Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 970
Filter
Add more filters

Publication year range
1.
Cell ; 145(7): 1049-61, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21703449

ABSTRACT

Atf1, the fission yeast homolog of activation transcription factor-2 (ATF-2), contributes to heterochromatin formation. However, the role of ATF-2 in chromatin assembly in higher organisms remains unknown. This study reveals that Drosophila ATF-2 (dATF-2) is required for heterochromatin assembly, whereas the stress-induced phosphorylation of dATF-2, via Mekk1-p38, disrupts heterochromatin. The dATF-2 protein colocalized with HP1, not only on heterochromatin but also at specific loci in euchromatin. Heat shock or osmotic stress induced phosphorylation of dATF-2 and resulted in its release from heterochromatin. This heterochromatic disruption was an epigenetic event that was transmitted to the next generation in a non-Mendelian fashion. When embryos were exposed to heat stress over multiple generations, the defective chromatin state was maintained over multiple successive generations, though it gradually returned to the normal state. The results suggest a mechanism by which the effects of stress are inherited epigenetically via the regulation of a tight chromatin structure.


Subject(s)
Activating Transcription Factor 2/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/genetics , Heterochromatin/metabolism , Activating Transcription Factor 2/analysis , Activating Transcription Factor 2/genetics , Animals , Drosophila Proteins/analysis , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Epigenomics , Euchromatin/metabolism , Female , Heterochromatin/chemistry , MAP Kinase Kinase Kinase 1/metabolism , Male , Mutagenesis , Phosphorylation , Signal Transduction , Stress, Physiological , p38 Mitogen-Activated Protein Kinases/metabolism
2.
J Biol Chem ; 300(7): 107486, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38897570

ABSTRACT

Aberrant regulation of signal transduction pathways can adversely derail biological processes for tissue development. One such process is the embryonic eyelid closure that is dependent on the mitogen-activated protein kinase kinase kinase 1 (MAP3K1). Map3k1 KO in mice results in defective eyelid closure and an autosomal recessive eye-open at birth phenotype. We have shown that in utero exposure to dioxin, a persistent environmental toxicant, induces the same eye defect in Map3k1+/- heterozygous but not WT pups. Here, we explore the mechanisms of the Map3k1 (gene) and dioxin (environment) interactions (GxE) underlying defective eyelid closure. We show that, acting through the aryl hydrocarbon receptor, dioxin activates epidermal growth factor receptor signaling, which in turn depresses MAP3K1-dependent Jun N-terminal kinase (JNK) activity. The dioxin-mediated JNK repression is moderate but is exacerbated by Map3k1 heterozygosity. Therefore, dioxin exposed Map3k1+/- embryonic eyelids have a marked reduction of JNK activity, accelerated differentiation and impeded polarization in the epithelial cells. Knocking out Ahr or Egfr in eyelid epithelium attenuates the open-eye defects in dioxin-treated Map3k1+/- pups, whereas knockout of Jnk1 and S1pr that encodes the sphigosin-1-phosphate (S1P) receptors upstream of the MAP3K1-JNK pathway potentiates the dioxin toxicity. Our novel findings show that the crosstalk of aryl hydrocarbon receptor, epidermal growth factor receptor, and S1P-MAP3K1-JNK pathways determines the outcome of dioxin exposure. Thus, gene mutations targeting these pathways are potential risk factors for the toxicity of environmental chemicals.


Subject(s)
Dioxins , ErbB Receptors , MAP Kinase Kinase Kinase 1 , Receptors, Aryl Hydrocarbon , Animals , Female , Mice , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Dioxins/toxicity , ErbB Receptors/metabolism , ErbB Receptors/genetics , Eyelids/metabolism , Eyelids/abnormalities , Gene-Environment Interaction , JNK Mitogen-Activated Protein Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Signaling System/drug effects , Mice, Knockout , Receptor Cross-Talk , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction/drug effects
3.
Crit Rev Immunol ; 44(6): 13-25, 2024.
Article in English | MEDLINE | ID: mdl-38848290

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia. Aberrant regulation of microRNAs (miRNAs) has been implicated in the pathogenesis of AD. In a large case-control study recruiting 208 patients with AD and 205 elderly control subjects, miRNA-let-7d-5p attracted our attention for its downregulated level in patients with AD. However, the biological functions of let-7d-5p in AD pathogenesis have not been investigated. This study emphasized the functions and mechanisms of let-7d-5p in the pathogenesis of AD. Mouse microglial BV2 cells treated with amyloid-ß (Aß)1-42 were used as in vitro AD inflammation models. We reported that let-7d-5p was downregulated in Aß1-42-stimulated BV2 cells, and upregulation of let-7d-5p promoted the transversion of microglial cells from Ml phenotype to M2 phenotype. Then, the binding relationship between let-7d-5p and Map3k1 was verified by luciferase reporter assays. Mechanistically, let-7d-5p could target Map3k1 3'UTR to inactivate ERK/p38 MAPK signaling. Therefore, it was suggested that let-7d-5p might be a novel modulator of microglial neuroinflammation and serve as a novel target for diagnosis and treatment of AD.


Subject(s)
Alzheimer Disease , MAP Kinase Signaling System , MicroRNAs , Microglia , p38 Mitogen-Activated Protein Kinases , MicroRNAs/genetics , MicroRNAs/metabolism , Microglia/metabolism , Microglia/immunology , Animals , Humans , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/immunology , Mice , p38 Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinase 1/genetics , Inflammation/genetics , Inflammation/immunology , Cell Line , Amyloid beta-Peptides/metabolism
4.
Proc Natl Acad Sci U S A ; 119(18): e2115071119, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35476515

ABSTRACT

Activation of inhibitor of nuclear factor NF-κB kinase subunit-ß (IKKß), characterized by phosphorylation of activation loop serine residues 177 and 181, has been implicated in the early onset of cancer. On the other hand, tissue-specific IKKß knockout in Kras mutation-driven mouse models stalled the disease in the precancerous stage. In this study, we used cell line models, tumor growth studies, and patient samples to assess the role of IKKß and its activation in cancer. We also conducted a hit-to-lead optimization study that led to the identification of 39-100 as a selective mitogen-activated protein kinase kinase kinase (MAP3K) 1 inhibitor. We show that IKKß is not required for growth of Kras mutant pancreatic cancer (PC) cells but is critical for PC tumor growth in mice. We also observed elevated basal levels of activated IKKß in PC cell lines, PC patient-derived tumors, and liver metastases, implicating it in disease onset and progression. Optimization of an ATP noncompetitive IKKß inhibitor resulted in the identification of 39-100, an orally bioavailable inhibitor with improved potency and pharmacokinetic properties. The compound 39-100 did not inhibit IKKß but inhibited the IKKß kinase MAP3K1 with low-micromolar potency. MAP3K1-mediated IKKß phosphorylation was inhibited by 39-100, thus we termed it IKKß activation modulator (IKAM) 1. In PC models, IKAM-1 reduced activated IKKß levels, inhibited tumor growth, and reduced metastasis. Our findings suggests that MAP3K1-mediated IKKß activation contributes to KRAS mutation-associated PC growth and IKAM-1 is a viable pretherapeutic lead that targets this pathway.


Subject(s)
MAP Kinase Kinase Kinase 1 , Pancreatic Neoplasms , Humans , I-kappa B Kinase/metabolism , Pancreatic Neoplasms/drug therapy , Protein Serine-Threonine Kinases , Pancreatic Neoplasms
5.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Article in English | MEDLINE | ID: mdl-34911761

ABSTRACT

Arterial remodeling is an important adaptive mechanism that maintains normal fluid shear stress in a variety of physiologic and pathologic conditions. Inward remodeling, a process that leads to reduction in arterial diameter, plays a critical role in progression of such common diseases as hypertension and atherosclerosis. Yet, despite its pathogenic importance, molecular mechanisms controlling inward remodeling remain undefined. Mitogen-activated protein kinases (MAPKs) perform a number of functions ranging from control of proliferation to migration and cell-fate transitions. While the MAPK ERK1/2 signaling pathway has been extensively examined in the endothelium, less is known about the role of the MEKK3/ERK5 pathway in vascular remodeling. To better define the role played by this signaling cascade, we studied the effect of endothelial-specific deletion of its key upstream MAP3K, MEKK3, in adult mice. The gene's deletion resulted in a gradual inward remodeling of both pulmonary and systematic arteries, leading to spontaneous hypertension in both vascular circuits and accelerated progression of atherosclerosis in hyperlipidemic mice. Molecular analysis revealed activation of TGFß-signaling both in vitro and in vivo. Endothelial-specific TGFßR1 knockout prevented inward arterial remodeling in MEKK3 endothelial knockout mice. These data point to the unexpected participation of endothelial MEKK3 in regulation of TGFßR1-Smad2/3 signaling and inward arterial remodeling in artery diseases.


Subject(s)
Hypertension, Pulmonary/pathology , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinase 3/metabolism , Transforming Growth Factor beta/metabolism , Vascular Remodeling/physiology , Animals , Gene Deletion , Gene Expression Regulation/drug effects , Genotype , Hindlimb/blood supply , Human Umbilical Vein Endothelial Cells , Humans , Hypertension, Pulmonary/metabolism , Ischemia , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Kinase Kinase 3/genetics , Mice , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Selective Estrogen Receptor Modulators/toxicity , Signal Transduction , Tamoxifen/toxicity , Transforming Growth Factor beta/genetics
6.
Proc Natl Acad Sci U S A ; 117(35): 21308-21318, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32817551

ABSTRACT

The MEKK1 protein is a pivotal kinase activator of responses to cellular stress. Activation of MEKK1 can trigger various responses, including mitogen-activated protein (MAP) kinases, NF-κB signaling, or cell migration. Notably, MEKK1 activity is triggered by microtubule-targeting chemotherapies, among other stressors. Here we show that MEKK1 contains a previously unidentified tumor overexpressed gene (TOG) domain. The MEKK1 TOG domain binds to tubulin heterodimers-a canonical function of TOG domains-but is unusual in that it appears alone rather than as part of a multi-TOG array, and has structural features distinct from previously characterized TOG domains. MEKK1 TOG demonstrates a clear preference for binding curved tubulin heterodimers, which exist in soluble tubulin and at sites of microtubule polymerization and depolymerization. Mutations disrupting tubulin binding decrease microtubule density at the leading edge of polarized cells, suggesting that tubulin binding may play a role in MEKK1 activity at the cellular periphery. We also show that MEKK1 mutations at the tubulin-binding interface of the TOG domain recur in patient-derived tumor sequences, suggesting selective enrichment of tumor cells with disrupted MEKK1-microtubule association. Together, these findings provide a direct link between the MEKK1 protein and tubulin, which is likely to be relevant to cancer cell migration and response to microtubule-modulating therapies.


Subject(s)
MAP Kinase Kinase Kinase 1/metabolism , Tubulin/metabolism , Humans , MAP Kinase Kinase Kinase 1/chemistry , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Kinase Kinase 1/ultrastructure , Neoplasms/genetics , Protein Domains
7.
Anim Biotechnol ; 34(3): 686-697, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37014133

ABSTRACT

MAP3K1 is a significant member of the MAPK family, and its expressed MEKK1 protein has a wide range of biological activities and is an essential node in the MAPK signaling pathway. A significant number of studies have revealed that MAP3K1 plays a complicated function in the control of cell proliferation, apoptosis, invasion and movement, participates in the regulation of the immune system, and plays an important role in wound healing, tumorigenesis and other processes. In this study, we looked at the involvement of MAP3K1 in the control of hair follicle stem cells (HFSCs). Overexpression of MAP3K1 significantly promoted the proliferation of HFSCs by inhibiting apoptosis and promoting the transition from S phase to G2 phase. The transcriptome identified 189 (MAP3K1_OE) and 414 (MAP3K1_sh) differential genes. The two pathways with the most significant enrichment of differentially expressed genes were the IL-17 signaling pathway and TNF signaling pathway, and the significantly enriched terms in the GO enrichment analysis involved regulation of response of external stimulus, inflammatory and cytokine. Indicate that MAP3K1 can function as a promoting factor in HFSCs through the induction of cell cycle transition from S phase to G2 phase can inhibition apoptosis by mediating crosstalk among several pathways and cytokines.HIGHLIGHTSAbnormal MAP3K1 expression in hair follicle stem cells (HFSCs) can impair HFSC proliferation and apoptosis.MAP3K1 controls hair follicle stem cell proliferation via modulating cell apoptosis and the ratio of cells in S phase/G2 phase.The differential genes shared by MAP3K1_sh and MAP3K1_OE are enriched in GO terms such as inflammation, adipocyte differentiation, acute inflammation, and so on.


Subject(s)
Hair Follicle , MAP Kinase Kinase Kinase 1 , Animals , Hair Follicle/metabolism , MAP Kinase Kinase Kinase 1/metabolism , Stem Cells/metabolism , Gene Expression Profiling , Cytokines/genetics , Cytokines/metabolism , Inflammation/metabolism
8.
Biochem Biophys Res Commun ; 612: 188-195, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35567901

ABSTRACT

Glioblastoma (GBM) stem cells (GSCs) are responsible for GBM initiation, progression, infiltration, standard therapy resistance, and recurrence. However, the mechanisms underlying GSC invasion remain incompletely understood. Using public single-cell RNA-Seq data, we identified MAP3K1 as a master regulator of infiltrative GSCs through c-JUN signaling regulation. MAP3K1 knockdown significantly decreased GSC invasion capacity, proliferation, and stemness in vitro. Moreover, in an orthotopic xenograft model, knockdown of MAP3K1 prominently suppressed GSC infiltration along the corpus callosum and tumor progression and prolonged mouse survival. Mechanistically, MAP3K1 regulates GSC invasion through phosphorylation of downstream c-JUN at serine 63 and 73, as confirmed using the CPTAC phosphoproteome dataset. Furthermore, the c-JUN inhibitor JNK-IN-8 significantly decreased GSC invasion, proliferation, and stemness. Taken together, our study demonstrates that MAP3K1 regulates GSC invasion and tumor progression via activation of c-JUN signaling and indicates that the MAP3K1/c-JUN signaling axis is a therapeutic target for infiltrative GBM.


Subject(s)
Brain Neoplasms , Glioblastoma , MAP Kinase Kinase Kinase 1 , Animals , Benzamides , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Glioblastoma/pathology , Humans , Mice , Neoplastic Stem Cells/pathology , Pyridines , Pyrimidines
9.
Circ Res ; 126(3): 330-346, 2020 01 31.
Article in English | MEDLINE | ID: mdl-31739742

ABSTRACT

Rationale: Genome-wide association studies have identified genetic loci associated with insulin resistance (IR) but pinpointing the causal genes of a risk locus has been challenging. Objective: To identify candidate causal genes for IR, we screened regional and biologically plausible genes (16 in total) near the top 10 IR-loci in risk-relevant cell types, namely preadipocytes and adipocytes. Methods and Results: We generated 16 human Simpson-Golabi-Behmel syndrome preadipocyte knockout lines each with a single IR-gene knocked out by lentivirus-mediated CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 system. We evaluated each gene knockout by screening IR-relevant phenotypes in the 3 insulin-sensitizing mechanisms, including adipogenesis, lipid metabolism, and insulin signaling. We performed genetic analyses using data on the genotype-tissue expression portal expression quantitative trait loci database and accelerating medicines partnership type 2 diabetes mellitus Knowledge Portal to evaluate whether candidate genes prioritized by our in vitro studies were expression quantitative trait loci genes in human subcutaneous adipose tissue, and whether expression of these genes is associated with risk of IR, type 2 diabetes mellitus, and cardiovascular diseases. We further validated the functions of 3 new adipose IR genes by overexpression-based phenotypic rescue in the Simpson-Golabi-Behmel syndrome preadipocyte knockout lines. Twelve genes, PPARG, IRS-1, FST, PEPD, PDGFC, MAP3K1, GRB14, ARL15, ANKRD55, RSPO3, COBLL1, and LYPLAL1, showed diverse phenotypes in the 3 insulin-sensitizing mechanisms, and the first 7 of these genes could affect all the 3 mechanisms. Five out of 6 expression quantitative trait loci genes are among the top candidate causal genes and the abnormal expression levels of these genes (IRS-1, GRB14, FST, PEPD, and PDGFC) in human subcutaneous adipose tissue could be associated with increased risk of IR, type 2 diabetes mellitus, and cardiovascular disease. Phenotypic rescue by overexpression of the candidate causal genes (FST, PEPD, and PDGFC) in the Simpson-Golabi-Behmel syndrome preadipocyte knockout lines confirmed their function in adipose IR. Conclusions: Twelve genes showed diverse phenotypes indicating differential roles in insulin sensitization, suggesting mechanisms bridging the association of their genomic loci with IR. We prioritized PPARG, IRS-1, GRB14, MAP3K1, FST, PEPD, and PDGFC as top candidate genes. Our work points to novel roles for FST, PEPD, and PDGFC in adipose tissue, with consequences for cardiometabolic diseases.


Subject(s)
Adipocytes/metabolism , Insulin Resistance/genetics , Quantitative Trait Loci , Adaptor Proteins, Signal Transducing/genetics , Cell Line , Dipeptidases/genetics , Follistatin/genetics , Genome-Wide Association Study/methods , Humans , Insulin Receptor Substrate Proteins/genetics , Loss of Function Mutation , Lymphokines/genetics , MAP Kinase Kinase Kinase 1/genetics , PPAR gamma/genetics , Platelet-Derived Growth Factor/genetics
10.
BMC Gastroenterol ; 22(1): 513, 2022 Dec 12.
Article in English | MEDLINE | ID: mdl-36510163

ABSTRACT

BACKGROUND/OBJECTIVES: The hormone-dependent effect of MAP3K1 gene polymorphisms may explain sex-specific differences in gastric cancer (GC) risk. Phytoestrogens have been shown to interact with this genetic factor. Here, we investigated the association between MAP3K1 gene polymorphisms and GC risk by sex and whether these associations differ depending on soy products intake. METHODS: Participants aged 20-79 years were recruited from two hospitals between December 2002 and September 2006. In all, 440 cases and 485 controls were recruited, among, 246 pairs of cases and controls, matched by sex, age (± 5 years), study admission period (± 1 years), and hospital, were included for the analysis. RESULTS: In dominant model, men with the A allele of rs252902 showed significantly increased GC risk (odd ratio; OR=2.19, 95% confidence interval; CI=1.31-3.64) compared to GG homozygotes. When stratified by intake of soy products, men with the A allele of rs252902 and low intake of soy products showed significantly higher GC risk (OR=3.29, 95% CI=1.55-6.78) than that in GG homozygotes. CONCLUSIONS: Men with the risk allele of MAP3K1 had a significantly increased GC risk compared to GG homozygotes; this trend was more pronounced in those with low intake of soy products.


Subject(s)
MAP Kinase Kinase Kinase 1 , Stomach Neoplasms , Male , Female , Humans , Stomach Neoplasms/genetics , Case-Control Studies , Polymorphism, Single Nucleotide , Alleles , Odds Ratio , Risk Factors , Genetic Predisposition to Disease , MAP Kinase Kinase Kinase 1/genetics
11.
Exp Cell Res ; 398(2): 112441, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33338478

ABSTRACT

The Yangtze River Delta white goat is a sole goat species that can naturally produce superior-quality brush hair. It's worth to mention that study the developmental mechanism of goat hair follicle stem cells is vital for future breed preservation and molecular breeding. In this study, we successfully isolated hair follicle stem cells from the skin tissue of fetal sheep neck spine, and harvested superior-quality and normal-quality brush hair goat tissue. The expression of miR-31-5p in goat hair follicle stem cells was verified by qPCR and Western blot. The effects of overexpression or inhibition of miR-31-5p on the proliferation and apoptosis of hair follicle stem cells were detected by EdU, CCK-8, flow cytometry, etc. miR-31-5p can significantly improve cell proliferation and inhibit cell apoptosis by targeting RASA1 and upregulating MAP3K1 level, whereas miR-31-5p knockdown led to an opposite effect. These results reveal a miR-31-5p-associated regulatory network between miR-31-5p and RASA1/MAP3K1 during the progression of superiorquality brush hair traits.


Subject(s)
Apoptosis , Hair Follicle/metabolism , MAP Kinase Kinase Kinase 1/metabolism , MicroRNAs/metabolism , Stem Cells/metabolism , p120 GTPase Activating Protein/metabolism , Animals , Cell Proliferation , Cells, Cultured , Goats
12.
J Clin Lab Anal ; 36(6): e24470, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35524422

ABSTRACT

BACKGROUND: Papillary thyroid carcinoma (PTC) grows slowly but has a great risk of metastasis. MicroRNAs are well known as vital tumor-related gene regulators. In PTC, the role of miR-203a-3p and the underlying mechanisms remain not completely understood. METHODS: We conducted CCK8 assay, wound healing assay, transwell experiment and flow cytometry analyses to investigate the function of miRNA-203a-3p. The interaction of miRNA-203a-3p with its gene MAP3K1 was characterized by quantitative real-time polymerase chain reaction, western blotting and luciferase assay. RESULTS: We found that the levels of miRNA-203a-3p were statistically decreased in PTC tissues. When mimics were delivered to TPC-1 and KTC-1 cells to upregulate miR-203a-3p, it was observed that cell proliferation, metastatic abilities and cell cycle process were prevented but cell apoptosis was enhanced. Furthermore, we proved the interaction between MAP3K1 and miR-203a-3p. Intriguingly, similar to miR-203a-3p mimics, siMAP3K1 showed a tumor-suppressive effect, and this effect could be reversed when miR-203a-3p was simultaneously inhibited. Finally, selected autophagy-linked proteins such as LC3 Beclin-1 were detected and found to be increased when miR-203a-3p was upregulated or MAP3K1 was inhibited. CONCLUSION: Overall, miR-203a-3p inhibits the oncogenic characteristics of TPC-1 and KTC-1 cells via suppressing MAP3K1 and activating autophagy. Our findings might enrich the understanding and the therapeutic strategies of PTC.


Subject(s)
Carcinoma, Papillary , MAP Kinase Kinase Kinase 1 , MicroRNAs , Thyroid Neoplasms , Autophagy/genetics , Carcinoma, Papillary/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Kinase Kinase 1/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology
13.
Hum Mol Genet ; 28(10): 1620-1628, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30608580

ABSTRACT

Missense mutations in the gene, MAP3K1, are a common cause of 46,XY gonadal dysgenesis, accounting for 15-20% of cases [Ostrer, 2014, Disorders of sex development (DSDs): an update. J. Clin. Endocrinol. Metab., 99, 1503-1509]. Functional studies demonstrated that all of these mutations cause a protein gain-of-function that alters co-factor binding and increases phosphorylation of the downstream MAP kinase pathway targets, MAPK11, MAP3K and MAPK1. This dysregulation of the MAP kinase pathway results in increased CTNNB1, increased expression of WNT4 and FOXL2 and decreased expression of SRY and SOX9. Unique and recurrent pathogenic mutations cluster in three semi-contiguous domains outside the kinase region of the protein, a newly identified N-terminal domain that shares homology with the Guanine Exchange Factor (residues Met164 to Glu231), a Plant HomeoDomain (residues Met442 to Trp495) and an ARMadillo repeat domain (residues Met566 to Glu862). Despite the presence of the mutation clusters and clinical data, there exists a dearth of mechanistic insights behind the development imbalance. In this paper, we use structural modeling and functional data of these mutations to understand alterations of the MAP3K1 protein and the effects on protein folding, binding and downstream target phosphorylation. We show that these mutations have differential effects on protein binding depending on the domains in which they occur. These mutations increase the binding of the RHOA, MAP3K4 and FRAT1 proteins and generally decrease the binding of RAC1. Thus, pathologies in MAP3K1 disrupt the balance between the pro-kinase activities of the RHOA and MAP3K4 binding partners and the inhibitory activity of RAC1.


Subject(s)
Disorders of Sex Development/genetics , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Kinase Kinase 4/genetics , rac1 GTP-Binding Protein/genetics , Adaptor Proteins, Signal Transducing/genetics , Armadillo Domain Proteins/genetics , Disorder of Sex Development, 46,XY , Disorders of Sex Development/pathology , Female , Forkhead Box Protein L2/genetics , Gene Expression Regulation/genetics , Gonadal Dysgenesis, 46,XY/genetics , Gonadal Dysgenesis, 46,XY/pathology , Humans , MAP Kinase Kinase Kinase 1/chemistry , MAP Kinase Kinase Kinase 4/chemistry , MAP Kinase Signaling System/genetics , Male , Mutation, Missense/genetics , Protein Binding/genetics , Proto-Oncogene Proteins/genetics , Sex-Determining Region Y Protein/genetics , rac1 GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/genetics
14.
Nat Immunol ; 10(9): 949-57, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19668222

ABSTRACT

All metazoan guts are in permanent contact with the microbial realm. However, understanding of the exact mechanisms by which the strength of gut immune responses is regulated to achieve gut-microbe mutualism is far from complete. Here we identify a signaling network composed of complex positive and negative mechanisms that controlled the expression and activity of dual oxidase (DUOX), which 'fine tuned' the production of microbicidal reactive oxygen species depending on whether the gut encountered infectious or commensal microbes. Genetic analyses demonstrated that negative and positive regulation of DUOX was required for normal host survival in response to colonization with commensal and infectious microbes, respectively. Thus, the coordinated regulation of DUOX enables the host to achieve gut-microbe homeostasis by efficiently combating infection while tolerating commensal microbes.


Subject(s)
Drosophila/immunology , NADPH Oxidases/physiology , Activating Transcription Factor 2/physiology , Animals , Caco-2 Cells , Calcineurin/physiology , Carrier Proteins/physiology , Gene Expression Regulation, Enzymologic , Humans , Intestines/immunology , Intestines/microbiology , MAP Kinase Kinase 3/physiology , MAP Kinase Kinase Kinase 1/physiology , NADPH Oxidases/genetics , Phospholipase C beta/physiology , Reactive Oxygen Species/metabolism , Signal Transduction , Transcription, Genetic , p38 Mitogen-Activated Protein Kinases/physiology
15.
Plant Physiol ; 183(1): 331-344, 2020 05.
Article in English | MEDLINE | ID: mdl-32165446

ABSTRACT

A wide variety of intrinsic and extrinsic cues lead to cell death with unclear mechanisms. The infertility of some death mutants often hurdles the classical suppressor screens for death regulators. We have developed a transient RNA interference (RNAi)-based screen using a virus-induced gene silencing approach to understand diverse cell death pathways in Arabidopsis (Arabidopsis thaliana). One death pathway is due to the depletion of a MAP kinase (MAPK) cascade, consisting of MAPK kinase kinase 1 (MEKK1), MKK1/2, and MPK4, which depends on a nucleotide-binding site Leu-rich repeat (NLR) protein SUMM2. Silencing of MEKK1 by virus-induced gene silencing resembles the mekk1 mutant with autoimmunity and defense activation. The RNAi-based screen toward Arabidopsis T-DNA insertion lines identified SUMM2, MEKK2, and Calmodulin-binding receptor-like cytoplasmic kinase 3 (CRCK3) to be vital regulators of RNAi MEKK1-induced cell death, consistent with the reports of their requirement in the mekk1-mkk1/2-mpk4 death pathway. Similar with MEKK2, overexpression of CRCK3 caused dosage- and SUMM2-dependent cell death, and the transcripts of CRCK3 were up-regulated in mekk1, mkk1/2, and mpk4 MEKK2-induced cell death depends on CRCK3. Interestingly, CRCK3-induced cell death also depends on MEKK2, consistent with the biochemical data that MEKK2 complexes with CRCK3. Furthermore, the kinase activity of CRCK3 is essential, whereas the kinase activity of MEKK2 is dispensable, for triggering cell death. Our studies suggest that MEKK2 and CRCK3 exert concerted functions in the control of NLR SUMM2 activation and MEKK2 may play a structural role, rather than function as a kinase, in regulating CRCK3 protein stability.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinase 2/genetics , MAP Kinase Kinase Kinase 2/metabolism , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Protein Stability , RNA Interference/physiology
16.
Int J Mol Sci ; 22(18)2021 Sep 17.
Article in English | MEDLINE | ID: mdl-34576208

ABSTRACT

Sex determination triggers the differentiation of the bi-potential gonad into either an ovary or testis. In non-mammalian vertebrates, the presence or absence of oestrogen dictates gonad differentiation, while in mammals, this mechanism has been supplanted by the testis-determining gene SRY. Exogenous oestrogen can override this genetic trigger to shift somatic cell fate in the gonad towards ovarian developmental pathways by limiting the bioavailability of the key testis factor SOX9 within somatic cells. Our previous work has implicated the MAPK pathway in mediating the rapid cellular response to oestrogen. We performed proteomic and phosphoproteomic analyses to investigate the precise mechanism through which oestrogen impacts these pathways to activate ß-catenin-a factor essential for ovarian development. We show that oestrogen can activate ß-catenin within 30 min, concomitant with the cytoplasmic retention of SOX9. This occurs through changes to the MAP3K1 cascade, suggesting this pathway is a mechanism through which oestrogen influences gonad somatic cell fate. We demonstrate that oestrogen can promote the shift from SOX9 pro-testis activity to ß-catenin pro-ovary activity through activation of MAP3K1. Our findings define a previously unknown mechanism through which oestrogen can promote a switch in gonad somatic cell fate and provided novel insights into the impacts of exogenous oestrogen exposure on the testis.


Subject(s)
MAP Kinase Kinase Kinase 1/metabolism , beta Catenin/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Estrogens/pharmacology , Humans , MAP Kinase Kinase Kinase 1/genetics , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism
17.
J Cell Physiol ; 235(5): 4466-4480, 2020 05.
Article in English | MEDLINE | ID: mdl-31643095

ABSTRACT

Bone remodeling is a strictly regulated dynamic process that cycles between bone formation and resorption, and interleukin-17 (IL-17) critically orchestrates the activation and differentiation of both osteoblasts and osteoclasts. Mesenchymal stem cells (MSCs) within their native environment receive biochemical stimuli from surrounding cells that influences their differentiation into bone precursors, while the roles of osteocytes in regulating the osteogenic differentiation of MSCs remain unclear. This study investigated the specific roles of IL-17 signaling cascades and osteocyte-specific pathways in the osteogenesis of MSCs. Using a transwell coculture (CC) system, we explored the effects of osteocytes and osteoblasts on the osteogenesis of MSCs with and without IL-17 supplementation. A polycaprolactone (PCL) three-dimensional (3D) culture model was used to evaluate their osteogenic potential in the presence of osteocytes and IL-17. Notably, IL-17 induced osteogenesis in MSCs, which could be attenuated by blocking IL-17 receptor A. The osteogenic differentiation of MSCs promoted by IL-17 was further enhanced by CC with osteocytes. Moreover, proinflammatory cytokines IL-6 and IL-1ß played an important role in IL-17-dependent differentiation, via the phosphorylation of AKT, signal transducer and activator of transcription 3, and extracellular signal-regulated kinase 1/2 signaling pathways in the MSC niche. The present study confirms a synergistic effect of osteocytes and IL-17 in the production of biochemical signals to stimulate the osteogenic differentiation of MSCs, which could be further promoted in the PCL 3D-scaffold. These findings provide important insight into the mechanisms of MSCs activation and osteogenic differentiation within the native stem cell niche, and suggest a possible role of IL-17 in bone tissue engineering.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells/physiology , Osteocytes/physiology , Osteogenesis/physiology , Animals , Antibodies , Bone Marrow Cells , Cell Culture Techniques , Cell Line , Interleukin-17/pharmacology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinase 2/genetics , MAP Kinase Kinase Kinase 2/metabolism , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Up-Regulation
18.
Int J Cancer ; 146(6): 1606-1617, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31310010

ABSTRACT

Using a method optimized in hepatocellular carcinoma (HCC), we established patient-derived xenograft (PDX) models with an increased take rate (42.2%) and demonstrated that FBS +10% dimethyl sulfoxide exhibited the highest tumor take rate efficacy. Among 254 HCC patients, 103 stably transplantable xenograft lines that could be serially passaged, cryopreserved and revived were established. These lines maintained the diversity of HCC and the essential features of the original specimens at the histological, transcriptome, proteomic and genomic levels. Tumor engraftment was associated with lack of encapsulation, poor tumor differentiation, large size and overexpression of cancer stem cell biomarkers, and was an independent predictor for overall survival and tumor recurrence after resection. To confirm the preclinical value of the PDX model in HCC treatment, several antitumor agents were tested in 16 selected PDX models. The results revealed a high degree of pharmacologic heterogeneity in the cohort, as well as heterogeneity to different agents in the same individual. The sorafenib responses observed between HCC patients and the corresponding PDXs were also consistent. After molecular characterization of the PDX models, we explored the predictive markers for sorafenib response and found that mitogen-activated protein kinase kinase kinase 1 (MAP3K1) might play an important role in sorafenib resistance and sorafenib response is impaired in patients with MAP3K1 downexpression. Our results indicated that PDX models could accurately reproduce patient tumors biology and could aid in the discovery of new treatments to advance in precision medicine.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/therapy , Xenograft Model Antitumor Assays , Animals , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Chemoradiotherapy, Adjuvant/methods , Down-Regulation , Drug Resistance, Neoplasm , Female , Follow-Up Studies , Gene Expression Profiling , Genomics , Hepatectomy , Humans , Liver Neoplasms/diagnosis , Liver Neoplasms/pathology , MAP Kinase Kinase Kinase 1/metabolism , Male , Middle Aged , Proof of Concept Study , Prospective Studies , Protein Kinase Inhibitors/administration & dosage , Sorafenib/administration & dosage , Treatment Outcome
19.
J Cell Sci ; 131(4)2018 02 20.
Article in English | MEDLINE | ID: mdl-29361535

ABSTRACT

Glomerular matrix accumulation is the hallmark of diabetic nephropathy. The metalloprotease ADAM17 mediates high glucose (HG)-induced matrix production by kidney mesangial cells through release of ligands for the epidermal growth factor receptor. Here, we study the mechanism by which HG activates ADAM17. We find that the C-terminus is essential for ADAM17 activation and the profibrotic response to HG. In the C-terminus, Src-mediated Y702 phosphorylation and PI3K-MEK-Erk-mediated T735 phosphorylation are crucial for ADAM17 activation, both are also required for the HG-induced increase in cell surface mature ADAM17. The non-receptor tyrosine kinase FAK is a central mediator of these processes. These data not only support a crucial role for the C-terminus in ADAM17 activation and downstream profibrotic responses to HG, but also highlight FAK as a potential alternative therapeutic target for diabetic nephropathy.


Subject(s)
ADAM17 Protein/genetics , Diabetic Nephropathies/genetics , Focal Adhesion Kinase 1/genetics , Glucose/metabolism , Diabetic Nephropathies/pathology , Diabetic Nephropathies/therapy , ErbB Receptors/genetics , Humans , Kidney/metabolism , Kidney/pathology , Ligands , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Signaling System/genetics , Mesangial Cells/metabolism , Mesangial Cells/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation , Transcriptional Activation/genetics , src-Family Kinases/genetics
20.
Breast Cancer Res Treat ; 179(2): 387-401, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31620934

ABSTRACT

PURPOSE: Hormone receptor-positive (HR+) and human epidermal growth factor receptor 2 negative (HER2-) early breast cancer (BC) is the most prevalent BC subtype with substantial biological heterogeneity. Although clinicopathological (CP) characteristics have a clear prognostic value, additional biomarkers could refine survival prediction and guide treatment decision. METHODS: Copy number aberrations and somatic driver mutations were obtained with OncoScan CGH array and sequencing of 36 genes on HR+/HER2- node-positive early BC patients treated with chemotherapy from the PACS04 trial. We built a two-gene genomic score (GS) associated with distant disease-free survival (DDFS), whose prognostic value was assessed on the external METABRIC data (n = 1413) using overall survival (OS) and breast cancer-specific survival (BCSS). RESULTS: In the PACS04 trial (n = 327), the median follow-up for DDFS (65 events) was 9.6 years. FGFR1 amplifications ([Formula: see text] = 2.44, 95% CI [1.25; 4.76], p = 0.009) and MAP3K1 mutations ([Formula: see text] = 0.10, [0.01; 0.78], p = 0.03) were associated with DDFS beyond CP characteristics. A prognostic GS combining FGFR1 amplifications and MAP3K1 mutations added more information to CP model ([Formula: see text] = 12.97, [Formula: see text] < 0.001 and [Formula: see text] = 11.52, [Formula: see text] < 0.001). In the METABRIC study (n = 1413), FGFR1 amplifications ([Formula: see text] = 2.00 [1.40; 2.87], p < 0.001) and MAP3K1 mutations ([Formula: see text] = 0.58 [0.41; 0.83], p = 0.003) were significantly associated with BCSS beyond CP characteristics. The prognostic GS added significant prognostic information to CP model ([Formula: see text] = 15.39, [Formula: see text] < 0.001 and [Formula: see text] = 5.62, [Formula: see text] = 0.02). CONCLUSION: In axillary node-positive, HR+, and HER2- early BC, amplifications of FGFR1 gene were strongly associated with increased risk for distant disease, while mutations of MAP3K1 gene were significantly associated with decreased risk.


Subject(s)
Biomarkers, Tumor , Breast Neoplasms/genetics , Breast Neoplasms/mortality , DNA Copy Number Variations , MAP Kinase Kinase Kinase 1/genetics , Mutation , Receptor, Fibroblast Growth Factor, Type 1/genetics , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Axilla/pathology , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Clinical Trials, Phase III as Topic , Female , Humans , Lymph Nodes/pathology , Lymphatic Metastasis , MAP Kinase Kinase Kinase 1/metabolism , Neoplasm Staging , Prognosis , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL