1
|
Escárcega-González CE, Hernández-Cuellar E, Ruiz Esparza-Juárez FD, Chávez-Reyes J. Molecular mechanisms associated with embryotoxic effects of heavy metals in the Sea Urchin. Reprod Toxicol 2025; 134:108898. [PMID: 40118295 DOI: 10.1016/j.reprotox.2025.108898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 02/24/2025] [Accepted: 03/15/2025] [Indexed: 03/23/2025]
Abstract
The sea urchin embryo model has become a classic model for studying the harmful effects of heavy metals and the molecular responses associated with exposure to these pollutants. In this context, several biochemical pathways have been associated with exposure to heavy metals in sea urchin embryos, such as autophagy, apoptosis, oxidative stress, activation of heat shock proteins, and induction of metallothioneins. These biochemical pathways are activated or altered in embryos after exposure to heavy metals; therefore, this review provides a comprehensive literature exploration, summarizing the main biochemical changes observed in sea urchin embryos following exposure to certain heavy metals, such as cadmium, gadolinium, arsenic, manganese, zinc, mercury, copper, nickel, and lead.
Collapse
Affiliation(s)
- Carlos E Escárcega-González
- Facultad de Ciencias Químicas Universidad Autónoma de Nuevo León Av., Universidad s/n, CD, Universitaria, San Nicolás de los Garza, NL 66455, Mexico
| | - Eduardo Hernández-Cuellar
- Laboratorio de Biología Celular y Tisular, Departamento de Morfología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Ciudad Universitaria 940, Aguascalientes 20100, Mexico
| | - Fabián D Ruiz Esparza-Juárez
- Departamento de Medicina, Centro de Ciencias de la Salud, Universidad Autónoma de Aguascalientes, Ciudad Universitaria 940, Aguascalientes 20100, Mexico
| | - Jesús Chávez-Reyes
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Ciudad Universitaria 940, Aguascalientes 20100, Mexico.
| |
Collapse
|
2
|
Xu G, Zhang W, Du J, Cong J, Wang P, Li X, Si X, Wei B. Binding mechanism of inhibitors to DFG-in and DFG-out P38α deciphered using multiple independent Gaussian accelerated molecular dynamics simulations and deep learning. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2025; 36:101-126. [PMID: 40110797 DOI: 10.1080/1062936x.2025.2475407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 02/27/2025] [Indexed: 03/22/2025]
Abstract
P38α has been identified as a key target for drug design to treat a wide range of diseases. In this study, multiple independent Gaussian accelerated molecular dynamics (GaMD) simulations, deep learning (DL), and the molecular mechanics generalized Born surface area (MM-GBSA) method were used to investigate the binding mechanism of inhibitors (SB2, SK8, and BMU) to DFG-in and DFG-out P38α and clarify the effect of conformational differences in P38α on inhibitor binding. GaMD trajectory-based DL effectively identified important functional domains, such as the A-loop and N-sheet. Post-processing analysis on GaMD trajectories showed that binding of the three inhibitors profoundly affected the structural flexibility and dynamical behaviour of P38α situated at the DFG-in and DFG-out states. The MM-GBSA calculations not only revealed that differences in the binding ability of inhibitors are affected by DFG-in and DFG-out conformations of P38α, but also confirmed that van der Waals interactions are the primary force driving inhibitor-P38α binding. Residue-based free energy estimation identifies hot spots of inhibitor-P38α binding across DFG-in and DFG-out conformations, providing potential target sites for drug design towards P38α. This work is expected to offer valuable theoretical support for the development of selective inhibitors of P38α family members.
Collapse
Affiliation(s)
- G Xu
- Center for Medical Artificial Intelligence, Research Institute for Marine Traditional Chinese Medicine (Qingdao Academy of Chinese Medical Sciences), Shandong University of Traditional Chinese Medicine, Qingdao, China
| | - W Zhang
- Center for Medical Artificial Intelligence, Research Institute for Marine Traditional Chinese Medicine (Qingdao Academy of Chinese Medical Sciences), Shandong University of Traditional Chinese Medicine, Qingdao, China
| | - J Du
- Center for Medical Artificial Intelligence, Research Institute for Marine Traditional Chinese Medicine (Qingdao Academy of Chinese Medical Sciences), Shandong University of Traditional Chinese Medicine, Qingdao, China
| | - J Cong
- Center for Medical Artificial Intelligence, Research Institute for Marine Traditional Chinese Medicine (Qingdao Academy of Chinese Medical Sciences), Shandong University of Traditional Chinese Medicine, Qingdao, China
| | - P Wang
- Center for Medical Artificial Intelligence, Research Institute for Marine Traditional Chinese Medicine (Qingdao Academy of Chinese Medical Sciences), Shandong University of Traditional Chinese Medicine, Qingdao, China
| | - X Li
- Center for Medical Artificial Intelligence, Research Institute for Marine Traditional Chinese Medicine (Qingdao Academy of Chinese Medical Sciences), Shandong University of Traditional Chinese Medicine, Qingdao, China
| | - X Si
- Center for Medical Artificial Intelligence, Research Institute for Marine Traditional Chinese Medicine (Qingdao Academy of Chinese Medical Sciences), Shandong University of Traditional Chinese Medicine, Qingdao, China
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - B Wei
- Center for Medical Artificial Intelligence, Research Institute for Marine Traditional Chinese Medicine (Qingdao Academy of Chinese Medical Sciences), Shandong University of Traditional Chinese Medicine, Qingdao, China
| |
Collapse
|
3
|
Pasula MB, Sylvester PW, Briski KP. GLUT2 regulation of p38 MAPK isoform protein expression and p38 phosphorylation in male versus female rat hypothalamic primary astrocyte Cultures. IBRO Neurosci Rep 2024; 16:635-642. [PMID: 38832087 PMCID: PMC11144729 DOI: 10.1016/j.ibneur.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 06/05/2024] Open
Abstract
Recent studies documented regulation of hypothalamic astrocyte mitogen-activated protein kinase (MAPK) pathways, including p38, by the plasma membrane glucose carrier/sensor glucose transporter-2 (GLUT2). Sex-specific GLUT2 control of p38 phosphorylation was observed, but effects on individual p38 family protein profiles were not investigated. Current research employed an established primary astrocyte culture model, gene knockdown tools, and selective primary antisera against p38-alpha, p38-beta, p38-gamma, and p38-delta isoforms to investigate whether GLUT2 governs expression of one or more of these variants in a glucose-dependent manner. Data show that GLUT2 inhibits baseline expression of each p38 protein in male cultures, yet stimulates p38-delta profiles without affecting other p38 proteins in female. Glucose starvation caused selective up-regulation of p38-delta profiles in male versus p38-alpha and -gamma proteins in female; these positive responses were amplified by GLUT2 siRNA pretreatment. GLUT2 opposes or enhances basal p38 phosphorylation in male versus female, respectively. GLUT2 siRNA pretreatment did not affect glucoprivic patterns of phospho-p38 protein expression in either sex. Outcomes document co-expression of the four principal p38 MAPK family proteins in hypothalamic astrocytes, and implicate GLUT2 in regulation of all (male) versus one (female) variant(s). Glucoprivation up-regulated expression of distinctive p38 isoforms in each sex; these stimulatory responses are evidently blunted by GLUT2. Glucoprivic-associated loss of GLUT2 gene silencing effects on p38 phosphorylation infers either that glucose status determines whether this sensor controls phosphorylation, or that decrements in screened glucose in each instance are of sufficient magnitude to abolish GLUT2 regulation of that function.
Collapse
Affiliation(s)
- Madhu Babu Pasula
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201, USA
| | - Paul W. Sylvester
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201, USA
| | - Karen P. Briski
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201, USA
| |
Collapse
|
4
|
Velasquez C, Gutierrez O, Carcelen M, Fernandez-Luna JL. The Invasion Factor ODZ1 Is Upregulated through an Epidermal Growth Factor Receptor-Induced Pathway in Primary Glioblastoma Cells. Cells 2024; 13:766. [PMID: 38727302 PMCID: PMC11083495 DOI: 10.3390/cells13090766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
We have previously shown that the transmembrane protein ODZ1 promotes cytoskeletal remodeling of glioblastoma (GBM) cells and invasion of the surrounding parenchyma through the activation of a RhoA-ROCK pathway. We also described that GBM cells can control the expression of ODZ1 through transcriptional mechanisms triggered by the binding of IL-6 to its receptor and a hypoxic environment. Epidermal growth factor (EGF) plays a key role in the invasive capacity of GBM. However, the molecular mechanisms that enable tumor cells to acquire the morphological changes to migrate out from the tumor core have not been fully characterized. Here, we show that EGF is able to induce the expression of ODZ1 in primary GBM cells. We analyzed the levels of the EGF receptor (EGFR) in 20 GBM primary cell lines and found expression in 19 of them by flow cytometry. We selected two cell lines that do or do not express the EGFR and found that EGFR-expressing cells responded to the EGF ligand by increasing ODZ1 at the mRNA and protein levels. Moreover, blockade of EGF-EGFR binding by Cetuximab, inhibition of the p38 MAPK pathway, or Additionally, the siRNA-mediated knockdown of MAPK11 (p38β MAPK) reduced the induction of ODZ1 in response to EGF. Overall, we show that EGF may activate an EGFR-mediated signaling pathway through p38β MAPK, to upregulate the invasion factor ODZ1, which may initiate morphological changes for tumor cells to invade the surrounding parenchyma. These data identify a new candidate of the EGF-EGFR pathway for novel therapeutic approaches.
Collapse
Affiliation(s)
- Carlos Velasquez
- Department of Neurosurgery, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain;
- Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39008 Santander, Spain; (O.G.); (M.C.)
- Department of Anatomy and Cellular Biology, Universidad de Cantabria, 39011 Santander, Spain
| | - Olga Gutierrez
- Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39008 Santander, Spain; (O.G.); (M.C.)
| | - Maria Carcelen
- Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39008 Santander, Spain; (O.G.); (M.C.)
| | - Jose L. Fernandez-Luna
- Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39008 Santander, Spain; (O.G.); (M.C.)
- Department of Genetics, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
- Centro de Investigación en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| |
Collapse
|
5
|
Wójcik M, Plata-Babula A, Głowaczewska A, Sirek T, Orczyk A, Małecka M, Grabarek BO. Expression profile of mRNAs and miRNAs related to mitogen-activated kinases in HaCaT cell culture treated with lipopolysaccharide a and adalimumab. Cell Cycle 2024; 23:385-404. [PMID: 38557266 PMCID: PMC11174132 DOI: 10.1080/15384101.2024.2335051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 02/20/2024] [Indexed: 04/04/2024] Open
Abstract
Studies indicate that mitogen-activated protein kinases (MAPKs) exhibit activation and overexpression within psoriatic lesions. This study aimed to investigate alterations in the expression patterns of genes encoding MAPKs and microRNA (miRNA) molecules that potentially regulate their expression in human adult low-calcium high-temperature (HaCaT) keratinocytes when exposed to bacterial lipopolysaccharide A (LPS) and adalimumab. HaCaT cells underwent treatment with 1 µg/mL LPS for 8 hours, followed by treatment with 8 µg/mL adalimumab for 2, 8, or 24 hours. Untreated cells served as controls. The molecular analysis involved microarray, quantitative real-time polymerase chain reaction (RTqPCR), and enzyme-linked immunosorbent assay (ELISA) analyses. Changes in the expression profile of seven mRNAs: dual specificity phosphatase 1 (DUSP1), dual specificity phosphatase 3 (DUSP3), dual specificity phosphatase 4 (DUSP4), mitogen-activated protein kinase 9 (MAPK9), mitogen-activated protein kinase kinase kinase 2 (MAP3K2), mitogen-activated protein kinase kinase 2 (MAP2K2), and MAP kinase-activated protein kinase 2 (MAPKAPK2, also known as MK2) in cell culture exposed to LPS or LPS and the drug compared to the control. It was noted that miR-34a may potentially regulate the activity of DUSP1, DUSP3, and DUSP4, while miR-1275 is implicated in regulating MAPK9 expression. Additionally, miR-382 and miR-3188 are potential regulators of DUSP4 levels, and miR-200-5p is involved in regulating MAPKAPK2 and MAP3K2 levels. Thus, the analysis showed that these mRNA molecules and the proteins and miRNAs they encode appear to be useful molecular markers for monitoring the efficacy of adalimumab therapy.
Collapse
Affiliation(s)
- Michał Wójcik
- Collegium Medicum, WSB University, Dabrowa Gornicza, Poland
| | - Aleksandra Plata-Babula
- Department of Nursing and Maternity, High School of Strategic Planning in Dabrowa Gornicza, Dabrowa Gornicza, Poland
| | - Amelia Głowaczewska
- Faculty of Health Sciences, University of Applied Sciences in Nysa, Nysa, Poland
| | - Tomasz Sirek
- Department of Plastic Surgery, Faculty of Medicine, Academia of Silesia, Katowice, Poland
- Department of Plastic and Reconstructive Surgery, Hospital for Minimally Invasive and Reconstructive Surgery in Bielsko-Biała, Bielsko-Biala, Poland
| | - Aneta Orczyk
- Collegium Medicum, WSB University, Dabrowa Gornicza, Poland
| | - Mariola Małecka
- Faculty of Medicine, Uczelnia Medyczna im. Marii Skłodowskiej-Curie, Warszawa, Poland
| | | |
Collapse
|
6
|
Ganguly P, Macleod T, Wong C, Harland M, McGonagle D. Revisiting p38 Mitogen-Activated Protein Kinases (MAPK) in Inflammatory Arthritis: A Narrative of the Emergence of MAPK-Activated Protein Kinase Inhibitors (MK2i). Pharmaceuticals (Basel) 2023; 16:1286. [PMID: 37765094 PMCID: PMC10537904 DOI: 10.3390/ph16091286] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
The p38 mitogen-activated protein kinase (p38-MAPK) is a crucial signaling pathway closely involved in several physiological and cellular functions, including cell cycle, apoptosis, gene expression, and responses to stress stimuli. It also plays a central role in inflammation and immunity. Owing to disparate p38-MAPK functions, it has thus far formed an elusive drug target with failed clinical trials in inflammatory diseases due to challenges including hepatotoxicity, cardiac toxicity, lack of efficacy, and tachyphylaxis, which is a brief initial improvement with rapid disease rebound. To overcome these limitations, downstream antagonism of the p38 pathway with a MAPK-activated protein kinase (MAPKAPK, also known as MK2) blockade has demonstrated the potential to abrogate inflammation without the prior recognized toxicities. Such MK2 inhibition (MK2i) is associated with robust suppression of key pro-inflammatory cytokines, including TNFα and IL-6 and others in experimental systems and in vitro. Considering this recent evidence regarding MK2i in inflammatory arthritis, we revisit the p38-MAPK pathway and discuss the literature encompassing the challenges of p38 inhibitors with a focus on this pathway. We then highlight how novel MK2i strategies, although encouraging in the pre-clinical arena, may either show evidence for efficacy or the lack of efficacy in emergent human trials data from different disease settings.
Collapse
Affiliation(s)
| | | | | | | | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds LS9 7JT, UK
| |
Collapse
|
7
|
Moreira J, Martins H, Saraiva M, Saraiva MJ. TLR2 and 4 signaling pathways are altered in macrophages from V30M TTR mice with down-regulated expression of chemokines. Clin Sci (Lond) 2023; 137:355-366. [PMID: 36852978 DOI: 10.1042/cs20220656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/15/2023] [Accepted: 02/28/2023] [Indexed: 03/01/2023]
Abstract
Hereditary amyloid transthyretin (ATTRv) amyloidosis is a fatal neurodegenerative disorder, first identified in Portugal. The most common transthyretin (TTR) mutation in ATTRv results from an exchange of a methionine for a valine at position 30 (V30M). ATTRv is characterized by the extracellular deposition of aggregates and fibrils of mutant forms of TTR, particularly in the nerves and ganglia of the peripheral nervous system (PNS). This phenotype is often accompanied by the lack of inflammatory infiltrates, despite the importance of macrophages in removal of TTR deposits in ATTRv patients. The mechanisms underlying this impairment of inflammatory responses in ATTRv patients are poorly understood. Here, we show a significant down-regulation in the expression of several chemokines by bone marrow-derived macrophages (BMDM) generated from V30M TTR mice upon stimulation with toll-like receptor 4 (TLR4) and TLR2 agonists. The phosphorylation of the MAP kinase p38, important for TLR4 and TLR2 signaling pathways, was also down-regulated in V30M macrophages, as compared with wild-type (WT) ones. The present study contributes with new insights to unravel the molecular mechanisms underlying the lack of inflammatory immune responses observed in ATTRv patients and may help in the development of new immune therapeutic strategies for the disease.
Collapse
Affiliation(s)
- João Moreira
- Molecular Neurobiology Group, i3S - Instituto de Investigação e Inovação em Saúde, Department of Neurobiology and Neurologic Disorders, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Department of Molecular Biology, Universidade do Porto, 4050-313 Porto, Portugal
| | - Helena Martins
- Molecular Neurobiology Group, i3S - Instituto de Investigação e Inovação em Saúde, Department of Neurobiology and Neurologic Disorders, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Margarida Saraiva
- Immune Regulation Group, i3S - Instituto de Investigação e Inovação em Saúde, Department of Infection, Immunity,and Regeneration, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Maria João Saraiva
- Molecular Neurobiology Group, i3S - Instituto de Investigação e Inovação em Saúde, Department of Neurobiology and Neurologic Disorders, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| |
Collapse
|
8
|
Hazegh K, Fang F, Kelly K, Sinchar D, Wang L, Zuchelkowski BE, Ufelle AC, Esparza O, Davizon-Castillo P, Page GP, Kanias T. Erythrocyte mitogen-activated protein kinases mediate hemolytic events under osmotic and oxidative stress and in hemolytic diseases. Cell Signal 2022; 99:110450. [PMID: 36029940 PMCID: PMC9530026 DOI: 10.1016/j.cellsig.2022.110450] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 11/20/2022]
Abstract
p38 MAPKs are key regulators of cellular adaptation to various stress stimuli, however, their role in mediating erythrocyte cell death and hemolysis is largely unknown. We hypothesized that activation of erythrocyte p38 MAPK is a common event in the stimulation of hemolysis, and that inhibition of p38 MAPK pathways could mitigate hemolysis in hemoglobinopathies. We exposed human erythrocytes to diamide-induced oxidative stress or to hypoosmotic shock in the presence or absence of p38 MAPK inhibitors (SCIO469, SB203580, CMPD1) and used immunoblotting to determine MAPK activity and to identify possible downstream effectors of p38 MAPK. We also evaluated the impact of p38 MAPK inhibitors on stress-induced hemolysis or hypoxia-induced sickling in erythrocytes from mouse models of sickle cell disease. We found that human erythrocytes express conventional MAPKs (MKK3, p38 MAPK, MAPKAPK2) and identified differential MAPK activation pathways in each stress condition. Specifically, p38 MAPK inhibition in diamide-treated erythrocytes was associated with decreased phosphorylation of Src tyrosine kinases and Band 3 protein. Conversely, hypoosmotic shock induced MAPKAPK2 and RSK2 phosphorylation, which was inhibited by SCIO469 or CMPD1. Relevant to hemoglobinopathies, sickle cell disease was associated with increased erythrocyte MKK3, p38 MAPK, and MAPKAPK2 expression and phosphorylation as compared with erythrocytes from healthy individuals. Furthermore, p38 MAPK inhibition was associated with decreased hemolysis in response to diamide treatments or osmotic shock, and with decreased erythrocyte sickling under experimental hypoxia. These findings provided insights into MAPK-mediated signaling pathways that regulate erythrocyte function and hemolysis in response to extracellular stressors or human diseases.
Collapse
Affiliation(s)
| | - Fang Fang
- RTI International, Research Triangle Park, NC, USA
| | | | - Derek Sinchar
- Vascular Medicine Institute, University of Pittsburg. Pittsburgh, PA, USA
| | - Ling Wang
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA, USA
| | | | - Alexander C Ufelle
- Department of Public Health, Slippery Rock University of Pennsylvania, Slippery Rock, PA, USA
| | - Orlando Esparza
- Department of Pediatric Hematology, Oncology, and Bone Marrow Transplant, Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | - Pavel Davizon-Castillo
- Department of Pediatrics, Anschutz Medical Campus and the Hemophilia and Thrombosis Center, University of Colorado, Aurora, CO, USA
| | | | - Tamir Kanias
- Vitalant Research Institute, Denver, CO, USA; Department of Pathology, Anschutz Medical Campus, University of Colorado Aurora, CO, USA.
| |
Collapse
|
9
|
Cheng Y, Chen J, Shi Y, Fang X, Tang Z. MAPK Signaling Pathway in Oral Squamous Cell Carcinoma: Biological Function and Targeted Therapy. Cancers (Basel) 2022; 14:cancers14194625. [PMID: 36230547 PMCID: PMC9563402 DOI: 10.3390/cancers14194625] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
Abstract
Oral squamous cell carcinoma accounts for 95% of human head and neck squamous cell carcinoma cases. It is highly malignant and aggressive, with a poor prognosis and a 5-year survival rate of <50%. In recent years, basic and clinical studies have been performed on the role of the mitogen-activated protein kinase (MAPK) signaling pathway in oral cancer. The MAPK signaling pathway is activated in over 50% of human oral cancer cases. Herein, we review research progress on the MAPK signaling pathway and its potential therapeutic mechanisms and discuss its molecular targeting to explore its potential as a therapeutic strategy for oral squamous cell carcinoma.
Collapse
Affiliation(s)
- Yuxi Cheng
- Xiangya Stomatological Hospital, Central South University, Changsha 410008, China
- Xiangya School of Stomatology, Central South University, Changsha 410008, China
| | - Juan Chen
- Xiangya Stomatological Hospital, Central South University, Changsha 410008, China
- Xiangya School of Stomatology, Central South University, Changsha 410008, China
| | - Yuxin Shi
- Xiangya Stomatological Hospital, Central South University, Changsha 410008, China
- Xiangya School of Stomatology, Central South University, Changsha 410008, China
| | - Xiaodan Fang
- Xiangya Stomatological Hospital, Central South University, Changsha 410008, China
- Xiangya School of Stomatology, Central South University, Changsha 410008, China
- Correspondence: (X.F.); (Z.T.)
| | - Zhangui Tang
- Xiangya Stomatological Hospital, Central South University, Changsha 410008, China
- Xiangya School of Stomatology, Central South University, Changsha 410008, China
- Correspondence: (X.F.); (Z.T.)
| |
Collapse
|
10
|
Liu Z, Demian W, Persaud A, Jiang C, Subramanaya AR, Rotin D. Regulation of the p38-MAPK pathway by hyperosmolarity and by WNK kinases. Sci Rep 2022; 12:14480. [PMID: 36008477 PMCID: PMC9411163 DOI: 10.1038/s41598-022-18630-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/16/2022] [Indexed: 12/01/2022] Open
Abstract
p38-MAPK is a stress-response kinase activated by hyperosmolarity. Here we interrogated the pathways involved. We show that p38-MAPK signaling is activated by hyperosmotic stimulation in various solutions, cell types and colonic organoids. Hyperosmolarity sensing is detected at the level of the upstream activators of p38-MAPK: TRAF2/ASK1 (but not Rac1) and MKK3/6/4. While WNK kinases are known osmo-sensors, we found, unexpectedly, that short (2 h) inhibition of WNKs (with WNK463) led to elevated p38-MAPK activity under hyperosmolarity, which was mediated by WNK463-dependent stimulation of TAK1 or TRAF2/ASK1, the upstream activators of MKK3/6/4. However, this effect was temporary and was reversed by long-term (2 days) incubation with WNK463. Accordingly, 2 days (but not 2 h) inhibition of p38-MAPK or its upstream activators ASK1 or TAK1, or WNKs, diminished regulatory volume increase (RVI) following cell shrinkage under hyperosmolarity. We also show that RVI mediated by the ion transporter NKCC1 is dependent on p38-MAPK. Since WNKs are known activators of NKCC1, we propose a WNK- > NKCC1- > p38-MAPK pathway that controls RVI. This pathway is augmented by NHE1. Additionally, hyperosmolarity inhibited mTORC1 activation and cell proliferation. Thus, activation of p38-MAPK and WNKs is important for RVI and for cell proliferation.
Collapse
Affiliation(s)
- Zetao Liu
- Cell Biology Program, The Hospital for Sick Children, PGCRL 19-9715, 686 Bay St., Toronto, ON, M5G 0A4, Canada
- Biochemistry Department, University of Toronto, Toronto, ON, Canada
| | - Wael Demian
- Cell Biology Program, The Hospital for Sick Children, PGCRL 19-9715, 686 Bay St., Toronto, ON, M5G 0A4, Canada
- Biochemistry Department, University of Toronto, Toronto, ON, Canada
| | - Avinash Persaud
- Cell Biology Program, The Hospital for Sick Children, PGCRL 19-9715, 686 Bay St., Toronto, ON, M5G 0A4, Canada
| | - Chong Jiang
- Cell Biology Program, The Hospital for Sick Children, PGCRL 19-9715, 686 Bay St., Toronto, ON, M5G 0A4, Canada
| | - Arohan R Subramanaya
- Department of Medicine and Cell Biology, University of Pittsburgh, Pittsburgh, USA
| | - Daniela Rotin
- Cell Biology Program, The Hospital for Sick Children, PGCRL 19-9715, 686 Bay St., Toronto, ON, M5G 0A4, Canada.
- Biochemistry Department, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
11
|
Chander Y, Kumar R, Verma A, Khandelwal N, Nagori H, Singh N, Sharma S, Pal Y, Puvar A, Pandit R, Shukla N, Chavada P, Tripathi BN, Barua S, Kumar N. Resistance evolution against host-directed antiviral agents: Buffalopox virus switches to use p38-ϒ under long-term selective pressure of an inhibitor targeting p38-α. Mol Biol Evol 2022; 39:6668988. [PMID: 35975687 PMCID: PMC9435063 DOI: 10.1093/molbev/msac177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Host-dependency factors have increasingly been targeted to minimize antiviral drug resistance. In this study, we have demonstrated that inhibition of p38 mitogen-activated protein kinase (a cellular protein) suppresses buffalopox virus (BPXV) protein synthesis by targeting p38-MNK1-eIF4E signaling pathway. In order to provide insights into the evolution of drug resistance, we selected resistant mutants by long-term sequential passages (P; n = 60) in the presence of p38 inhibitor (SB239063). The P60-SB239063 virus exhibited significant resistance to SB239063 as compared to the P60-Control virus. To provide mechanistic insights on the acquisition of resistance by BPXV-P60-SB239063, we generated p38-α and p38-ϒ (isoforms of p38) knockout Vero cells by CRISPR/Cas9-mediated genome editing. It was demonstrated that unlike the wild type (WT) virus which is dependent on p38-α isoform, the resistant virus (BPXV-P60-SB239063) switches over to use p38-ϒ so as to efficiently replicate in the target cells. This is a rare evidence wherein a virus was shown to bypass the dependency on a critical cellular factor under selective pressure of a drug.
Collapse
Affiliation(s)
- Yogesh Chander
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India.,Department of Bio and Nano Technology, Guru Jambeshwar University of Science and Technology, Hisar, Haryana, India
| | - Ram Kumar
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| | - Assim Verma
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India.,Department of Bio and Nano Technology, Guru Jambeshwar University of Science and Technology, Hisar, Haryana, India
| | - Nitin Khandelwal
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| | - Himanshu Nagori
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| | - Namita Singh
- Department of Bio and Nano Technology, Guru Jambeshwar University of Science and Technology, Hisar, Haryana, India
| | - Shalini Sharma
- Department of Veterinary Physiology and Biochemistry, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hiar, Haryana, India
| | - Yash Pal
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| | - Apurvasinh Puvar
- Gujarat Biotechnology Research Centre, Department of Science & Technology, Government of Gujarat, India
| | - Rameshchandra Pandit
- Gujarat Biotechnology Research Centre, Department of Science & Technology, Government of Gujarat, India
| | - Nitin Shukla
- Gujarat Biotechnology Research Centre, Department of Science & Technology, Government of Gujarat, India
| | - Priyank Chavada
- Gujarat Biotechnology Research Centre, Department of Science & Technology, Government of Gujarat, India
| | - Bhupendra N Tripathi
- Gujarat Biotechnology Research Centre, Department of Science & Technology, Government of Gujarat, India
| | - Sanjay Barua
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| | - Naveen Kumar
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| |
Collapse
|
12
|
Oladejo AO, Li Y, Imam BH, Ma X, Shen W, Wu X, Jiang W, Yang J, Lv Y, Ding X, Wang S, Yan Z. MicroRNA miR-24-3p Mediates the Negative Regulation of Lipopolysaccharide-Induced Endometrial Inflammatory Response by Targeting TNF Receptor-Associated Factor 6 (TRAF6). J Inflamm Res 2022; 15:807-825. [PMID: 35173455 PMCID: PMC8831117 DOI: 10.2147/jir.s347293] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/22/2021] [Indexed: 12/16/2022] Open
Abstract
Purpose Endometritis is a female reproductive disease that affects the cattle industries development and microRNAs (miRNAs) play a pivotal role and critical regulators of the innate immune response in varieties of diseases. The present study intends to investigate the regulatory role of miR‐24-3p in the innate immune response involved in endometritis and evaluate its therapeutic potential. Methods Whole mice uteri and bovine endometrial epithelial cells (BEECs) were separately stimulated with LPS. The BEECs were also transfected with miR-24-3p mimic and negative control; siTRAF6 and siNC; pcDNA3.1 empty and pcDNA3.1(+)TRAF6 separately with LPS stimulation. The expression levels of miR‐24-3p and TRAF6 were measured via quantitative real‐time polymerase chain reaction (qRT-PCR) and Western blot, respectively. LPS‐induced inflammatory response assessed by inflammatory cytokines secretion and expression via ELISA and qRT-PCR. Bioinformatics analysis and luciferase reporter assay validated the interaction between miR‐24-3p and TRAF6. The activation of the NF‐ĸB/MAPK pathway and p65 phosphorylation was investigated by Western blot and immunofluorescence assay, respectively. Results The expression of miR‐24-3p was decreased, and TRAF6 was elevated with an increased level of pro-inflammatory cytokines in LPS‐treated BEECs and mice uterus. The overexpression of miR‐24-3p suppressed LPS‐induced secretion of inflammatory cytokines (IL‐1β, IL‐6, IL-8 and TNF-α) and deactivation of NF‐ĸB/MAPK pathways. The downregulation of TRAF6 inhibited LPS‐induced inflammatory response in BEECs. TRAF6 is validated as a target of miR‐24-3p, and miR‐24-3p reversed the overexpression of cloned TRAF6 on inflammation response in BEECs. Conclusion Our findings demonstrate that the overexpression of miR‐24-3p attenuates endometrial inflammation and the expression of pro-inflammatory mediators via suppressing TRAF6. Therefore, modulating the pathogenesis of endometritis and possibly, a therapeutic potential against endometritis.
Collapse
Affiliation(s)
- Ayodele Olaolu Oladejo
- Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, 730050, People’s Republic of China
- Department of Animal Health Technology, Oyo State College of Agriculture and Technology, Igboora, 201103, Nigeria
| | - Yajuan Li
- Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, 730050, People’s Republic of China
| | - Bereket Habte Imam
- Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, 730050, People’s Republic of China
- Department of Veterinary Science, Hamelmalo Agricultural College, Keren, 397, Eritrea
| | - Xiaoyu Ma
- Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, 730050, People’s Republic of China
| | - Wenxiang Shen
- Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, 730050, People’s Republic of China
| | - Xiaohu Wu
- Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, 730050, People’s Republic of China
| | - Wei Jiang
- Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, 730050, People’s Republic of China
| | - Jie Yang
- Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, 730050, People’s Republic of China
| | - Yanan Lv
- Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, 730050, People’s Republic of China
| | - Xuezhi Ding
- Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, 730050, People’s Republic of China
| | - Shengyi Wang
- Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, 730050, People’s Republic of China
| | - Zuoting Yan
- Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, 730050, People’s Republic of China
- Correspondence: Zuoting Yan, Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, 730050, People’s Republic of China, Tel +8613919067215, Email
| |
Collapse
|
13
|
de Klerk DJ, de Keijzer MJ, Dias LM, Heemskerk J, de Haan LR, Kleijn TG, Franchi LP, Heger M. Strategies for Improving Photodynamic Therapy Through Pharmacological Modulation of the Immediate Early Stress Response. Methods Mol Biol 2022; 2451:405-480. [PMID: 35505025 DOI: 10.1007/978-1-0716-2099-1_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Photodynamic therapy (PDT) is a minimally to noninvasive treatment modality that has emerged as a promising alternative to conventional cancer treatments. PDT induces hyperoxidative stress and disrupts cellular homeostasis in photosensitized cancer cells, resulting in cell death and ultimately removal of the tumor. However, various survival pathways can be activated in sublethally afflicted cancer cells following PDT. The acute stress response is one of the known survival pathways in PDT, which is activated by reactive oxygen species and signals via ASK-1 (directly) or via TNFR (indirectly). The acute stress response can activate various other survival pathways that may entail antioxidant, pro-inflammatory, angiogenic, and proteotoxic stress responses that culminate in the cancer cell's ability to cope with redox stress and oxidative damage. This review provides an overview of the immediate early stress response in the context of PDT, mechanisms of activation by PDT, and molecular intervention strategies aimed at inhibiting survival signaling and improving PDT outcome.
Collapse
Affiliation(s)
- Daniel J de Klerk
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Mark J de Keijzer
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Lionel M Dias
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Faculdade de Ciências da Saúde (FCS-UBI), Universidade da Beira Interior, Covilhã, Portugal
| | - Jordi Heemskerk
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
| | - Lianne R de Haan
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Tony G Kleijn
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Leonardo P Franchi
- Departamento de Bioquímica e Biologia Molecular, Instituto de Ciências Biológicas (ICB) 2, Universidade Federal de Goiás (UFG), Goiânia, GO, Brazil
- Faculty of Philosophy, Department of Chemistry, Center of Nanotechnology and Tissue Engineering-Photobiology and Photomedicine Research Group, Sciences, and Letters of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Michal Heger
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China.
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands.
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
14
|
Zhang Y, Xiao F, Zhou Q, Diao T, Zhang M, Liu D, Wang Z, Huang T, Wu Y, Bai Y, Min Q. The Potential Protective Effect of Iridoid Glycosides Isolated From Osmanthus fragrans Seeds Against the Development of Immune Liver Injury in Mice. Front Pharmacol 2021; 12:760338. [PMID: 34819861 PMCID: PMC8606819 DOI: 10.3389/fphar.2021.760338] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/12/2021] [Indexed: 12/04/2022] Open
Abstract
Objective: The iridoid glycosides were extracted and separated from Osmanthus fragrans seeds, and the potential protective effect of Osmanthus fragrans seed extract on concanavalin A-induced immune liver injury in mice was studied. Methods:Osmanthus fragrans seeds were extracted by 95% ethanol reflux. Then, the iridoid glycosides were enriched by extraction refined through petroleum ether (60°C–90°C), ethyl acetate, and water-saturated n-butanol in sequence, so as to purify the n-butanol part (Osmanthus fragrans seed’s n-butanol extraction, OFSN) by macroporous resin. Specnuezhenide and Nuezhenoside G13 were used as the reference substances to determine the concentration of iridoid glycosides by HPLC. On this basis, a mouse immune liver injury model was established by tail intravenous concanavalin A (20 mg/kg); the contents of serum ALT, AST, IFN-γ, and TNF-α and the contents of liver tissue MDA and SOD were determined; the pathological changes of the liver by HE staining were observed; and the expression levels of p38MAPK and p-p38mapk in liver tissue were detected by WB. Results: The linearity, precision, repeatability, recovery, and stability of HPLC all met the requirements by validating with the methodology. The contents of Specnuezhenide and Nuezhenoside G13 in the n-butanol extracts were 39.20% and 39.88%, respectively. Actually, their contents can reach up to 82.56% and 87.9% after being purified by macroporous resin. The results of animal experiments show that OFSN could significantly reduce the liver and spleen index, reduce the ALT and AST contents in plasma and the MDA content in liver tissue, and then increase the SOD content. Besides, OFSN could also reduce the plasma IFN-γ and TNF-α levels. The HE staining result indicates that the pathological changes in the liver tissues of mice treated with OFSN are alleviated to different degrees while the WB result suggests that OFSN could significantly inhibit the expression of p-p38mapk. Conclusion:Osmanthus fragrans seeds are rich in iridoid glycosides, which has a good protective effect on mouse immune liver injury caused by concanavalin A. The mechanism may be related to inhibiting the phosphorylation of p38MAPK, inhibiting the release of inflammatory mediators, improving the antioxidant capacity of liver cells, and weakening the occurrence of lipid peroxidation.
Collapse
Affiliation(s)
- Yuchen Zhang
- School of Pharmcy, Hubei University of Science and Technology, Xianning, China
| | - Feng Xiao
- School of Pharmcy, Hubei University of Science and Technology, Xianning, China
| | - Qiqi Zhou
- School of Pharmcy, Hubei University of Science and Technology, Xianning, China
| | - Tingting Diao
- School of Biological and Pharmaceutical Engineering, Xinyang Agricultural and Forestry University, Xinyang, China
| | - Meng Zhang
- School of Pharmcy, Hubei University of Science and Technology, Xianning, China
| | - Dongyang Liu
- School of Pharmcy, Hubei University of Science and Technology, Xianning, China
| | - Zhuowen Wang
- School of Pharmcy, Hubei University of Science and Technology, Xianning, China
| | - Ting Huang
- School of Pharmcy, Hubei University of Science and Technology, Xianning, China
| | - Yupei Wu
- School of Pharmcy, Hubei University of Science and Technology, Xianning, China
| | - Yuting Bai
- School of Pharmcy, Hubei University of Science and Technology, Xianning, China
| | - Qing Min
- School of Pharmcy, Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
15
|
Xu W, Liu R, Dai Y, Hong S, Dong H, Wang H. The Role of p38γ in Cancer: From review to outlook. Int J Biol Sci 2021; 17:4036-4046. [PMID: 34671218 PMCID: PMC8495394 DOI: 10.7150/ijbs.63537] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/16/2021] [Indexed: 01/20/2023] Open
Abstract
p38γ is a member of the p38 Mitogen Activated Protein Kinases (p38 MAPKs). It contains four subtypes in mammalian cells encoded by different genes including p38α (MAPK14), p38β (MAPK11), p38γ (MAPK12), and p38δ (MAPK13). Recent studies revealed that p38γ may exhibit a crucial role in tumorigenesis and cancer aggressiveness. Despite the large number of published literatures, further researches are demanded to clarify its role in cancer development, the tissue-specific function and associated novel treatment strategies. In this article, we provide the latest view on the connection between p38γ and malignant tumors, highlighting the function of p38γ. The clinical value of p38γ is also discussed, helping the translation into the remarkable therapeutic strategy in tumor diseases.
Collapse
Affiliation(s)
- Wentao Xu
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.,First Clinical Medical College of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Rui Liu
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Ying Dai
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Shaocheng Hong
- First Clinical Medical College of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Huke Dong
- First Clinical Medical College of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, Anhui, China
| |
Collapse
|
16
|
Zhao J, Yin L, Jiang L, Hou L, He L, Zhang C. PTEN nuclear translocation enhances neuronal injury after hypoxia-ischemia via modulation of the nuclear factor-κB signaling pathway. Aging (Albany NY) 2021; 13:16165-16177. [PMID: 34114972 PMCID: PMC8266328 DOI: 10.18632/aging.203141] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 04/29/2021] [Indexed: 11/25/2022]
Abstract
The occurrence of hypoxia-ischemia (HI) in the developing brain is closely associated with neuronal injury and even death. However, the underlying molecular mechanism is not fully understood. This study was designed to investigate phosphatase and tensin homolog (PTEN) nuclear translocation and its possible role in rat cortical neuronal damage following oxygen-glucose deprivation (OGD) in vitro. An in vitro OGD model was established using primary cortical neurons dissected from newborn Sprague-Dawley rats to mimic HI conditions. The PTENK13R mutant plasmid, which contains a lysine-to-arginine mutation at the lysine 13 residue, was constructed. The nuclei and cytoplasm of neurons were separated. Neuronal injury following OGD was evidenced by increased lactate dehydrogenase (LDH) release and apoptotic cell counts. In addition, PTEN expression was increased and the phosphorylation of extracellular signal-regulated kinase 1/2 (p-ERK1/2) and activation of nuclear factor kappa B (NF-κB) were decreased following OGD. PTENK13R transfection prevented PTEN nuclear translocation; attenuated the effect of OGD on nuclear p-ERK1/2 and NF-κB, apoptosis, and LDH release; and increased the expression of several anti-apoptotic proteins. We conclude that PTEN nuclear translocation plays an essential role in neuronal injury following OGD via modulation of the p-ERK1/2 and NF-κB pathways. Prevention of PTEN nuclear translocation might be a candidate strategy for preventing brain injury following HI.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Linlin Yin
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Lin Jiang
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Li Hou
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Ling He
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Chunyan Zhang
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| |
Collapse
|
17
|
Anton DB, Ducati RG, Timmers LFSM, Laufer S, Goettert MI. A Special View of What Was Almost Forgotten: p38δ MAPK. Cancers (Basel) 2021; 13:2077. [PMID: 33923030 PMCID: PMC8123357 DOI: 10.3390/cancers13092077] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 12/22/2022] Open
Abstract
The p38δ mitogen-activated protein kinase is an important signal transduction enzyme. p38δ has recently emerged as a drug target due to its tissue-specific expression patterns and its critical roles in regulation of cellular processes related to cancer and inflammatory diseases, such as cell proliferation, cell migration, apoptosis, and inflammatory responses. However, potent and specific p38δ inhibitors have not been defined so far. Moreover, in cancer disease, p38δ appears to act as a tumor suppressor or tumor promoter according to cancer and cell type studied. In this review, we outline the current understanding of p38δ roles in each cancer type, to define whether it is possible to delineate new cancer therapies based on small-molecule p38δ inhibitors. We also highlight recent advances made in the design of molecules with potential to inhibit p38 isoforms and discuss structural approaches to guide the search for p38δ inhibitors.
Collapse
Affiliation(s)
- Débora Bublitz Anton
- Biotechnology Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado, Rio Grande do Sul CEP 95914-014, Brazil; (D.B.A.); (R.G.D.); (L.F.S.M.T.)
| | - Rodrigo Gay Ducati
- Biotechnology Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado, Rio Grande do Sul CEP 95914-014, Brazil; (D.B.A.); (R.G.D.); (L.F.S.M.T.)
| | - Luís Fernando Saraiva Macedo Timmers
- Biotechnology Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado, Rio Grande do Sul CEP 95914-014, Brazil; (D.B.A.); (R.G.D.); (L.F.S.M.T.)
- Medical Science Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado, Rio Grande do Sul CEP 95914-014, Brazil
| | - Stefan Laufer
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Faculty of Sciences, University of Tuebingen, D-72076 Tuebingen, Germany
| | - Márcia Inês Goettert
- Biotechnology Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado, Rio Grande do Sul CEP 95914-014, Brazil; (D.B.A.); (R.G.D.); (L.F.S.M.T.)
- Medical Science Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado, Rio Grande do Sul CEP 95914-014, Brazil
| |
Collapse
|
18
|
Cicuéndez B, Ruiz-Garrido I, Mora A, Sabio G. Stress kinases in the development of liver steatosis and hepatocellular carcinoma. Mol Metab 2021; 50:101190. [PMID: 33588102 PMCID: PMC8324677 DOI: 10.1016/j.molmet.2021.101190] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/31/2020] [Accepted: 02/09/2021] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is an important component of metabolic syndrome and one of the most prevalent liver diseases worldwide. This disorder is closely linked to hepatic insulin resistance, lipotoxicity, and inflammation. Although the mechanisms that cause steatosis and chronic liver injury in NAFLD remain unclear, a key component of this process is the activation of stress-activated kinases (SAPKs), including p38 and JNK in the liver and immune system. This review summarizes findings which indicate that the dysregulation of stress kinases plays a fundamental role in the development of steatosis and are important players in inducing liver fibrosis. To avoid the development of steatohepatitis and liver cancer, SAPK activity must be tightly regulated not only in the hepatocytes but also in other tissues, including cells of the immune system. Possible cellular mechanisms of SAPK actions are discussed. Hepatic JNK triggers steatosis and insulin resistance, decreasing lipid oxidation and ketogenesis in HFD-fed mice. Decreased liver expression of p38α/β in HFD increases lipogenesis. Hepatic p38γ/δ drive insulin resistance and inhibit autophagy, which may lead to steatosis. Macrophage p38α/β promote cytokine production and M1 polarization, leading to lipid accumulation in hepatocytes. Myeloid p38γ/δ contribute to cytokine production and neutrophil migration, protecting against steatosis, diabetes and NAFLD. JNK1 and p38γ induce HCC while p38α blocks it. However, deletion of hepatic JNK1/2 induces cholangiocarcinoma. SAPK are potential therapeutic target for metabolic disorders, steatohepatitis and liver cancer.
Collapse
Affiliation(s)
- Beatriz Cicuéndez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Irene Ruiz-Garrido
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Alfonso Mora
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain.
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain.
| |
Collapse
|
19
|
Ittner A, Asih PR, Tan ARP, Prikas E, Bertz J, Stefanoska K, Lin Y, Volkerling AM, Ke YD, Delerue F, Ittner LM. Reduction of advanced tau-mediated memory deficits by the MAP kinase p38γ. Acta Neuropathol 2020; 140:279-294. [PMID: 32725265 DOI: 10.1007/s00401-020-02191-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 07/02/2020] [Accepted: 07/02/2020] [Indexed: 01/12/2023]
Abstract
Hyperphosphorylation of the neuronal tau protein contributes to Alzheimer's disease (AD) by promoting tau pathology and neuronal and cognitive deficits. In contrast, we have previously shown that site-specific tau phosphorylation can inhibit toxic signals induced by amyloid-β (Aβ) in mouse models. The post-synaptic mitogen-activated protein (MAP) kinase p38γ mediates this site-specific phosphorylation on tau at Threonine-205 (T205). Using a gene therapeutic approach, we draw on this neuroprotective mechanism to improve memory in two Aβ-dependent mouse models of AD at stages when advanced memory deficits are present. Increasing activity of post-synaptic kinase p38γ that targets T205 in tau reduced memory deficits in symptomatic Aβ-induced AD models. Reconstitution experiments with wildtype human tau or phosphorylation-deficient tauT205A showed that T205 modification is critical for downstream effects of p38γ that prevent memory impairment in APP-transgenic mice. Furthermore, genome editing of the T205 codon in the murine Mapt gene showed that this single side chain in endogenous tau critically modulates memory deficits in APP-transgenic Alzheimer's mice. Ablating the protective effect of p38γ activity by genetic p38γ deletion in a tau transgenic mouse model that expresses non-pathogenic tau rendered tau toxic and resulted in impaired memory function in the absence of human Aβ. Thus, we propose that modulating neuronal p38γ activity serves as an intrinsic tau-dependent therapeutic approach to augment compromised cognition in advanced dementia.
Collapse
|
20
|
Blanc RS, Kallenbach JG, Bachman JF, Mitchell A, Paris ND, Chakkalakal JV. Inhibition of inflammatory CCR2 signaling promotes aged muscle regeneration and strength recovery after injury. Nat Commun 2020; 11:4167. [PMID: 32820177 PMCID: PMC7441393 DOI: 10.1038/s41467-020-17620-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 07/06/2020] [Indexed: 02/07/2023] Open
Abstract
Muscle regeneration depends on a robust albeit transient inflammatory response. Persistent inflammation is a feature of age-related regenerative deficits, yet the underlying mechanisms are poorly understood. Here, we find inflammatory-related CC-chemokine-receptor 2 (Ccr2) expression in non-hematopoietic myogenic progenitors (MPs) during regeneration. After injury, the expression of Ccr2 in MPs corresponds to the levels of its ligands, the chemokines Ccl2, 7, and 8. We find stimulation of Ccr2-activity inhibits MP fusion and contribution to myofibers. This occurs in association with increases in MAPKp38δ/γ signaling, MyoD phosphorylation, and repression of the terminal myogenic commitment factor Myogenin. High levels of Ccr2-chemokines are a feature of regenerating aged muscle. Correspondingly, deletion of Ccr2 in MPs is necessary for proper fusion into regenerating aged muscle. Finally, opportune Ccr2 inhibition after injury enhances aged regeneration and functional recovery. These results demonstrate that inflammatory-induced activation of Ccr2 signaling in myogenic cells contributes to aged muscle regenerative decline. Chronic inflammation is a feature of age-related regenerative decline in skeletal muscles, but how it directly affects resident muscle stem cell fate and function is unclear. Here, the authors show that Ccr2 signaling in muscle stem cell derived progenitors represses terminal myogenic differentiation, and that targeting Ccr2 on aged myogenic progenitors rejuvenates aged skeletal muscle healing and function.
Collapse
Affiliation(s)
- Roméo S Blanc
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.,Wilmot Cancer Institute, Stem Cell and Regenerative Medicine Institute, and The Rochester Aging Research Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Jacob G Kallenbach
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.,Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
| | - John F Bachman
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.,Department of Pathology and Laboratory Medicine, Cell Biology of Disease Graduate Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Amanda Mitchell
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Nicole D Paris
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.,Wilmot Cancer Institute, Stem Cell and Regenerative Medicine Institute, and The Rochester Aging Research Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Joe V Chakkalakal
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA. .,Wilmot Cancer Institute, Stem Cell and Regenerative Medicine Institute, and The Rochester Aging Research Center, University of Rochester Medical Center, Rochester, NY, USA. .,Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
21
|
Regulation of the Mammalian SWI/SNF Family of Chromatin Remodeling Enzymes by Phosphorylation during Myogenesis. BIOLOGY 2020; 9:biology9070152. [PMID: 32635263 PMCID: PMC7407365 DOI: 10.3390/biology9070152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/24/2020] [Accepted: 07/01/2020] [Indexed: 11/16/2022]
Abstract
Myogenesis is the biological process by which skeletal muscle tissue forms. Regulation of myogenesis involves a variety of conventional, epigenetic, and epigenomic mechanisms that control chromatin remodeling, DNA methylation, histone modification, and activation of transcription factors. Chromatin remodeling enzymes utilize ATP hydrolysis to alter nucleosome structure and/or positioning. The mammalian SWItch/Sucrose Non-Fermentable (mSWI/SNF) family of chromatin remodeling enzymes is essential for myogenesis. Here we review diverse and novel mechanisms of regulation of mSWI/SNF enzymes by kinases and phosphatases. The integration of classic signaling pathways with chromatin remodeling enzyme function impacts myoblast viability and proliferation as well as differentiation. Regulated processes include the assembly of the mSWI/SNF enzyme complex, choice of subunits to be incorporated into the complex, and sub-nuclear localization of enzyme subunits. Together these processes influence the chromatin remodeling and gene expression events that control myoblast function and the induction of tissue-specific genes during differentiation.
Collapse
|
22
|
Wei TH, Hsieh CL. Effect of Acupuncture on the p38 Signaling Pathway in Several Nervous System Diseases: A Systematic Review. Int J Mol Sci 2020; 21:E4693. [PMID: 32630156 PMCID: PMC7370084 DOI: 10.3390/ijms21134693] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 06/26/2020] [Accepted: 06/28/2020] [Indexed: 12/16/2022] Open
Abstract
Acupuncture is clinically used to treat various diseases and exerts positive local and systemic effects in several nervous system diseases. Advanced molecular and clinical studies have continually attempted to decipher the mechanisms underlying these effects of acupuncture. While a growing understanding of the pathophysiology underlying several nervous system diseases shows it to be related to inflammation and impair cell regeneration after ischemic events, the relationship between the therapeutic mechanism of acupuncture and the p38 MAPK signal pathway has yet to be elucidated. This review discusses the latest advancements in the identification of the effect of acupuncture on the p38 signaling pathway in several nervous system diseases. We electronically searched databases including PubMed, Embase, and the Cochrane Library from their inception to April 2020, using the following keywords alone or in various combinations: "acupuncture", "p38 MAPK pathway", "signaling", "stress response", "inflammation", "immune", "pain", "analgesic", "cerebral ischemic injury", "epilepsy", "Alzheimer's disease", "Parkinson's disease", "dementia", "degenerative", and "homeostasis". Manual acupuncture and electroacupuncture confer positive therapeutic effects by regulating proinflammatory cytokines, ion channels, scaffold proteins, and transcription factors including TRPV1/4, Nav, BDNF, and NADMR1; consequently, p38 regulates various phenomena including cell communication, remodeling, regeneration, and gene expression. In this review article, we found the most common acupoints for the relief of nervous system disorders including GV20, GV14, ST36, ST37, and LI4. Acupuncture exhibits dual regulatory functions of activating or inhibiting different p38 MAPK pathways, contributing to an overall improvement of clinical symptoms and function in several nervous system diseases.
Collapse
Affiliation(s)
- Tzu-Hsuan Wei
- Department of Chinese Medicine, China Medical University Hospital, Taichung 40447, Taiwan;
| | - Ching-Liang Hsieh
- Department of Chinese Medicine, China Medical University Hospital, Taichung 40447, Taiwan;
- Chinese Medicine Research Center, China Medical University, Taichung 40402, Taiwan
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
23
|
Antiprolactinoma Effect of Hordenine by Inhibiting MAPK Signaling Pathway Activation in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:3107290. [PMID: 32382283 PMCID: PMC7195642 DOI: 10.1155/2020/3107290] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 02/20/2020] [Accepted: 03/09/2020] [Indexed: 11/17/2022]
Abstract
Prolactinomas are harmful to human health, and the clinical first-line treatment drug is bromocriptine. However, 20% prolactinomas patients did not respond to bromocriptine. Hordenine is an alkaloid separated from Fructus Hordei Germinatus, which showed significant antihyperprolactinemia activity in rats. The aim of this study was to explore the effect and mechanism of hordenine on prolactinomas in rats. The study used estradiol to induce prolactinomas, which caused the activation of the pituitary mitogen-activated protein kinase (MAPK) pathway in rats significantly. The treatment of hordenine restored estradiol, induced the overgrowth of pituitary gland, and reduced the prolactin (PRL) accumulation in the serum and pituitary gland of rats by blocking the MAPK (p38, ERK1/2, and JNK) activation and production of inflammatory cytokines, tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6). The antiprolactinoma effect of hordenine was mediated by inhibiting the MAPK signaling pathway activation in rats.
Collapse
|
24
|
Li Z, Liu FY, Kirkwood KL. The p38/MKP-1 signaling axis in oral cancer: Impact of tumor-associated macrophages. Oral Oncol 2020; 103:104591. [PMID: 32058294 PMCID: PMC7136140 DOI: 10.1016/j.oraloncology.2020.104591] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 02/04/2020] [Indexed: 02/07/2023]
Abstract
Oral squamous cell carcinomas (OSCC) constitute over 95% of all head and neck malignancies. As a key component of the tumor microenvironment (TME), chronic inflammation contributes towards the development, progression, and regional metastasis of OSCC. Tumor associated macrophages (TAMs) associated with OSSC promote tumorigenesis through the production of cytokines and pro-inflammatory factors that are critical role in the various steps of malignant transformation, including tumor growth, survival, invasion, angiogenesis, and metastasis. The mitogen-activated protein kinases (MAPKs) can regulate inflammation along with a wide range of cellular processes including cell metabolism, proliferation, motility, apoptosis, survival, differentiation and play a crucial role in cell growth and survival in physiological and pathological processes including innate and adaptive immune responses. Dual specificity MAPK phosphatases (MKPs) deactivates MAPKs. MKPs are considered as an important feedback control mechanism that limits MAPK signaling and subsequent target gene expression. This review outlines the role of MKP-1, the founding member of the MKP family, in OSCC and the TME. Herein, we summarize recent progress in understanding the regulation of p38 MAPK/MKP-1 signaling pathways via TAM-related immune responses in OSCC development, progression and treatment outcomes.
Collapse
Affiliation(s)
- Zhenning Li
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, China
- Department of Medical Genetics, China Medical University, Shenyang, China
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, USA
| | - Fa-yu Liu
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, China
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, USA
| | - Keith L. Kirkwood
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, USA
- Department of Head and Neck/Plastic and Reconstructive Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
25
|
Ewendt F, Föller M. p38MAPK controls fibroblast growth factor 23 (FGF23) synthesis in UMR106-osteoblast-like cells and in IDG-SW3 osteocytes. J Endocrinol Invest 2019; 42:1477-1483. [PMID: 31201665 DOI: 10.1007/s40618-019-01073-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 06/10/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND p38 mitogen-activated protein kinase (p38MAPK) is a serine/threonine kinase activated by cellular stress stimuli including radiation, osmotic shock, and inflammation and influencing apoptosis, cell proliferation, and autophagy. Moreover, p38MAPK induces transcriptional activity of the transcription factor complex NFκB mediating multiple pro-inflammatory cellular responses. Fibroblast growth factor 23 (FGF23) is produced by bone cells, and regulates renal phosphate and vitamin D metabolism as a hormone. FGF23 expression is enhanced by NFκB. Here, we analyzed the relevance of p38MAPK activity for the production of FGF23. METHODS Fgf23 expression was analyzed by qRT-PCR and FGF23 protein by ELISA in UMR106 osteoblast-like cells and in IDG-SW3 osteocytes. RESULTS Inhibition of p38MAPK with SB203580 or SB202190 significantly down-regulated Fgf23 expression and FGF23 protein expression. Conversely, p38MAPK activator anisomycin increased the abundance of Fgf23 mRNA. NFκB inhibitors wogonin and withaferin A abrogated the stimulatory effect of anisomycin on Fgf23 gene expression. CONCLUSION p38MAPK induces FGF23 formation, an effect at least in part dependent on NFκB activity.
Collapse
Affiliation(s)
- F Ewendt
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, 06120, Halle (Saale), Germany
| | - M Föller
- Institute of Physiology, University of Hohenheim, Garbenstraße 30, 70599, Stuttgart, Germany.
| |
Collapse
|
26
|
Ng CT, Fong LY, Yong YK, Hakim MN, Ahmad Z. Interferon-γ induces biphasic changes in caldesmon localization as well as adherens junction organization and expression in HUVECs. Cytokine 2018; 111:541-550. [PMID: 29909980 DOI: 10.1016/j.cyto.2018.06.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 06/06/2018] [Accepted: 06/08/2018] [Indexed: 01/11/2023]
Abstract
Endothelial barrier dysfunction leads to increased endothelial permeability and is an early step in the development of vascular inflammatory diseases such as atherosclerosis. Interferon-γ (IFN-γ), a proinflammatory cytokine, is known to cause increased endothelial permeability. However, the mechanisms by which IFN-γ disrupts the endothelial barrier have not been clarified. This study aimed to investigate how IFN-γ impairs the endothelial barrier integrity by specifically examining the roles of caldesmon, adherens junctions (AJs) and p38 mitogen-activated protein (MAP) kinase in IFN-γ-induced endothelial barrier dysfunction. IFN-γ exhibited a biphasic effect on caldesmon localization and both the structural organization and protein expression of AJs. In the early phase (4-8 h), IFN-γ induced the formation of peripheral caldesmon bands and discontinuous AJs, while AJ protein expression was unchanged. Interestingly, IFN-γ also stimulated caldesmon phosphorylation, resulting in actin dissociation from caldesmon at 8 h. Conversely, changes seen in the late phase (16-24 h) included cytoplasmic caldesmon dispersal, AJ linearization and junctional area reduction, which were associated with reduced membrane, cytoskeletal and total AJ protein expression. In addition, IFN-γ enhanced myosin binding to caldesmon at 12 h and persisted up to 24 h. Furthermore, inhibition of p38 MAP kinase by SB203580 did not reverse either the early or late phase changes observed. These data suggest that IFN-γ may activate signaling molecules other than p38 MAP kinase. In conclusion, our findings enhance the current understanding of how IFN-γ disrupts endothelial barrier function and reveal potential therapeutic targets, such as caldesmon and AJs, for the treatment of IFN-γ-associated vascular inflammatory diseases.
Collapse
Affiliation(s)
- Chin Theng Ng
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia; Physiology Unit, Faculty of Medicine, AIMST University, 08100 Bedong, Kedah, Malaysia.
| | - Lai Yen Fong
- Department of Pre-clinical Sciences, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, 43000 Kajang, Selangor, Malaysia.
| | - Yoke Keong Yong
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia.
| | - Muhammad Nazrul Hakim
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia.
| | - Zuraini Ahmad
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| |
Collapse
|
27
|
Roy S, Roy S, Rana A, Akhter Y, Hande MP, Banerjee B. The role of p38 MAPK pathway in p53 compromised state and telomere mediated DNA damage response. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2018; 836:89-97. [PMID: 30389168 DOI: 10.1016/j.mrgentox.2018.05.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 04/17/2018] [Accepted: 05/26/2018] [Indexed: 12/19/2022]
Abstract
There is an intricate balance of DNA damage response and repair which determines the homeostasis of human genome function. p53 protein is widely known for its role in cell cycle regulation and tumor suppressor activity. In case of several cancers where function of p53 gene gets compromised either by mutation or partial inactivation, the role of p53 in response to DNA damage needs to be supplemented by another molecule or pathway. Due to sedentary lifestyle and exposure to genotoxic agents, genome is predisposed to chronic stress, which ultimately leads to unrepaired or background DNA damage. p38 MAPK signaling pathway is strongly activated in response to various environmental and cellular stresses. DNA damage response and the repair options have crucial links with chromosomal integrity. Telomere that regulates integrity of genome is protected by a six member shielding unit called shelterin complex which communicates with other pathways for functionality of telomeres. There are evidences that p38 gets activated through ATM in response to DNA damage. Dysfunctional telomere leads to activation of ATM which subsequently activates p38 suggesting a crosstalk between p38, ATM and shelterin complex. This review focuses on activation of p38 in response to genotoxic stress induced DNA damage in p53 mutated or compromised state and its possible cross talk with telomere shelterin proteins. Thus p38 may act as an important target to treat various diseases and in majority of cancers in p53 mutated state.
Collapse
Affiliation(s)
- Shomereeta Roy
- Molecular Stress and Stem Cell Biology Group, School of Biotechnology, KIIT University, Bhubaneswar, Odisha-751024, India
| | - Souvick Roy
- Molecular Stress and Stem Cell Biology Group, School of Biotechnology, KIIT University, Bhubaneswar, Odisha-751024, India
| | - Aarti Rana
- Centre for Computational Biology and Bioinformatics, School of Life Sciences, Central University of Himachal Pradesh, Shahpur, Himachal Pradesh-176206, India
| | - Yusuf Akhter
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025, India
| | - Manoor Prakash Hande
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Birendranath Banerjee
- Molecular Stress and Stem Cell Biology Group, School of Biotechnology, KIIT University, Bhubaneswar, Odisha-751024, India.
| |
Collapse
|
28
|
Sahu V, Nigam L, Agnihotri V, Gupta A, Shekhar S, Subbarao N, Bhaskar S, Dey S. Diagnostic Significance of p38 Isoforms (p38α, p38β, p38γ, p38δ) in Head and Neck Squamous Cell Carcinoma: Comparative Serum Level Evaluation and Design of Novel Peptide Inhibitor Targeting the Same. Cancer Res Treat 2018; 51:313-325. [PMID: 29747487 PMCID: PMC6333999 DOI: 10.4143/crt.2018.105] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 05/04/2018] [Indexed: 01/12/2023] Open
Abstract
Purpose The p38 mitogen-activated protein kinase (MAPKs) play a crucial role in the production of pro-inflammatory cytokines and over-expression of it increase cytokines which promote cancer. Among four isoforms, p38α has been well studied in head and neck squamous cell carcinoma (HNSCC) and other cancers as a therapeutic target. p38δ has recently emerged as a potential disease-specific drug target. Elevated serum p38α level in HNSCC was reported earlier from our lab. This study aims to estimate the levels of p38 MAPK-isoforms in the serum of HNSCC and design peptide inhibitor targeting the same. Materials and Methods Levels of p38 MAPK isoforms in the serum of HNSCC and healthy controls were quantified by surface plasmon resonance technology. The peptide inhibitor for p38 MAPK was designed by molecular modeling using Grid-based Ligand Docking with Energetics tools and compared with known specific inhibitors. Results We have observed highly elevated levels of all four isoforms of p38 MAPK in serum of HNSCC patients compared to the control group. Further, serum p38α, p38β, and p38δ levels were down regulated after therapy in follow-up patients, while p38γ showed no response to the therapy. Present study screened designed peptide WFYH as a specific inhibitor against p38δ. The specific inhibitor of p38δ was found to have no effect on p38α due to great structural difference at ATP binding pocket. Conclusion In this study, first time estimated the levels of p38 MAPK isoforms in the serum of HNSCC. It can be concluded that p38 MAPK isoforms can be a diagnostic and prognostic marker for HNSCC and p38δ as a therapeutic target.
Collapse
Affiliation(s)
- Vishal Sahu
- Department of Biophysics, All India Institute of Medical Sciences, Ansari Nagar, India
| | - Lokesh Nigam
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Vertica Agnihotri
- Department of Biophysics, All India Institute of Medical Sciences, Ansari Nagar, India
| | - Abhishek Gupta
- Department of Biophysics, All India Institute of Medical Sciences, Ansari Nagar, India
| | - Shashank Shekhar
- Department of Biophysics, All India Institute of Medical Sciences, Ansari Nagar, India
| | - Naidu Subbarao
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Suman Bhaskar
- Department of Radiation Oncology, All India Institute of Medical Sciences, Ansari Nagar, India
| | - Sharmistha Dey
- Department of Biophysics, All India Institute of Medical Sciences, Ansari Nagar, India
| |
Collapse
|
29
|
Inactivation of p38 MAPK contributes to stem cell-like properties of non-small cell lung cancer. Oncotarget 2018; 8:26702-26717. [PMID: 28460458 PMCID: PMC5432291 DOI: 10.18632/oncotarget.15804] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 02/15/2017] [Indexed: 01/07/2023] Open
Abstract
Cancer stem cells (CSCs) are recognized as the major source for cancer initiation and recurrence. Yet, the mechanism by which the cancer stem cell properties are acquired and maintained in a cancer cell population is not well understood. In the current study, we observed that the level of active p38 MAPK is downregulated, while the level of the stemness marker SOX2 is upregulated in lung cancer tissues as compared to normal tissues. We further demonstrated that inactivation of p38 is a potential mechanism contributing to acquisition and maintenance of cancer stem cell properties in non-small cell lung cancer (NSCLC) cells. p38, in particular the p38γ and p38δ isoforms, suppresses the cancer stem cell properties and tumor initiating ability of NSCLC cells by promoting the ubiquitylation and degradation of stemness proteins such as SOX2, Oct4, Nanog, Klf4 and c-Myc, through MK2-mediated phosphorylation of Hsp27 that is an essential component of the proteasomal degradation machinery. In contrast, inactivation of p38 in lung cancer cells leads to upregulation of the stemness proteins, thus promoting the cancer stem cell properties of these cells. These findings have demonstrated a novel mechanism by which cancer stem cell properties are acquired and maintained in a cancer cell population, and have revealed a new function of the p38 pathway in suppressing cancer development. These studies have also identified a new pathway that can potentially serve as a target for cancer therapies aimed at eliminating CSCs.
Collapse
|
30
|
Ruiz M, Coderre L, Allen BG, Des Rosiers C. Protecting the heart through MK2 modulation, toward a role in diabetic cardiomyopathy and lipid metabolism. Biochim Biophys Acta Mol Basis Dis 2017; 1864:1914-1922. [PMID: 28735097 DOI: 10.1016/j.bbadis.2017.07.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 07/11/2017] [Accepted: 07/14/2017] [Indexed: 12/20/2022]
Abstract
Various signaling pathways have been identified in the heart as important players during development, physiological adaptation or pathological processes. This includes the MAPK families, particularly p38MAPK, which is involved in several key cellular processes, including differentiation, proliferation, apoptosis, inflammation, metabolism and survival. Disrupted p38MAPK signaling has been associated with several diseases, including cardiovascular diseases (CVD) as well as diabetes and its related complications. Despite efforts to translate this knowledge into therapeutic avenues, p38 inhibitors have failed in clinical trials due to adverse effects. Inhibition of MK2, a downstream target of p38, appears to be a promising alternative strategy. Targeting MK2 activity may avoid the adverse effects linked to p38 inhibition, while maintaining its beneficial effects. MK2 was first considered as a therapeutic target in inflammatory diseases such as rheumatoid polyarthritis. A growing body of evidence now supports a key role of MK2 signaling in the pathogenesis of CVD, particularly ischemia/reperfusion injury, hypertrophy, and hypertension and that its inhibition or inactivation is associated with improved heart and vascular functions. More recently, MK2 was shown to be a potential player in diabetes and related complications, particularly in liver and heart, and perturbations in calcium handling and lipid metabolism. In this review, we will discuss recent advances in our knowledge of the role of MK2 in p38MAPK-mediated signaling and the benefits of its loss of function in CVD and diabetes, with an emphasis on the roles of MK2 in calcium handling and lipid metabolism. This article is part of a Special issue entitled Cardiac adaptations to obesity, diabetes and insulin resistance, edited by Professors Jan F.C. Glatz, Jason R.B. Dyck and Christine Des Rosiers.
Collapse
Affiliation(s)
- Matthieu Ruiz
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada; Montreal Heart Institute, Research Center, 5000 Belanger Street, Montreal, Quebec, Canada
| | - Lise Coderre
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; Montreal Heart Institute, Research Center, 5000 Belanger Street, Montreal, Quebec, Canada
| | - Bruce Gordon Allen
- Department of Biochemistry, Université de Montréal, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; Montreal Heart Institute, Research Center, 5000 Belanger Street, Montreal, Quebec, Canada.
| | - Christine Des Rosiers
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; Montreal Heart Institute, Research Center, 5000 Belanger Street, Montreal, Quebec, Canada.
| |
Collapse
|
31
|
Reappraisal to the study of 4E-BP1 as an mTOR substrate – A normative critique. Eur J Cell Biol 2017; 96:325-336. [DOI: 10.1016/j.ejcb.2017.03.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/31/2017] [Accepted: 03/31/2017] [Indexed: 12/20/2022] Open
|
32
|
Cuenda A, Sanz-Ezquerro JJ. p38γ and p38δ: From Spectators to Key Physiological Players. Trends Biochem Sci 2017; 42:431-442. [PMID: 28473179 DOI: 10.1016/j.tibs.2017.02.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/14/2017] [Accepted: 02/22/2017] [Indexed: 12/20/2022]
Abstract
Although the physiological roles of p38γ and p38δ signalling pathways are largely unknown, new genetic and pharmacological tools are providing groundbreaking information on the function of these two stress-activated protein kinases. Recent studies show the importance of p38γ and p38δ in the regulation of processes as diverse as cytokine production, protein synthesis, exocytosis, cell migration, gene expression, and neuron activity, which have an acute impact on the development of pathologies related to inflammation, diabetes, neurodegeneration, and cancer. These recent breakthroughs are resolving some of the questions that have long been asked regarding the function of p38γ and p38δ in biology and pathology.
Collapse
Affiliation(s)
- Ana Cuenda
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Campus de Cantoblanco, 28049 Madrid, Spain.
| | - Juan José Sanz-Ezquerro
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología/CSIC, Campus de Cantoblanco, 28049 Madrid, Spain
| |
Collapse
|
33
|
Cook PR, Tabor GT. Deciphering fact from artifact when using reporter assays to investigate the roles of host factors on L1 retrotransposition. Mob DNA 2016; 7:23. [PMID: 27895722 PMCID: PMC5120415 DOI: 10.1186/s13100-016-0079-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 11/04/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The Long INterspersed Element-1 (L1, LINE-1) is the only autonomous mobile DNA element in humans and has generated as much as half of the genome. Due to increasing clinical interest in the roles of L1 in cancer, embryogenesis and neuronal development, it has become a priority to understand L1-host interactions and identify host factors required for its activity. Apropos to this, we recently reported that L1 retrotransposition in HeLa cells requires phosphorylation of the L1 protein ORF1p at motifs targeted by host cell proline-directed protein kinases (PDPKs), which include the family of mitogen-activated protein kinases (MAPKs). Using two engineered L1 reporter assays, we continued our investigation into the roles of MAPKs in L1 activity. RESULTS We found that the MAPK p38δ phosphorylated ORF1p on three of its four PDPK motifs required for L1 activity. In addition, we found that a constitutively active p38δ mutant appeared to promote L1 retrotransposition in HeLa cells. However, despite the consistency of these findings with our earlier work, we identified some technical concerns regarding the experimental methodology. Specifically, we found that exogenous expression of p38δ appeared to affect at least one heterologous promoter in an engineered L1 reporter, as well as generate opposing effects on two different reporters. We also show that two commercially available non-targeting control (NTC) siRNAs elicit drastically different effects on the apparent retrotransposition reported by both L1 assays, which raises concerns about the use of NTCs as normalizing controls. CONCLUSIONS Engineered L1 reporter assays have been invaluable for determining the functions and critical residues of L1 open reading frames, as well as elucidating many aspects of L1 replication. However, our results suggest that caution is required when interpreting data obtained from L1 reporters used in conjunction with exogenous gene expression or siRNA.
Collapse
Affiliation(s)
- Pamela R. Cook
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 8 Center Drive, Bethesda, MD 20892 USA
| | - G. Travis Tabor
- National Institute of Child Health and Human Development, National Institutes of Health, 35 Convent Drive, Bethesda, MD 20892 USA
| |
Collapse
|
34
|
Cai W, Tucholski TM, Gregorich ZR, Ge Y. Top-down Proteomics: Technology Advancements and Applications to Heart Diseases. Expert Rev Proteomics 2016; 13:717-30. [PMID: 27448560 DOI: 10.1080/14789450.2016.1209414] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Heart diseases are a leading cause of morbidity and mortality for both men and women worldwide, and impose significant economic burdens on the healthcare systems. Despite substantial effort over the last several decades, the molecular mechanisms underlying diseases of the heart remain poorly understood. AREAS COVERED Altered protein post-translational modifications (PTMs) and protein isoform switching are increasingly recognized as important disease mechanisms. Top-down high-resolution mass spectrometry (MS)-based proteomics has emerged as the most powerful method for the comprehensive analysis of PTMs and protein isoforms. Here, we will review recent technology developments in the field of top-down proteomics, as well as highlight recent studies utilizing top-down proteomics to decipher the cardiac proteome for the understanding of the molecular mechanisms underlying diseases of the heart. Expert commentary: Top-down proteomics is a premier method for the global and comprehensive study of protein isoforms and their PTMs, enabling the identification of novel protein isoforms and PTMs, characterization of sequence variations, and quantification of disease-associated alterations. Despite significant challenges, continuous development of top-down proteomics technology will greatly aid the dissection of the molecular mechanisms underlying diseases of the hearts for the identification of novel biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Wenxuan Cai
- a Department of Cell and Regenerative Biology , University of Wisconsin-Madison , Madison , WI , USA.,b Molecular and Cellular Pharmacology Training Program , University of Wisconsin-Madison , Madison , WI , USA
| | - Trisha M Tucholski
- c Department of Chemistry , University of Wisconsin-Madison , Madison , WI , USA
| | - Zachery R Gregorich
- a Department of Cell and Regenerative Biology , University of Wisconsin-Madison , Madison , WI , USA.,b Molecular and Cellular Pharmacology Training Program , University of Wisconsin-Madison , Madison , WI , USA
| | - Ying Ge
- a Department of Cell and Regenerative Biology , University of Wisconsin-Madison , Madison , WI , USA.,c Department of Chemistry , University of Wisconsin-Madison , Madison , WI , USA.,d Human Proteomics Program , University of Wisconsin-Madison , Madison , WI , USA
| |
Collapse
|
35
|
Munkácsy E, Khan MH, Lane RK, Borror MB, Park JH, Bokov AF, Fisher AL, Link CD, Rea SL. DLK-1, SEK-3 and PMK-3 Are Required for the Life Extension Induced by Mitochondrial Bioenergetic Disruption in C. elegans. PLoS Genet 2016; 12:e1006133. [PMID: 27420916 PMCID: PMC4946786 DOI: 10.1371/journal.pgen.1006133] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 05/27/2016] [Indexed: 12/17/2022] Open
Abstract
Mitochondrial dysfunction underlies numerous age-related pathologies. In an effort to uncover how the detrimental effects of mitochondrial dysfunction might be alleviated, we examined how the nematode C. elegans not only adapts to disruption of the mitochondrial electron transport chain, but in many instances responds with extended lifespan. Studies have shown various retrograde responses are activated in these animals, including the well-studied ATFS-1-dependent mitochondrial unfolded protein response (UPRmt). Such processes fall under the greater rubric of cellular surveillance mechanisms. Here we identify a novel p38 signaling cascade that is required to extend life when the mitochondrial electron transport chain is disrupted in worms, and which is blocked by disruption of the Mitochondrial-associated Degradation (MAD) pathway. This novel cascade is defined by DLK-1 (MAP3K), SEK-3 (MAP2K), PMK-3 (MAPK) and the reporter gene Ptbb-6::GFP. Inhibition of known mitochondrial retrograde responses does not alter induction of Ptbb-6::GFP, instead induction of this reporter often occurs in counterpoint to activation of SKN-1, which we show is under the control of ATFS-1. In those mitochondrial bioenergetic mutants which activate Ptbb-6::GFP, we find that dlk-1, sek-3 and pmk-3 are all required for their life extension.
Collapse
Affiliation(s)
- Erin Munkácsy
- The Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Cellular & Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Maruf H. Khan
- The Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Medicine (Division of Geriatrics, Gerontology, and Palliative Medicine), University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Rebecca K. Lane
- The Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Megan B. Borror
- The Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Jae H. Park
- The Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Alex F. Bokov
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Alfred L. Fisher
- Department of Medicine (Division of Geriatrics, Gerontology, and Palliative Medicine), University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Geriatric Research, Education and Clinical Center, South Texas VA Health Care System, San Antonio, Texas, United States of America
- Center for Healthy Aging, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Christopher D. Link
- Institute for Behavioral Genetics & Department of Integrative Physiology, University of Colorado at Boulder, Boulder, Colorado, United States of America
| | - Shane L. Rea
- The Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| |
Collapse
|
36
|
Yurtsever Z, Patel DA, Kober DL, Su A, Miller CA, Romero AG, Holtzman MJ, Brett TJ. First comprehensive structural and biophysical analysis of MAPK13 inhibitors targeting DFG-in and DFG-out binding modes. Biochim Biophys Acta Gen Subj 2016; 1860:2335-2344. [PMID: 27369736 DOI: 10.1016/j.bbagen.2016.06.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/13/2016] [Accepted: 06/26/2016] [Indexed: 12/24/2022]
Abstract
BACKGROUND P38 MAP kinases are centrally involved in mediating extracellular signaling in various diseases. While much attention has previously been focused on the ubiquitously expressed family member MAPK14 (p38α), recent studies indicate that family members such as MAPK13 (p38δ) display a more selective cellular and tissue expression and might therefore represent a specific kinase to target in certain diseases. METHODS To facilitate the design of potent and specific inhibitors, we present here the structural, biophysical, and functional characterization of two new MAPK13-inhibitor complexes, as well as the first comprehensive structural, biophysical, and functional analysis of MAPK13 complexes with four different inhibitor compounds of greatly varying potency. RESULTS These inhibitors display IC50 values either in the nanomolar range or micromolar range (>800-fold range). The nanomolar inhibitors exhibit much longer ligand-enzyme complex half-lives compared to the micromolar inhibitors as measured by biolayer interferometry. Crystal structures of the MAPK13 inhibitor complexes reveal that the nanomolar inhibitors engage MAPK13 in the DFG-out binding mode, while the micromolar inhibitors are in the DFG-in mode. Detailed structural and computational docking analyses suggest that this difference in binding mode engagement is driven by conformational restraints imposed by the chemical structure of the inhibitors, and may be fortified by an additional hydrogen bond to MAPK13 in the nanomolar inhibitors. CONCLUSIONS These studies provide a structural basis for understanding the differences in potency exhibited by these inhibitors. GENERAL SIGNIFICANCE They also provide the groundwork for future studies to improve specificity, potency, pharmacodynamics, and pharmacokinetic properties.
Collapse
Affiliation(s)
- Zeynep Yurtsever
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, United States; Biochemistry Program, Washington University School of Medicine, St. Louis, MO 63110, United States; Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Dhara A Patel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Daniel L Kober
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, United States; Microbiology Program, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Alvin Su
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Chantel A Miller
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Arthur G Romero
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Michael J Holtzman
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, United States; Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Tom J Brett
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, United States; Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO 63110, United States; Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, United States; Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, United States.
| |
Collapse
|
37
|
Saba-El-Leil MK, Frémin C, Meloche S. Redundancy in the World of MAP Kinases: All for One. Front Cell Dev Biol 2016; 4:67. [PMID: 27446918 PMCID: PMC4921452 DOI: 10.3389/fcell.2016.00067] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/10/2016] [Indexed: 11/13/2022] Open
Abstract
The protein kinases ERK1 and ERK2 are the effector components of the prototypical ERK1/2 mitogen-activated protein (MAP) kinase pathway. This signaling pathway regulates cell proliferation, differentiation and survival, and is essential for embryonic development and cellular homeostasis. ERK1 and ERK2 homologs share similar biochemical properties but whether they exert specific physiological functions or act redundantly has been a matter of controversy. However, recent studies now provide compelling evidence in support of functionally redundant roles of ERK1 and ERK2 in embryonic development and physiology. In this review, we present a critical assessment of the evidence for the functional specificity or redundancy of MAP kinase isoforms. We focus on the ERK1/ERK2 pathway but also discuss the case of JNK and p38 isoforms.
Collapse
Affiliation(s)
- Marc K Saba-El-Leil
- Institute for Research in Immunology and Cancer, Université de Montréal Montréal, QC, Canada
| | - Christophe Frémin
- Institute for Research in Immunology and Cancer, Université de MontréalMontréal, QC, Canada; Institute for Research in Cancer of MontpellierMontpellier, France
| | - Sylvain Meloche
- Institute for Research in Immunology and Cancer, Université de MontréalMontréal, QC, Canada; Molecular Biology Program, Université de MontréalMontréal, QC, Canada; Department of Pharmacology, Université de MontréalMontréal, QC, Canada
| |
Collapse
|
38
|
Zheng S, Yang C, Liu T, Liu Q, Dai F, Sheyhidin I, Lu X. Clinicopathological significance of p38β, p38γ, and p38δ and its biological roles in esophageal squamous cell carcinoma. Tumour Biol 2016; 37:7255-7266. [PMID: 26666822 DOI: 10.1007/s13277-015-4610-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 12/07/2015] [Indexed: 01/09/2023] Open
Abstract
P38β, p38γ, and p38δ have been sporadically and scarcely reported to be involved in the carcinogenesis of cancers, compared with p38α isoform. However, little has been known regarding their clinicopathological significance and biological roles in esophageal squamous cell carcinoma (ESCC). Expression status of p38β, p38γ, and p38δ was assayed using immunohistochemistry with ESCC tissue microarray; ensuing clinicopathological significance was statistically analyzed. To define its biological roles on proliferation, migration and invasion of ESCC cell line Eca109 in vitro, MTT, wound healing, and Transwell assays were employed, respectively. As confirmation, athymic nude mice were taken to verify the effect over proliferation in vivo. It was found that both p38β and p38δ expression, other than p38γ, were significantly higher in ESCC tissues compared with paired normal controls. In terms of prognosis, only p38β expression was observed to be significantly associated with overall prognosis. Clinicopathologically, there was significant association between p38γ expression and clinical stage, lymph nodes metastases, and tumor volume. No significant association was found for p38β and p38δ between its expression and other clinicopathological parameters other than significant difference of expression between ESCC versus normal control. In Eca109, it was observed that p38β, p38γ, and p38δ can promote the cell growth and motility. As verification, over-expression of p38δ can promote, whereas knockdown of p38γ can prevent, the tumorigenesis in nude mice model xenografted with Eca109 cells whose basal level of p38δ was stably over-expressed and p38γ was stably knocked down. Together, our results demonstrate that p38β, p38γ, and p38δ played oncogenic roles in ESCC.
Collapse
Affiliation(s)
- Shutao Zheng
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
- State Key Lab Incubation Base of Xinjiang Major Diseases Research, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
| | - Chenchen Yang
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
- State Key Lab Incubation Base of Xinjiang Major Diseases Research, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
| | - Tao Liu
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
- State Key Lab Incubation Base of Xinjiang Major Diseases Research, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
| | - Qing Liu
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
- State Key Lab Incubation Base of Xinjiang Major Diseases Research, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
| | - Fang Dai
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
- State Key Lab Incubation Base of Xinjiang Major Diseases Research, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
| | - Ilyar Sheyhidin
- State Key Lab Incubation Base of Xinjiang Major Diseases Research, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
| | - Xiaomei Lu
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China.
- State Key Lab Incubation Base of Xinjiang Major Diseases Research, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China.
- Clinical Medical Research Institute, State Key Lab Breeding Base of Xinjiang Major Diseases Research, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang Uygur Autonomous Region, People's Republic of China.
| |
Collapse
|
39
|
Escós A, Risco A, Alsina-Beauchamp D, Cuenda A. p38γ and p38δ Mitogen Activated Protein Kinases (MAPKs), New Stars in the MAPK Galaxy. Front Cell Dev Biol 2016; 4:31. [PMID: 27148533 PMCID: PMC4830812 DOI: 10.3389/fcell.2016.00031] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/28/2016] [Indexed: 01/04/2023] Open
Abstract
The protein kinases p38γ and p38δ belong to the p38 mitogen-activated protein kinase (MAPK) family. p38MAPK signaling controls many cellular processes and is one of the most conserved mechanisms in eukaryotes for the cellular response to environmental stress and inflammation. Although p38γ and p38δ are widely expressed, it is likely that they perform specific functions in different tissues. Their involvement in human pathologies such as inflammation-related diseases or cancer is starting to be uncovered. In this article we give a general overview and highlight recent advances made in defining the functions of p38γ and p38δ, focusing in innate immunity and inflammation. We consider the potential of the pharmacological targeting of MAPK pathways to treat autoimmune and inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Alejandra Escós
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Spanish National Research Council (CSIC) Madrid, Spain
| | - Ana Risco
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Spanish National Research Council (CSIC) Madrid, Spain
| | - Dayanira Alsina-Beauchamp
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Spanish National Research Council (CSIC) Madrid, Spain
| | - Ana Cuenda
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Spanish National Research Council (CSIC) Madrid, Spain
| |
Collapse
|
40
|
Zur R, Garcia-Ibanez L, Nunez-Buiza A, Aparicio N, Liappas G, Escós A, Risco A, Page A, Saiz-Ladera C, Alsina-Beauchamp D, Montans J, Paramio JM, Cuenda A. Combined deletion of p38γ and p38δ reduces skin inflammation and protects from carcinogenesis. Oncotarget 2016; 6:12920-35. [PMID: 26079427 PMCID: PMC4536989 DOI: 10.18632/oncotarget.4320] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 05/25/2015] [Indexed: 12/21/2022] Open
Abstract
The contribution of chronic skin inflammation to the development of squamous cell carcinoma (SCC) is poorly understood. While the mitogen-activated protein kinase p38α regulates inflammatory responses and tumour development, little is known about the role of p38γ and p38δ in these processes. Here we show that combined p38γ and p38δ (p38γ/δ) deletion blocked skin tumour development in a chemically induced carcinogenesis model. p38γ/δ deletion reduced TPA-induced epidermal hyperproliferation and inflammation; it inhibited expression of proinflammatory cytokines and chemokines in keratinocytes in vitro and in whole skin in vivo, resulting in decreased neutrophil recruitment to skin. Our data indicate that p38γ/δ in keratinocytes promote carcinogenesis by enabling formation of a proinflammatory microenvironment that fosters epidermal hyperproliferation and tumourigenesis. These findings provide genetic evidence that p38γ and p38δ have essential roles in skin tumour development, and suggest that targeting inflammation through p38γ/δ offers a therapeutic strategy for SCC treatment and prevention.
Collapse
Affiliation(s)
- Rafal Zur
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Laura Garcia-Ibanez
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Angel Nunez-Buiza
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Noelia Aparicio
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | | | - Alejandra Escós
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Ana Risco
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Angustias Page
- Molecular Oncology Unit, CIEMAT and I+12 Biomedical Research Institute, University Hospital 12 de Octubre, Madrid, Spain
| | - Cristina Saiz-Ladera
- Molecular Oncology Unit, CIEMAT and I+12 Biomedical Research Institute, University Hospital 12 de Octubre, Madrid, Spain
| | | | - José Montans
- Centro Anatomopatológico, Camino de Vinateros, Madrid, Spain
| | - Jesús M Paramio
- Molecular Oncology Unit, CIEMAT and I+12 Biomedical Research Institute, University Hospital 12 de Octubre, Madrid, Spain
| | - Ana Cuenda
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| |
Collapse
|
41
|
Maiden SL, Petrova YI, Gumbiner BM. Microtubules Inhibit E-Cadherin Adhesive Activity by Maintaining Phosphorylated p120-Catenin in a Colon Carcinoma Cell Model. PLoS One 2016; 11:e0148574. [PMID: 26845024 PMCID: PMC4742228 DOI: 10.1371/journal.pone.0148574] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 01/19/2016] [Indexed: 01/06/2023] Open
Abstract
Tight regulation of cadherin-mediated intercellular adhesions is critical to both tissue morphogenesis during development and tissue homeostasis in adults. Cell surface expression of the cadherin-catenin complex is often directly correlated with the level of adhesion, however, examples exist where cadherin appears to be inactive and cells are completely non-adhesive. The state of p120-catenin phosphorylation has been implicated in regulating the adhesive activity of E-cadherin but the mechanism is currently unclear. We have found that destabilization of the microtubule cytoskeleton, independent of microtubule plus-end dynamics, dephosphorylates p120-catenin and activates E-cadherin adhesion in Colo 205 cells. Through chemical screening, we have also identified several kinases as potential regulators of E-cadherin adhesive activity. Analysis of several p120-catenin phosphomutants suggests that gross dephosphorylation of p120-catenin rather than that of specific amino acids may trigger E-cadherin adhesion. Uncoupling p120-catenin binding to E-cadherin at the membrane causes constitutive adhesion in Colo 205 cells, further supporting an inhibitory role of phosphorylated p120-catenin on E-cadherin activity.
Collapse
Affiliation(s)
- Stephanie L. Maiden
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- Department of Biology, Truman State University, Kirksville, Missouri, United States of America
| | - Yuliya I. Petrova
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Barry M. Gumbiner
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- Seattle Children’s Research Institute and University of Washington School of Medicine, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
42
|
Fekete T, Koncz G, Szabo B, Gregus A, Rajnavölgyi E. Interferon gamma boosts the nucleotide oligomerization domain 2-mediated signaling pathway in human dendritic cells in an X-linked inhibitor of apoptosis protein and mammalian target of rapamycin-dependent manner. Cell Mol Immunol 2015; 14:380-391. [PMID: 26521691 DOI: 10.1038/cmi.2015.90] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 08/31/2015] [Accepted: 09/06/2015] [Indexed: 11/09/2022] Open
Abstract
The cytoplasmic nucleotide oligomerization domain 2 (NOD2) receptor recognizes the bacterial cell wall component muramyl dipeptide (MDP). NOD2 ligation initiates the nuclear factor kappa B and the mitogen-activated protein kinase cascades. However, administering MDP alone is insufficient to elicit strong cytokine responses in various immune cells, including dendritic cells (DCs). Because the simultaneous presence of various microbial products and cytokines in inflamed tissues modulates DC function, we initiated this study to examine how interferon gamma (IFNγ), a central modulator of inflammation, affects the NOD2-mediated signaling pathway in human conventional DCs (cDCs). Synergistic stimulation of DCs with MDP and IFNγ increased the expression of CD40, CD80, CD83, CD86, and human leukocyte antigen DQ proteins and significantly elevated the production of pro-inflammatory cytokines IL-1β, IL-6, IL-12, and tumour necrosis factor (TNF), as well as anti-inflammatory cytokine IL-10. Furthermore, the simultaneous presence of MDP and IFNγ was necessary to decrease IkBα protein levels. By investigating various mechanisms implicated in MDP- and IFNγ-mediated signaling pathways, we revealed that the increased production of pro-inflammatory cytokines is highly dependent on the X-linked inhibitor of apoptosis protein (XIAP) but not on cellular IAP1 and IAP2. We also found that the NOD2 signaling pathway is regulated by the mammalian target of rapamycin (mTOR) but is not affected by phosphatidylinositol-3 kinase or signal transducer and activator of transcription 1 inhibition. Our results demonstrate, for the first time, that IFNγ positively affects NOD2-mediated signaling in human cDCs, in a manner considerably dependent on XIAP and partially dependent on mTOR.
Collapse
Affiliation(s)
- Tünde Fekete
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gabor Koncz
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Bioengineering, Sapientia Hungarian University of Transylvania, Cluj-Napoca, Romania
| | - Brigitta Szabo
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Andrea Gregus
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Eva Rajnavölgyi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Bioengineering, Sapientia Hungarian University of Transylvania, Cluj-Napoca, Romania
| |
Collapse
|
43
|
Prashanth Kumar B, Rajput S, Bharti R, Parida S, Mandal M. BI2536 – A PLK inhibitor augments paclitaxel efficacy in suppressing tamoxifen induced senescence and resistance in breast cancer cells. Biomed Pharmacother 2015; 74:124-32. [DOI: 10.1016/j.biopha.2015.07.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Accepted: 07/09/2015] [Indexed: 02/02/2023] Open
|
44
|
Yurtsever Z, Scheaffer SM, Romero AG, Holtzman MJ, Brett TJ. The crystal structure of phosphorylated MAPK13 reveals common structural features and differences in p38 MAPK family activation. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2015; 71:790-9. [PMID: 25849390 PMCID: PMC4388263 DOI: 10.1107/s1399004715001212] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/20/2015] [Indexed: 11/10/2022]
Abstract
The p38 MAP kinases (p38 MAPKs) represent an important family centrally involved in mediating extracellular signaling. Recent studies indicate that family members such as MAPK13 (p38δ) display a selective cellular and tissue expression and are therefore involved in specific diseases. Detailed structural studies of all p38 MAPK family members are crucial for the design of specific inhibitors. In order to facilitate such ventures, the structure of MAPK13 was determined in both the inactive (unphosphorylated; MAPK13) and active (dual phosphorylated; MAPK13/pTpY) forms. Here, the first preparation, crystallization and structure determination of MAPK13/pTpY are presented and the structure is compared with the previously reported structure of MAPK13 in order to facilitate studies for structure-based drug design. A comprehensive analysis of inactive versus active structures for the p38 MAPK family is also presented. It is found that MAPK13 undergoes a larger interlobe configurational rearrangement upon activation compared with MAPK14. Surprisingly, the analysis of activated p38 MAPK structures (MAP12/pTpY, MAPK13/pTpY and MAPK14/pTpY) reveals that, despite a high degree of sequence similarity, different side chains are used to coordinate the phosphorylated residues. There are also differences in the rearrangement of the hinge region that occur in MAPK14 compared with MAPK13 which would affect inhibitor binding. A thorough examination of all of the active (phosphorylated) and inactive (unphosphorylated) p38 MAPK family member structures was performed to reveal a common structural basis of activation for the p38 MAP kinase family and to identify structural differences that may be exploited for developing family member-specific inhibitors.
Collapse
Affiliation(s)
- Zeynep Yurtsever
- Biochemistry Program, Washington University School of Medicine, St Louis, MO 63110, USA
- Department of Internal Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St Louis, MO 63110, USA
- Drug Discovery Program in Pulmonary and Critical Care Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Suzanne M. Scheaffer
- Department of Internal Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Arthur G. Romero
- Department of Internal Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
- Drug Discovery Program in Pulmonary and Critical Care Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Michael J. Holtzman
- Department of Internal Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
- Drug Discovery Program in Pulmonary and Critical Care Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
- Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Tom J. Brett
- Department of Internal Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St Louis, MO 63110, USA
- Drug Discovery Program in Pulmonary and Critical Care Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
- Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO 63110, USA
- Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO 63110, USA
| |
Collapse
|
45
|
Martin EC, Krebs AE, Burks HE, Elliott S, Baddoo M, Collins-Burow BM, Flemington EK, Burow ME. miR-155 induced transcriptome changes in the MCF-7 breast cancer cell line leads to enhanced mitogen activated protein kinase signaling. Genes Cancer 2014; 5:353-64. [PMID: 25352952 PMCID: PMC4209600 DOI: 10.18632/genesandcancer.33] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 10/01/2014] [Indexed: 11/25/2022] Open
Abstract
A single microRNA (miRNA) has the potential to regulate thousands of genes and thus govern multiple signaling pathways at once. miR-155 is an oncogenic miRNA which regulates many cellular pathways, designating it as a multifaceted regulator of proliferation, chemo-resistance, and apoptosis. While many singular targeted effects of miR-155 have been defined and an oncogenic role has been attributed to miR-155 expression, the global effect of miR-155 on the cellular transcriptomes of an ER+ breast cancer cell line has yet to be determined. Here we demonstrate that miR-155 expression increases tumorigenesis in vivo and we determine miR-155 mediated transcriptome changes through next generation sequencing analysis. miR-155 expression alters many signaling pathways, with the chief altered pathway being the MAPK signaling cascade and miR-155 induces shortening of target mRNA 3′UTRs and alternative isoform expression of MAPK related genes. In addition there is an observed increase in protein phosphorylation of components of MAPK signaling including ERK1/2 and AP-1 complex members (Fra-1 and c-Fos) as well as elevated gene expression of MAPK regulated genes Zeb1, Snail, Plaur, and SerpinE1.
Collapse
Affiliation(s)
- Elizabeth C Martin
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA
| | - Adrienne E Krebs
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA
| | - Hope E Burks
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA
| | - Steven Elliott
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA
| | - Melody Baddoo
- Tulane Cancer Center, Tulane University, New Orleans, LA ; Department of Pathology, Tulane University, New Orleans, LA
| | - Bridgette M Collins-Burow
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA
| | - Erik K Flemington
- Tulane Cancer Center, Tulane University, New Orleans, LA ; Department of Pathology, Tulane University, New Orleans, LA
| | - Matthew E Burow
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA ; Tulane Cancer Center, Tulane University, New Orleans, LA ; Department of Pharmacology, Tulane University, New Orleans, LA
| |
Collapse
|
46
|
p38δ MAPK: Emerging Roles of a Neglected Isoform. Int J Cell Biol 2014; 2014:272689. [PMID: 25313309 PMCID: PMC4182853 DOI: 10.1155/2014/272689] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 08/29/2014] [Accepted: 08/31/2014] [Indexed: 12/19/2022] Open
Abstract
p38δ mitogen activated protein kinase (MAPK) is a unique stress responsive protein kinase. While the p38 MAPK family as a whole has been implicated in a wide variety of biological processes, a specific role for p38δ MAPK in cellular signalling and its contribution to both physiological and pathological conditions are presently lacking. Recent emerging evidence, however, provides some insights into specific p38δ MAPK signalling. Importantly, these studies have helped to highlight functional similarities as well as differences between p38δ MAPK and the other members of the p38 MAPK family of kinases. In this review we discuss the current understanding of the molecular mechanisms underlying p38δ MAPK activity. We outline a role for p38δ MAPK in important cellular processes such as differentiation and apoptosis as well as pathological conditions such as neurodegenerative disorders, diabetes, and inflammatory disease. Interestingly, disparate roles for p38δ MAPK in tumour development have also recently been reported. Thus, we consider evidence which characterises p38δ MAPK as both a tumour promoter and a tumour suppressor. In summary, while our knowledge of p38δ MAPK has progressed somewhat since its identification in 1997, our understanding of this particular isoform in many cellular processes still strikingly lags behind that of its counterparts.
Collapse
|
47
|
del Reino P, Alsina-Beauchamp D, Escós A, Cerezo-Guisado MI, Risco A, Aparicio N, Zur R, Fernandez-Estévez M, Collantes E, Montans J, Cuenda A. Pro-Oncogenic Role of Alternative p38 Mitogen-Activated Protein Kinases p38γ and p38δ, Linking Inflammation and Cancer in Colitis-Associated Colon Cancer. Cancer Res 2014; 74:6150-60. [DOI: 10.1158/0008-5472.can-14-0870] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
48
|
Johnson JC, Martinez O, Honko AN, Hensley LE, Olinger GG, Basler CF. Pyridinyl imidazole inhibitors of p38 MAP kinase impair viral entry and reduce cytokine induction by Zaire ebolavirus in human dendritic cells. Antiviral Res 2014; 107:102-9. [PMID: 24815087 DOI: 10.1016/j.antiviral.2014.04.014] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 04/23/2014] [Accepted: 04/25/2014] [Indexed: 01/03/2023]
Abstract
Antigen presenting cells (APCs), including macrophages and dendritic cells, are early and sustained targets of Ebola virus (EBOV) infection in vivo. Because EBOV activates mitogen-activated protein kinase (MAPK) signaling upon infection of APCs, we evaluated the effect of pyridinyl imidazole inhibitors of p38 MAPK on EBOV infection of human APCs and EBOV mediated cytokine production from human DCs. The p38 MAPK inhibitors reduced viral replication in PMA-differentiated macrophage-like human THP-1 cells with an IC50 of 4.73μM (SB202190), 8.26μM (p38kinhIII) and 8.21μM (SB203580) and primary human monocyte-derived dendritic cells (MDDCs) with an IC50 of 2.67μM (SB202190). Furthermore, cytokine production from EBOV-treated MDDCs was inhibited in a dose-dependent manner. A control pyridinyl imidazole compound failed to inhibit either EBOV infection or cytokine induction. Using an established EBOV virus-like particle (VLP) entry assay, we demonstrate that inhibitor pretreatment blocked VLP entry suggesting that the inhibitors blocked infection and replication at least in part by blocking EBOV entry. Taken together, our results indicate that pyridinyl imidazole p38 MAPK inhibitors may serve as leads for the development of therapeutics to treat EBOV infection.
Collapse
Affiliation(s)
- Joshua C Johnson
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD 21702, United States
| | - Osvaldo Martinez
- Dept. of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Anna N Honko
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD 21702, United States
| | - Lisa E Hensley
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD 21702, United States
| | - Gene G Olinger
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD 21702, United States
| | - Christopher F Basler
- Dept. of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States.
| |
Collapse
|
49
|
Abstract
The binding of tumour necrosis factor α (TNFα) to cell surface receptors engages multiple signal transduction pathways, including three groups of mitogen-activated protein (MAP) kinases: extracellular-signal-regulated kinases (ERKs); the cJun NH2-terminal kinases (JNKs); and the p38 MAP kinases. These MAP kinase signalling pathways induce a secondary response by increasing the expression of several inflammatory cytokines (including TNFα) that contribute to the biological activity of TNFα. MAP kinases therefore function both upstream and down-stream of signalling by TNFα receptors. Here we review mechanisms that mediate these actions of MAP kinases during the response to TNFα.
Collapse
Affiliation(s)
- Guadalupe Sabio
- Department of Vascular Biology and Inflammation, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain
| | - Roger J Davis
- Howard Hughes Medical Institute and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
50
|
Pinsino A, Roccheri MC, Matranga V. Manganese overload affects p38 MAPK phosphorylation and metalloproteinase activity during sea urchin embryonic development. MARINE ENVIRONMENTAL RESEARCH 2014; 93:64-69. [PMID: 23998794 DOI: 10.1016/j.marenvres.2013.08.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 07/22/2013] [Accepted: 08/07/2013] [Indexed: 06/02/2023]
Abstract
In the marine environment, manganese represents a potential emerging contaminant, resulting from an increased production of manganese-containing compounds. In earlier reports we found that the exposure of Paracentrotus lividus sea urchin embryos to manganese produced phenotypes with no skeleton. In addition, manganese interfered with calcium uptake, perturbed extracellular signal-regulated kinase (ERK) signaling, affected the expression of skeletogenic genes, and caused an increase of the hsc70 and hsc60 protein levels. Here, we extended our studies focusing on the temporal activation of the p38 mitogen-activated protein kinase (p38 MAPK) and the proteolytic activity of metalloproteinases (MMPs). We found that manganese affects the stage-dependent dynamics of p38 MAPK activation and inhibits the total gelatin-auto-cleaving activity of MMPs, with the exclusion of the 90-85 kDa and 68-58 kDa MMPs, whose levels remain high all throughout development. Our findings correlate, for the first time to our knowledge, an altered activation pattern of the p38 MAPK with an aberrant MMP proteolytic activity in the sea urchin embryo.
Collapse
Affiliation(s)
- A Pinsino
- Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Consiglio Nazionale delle Ricerche, Palermo, Italy.
| | - M C Roccheri
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Italy
| | - V Matranga
- Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Consiglio Nazionale delle Ricerche, Palermo, Italy
| |
Collapse
|