Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
J Biol Chem ; 298(9): 102310, 2022 09.
Article in English | MEDLINE | ID: mdl-35921893

ABSTRACT

Disruption of fetal growth results in severe consequences to human health, including increased fetal and neonatal morbidity and mortality, as well as potential lifelong health problems. Molecular mechanisms promoting fetal growth represent potential therapeutic strategies to treat and/or prevent fetal growth restriction (FGR). Here, we identify a previously unknown role for the mitogen-activated protein kinase kinase kinase 4 (MAP3K4) in promoting fetal and placental growth. We demonstrate that inactivation of MAP3K4 kinase activity causes FGR due in part to placental insufficiency. Significantly, MAP3K4 kinase-inactive mice display highly penetrant lethality prior to weaning and persistent growth reduction of surviving adults. Additionally, we elucidate molecular mechanisms by which MAP3K4 promotes growth through control of the insulin-like growth factor 1 receptor (IGF1R), insulin receptor (IR), and Akt signaling pathway. Specifically, MAP3K4 kinase inactivation in trophoblast stem (TS) cells results in reduced IGF1R and IR expression and decreased Akt activation. We observe these changes in TS cells also occur in differentiated trophoblasts created through in vitro differentiation of cultured TS cells and in vivo in placental tissues formed by TS cells. Furthermore, we show that MAP3K4 controls this pathway by promoting Igf1r transcript expression in TS cells through activation of CREB-binding protein (CBP). In the MAP3K4 kinase-inactive TS cells, Igf1r transcripts are repressed because of reduced CBP activity and increased histone deacetylase 6 expression and activity. Together, these data demonstrate a critical role for MAP3K4 in promoting fetal and placental growth by controlling the activity of the IGF1R/IR and Akt signaling pathway.


Subject(s)
Fetal Development , MAP Kinase Kinase Kinase 4 , Placenta , Placentation , Receptor, IGF Type 1 , Receptor, Insulin , Adult , Animals , CREB-Binding Protein/metabolism , Female , Fetal Growth Retardation/genetics , Fetal Growth Retardation/metabolism , Histone Deacetylase 6/metabolism , Humans , MAP Kinase Kinase Kinase 4/genetics , MAP Kinase Kinase Kinase 4/metabolism , Mice , Placenta/enzymology , Pregnancy , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Signal Transduction
2.
FASEB J ; 35(10): e21948, 2021 10.
Article in English | MEDLINE | ID: mdl-34569098

ABSTRACT

Aminoacyl-tRNA synthetases (aaRSs) are house-keeping enzymes that are essential for protein synthesis. However, it has become increasingly evident that some aaRSs also have non-translational functions. Here we report the identification of a non-translational function of threonyl-tRNA synthetase (ThrRS) in myogenic differentiation. We find that ThrRS negatively regulates myoblast differentiation in vitro and injury-induced skeletal muscle regeneration in vivo. This function is independent of amino acid binding or aminoacylation activity of ThrRS, and knockdown of ThrRS leads to enhanced differentiation without affecting the global protein synthesis rate. Furthermore, we show that the non-catalytic new domains (UNE-T and TGS) of ThrRS are both necessary and sufficient for the myogenic function. In searching for a molecular mechanism of this new function, we find the kinase JNK to be a downstream target of ThrRS. Our data further reveal MEKK4 and MKK4 as upstream regulators of JNK in myogenesis and the MEKK4-MKK4-JNK pathway to be a mediator of the myogenic function of ThrRS. Finally, we show that ThrRS physically interacts with Axin1, disrupts Axin1-MEKK4 interaction and consequently inhibits JNK signaling. In conclusion, we uncover a non-translational function for ThrRS in the maintenance of homeostasis of skeletal myogenesis and identify the Axin1-MEKK4-MKK4-JNK signaling axis to be an immediate target of ThrRS action.


Subject(s)
JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Muscle Development , Threonine-tRNA Ligase/metabolism , Animals , Axin Protein/metabolism , Female , MAP Kinase Kinase 4/metabolism , MAP Kinase Kinase Kinase 4/metabolism , Male , Mice , Mice, Inbred C57BL , Protein Binding , Protein Biosynthesis , Protein Domains , Threonine-tRNA Ligase/chemistry
3.
Biol Reprod ; 105(2): 491-502, 2021 08 03.
Article in English | MEDLINE | ID: mdl-33912929

ABSTRACT

Sex determination requires the commitment of bipotential gonads to either a testis or an ovarian fate. Gene deletion of the kinase Map3k4 results in gonadal sex reversal in XY mice, and transgenic re-expression of Map3k4 rescues the sex reversal phenotype. Map3k4 encodes a large, multi-functional protein possessing a kinase domain and several, additional protein-protein interaction domains. Although MAP3K4 plays a critical role in male gonadal sex determination, it is unknown if the kinase activity of MAP3K4 is required. Here, we use mice expressing full-length, kinase-inactive MAP3K4 from the endogenous Map3k4 locus to examine the requirement of MAP3K4 kinase activity in sex determination. Although homozygous kinase-inactivation of MAP3K4 (Map3k4KI/KI) is lethal, a small fraction survive to adulthood. We show Map3k4KI/KI adults exhibit a 4:1 female-biased sex ratio. Many adult Map3k4KI/KI phenotypic females have a Y chromosome. XY Map3k4KI/KI adults with sex reversal display female mating behavior, but do not give rise to offspring. Reproductive organs are overtly female, but there is a broad spectrum of ovarian phenotypes, including ovarian absence, primitive ovaries, reduced ovarian size, and ovaries having follicles in all stages of development. Further, XY Map3k4KI/KI adults are smaller than either male or female Map3k4WT/WT mice. Examination of the critical stage of gonadal sex determination at E11.5 shows that loss of MAP3K4 kinase activity results in the loss of Sry expression in XY Map3k4KI/KI embryos, indicating embryonic male gonadal sex reversal. Together, these findings demonstrate the essential role for kinase activity of MAP3K4 in male gonadal sex determination.


Subject(s)
MAP Kinase Kinase Kinase 4/genetics , Mice/genetics , Ovary/embryology , Sex Determination Processes/genetics , Testis/embryology , Animals , Female , MAP Kinase Kinase Kinase 4/metabolism , Male , Mice/embryology
4.
Oncol Rep ; 45(4)2021 04.
Article in English | MEDLINE | ID: mdl-33649790

ABSTRACT

Chondroitin sulfate proteoglycan 4 (CSPG4) is a multifunctional transmembrane proteoglycan involved in spreading, migration and invasion of melanoma. In addition to the activating BRAF V600E mutation, CSPG4 was shown to promote MAPK signaling by mediating the growth­factor induced activation of receptor tyrosine kinases. However, it remains elusive which factors regulate CSPG4 expression. Therefore, the aim of the present study was to examine whether BRAF and MEK inhibitors have an effect on the expression of CSPG4. We exposed a panel of BRAF­mutant CSPG4­positive or ­negative melanoma cell lines to BRAF and MEK inhibitors. Protein levels of CSPG4 were analyzed by flow cytometry (FACS), immunofluorescence microscopy (IF), and western blotting. CSPG4 mRNA levels were determined by quantitative PCR (qPCR). The prolonged exposure of cells to BRAF and MEK inhibitors resulted in markedly reduced levels of the CSPG4 protein in permanent resistant melanoma cells as well as decreased levels of its mRNA. We did not observe increasing levels of CSPG4 shedding into the culture supernatants. In addition, patient­derived matched tumor samples following therapy with kinase inhibitors showed decreased numbers of CSPG4­positive cells as compared to pre­therapy tumor samples. Our results indicate that BRAF and MEK inhibition downregulates CSPG4 expression until the cells have developed permanent resistance. Our findings provide the basis for further investigation of the role of CSPG4 in the development of drug­resistance in melanoma cells.


Subject(s)
Chondroitin Sulfate Proteoglycans/metabolism , Melanoma/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Cell Line, Tumor , Chondroitin Sulfate Proteoglycans/genetics , Disease Progression , Down-Regulation , Drug Resistance, Neoplasm , Humans , MAP Kinase Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Kinase Kinase 4/metabolism , Melanoma/drug therapy , Melanoma/genetics , Membrane Proteins/genetics , Mutation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
FASEB J ; 35(1): e21133, 2021 01.
Article in English | MEDLINE | ID: mdl-33184917

ABSTRACT

Chronic vascular inflammation plays a key role in the pathogenesis of atherosclerosis. Long non-coding RNAs (lncRNAs) have emerged as essential inflammation regulators. We identify a novel lncRNA termed lncRNA-MAP3K4 that is enriched in the vessel wall and regulates vascular inflammation. In the aortic intima, lncRNA-MAP3K4 expression was reduced by 50% during the progression of atherosclerosis (chronic inflammation) and 70% during endotoxemia (acute inflammation). lncRNA-MAP3K4 knockdown reduced the expression of key inflammatory factors (eg, ICAM-1, E-selectin, MCP-1) in endothelial cells or vascular smooth muscle cells and decreased monocytes adhesion to endothelium, as well as reducing TNF-α, IL-1ß, COX2 expression in macrophages. Mechanistically, lncRNA-MAP3K4 regulates inflammation through the p38 MAPK signaling pathway. lncRNA-MAP3K4 shares a bidirectional promoter with MAP3K4, an upstream regulator of the MAPK signaling pathway, and regulates its transcription in cis. lncRNA-MAP3K4 and MAP3K4 show coordinated expression in response to inflammation in vivo and in vitro. Similar to lncRNA-MAP3K4, MAP3K4 knockdown reduced the expression of inflammatory factors in several different vascular cells. Furthermore, lncRNA-MAP3K4 and MAP3K4 knockdown showed cooperativity in reducing inflammation in endothelial cells. Collectively, these findings unveil the role of a novel lncRNA in vascular inflammation by cis-regulating MAP3K4 via a p38 MAPK pathway.


Subject(s)
Gene Expression Regulation , MAP Kinase Kinase Kinase 4/metabolism , MAP Kinase Signaling System , RNA, Long Noncoding/metabolism , Vasculitis/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , MAP Kinase Kinase Kinase 4/genetics , Mice , RNA, Long Noncoding/genetics , Vasculitis/genetics , Vasculitis/pathology , p38 Mitogen-Activated Protein Kinases/genetics
6.
Nat Commun ; 11(1): 5573, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33149122

ABSTRACT

Non-coding mutations can create splice sites, however the true extent of how such somatic non-coding mutations affect RNA splicing are largely unexplored. Here we use the MiSplice pipeline to analyze 783 cancer cases with WGS data and 9494 cases with WES data, discovering 562 non-coding mutations that lead to splicing alterations. Notably, most of these mutations create new exons. Introns associated with new exon creation are significantly larger than the genome-wide average intron size. We find that some mutation-induced splicing alterations are located in genes important in tumorigenesis (ATRX, BCOR, CDKN2B, MAP3K1, MAP3K4, MDM2, SMAD4, STK11, TP53 etc.), often leading to truncated proteins and affecting gene expression. The pattern emerging from these exon-creating mutations suggests that splice sites created by non-coding mutations interact with pre-existing potential splice sites that originally lacked a suitable splicing pair to induce new exon formation. Our study suggests the importance of investigating biological and clinical consequences of noncoding splice-inducing mutations that were previously neglected by conventional annotation pipelines. MiSplice will be useful for automatically annotating the splicing impact of coding and non-coding mutations in future large-scale analyses.


Subject(s)
Neoplasms/genetics , RNA Precursors/genetics , RNA Splice Sites , RNA Splicing , AMP-Activated Protein Kinase Kinases , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Databases, Genetic , Exons , Gene Expression Regulation, Neoplastic/genetics , Humans , Introns , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinase 4/genetics , MAP Kinase Kinase Kinase 4/metabolism , Mutation , Neoplasms/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , RNA, Untranslated , RNA-Seq , Repressor Proteins/genetics , Repressor Proteins/metabolism , Smad4 Protein/genetics , Smad4 Protein/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Exome Sequencing , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism
7.
Commun Biol ; 3(1): 475, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32859943

ABSTRACT

Coordinated gene expression is required for phenotypic switching between epithelial and mesenchymal phenotypes during normal development and in disease states. Trophoblast stem (TS) cells undergo epithelial-mesenchymal transition (EMT) during implantation and placentation. Mechanisms coordinating gene expression during these processes are poorly understood. We have previously demonstrated that MAP3K4-regulated chromatin modifiers CBP and HDAC6 each regulate thousands of genes during EMT in TS cells. Here we show that CBP and HDAC6 coordinate expression of only 183 genes predicted to be critical regulators of phenotypic switching. The highest-ranking co-regulated gene is the NF-κB family member Rel. Although NF-κB is primarily regulated post-transcriptionally, CBP and HDAC6 control Rel transcript levels by binding Rel regulatory regions and controlling histone acetylation. REL re-expression in mesenchymal-like TS cells induces a mesenchymal-epithelial transition. Importantly, REL forms a feedback loop, blocking HDAC6 expression and nuclear localization. Together, our work defines a developmental program coordinating phenotypic switching.


Subject(s)
Gene Expression Regulation , Histone Deacetylase 6/metabolism , MAP Kinase Kinase Kinase 4/metabolism , Oncogene Proteins v-rel/genetics , Peptide Fragments/metabolism , Phenotype , Sialoglycoproteins/metabolism , Animals , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Male , Mice , Models, Biological , Protein Transport , Proto-Oncogene Proteins c-met/metabolism , Stem Cells/metabolism , Transcription Factors
8.
Cell Death Dis ; 11(8): 684, 2020 08 12.
Article in English | MEDLINE | ID: mdl-32826872

ABSTRACT

Autophagy and apoptosis, which are important processes for host immunity, are commonly exploited by viruses to facilitate their survival. However, to the best of our knowledge, very few studies have researched the mechanisms of action of the autophagic and apoptotic signaling pathways following viral infection. Thus, the present study aimed to investigate the mechanisms of action of growth arrest and DNA-damage-inducible ß (GADD45ß), an important resistance gene involved in the host resistance to ALV-J. Both ALV-J infection and the overexpression of GADD45ß inhibited autophagy during the early stages, which prevented the autophagosomes from binding to the lysosomes and resulted in an incomplete autophagic flux. Notably, GADD45ß was discovered to interact with MEKK4 in DF-1 cells. The genetic knockdown of GADD45ß and MEKK4 using small interfering RNA-affected ALV-J infection, which suggested that ALV-J may promote the binding of GADD45ß to MEKK4 to activate the p38MAPK signaling pathway, which subsequently inhibits autophagy. Furthermore, ALV-J was revealed to affect the autophagic pathway prior to affecting the apoptotic pathway. In conclusion, to the best of our knowledge, the present study was the first to investigate the combined effects of ALV-J infection on autophagy and apoptosis, and to suggest that ALV-J inhibits autophagy via the GADD45ß/MEKK4/p38MAPK signaling pathway.


Subject(s)
Antigens, Differentiation/metabolism , Autophagy/physiology , Avian Leukosis Virus/metabolism , Animals , Apoptosis/physiology , Avian Leukosis Virus/genetics , Avian Leukosis Virus/pathogenicity , Cell Line , Chick Embryo , Chickens/genetics , Host-Pathogen Interactions/physiology , MAP Kinase Kinase Kinase 4/metabolism , MAP Kinase Signaling System/physiology , RNA, Small Interfering/metabolism , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Mol Med Rep ; 22(2): 1195-1204, 2020 08.
Article in English | MEDLINE | ID: mdl-32468015

ABSTRACT

Interleukin (IL)­1ß is a key promotor in the pathogenesis of temporomandibular joint osteoarthritis. Differentiation of stem cells to cartilage is a crucial repair mechanism of articular cartilage damage, and IL­1ß has been reported to impede the differentiation by upregulating the secretion of IL­6, an important inflammatory factor. Long non­coding RNAs (lncRNAs) regulate a number of physiological and pathological processes, but whether lncRNA AK094629 contributes to the IL­1ß mediated induction of inflammation remains unclear. Therefore, the aim of the present study was to investigate the effect of AK094629 on IL­1ß­induced IL­6 expression in synovial­derived mesenchymal stem cells (SMSCs) of the temporomandibular joints. The results of the present study demonstrated that the expression of AK094629 in the synovial tissue of patients with osteoarthritis was positively correlated with IL­1ß. In addition, IL­1ß upregulated the expression of AK094629 in the SMSCs in vitro, and AK094629 knockdown inhibited the IL­1ß mediated upregulation of IL­6. The present study also demonstrated that AK094629 knockdown downregulated the expression of the mitogen­activated protein kinase kinase kinase 4 (MAP3K4), which is upregulated by IL­1ß, whereas knockdown of MAP3K4 did not affect the expression of AK094629, but reversed the upregulation of IL­6 in SMSCs. In conclusion, AK094629 knockdown attenuated the expression of IL­1ß­regulated IL­6 in the SMSCs of the temporomandibular joint by inhibiting MAP3K4. Therefore, AK094629 may be a potential novel therapeutic target for the treatment of temporomandibular joint osteoarthritis.


Subject(s)
Interleukin-1beta/metabolism , Interleukin-6/metabolism , Mesenchymal Stem Cells/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Synovial Membrane/metabolism , Temporomandibular Joint/metabolism , Cells, Cultured , Gene Knockdown Techniques , Humans , Interleukin-6/genetics , MAP Kinase Kinase Kinase 4/genetics , MAP Kinase Kinase Kinase 4/metabolism , Mesenchymal Stem Cells/pathology , Osteoarthritis/etiology , Osteoarthritis/metabolism , Osteoarthritis/pathology , Synovial Membrane/cytology , Temporomandibular Joint/pathology , Temporomandibular Joint Disorders/etiology , Temporomandibular Joint Disorders/metabolism , Temporomandibular Joint Disorders/pathology , Transcriptional Activation/genetics , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Sci Rep ; 9(1): 10823, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31346208

ABSTRACT

One of the major features of cancer is Otto Warburg's observation that many tumors have increased extracellular acidification compared to healthy tissues. Since Warburg's observation, the importance of extracellular acidification in cancer is now considered a hallmark of cancer. Human MAP3K4 functions upstream of the p38 and JNK mitogen activated protein kinases (MAPKs). Additionally, MAP3K4 is required for cell migration and extracellular acidification of breast cancer cells in response to HER2/HER3 signaling. Here, we demonstrate that GIT1 interacts with MAP3K4 by immunoprecipitation, while cellular lactate production and the capacity of MCF-7 cells for anchorage independent growth in soft agar were dependent on GIT1. Additionally, we show that activation of HER2/HER3 signaling leads to reduced expression of lactate receptor (GPR81) mRNA and that both, GIT1 and MAP3K4, are necessary for constitutive expression of GPR81 mRNA. Our study suggests that targeting downstream proteins in the HER2/HER3-induced extracellular lactate signaling pathway may be a way to inhibit the Warburg Effect to disrupt tumor growth.


Subject(s)
Lactic Acid/metabolism , MAP Kinase Kinase Kinase 4/metabolism , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Signal Transduction/physiology , Tumor Microenvironment/physiology , Animals , Cell Movement/physiology , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mice , Muscle, Skeletal/metabolism , Phosphorylation , RNA, Messenger
11.
Sex Dev ; 13(4): 195-204, 2019.
Article in English | MEDLINE | ID: mdl-32008010

ABSTRACT

MAPKs affect gonadal differentiation in mice and humans, but whether this applies to all mammals is as yet unknown. Thus, we investigated MAPK expression during gonadal differentiation and after treatment with oestrogen in a distantly related mammal, the marsupial tammar wallaby, using our model of oestrogen-induced gonadal sex reversal. High-throughput RNA-sequencing was carried out on gonads collected from developing tammar 2 days before birth to 8 days after birth to characterise MAPK and key sexual differentiation markers. Day 25 foetal testes were cultured for 120 h in control medium or medium supplemented with exogenous oestrogen and processed for RNA-seq to identify changes in gene expression in response to oestrogen. MAPK pathway genes in the tammar were highly conserved at the sequence and amino acid level with those of mice and humans. Marsupial MAP3K1 and MAP3K4 clustered together in a separate branch from eutherian mammals. There was a marked decrease in the expression of male-determining genes SOX9 and AMH and increase in the female marker FOXL2 in oestrogen-treated male gonads. Only MAP3K1 expression increased in male gonads in response to oestrogen while other MAPK genes remained unaffected. This study suggests that MAP3K1 can be influenced by exogenous oestrogens during gonadal differentiation in this marsupial.


Subject(s)
Gene Expression Profiling , Gonads/embryology , Gonads/enzymology , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Kinase Kinase 4/genetics , Macropodidae/embryology , Macropodidae/genetics , Animals , Estrogens/pharmacology , Female , Gene Expression Regulation, Developmental/drug effects , Genetic Markers , Gonads/drug effects , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinase 4/metabolism , Male , Phylogeny , Sex Differentiation/drug effects , Sex Differentiation/genetics , Transcriptome/drug effects , Transcriptome/genetics
12.
Biochem Biophys Res Commun ; 504(4): 771-776, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30217450

ABSTRACT

The age-related reduction in the function of osteoblasts plays a central role in the pathogenesis of bone loss and osteoporosis. Collagen synthesis is a primary function of differentiated osteoblasts, however, the mechanisms for age-related changes in collagen synthesis in human osteoblasts remain elusive. We use Gene Ontology (GO) analysis and Gene Set Enrichment Analysis (GSEA) analysis to exploit the transcriptional profiles of osteoblasts from young and old donors. A panel of collagen members was downregulated in aged osteoblasts, including COL12A1, COL5A1, COL5A3, COL8A1 and COL8A2. Co-expression analysis followed by GO analysis revealed that oxidoreductase activity and kinase activity were inversely correlated with collagen synthesis in osteoblasts. GESA analysis further showed that JNK signaling was upregulated in aged osteoblasts. Consistently, MAP3K4 and MAP4K2, upstream of JNK, were also increased in aged osteoblasts. Moreover, expression levels of MAP3K4 were significantly inversely correlated with levels of the collagen genes. Those transcriptomic results were further verified by examining clinical specimens of osteoporosis by immunohistochemistry. These results provide transcriptomic evidence that deregulated JNK signaling may impair collagen synthesis in osteoblasts and imply a therapeutic value of JNK inhibitors for treating osteoporosis and preventing skeletal aging by counteracting the age-related reduction in the function of osteoblasts.


Subject(s)
Collagen/biosynthesis , Gene Expression Regulation , MAP Kinase Signaling System/physiology , Osteoblasts/metabolism , Osteoporosis/metabolism , Adult , Age Factors , Aged , Collagen/genetics , Collagen Type VIII/genetics , Collagen Type VIII/metabolism , Collagen Type XII/genetics , Collagen Type XII/metabolism , Germinal Center Kinases , Humans , MAP Kinase Kinase Kinase 4/genetics , MAP Kinase Kinase Kinase 4/metabolism , Middle Aged , Osteoblasts/physiology , Osteoporosis/pathology , Oxidoreductases/genetics , Oxidoreductases/metabolism , Protein Serine-Threonine Kinases/metabolism , Sequence Analysis, RNA
13.
Fish Shellfish Immunol ; 66: 372-381, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28476674

ABSTRACT

The mitogen-activated protein kinase (MAPK) cascades stand for one of the most important signaling mechanisms in response to environmental stimuli. In the present study, we cloned and identified for the first time the full-length cDNA of MAPK kinase kinase 4 (TgMEKK4) from Blood clam Tegillarca granosa using rapid amplification of cDNA ends method. The full-length cDNA of TgMEKK4 was of 1605 bp in length, encoding a polypeptide of 364 amino acids with a predicted molecular mass of 41.22 kDa and theoretical isoelectric point of 6.29. The conserved MEKK4-domain was identified in TgMEKK4 by SMART program analysis. Homology analysis of the deduced amino acid sequence of TgMEKK4 with other known sequences revealed that TgMEKK4 shared 58%-80% identity to MEKK4s from other species. TgMEKK4 mRNA transcripts could be detected in all tissues examined with the highest expression level in the gill by qRT-PCR. The mRNA expression of TgMEKK4 was up-regulated significantly in hemocytes after Vibrio parahaemolyticus, Vibrio alginolyticus and Lipopolysaccharide (LPS) challenges. Overexpression of TgMEKK4 in HEK 293T cells resulted in the activation of JNK and ERK, but not p38. Consistently, In vivo study indicated that LPS stimulation enhanced JNK, ERK and p38 phosphorylation in blood clams. These results suggest that TgMEKK4 is a powerful factor in the regulation of genes that may be involved in innate immune response of blood clam.


Subject(s)
Arcidae/genetics , Arcidae/immunology , Immunity, Innate , MAP Kinase Kinase Kinase 4/genetics , Amino Acid Sequence , Animals , Arcidae/microbiology , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , DNA, Complementary/metabolism , Lipopolysaccharides/pharmacology , MAP Kinase Kinase Kinase 4/chemistry , MAP Kinase Kinase Kinase 4/metabolism , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Alignment , Vibrio alginolyticus/physiology , Vibrio parahaemolyticus/physiology
14.
Cell Rep ; 18(10): 2387-2400, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28273454

ABSTRACT

The first epithelial-to-mesenchymal transition (EMT) occurs in trophoblast stem (TS) cells during implantation. Inactivation of the serine/threonine kinase MAP3K4 in TS cells (TSKI4 cells) induces an intermediate state of EMT, where cells retain stemness, lose epithelial markers, and gain mesenchymal characteristics. Investigation of relationships among MAP3K4 activity, stemness, and EMT in TS cells may reveal key regulators of EMT. Here, we show that MAP3K4 activity controls EMT through the ubiquitination and degradation of HDAC6. Loss of MAP3K4 activity in TSKI4 cells results in elevated HDAC6 expression and the deacetylation of cytoplasmic and nuclear targets. In the nucleus, HDAC6 deacetylates the promoters of tight junction genes, promoting the dissolution of tight junctions. Importantly, HDAC6 knockdown in TSKI4 cells restores epithelial features, including cell-cell adhesion and barrier formation. These data define a role for HDAC6 in regulating gene expression during transitions between epithelial and mesenchymal phenotypes.


Subject(s)
Chromatin/metabolism , Epithelial-Mesenchymal Transition , Histone Deacetylase 6/metabolism , Stem Cells/cytology , Trophoblasts/metabolism , Acetylation , Animals , Cell Differentiation , Cell Nucleus/metabolism , Epithelial-Mesenchymal Transition/genetics , MAP Kinase Kinase Kinase 4/metabolism , Mice , Phenotype , Promoter Regions, Genetic/genetics , Protein Binding , Proteolysis , Tight Junction Proteins/metabolism , Ubiquitination
15.
J Biol Chem ; 291(34): 17496-17509, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27358404

ABSTRACT

Skeletal muscle atrophy is a serious and highly prevalent condition that remains poorly understood at the molecular level. Previous work found that skeletal muscle atrophy involves an increase in skeletal muscle Gadd45a expression, which is necessary and sufficient for skeletal muscle fiber atrophy. However, the direct mechanism by which Gadd45a promotes skeletal muscle atrophy was unknown. To address this question, we biochemically isolated skeletal muscle proteins that associate with Gadd45a as it induces atrophy in mouse skeletal muscle fibers in vivo We found that Gadd45a interacts with multiple proteins in skeletal muscle fibers, including, most prominently, MEKK4, a mitogen-activated protein kinase kinase kinase that was not previously known to play a role in skeletal muscle atrophy. Furthermore, we found that, by forming a complex with MEKK4 in skeletal muscle fibers, Gadd45a increases MEKK4 protein kinase activity, which is both sufficient to induce skeletal muscle fiber atrophy and required for Gadd45a-mediated skeletal muscle fiber atrophy. Together, these results identify a direct biochemical mechanism by which Gadd45a induces skeletal muscle atrophy and provide new insight into the way that skeletal muscle atrophy occurs at the molecular level.


Subject(s)
Cell Cycle Proteins/metabolism , MAP Kinase Kinase Kinase 4/metabolism , Multiprotein Complexes/metabolism , Muscle Fibers, Skeletal/metabolism , Muscular Atrophy/metabolism , Nuclear Proteins/metabolism , Animals , Cell Cycle Proteins/genetics , MAP Kinase Kinase Kinase 4/genetics , Mice , Multiprotein Complexes/genetics , Muscle Fibers, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Nuclear Proteins/genetics
16.
J Neurosci ; 36(4): 1347-61, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26818521

ABSTRACT

Mechanosensory hair cells (HCs) residing in the inner ear are critical for hearing and balance. Precise coordination of proliferation, sensory specification, and differentiation during development is essential to ensure the correct patterning of HCs in the cochlear and vestibular epithelium. Recent studies have revealed that FGF20 signaling is vital for proper HC differentiation. However, the mechanisms by which FGF20 signaling promotes HC differentiation remain unknown. Here, we show that mitogen-activated protein 3 kinase 4 (MEKK4) expression is highly regulated during inner ear development and is critical to normal cytoarchitecture and function. Mice homozygous for a kinase-inactive MEKK4 mutation exhibit significant hearing loss. Lack of MEKK4 activity in vivo also leads to a significant reduction in the number of cochlear and vestibular HCs, suggesting that MEKK4 activity is essential for overall development of HCs within the inner ear. Furthermore, we show that loss of FGF20 signaling in vivo inhibits MEKK4 activity, whereas gain of Fgf20 function stimulates MEKK4 expression, suggesting that Fgf20 modulates MEKK4 activity to regulate cellular differentiation. Finally, we demonstrate, for the first time, that MEKK4 acts as a critical node to integrate FGF20-FGFR1 signaling responses to specifically influence HC development and that FGFR1 signaling through activation of MEKK4 is necessary for outer hair cell differentiation. Collectively, this study provides compelling evidence of an essential role for MEKK4 in inner ear morphogenesis and identifies the requirement of MEKK4 expression in regulating the specific response of FGFR1 during HC development and FGF20/FGFR1 signaling activated MEKK4 for normal sensory cell differentiation. SIGNIFICANCE STATEMENT: Sensory hair cells (HCs) are the mechanoreceptors within the inner ear responsible for our sense of hearing. HCs are formed before birth, and mammals lack the ability to restore the sensory deficits associated with their loss. In this study, we show, for the first time, that MEKK4 signaling is essential for the development of normal cytoarchitecture and hearing function as MEKK4 signaling-deficient mice exhibit a significant reduction of HCs and a hearing loss. We also identify MEKK4 as a critical hub kinase for FGF20-FGFR1 signaling to induce HC differentiation in the mammalian cochlea. These results reveal a new paradigm in the regulation of HC differentiation and provide significant new insights into the mechanism of Fgf signaling governing HC formation.


Subject(s)
Ear, Inner , Gene Expression Regulation, Developmental/physiology , MAP Kinase Kinase Kinase 4/metabolism , Sensory Receptor Cells/physiology , Signal Transduction/physiology , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Ear, Inner/cytology , Ear, Inner/enzymology , Ear, Inner/growth & development , Embryo, Mammalian , Evoked Potentials, Auditory, Brain Stem/genetics , Female , Gene Expression Regulation, Developmental/genetics , Hair Cells, Auditory, Inner/physiology , MAP Kinase Kinase Kinase 4/genetics , Male , Mice , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/metabolism , Pregnancy , Repressor Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Signal Transduction/genetics , Spiral Ganglion/cytology , Tubulin/metabolism
18.
Retrovirology ; 12: 102, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26654242

ABSTRACT

BACKGROUND: The HIV-1 infection is characterized by profound CD4(+) T cell destruction and a marked Th17 dysfunction at the mucosal level. Viral suppressive antiretroviral therapy restores Th1 but not Th17 cells. Although several key HIV dependency factors (HDF) were identified in the past years via genome-wide siRNA screens in cell lines, molecular determinants of HIV permissiveness in primary Th17 cells remain to be elucidated. RESULTS: In an effort to orient Th17-targeted reconstitution strategies, we investigated molecular mechanisms of HIV permissiveness in Th17 cells. Genome-wide transcriptional profiling in memory CD4(+) T-cell subsets enriched in cells exhibiting Th17 (CCR4(+)CCR6(+)), Th1 (CXCR3(+)CCR6(-)), Th2 (CCR4(+)CCR6(-)), and Th1Th17 (CXCR3(+)CCR6(+)) features revealed remarkable transcriptional differences between Th17 and Th1 subsets. The HIV-DNA integration was superior in Th17 versus Th1 upon exposure to both wild-type and VSV-G-pseudotyped HIV; this indicates that post-entry mechanisms contribute to viral replication in Th17. Transcripts significantly enriched in Th17 versus Th1 were previously associated with the regulation of TCR signaling (ZAP-70, Lck, and CD96) and Th17 polarization (RORγt, ARNTL, PTPN13, and RUNX1). A meta-analysis using the NCBI HIV Interaction Database revealed a set of Th17-specific HIV dependency factors (HDFs): PARG, PAK2, KLF2, ITGB7, PTEN, ATG16L1, Alix/AIP1/PDCD6IP, LGALS3, JAK1, TRIM8, MALT1, FOXO3, ARNTL/BMAL1, ABCB1/MDR1, TNFSF13B/BAFF, and CDKN1B. Functional studies demonstrated an increased ability of Th17 versus Th1 cells to respond to TCR triggering in terms of NF-κB nuclear translocation/DNA-binding activity and proliferation. Finally, RNA interference studies identified MAP3K4 and PTPN13 as two novel Th17-specific HDFs. CONCLUSIONS: The transcriptional program of Th17 cells includes molecules regulating HIV replication at multiple post-entry steps that may represent potential targets for novel therapies aimed at protecting Th17 cells from infection and subsequent depletion in HIV-infected subjects.


Subject(s)
HIV Infections/virology , HIV-1/physiology , Receptors, Antigen, T-Cell/immunology , Th17 Cells/immunology , Th17 Cells/virology , Virus Replication , Adult , Cells, Cultured , Female , Gene Expression Profiling , Humans , Immunity, Mucosal , Immunologic Memory , MAP Kinase Kinase Kinase 4/genetics , MAP Kinase Kinase Kinase 4/metabolism , Male , NF-kappa B/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 13/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 13/metabolism , RNA Interference , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, CCR4/immunology , Receptors, CCR6/immunology , T-Lymphocyte Subsets/virology , Th1 Cells/immunology , Th1 Cells/virology , Th17 Cells/classification , Transcriptome
19.
Biochem Biophys Res Commun ; 467(4): 792-7, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26498521

ABSTRACT

MAPKKK is the largest family of MAPK cascade, which is known to play important roles in plant growth, development and immune responses. So far, only a few have been functionally characterized even in the model plant, Arabidopsis due to the potential functional redundancy of MAPKKK. We previously identified and cloned a few MAPKKK family genes from rapeseed. In this study, BnaMAPKKK4 was characterized as a member in eliciting accumulation of reactive oxygen species (ROS) and hypersensitive response (HR)-like cell death. This is accompanied with accumulation of malondialdehyde (MDA), anthocyanin as well as nuclear DNA fragmentation. The transcript abundance of a series of ROS accumulation, cell death, and defense response related genes were up-regulated by the expression of MAPKKK4. Further investigation identified BnaMAPKKK4 elicited ROS through the downstream MPK3. These results indicate that BnaMAPKKK4 and its downstream components function in the ROS-induced cell death.


Subject(s)
Brassica napus/metabolism , MAP Kinase Kinase Kinase 4/metabolism , Plant Proteins/metabolism , Reactive Oxygen Species/metabolism , Brassica napus/cytology , Brassica napus/genetics , Cell Death , Gene Expression Regulation, Plant , Hydrogen Peroxide/metabolism , MAP Kinase Kinase Kinase 4/genetics , Phylogeny , Plant Cells/metabolism , Plant Proteins/genetics , Plants, Genetically Modified , Nicotiana/genetics
20.
Stem Cell Reports ; 3(1): 34-43, 2014 Jul 08.
Article in English | MEDLINE | ID: mdl-25068120

ABSTRACT

The balance of self-renewal and differentiation in long-term repopulating hematopoietic stem cells (LT-HSC) must be strictly controlled to maintain blood homeostasis and to prevent leukemogenesis. Hematopoietic cytokines can induce differentiation in LT-HSCs; however, the molecular mechanism orchestrating this delicate balance requires further elucidation. We identified the tumor suppressor GADD45G as an instructor of LT-HSC differentiation under the control of differentiation-promoting cytokine receptor signaling. GADD45G immediately induces and accelerates differentiation in LT-HSCs and overrides the self-renewal program by specifically activating MAP3K4-mediated MAPK p38. Conversely, the absence of GADD45G enhances the self-renewal potential of LT-HSCs. Videomicroscopy-based tracking of single LT-HSCs revealed that, once GADD45G is expressed, the development of LT-HSCs into lineage-committed progeny occurred within 36 hr and uncovered a selective lineage choice with a severe reduction in megakaryocytic-erythroid cells. Here, we report an unrecognized role of GADD45G as a central molecular linker of extrinsic cytokine differentiation and lineage choice control in hematopoiesis.


Subject(s)
Cytokines/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Cell Differentiation/physiology , Flow Cytometry , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Kinase Kinase 4/genetics , MAP Kinase Kinase Kinase 4/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Video , GADD45 Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...