult to distinguish from other phospho-sites by sequence alone. Furthermore, trial-and-error molecular approaches to identify these phosphorylations are costly and can take months or years to identify even a single 14-3-3 docking site phosphorylation. To help overcome this challenge, we used machine learning to analyze predictive features of 14-3-3 binding sites. We found that accounting for intrinsic protein disorder and the unbiased mass spectrometry identification rate of a given phosphorylation significantly improves the identification of 14-3-3 docking site phosphorylations across the proteome. We incorporated these features, coupled with consensus sequence prediction, into a publicly available web app, called "14-3-3 site-finder". We demonstrate the strength of this approach through its ability to identify 14-3-3 binding sites that do not conform to the loose consensus sequence of 14-3-3 docking phosphorylations, which we validate with 14-3-3 client proteins, including TNK1, CHEK1, MAPK7, and others. In addition, by using this approach, we identify a phosphorylation on A-kinase anchor protein-13 (AKAP13) at Ser2467 that dominantly controls its interaction with 14-3-3.


Subject(s)
14-3-3 Proteins , Protein Interaction Maps , Humans , 14-3-3 Proteins/metabolism , Binding Sites , Fetal Proteins/metabolism , Machine Learning , Mitogen-Activated Protein Kinase 7/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Proteome/metabolism , Serine/metabolism , Threonine/metabolism
19.
Cell Chem Biol ; 29(11): 1630-1638.e7, 2022 11 17.
Article in English | MEDLINE | ID: mdl-36220104

ABSTRACT

Recent interest in the role that extracellular signal-regulated kinase 5 (ERK5) plays in various diseases, particularly cancer and inflammation, has grown. Phenotypes observed from genetic knockdown or deletion of ERK5 suggested that targeting ERK5 could have therapeutic potential in various disease settings, motivating the development ATP-competitive ERK5 inhibitors. However, these inhibitors were unable to recapitulate the effects of genetic loss of ERK5, suggesting that ERK5 may have key kinase-independent roles. To investigate potential non-catalytic functions of ERK5, we report the development of INY-06-061, a potent and selective heterobifunctional degrader of ERK5. In contrast to results reported through genetic knockdown of ERK5, INY-06-061-induced ERK5 degradation did not induce anti-proliferative effects in multiple cancer cell lines or suppress inflammatory responses in primary endothelial cells. Thus, we developed and characterized a chemical tool useful for validating phenotypes reported to be associated with genetic ERK5 ablation and for guiding future ERK5-directed drug discovery efforts.


Subject(s)
Endothelial Cells , Mitogen-Activated Protein Kinase 7 , Humans , Mitogen-Activated Protein Kinase 7/genetics , Mitogen-Activated Protein Kinase 7/metabolism , Endothelial Cells/metabolism , Immunity, Cellular , Inflammation/drug therapy , Inflammation/metabolism , Cell Proliferation
20.
Nat Commun ; 13(1): 5124, 2022 08 31.
Article in English | MEDLINE | ID: mdl-36045118

ABSTRACT

The Pattern Of Invasion (POI) of tumor cells into adjacent normal tissues clinically predicts postoperative tumor metastasis/recurrence of early oral squamous cell carcinoma (OSCC), but the mechanisms underlying the development of these subtypes remain unclear. Focusing on the highest score of POIs (Worst POI, WPOI) present within each tumor, we observe a disease progression-driven shift of WPOI towards the high-risk type 4/5, associated with a mesenchymal phenotype in advanced OSCC. WPOI 4-5-derived cancer-associated fibroblasts (CAFsWPOI4-5), characterized by high oxytocin receptor expression (OXTRHigh), contribute to local-regional metastasis. OXTRHigh CAFs induce a desmoplastic stroma and CCL26 is required for the invasive phenotype of CCR3+ tumors. Mechanistically, OXTR activates nuclear ERK5 transcription signaling via Gαq and CDC37 to maintain high levels of OXTR and CCL26. ERK5 ablation reprograms the pro-invasive phenotype of OXTRHigh CAFs. Therefore, targeting ERK5 signaling in OXTRHigh CAFs is a potential therapeutic strategy for OSCC patients with WPOI 4-5.


Subject(s)
Head and Neck Neoplasms , Mitogen-Activated Protein Kinase 7 , Mouth Neoplasms , Squamous Cell Carcinoma of Head and Neck , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Fibroblasts/metabolism , Head and Neck Neoplasms/pathology , Humans , Mitogen-Activated Protein Kinase 7/metabolism , Mouth Neoplasms/pathology , Neoplasm Invasiveness/pathology , Receptors, Oxytocin/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology
SELECTION OF CITATIONS
SEARCH DETAIL