Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Mol Oncol ; 15(5): 1566-1583, 2021 05.
Article in English | MEDLINE | ID: mdl-33660414

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is one of the most refractory malignancies worldwide. Mitogen-activated protein kinase 3 (MAP2K3) has a contradictory role in tumor progression, and the function and expression patterns of MAP2K3 in ESCC remain to be determined. We found that MAP2K3 expression to be downregulated in ESCC, and MAP2K3 downregulation correlated with clinically poor survival. MAP2K3 inhibited ESCC cell proliferation and invasion in vitro and in vivo. MAP2K3 suppressed STAT3 expression and activation. Mechanistically, MAPSK3 interacted with MDM2 to promote STAT3 degradation via the ubiquitin-proteasome pathway. Furthermore, exosomal miR-19b-3p derived from the plasma of patients with ESCC could suppress MAP2K3 expression to promote ESCC tumorigenesis. STAT3 was found to bind to the MIR19B promoter and increased the expression of miR-19b-3p in ESCC cells. In summary, our results demonstrated that the miR-19b-3p-MAP2K3-STAT3 feedback loop regulates ESCC tumorigenesis and elucidates the potential of therapeutically targeting this pathway in ESCC.


Subject(s)
Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , MAP Kinase Kinase 3/physiology , MicroRNAs/physiology , STAT3 Transcription Factor/physiology , Adult , Aged , Animals , Case-Control Studies , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/genetics , Feedback, Physiological/physiology , Female , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Kinase 3/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Middle Aged , Neoplasm Invasiveness , STAT3 Transcription Factor/genetics
2.
Blood ; 136(6): 749-754, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32548640

ABSTRACT

Several studies demonstrate that hemolysis and free heme in circulation cause endothelial barrier dysfunction and are associated with severe pathological conditions such as acute respiratory distress syndrome, acute chest syndrome, and sepsis. However, the precise molecular mechanisms involved in the pathology of heme-induced barrier disruption remain to be elucidated. In this study, we investigated the role of free heme in the endothelial barrier integrity and mechanisms of heme-mediated intracellular signaling of human lung microvascular endothelial cells (HLMVECs). Heme, in a dose-dependent manner, induced a rapid drop in the endothelial barrier integrity of HLMVECs. An investigation into barrier proteins revealed that heme primarily affected the tight junction proteins zona occludens-1, claudin-1, and claudin-5, which were significantly reduced after heme exposure. The p38MAPK/HSP27 pathway, involved in the regulation of endothelial cytoskeleton remodeling, was also significantly altered after heme treatment, both in HLMVECs and mice. By using a knockout (KO) mouse for MKK3, a key regulator of the p38MAPK pathway, we showed that this KO effectively decreased heme-induced endothelial barrier dysfunction. Taken together, our results indicate that targeting the p38MAPK pathway may represent a crucial treatment strategy in alleviating hemolytic diseases.


Subject(s)
Capillary Permeability/drug effects , Endothelial Cells/drug effects , Heme/pharmacology , MAP Kinase Kinase 3/physiology , MAP Kinase Signaling System/drug effects , Animals , Antigens, CD/analysis , Cadherins/analysis , Capillary Permeability/physiology , Cells, Cultured , Claudins/analysis , Endothelial Cells/physiology , HSP27 Heat-Shock Proteins/physiology , Heat-Shock Proteins/physiology , Hemolysis , Humans , Lung/blood supply , MAP Kinase Kinase 3/deficiency , MAP Kinase Signaling System/physiology , Mice , Mice, Knockout , Microvessels/cytology , Molecular Chaperones/physiology , Tight Junctions/drug effects , Zonula Occludens-1 Protein/analysis , p38 Mitogen-Activated Protein Kinases
3.
Biol Reprod ; 94(5): 103, 2016 05.
Article in English | MEDLINE | ID: mdl-27009039

ABSTRACT

Testis determination in mammals is initiated by expression of SRY in somatic cells of the embryonic gonad. Genetic analyses in the mouse have revealed a requirement for mitogen-activated protein kinase (MAPK) signaling in testis determination: targeted loss of the kinases MAP3K4 and p38 MAPK causes complete XY embryonic gonadal sex reversal. These kinases occupy positions at the top and bottom level, respectively, in the canonical three-tier MAPK-signaling cascade: MAP3K, MAP2K, MAPK. To date, no role in sex determination has been attributed to a MAP2K, although such a function is predicted to exist. Here, we report roles for the kinases MAP2K3 and MAP2K6 in testis determination. C57BL/6J (B6) embryos lacking MAP2K3 exhibited no significant abnormalities of testis development, whilst those lacking MAP2K6 exhibited a minor delay in testis determination. Compound mutants lacking three out of four functional alleles at the two loci also exhibited delayed testis determination and transient ovotestis formation as a consequence, suggestive of partially redundant roles for these kinases in testis determination. Early lethality of double-knockout embryos precludes analysis of sexual development. To reveal their roles in testis determination more clearly, we generated Map2k mutant B6 embryos using a weaker Sry allele (Sry(AKR)). Loss of Map2k3 on this highly sensitized background exacerbates ovotestis development, whilst loss of Map2k6 results in complete XY gonadal sex reversal associated with reduction of Sry expression at 11.25 days postcoitum. Our data suggest that MAP2K6 functions in mouse testis determination, via positive effects on Sry, and also indicate a minor role for MAP2K3.


Subject(s)
MAP Kinase Kinase 3/physiology , MAP Kinase Kinase 6/physiology , Sex Determination Processes/genetics , Sex-Determining Region Y Protein/metabolism , Testis/embryology , Animals , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , MAP Kinase Kinase 3/genetics , MAP Kinase Kinase 6/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Sex Differentiation/genetics , Sex-Determining Region Y Protein/genetics , Testis/metabolism
4.
Free Radic Biol Med ; 83: 139-48, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25697779

ABSTRACT

Mitochondria are increasingly recognized as drivers of inflammatory responses. MAP kinase kinase 3 (MKK3), a dual-specificity protein kinase, is activated in inflammation and in turn activates p38 MAP kinase signaling. Here we show that MKK3 influences mitochondrial function and acts as a critical mediator of inflammation. MKK3-deficient (MKK3(-/-)) mice and bone marrow-derived macrophages (BMDMs) secreted smaller amounts of cytokines than wild type (WT) after lipopolysaccharide (LPS) exposure. There was improved mitochondrial function, as measured by basal oxygen consumption rate, mitochondrial membrane potential, and ATP production, in MKK3(-/-) BMDMs. After LPS exposure, MKK3(-/-) BMDMs did not show a significant increase in cellular reactive oxygen species production or in mitochondrial superoxide compared to WT. Activation of two important inflammatory mediators, i.e., the nuclear translocation of NF-κB and caspase-1 activity (a key component of the inflammasome), was lower in MKK3(-/-) BMDMs. p38 and JNK activation was lower in MKK3(-/-) BMDMs compared to WT after exposure to LPS. Knockdown of MKK3 by siRNA in wild-type BMDMs improved mitochondrial membrane potential, reduced LPS-induced caspase-1 activation, and attenuated cytokine secretion. Our studies establish MKK3 as a regulator of mitochondrial function and inflammatory responses to LPS and suggest that MKK3 may be a therapeutic target in inflammatory disorders such as sepsis.


Subject(s)
Bone Marrow/immunology , Inflammation/immunology , MAP Kinase Kinase 3/physiology , Macrophages/immunology , Mitochondria/immunology , Animals , Blotting, Western , Bone Marrow/drug effects , Bone Marrow/metabolism , Cells, Cultured , Cytokines/metabolism , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/pharmacology , MAP Kinase Kinase 3/antagonists & inhibitors , Macrophages/drug effects , Macrophages/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Phosphorylation/drug effects , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
5.
Plant Cell ; 26(8): 3343-57, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25139007

ABSTRACT

Mitogen-activated protein kinase (MAPK) pathways are involved in several signal transduction processes in eukaryotes. Light signal transduction pathways have been extensively studied in plants; however, the connection between MAPK and light signaling pathways is currently unknown. Here, we show that MKK3-MPK6 is activated by blue light in a MYC2-dependent manner. MPK6 physically interacts with and phosphorylates a basic helix-loop-helix transcription factor, MYC2, and is phosphorylated by a MAPK kinase, MKK3. Furthermore, MYC2 binds to the MPK6 promoter and regulates its expression in a feedback regulatory mechanism in blue light signaling. We present mutational and physiological studies that illustrate the function of the MKK3-MPK6-MYC2 module in Arabidopsis thaliana seedling development and provide a revised mechanistic view of photomorphogenesis.


Subject(s)
Arabidopsis Proteins/physiology , Arabidopsis/growth & development , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Light , MAP Kinase Kinase 3/physiology , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/physiology , Arabidopsis/genetics , Arabidopsis/radiation effects , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Gene Expression Regulation, Plant , MAP Kinase Kinase 3/genetics , MAP Kinase Kinase 3/metabolism , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Seedlings/genetics , Seedlings/growth & development , Seedlings/radiation effects
6.
Am J Physiol Lung Cell Mol Physiol ; 306(7): L604-19, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24487387

ABSTRACT

Sepsis is a systemic inflammatory response to infection and a major cause of death worldwide. Because specific therapies to treat sepsis are limited, and underlying pathogenesis is unclear, current medical care remains purely supportive. Therefore targeted therapies to treat sepsis need to be developed. Although an important mediator of sepsis is thought to be mitochondrial dysfunction, the underlying molecular mechanism is unclear. Modulation of mitochondrial processes may be an effective therapeutic strategy in sepsis. Here, we investigated the role of the kinase MKK3 in regulation of mitochondrial function in sepsis. Using clinically relevant animal models, we examined mitochondrial function in primary mouse lung endothelial cells exposed to LPS. MKK3 deficiency reduces lethality of sepsis in mice and by lowering levels of lung and mitochondrial injury as well as reactive oxygen species. Furthermore, MKK3 deficiency appeared to simultaneously increase mitochondrial biogenesis and mitophagy through the actions of Sirt1, Pink1, and Parkin. This led to a more robust mitochondrial network, which we propose provides protection against sepsis. We also detected higher MKK3 activation in isolated peripheral blood mononuclear cells from septic patients compared with nonseptic controls. Our findings demonstrate a critical role for mitochondria in the pathogenesis of sepsis that involves a previously unrecognized function of MKK3 in mitochondrial quality control. This mitochondrial pathway may help reveal new diagnostic markers and therapeutic targets against sepsis.


Subject(s)
Lung Injury/etiology , MAP Kinase Kinase 3/blood , MAP Kinase Kinase 3/deficiency , Mitochondria/physiology , Mitophagy , Sepsis/physiopathology , Aged , Aged, 80 and over , Animals , Endothelial Cells/metabolism , Female , Humans , Lipopolysaccharides , Lung/metabolism , MAP Kinase Kinase 3/physiology , Male , Mice , Middle Aged , Mitochondria/drug effects , Mitophagy/drug effects , Protein Kinases/metabolism , Sepsis/complications , Sirtuin 1/antagonists & inhibitors , Ubiquitin-Protein Ligases/metabolism
7.
J Leukoc Biol ; 95(6): 903-15, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24453276

ABSTRACT

Mast cells are critical immune effectors abundant in tissues interfacing with the environment and have major roles in allergen-induced inflammation and host responses to infection. SCF is a regulator of mast cell function and growth. However, the critical mechanisms in SCF-directed events remain incompletely defined. Here, we have investigated the role of MKK3 in mast cell SCF signaling-dependent functions by using BMMCs from MKK3-deficient mice. MKK3 was phosphorylated rapidly and persistently following SCF-induced activation and contributed to mast cell proliferation but not survival or migration in response to SCF. Analysis of SCF-induced mast cell mediator secretion demonstrated that IL-6 production is specifically dependent on MKK3 signals, both independently and in concert with IgE. Analysis of SCF-induced signaling showed that sustained p38 phosphorylation was impaired in MKK3-deficient mast cells, where as early JNK and IκBα activation were enhanced. Notably, SCF-inducible expression and activation of c-Jun, a component of the AP-1 transcription factor, was significantly dependent on MKK3. Accordingly, AP-1 DNA-binding activity and interaction with the IL6 gene promoter was markedly impaired in MKK3-deficient mast cells, whereas transcription factors of the Egr family, NF-κB, and NFAT retained near-full activity. These results designate MKK3 as a novel, positive regulator of SCF-induced mast cell proliferation and a critical signaling protein for AP-1-dependent IL-6 production.


Subject(s)
Interleukin-6/biosynthesis , MAP Kinase Kinase 3/physiology , Mast Cells/immunology , Stem Cell Factor/pharmacology , Transcription Factor AP-1/physiology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/biosynthesis , I-kappa B Proteins/metabolism , Immunoglobulin E/physiology , JNK Mitogen-Activated Protein Kinases/metabolism , Mast Cells/drug effects , Mice , Phosphorylation , p38 Mitogen-Activated Protein Kinases/metabolism
8.
J Immunol ; 190(3): 1264-75, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23275604

ABSTRACT

Sepsis is a leading cause of intensive care unit admissions, with high mortality and morbidity. Although outcomes have improved with better supportive care, specific therapies are limited. Endothelial activation and oxidant injury are key events in the pathogenesis of sepsis-induced lung injury. The signaling pathways leading to these events remain poorly defined. We sought to determine the role of MAPK kinase 3 (MKK3), a kinase of the p38 group, in the pathogenesis of sepsis. We used a murine i.p. LPS model of systemic inflammation to mimic sepsis. Lung injury parameters were assessed in lung tissue and bronchoalveolar lavage specimens. Primary lung endothelial cells were cultured and assessed for mediators of inflammation and injury, such as ICAM-1, AP-1, NF-κB, and mitochondrial reactive oxygen species. Our studies demonstrate that MKK3 deficiency confers virtually complete protection against organ injury after i.p. LPS. Specifically, MKK3(-/-) mice were protected against acute lung injury, as assessed by reduced inflammation, mitochondrial reactive oxygen species generation, endothelial injury, and ICAM-1 expression after LPS administration. Our results show that endothelial MKK3 is required for inflammatory cell recruitment to the lungs, mitochondrial oxidant-mediated AP-1, NF-κB activation, and ICAM-1 expression during LPS challenge. Collectively, these studies identify a novel role for MKK3 in lethal LPS responses and provide new therapeutic targets against sepsis and acute lung injury.


Subject(s)
Acute Lung Injury/enzymology , Endothelial Cells/enzymology , Endotoxemia/enzymology , MAP Kinase Kinase 3/physiology , Acute Lung Injury/pathology , Acute Lung Injury/prevention & control , Animals , Apoptosis , Bronchoalveolar Lavage Fluid , Chemotaxis, Leukocyte/physiology , Disease Models, Animal , Endothelial Cells/pathology , Endotoxemia/pathology , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Lipopolysaccharides/toxicity , Lung/pathology , MAP Kinase Kinase 3/antagonists & inhibitors , MAP Kinase Kinase 3/deficiency , MAP Kinase Kinase 3/genetics , Mice , Mice, Knockout , Mitochondria/metabolism , NF-kappa B/metabolism , Neutrophil Activation , Peritonitis/chemically induced , Peritonitis/enzymology , RNA, Small Interfering/pharmacology , Radiation Chimera , Reactive Oxygen Species/metabolism , Sepsis/enzymology , Transcription Factor AP-1/metabolism
9.
Brain Res ; 1422: 1-12, 2011 Nov 08.
Article in English | MEDLINE | ID: mdl-21981804

ABSTRACT

Stimulation of rat microglia with lipopolysaccharide (LPS) in vitro induces production of the inflammatory/cytotoxic cytokine tumor necrosis factor alpha (TNFα) along with superoxide anion (O(2)(-)) and nitric oxide (NO). In this study, we investigated the role of O(2)(-) and NO in the induction of TNFα in microglia. The LPS-inducible TNFα was significantly suppressed by pretreatment with the O(2)(-) scavenger N-acetyl cysteine (NAC), but not by the NO scavenger 2-(4-Carboxyphenyl)-4,4,5,5-tetramethyl imidazoline-1-oxyl 3-oxide, suggesting the close association of O(2)(-) with TNFα induction. NAC strongly depressed phosphorylation of p38 mitogen-activated protein kinase (p38 MAPK), which is necessary for inducing TNFα in microglia. On the other hand, an O(2)(-) donor, 3-(4-Morpholinyl)sydnonimine (SIN-1), induced TNFα in microglia, and the effects of SIN-1 were completely abolished in the presence of superoxide dismutase. There is little likelihood that the NO produced in SIN-1 degradation induces TNFα in microglia, because TNFα was not induced in microglia exposed to the NO-donor S-nitroso-N-acetyl-dl-penicillamine. Moreover, the addition of SIN-1 to microglia resulted in activation of p38 MAPK and its upstream kinase MKK3/6. Taken together, these results showed that O(2)(-) is an important signaling molecule for activating the MKK3/6-p38 cascade, which is requisite for inducing TNFα in microglia.


Subject(s)
MAP Kinase Kinase 3/metabolism , MAP Kinase Signaling System/physiology , Microglia/enzymology , Superoxides/pharmacology , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Animals, Newborn , Female , MAP Kinase Kinase 3/physiology , MAP Kinase Signaling System/drug effects , Microglia/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Pregnancy , Primary Cell Culture , Rats , Rats, Wistar
10.
J Immunol ; 186(7): 4140-6, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21368234

ABSTRACT

NKT cells are known to rapidly produce a large amount of cytokines upon activation. Although a number of signaling pathways that regulate the development of NKT cells have been identified, the signaling pathways involved in the regulation of NKT cell cytokine production remain unclear. In this study, we show that the p38 MAPK pathway is dispensable for the development of NKT cells. However, NKT cell cytokine production and NKT-mediated liver damage are highly dependent on activation of this pathway. p38 MAPK does not substantially affect cytokine gene expression in NKT cells, but it regulates the synthesis of cytokines through the Mnk-eIF4E pathway. Thus, in addition to gene expression, translational regulation by p38 MAPK could be a novel mechanism that contributes to the overall production of cytokine by NKT cells.


Subject(s)
Cytokines/biosynthesis , Cytokines/genetics , MAP Kinase Signaling System/immunology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Protein Modification, Translational/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Enzyme Activation/genetics , Enzyme Activation/immunology , Liver Diseases/enzymology , Liver Diseases/genetics , Liver Diseases/immunology , MAP Kinase Kinase 3/deficiency , MAP Kinase Kinase 3/genetics , MAP Kinase Kinase 3/physiology , MAP Kinase Kinase 6/deficiency , MAP Kinase Kinase 6/genetics , MAP Kinase Kinase 6/physiology , MAP Kinase Signaling System/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Natural Killer T-Cells/enzymology
11.
Aging (Albany NY) ; 2(9): 597-611, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20844314

ABSTRACT

Reactive oxygen species (ROS) and elevated levels of p38 MAPK activity accelerate physiological aging. This emphasizes the importance of understanding the molecular mechanism(s) that link ROS production to activation of the p38 mediated promotion of aging, longevity, and resistance to oxidative stress. We examined Klotho(-/-) (elevated ROS) and Klotho overexpressing mice (low ROS and resistance to ROS) to determine whether the ROS-sensitive apoptosis signal-regulating kinase (ASK1)-signalosome -> p38 MAPK pathway plays a role in the accelerated aging of Klotho(-/-), and resistance to oxidative stress and extended lifespan in the Klotho overexpressing models. Our results suggest that increased endogenous ROS generated by Klotho(-/-) and resistance to oxidative stress in Klotho overexpression are linked to the regulation of ASK1-signalosome -> p38 activity. We propose that (a) the ASK1-signalosome -> p38 MAPK pathway is activated by oxidative stress due to ablation of the Klotho gene; (b) increased longevity by Klotho overexpression is linked to suppression of the ASK1-signalosome-p38 MAPK activity; (c) the ROS-responsive ASK1-signalosome regulates physiological aging via its regulation of p38 MAPK, through a mechanism that balances the levels of inhibitory vs. activating ASK1-signalosomes. We conclude that the Klotho suppressor-of-aging activity is linked to the ASK1-signalsome, a physiological ROS-sensitive signaling center.


Subject(s)
Aging/physiology , Glucuronidase/physiology , MAP Kinase Kinase Kinase 5/physiology , Oxidative Stress/physiology , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/physiology , 14-3-3 Proteins/physiology , Animals , Glucuronidase/genetics , Klotho Proteins , Longevity/physiology , MAP Kinase Kinase 3/physiology , MAP Kinase Kinase 6/physiology , Mice , Mice, Knockout , Models, Animal , NF-E2-Related Factor 2/physiology , Reactive Oxygen Species/metabolism , Thioredoxins/physiology
12.
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
13.
Neuroscience ; 162(2): 462-71, 2009 Aug 18.
Article in English | MEDLINE | ID: mdl-19427893

ABSTRACT

Spinal p38 mitogen activated (MAP) kinase plays a key role in chronic pain behavior. However, clinical development of p38 inhibitors has been hindered by significant toxicity. To evaluate alternative strategies of p38 regulation, we determined if known upstream activators of p38 (mitogen activated kinase kinase [MKK] 3 and MKK6), are involved in development and maintenance of pain and spinal p38 phosphorylation. Acute pain behaviors were not altered in MKK3 or MKK6 deficient mice. The phase 2 formalin response was delayed in MKK3-/- mice, but unchanged in magnitude, while the response remained normal in MKK6-/- mice. More striking, late formalin allodynia (3-18 days post-injection) was prominent in wild type and MKK6-/- mice, but was delayed for several days in MKK3-/- mice. In wild type, but not MKK3-/- mice, intraplantar formalin elicited increases in ipsilateral spinal MKK3/6 phosphorylation acutely and again at 9 days postinjection. Phosphorylation of MKK3/6 correlated with phase 2 formalin behavior. Wild type (WT) and MKK3-/- mice both expressed increases in spinal phosphorylated p38, however in WT mice this response began several days earlier, and was of higher magnitude and duration than in MKK3-/- mice. This phosphorylation correlated with the late allodynia. Phosphorylated MKK3/6 was detected only in astrocytes, given that phosphorylated p38 (P-p38) is usually not seen in astrocytes this argues for astrocytic release of soluble mediators that affect p38 phosphorylation in microglia. Taking these data together, MKK3, but not MKK6, is necessary for normal development of chronic pain behavior and phosphorylation of spinal p38.


Subject(s)
MAP Kinase Kinase 3/physiology , Pain/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Acute Disease , Animals , Astrocytes/enzymology , Chronic Disease , Enzyme Activation , Formaldehyde , MAP Kinase Kinase 3/genetics , MAP Kinase Kinase 6/genetics , MAP Kinase Kinase 6/physiology , Mice , Mice, Knockout , Pain/physiopathology , Pain Measurement , Phosphorylation , Physical Stimulation , Spinal Cord/metabolism
14.
J Endocrinol ; 198(1): 193-207, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18417530

ABSTRACT

Previous studies from this laboratory identified excessive oxidative stress as an important mediator of age-related decline in steroid hormone production. Here, we investigated whether oxidative stress exerts its antisteroidogenic action through modulation of oxidant-sensitive mitogen-activated protein kinase (MAPK) signaling pathways. To accomplish these studies, we employed a highly responsive mouse adrenocortical cell line, Y1-BS1 cells that secrete large quantities of steroids when stimulated with lipoprotein plus hormone. Treatment of these cells with superoxide, H(2)O(2) or 4-hydroxy-2-nonenal (HNE) significantly inhibited steroid production and increased phosphorylation and activation of p38 MAPK. None of the treatments altered the phosphorylation of either extracellular signal-regulated kinases or c-Jun N-terminal kinases (JNKs). Pretreatment of Y1-BS1 cells with MnTMPyP, a cell-permeable superoxide-dismutase/catalase mimetic reactive oxygen species (ROS scavenger), completely prevented the superoxide- and H(2)O(2)-mediated inhibition of steroid production. Likewise, antioxidant N-acetylcysteine completely blocked the HNE-induced loss of steroidogenic response. Incubation of Y1-BS1 cells with either MnTMPyP or NAC also upregulated Bt(2)cAMP and Bt(2)cAMP+hHDL(3)-stimulated steroid synthesis, indicating that endogenously produced ROS can inhibit steroidogenesis. Inhibition of p38 MAPK with SB203580 or SB202190 upregulated the basal steroid production and also prevented the oxidant-mediated inhibition of steroid production. mRNA measurements by qPCR indicated that Y1-BS1 adrenal cells predominantly express p38 MAPKalpha isoform, along with relatively low-level expression of p38 MAPKgamma. By contrast, little or no expression was detected for p38 MAPKbeta and p38 MAPKdelta isoforms in these cells. Transfection of Y1-BS1 cells with either caMKK3 or caMMK6 construct, the upstream p38 MAPK activators, decreased steroidogenesis, whereas transfection with dnMKK3 or dnMKK6 plasmid DNA increased steroidogenesis. Similarly, transfection of cells with a dnp38 MAPKalpha or dnp38 MAPKbeta construct also increased steroid hormone production; however, the effect was less pronounced after expression of either dnp38 MAPKgamma or dnp38 MAPKdelta construct. These results indicate that activated p38 MAPK mediates oxidant (excessive oxidative stress)-induced inhibition of adrenal steroidogenesis.


Subject(s)
20-alpha-Dihydroprogesterone/biosynthesis , Adrenal Glands/metabolism , MAP Kinase Signaling System , Oxidative Stress , p38 Mitogen-Activated Protein Kinases/physiology , Adrenocorticotropic Hormone/pharmacology , Animals , Cell Line, Tumor , Cyclic CMP/analogs & derivatives , Cyclic CMP/pharmacology , MAP Kinase Kinase 3/physiology , MAP Kinase Kinase 6/physiology , Mice , Phosphoproteins/genetics , Phosphorylation , Superoxides/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
15.
Mol Cell ; 27(3): 498-508, 2007 Aug 03.
Article in English | MEDLINE | ID: mdl-17679097

ABSTRACT

Saturated free fatty acid (FFA) is a major source of metabolic stress that activates the c-Jun NH(2)-terminal kinase (JNK). This FFA-stimulated JNK pathway is relevant to hallmarks of metabolic syndrome, including insulin resistance. Here we used gene ablation studies in mice to demonstrate a central role for mixed-lineage protein kinases (MLK) in this signaling pathway. Saturated FFA causes protein kinase C (PKC)-dependent activation of MLK3 that subsequently causes increased JNK activity by a mechanism that requires the MAP kinase kinases MKK4 and MKK7. Loss of PKC, MLK3, MKK4, or MKK7 expression prevents FFA-stimulated JNK activation. Together, these data establish a signaling pathway that mediates effects of metabolic stress on insulin resistance.


Subject(s)
Fatty Acids, Nonesterified/pharmacology , MAP Kinase Kinase Kinases/physiology , Signal Transduction , Animals , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Insulin Receptor Substrate Proteins , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase 3/genetics , MAP Kinase Kinase 3/physiology , MAP Kinase Kinase 7/genetics , MAP Kinase Kinase 7/physiology , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins , Protein Kinase C/metabolism , Mitogen-Activated Protein Kinase Kinase Kinase 11
16.
J Biol Chem ; 282(37): 27020-27029, 2007 Sep 14.
Article in English | MEDLINE | ID: mdl-17631503

ABSTRACT

Previously, we identified five active phosphatidylinositol ether lipid analogues (PIAs) that target the pleckstrin homology domain of Akt and selectively induce apoptosis in cancer cells with high levels of Akt activity. To examine specificity, PIAs were screened against a panel of 29 purified kinases. No kinase was inhibited, but one isoform of p38, p38alpha, was uniformly activated 2-fold. Molecular modeling of p38alpha revealed the presence of two regions that could interact with PIAs, one in the activation loop and a heretofore unappreciated region in the upper lobe that resembles a pleckstrin homology domain. In cells, two phases of activation were observed, an early phase that was independent of the upstream kinase MKK3/6 and inhibited by the p38 inhibitor SB203580 and a latter phase that was coincident with MKK3/6 activation. In short term xenograft experiments that employed immunohistochemistry and immunoblotting, PIA administration increased phosphorylation of p38 but not MKK3/6 in tumors in a statistically significant manner. Although PIAs rapidly activated p38 with similar time and dose dependence as Akt inhibition, p38 activation and Akt inhibition were independent events induced by PIAs. Using SB203580 or p38alpha(-/-) cells, we showed that p38alpha is not required for PIA-induced apoptosis but is required for H(2)O(2)- and anisomycin-induced apoptosis. Nonetheless, activation of p38a contributes to PIA-induced apoptosis, because reconstitution of p38a into p38alpha(-/-) cells increased apoptosis. These studies indicate that p38alpha is activated by PIAs through a novel mechanism and show that p38alpha activation contributes to PIA-induced cell death. Independent modulation of Akt and p38alpha could account for the profound cytotoxicity of PIAs.


Subject(s)
MAP Kinase Kinase 3/physiology , MAP Kinase Kinase 6/physiology , Mitogen-Activated Protein Kinase 14/metabolism , Phosphatidylinositols/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Humans , Hydrogen Peroxide/pharmacology , Mice , Mitogen-Activated Protein Kinase 14/chemistry , Models, Molecular , Phosphorylation
17.
FEMS Immunol Med Microbiol ; 50(3): 401-10, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17521390

ABSTRACT

Human anaplasmosis is an emerging infectious disease transmitted by ticks that can be potentially fatal in the immunocompromised and the elderly. The mechanisms of defense against the causative agent, Anaplasma phagocytophilum, are not completely understood; however, interferon (IFN)-gamma plays an important role in pathogen clearance. Here, we show that IFN-gamma is regulated through an early IL-12/23p40-dependent mechanism. Interleukin (IL)-12/23p40 is regulated in macrophages and dendritic cells after activation by microbial agonists and cytokines and constitutes a subunit of IL-12 and IL-23. IL-12/23p40-deficient mice displayed an increased A. phagocytophilum burden, accelerated thrombocytopenia and increased neutrophil numbers in the spleen at day 6 postinfection. Infection of MyD88- and mitogen-activated kinase kinase 3 (MKK3)-deficient mice suggested that the early susceptibility due to IL-12/23p40 deficiency was not dependent on signaling through MyD88 or MKK3. The lack of IL-12/23p40 reduced IFN-gamma production in both CD4(+) and CD8(+) T cells although the effect was more pronounced in CD4(+) T cells. Our data suggest that the immune response against A. phagocytophilum is a multifactorial and cooperative process. The IL-12/23p40 subunit drives the CD4(+) Th1 immune response in the early phase of infection and IL-12/23p40-independent mechanisms ultimately contribute to pathogen elimination from the host.


Subject(s)
Anaplasma phagocytophilum , Anaplasmosis/immunology , Interferon-gamma/metabolism , Interleukin-12 Subunit p40/physiology , Th1 Cells/immunology , Anaplasmosis/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , Genetic Predisposition to Disease , Humans , Interferon-gamma/blood , Interleukin-12 Subunit p40/genetics , Leukocyte Count , MAP Kinase Kinase 3/genetics , MAP Kinase Kinase 3/physiology , Mice , Mice, Mutant Strains , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/physiology , Neutrophils , Spleen/immunology , Thrombocytopenia/microbiology
18.
Cardiovasc Res ; 74(3): 466-70, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17399693

ABSTRACT

OBJECTIVES: Our aim was to examine the role of mitogen-activated protein kinase kinase 3 (MKK3) in the development of left ventricular (LV) remodeling following myocardial infarction (MI). METHODS: MKK3-null mice were subjected to permanent coronary artery ligation. Twenty-eight days after MI, haemodynamics in male mkk3+/+(WT) and mkk3-/-(KO) littermates were assessed using a pressure-conductance catheter. MI groups were compared to un-operated time-matched WT and KO controls. RESULTS: MI caused significant LV contractile dysfunction and dilatation which did not differ by genotype. Detailed morphometric analysis of excised hearts confirmed these similar global indices of remodeling and also demonstrated that pathological changes within remote myocardium and scar did not differ between KO and WT hearts. CONCLUSIONS: Despite numerous lines of evidence suggesting MKK3 is the relevant kinase upstream of p38 mitogen-activated protein kinase in LV remodeling these processes can continue in its absence.


Subject(s)
MAP Kinase Kinase 3/physiology , Myocardial Infarction/enzymology , Myocardium/enzymology , Ventricular Remodeling/physiology , Animals , Dilatation, Pathologic , Immunoblotting , MAP Kinase Kinase 3/genetics , Male , Mice , Mice, Knockout , Myocardial Contraction , Ventricular Function, Left
19.
Mol Cell Biol ; 27(1): 170-81, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17030606

ABSTRACT

MK2 and MK3 represent protein kinases downstream of p38 mitogen-activated protein kinase (MAPK). Deletion of the MK2 gene in mice resulted in an impaired inflammatory response although MK3, which displays extensive structural similarities and identical functional properties in vitro, is still present. Here, we analyze tumor necrosis factor (TNF) production and expression of p38 MAPK and tristetraprolin (TTP) in MK3-deficient mice and demonstrate that there are no significant differences with wild-type animals. We show that in vivo MK2 and MK3 are expressed and activated in parallel. However, the level of activity of MK2 is always significantly higher than that of MK3. Accordingly, we hypothesized that MK3 could have significant effects only in an MK2-free background and generated MK2/MK3 double-knockout mice. Unexpectedly, these mice are viable and show no obvious defects due to loss of compensation between MK2 and MK3. However, there is a further reduction of TNF production and expression of p38 and TTP in double-knockout mice compared to MK2-deficient mice. This finding, together with the observation that ectopically expressed MK3 can rescue MK2 deficiency similarly to MK2, indicates that both kinases share the same physiological function in vivo but are expressed to different levels.


Subject(s)
Gene Expression Regulation , MAP Kinase Kinase 3/physiology , MAP Kinase Signaling System , Protein Kinases/physiology , Tumor Necrosis Factor-alpha/biosynthesis , p38 Mitogen-Activated Protein Kinases/biosynthesis , Animals , CHO Cells , Cricetinae , Gene Deletion , Inflammation , Intracellular Signaling Peptides and Proteins , MAP Kinase Kinase 3/genetics , Macrophages/metabolism , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Protein Kinases/genetics , Protein Serine-Threonine Kinases , p38 Mitogen-Activated Protein Kinases/chemistry , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Cell Signal ; 18(3): 349-58, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16005186

ABSTRACT

The retinoid-inducible gene 1 (RIG1) protein is a retinoid-inducible growth regulator. Previous studies have shown that the RIG1 protein inhibits the signaling pathways of Ras/mitogen-activated protein kinases. However, neither the mode of action nor the site of inhibition of RIG1 is known. This study investigated the effects of RIG1, and the mechanisms responsible for these effects, on the activation of Ras proteins in HtTA cervical cancer cells. RIG1 reduced the levels of activated Ras (Ras-GTP) and total Ras protein in cells transfected with mutated H-, N-, or K-Ras(G12V), or in cells transfected with the wild type H- or N-Ras followed by stimulation with epidermal growth factor. The half-life of Ras protein decreased from more than 36 h in control cells to 18 h in RIG1-transfected cells. RIG1 immunoprecipitated with the Ras protein in co-transfected cellular lysates. In contrast to the predominant plasma membrane localization in control cells, the H-Ras fusion protein EGFP-H-Ras was localized within a discrete cytoplasmic compartment where it co-localized with RIG1. RIG1 inhibited more than 93% of the Elk- and CHOP-mediated transactivation induced by H- or K-Ras(G12V). However, RIG1 did not inhibit the transactivation induced by MEK1 or MEK3, and failed to suppress the phosphorylation of extracellular signal-regulated kinases 1 and 2 induced by the constitutively activated B-Raf(V599E). The RIG1 with carboxyl terminal truncation (RIG1DeltaC) did not immunoprecipitate with Ras and had no effect on Ras activation or transactivation of the downstream signal pathways. These data indicate that RIG1 exerts its inhibitory effect at the level of Ras activation, which is independent of Ras subtype but dependent on the membrane localization of the RIG1 protein. This inhibition of Ras activation may be mediated through downregulation of Ras levels and alteration of Ras subcellular distribution.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , RNA Helicases/metabolism , Signal Transduction/drug effects , ras Proteins/metabolism , Blotting, Western , Cell Line, Tumor , Cell Membrane/chemistry , DEAD Box Protein 58 , DEAD-box RNA Helicases , Down-Regulation , Epidermal Growth Factor/pharmacology , Female , Genes, ras , Humans , Intracellular Signaling Peptides and Proteins , MAP Kinase Kinase 1/physiology , MAP Kinase Kinase 3/physiology , MAP Kinase Signaling System/physiology , Mutation , Phospholipases A2, Calcium-Independent , Phosphorylation , Protein Isoforms , Proteins/metabolism , RNA Helicases/chemistry , Receptors, Immunologic , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Transfection , Tumor Suppressor Proteins , ras Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...