Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
Am J Physiol Lung Cell Mol Physiol ; 325(6): L726-L740, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37847710

ABSTRACT

Common respiratory diseases continue to represent a major public health problem, and much of the morbidity and mortality is due to airway inflammation and mucus production. Previous studies indicated a role for mitogen-activated protein kinase 14 (MAPK14) in this type of disease, but clinical trials are unsuccessful to date. Our previous work identified a related but distinct kinase known as MAPK13 that is activated in respiratory airway diseases and is required for mucus production in human cell-culture models. Support for MAPK13 function in these models came from effectiveness of MAPK13 versus MAPK14 gene-knockdown and from first-generation MAPK13-14 inhibitors. However, these first-generation inhibitors were incompletely optimized for blocking activity and were untested in vivo. Here we report the next generation and selection of a potent MAPK13-14 inhibitor (designated NuP-3) that more effectively downregulates type-2 cytokine-stimulated mucus production in air-liquid interface and organoid cultures of human airway epithelial cells. We also show that NuP-3 treatment prevents respiratory airway inflammation and mucus production in new minipig models of airway disease triggered by type-2 cytokine challenge or respiratory viral infection. The results thereby provide the next advance in developing a small-molecule kinase inhibitor to address key features of respiratory disease.NEW & NOTEWORTHY This study describes the discovery of a potent mitogen-activated protein kinase 13-14 (MAPK13-14) inhibitor and its effectiveness in models of respiratory airway disease. The findings thereby provide a scheme for pathogenesis and therapy of lung diseases [e.g., asthma, chronic obstructive pulmonary disease (COPD), Covid-19, postviral, and allergic respiratory disease] and related conditions that implicate MAPK13-14 function. The findings also refine a hypothesis for epithelial and immune cell functions in respiratory disease that features MAPK13 as a possible component of this disease process.


Subject(s)
Mitogen-Activated Protein Kinase 14 , Pulmonary Disease, Chronic Obstructive , Animals , Humans , Swine , Mitogen-Activated Protein Kinase 14/metabolism , Swine, Miniature/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Mucus/metabolism , Cytokines/metabolism , Mitogen-Activated Protein Kinase 13/metabolism
2.
Elife ; 122023 07 17.
Article in English | MEDLINE | ID: mdl-37458356

ABSTRACT

Evidence implicating p38γ and p38δ (p38γ/p38δ) in inflammation are mainly based on experiments using Mapk12/Mapk13-deficient (p38γ/δKO) mice, which show low levels of TPL2, the kinase upstream of MKK1-ERK1/2 in myeloid cells. This could obscure p38γ/p38δ roles, since TPL2 is essential for regulating inflammation. Here, we generated a Mapk12D171A/D171A/Mapk13-/- (p38γ/δKIKO) mouse, expressing kinase-inactive p38γ and lacking p38δ. This mouse exhibited normal TPL2 levels, making it an excellent tool to elucidate specific p38γ/p38δ functions. p38γ/δKIKO mice showed a reduced inflammatory response and less susceptibility to lipopolysaccharide (LPS)-induced septic shock and Candida albicans infection than wild-type (WT) mice. Gene expression analyses in LPS-activated wild-type and p38γ/δKIKO macrophages revealed that p38γ/p38δ-regulated numerous genes implicated in innate immune response. Additionally, phospho-proteomic analyses and in vitro kinase assays showed that the transcription factor myocyte enhancer factor-2D (MEF2D) was phosphorylated at Ser444 via p38γ/p38δ. Mutation of MEF2D Ser444 to the non-phosphorylatable residue Ala increased its transcriptional activity and the expression of Nos2 and Il1b mRNA. These results suggest that p38γ/p38δ govern innate immune responses by regulating MEF2D phosphorylation and transcriptional activity.


Subject(s)
Lipopolysaccharides , Mitogen-Activated Protein Kinase 13 , Animals , Mice , Mitogen-Activated Protein Kinase 13/metabolism , Proteomics , Immunity, Innate , Mitogen-Activated Protein Kinase 12/genetics , Mitogen-Activated Protein Kinase 12/metabolism , Inflammation
3.
Cancer Gene Ther ; 30(9): 1181-1189, 2023 09.
Article in English | MEDLINE | ID: mdl-37248432

ABSTRACT

Gastrointestinal cancers are a leading cause of cancer morbidity and mortality worldwide with 4.2 million new cases and 3.2 million deaths estimated in 2020. Despite the advances in primary and adjuvant therapies, patients still develop distant metastases and require novel therapies. Mitogen­activated protein kinase (MAPK) cascades are crucial signaling pathways that regulate many cellular processes, including proliferation, differentiation, apoptosis, stress responses and cancer development. p38 Mitogen Activated Protein Kinases (p38 MAPKs) includes four isoforms: p38α (MAPK14), p38ß (MAPK11), p38γ (MAPK12), and p38δ (MAPK13). p38 MAPK was first identified as a stress response protein kinase that phosphorylates different transcriptional factors. Dysregulation of p38 pathways, in particular p38γ, are associated with cancer development, metastasis, autophagy and tumor microenvironment. In this article, we provide an overview of p38 and p38γ with respect to gastrointestinal cancers. Furthermore, targeting p38γ is also discussed as a potential therapy for gastrointestinal cancers.


Subject(s)
Gastrointestinal Neoplasms , Mitogen-Activated Protein Kinase 11 , Humans , Mitogen-Activated Protein Kinase 11/metabolism , Mitogen-Activated Protein Kinase 12/genetics , Mitogen-Activated Protein Kinase 12/metabolism , Mitogen-Activated Protein Kinase 13/metabolism , Signal Transduction , Gastrointestinal Neoplasms/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , Tumor Microenvironment
4.
Elife ; 112022 08 16.
Article in English | MEDLINE | ID: mdl-35971771

ABSTRACT

Stress-activated p38 kinases control a plethora of functions, and their dysregulation has been linked to the development of steatosis, obesity, immune disorders, and cancer. Therefore, they have been identified as potential targets for novel therapeutic strategies. There are four p38 family members (p38α, p38ß, p38γ, and p38δ) that are activated by MKK3 and MKK6. Here, we demonstrate that lack of MKK6 reduces the lifespan in mice. Longitudinal study of cardiac function in MKK6 KO mice showed that young mice develop cardiac hypertrophy which progresses to cardiac dilatation and fibrosis with age. Mechanistically, lack of MKK6 blunts p38α activation while causing MKK3-p38γ/δ hyperphosphorylation and increased mammalian target of rapamycin (mTOR) signaling, resulting in cardiac hypertrophy. Cardiac hypertrophy in MKK6 KO mice is reverted by knocking out either p38γ or p38δ or by inhibiting the mTOR pathway with rapamycin. In conclusion, we have identified a key role for the MKK3/6-p38γ/δ pathway in the development of cardiac hypertrophy, which has important implications for the clinical use of p38α inhibitors in the long-term treatment since they might result in cardiotoxicity.


The human heart can increase its size to supply more blood to the body's organs. This process, called hypertrophy, can happen during exercise or be caused by medical conditions, such as high blood pressure or inherited genetic diseases. If hypertrophy is continually driven by illness, this can cause the heart to fail and no longer be able to properly pump blood around the body. For hypertrophy to happen, several molecular changes occur in the cells responsible for contracting the heart, including activation of the p38 pathway. Within this pathway is a p38 enzyme as well as a series of other proteins which are sequentially turned on in response to stress, such as inflammatory molecules or mechanical forces that alter the cell's shape. There are different types of p38 enzyme which have been linked to other diseases, making them a promising target for drug development. However, clinical trials blocking individual members of the p38 family have had disappointing results. An alternative approach is to target other proteins involved in the p38 pathway, such as MKK6, but it is not known what effect this might have. To investigate, Romero-Becerra et al. genetically modified mice to not have any MKK6 protein. As a result, these mice had a shorter lifespan, with hypertrophy developing at a young age that led to heart problems. Romero-Becerra et al. used different mice models to understand why this happened, showing that a lack of MKK6 reduces the activity of a specific member of the p38 family called p38α. However, this blockage boosted a different branch of the pathway which involved two other p38 proteins, p38γ and p38δ. This, in turn, triggered another key pathway called mTOR which also promotes hypertrophy of the heart. These results suggest that drugs blocking MKK6 and p38α could lead to side effects that cause further harm to the heart. A more promising approach for treating hypertrophic heart conditions could be to inhibit p38γ and/or p38δ. However, before this can be fully explored, further work is needed to generate compounds that specifically target these proteins.


Subject(s)
Heart Diseases , MAP Kinase Kinase 6 , Mitogen-Activated Protein Kinase 13 , Animals , Cardiomegaly , Heart Diseases/genetics , Heart Diseases/pathology , Longitudinal Studies , MAP Kinase Kinase 3/metabolism , MAP Kinase Kinase 6/genetics , Mice , Mitogen-Activated Protein Kinase 13/metabolism , TOR Serine-Threonine Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Proc Natl Acad Sci U S A ; 119(35): e2204752119, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35994673

ABSTRACT

p38γ and p38δ (p38γ/p38δ) regulate inflammation, in part by controlling tumor progression locus 2 (TPL2) expression in myeloid cells. Here, we demonstrate that TPL2 protein levels are dramatically reduced in p38γ/p38δ-deficient (p38γ/δ-/-) cells and tissues without affecting TPL2 messenger ribonucleic acid (mRNA) expression. We show that p38γ/p38δ posttranscriptionally regulates the TPL2 amount at two different levels. p38γ/p38δ interacts with the TPL2/A20 Binding Inhibitor of NF-κB2 (ABIN2)/Nuclear Factor κB1p105 (NF-κB1p105) complex, increasing TPL2 protein stability. Additionally, p38γ/p38δ regulates TPL2 mRNA translation by modulating the repressor function of TPL2 3' Untranslated region (UTR) mediated by its association with aconitase-1 (ACO1). ACO1 overexpression in wild-type cells increases the translational repression induced by TPL2 3'UTR and severely decreases TPL2 protein levels. p38δ binds to ACO1, and p38δ expression in p38γ/δ-/- cells fully restores TPL2 protein to wild-type levels by reducing the translational repression of TPL2 mRNA. This study reveals a unique mechanism of posttranscriptional regulation of TPL2 expression, which given its central role in innate immune response, likely has great relevance in physiopathology.


Subject(s)
Aconitate Hydratase , MAP Kinase Kinase Kinases , Mitogen-Activated Protein Kinase 12 , Mitogen-Activated Protein Kinase 13 , Aconitate Hydratase/genetics , Aconitate Hydratase/metabolism , Gene Expression Regulation , Immunity, Innate , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase 12/genetics , Mitogen-Activated Protein Kinase 12/metabolism , Mitogen-Activated Protein Kinase 13/genetics , Mitogen-Activated Protein Kinase 13/metabolism , RNA, Messenger/genetics
6.
PLoS Biol ; 19(11): e3001447, 2021 11.
Article in English | MEDLINE | ID: mdl-34758018

ABSTRACT

During the first weeks of postnatal heart development, cardiomyocytes undergo a major adaptive metabolic shift from glycolytic energy production to fatty acid oxidation. This metabolic change is contemporaneous to the up-regulation and activation of the p38γ and p38δ stress-activated protein kinases in the heart. We demonstrate that p38γ/δ contribute to the early postnatal cardiac metabolic switch through inhibitory phosphorylation of glycogen synthase 1 (GYS1) and glycogen metabolism inactivation. Premature induction of p38γ/δ activation in cardiomyocytes of newborn mice results in an early GYS1 phosphorylation and inhibition of cardiac glycogen production, triggering an early metabolic shift that induces a deficit in cardiomyocyte fuel supply, leading to whole-body metabolic deregulation and maladaptive cardiac pathogenesis. Notably, the adverse effects of forced premature cardiac p38γ/δ activation in neonate mice are prevented by maternal diet supplementation of fatty acids during pregnancy and lactation. These results suggest that diet interventions have a potential for treating human cardiac genetic diseases that affect heart metabolism.


Subject(s)
Glycogen Synthase/metabolism , Mitogen-Activated Protein Kinase 12/metabolism , Mitogen-Activated Protein Kinase 13/metabolism , Myocardium/enzymology , Animals , Animals, Newborn , Cardiomegaly/enzymology , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Diet, High-Fat , Enzyme Activation , Feeding Behavior , Female , Gene Deletion , Glucose Intolerance/enzymology , Glycogen/metabolism , Glycogen Synthase Kinase 3/metabolism , Insulin Resistance , Lipid Metabolism , MAP Kinase Signaling System , Mice, Inbred C57BL , Myocytes, Cardiac/enzymology , Organ Specificity , Phosphorylation
7.
PLoS One ; 16(9): e0256701, 2021.
Article in English | MEDLINE | ID: mdl-34473747

ABSTRACT

The developmental competence of male and female gametes is frequently reduced under in vitro conditions, mainly due to oxidative stress during handling. The amino-acid derived hormone melatonin has emerged as a potent non-enzymatic antioxidant in many biological systems. The goal of the present study was to evaluate the effects of melatonin on post-thaw sperm quality, fertilizing ability, and embryo development and competence in vitro after in vitro fertilization. Frozen-thawed bovine spermatozoa were incubated either in the presence of 10-11 M melatonin (MT), or its solvent (ethanol; Sham-Control), or plain Tyrode's Albumin Lactate Pyruvate medium (TALP, Control). Computer-Assisted Sperm Analysis (CASA) and flow cytometry data after 30 min, 120 min, and 180 min incubation did not reveal any significant effects of melatonin on average motility parameters, sperm subpopulation structure as determined by hierarchical cluster, or on the percentage of viable, acrosome intact sperm, or viable sperm with active mitochondria. Nevertheless, in vitro matured cumulus-oocyte-complexes fertilized with spermatozoa which had been preincubated with 10-11 M melatonin (MT-Sperm) showed higher (P < 0.01) rates of monospermic fertilization, reduced (P < 0.05) polyspermy and enhanced (P < 0.05) embryo development compared to the Control group. Moreover, the relative abundance of MAPK13 in the in vitro-derived blastocysts was greater (P < 0.05) than observed in the Control group. In conclusion, adding melatonin to the sperm-preparation protocol for bovine IVF improved proper fertilization and enhanced embryonic development and competence in vitro.


Subject(s)
Cryopreservation/methods , Embryonic Development/drug effects , In Vitro Oocyte Maturation Techniques , Melatonin/pharmacology , Oocytes/drug effects , Spermatozoa/drug effects , Animals , Biomarkers/metabolism , Blastocyst/cytology , Blastocyst/drug effects , Blastocyst/metabolism , Cattle , Culture Media/chemistry , Culture Media/pharmacology , Female , Fertilization in Vitro/methods , Gene Expression , Male , Mitogen-Activated Protein Kinase 13/genetics , Mitogen-Activated Protein Kinase 13/metabolism , Oocytes/cytology , Oocytes/metabolism , Spermatozoa/cytology , Spermatozoa/metabolism
8.
Inflamm Res ; 69(11): 1143-1156, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32852592

ABSTRACT

OBJECTIVE: IL-6-induced STAT3 activation is associated with various chronic inflammatory diseases. In this study, we investigated the anti-STAT3 mechanism of the dietary polyphenol, biochanin A (BCA), in IL-6-treated macrophages. METHODS: The effect of BCA on STAT3 and p38 MAPK was analyzed by immunoblot. The localization of both these transcription factors was determined by immunofluorescence and fractionation studies. The impact on DNA-binding activity of STAT3 was studied by luciferase assay. To understand which of the isoforms of p38 MAPK was responsible for BCA-mediated regulation of STAT3, overexpression of the proteins, site-directed mutagenesis, pull-down assays and computational analysis were performed. Finally, adhesion-migration assays and semi-quantitative PCR were employed to understand the biological effects of BCA-mediated regulation of STAT3. RESULTS: BCA prevented STAT3 phosphorylation (Tyr705) and increased p38 MAPK phosphorylation (Thr180/Tyr182) in IL-6-stimulated differentiated macrophages. This opposing modulatory effect of BCA was not observed in cells treated with other stress-inducing stimuli that activate p38 MAPK. BCA abrogated IL-6-induced nuclear translocation of phospho-STAT3 and its transcriptional activity, while increasing the cellular abundance of phospho-p38 MAPK. BCA-induced phosphorylation of p38δ, but not α, ß, or γ was responsible for impeding IL-6-induced STAT3 phosphorylation. Interestingly, interaction with phospho-p38δ masked the Tyr705 residue of STAT3, preventing its phosphorylation. BCA significantly reduced STAT3-dependent expression of icam-1 and mcp-1 diminishing IL-6-mediated monocyte adhesion and migration. CONCLUSION: This differential regulation of STAT3 and p38 MAPK in macrophages establishes a novel anti-inflammatory mechanism of BCA which could be important for the prevention of IL-6-associated chronic inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Genistein/pharmacology , Interleukin-6/pharmacology , Macrophages/drug effects , Mitogen-Activated Protein Kinase 13/metabolism , STAT3 Transcription Factor/metabolism , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , HEK293 Cells , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/physiology , Macrophages/physiology , Mitogen-Activated Protein Kinase 13/genetics , Phosphorylation/drug effects , THP-1 Cells
9.
Clin Sci (Lond) ; 134(12): 1449-1456, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32556178

ABSTRACT

Hepatitis C virus (HCV) infection and chronic hepatitis C (CHC) are associated with a measurable risk of insulin resistance (IR)/impaired glucose tolerance (IGT)/diabetes mellitus (DM). While loss of hepatic endocrine function contributes to liver cirrhosis in diabetic patients, onset and progression of IR/IGT to diabetes and exacerbation of incident hyperglycemia are ostensibly linked with chronic HCV infection. In this regard, the study by Chen J et al. appearing in Clinical Science (2020) (134(5) https://doi.org/10.1042/CS20190900) attempts to understand the mechanisms underlying the savaging effects of chronic HCV infection on insulin-producing pancreatic ß-cells and hence diabetic onset. The study investigated the role of mitogen-activated protein kinase (MAPK) p38δ-protein kinase D (PKD)-golgi complex axis in impacting insulin exocytosis. It was inferred that an insulin secretory defect of pancreatic ß-cells, owing to disrupted insulin exocytosis, to an extent explains ß-cell dysfunction in HCV-infected or CHC milieu. HCV infection negatively regulates first-phase and second-phase insulin secretion by impinging on PKD-dependent insulin secretory granule fission at trans-golgi network and insulin secretory vesicle membrane fusion events. This commentary highlights the study in question, that deciphered the contribution of p38δ MAPK-PKD-golgi complex axis to ß-cell dysfunction in CHC milieu. This pivotal axis proffers a formidable therapeutic opportunity for alleviation of double burden of glucose abnormalities/DM and CHC.


Subject(s)
Exocytosis , Golgi Apparatus/metabolism , Hepatitis C, Chronic/metabolism , Hepatitis C, Chronic/pathology , Insulin/metabolism , Mitogen-Activated Protein Kinase 13/metabolism , Protein Kinase C/metabolism , Animals , Humans , Mice, Inbred C57BL , Models, Biological
10.
Aging (Albany NY) ; 12(8): 6774-6792, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32299063

ABSTRACT

BACKGROUND: Circular RNA is a type of non-coding RNA with great potential in regulating gene expression and associated with disease progression. However, the role of circular RNA in endometrial carcinoma (EC) remains largely unknown. RESULTS: In this study, we found that circTNFRSF21 was highly expressed in EC cells and tumor tissues. In vitro and in vivo results showed that circTNFRSF21 was linked to increased EC cell growth and EC xenografts formation in nude mice. Mechanically, we showed that circTNFRSF21 acts as a sponge of miR-1227 in EC cells to rescue MAPK13/ATF2 signaling pathway activity. CONCLUSIONS: Our studies suggested that in the EC, circTNFRSF21 promotes EC formation through downregulating miR-1227 expression and activating MAPK13/ATF2 signaling pathway. These findings provide strong evidence that circTNFRSF21-miR-1227-MAPK13/ATF2 axis is a promising target for EC treatment. METHODS: qRT-PCR was used to detect circTNFRSF21expression in EC patients and EC cell lines. Cell growth, cell colony formation, cell apoptosis, cell cycle progression, and in vivo tumor formation assays were used to evaluate the roles of circTNFRSF21 in EC. Western blot, luciferase assay, RNA pull-down, siRNA knockdown, and CRISPR gene knock out assays were applied to study the mechanisms through which circTNFRSF21 regulates EC formation.


Subject(s)
Carcinoma/genetics , Carcinoma/pathology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , RNA, Circular/genetics , Receptors, Tumor Necrosis Factor/genetics , Activating Transcription Factor 2/metabolism , Animals , Apoptosis , Carcinoma/metabolism , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Endometrial Neoplasms/metabolism , Endometrium/metabolism , Female , Humans , Mice , Middle Aged , Mitogen-Activated Protein Kinase 13/metabolism , Neoplasm Transplantation , RNA, Circular/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction
11.
Clin Sci (Lond) ; 134(5): 529-542, 2020 03 13.
Article in English | MEDLINE | ID: mdl-32100852

ABSTRACT

Chronic hepatitis C virus (HCV) infection has a close association with type 2 diabetes mellitus. Although the mechanisms of insulin resistance in chronic hepatitis C (CHC) patients have been extensively studied, little attention has been given to the role of ß-cell function in HCV-associated diabetes. Here, we analysed ß-cell function in CHC patients and HCV-infected mouse model and found in addition to insulin resistance, impaired pancreatic ß-cell function occurred in CHC patients and HCV-infected C/OTg mice, not only in diabetic individuals but also in individuals with impaired fasting glucose levels. Both first-phase and second-phase insulin secretion were impaired, at least partially due to the reduction of exocytosis of secretory insulin-containing granules following HCV infection. Up-regulated p38δ in HCV-infected ß-cells resulted in inactivation of protein kinase D (PKD), which was responsible for impaired insulin secretory capacity of ß-cells. Thus, impaired insulin secretion due to HCV infection in ß-cells contributes to HCV-associated type 2 diabetes. These findings provided a new inspiration for the important prognostic and therapeutic implications in the management of CHC patients with impaired fasting glucose.


Subject(s)
Exocytosis , Hepatitis C, Chronic/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Mitogen-Activated Protein Kinase 13/metabolism , Animals , Cell Line, Tumor , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Glucose Intolerance/blood , Glucose Intolerance/metabolism , Glucose Intolerance/virology , Hepatitis C, Chronic/virology , Humans , Insulin/metabolism , Insulin-Secreting Cells/virology , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Protein Kinase C/metabolism
12.
J Exp Clin Cancer Res ; 38(1): 504, 2019 Dec 27.
Article in English | MEDLINE | ID: mdl-31881903

ABSTRACT

MKK3 is a member of the dual specificity kinase group specific upstream activator of p38 MAPK proteins. We originally identified MKK3 as mutant p53 (mutp53) gain-of-function (GOF) upregulated target gene in different tumor models. To deeply investigate the MKK3 functions in cancer, taking advantage of a panel of authenticated colorectal cancer (CRC) lines and primary colonocytes, we found that MKK3 activates specifically p38delta MAPK protein, which signaling is further triggered by 5-fluorouracil (5-FU) treatments, a largely adopted chemotherapeutic drug in CRC clinical practice. The overall achieved results proposed the MKK3/p38delta MAPK as relevant molecular axis involved in abrogating efficacy to 5-FU treatments in CRC. This commentary will provide an overall discussion of the results that have been achieved contextualizing them in the overview of the knowledge in the p38 MAPK field in cancer disease.


Subject(s)
Colorectal Neoplasms/metabolism , MAP Kinase Kinase 3/metabolism , Mitogen-Activated Protein Kinase 13/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Humans , Molecular Targeted Therapy , Protein Kinase Inhibitors/therapeutic use , Treatment Outcome
13.
Int J Mol Sci ; 20(7)2019 Mar 27.
Article in English | MEDLINE | ID: mdl-30934690

ABSTRACT

Activation and/or upregulated expression of p38δ are demonstrated in human skin malignancies including cutaneous squamous cell carcinoma, suggesting a role for p38δ in skin carcinogenesis. We previously reported that mice with germline deletion of the p38δ gene are significantly protected from chemical skin carcinogenesis. Here, we investigated the effects of cell-selective targeted ablation of p38δ in keratinocytes and in immune (myeloid) cells on skin tumor development in a two-stage 7,12-dimethylbenz(a)anthracene (DMBA)/12-O-tetradecanoylphorbol-13-acetate (TPA) chemical mouse skin carcinogenesis model. Conditional keratinocyte-specific p38δ ablation (p38δ-cKO∆K) did not influence the latency, incidence, or multiplicity of chemically-induced skin tumors, but led to increased tumor volume in females during the TPA promotion stage, and reduced malignant progression in males and females relative to their wild-type counterparts. In contrast, conditional myeloid cell-specific p38δ deletion (p38δ-cKO∆M) inhibited DMBA/TPA-induced skin tumorigenesis in male but not female mice. Thus, tumor onset was delayed, and tumor incidence, multiplicity, and volume were reduced in p38δ-cKO∆M males compared with control wild-type males. Moreover, the percentage of male mice with malignant tumors was decreased in the p38δ-cKO∆M group relative to their wild-type counterparts. Collectively, these results reveal that cell-specific p38δ targeting modifies susceptibility to chemical skin carcinogenesis in a context-, stage-, and sex-specific manner.


Subject(s)
Mitogen-Activated Protein Kinase 13/metabolism , Sex Characteristics , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , 9,10-Dimethyl-1,2-benzanthracene , Animals , Carcinogenesis/pathology , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Cytokines/metabolism , Disease Progression , Female , Gene Deletion , Inflammation Mediators/metabolism , Keratinocytes/enzymology , Male , Mice, Knockout , Myeloid Cells/metabolism , Neoplasm Staging , Phenotype , Skin/pathology , Tetradecanoylphorbol Acetate
14.
J Exp Clin Cancer Res ; 38(1): 124, 2019 Mar 11.
Article in English | MEDLINE | ID: mdl-30871606

ABSTRACT

BACKGROUND: Gastric cancer is the second most common cause of cancer-related mortality; thus, the mechanisms underlying tumor metastasis and growth in gastric cancer need to be extensively explored. METHODS: Differentially expressed genes were examined in gastric cancer samples with lymph node metastasis (LNM) and without LNM using mRNA microarray and RT-qPCR. The effects of G antigen 7B (GAGE7B) on the metastasis, growth, and angiogenesis of gastric cancer were investigated in vitro and in vivo. GAGE7B protein expression was detected by immunohistochemical (IHC) analysis. Microarray, RT-qPCR, and western blot assays were performed to detect downstream target genes of GAGE7B. Dual-luciferase reporter and western blot assays were used to identify miRNAs that could negatively regulate GAGE7B. RESULTS: GAGE7B was significantly overexpressed in samples with LNM. High expression levels of GAGE7B were associated with advanced clinical stage and poor patient survival. GAGE7B dramatically enhanced the metastasis, growth, and angiogenesis ability of gastric cancer. GAGE7B was further demonstrated to promote the progression of gastric cancer by activating the p38δ/pMAPKAPK2/pHSP27 pathway. However, the GAGE7B-induced p38δ/pMAPKAPK2/pHSP27 pathway was inactivated by miR-30c, as the expression levels of both GAGE7B and p38δ were found to be directly suppressed by miR-30c. Intriguingly, GAGE7B was found to be a ceRNA for p38δ, as it activated the p38δ/pMAPKAPK2/pHSP27 pathway by competitively binding miR-30c. CONCLUSIONS: GAGE7B may serve as a prognostic indicator in gastric cancer. GAGE7B significantly promotes gastric cancer progression by upregulating the p38δ/pMAPKAPK2/pHSP27 pathway, but it is negatively regulated by miR-30c. GAGE7B and miR-30c may be potential therapeutic targets in gastric cancer.


Subject(s)
Antigens, Neoplasm/metabolism , HSP27 Heat-Shock Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinase 13/metabolism , Neoplasm Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Stomach Neoplasms/metabolism , Animals , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Cell Growth Processes/physiology , Cell Line, Tumor , Female , Heat-Shock Proteins , Heterografts , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Nude , Middle Aged , Mitogen-Activated Protein Kinase 13/genetics , Molecular Chaperones , Neoplasm Metastasis , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Phosphorylation , Prognosis , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Stomach Neoplasms/blood supply , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transfection
15.
Pharmacol Res ; 139: 431-439, 2019 01.
Article in English | MEDLINE | ID: mdl-30503840

ABSTRACT

Inflammatory reaction and cell apoptosis are two important processes in intestinal ischemia/reperfusion (II/R) injury, and exploration of effective lead compounds against II/R injury via regulating inflammation and apoptosis is critical important. In this paper, the results indicated that dioscin significantly increased cell viability, and inhibited inflammation and apoptosis caused by hypoxia-reoxygenation (H/R) injury in IEC-6 cells. in vivo II/R injury, dioscin markedly suppressed inflamma- tion and apoptosis, improved pathological changes, and depressed chiu' score in rats. Mechanistic studies indicated that dioscin notably up-regulated the expression level of MAPK13 through decreasing miR-351-5p level, and thereby decreased the expression levels of p-PKD1, NF-κB, Apaf-1, cleaved Caspase-3 and cleaved Caspase-9. Furthermore, miR-351-5p mimic and inhibitor experiments in IEC-6 cells further proved that dioscin up-regulated MAPK13 expression by decreasing miR-351-5p level to inhibit inflammation and apoptosis. Therefore, dioscin showed protective effect against II/R injury via adjusting miR-351-5/MAPK13-mediated inflammation and apoptosis. Dioscin should be considered as one potent candidate and miR-351-5/ MAPK13 should be one effective drug target for the treatment of II/R injury.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Diosgenin/analogs & derivatives , Protective Agents/therapeutic use , Reperfusion Injury/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Cell Hypoxia/drug effects , Cell Line , Diosgenin/pharmacology , Diosgenin/therapeutic use , Intestinal Mucosa/metabolism , Male , MicroRNAs/metabolism , Mitogen-Activated Protein Kinase 13/metabolism , Protective Agents/pharmacology , Rats, Sprague-Dawley , Reperfusion Injury/metabolism
16.
PLoS Biol ; 16(7): e2004455, 2018 07.
Article in English | MEDLINE | ID: mdl-29979672

ABSTRACT

Adipose tissue has emerged as an important regulator of whole-body metabolism, and its capacity to dissipate energy in the form of heat has acquired a special relevance in recent years as potential treatment for obesity. In this context, the p38MAPK pathway has arisen as a key player in the thermogenic program because it is required for the activation of brown adipose tissue (BAT) thermogenesis and participates also in the transformation of white adipose tissue (WAT) into BAT-like depot called beige/brite tissue. Here, using mice that are deficient in p38α specifically in adipose tissue (p38αFab-KO), we unexpectedly found that lack of p38α protected against high-fat diet (HFD)-induced obesity. We also showed that p38αFab-KO mice presented higher energy expenditure due to increased BAT thermogenesis. Mechanistically, we found that lack of p38α resulted in the activation of the related protein kinase family member p38δ. Our results showed that p38δ is activated in BAT by cold exposure, and lack of this kinase specifically in adipose tissue (p38δ Fab-KO) resulted in overweight together with reduced energy expenditure and lower body and skin surface temperature in the BAT region. These observations indicate that p38α probably blocks BAT thermogenesis through p38δ inhibition. Consistent with the results obtained in animals, p38α was reduced in visceral and subcutaneous adipose tissue of subjects with obesity and was inversely correlated with body mass index (BMI). Altogether, we have elucidated a mechanism implicated in physiological BAT activation that has potential clinical implications for the treatment of obesity and related diseases such as diabetes.


Subject(s)
Adipose Tissue, Brown/enzymology , Adipose Tissue, Brown/physiology , Mitogen-Activated Protein Kinase 13/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/metabolism , Thermogenesis , Adipocytes, Brown/enzymology , Adult , Animals , Body Mass Index , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/prevention & control , Diet , Energy Metabolism , Enzyme Activation , Humans , MAP Kinase Signaling System , Male , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 13/metabolism , Models, Biological , Obesity/enzymology , Obesity/prevention & control , Uncoupling Protein 1/metabolism
17.
J Cell Sci ; 131(14)2018 07 26.
Article in English | MEDLINE | ID: mdl-29930081

ABSTRACT

Aggresome formation is a major strategy to enable cells to cope with proteasomal stress. Misfolded proteins are assembled into micro-aggregates and transported to the microtubule organizing center (MTOC) to form perinuclear aggresomes before their degradation through autophagy. So far, multiple factors have been identified as the activators of micro-aggregate formation, but much less is known about the regulatory mechanisms of their transport. Here, we report that proteasomal stress leads to the activation of p38 MAPK family members. Two of them, p38γ (MAPK12) and p38δ (MAPK13), are dispensable for micro-aggregate formation but are required for their targeting to the MTOC. Interestingly, p38δ promotes micro-aggregate transport by phosphorylating SQSTM1, a major scaffold protein that assembles soluble ubiquitylated proteins into micro-aggregates. Expression of the phospho-mimetic mutant of SQSTM1 in p38δ-knockout cells completely rescued their aggresome formation defects and enhanced their resistance to proteasomal stress to wild-type levels. This study reveals p38δ-mediated SQSTM1 phosphorylation as a critical signal for the targeting of micro-aggregates to the MTOC and provides direct evidence for the survival advantages associated with aggresome formation in cells under proteasomal stress.


Subject(s)
Mitogen-Activated Protein Kinase 13/metabolism , Proteasome Endopeptidase Complex/metabolism , Sequestosome-1 Protein/metabolism , Humans , Microtubule-Organizing Center/enzymology , Microtubule-Organizing Center/metabolism , Mitogen-Activated Protein Kinase 12/genetics , Mitogen-Activated Protein Kinase 12/metabolism , Mitogen-Activated Protein Kinase 13/genetics , Phosphorylation , Proteasome Endopeptidase Complex/genetics , Protein Aggregates , Protein Transport , Sequestosome-1 Protein/genetics
18.
Front Immunol ; 9: 65, 2018.
Article in English | MEDLINE | ID: mdl-29434594

ABSTRACT

p38 mitogen-activated protein kinase (MAPK) signal transduction pathways are essential regulators of the immune response. Particularly, p38γ and p38δ regulate many immune cell functions such as cytokine production, migration, or T cell activation; however, their involvement in immune cell development is largely unknown. Here, we analysed the role of p38 MAPK isoforms p38γ and p38δ in T cell differentiation in the thymus and in lymph nodes, using mice deficient in p38γ, p38δ, or in both. We found that the T cell differentiation program in the thymus was affected at different stages in p38γ-, p38δ-, and p38γ/δ-deficient mice, and also peripheral T cell homaeostasis was compromised. Particularly, p38δ deletion affects different stages of early CD4-CD8- double-negative thymocyte development, whereas lack of p38γ favours thymocyte positive selection from CD4+CD8+ double-positive to CD4+ or CD8+ single-positive cells. Our results identify unreported functions for p38γ and p38δ in T cells.


Subject(s)
Cell Differentiation , Mitogen-Activated Protein Kinase 12/metabolism , Mitogen-Activated Protein Kinase 13/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Animals , Biomarkers , Gene Knockdown Techniques , Lymphoid Tissue/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 12/genetics , Mitogen-Activated Protein Kinase 13/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Thymocytes/cytology , Thymocytes/metabolism
19.
Mol Med Rep ; 17(3): 4554-4560, 2018 03.
Article in English | MEDLINE | ID: mdl-29344643

ABSTRACT

The molecular mechanisms of osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) remain to be fully elucidated. MicroRNAs (miRs) serve vital roles in the process of regulating osteogenic differentiation of BMSCs. The present study aimed to investigate the role of miR­23a­5p in osteogenic differentiation of human (h)BMSCs, and the underlying molecular mechanism. The results of reverse transcription­quantitative polymerase chain reaction demonstrated that miR­23a­5p was significantly downregulated in the process of osteogenic differentiation. Upregulation of miR­23a­5p inhibited osteogenic differentiation of hBMSCs, and down­regulated expression of miR­23a­5p enhanced this process, which was confirmed by alkaline phosphatase (ALP) and Alizarin Red S staining. A dual­luciferase reporter assay confirmed that mitogen­activated protein kinase 13 (MAPK13) was a direct target of miR­23a­5p. In addition, knockdown of MAPK13 inhibited osteogenic differentiation of hBMSCs, similar to the effect of upregulation of miR­23a­5p. Finally, the knockdown of MAPK13 also blocked the effect of miR­23a­5p in osteogenic differentiation of hBMSCs, which was also confirmed by ALP and Alizarin Red S staining. These results indicated that by targeting MAPK13, miR­23a­5p serves a vital role in osteogenic differentiation of hBMSCs, which may provide novel clinical treatments for bone injury however, further studies are required.


Subject(s)
MicroRNAs/metabolism , Mitogen-Activated Protein Kinase 13/metabolism , 3' Untranslated Regions , Adult , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Antagomirs/metabolism , Bone Marrow Cells/cytology , Cell Differentiation , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Humans , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 13/antagonists & inhibitors , Mitogen-Activated Protein Kinase 13/genetics , Osteogenesis , Osteopontin/genetics , Osteopontin/metabolism , RNA Interference , RNA, Small Interfering/metabolism
20.
Assay Drug Dev Technol ; 16(1): 15-26, 2018 01.
Article in English | MEDLINE | ID: mdl-29148820

ABSTRACT

Protease-activated receptor 2 (PAR2) is a proinflammatory G-protein coupled receptor (GPCR) that is activated by inflammatory proteases, and its activation initiates signaling pathways that modulate the nonselective cation channel transient receptor potential vanilloid-4 (TRPV4). PAR2-dependent opening of TRPV4 has been attributed to kinase activation, but the identity of the responsible enzymes is unknown. Deciphering the signaling pathways involved in the PAR2-dependent opening of TRPV4 may yield new targets for pain treatment. This study has identified specific kinases that are involved in opening TRPV4, using a selective screen of short interfering ribonucleic acid (siRNA) SMARTpools, which individually targeted all human kinases, in human embryonic kidney 293 (HEK293) cells that stably express inducible TRPV4. This screen is unique because it uses a real-time assay measuring intracellular calcium with Fura-2AM dye. From the primary screen, subsequent confirmation screen, and on-target messenger ribonucleic acid expression analysis, we identified two kinases as crucial to the PAR2-dependent opening of TRPV4 in HEK293 cells, mitogen-activated protein kinase 13 and with no lysine kinase 4. In conclusion, this study describes a powerful new application of siRNA knockdown to identity signaling molecules that are responsible for the PAR2-dependent opening of TRPV4, which will help elucidate this signaling process.


Subject(s)
Mitogen-Activated Protein Kinase 13/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA/metabolism , Receptor, PAR-2/metabolism , TRPV Cation Channels/metabolism , Calcium/analysis , Calcium/metabolism , Drug Evaluation, Preclinical , HEK293 Cells , High-Throughput Screening Assays , Humans , Mitogen-Activated Protein Kinase 13/analysis , Protein Serine-Threonine Kinases/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...