Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Int J Mol Sci ; 22(17)2021 Sep 02.
Article in English | MEDLINE | ID: mdl-34502457

ABSTRACT

(1) Background: The c-Jun-NH2-terminal protein kinase (JNK) is a mitogen-activated protein kinase involved in regulating physiological processes in the central nervous system. However, the dual genetic deletion of Mkk4 and Mkk7 (upstream activators of JNK) in adult mice is not reported. The aim of this study was to induce the genetic deletion of Mkk4/Mkk7 in adult mice and analyze their effect in hippocampal neurogenesis. (2) Methods: To achieve this goal, Actin-CreERT2 (Cre+/-), Mkk4flox/flox, Mkk7flox/flox mice were created. The administration of tamoxifen in these 2-month-old mice induced the gene deletion (Actin-CreERT2 (Cre+/-), Mkk4∆/∆, Mkk7∆/∆ genotype), which was verified by PCR, Western blot, and immunohistochemistry techniques. (3) Results: The levels of MKK4/MKK7 at 7 and 14 days after tamoxifen administration were not eliminated totally in CNS, unlike what happens in the liver and heart. These data could be correlated with the high levels of these proteins in CNS. In the hippocampus, the deletion of Mkk4/Mkk7 induced a misalignment position of immature hippocampal neurons together with alterations in their dendritic architecture pattern and maturation process jointly to the diminution of JNK phosphorylation. (4) Conclusion: All these data supported that the MKK4/MKK7-JNK pathway has a role in adult neurogenic activity.


Subject(s)
Hippocampus/physiology , MAP Kinase Kinase 4/physiology , MAP Kinase Kinase 7/physiology , MAP Kinase Signaling System , Neurogenesis , Animals , Doublecortin Protein , Gene Deletion , Mice, Transgenic
2.
Int J Mol Sci ; 22(17)2021 Sep 04.
Article in English | MEDLINE | ID: mdl-34502507

ABSTRACT

p38 and c-Jun N-terninal kinase (JNK) are activated in response to acute stress and inflammatory signals. Through modification of a plethora of substrates, these kinases profoundly re-shape cellular physiology for the optimal response to a harmful environment and/or an inflammatory state. Here, we utilized phospho-proteomics to identify several hundred substrates for both kinases. Our results indicate that the scale of signaling from p38 and JNK are of a similar magnitude. Among the many new targets, we highlight the regulation of the transcriptional regulators grb10-interacting GYF protein 1 and 2 (GIGYF1/2) by p38-dependent MAP kinase-activated protein kinase 2 (MK2) phosphorylation and 14-3-3 binding. We also show that the Golgi apparatus contains numerous substrates, and is a major target for regulation by p38 and JNK. When activated, these kinases mediate structural rearrangement of the Golgi apparatus, which positively affects protein flux through the secretory system. Our work expands on our knowledge about p38 and JNK signaling with important biological ramifications.


Subject(s)
MAP Kinase Kinase 4/metabolism , Stress, Physiological/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Carrier Proteins/metabolism , Cell Line, Tumor , Golgi Apparatus/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase 4/physiology , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/physiology
3.
Carcinogenesis ; 40(1): 41-51, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30475985

ABSTRACT

High-grade serous ovarian cancer (HGSOC) can originate in the fallopian tube epithelium (FTE), but the role of the ovary in these tumors is unclear. Tumorigenic murine oviductal epithelial (MOE) cells allografted in the ovarian bursa resulted in aggressive tumors that spread throughout the peritoneum whereas intraperitoneal xenografting the same number of cells did not form tumors, indicating that colonization of the ovary may play a role in metastasis. Physical tearing of the ovarian surface to mimic rupture of the ovary during ovulation (independent of hormonal changes) resulted in more MOE and HGSOC cells adhering to the ovary compared with intact ovaries. More MOE cells also adhered to three-dimensional (3D) collagen and primary ovarian stromal cells than to ovarian surface epithelia, indicating that FTE cells adhered to the extracellular matrix exposed during ovulation. However, plating cells on 3D collagen reduced the viability of normal FTE but not cancer cells. Mutation of p53 (R273H or R248W) and activation of Kirsten Rat Sarcoma Viral Oncogene Homolog (KRAS) (G12V) did not increase the viability of MOE cells on 3D collagen. In contrast, loss of phosphatase and tensin homolog (PTEN) allowed MOE cells to retain normal viability on 3D collagen. Loss of PTEN activated AKT and RAC1/c-jun N-terminal kinase signaling that each contributed to the increased viability, invasion and attachment in the collagen rich ovarian microenvironment. These results show that loss of PTEN activates multiple pathways that together enhance colonization of the ovary due to access to 3D collagen, which is a critical organ in the colonization of FTE-derived HGSOC.


Subject(s)
Extracellular Matrix/metabolism , Fallopian Tube Neoplasms/pathology , Ovary/pathology , Animals , Cell Line, Tumor , Female , MAP Kinase Kinase 4/physiology , Mice , Neoplasm Metastasis , PTEN Phosphohydrolase/physiology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Proto-Oncogene Proteins p21(ras)/physiology , rac1 GTP-Binding Protein/physiology
4.
J Neuroinflammation ; 13(1): 59, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26957005

ABSTRACT

BACKGROUND: Secretion of proteopathic α-synuclein (α-SNC) species from neurons is a suspected driving force in the propagation of Parkinson's disease (PD). We have previously implicated exophagy, the exocytosis of autophagosomes, as a dominant mechanism of α-SNC secretion in differentiated PC12 or SH-SY5Y nerve cells. Here we have examined the regulation of exophagy associated with different forms of nerve cell stress relevant to PD. RESULTS: We identify cJUN-N-terminal kinase (JNK) activity as pivotal in the secretory fate of autophagosomes containing α-SNC. Pharmacological inhibition or genetic (shRNA) knockdown of JNK2 or JNK3 decreases α-SNC secretion in differentiated PC12 and SH-SY5Y cells, respectively. Conversely, expression of constitutively active mitogen-activated protein kinase kinase 7 (MKK7)-JNK2 and -JNK3 constructs augment secretion. The transcriptional activity of cJUN was not required for the observed effects. We establish a causal relationship between increased α-SNC release by exophagy and JNK activation subsequent to lysosomal fusion deficiency (overexpression of Lewy body-localized protein p25α or bafilomycin A1). JNK activation following neuronal ER or oxidative stress was not correlated with exophagy, but of note, we demonstrate that reciprocal signaling between microglia and neurons modulates α-SNC secretion. NADPH oxidase activity of microglia cell lines was upregulated by direct co-culture with α-SNC-expressing PC12 neurons or by passive transfer of nerve cell-conditioned medium. Conversely, inflammatory factors secreted from activated microglia increased JNK activation and α-SNC secretion several-fold in PC12 cells. While we do not identify these factors, we extend our observations by showing that exposure of neurons in monoculture to TNFα, a classical pro-inflammatory mediator of activated microglia, is sufficient to increase α-SNC secretion in a mechanism dependent on JNK2 or JNK3. In continuation hereof, we show that also IFNß and TGFß increase the release of α-SNC from PC12 neurons. CONCLUSIONS: We implicate stress kinases of the JNK family in the regulation of exophagy and release of α-SNC following endogenous or exogenous stimulation. In a wider scope, our results imply that microglia not only inflict bystander damage to neurons in late phases of inflammatory brain disease but may also be active mediators of disease propagation.


Subject(s)
MAP Kinase Kinase 4/physiology , Microglia/physiology , Neurons/physiology , alpha-Synuclein/metabolism , Animals , Autophagy/physiology , Cell Communication/physiology , Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum Stress/physiology , Enzyme Activation , Gene Knockdown Techniques , Humans , Microglia/enzymology , Mitogen-Activated Protein Kinase 7/metabolism , Neurons/enzymology , PC12 Cells , Rats , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/pharmacology
5.
Mediators Inflamm ; 2015: 372931, 2015.
Article in English | MEDLINE | ID: mdl-26664149

ABSTRACT

The roots and rhizomes of Glycyrrhiza species (licorice) have been widely used as natural sweeteners and herbal medicines. The aim of this study is to investigate the effect of glycyrrhizic acid (GA) from licorice on macrophage polarization. Both phenotypic and functional activities of murine bone marrow-derived macrophages (BMDMs) treated by GA were assessed. Our results showed that GA obviously increased the cell surface expression of CD80, CD86, and MHCII molecules. Meanwhile, GA upregulated the expression of CCR7 and the production of TNF-α, IL-12, IL-6, and NO (the markers of classically activated (M1) macrophages), whereas it downregulated the expression of MR, Ym1, and Arg1 (the markers of alternatively activated (M2) macrophage). The functional tests showed that GA dramatically enhanced the uptake of FITC-dextran and E. coli K88 by BMDMs and decreased the intracellular survival of E. coli K88 and S. typhimurium. Moreover, we demonstrated that JNK and NF-κB activation are required for GA-induced NO and M1-related cytokines production, while ERK1/2 pathway exhibits a regulatory effect via induction of IL-10. Together, these findings indicated that GA promoted polarization of M1 macrophages and enhanced its phagocytosis and bactericidal capacity. The results expanded our knowledge about the role of GA in macrophage polarization.


Subject(s)
Bone Marrow Cells/drug effects , Cell Polarity/drug effects , Glycyrrhizic Acid/pharmacology , MAP Kinase Kinase 4/physiology , Macrophages/drug effects , NF-kappa B/physiology , Animals , Bone Marrow Cells/physiology , Enzyme Activation , Macrophages/physiology , Mice , Mice, Inbred C57BL , Nitric Oxide/biosynthesis , Phagocytosis/drug effects , Signal Transduction/drug effects
6.
Arthritis Rheumatol ; 67(7): 1826-36, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25776267

ABSTRACT

OBJECTIVE: Aggrecan enables articular cartilage to bear load and resist compression. Aggrecan loss occurs early in osteoarthritis and rheumatoid arthritis and can be induced by inflammatory cytokines such as interleukin-1 (IL-1). IL-1 induces cleavage of specific aggrecans characteristic of the ADAMTS proteinases. The aim of this study was to identify the intracellular signaling pathways by which IL-1 causes aggrecan degradation by human chondrocytes and to investigate how aggrecanase activity is controlled by chondrocytes. METHODS: We developed a cell-based assay combining small interfering RNA (siRNA)-induced knockdown with aggrecan degradation assays. Human articular chondrocytes were overlaid with bovine aggrecan after transfection with siRNAs against molecules of the IL-1 signaling pathway. After IL-1 stimulation, released aggrecan fragments were detected with AGEG and ARGS neoepitope antibodies. Aggrecanase activity and tissue inhibitor of metalloproteinases 3 levels were measured by enzyme-linked immunosorbent assay. Low-density lipoprotein receptor-related protein 1 (LRP-1) shedding was analyzed by Western blotting. RESULTS: ADAMTS-5 is a major aggrecanase in human chondrocytes, regulating aggrecan degradation in response to IL-1. The tumor necrosis factor receptor-associated 6 (TRAF-6)/transforming growth factor ß-activated kinase 1 (TAK-1)/MKK-4 signaling axis is essential for IL-1-induced aggrecan degradation, while NF-κB is not. Of the 3 MAPKs (ERK, p38, and JNK), only JNK-2 showed a significant role in aggrecan degradation. Chondrocytes constitutively secreted aggrecanase, which was continuously endocytosed by LRP-1, keeping the extracellular level of aggrecanase low. IL-1 induced aggrecanase activity in the medium in a JNK-2-dependent manner, possibly by reducing aggrecanase endocytosis, because IL-1 caused JNK-2-dependent shedding of LRP-1. CONCLUSION: The signaling axis TRAF-6/TAK-1/MKK-4/JNK-2 mediates IL-1-induced aggrecanolysis. The level of aggrecanase is controlled by its endocytosis, which may be reduced upon IL-1 stimulation because of LRP-1 shedding.


Subject(s)
Aggrecans/metabolism , Chondrocytes/metabolism , Interleukin-1/physiology , MAP Kinase Kinase 7/physiology , Signal Transduction/physiology , ADAM Proteins/physiology , ADAMTS5 Protein , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/pathology , Humans , Interleukin-1/pharmacology , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , MAP Kinase Kinase 4/physiology , MAP Kinase Kinase Kinases/physiology , RNA, Small Interfering/pharmacology , TNF Receptor-Associated Factor 6/physiology
7.
J Invest Dermatol ; 135(5): 1435-1444, 2015 May.
Article in English | MEDLINE | ID: mdl-25634355

ABSTRACT

Previous wound healing studies have failed to define a role for either α1ß1 or α2ß1 integrin in fibroblast-mediated wound contraction, suggesting the involvement of another collagen receptor in this process. Our previous work demonstrated that the integrin subunit α11 is highly induced during wound healing both at the mRNA and protein level, prompting us to investigate and dissect the role of the integrin α11ß1 during this process. Therefore, we used mice with a global ablation of either α2 or α11 or both integrin subunits and investigated the repair of excisional wounds. Analyses of wounds demonstrated that α11ß1 deficiency results in reduced granulation tissue formation and impaired wound contraction, independently of the presence of α2ß1. Our combined in vivo and in vitro data further demonstrate that dermal fibroblasts lacking α11ß1 are unable to efficiently convert to myofibroblasts, resulting in scar tissue with compromised tensile strength. Moreover, we suggest that the reduced stability of the scar is a consequence of poor collagen remodeling in α11(-/-) wounds associated with defective transforming growth factor-ß-dependent JNK signaling.


Subject(s)
Cicatrix/pathology , Cicatrix/physiopathology , Granulation Tissue/physiology , Integrins/deficiency , Receptors, Collagen/deficiency , Tensile Strength/physiology , Wound Healing/physiology , Animals , Cell Differentiation/physiology , Cells, Cultured , Collagen/physiology , Female , Granulation Tissue/pathology , In Vitro Techniques , Integrins/genetics , Integrins/physiology , MAP Kinase Kinase 4/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Myofibroblasts/pathology , Myofibroblasts/physiology , Receptors, Collagen/genetics , Receptors, Collagen/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/physiology
8.
J Am Heart Assoc ; 3(3): e000747, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24811612

ABSTRACT

BACKGROUND: Although increased amounts of reactive oxygen species in the pathogenesis of abdominal aortic aneurysm (AAA) are well documented, the precise molecular mechanisms by which reactive oxygen species induce AAAs have not been fully elucidated. This study focused on the role of hydrogen peroxide-inducible clone 5 (Hic-5), which is induced by hydrogen peroxide and transforming growth factor-ß, in the cellular signaling of AAA pathogenesis. METHODS AND RESULTS: Using the angiotensin II-induced AAA model in Apoe(-/-) mice, we showed that Apoe(-/-)Hic-5(-/-) mice were completely protected from AAA formation and aortic rupture, whereas Apoe(-/-) mice were not. These features were similarly observed in smooth muscle cell-specific Hic-5-deficient mice. Furthermore, angiotensin II treatment induced Hic-5 expression in a reactive oxygen species-dependent manner in aortic smooth muscle cells in the early stage of AAA development. Mechanistic studies revealed that Hic-5 interacted specifically with c-Jun N-terminal kinase p54 and its upstream regulatory molecule mitogen-activated protein kinase kinase 4 as a novel scaffold protein, resulting in the expression of membrane type 1 matrix metalloproteinase and matrix metalloproteinase 2 activation in aortic smooth muscle cells. CONCLUSION: Hic-5 serves as a novel scaffold protein that specifically activates the mitogen-activated protein kinase kinase 4/p54 c-Jun N-terminal kinase pathway, thereby leading to the induction and activation of matrix metalloproteinases in smooth muscle cells and subsequent AAA formation. Our study provided a novel therapeutic option aimed at inhibiting the mitogen-activated protein kinase kinase 4-Hic-5-p54 c-Jun N-terminal kinase pathway in the vessel wall, particularly through Hic-5 inhibition, which may be used to produce more precise and effective therapies.


Subject(s)
Aortic Aneurysm, Abdominal/etiology , Cytoskeletal Proteins/physiology , DNA-Binding Proteins/physiology , LIM Domain Proteins/physiology , MAP Kinase Kinase 4/physiology , Angiotensin II/pharmacology , Animals , Aorta, Abdominal/drug effects , Aorta, Abdominal/physiopathology , Disease Models, Animal , MAP Kinase Signaling System/physiology , Male , Membrane Proteins , Mice, Knockout , Phosphoproteins , Reactive Oxygen Species/metabolism , Signal Transduction/physiology
9.
Rheumatology (Oxford) ; 53(6): 1022-33, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24501244

ABSTRACT

OBJECTIVE: Autophagy is a potential protective mechanism that is involved in several degenerative diseases. Nitric oxide (NO) is associated with programmed cellular death in meniscal cells, but whether it can induce autophagy is still undetermined. This study aims to investigate the interaction between autophagy and NO in normal human meniscal cells. METHODS: Normal meniscal cells were harvested from female patients. NO donors and NO synthase inhibitors were used to regulate the level of NO. Changes in the incidence of autophagy and apoptosis were examined using flow cytometry, western blot and immunofluorescence methods. The effects of NO-mediated autophagy regulation of the expression of MMPs and aggrecanases (ADAMTS-4 and -5) were analysed by real-time PCR. RESULTS: NO donors inhibited autophagy as well as augmented apoptosis in human meniscal cells with serum deprivation. Conversely, treatment with NOS inhibitors resulted in up-regulation of the autophagy level while repressing apoptosis. NOS inhibitor treatment also resulted in down-regulation of MMPs and aggrecanase mRNA expression. This effect of NOS inhibitor was also blocked by autophagy inhibitors. Our results also showed that NOS inhibitor enhanced Jun-N-terminal kinase (JNK) activation. Furthermore, SP600125, a selective JNK inhibitor, blocked up-regulation of autophagy by NOS inhibitor. CONCLUSION: Our results demonstrated that NO augmented serum deprivation-induced apoptosis of meniscal cells via inhibition of autophagy through inactivation of JNK. Up-regulation of autophagy may be a potential approach in the treatment of meniscal tissue degeneration.


Subject(s)
Autophagy/physiology , MAP Kinase Kinase 4/antagonists & inhibitors , Menisci, Tibial/enzymology , Nitric Oxide/physiology , Adolescent , Adult , Apoptosis/drug effects , Apoptosis/physiology , Autophagy/drug effects , Biomarkers/metabolism , Cells, Cultured , Culture Media, Serum-Free , Down-Regulation/drug effects , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Enzymologic/drug effects , Humans , MAP Kinase Kinase 4/physiology , Menisci, Tibial/cytology , Menisci, Tibial/drug effects , Menisci, Tibial/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , RNA, Messenger/genetics , Young Adult
10.
J Neurochem ; 126(1): 58-69, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23646899

ABSTRACT

The α2-Antiplasmin (α2AP) protein is known as a principal physiological inhibitor of plasmin, but we previously demonstrated that it acts as a regulatory factor for cellular functions independent of plasmin. α2AP is highly expressed in the hippocampus, suggesting a potential role for α2AP in hippocampal neuronal functions. However, the role for α2AP was unclear. This study is the first to investigate the involvement of α2AP in the dendritic growth of hippocampal neurons. The expression of microtubule-associated protein 2, which contributes to neurite initiation and neuronal growth, was lower in the neurons from α2AP⁻/⁻ mice than in the neurons from α2AP⁺/⁺ mice. Exogenous treatment with α2AP enhanced the microtubule-associated protein 2 expression, dendritic growth and filopodia formation in the neurons. This study also elucidated the mechanism underlying the α2AP-induced dendritic growth. Aprotinin, another plasmin inhibitor, had little effect on the dendritic growth of neurons, and α2AP induced its expression in the neurons from plaminogen⁻/⁻ mice. The activation of p38 MAPK was involved in the α2AP-induced dendritic growth. Therefore, our findings suggest that α2AP induces dendritic growth in hippocampal neurons through p38 MAPK activation, independent of plasmin, providing new insights into the role of α2AP in the CNS.


Subject(s)
Dendrites/physiology , Hippocampus/cytology , Hippocampus/growth & development , Neurons/physiology , alpha-2-Antiplasmin/physiology , Animals , Blotting, Western , Cells, Cultured , Fibrinolysin/physiology , Hippocampus/physiology , Immunohistochemistry , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/physiology , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/physiology , Plasminogen/genetics , Plasminogen/physiology , Real-Time Polymerase Chain Reaction , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/physiology
11.
Arch Oral Biol ; 58(6): 731-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23332208

ABSTRACT

Matrix metalloproteinases (MMPs) are known to play a key role during orthodontic treatment leading to periodontal remodelling and tooth movement. MMPs may be induced by mechanical forces. However, the role played by toll-like receptors (TLRs) in modulating the effects of the mechanical force on periodontal fibroblasts is not known. To investigate the interaction between mechanical force and TLR stimulation, primary cultures of human periodontal fibroblasts were submitted to centrifugation in the presence of LPS and Pam3Cys, which are known TLR-4 and TLR-2 ligands, respectively. The expression of MMP-1, -2, -3, -8, -9, -10 and -13; TIMP (Tissue Inhibitor of Metalloproteinases) -1, -2 and -4; TNF-α (Tumour Necrosis Factor alpha); IL-1ß (Interleukin 1 beta); ERK 1/2 (Extracellular Signal-Regulated Kinase 1/2); p38; JNK (c-jun N-terminal Kinase); IRAK1 (Interleukin-1 Receptor-Associated Kinase); and NF-κB (Nuclear Factor kappa B) were measured by antibody array, ELISA and immunoblotting methods. The activation of TLRs associated with centrifugation induced an increase in the secretion of MMPs 1, 3 and 10, with no increase in TNF-α or IL-1ß. An increase in the phosphorylation of the MAP kinases p38 and JNK and the transcription factor NF-κB, without an increase in TIMPs was also observed. These findings suggest that the secretion of MMPs by cultured periodontal fibroblasts that is induced by combined TLR activation and mechanical force stimulation is regulated via the p38, JNK and NF-κB pathways. The increased secretion of MMPs by TLR activation may be an important factor that should be considered during orthodontic treatment.


Subject(s)
Fibroblasts/enzymology , MAP Kinase Kinase 4/physiology , Matrix Metalloproteinase 10/metabolism , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/metabolism , NF-kappa B/physiology , Periodontal Ligament/cytology , Toll-Like Receptor 2/physiology , Toll-Like Receptor 4/physiology , p38 Mitogen-Activated Protein Kinases/physiology , Biomechanical Phenomena , Cell Culture Techniques , Cells, Cultured , Centrifugation , Cytotoxins/pharmacology , Fibroblasts/metabolism , Humans , Interleukin-1beta/analysis , Lipopolysaccharides/pharmacology , Lipoproteins/pharmacology , MAP Kinase Signaling System/physiology , Matrix Metalloproteinase 1/analysis , Matrix Metalloproteinase 10/analysis , Matrix Metalloproteinase 3/analysis , Periodontal Ligament/enzymology , Stress, Mechanical , Tissue Inhibitor of Metalloproteinase-1/analysis , Tissue Inhibitor of Metalloproteinase-2/analysis , Tissue Inhibitor of Metalloproteinases/analysis , Toll-Like Receptor 2/drug effects , Toll-Like Receptor 4/drug effects , Tumor Necrosis Factor-alpha/analysis , Tissue Inhibitor of Metalloproteinase-4
12.
J Gastroenterol ; 48(2): 222-37, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22722906

ABSTRACT

BACKGROUND: Butein has been reported to prevent and partly reverse liver fibrosis in vivo; however, the mechanisms of its action are poorly understood. We, therefore, aimed to determine the antifibrotic potential of butein. METHODS: We assessed the influence of the incubation of hepatic stellate cells (HSCs) and hepatoma cells (HepG2) with butein on sensitivity to ethanol- or acetaldehyde-induced toxicity; the production of reactive oxygen species (ROS); the expression of markers of HSC activation, including smooth muscle α-actin (α-SMA) and procollagen I; and the production of transforming growth factor-ß1 (TGF-ß1), metalloproteinases-2 and -13 (MMP-2and MMP-13), and tissue inhibitors of metalloproteinases (TIMPs). The influence of butein on intracellular signals in HSCs; i.e., nuclear factor-κB (NFκB), c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase (p38 MAPK) induced by ethanol was estimated. RESULTS: Butein protected HSCs and HepG2 cells against ethanol toxicity by the inhibition of ethanol- or acetaldehyde-induced production of ROS when cells were incubated separately or in co-cultures; butein also inhibited HSC activation measured as the production of α-SMA and procollagen I. As well, butein downregulated ethanol- or acetaldehyde-induced HSC migration and the production of TGF-ß, TIMP-1, and TIMP-2; decreased the activity of MMP-2; and increased the activity of MMP-13. In ethanol-induced HSCs, butein inhibited the activation of the p38 MAPK and JNK transduction pathways as well as significantly inhibiting the phosphorylation of NF κB inhibitor (IκB) and Smad3. CONCLUSIONS: The results indicated that butein inhibited ethanol- and acetaldehyde-induced activation of HSCs at different levels, acting as an antioxidant and inhibitor of ethanol-induced MAPK, TGF-ß, and NFκB/IκB transduction signaling; this result makes butein a promising agent for antifibrotic therapies.


Subject(s)
Antioxidants/pharmacology , Chalcones/pharmacology , Ethanol/antagonists & inhibitors , Hepatic Stellate Cells/drug effects , Oxidative Stress/drug effects , Acetaldehyde/antagonists & inhibitors , Acetaldehyde/pharmacology , Actins/biosynthesis , Animals , Cell Death/drug effects , Cells, Cultured , Coculture Techniques , Collagen Type I/biosynthesis , Drug Evaluation, Preclinical/methods , Ethanol/pharmacology , Hep G2 Cells , Hepatic Stellate Cells/metabolism , Humans , MAP Kinase Kinase 4/physiology , NF-kappa B/physiology , Oxidative Stress/physiology , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , p38 Mitogen-Activated Protein Kinases/physiology
13.
Gastroenterology ; 143(6): 1530-43, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22967725

ABSTRACT

BACKGROUND & AIMS: Stimulation of nucleotide-binding oligomerization domain-containing (Nod)2 and other pattern recognition receptors (PRR) in human monocyte-derived macrophages induces interleukin (IL)-1, which increases mitogen-activated protein kinase (MAPK) activation and cytokine secretion. Activation of MAPK by PRR has varied effects on inflammatory cytokine secretion. We investigated whether different levels of autocrine IL-1 mediate these varied effects. METHODS: Macrophage responses to PRR ligands were analyzed by enzyme-linked immunosorbent assay and flow cytometry. We overexpressed or reduced MAPK levels (using small inhibitory RNA). RESULTS: Nod2 and other PRR activated signaling via extracellular signal-related kinase (ERK) and p38 that inhibited inflammatory cytokine production by human monocyte-derived macrophages; autocrine IL-1 production prevented this inhibition. ERK and p38 inhibited inflammatory cytokine production by human macrophages that produce low levels of IL-1 (such as M2, endotoxin-tolerant, and intestinal macrophages); adding exogenous IL-1 caused ERK and p38 to stimulate production of inflammatory cytokines in these cells. In mouse macrophages, which do not produce IL-1 in response to PRR stimulation alone, addition of exogenous IL-1 reversed the ERK-mediated inhibition of IL-12p40. Increasing activation of c-Jun N-terminal kinase in Nod2-stimulated human monocyte-derived macrophages, in the absence of autocrine IL-1 signaling, caused ERK and p38 to stimulate inflammatory cytokines secretion. Importantly, infection of human intestinal macrophages with pathogens that induce IL-1 production reversed the inhibition of inflammatory cytokine production by ERK and p38. CONCLUSIONS: In response to PRR stimulation of macrophages, the level of MAPK signaling is regulated by autocrine IL-1 and determines whether production of inflammatory cytokines is inhibited or stimulated. This mechanism could account for reported differences in MAPK regulation of inflammatory cytokines and propagate the inflammatory response to pathogens.


Subject(s)
Cytokines/metabolism , Extracellular Signal-Regulated MAP Kinases/physiology , Interleukin-1/physiology , Macrophages/metabolism , Nod2 Signaling Adaptor Protein/pharmacology , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Cells, Cultured , Humans , Ligands , MAP Kinase Kinase 4/physiology , Macrophages/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Animal , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Receptors, Pattern Recognition/physiology , Salmonella typhimurium/physiology
14.
Int J Radiat Oncol Biol Phys ; 84(4): e515-23, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22901381

ABSTRACT

PURPOSE: Cell invasion represents one of the major determinants that treatment has failed for patients suffering from glioblastoma. Contrary findings have been reported for cell migration upon exposure to ionizing radiation. Here, the migration and invasion capability of glioblastoma cells on and in collagen type I were evaluated upon irradiation with X-rays or carbon ions. METHODS AND MATERIALS: Migration on and invasion in collagen type I were evaluated in four established human glioblastoma cell lines exposed to either X-rays or carbon ions. Furthermore, clonogenic radiation survival, proliferation (5-bromo-2-deoxyuridine positivity), DNA double-strand breaks (γH2AX/53BP1-positive foci), and expression of invasion-relevant proteins (eg, ß1 integrin, FAK, MMP2, and MMP9) were explored. Migration and invasion assays for primary glioblastoma cells also were carried out with X-ray irradiation. RESULTS: Neither X-ray nor carbon ion irradiation affected glioblastoma cell migration and invasion, a finding similarly observed in primary glioblastoma cells. Intriguingly, irradiated cells migrated unhampered, despite DNA double-strand breaks and reduced proliferation. Clonogenic radiation survival was increased when cells had contact with extracellular matrix. Specific inhibition of the ß1 integrin or proliferation-associated signaling molecules revealed a critical function of JNK, PI3K, and p38 MAPK in glioblastoma cell invasion. CONCLUSIONS: These findings indicate that X-rays and carbon ion irradiation effectively reduce proliferation and clonogenic survival without modifying the migration and invasion ability of glioblastoma cells in a collagen type I environment. Addition of targeted agents against members of the MAPK and PI3K signaling axis to conventional chemoradiation therapy seems potentially useful to optimize glioblastoma therapy.


Subject(s)
Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Cell Movement/radiation effects , Glioblastoma/pathology , Glioblastoma/radiotherapy , Neoplasm Invasiveness , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Bromodeoxyuridine/analysis , Carbon , Cell Migration Assays/methods , Cell Proliferation/radiation effects , Collagen Type I , DNA Breaks, Double-Stranded , Glioblastoma/genetics , Glioblastoma/metabolism , Histones/analysis , Humans , Integrin beta1/physiology , Intracellular Signaling Peptides and Proteins/analysis , MAP Kinase Kinase 4/physiology , Phosphatidylinositol 3-Kinases/physiology , Tumor Suppressor p53-Binding Protein 1 , p38 Mitogen-Activated Protein Kinases/physiology
15.
J Immunol ; 189(2): 860-6, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22685314

ABSTRACT

Nucleated cells are equipped with several mechanisms that support their resistance to complement-dependent cytotoxicity (CDC). The role of the NF-κB pathway in cell protection from CDC was examined. Elevated sensitivity to CDC was demonstrated in cells lacking the p65 subunit of NF-κB or the IκB kinases IKKα or IKKß, and in cells treated with p65 small interfering RNA. Pretreatment with the IKK inhibitor PS-1145 also enhanced CDC of wild-type cells (WT) but not of p65(-/-) cells. Furthermore, reconstitution of p65 into p65(-/-) cells and overexpression of p65 in WT cells lowered their sensitivity to CDC. The postulated effect of p65 on the JNK-mediated death-signaling pathway activated by complement was examined. p65 small interfering RNA enhanced CDC in WT cells but not in cells lacking JNK. JNK phosphorylation induced by complement was more pronounced in p65(-/-) cells than in WT cells. The results indicate that the NF-κB pathway mediates cell resistance to CDC, possibly by suppressing JNK-dependent programmed necrotic cell death.


Subject(s)
Complement Activation/immunology , Complement Membrane Attack Complex/physiology , Cytotoxicity, Immunologic/immunology , Signal Transduction/immunology , Transcription Factor RelA/physiology , Animals , Cell Communication/genetics , Cell Communication/immunology , Complement Activation/genetics , Complement Membrane Attack Complex/deficiency , Complement Membrane Attack Complex/metabolism , Cytotoxicity, Immunologic/genetics , Embryonic Stem Cells/enzymology , Embryonic Stem Cells/immunology , Embryonic Stem Cells/metabolism , Fibroblasts/enzymology , Fibroblasts/immunology , Fibroblasts/metabolism , HEK293 Cells , HeLa Cells , Humans , I-kappa B Kinase/deficiency , MAP Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Kinase 4/metabolism , MAP Kinase Kinase 4/physiology , Mice , Mice, Knockout , Protein Subunits/deficiency , Signal Transduction/genetics , Transcription Factor RelA/deficiency , Transcription Factor RelA/metabolism
16.
Cancer Metastasis Rev ; 31(3-4): 605-13, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22706843

ABSTRACT

Host tissue microenvironment plays key roles in cancer progression and colonization of secondary organs. One example is ovarian cancer, which colonizes the peritoneal cavity and especially the omentum. Our research indicates that the interaction of ovarian cancer cells with the omental microenvironment can activate a stress-kinase pathway involving the mitogen-activated protein kinase kinase 4 (MKK4). A combination of clinical correlative and functional data suggests that MKK4 activation suppresses growth of ovarian cancer cells lodged in omentum. These findings prompted us to turn our focus to the cellular composition of the omental microenvironment and its role in regulating cancer growth. In this review, in addition to providing an overview of MKK4 function, we highlight a use for metastasis suppressors as a molecular tool to study cancer cell interaction with its microenvironment. We review features of the omentum that makes it a favorable microenvironment for metastatic colonization. In conclusion, a broader, evolutionary biology perspective is presented which we believe needs to be considered when studying the evolution of cancer cells within a defined microenvironment. Taken together, this approach can direct new multi-dimensional lines of research aimed at a mechanistic understanding of host tissue microenvironment, which could be used to realize novel targets for future research.


Subject(s)
MAP Kinase Kinase 4/physiology , Neoplasm Metastasis/prevention & control , Tumor Microenvironment , Tumor Suppressor Proteins/physiology , Animals , Female , Humans , Neoplasm Metastasis/pathology , Ovarian Neoplasms/pathology
17.
Brain Res ; 1463: 21-9, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22575563

ABSTRACT

Basic fibroblast growth factor (FGF-2) has a neuroprotective effect. Astrocytes support neurons by releasing neurotrophic factors including glial cell line-derived neurotrophic factor (GDNF). FGF-2 stimulates GDNF synthesis in astrocytes and the release. It has been reported that FGF-2 induces the activation of p44/p42 mitogen-activated protein (MAP) kinase, stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) and p38 MAP kinase in C6 glioma cells, and that FGF-2 stimulates GDNF release through p44/p42 MAP kinase or SAPK/JNK, but not p38 MAP kinase. In the present study, we investigated the exact mechanism of FGF-2-induced GDNF release from C6 cells. FGF-2 induced the phosphorylation of Akt and its substrate, glycogen synthase kinase 3ß (GSK3ß) in addition to three MAP kinases in these cells. FGF-2-stimulated release of GDNF was suppressed by wortmannin (a phosphatidylinositol 3 (PI3)-kinase inhibitor) or LY294002 (another PI3-kinase inhibitor). The FGF-2-induced GDNF release from PI3-kinase-downregulated C6 cells was decreased compared with that in control siRNA-transfected cells. PD98059 (an inhibitor of MEK 1/2) or SP600125 (an inhibitor of SAPK/JNK), which suppressed FGF-2-induced phosphorylation of p44/p42 MAP kinase or SAPK/JNK respectively, did not affect FGF-2-induced Akt phosphorylation. Wortmannin or LY294002, which attenuated FGF-2-induced phosphorylation of Akt and GSK3ß, had no effect on FGF-2-induced phosphorylation of p44/p42 MAP kinase or SAPK/JNK. These results strongly suggest that the PI3-kinase/Akt pathway plays a positive role in FGF-2-stimulated GDNF release independently of p44/p42 MAP kinase or SAPK/JNK in C6 glioma cells.


Subject(s)
Fibroblast Growth Factor 2/physiology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glioma/metabolism , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Animals , Cell Line , Cell Line, Tumor , Chromones/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/agonists , Glioma/enzymology , MAP Kinase Kinase 4/physiology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/physiology , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Plant Proteins , Rats , Up-Regulation/drug effects , Up-Regulation/physiology
18.
Acta Neurochir (Wien) ; 154(7): 1197-203, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22638597

ABSTRACT

BACKGROUND: Recent studies suggest that endoplasmic reticulum stress (ERS) is the key process in ischemic brain injury. The JNK pathway is also involved in the process of ischemic brain injury. METHOD: We established a middle cerebral artery occlusion/reperfusion (MCAO/R) model in rats; detected the changes in c-Jun N-terminal kinase (JNK), GADD153 and caspase-12 at different reperfusion time points by immunohistochemistry, Western blot and double-label immunofluorescence; and observed the effect of JNK inhibitor SP600125 on the expression of JNK, GADD153 and caspase-12 to explore the relationship between the endoplasmic reticulum road and JNK pathway. RESULTS: The expression of the two hallmarks of ERS-GADD153 and caspase-12-significantly increased, and the activation of JNK also obviously increased. After interference by SP600125, the expression of p-JNk and caspase-12 obviously decreased, whereas the decrease of GADD153 occurred only after 24 h reperfusion. CONCLUSIONS: Both ERS and JNK pathways are involved in the pathological process of ischemic brain injury. The JNK pathway may be involved in the process of ERS, but perhaps has more effect on the caspase-12 pathway.


Subject(s)
Apoptosis/physiology , Brain/blood supply , Endoplasmic Reticulum/physiology , MAP Kinase Kinase 4/physiology , Reperfusion Injury/physiopathology , Animals , Blotting, Western , Brain/pathology , Caspase 12/metabolism , Corpus Striatum/pathology , Corpus Striatum/physiopathology , Endoplasmic Reticulum/pathology , Frontal Lobe/pathology , Frontal Lobe/physiopathology , Image Processing, Computer-Assisted , Male , Microscopy, Fluorescence , Parietal Lobe/pathology , Parietal Lobe/physiopathology , Rats , Rats, Wistar , Reperfusion Injury/pathology , Transcription Factor CHOP/metabolism
19.
Am J Physiol Renal Physiol ; 302(9): F1210-23, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22301622

ABSTRACT

We investigated the signaling basis for tubule pathology during fibrosis after renal injury. Numerous signaling pathways are activated physiologically to direct tubule regeneration after acute kidney injury (AKI) but several persist pathologically after repair. Among these, transforming growth factor (TGF)-ß is particularly important because it controls epithelial differentiation and profibrotic cytokine production. We found that increased TGF-ß signaling after AKI is accompanied by PTEN loss from proximal tubules (PT). With time, subpopulations of regenerating PT with persistent loss of PTEN (phosphate and tension homolog) failed to differentiate, became growth arrested, expressed vimentin, displayed profibrotic JNK activation, and produced PDGF-B. These tubules were surrounded by fibrosis. In contrast, PTEN recovery was associated with epithelial differentiation, normal tubule repair, and less fibrosis. This beneficial outcome was promoted by TGF-ß antagonism. Tubule-specific induction of TGF-ß led to PTEN loss, JNK activation, and fibrosis even without prior AKI. In PT culture, high TGF-ß depleted PTEN, inhibited differentiation, and activated JNK. Conversely, TGF-ß antagonism increased PTEN, promoted differentiation, and decreased JNK activity. Cre-Lox PTEN deletion suppressed differentiation, induced growth arrest, and activated JNK. The low-PTEN state with JNK signaling and fibrosis was ameliorated by contralateral nephrectomy done 2 wk after unilateral ischemia, suggesting reversibility of the low-PTEN dysfunctional tubule phenotype. Vimentin-expressing tubules with low-PTEN and JNK activation were associated with fibrosis also after tubule-selective AKI, and with human chronic kidney diseases of diverse etiology. By preventing tubule differentiation, the low-PTEN state may provide a platform for signals initiated physiologically to persist pathologically and cause fibrosis after injury.


Subject(s)
Cell Differentiation , Kidney Tubules, Proximal/pathology , MAP Kinase Kinase 4/physiology , PTEN Phosphohydrolase/deficiency , Phenotype , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Animals , Cells, Cultured , Chronic Disease , Fibrosis , Humans , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Kidney Tubules, Proximal/physiopathology , Male , Mice , Mice, Transgenic , Models, Animal , Rats , Rats, Sprague-Dawley , Regeneration/physiology , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology
20.
Biochemistry ; 50(48): 10520-9, 2011 Dec 06.
Article in English | MEDLINE | ID: mdl-22047447

ABSTRACT

Arrestins make up a small family of proteins with four mammalian members that play key roles in the regulation of multiple G protein-coupled receptor-dependent and -independent signaling pathways. Although arrestins were reported to serve as scaffolds for MAP kinase cascades, promoting the activation of JNK3, ERK1/2, and p38, the molecular mechanisms involved were not elucidated, and even the direct binding of arrestins with MAP kinases was never demonstrated. Here, using purified proteins, we show that both nonvisual arrestins directly bind JNK3α2 and its upstream activator MKK4, and that the affinity of arrestin-3 for these kinases is higher than that of arrestin-2. Reconstitution of the MKK4-JNK3α2 signaling module from pure proteins in the presence of different arrestin-3 concentrations showed that arrestin-3 acts as a "true" scaffold, facilitating JNK3α2 phosphorylation by bringing the two kinases together. Both the level of JNK3α2 phosphorylation by MKK4 and JNK3α2 activity toward its substrate ATF2 increase at low and then decrease at high arrestin-3 levels, yielding a bell-shaped concentration dependence expected with true scaffolds that do not activate the upstream kinase or its substrate. Thus, direct binding of both kinases and true scaffolding is the molecular mechanism of action of arrestin-3 on the MKK4-JNK3α2 signaling module.


Subject(s)
Arrestins/physiology , MAP Kinase Kinase 4/physiology , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 10/metabolism , Animals , Arrestins/metabolism , Cattle , Cells, Cultured , Humans , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 4/metabolism , Mitogen-Activated Protein Kinase 10/physiology , Phosphorylation/physiology , Protein Binding/physiology , Up-Regulation/physiology , beta-Arrestins
SELECTION OF CITATIONS
SEARCH DETAIL
...