Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Cell Mol Life Sci ; 79(4): 198, 2022 Mar 21.
Article in English | MEDLINE | ID: mdl-35313355

ABSTRACT

The dual specificity protein phosphatases (Dusps) control dephosphorylation of mitogen-activated protein kinases (MAPKs) as well as other substrates. Here, we report that Dusp26, which is highly expressed in neuroblastoma cells and primary neurons is targeted to the mitochondrial outer membrane via its NH2-terminal mitochondrial targeting sequence. Loss of Dusp26 has a significant impact on mitochondrial function that is associated with increased levels of reactive oxygen species (ROS), reduction in ATP generation, reduction in mitochondria motility and release of mitochondrial HtrA2 protease into the cytoplasm. The mitochondrial dysregulation in dusp26-deficient neuroblastoma cells leads to the inhibition of cell proliferation and cell death. In vivo, Dusp26 is highly expressed in neurons in different brain regions, including cortex and midbrain (MB). Ablation of Dusp26 in mouse model leads to dopaminergic (DA) neuronal cell loss in the substantia nigra par compacta (SNpc), inflammatory response in MB and striatum, and phenotypes that are normally associated with Neurodegenerative diseases. Consistent with the data from our mouse model, Dusp26 expressing cells are significantly reduced in the SNpc of Parkinson's Disease patients. The underlying mechanism of DA neuronal death is that loss of Dusp26 in neurons increases mitochondrial ROS and concurrent activation of MAPK/p38 signaling pathway and inflammatory response. Our results suggest that regulation of mitochondrial-associated protein phosphorylation is essential for the maintenance of mitochondrial homeostasis and dysregulation of this process may contribute to the initiation and development of neurodegenerative diseases.


Subject(s)
Dopaminergic Neurons/physiology , Dual-Specificity Phosphatases/physiology , Mitochondria/metabolism , Mitogen-Activated Protein Kinase Phosphatases/physiology , Animals , Cell Death/genetics , Cell Respiration/genetics , Cells, Cultured , Cytoprotection/genetics , HEK293 Cells , Humans , Male , Mice , Mice, 129 Strain , Mice, Knockout , Mitochondria/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Oxidative Stress/genetics , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology
2.
Biosci Biotechnol Biochem ; 85(8): 1839-1845, 2021 Jul 23.
Article in English | MEDLINE | ID: mdl-34143206

ABSTRACT

DUSP4 is considered as an oncogenic gene. However, the effect of DUSP4 on the carcinogenesis of clear cell Renal cell carcinoma (CCRCC) is still unclear. In this study, DUSP4 mRNA levels were significantly increased in CCRCC tissues and cell lines. Furthermore, DUSP4 overexpression promotes the proliferation, migration, and tumorigenicity of CCRCC cells while DUSP4 silencing showed the opposite effects. Importantly, both autophagic activity (LC3 conversion rate and LC3 puncta formation) and total death level promoted by DUSP4 silencing were reversed by treatment with 3-MA in CCRCC cells. Moreover, the proliferation and migration of CCRCC cells inhibited by DUSP4 silencing were also recovered by suppression of autophagy with 3-MA. In conclusion, DUSP4 serves as an oncogenic gene in CCRCC carcinogenesis due to its inhibitory effect on autophagic death, indicating the potential value of DUSP4 in the diagnosis and treatment of CCRCC.


Subject(s)
Autophagy/genetics , Carcinogenesis , Carcinoma, Renal Cell/pathology , Cell Death/genetics , Dual-Specificity Phosphatases/genetics , Kidney Neoplasms/pathology , Mitogen-Activated Protein Kinase Phosphatases/genetics , Aged , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , Cell Survival/genetics , Dual-Specificity Phosphatases/physiology , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Kidney Neoplasms/genetics , Middle Aged , Mitogen-Activated Protein Kinase Phosphatases/physiology
3.
Hepatology ; 69(1): 76-93, 2019 01.
Article in English | MEDLINE | ID: mdl-30063256

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD), ranging from nonalcoholic fatty liver to nonalcoholic steatohepatitis (NASH), is the leading cause of chronic liver diseases. Until now, no medications for NAFLD have been approved by relevant governmental agencies. Dual-specificity phosphatase 9 (Dusp9) is a member of the DUSP protein family. Dusp9 is expressed in insulin-sensitive tissues, and its expression may be modified with the development of insulin resistance (IR). However, the molecular targets and mechanisms of Dusp9 action on NAFLD and NASH remain poorly understood. In this study, using conditional liver-specific Dusp9-knockout (Dusp9-CKO) mice and Dusp9-transgenic mice, we showed that Dusp9 was a key suppressor of high-fat diet-induced hepatic steatosis and inflammatory responses and that Dusp9 deficiency aggravated high-fat high-cholesterol diet-induced liver fibrosis. Dusp9 was shown to exert its effects by blocking apoptosis signal-regulating kinase 1 (ASK1) phosphorylation and the subsequent activation of p38 and c-Jun NH2-terminal kinase signaling. Conclusion: Hepatocyte Dusp9 prevents NAFLD and NASH progression in mice, including lipid accumulation, glucose metabolism disorders, and enhanced inflammation and liver fibrosis, in an ASK1-dependent manner; these findings suggest that Dusp9 may be a promising therapeutic target for the treatment of NAFLD and NASH.


Subject(s)
Dual-Specificity Phosphatases/physiology , MAP Kinase Kinase Kinase 5/antagonists & inhibitors , Non-alcoholic Fatty Liver Disease/enzymology , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
4.
FASEB J ; 33(11): 12941-12959, 2019 11.
Article in English | MEDLINE | ID: mdl-31480854

ABSTRACT

Gliomas are the most common primary brain tumors. Their highly invasive character and the heterogeneity of active oncogenic pathways within single tumors complicate the development of curative therapies and cause poor patient prognosis. Glioma cells express the intermediate filament protein glial fibrillary acidic protein (GFAP), and the level of its alternative splice variant GFAP-δ, relative to its canonical splice variant GFAP-α, is higher in grade IV compared with lower-grade and lower malignant glioma. In this study we show that a high GFAP-δ/α ratio induces the expression of the dual-specificity phosphatase 4 (DUSP4) in focal adhesions. By focusing on pathways up- and downstream of DUSP4 that are involved in the cell-extracellular matrix interaction, we show that a high GFAP-δ/α ratio equips glioma cells to better invade the brain. This study supports the hypothesis that glioma cells with a high GFAP-δ/α ratio are highly invasive and more malignant cells, thus making GFAP alternative splicing a potential therapeutic target.-Van Bodegraven, E. J., van Asperen, J. V., Sluijs, J. A., van Deursen, C. B. J., van Strien, M. E., Stassen, O. M. J. A., Robe, P. A. J., Hol, E. M. GFAP alternative splicing regulates glioma cell-ECM interaction in a DUSP4-dependent manner.


Subject(s)
Alternative Splicing , Brain Neoplasms/pathology , Dual-Specificity Phosphatases/physiology , Extracellular Matrix/pathology , Glial Fibrillary Acidic Protein/genetics , Glioma/pathology , Mitogen-Activated Protein Kinase Phosphatases/physiology , Brain Neoplasms/metabolism , CRISPR-Cas Systems , Cell Line, Tumor , Dual-Specificity Phosphatases/genetics , Extracellular Matrix/metabolism , Gene Knockdown Techniques , Glioma/metabolism , Humans , Laminin/metabolism , MAP Kinase Kinase 4/metabolism , Mitogen-Activated Protein Kinase Phosphatases/genetics , Phosphorylation
5.
FASEB J ; 33(6): 7331-7347, 2019 06.
Article in English | MEDLINE | ID: mdl-30884976

ABSTRACT

Osteoblasts are versatile cells involved in multiple whole-body processes, including bone formation and immune response. Secretory amounts and patterns of osteoblast-derived proteins such as osteopontin (OPN) and osteocalcin (OCN) modulate osteoblast function. However, the regulatory mechanism of OPN and OCN expression remains unknown. Here, we demonstrate that p54/p46 c-jun N-terminal kinase (JNK) inhibition suppresses matrix mineralization and OCN expression but increases OPN expression in MC3T3-E1 cells and primary osteoblasts treated with differentiation inducers, including ascorbic acid, bone morphogenic protein-2, or fibroblast growth factor 2. Preinhibition of JNK before the onset of differentiation increased the number of osteoblasts that highly express OPN but not OCN (OPN-OBs), indicating that JNK affects OPN secretory phenotype at the early stage of osteogenic differentiation. Additionally, we identified JNK2 isoform as being critically involved in OPN-OB differentiation. Microarray analysis revealed that OPN-OBs express characteristic transcription factors, cell surface markers, and cytokines, including glycoprotein hormone α2 and endothelial cell-specific molecule 1. Moreover, we found that inhibitor of DNA binding 4 is an important regulator of OPN-OB differentiation and that dual-specificity phosphatase 16, a JNK-specific phosphatase, functions as an endogenous regulator of OPN-OB induction. OPN-OB phenotype was also observed following LPS from Porphyromonas gingivalis stimulation during osteogenic differentiation. Collectively, these results suggest that the JNK-Id4 signaling axis is crucial in the control of OPN and OCN expression during osteoblastic differentiation.-Kusuyama, J., Amir, M. S., Albertson, B. G., Bandow, K., Ohnishi, T., Nakamura, T., Noguchi, K., Shima, K., Semba, I., Matsuguchi, T. JNK inactivation suppresses osteogenic differentiation, but robustly induces osteopontin expression in osteoblasts through the induction of inhibitor of DNA binding 4 (Id4).


Subject(s)
Inhibitor of Differentiation Proteins/physiology , JNK Mitogen-Activated Protein Kinases/physiology , MAP Kinase Signaling System/physiology , Osteoblasts/metabolism , Osteogenesis/physiology , Osteopontin/biosynthesis , Animals , Cells, Cultured , Dual-Specificity Phosphatases/deficiency , Dual-Specificity Phosphatases/physiology , Gene Expression Regulation, Developmental/drug effects , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 9/antagonists & inhibitors , Mitogen-Activated Protein Kinase 9/physiology , Mitogen-Activated Protein Kinase Phosphatases/deficiency , Mitogen-Activated Protein Kinase Phosphatases/physiology , Osteocalcin/biosynthesis , Osteocalcin/genetics , Osteogenesis/drug effects , Osteopontin/genetics , Protein Isoforms/physiology , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology
6.
Exp Cell Res ; 382(1): 111467, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31202710

ABSTRACT

Hyperglycemia and hyperlipidemia (glycolipotoxicity)-triggered islet ß-cell dysfunction is known to drive the progression of obesity-related type 2 diabetes, however the underlying mechanisms have not been clearly elucidated. The current study aimed to investigate the role of mitogen-activated protein kinase phosphatase 5 (MKP-5) in islet cells under glucolipotoxic conditions. Using gene overexpression and knockdown approaches, we demonstrated that MKP-5 could alleviate glucolipotoxicity-induced apoptosis via the endoplasmic reticulum (ER) stress and mitochondrial apoptosis pathways owing to the altered regulation of caspase family members and ER stress-related molecules in MIN6 and primary islet cells. Overexpression of MKP-5 reversed the glucose and palmitic acid (GP)-induced impairment of insulin secretion as well as the abnormal decreases in the expression of islet functional genes, thereby maintaining the normal insulin secretory functionality, whereas the absence of MKP-5 aggravated islet cell dysfunction. In parallel, the production of ROS and increased inflammation-associated genes in response to GP were also reduced upon MKP-5 overexpression. Further, inhibition of JNK or P38 MAPK pathways resisted to glucolipotoxicity observed in MKP-5 knockdown MIN6 cells. These findings indicate that MKP-5 is an important mediator for glucolipotoxicity-induced islet cell dysfunction and apoptosis, with JNK and P38 as the critical downstream pathways.


Subject(s)
Apoptosis/physiology , Dual-Specificity Phosphatases/physiology , Endoplasmic Reticulum Stress/physiology , Glucose/toxicity , Islets of Langerhans/drug effects , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase Phosphatases/physiology , Palmitates/toxicity , Animals , Cell Line, Tumor , Diet, High-Fat/adverse effects , Dual-Specificity Phosphatases/genetics , Gene Knockdown Techniques , Humans , Insulin/metabolism , Insulinoma/pathology , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitogen-Activated Protein Kinase Phosphatases/genetics , Pancreatic Neoplasms/pathology , Recombinant Proteins/metabolism , Up-Regulation
7.
Mol Cell ; 45(1): 123-31, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22178396

ABSTRACT

Both the DNA damage response (DDR) and epigenetic mechanisms play key roles in the implementation of senescent phenotypes, but very little is known about how these two mechanisms are integrated to establish senescence-associated gene expression. Here we show that, in senescent cells, the DDR induces proteasomal degradation of G9a and GLP, major histone H3K9 mono- and dimethyltransferases, through Cdc14B- and p21(Waf1/Cip1)-dependent activation of APC/C(Cdh1) ubiquitin ligase, thereby causing a global decrease in H3K9 dimethylation, an epigenetic mark for euchromatic gene silencing. Interestingly, induction of IL-6 and IL-8, major players of the senescence-associated secretory phenotype (SASP), correlated with a decline of H3K9 dimethylation around the respective gene promoters and knockdown of Cdh1 abolished IL-6/IL-8 expression in senescent cells, suggesting that the APC/C(Cdh1)-G9a/GLP axis plays crucial roles in aspects of senescent phenotype. These findings establish a role for APC/C(Cdh1) and reveal how the DDR integrates with epigenetic processes to induce senescence-associated gene expression.


Subject(s)
Cellular Senescence , DNA Damage , Histone-Lysine N-Methyltransferase/metabolism , Ubiquitin-Protein Ligase Complexes/physiology , Anaphase-Promoting Complex-Cyclosome , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/physiology , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , Dual-Specificity Phosphatases/genetics , Dual-Specificity Phosphatases/metabolism , Dual-Specificity Phosphatases/physiology , Histocompatibility Antigens/metabolism , Histone Methyltransferases , Histones/metabolism , Humans , Methylation , Signal Transduction
8.
Am J Physiol Lung Cell Mol Physiol ; 317(5): L678-L689, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31483681

ABSTRACT

Mitogen-activated protein kinase (MAPK) phosphatase 5 (MKP-5) is a member of the dual-specificity family of protein tyrosine phosphatases that negatively regulates p38 MAPK and the JNK. MKP-5-deficient mice exhibit improved muscle repair and reduced fibrosis in an animal model of muscular dystrophy. Here, we asked whether the effects of MKP-5 on muscle fibrosis extend to other tissues. Using a bleomycin-induced model of pulmonary fibrosis, we found that MKP-5-deficient mice were protected from the development of lung fibrosis, expressed reduced levels of hydroxyproline and fibrogenic genes, and displayed marked polarization towards an M1-macrophage phenotype. We showed that the profibrogenic effects of the transforming growth factor-ß1 (TGF-ß1) were inhibited in MKP-5-deficient lung fibroblasts. MKP-5-deficient fibroblasts exhibited enhanced p38 MAPK activity, impaired Smad3 phosphorylation, increased Smad7 levels, and decreased expression of fibrogenic genes. Myofibroblast differentiation was attenuated in MKP-5-deficient fibroblasts. Finally, we found that MKP-5 expression was increased in idiopathic pulmonary fibrosis (IPF)-derived lung fibroblasts but not in whole IPF lungs. These data suggest that MKP-5 plays an essential role in promoting lung fibrosis. Our results couple MKP-5 with the TGF-ß1 signaling machinery and imply that MKP-5 inhibition may serve as a therapeutic target for human lung fibrosis.


Subject(s)
Dual-Specificity Phosphatases/metabolism , Dual-Specificity Phosphatases/physiology , Fibroblasts/pathology , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Pulmonary Fibrosis/pathology , Transforming Growth Factor beta1/pharmacology , Animals , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Dual-Specificity Phosphatases/genetics , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase Phosphatases/genetics , Phosphorylation , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Signal Transduction
9.
EMBO J ; 34(2): 218-35, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25398911

ABSTRACT

Mitogen-activated protein kinase (MAPK) activation controls diverse cellular functions including cellular survival, proliferation, and apoptosis. Tuning of MAPK activation is counter-regulated by a family of dual-specificity phosphatases (DUSPs). IL-33 is a recently described cytokine that initiates Th2 immune responses through binding to a heterodimeric IL-33Rα (ST2L)/IL-1α accessory protein (IL-1RAcP) receptor that coordinates activation of ERK and NF-κB pathways. We demonstrate here that DUSP5 is expressed in eosinophils, is upregulated following IL-33 stimulation and regulates IL-33 signaling. Dusp5(-/-) mice have prolonged eosinophil survival and enhanced eosinophil effector functions following infection with the helminth Nippostrongylus brasiliensis. IL-33-activated Dusp5(-/-) eosinophils exhibit increased cellular ERK1/2 activation and BCL-XL expression that results in enhanced eosinophil survival. In addition, Dusp5(-/-) eosinophils demonstrate enhanced IL-33-mediated activation and effector functions. Together, these data support a role for DUSP5 as a novel negative regulator of IL-33-dependent eosinophil function and survival.


Subject(s)
Dual-Specificity Phosphatases/physiology , Eosinophils/immunology , Interleukins/pharmacology , Killer Cells, Natural/immunology , Strongylida Infections/immunology , Animals , Blotting, Western , Cells, Cultured , DNA-Binding Proteins/physiology , Enzyme-Linked Immunosorbent Assay , Eosinophils/cytology , Eosinophils/drug effects , Eosinophils/parasitology , Female , Humans , Interleukin-33 , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Killer Cells, Natural/parasitology , Mice , Mice, Knockout , Nippostrongylus/pathogenicity , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Strongylida Infections/drug therapy , Strongylida Infections/mortality , Strongylida Infections/parasitology
10.
Circ Res ; 119(2): 249-60, 2016 07 08.
Article in English | MEDLINE | ID: mdl-27225478

ABSTRACT

RATIONALE: Mitogen-activated protein kinase (MAPK) signaling regulates the growth response of the adult myocardium in response to increased cardiac workload or pathological insults. The dual-specificity phosphatases (DUSPs) are critical effectors, which dephosphorylate the MAPKs to control the basal tone, amplitude, and duration of MAPK signaling. OBJECTIVE: To examine DUSP8 as a regulator of MAPK signaling in the heart and its impact on ventricular and cardiac myocyte growth dynamics. METHODS AND RESULTS: Dusp8 gene-deleted mice and transgenic mice with inducible expression of DUSP8 in the heart were used here to investigate how this MAPK-phosphatase might regulate intracellular signaling and cardiac growth dynamics in vivo. Dusp8 gene-deleted mice were mildly hypercontractile at baseline with a cardiac phenotype of concentric ventricular remodeling, which protected them from progressing towards heart failure in 2 surgery-induced disease models. Cardiac-specific overexpression of DUSP8 produced spontaneous eccentric remodeling and ventricular dilation with heart failure. At the cellular level, adult cardiac myocytes from Dusp8 gene-deleted mice were thicker and shorter, whereas DUSP8 overexpression promoted cardiac myocyte lengthening with a loss of thickness. Mechanistically, activation of extracellular signal-regulated kinases 1/2 were selectively increased in Dusp8 gene-deleted hearts at baseline and following acute pathological stress stimulation, whereas p38 MAPK and c-Jun N-terminal kinases were mostly unaffected. CONCLUSIONS: These results indicate that DUSP8 controls basal and acute stress-induced extracellular signal-regulated kinases 1/2 signaling in adult cardiac myocytes that then alters the length-width growth dynamics of individual cardiac myocytes, which further alters contractility, ventricular remodeling, and disease susceptibility.


Subject(s)
Dual-Specificity Phosphatases/physiology , MAP Kinase Signaling System/physiology , Myocytes, Cardiac/physiology , Ventricular Remodeling/physiology , Animals , Animals, Newborn , Cells, Cultured , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Rats
11.
J Neurochem ; 140(3): 368-382, 2017 02.
Article in English | MEDLINE | ID: mdl-27891578

ABSTRACT

Schwann cells and oligodendrocytes are the myelinating cells of the peripheral and central nervous system, respectively. Despite having different myelin components and different transcription factors driving their terminal differentiation there are shared molecular mechanisms between the two. Sox10 is one common transcription factor required for several steps in development of myelinating glia. However, other factors are divergent as Schwann cells need the transcription factor early growth response 2/Krox20 and oligodendrocytes require Myrf. Likewise, some signaling pathways, like the Erk1/2 kinases, are necessary in both cell types for proper myelination. Nonetheless, the molecular mechanisms that control this shared signaling pathway in myelinating cells remain only partially characterized. The hypothesis of this study is that signaling pathways that are similarly regulated in both Schwann cells and oligodendrocytes play central roles in coordinating the differentiation of myelinating glia. To address this hypothesis, we have used genome-wide binding data to identify a relatively small set of genes that are similarly regulated by Sox10 in myelinating glia. We chose one such gene encoding Dual specificity phosphatase 15 (Dusp15) for further analysis in Schwann cell signaling. RNA interference and gene deletion by genome editing in cultured RT4 and primary Schwann cells showed Dusp15 is necessary for full activation of Erk1/2 phosphorylation. In addition, we show that Dusp15 represses expression of several myelin genes, including myelin basic protein. The data shown here support a mechanism by which early growth response 2 activates myelin genes, but also induces a negative feedback loop through Dusp15 to limit over-expression of myelin genes.


Subject(s)
Dual-Specificity Phosphatases/physiology , MAP Kinase Signaling System/physiology , Myelin Sheath/enzymology , Schwann Cells/enzymology , Animals , Cell Line , Enzyme Activation/physiology , Female , Male , Mice , Mice, Inbred C57BL , Myelin Sheath/genetics , Rats
12.
J Am Soc Nephrol ; 27(5): 1465-77, 2016 05.
Article in English | MEDLINE | ID: mdl-26407593

ABSTRACT

Integrin-linked kinase (ILK) is an intracellular scaffold protein with critical cell-specific functions in the embryonic and mature mammalian kidney. Previously, we demonstrated a requirement for Ilk during ureteric branching and cell cycle regulation in collecting duct cells in vivo Although in vitro data indicate that ILK controls p38 mitogen-activated protein kinase (p38MAPK) activity, the contribution of ILK-p38MAPK signaling to branching morphogenesis in vivo is not defined. Here, we identified genes that are regulated by Ilk in ureteric cells using a whole-genome expression analysis of whole-kidney mRNA in mice with Ilk deficiency in the ureteric cell lineage. Six genes with expression in ureteric tip cells, including Wnt11, were downregulated, whereas the expression of dual-specificity phosphatase 8 (DUSP8) was upregulated. Phosphorylation of p38MAPK was decreased in kidney tissue with Ilk deficiency, but no significant decrease in the phosphorylation of other intracellular effectors previously shown to control renal morphogenesis was observed. Pharmacologic inhibition of p38MAPK activity in murine inner medullary collecting duct 3 (mIMCD3) cells decreased expression of Wnt11, Krt23, and Slo4c1 DUSP8 overexpression in mIMCD3 cells significantly inhibited p38MAPK activation and the expression of Wnt11 and Slo4c1. Adenovirus-mediated overexpression of DUSP8 in cultured embryonic murine kidneys decreased ureteric branching and p38MAPK activation. Together, these data demonstrate that Ilk controls branching morphogenesis by regulating the expression of DUSP8, which inhibits p38MAPK activity and decreases branching morphogenesis.


Subject(s)
Dual-Specificity Phosphatases/physiology , Kidney/embryology , Kidney/enzymology , Morphogenesis , Protein Serine-Threonine Kinases/physiology , Animals , Mice , p38 Mitogen-Activated Protein Kinases/physiology
13.
Int J Mol Sci ; 18(9)2017 Sep 13.
Article in English | MEDLINE | ID: mdl-28902166

ABSTRACT

Protein homeostasis or proteostasis is a fundamental cellular property that encompasses the dynamic balancing of processes in the proteostasis network (PN). Such processes include protein synthesis, folding, and degradation in both non-stressed and stressful conditions. The role of the PN in neurodegenerative disease is well-documented, where it is known to respond to changes in protein folding states or toxic gain-of-function protein aggregation. Dual-specificity phosphatases have recently emerged as important participants in maintaining balance within the PN, acting through modulation of cellular signaling pathways that are involved in neurodegeneration. In this review, we will summarize recent findings describing the roles of dual-specificity phosphatases in neurodegeneration and offer perspectives on future therapeutic directions.


Subject(s)
Dual-Specificity Phosphatases/physiology , Neurodegenerative Diseases/metabolism , Proteostasis/physiology , Apoptosis , Autophagy , Dual-Specificity Phosphatases/classification , Endoplasmic Reticulum Stress , Heat-Shock Response/physiology , Homeostasis/physiology , Humans , Oxidative Stress/physiology , Protein Aggregates , Protein Biosynthesis , Protein Folding , Protein Kinases/metabolism
14.
Hepatology ; 59(2): 518-30, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23929653

ABSTRACT

UNLABELLED: Cancer/testis (CT) antigens have been considered therapeutic targets for treating cancers. However, a central question is whether their expression contributes to tumorigenesis or if they are functionally irrelevant by-products derived from the process of cellular transformation. In any case, these CT antigens are essential for cancer cell survival and may serve as potential therapeutic targets. Recently, the cell-based RNA interference (RNAi) screen has proven to be a powerful approach for identifying potential therapeutic targets. In this study we sought to identify new CT antigens as potential therapeutic targets for human hepatocellular carcinoma (HCC), and 179 potential CT genes on the X chromosome were screened through a bioinformatics analysis of gene expression profiles. Then an RNAi screen against these potential CT genes identified nine that were required for sustaining the survival of Focus and PLC/PRF/5 cells. Among the nine genes, the physiologically testis-restricted dual specificity phosphatase 21 (DUSP21) encoding a dual specificity phosphatase was up-regulated in 39 (33%) of 118 human HCC specimens. Ectopic DUSP21 had no obvious impact on proliferation and colony formation in HCC cells. However, DUSP21 silencing significantly suppressed cell proliferation, colony formation, and in vivo tumorigenicity in HCC cells. The administration of adenovirus-mediated RNAi and an atelocollagen/siRNA mixture against endogenous DUSP21 significantly suppressed xenograft HCC tumors in mice. Further investigations showed that DUSP21 knockdown led to arrest of the cell cycle in G1 phase, cell senescence, and expression changes of some factors with functions in the cell cycle and/or senescence. Furthermore, the antiproliferative role of DUSP21 knockdown is through activation of p38 mitogen-activated protein kinase in HCC. CONCLUSION: DUSP21 plays an important role in sustaining HCC cell proliferation and may thus act as a potential therapeutic target in HCC treatment.


Subject(s)
Antigens, Neoplasm/genetics , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Dual-Specificity Phosphatases/physiology , Genes, Neoplasm/genetics , Liver Neoplasms/drug therapy , RNA Interference/physiology , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/physiopathology , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Cellular Senescence/physiology , Dual-Specificity Phosphatases/drug effects , Dual-Specificity Phosphatases/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Humans , Liver Neoplasms/pathology , Liver Neoplasms/physiopathology , Mice , Mice, Nude , RNA, Small Interfering/pharmacology , Tumor Cells, Cultured , Up-Regulation/drug effects , Up-Regulation/physiology , Xenograft Model Antitumor Assays
15.
Arterioscler Thromb Vasc Biol ; 34(9): 2023-32, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24990230

ABSTRACT

OBJECTIVE: Reactive oxygen species (ROS) act as signaling molecules during angiogenesis; however, the mechanisms used for such signaling events remain unclear. Stromal cell-derived factor-1α (SDF-1α) is one of the most potent angiogenic chemokines. Here, we examined the role of ROS in the regulation of SDF-1α-dependent angiogenesis. APPROACH AND RESULTS: Bovine aortic endothelial cells were treated with SDF-1α, and intracellular ROS generation was monitored. SDF-1α treatment induced bovine aortic endothelial cell migration and ROS generation, with the majority of ROS generated by bovine aortic endothelial cells at the leading edge of the migratory cells. Antioxidants and nicotinamide adenine dinucleotide phosphate oxidase (NOX) inhibitors blocked SDF-1α-induced endothelial migration. Furthermore, knockdown of either NOX5 or p22phox (a requisite subunit for NOX1/2/4 activation) significantly impaired endothelial motility and tube formation, suggesting that multiple NOXs regulate SDF-1α-dependent angiogenesis. Our previous study demonstrated that c-Jun N-terminal kinase 3 activity is essential for SDF-1α-dependent angiogenesis. Here, we identified that NOX5 is the dominant NOX required for SDF-1α-induced c-Jun N-terminal kinase 3 activation and that NOX5 and MAP kinase phosphatase 7 (MKP7; the c-Jun N-terminal kinase 3 phosphatase) associate with one another but decrease this interaction on SDF-1α treatment. Furthermore, MKP7 activity was inhibited by SDF-1α, and this inhibition was relieved by NOX5 knockdown, indicating that NOX5 promotes c-Jun N-terminal kinase 3 activation by blocking MKP7 activity. CONCLUSIONS: We conclude that NOX is required for SDF-1α signaling and that intracellular redox balance is critical for SDF-1α-induced endothelial migration and angiogenesis.


Subject(s)
Chemokine CXCL12/physiology , Membrane Proteins/physiology , NADPH Oxidases/physiology , Neovascularization, Physiologic/physiology , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Aorta , Azoles/pharmacology , Cattle , Cell Movement/drug effects , Chemokine CXCL12/pharmacology , Dual-Specificity Phosphatases/physiology , Endothelial Cells/drug effects , Endothelium, Vascular/cytology , Gene Knockdown Techniques , Hyperglycemia/metabolism , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/physiology , Isoindoles , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mitogen-Activated Protein Kinase 10/physiology , Mitogen-Activated Protein Kinase Phosphatases/physiology , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , Neovascularization, Physiologic/drug effects , Organoselenium Compounds/pharmacology , Oxidation-Reduction , Signal Transduction/drug effects , Signal Transduction/physiology
16.
J Biol Chem ; 288(2): 819-25, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-23229544

ABSTRACT

The transcription factor NF-κB is critically involved in the inflammatory response triggered by the proinflammatory cytokines TNF and IL-1. Various studies have demonstrated that activation of TAK1 (TGF-ß-activated kinase 1) is an essential step in TNF- and IL-1-induced NF-κB activation pathways. In this study, we identified a member of the dual-specificity phosphatase family, DUSP14, as a negative regulator of TNF- and IL-1-triggered NF-κB activation by expression screens. We found that DUSP14 interacted with TAK1 and that this interaction was enhanced by TNF or IL-1 stimulation. Overexpression of DUSP14 dephosphorylated TAK1 at Thr-187, a residue in the activation loop critically involved in TAK1 activation. Knockdown of DUSP14 increased basal as well as TNF- and IL-1-induced TAK1 phosphorylation at Thr-187. Overexpression of DUSP14, but not its phosphatase-deficient mutant, inhibited TNF- and IL-1-induced as well as TAK1-mediated NF-κB activation, whereas knockdown of DUSP14 had opposite effects. These findings suggest that DUSP14 negatively regulates TNF- or IL-1-induced NF-κB activation by dephosphorylating TAK1 at Thr-187. Our study reveals a new post-translational regulatory mechanism of NF-κB activation triggered by the proinflammatory cytokines.


Subject(s)
Dual-Specificity Phosphatases/physiology , Interleukin-1/physiology , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Phosphatases/physiology , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/physiology , Cell Line , Dual-Specificity Phosphatases/genetics , Gene Knockdown Techniques , Humans , Mitogen-Activated Protein Kinase Phosphatases/genetics , Phosphorylation
17.
J Pharmacol Sci ; 122(2): 93-102, 2013.
Article in English | MEDLINE | ID: mdl-23698110

ABSTRACT

AMP-activated protein kinase (AMPK) contributes to the acceleration of insulin signaling. However, the mechanism by which AMPK regulates insulin signaling remains unclear. Serine phosphorylation of insulin receptor substrate (IRS)-1 negatively regulates insulin signaling. Here we investigated the role of AMPK in serine phosphorylation of IRS-1 at 636/639 and 307, which is induced by tumor necrosis factor (TNF)-α in 3T3L1 adipocytes. We demonstrated that the AMPK activator 5-aminoimidazole-4-carboxamide-1-d-ribofuranoside (AICAR) significantly inhibited the TNF-α-induced serine phosphorylation of IRS-1 at 636/639 and 307 by suppression of extracellular signal-regulated kinase (ERK) phosphorylation but not c-Jun-NH2-terminal kinase (JNK) phosphorylation. In addition, AICAR stimulation resulted in enhanced interaction between ERK and MAP kinase phosphatase-4 (DUSP9/MKP-4) without affecting DUSP9/MPK4 mRNA synthesis. Moreover, intraperitoneal administration (0.25 g/kg) of AICAR to db/db mice improved blood glucose levels and inhibited the phosphorylation of ERK in adipose tissue. In conclusion, we propose a new mechanism in which AICAR suppresses TNF-α-induced serine phosphorylation of IRS-1 at 636/639 and 307 by enhancing the interaction between ERK and DUSP9/MKP-4. Taken together, these findings provide evidence that AMPK plays a crucial role in improving of type 2 diabetes.


Subject(s)
3T3-L1 Cells/metabolism , AMP-Activated Protein Kinases/physiology , Aminoimidazole Carboxamide/analogs & derivatives , Hypoglycemic Agents/pharmacology , Insulin Receptor Substrate Proteins/metabolism , Ribonucleotides/pharmacology , Serine/metabolism , Tumor Necrosis Factor-alpha/physiology , Aminoimidazole Carboxamide/administration & dosage , Aminoimidazole Carboxamide/pharmacology , Animals , Cells, Cultured , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/etiology , Dual-Specificity Phosphatases/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Hypoglycemic Agents/administration & dosage , Injections, Intraperitoneal , Insulin Resistance/physiology , Mice , Mice, Inbred Strains , Mitogen-Activated Protein Kinase Phosphatases/physiology , Phosphorylation/drug effects , Ribonucleotides/administration & dosage , Signal Transduction , Stimulation, Chemical , Tumor Necrosis Factor-alpha/antagonists & inhibitors
18.
Am J Physiol Lung Cell Mol Physiol ; 302(9): L866-74, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22307906

ABSTRACT

Mitogen-activated protein kinases (MAPKs) play a critical role in inflammation. Although activation of MAPK in inflammatory cells has been studied extensively, much less is known about the inactivation of these kinases. MAPK phosphatase 5 (MKP5) is a member of the dual-specificity phosphatase family that dephosphorylates activated MAPKs. Here we report that MKP5 protects sepsis-induced acute lung injury. Mice lacking MKP5 displayed severe lung tissue damage following LPS challenge, characterized with increased neutrophil infiltration and edema compared with wild-type (WT) controls. In response to LPS, MKP5-deficient macrophages produced significantly more inflammatory factors including inflammatory cytokines, nitric oxide, and superoxide. Phosphorylation of p38 MAPK, JNK, and ERK were enhanced in MKP5-deficient macrophages upon LPS stimulation. Adoptive transfer of MKP5-deficient macrophages led to more severe lung inflammation than transfer of WT macrophages, suggesting that MKP5-deficient macrophages directly contribute to acute lung injury. Taken together, these results suggest that MKP5 is crucial to homeostatic regulation of MAPK activation in inflammatory responses.


Subject(s)
Acute Lung Injury/enzymology , Dual-Specificity Phosphatases/physiology , Sepsis/complications , Acute Lung Injury/etiology , Acute Lung Injury/immunology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cells, Cultured , Cytokines/biosynthesis , Dual-Specificity Phosphatases/deficiency , Dual-Specificity Phosphatases/genetics , Escherichia coli , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System , Macrophages/immunology , Macrophages/metabolism , Macrophages/physiology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Nitric Oxide/biosynthesis , Phagocytosis , Phosphorylation , Protein Processing, Post-Translational , Sepsis/immunology , Superoxides/metabolism
19.
Biochim Biophys Acta ; 1803(9): 1003-12, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20347885

ABSTRACT

Dual specificity phosphatases are characterised by their ability to dephosphorylate both phosphotyrosine and phosphoserine/threonine residues within the one substrate. The aim of this study was to characterise the phosphatase activity of the atypical dual specificity phosphatase, DUSP26 on MAP kinases, and to determine its expression, regulation and function in cancer cells. Overexpression and knockdown of DUSP26 in epithelial cells and in vitro phosphatase assays were used to demonstrate that, contrary to several published reports, DUSP26 does not act as a dual specificity phosphatase on ERK, JNK or p38 MAPKs. However, overexpression of DUSP26 in MCF10A epithelial cells suppressed colony formation and acinar growth in 3D culture, effects dependent on its phosphatase activity, while knockdown of DUSP26 in HOSE17.1 cells enhanced colony formation and cellular proliferation. DUSP26 mRNA expression was reduced in neuroblastoma, brain and ovarian cancer cell lines. Consistent with epigenetic silencing of DUSP26, expression was enhanced by treatment of cells with 5-aza-2-deoxycitidine and trichostatin A, and a CpG island upstream of the DUSP26 transcriptional start site was variably methylated in cancer cell lines. Together, these results help to clarify confusion in the literature relating to DUSP26 substrate specificity and support recent reports that substrates other than MAPKs are the primary substrates of this phosphatase. In addition, they indicate that DUSP26 may function as a tumour suppressor in particular cancers.


Subject(s)
Cell Proliferation , Dual-Specificity Phosphatases/physiology , Epithelial Cells/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Mitogen-Activated Protein Kinase Phosphatases/physiology , Animals , CHO Cells , COS Cells , Cells, Cultured , Chlorocebus aethiops , CpG Islands/genetics , Cricetinae , Cricetulus , Dual-Specificity Phosphatases/antagonists & inhibitors , Dual-Specificity Phosphatases/genetics , Dual-Specificity Phosphatases/metabolism , Epithelial Cells/metabolism , Gene Knockdown Techniques , Genes, Tumor Suppressor/physiology , Humans , Mitogen-Activated Protein Kinase Phosphatases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Phosphatases/genetics , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Phosphorylation/physiology
20.
Mol Cell Biochem ; 352(1-2): 155-62, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21360282

ABSTRACT

The dual-specificity phosphatase (DUSP) 13 gene encodes two atypical DUSPs, DUSP13B/TMDP and DUSP13A/MDSP using alternative exons. DUSP13B protein is most highly expressed in testis, particularly in spermatocytes and round spermatids of the seminiferous tubules, while that of DUSP13A is restricted to skeletal muscle. Here, we show that DUSP13B inactivated MAPK activation in the order of selectivity, JNK = p38>ERK in cells, while DUSP13A did not show MAPK phosphatase activity. Reporter gene analysis showed that DUSP13B had significant inhibitory effect on AP-1-dependent gene expression, but DUSP13A did not. To our knowledge, DUSP13B is the first identified testis-specific phosphatase that inhibits stress-activated MAPKs. These data suggest an important role for DUSP13B in protection from external stress during spermatogenesis.


Subject(s)
Dual-Specificity Phosphatases/physiology , Gene Expression Regulation/physiology , Mitogen-Activated Protein Kinases/metabolism , Transcription Factor AP-1/physiology , Animals , Cell Line , Enzyme Activation , Exons , Humans , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL