Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Cell Death Dis ; 11(5): 343, 2020 05 11.
Article in English | MEDLINE | ID: mdl-32393755

ABSTRACT

Lipid-based RNA nanocarriers have been recently accepted as a novel therapeutic option in humans, thus increasing the therapeutic options for patients. Tailored nanomedicines will enable to treat chronic liver disease (CLD) and end-stage liver cancer, disorders with high mortality and few treatment options. Here, we investigated the curative potential of gene therapy of a key molecule in CLD, the c-Jun N-terminal kinase-2 (Jnk2). Delivery to hepatocytes was achieved using a lipid-based clinically employable siRNA formulation that includes a cationic aminolipid to knockdown Jnk2 (named siJnk2). After assessing the therapeutic potential of siJnk2 treatment, non-invasive imaging demonstrated reduced apoptotic cell death and improved hepatocarcinogenesis was evidenced by improved liver parenchyma as well as ameliorated markers of hepatic damage, reduced fibrogenesis in 1-year-old mice. Strikingly, chronic siJnk2 treatment reduced premalignant nodules, indicative of tumor initiation. Furthermore, siJnk2 treatment led to a significant activation of the immune cell compartment. In conclusion, Jnk2 knockdown in hepatocytes ameliorated hepatitis, fibrogenesis, and initiation of hepatocellular carcinoma (HCC), and hence might be a suitable therapeutic option, to define novel molecular targets for precision medicine in CLD.


Subject(s)
Gene Transfer Techniques , Hepatocytes/enzymology , Lipids/chemistry , Liver Cirrhosis/therapy , Liver Neoplasms/prevention & control , Liver/enzymology , Mitogen-Activated Protein Kinase 9/metabolism , RNA, Small Interfering/metabolism , RNAi Therapeutics , Animals , Apoptosis , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Hepatocytes/pathology , Intracellular Signaling Peptides and Proteins/genetics , Liver/pathology , Liver Cirrhosis/enzymology , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Nanoparticles , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics
2.
Am J Physiol Heart Circ Physiol ; 317(2): H364-H374, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31149833

ABSTRACT

Reduced vasodilator properties of insulin in obesity are caused by changes in perivascular adipose tissue and contribute to microvascular dysfunction in skeletal muscle. The causes of this dysfunction are unknown. The effects of a short-term Western diet on JNK2-expressing cells in perivascular adipose tissue (PVAT) on insulin-induced vasodilation and perfusion of skeletal muscle were assessed. In vivo, 2 wk of Western diet (WD) reduced whole body insulin sensitivity and insulin-stimulated muscle perfusion, determined using contrast ultrasonography during the hyperinsulinemic clamp. Ex vivo, WD triggered accumulation of PVAT in skeletal muscle and blunted its ability to facilitate insulin-induced vasodilation. Labeling of myeloid cells with green fluorescent protein identified bone marrow as a source of PVAT in muscle. To study whether JNK2-expressing inflammatory cells from bone marrow were involved, we transplanted JNK2-/- bone marrow to WT mice. Deletion of JNK2 in bone marrow rescued the vasodilator phenotype of PVAT during WD exposure. JNK2 deletion in myeloid cells prevented the WD-induced increase in F4/80 expression. Even though WD and JNK2 deletion resulted in specific changes in gene expression of PVAT; epididymal and subcutaneous adipose tissue; expression of tumor necrosis factor-α, interleukin-1ß, interleukin-6, or protein inhibitor of STAT1 was not affected. In conclusion, short-term Western diet triggers infiltration of JNK2-positive myeloid cells into PVAT, resulting in PVAT dysfunction, nonclassical inflammation, and loss of insulin-induced vasodilatation in vivo and ex vivo.NEW & NOTEWORTHY We demonstrate that in the earliest phase of weight gain, changes in perivascular adipose tissue in muscle impair insulin-stimulated muscle perfusion. The hallmark of these changes is infiltration by inflammatory cells. Deletion of JNK2 from the bone marrow restores the function of perivascular adipose tissue to enhance insulin's vasodilator effects in muscle, showing that the bone marrow contributes to regulation of muscle perfusion.


Subject(s)
Adipose Tissue/drug effects , Insulin Resistance , Insulin/pharmacology , Microvessels/drug effects , Mitogen-Activated Protein Kinase 9/metabolism , Muscle, Skeletal/blood supply , Myeloid Cells/enzymology , Obesity/enzymology , Vasodilation/drug effects , Adipose Tissue/metabolism , Adipose Tissue/physiopathology , Animals , Bone Marrow Transplantation , Diet, High-Fat , Disease Models, Animal , Male , Mice, Inbred C57BL , Mice, Knockout , Microvessels/physiopathology , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Obesity/etiology , Obesity/physiopathology , Regional Blood Flow , Time Factors , Weight Gain
3.
J Pathol ; 247(1): 110-122, 2019 01.
Article in English | MEDLINE | ID: mdl-30264435

ABSTRACT

Ibuprofen is a worldwide used non-steroidal anti-inflammatory drug which may cause acute liver injury (ALI) requiring liver transplantation. We aimed to unveil the molecular pathways involved in triggering ibuprofen-induced ALI, which, at present, remain elusive. First, we investigated activation of essential pathways in human liver sections of ibuprofen-induced ALI. Next, we assessed the cytotoxicity of ibuprofen in vitro and developed a novel murine model of ibuprofen intoxication. To assess the role of JNK, we used animals carrying constitutive deletion of c-Jun N-terminal kinase 1 (Jnk1-/- ) or Jnk2 (Jnk2-/- ) expression and included investigations using animals with hepatocyte-specific Jnk deletion either genetically (Jnk1Δhepa ) or by siRNA (siJnk2Δhepa ). We found in human and murine samples of ibuprofen-induced acute liver failure that JNK phosphorylation was increased in the cytoplasm of hepatocytes and other non-liver parenchymal cells (non-LPCs) compared with healthy tissue. In mice, ibuprofen intoxication resulted in a significantly stronger degree of liver injury compared with vehicle-treated controls as evidenced by serum transaminases, and hepatic histopathology. Next, we investigated molecular pathways. PKCα, AKT, JNK and RIPK1 were significantly increased 8 h after ibuprofen intoxication. Constitutive Jnk1-/- and Jnk2-/- deficient mice exhibited increased liver dysfunction compared to wild-type (WT) animals. Furthermore, siJnk2Δhepa animals showed a dramatic increase in biochemical markers of liver function, which correlated with significantly higher serum liver enzymes and worsened liver histology, and MAPK activation compared to Jnk1Δhepa or WT animals. In our study, cytoplasmic JNK activation in hepatocytes and other non-LPCs is a hallmark of human and murine ibuprofen-induced ALI. Functional in vivo analysis demonstrated a protective role of hepatocyte-specific Jnk2 during ibuprofen ALI. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Hepatocytes/enzymology , Ibuprofen , Liver Failure, Acute/prevention & control , Liver/enzymology , Mitogen-Activated Protein Kinase 9/metabolism , Animals , Cell Death , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Enzyme Activation , Hepatocytes/pathology , Humans , Liver/pathology , Liver Failure, Acute/enzymology , Liver Failure, Acute/genetics , Liver Failure, Acute/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Phosphorylation , Signal Transduction
4.
Int J Biol Macromol ; 120(Pt B): 2215-2224, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30003914

ABSTRACT

c-Jun N-terminal kinase 2 (JNK2) is primarily responsible for the oncogenic transformation of the transcription factor c-Jun. Expression of the proto-oncogene c-Jun progresses the cell cycle from G1 to S phase, but when its expression becomes awry it leads to uncontrolled proliferation and angiogenesis. Delivering a JNK2 siRNA (siJNK2) in tumor tissue was anticipated to reverse the condition with subsequent onset of apoptosis which predominantly requires an efficient delivering system capable of penetrating through the compact tumor mass. In the present study, it was demonstrated that polymannitol-based vector (PMGT) with inherent hyperosmotic properties was able to penetrate through and deliver the siJNK2 in the subcutaneous tumor of xenograft mice. Hyperosmotic activity of polymannitol was shown to account for the enhanced therapeutic delivery both in vitro and in vivo because of the induction of cyclooxygenase-2 (COX-2) which stimulates caveolin-1 for caveolae-mediated endocytosis of the polyplexes. Further suppression of JNK2 and hence c-Jun expression led to the activation of caspase-9 to induce apoptosis and inhibition of tumor growth in xenograft mice model. The study exemplifies PMGT as an efficient vector for delivering therapeutic molecules in compact tumor tissue and suppression of JNK2 introduces a strategy to inhibit tumor progression.


Subject(s)
Caspase 9/metabolism , Disease Progression , Gene Silencing , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Osmosis , Polymers/chemistry , A549 Cells , Animals , Apoptosis/genetics , Cell Transformation, Neoplastic , Cyclooxygenase 2/biosynthesis , Drug Carriers/chemistry , Drug Carriers/metabolism , Endocytosis/genetics , Enzyme Activation/drug effects , Enzyme Induction/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Humans , Mannitol/chemistry , Mice , Mice, Inbred BALB C , Polymers/metabolism , Proto-Oncogene Mas , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , Tumor Burden/genetics
5.
Cell Death Dis ; 9(6): 705, 2018 06 13.
Article in English | MEDLINE | ID: mdl-29899326

ABSTRACT

The cJun N-terminal kinases (JNKs; JNK1, JNK2, and JNK3) promote degenerative processes after neuronal injury and in disease. JNK2 and JNK3 have been shown to promote retinal ganglion cell (RGC) death after optic nerve injury. In their absence, long-term survival of RGC somas is significantly increased after mechanical optic nerve injury. In glaucoma, because optic nerve damage is thought to be a major cause of RGC death, JNKs are an important potential target for therapeutic intervention. To assess the role of JNK2 and JNK3 in an ocular hypertensive model of glaucoma, null alleles of Jnk2 and Jnk3 were backcrossed into the DBA/2J (D2) mouse. JNK activation occurred in RGCs following increased intraocular pressure in D2 mice. However, deficiency of both Jnk2 and Jnk3 together did not lessen optic nerve damage or RGC death. These results differentiate the molecular pathways controlling cell death in ocular hypertensive glaucoma compared with mechanical optic nerve injury. It is further shown that JUN, a pro-death component of the JNK pathway in RGCs, can be activated in glaucoma in the absence of JNK2 and JNK3. This implicates JNK1 in glaucomatous RGC death. Unexpectedly, at younger ages, Jnk2-deficient mice were more likely to develop features of glaucomatous neurodegeneration than D2 mice expressing Jnk2. This appears to be due to a neuroprotective effect of JNK2 and not due to a change in intraocular pressure. The Jnk2-deficient context also unmasked a lesser role for Jnk3 in glaucoma. Jnk2 and Jnk3 double knockout mice had a modestly increased risk of neurodegeneration compared with mice only deficient in Jnk2. Overall, these findings are consistent with pleiotropic effects of JNK isoforms in glaucoma and suggest caution is warranted when using JNK inhibitors to treat chronic neurodegenerative conditions.


Subject(s)
Glaucoma/enzymology , Glaucoma/pathology , Mitogen-Activated Protein Kinase 9/deficiency , Nerve Degeneration/enzymology , Nerve Degeneration/pathology , Ocular Hypertension/enzymology , Ocular Hypertension/pathology , Animals , Axons/metabolism , Cell Death , Enzyme Activation , Gene Expression Regulation , Glaucoma/physiopathology , Intraocular Pressure , Mice, Inbred DBA , Mitogen-Activated Protein Kinase 10/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Nerve Degeneration/physiopathology , Ocular Hypertension/physiopathology , Optic Nerve/enzymology , Optic Nerve/pathology , Optic Nerve/physiopathology , Retina/enzymology , Retina/pathology , Retina/physiopathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology
6.
Thromb Haemost ; 117(1): 83-89, 2017 01 05.
Article in English | MEDLINE | ID: mdl-27761579

ABSTRACT

Despite public awareness of its deleterious effects, smoking remains a major cause of death. Indeed, it is a risk factor for atherothrombotic complications and in line with this, the introduction of smoking ban in public areas reduced smoking-associated cardiovascular complications. Nonetheless, smoking remains a major concern, and molecular mechanisms by which it causes cardiovascular disease are not known. Peripheral blood monocytes from healthy smokers displayed increased JNK2 and tissue factor (TF) gene expression compared to non-smokers (n=15, p<0.05). Similarly, human aortic endothelial cells exposed to cigarette smoke total particulate matter (CS-TPM) revealed increased TF expression mediated by JNK2 (n=4; p<0.05). Wild-type and JNK2-/- mice were exposed to cigarette smoke for two weeks after which arterial thrombosis was investigated. Wild-type mice exposed to smoke displayed reduced time to thrombotic arterial occlusion (n=8; p<0.05) and increased tissue factor activity (n=7; p<0.05) as compared to wild-type controls (n=6), while JNK2-/-mice exposed to smoke maintained an unaltered thrombotic potential (n=8; p=NS) and tissue factor activity (n=8) comparable to that of JNK2-/- and wild-type controls (n=6; p=NS). Smoking caused an increased production of reactive oxygen species (ROS) in wild-type but not in JNK2-/- mice (n=7; p<0.05 for wild-type mice and n=5-6; p=NS for JNK2-/- mice). In conclusion, the MAP kinase JNK2 mediates cigarette smoke-induced TF activation, arterial thrombosis and ROS production. These results underscore a major role of JNK2 in smoke-mediated thrombus formation and may offer an attractive target to prevent smoke-related thrombosis in those subjects which do not manage quitting.


Subject(s)
Arterial Occlusive Diseases/etiology , Blood Coagulation , Mitogen-Activated Protein Kinase 9/metabolism , Smoke/adverse effects , Smoking/adverse effects , Thrombosis/etiology , Animals , Arterial Occlusive Diseases/blood , Arterial Occlusive Diseases/enzymology , Arterial Occlusive Diseases/genetics , Carotid Artery Injuries/blood , Carotid Artery Injuries/enzymology , Carotid Artery Injuries/genetics , Cells, Cultured , Endothelial Cells/enzymology , Female , Genetic Predisposition to Disease , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Phenotype , Reactive Oxygen Species/metabolism , Signal Transduction , Smoking/blood , Smoking/genetics , Thromboplastin/genetics , Thromboplastin/metabolism , Thrombosis/blood , Thrombosis/enzymology , Thrombosis/genetics
7.
Arterioscler Thromb Vasc Biol ; 36(6): 1122-31, 2016 06.
Article in English | MEDLINE | ID: mdl-27102962

ABSTRACT

OBJECTIVE: The c-Jun NH2-terminal kinases (JNK) are regulated by a wide variety of cellular stresses and have been implicated in apoptotic signaling. Macrophages express 2 JNK isoforms, JNK1 and JNK2, which may have different effects on cell survival and atherosclerosis. APPROACH AND RESULTS: To dissect the effect of macrophage JNK1 and JNK2 on early atherosclerosis, Ldlr(-/-) mice were reconstituted with wild-type, Jnk1(-/-), and Jnk2(-/-) hematopoietic cells and fed a high cholesterol diet. Jnk1(-/-)→Ldlr(-/-) mice have larger atherosclerotic lesions with more macrophages and fewer apoptotic cells than mice transplanted with wild-type or Jnk2(-/-) cells. Moreover, genetic ablation of JNK to a single allele (Jnk1(+/-)/Jnk2(-/-) or Jnk1(-/-)/Jnk2(+/-)) in marrow of Ldlr(-/-) recipients further increased atherosclerosis compared with Jnk1(-/-)→Ldlr(-/-) and wild-type→Ldlr(-/-) mice. In mouse macrophages, anisomycin-mediated JNK signaling antagonized Akt activity, and loss of Jnk1 gene obliterated this effect. Similarly, pharmacological inhibition of JNK1, but not JNK2, markedly reduced the antagonizing effect of JNK on Akt activity. Prolonged JNK signaling in the setting of endoplasmic reticulum stress gradually extinguished Akt and Bad activity in wild-type cells with markedly less effects in Jnk1(-/-) macrophages, which were also more resistant to apoptosis. Consequently, anisomycin increased and JNK1 inhibitors suppressed endoplasmic reticulum stress-mediated apoptosis in macrophages. We also found that genetic and pharmacological inhibition of phosphatase and tensin homolog abolished the JNK-mediated effects on Akt activity, indicating that phosphatase and tensin homolog mediates crosstalk between these pathways. CONCLUSIONS: Loss of Jnk1, but not Jnk2, in macrophages protects them from apoptosis, increasing cell survival, and this accelerates early atherosclerosis.


Subject(s)
Aorta/enzymology , Aortic Diseases/enzymology , Apoptosis , Atherosclerosis/enzymology , Bone Marrow Cells/enzymology , Macrophages/enzymology , Mitogen-Activated Protein Kinase 8/deficiency , Receptors, LDL/deficiency , Animals , Aorta/drug effects , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Apoptosis/drug effects , Atherosclerosis/genetics , Atherosclerosis/pathology , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Bone Marrow Transplantation , Cell Survival , Cells, Cultured , Diet, High-Fat , Disease Models, Animal , Endoplasmic Reticulum Stress , Genetic Predisposition to Disease , Hypercholesterolemia/enzymology , Hypercholesterolemia/genetics , Macrophages/drug effects , Macrophages/pathology , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/antagonists & inhibitors , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/metabolism , Phenotype , Plaque, Atherosclerotic , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, LDL/genetics , Signal Transduction , bcl-Associated Death Protein/metabolism
8.
Gastroenterology ; 150(4): 968-81, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26708719

ABSTRACT

BACKGROUND & AIMS: c-Jun N-terminal kinase (JNK) 1 and JNK2 are expressed in hepatocytes and have overlapping and distinct functions. JNK proteins are activated via phosphorylation in response to acetaminophen- or carbon tetrachloride (CCl4)-induced liver damage; the level of activation correlates with the degree of injury. SP600125, a JNK inhibitor, has been reported to block acetaminophen-induced liver injury. We investigated the role of JNK in drug-induced liver injury (DILI) in liver tissue from patients and in mice with genetic deletion of JNK in hepatocytes. METHODS: We studied liver sections from patients with DILI (due to acetaminophen, phenprocoumon, nonsteroidal anti-inflammatory drugs, or autoimmune hepatitis) or patients without acute liver failure (controls) collected from a DILI Biobank in Germany. Levels of total and activated (phosphorylated) JNK were measured by immunohistochemistry and Western blotting. Mice with hepatocyte-specific deletion of Jnk1 (Jnk1(Δhepa)) or combination of Jnk1 and Jnk2 (Jnk(Δhepa)), as well as Jnk1-floxed C57BL/6 (control) mice, were given injections of CCl4 (to induce fibrosis) or acetaminophen (to induce toxic liver injury). We performed gene expression microarray and phosphoproteomic analyses to determine mechanisms of JNK activity in hepatocytes. RESULTS: Liver samples from DILI patients contained more activated JNK, predominantly in nuclei of hepatocytes and in immune cells, than healthy tissue. Administration of acetaminophen to Jnk(Δhepa) mice produced a greater level of liver injury than that observed in Jnk1(Δhepa) or control mice, based on levels of serum markers and microscopic and histologic analysis of liver tissues. Administration of CCl4 also induced stronger hepatic injury in Jnk(Δhepa) mice, based on increased inflammation, cell proliferation, and fibrosis progression, compared with Jnk1(Δhepa) or control mice. Hepatocytes from Jnk(Δhepa) mice given acetaminophen had an increased oxidative stress response, leading to decreased activation of adenosine monophosphate-activated protein kinase, total protein adenosine monophosphate-activated protein kinase levels, and pJunD and subsequent necrosis. Administration of SP600125 before or with acetaminophen protected Jnk(Δhepa) and control mice from liver injury. CONCLUSIONS: In hepatocytes, JNK1 and JNK2 appear to have combined effects in protecting mice from CCl4- and acetaminophen-induced liver injury. It is important to study the tissue-specific functions of both proteins, rather than just JNK1, in the onset of toxic liver injury. JNK inhibition with SP600125 shows off-target effects.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Hepatocytes/enzymology , Liver Failure, Acute/prevention & control , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , AMP-Activated Protein Kinases/metabolism , Acetaminophen , Animals , Carbon Tetrachloride , Case-Control Studies , Cell Death , Cell Proliferation , Cells, Cultured , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Enzyme Activation , Female , Gene Expression Profiling , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Liver Failure, Acute/chemically induced , Liver Failure, Acute/enzymology , Liver Failure, Acute/genetics , Liver Failure, Acute/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/deficiency , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/antagonists & inhibitors , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Oxidative Stress , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Time Factors , Young Adult
9.
PLoS One ; 9(10): e109750, 2014.
Article in English | MEDLINE | ID: mdl-25285524

ABSTRACT

Stathmin 1 performs a critical function in cell proliferation by regulating microtubule polymerization. This proliferative function is thought to explain the frequent overexpression of stathmin in human cancer and its correlation with a bad prognosis. Whether stathmin also functions in cell death pathways is unclear. Stathmin regulates microtubules in part by binding free tubulin, a process inhibited by stathmin phosphorylation from kinases including c-Jun N-terminal kinase (JNK). The involvement of JNK activation both in stathmin phosphorylation, and in hepatocellular resistance to oxidative stress, led to an examination of the role of stathmin/JNK crosstalk in oxidant-induced hepatocyte death. Oxidative stress from menadione-generated superoxide induced JNK-dependent stathmin phosphorylation at Ser-16, Ser-25 and Ser-38 in hepatocytes. A stathmin knockdown sensitized hepatocytes to both apoptotic and necrotic cell death from menadione without altering levels of oxidant generation. The absence of stathmin during oxidative stress led to JNK overactivation that was the mechanism of cell death as a concomitant knockdown of JNK1 or JNK2 blocked death. Hepatocyte death from JNK overactivation was mediated by the effects of JNK on mitochondria. Mitochondrial outer membrane permeabilization occurred in stathmin knockdown cells at low concentrations of menadione that triggered apoptosis, whereas mitochondrial ß-oxidation and ATP homeostasis were compromised at higher, necrotic menadione concentrations. Stathmin therefore mediates hepatocyte resistance to death from oxidative stress by down regulating JNK and maintaining mitochondrial integrity. These findings demonstrate a new mechanism by which stathmin promotes cell survival and potentially tumor growth.


Subject(s)
Down-Regulation , Hepatocytes/cytology , Hepatocytes/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Oxidative Stress , Stathmin/metabolism , Animals , Apoptosis/drug effects , Cell Line , Down-Regulation/drug effects , Gene Knockdown Techniques , HEK293 Cells , Hepatocytes/drug effects , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Mitogen-Activated Protein Kinase 8/deficiency , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Necrosis/chemically induced , Oxidative Stress/drug effects , Rats , Signal Transduction/drug effects , Stathmin/deficiency , Stathmin/genetics , Vitamin K 3/pharmacology
10.
J Hepatol ; 61(6): 1376-84, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25064435

ABSTRACT

BACKGROUND & AIMS: Free cholesterol (FC) accumulates in non-alcoholic steatohepatitis (NASH) but not in simple steatosis. We sought to establish how FC causes hepatocyte injury. METHODS: In NASH-affected livers from diabetic mice, subcellular FC distribution (filipin fluorescence) was established by subcellular marker co-localization. We loaded murine hepatocytes with FC by incubation with low-density lipoprotein (LDL) and studied the effects of FC on JNK1 activation, mitochondrial injury and cell death and on the amplifying roles of the high-mobility-group-box 1 (HMGB1) protein and the Toll-like receptor 4 (TLR4). RESULTS: In NASH, FC localized to hepatocyte plasma membrane, mitochondria and ER. This was reproduced in FC-loaded hepatocytes. At 40 µM LDL, hepatocyte FC increased to cause LDH leakage, apoptosis and necrosis associated with JNK1 activation (c-Jun phosphorylation), mitochondrial membrane pore transition, cytochrome c release, oxidative stress (GSSG:GSH ratio) and ATP depletion. Mitochondrial swelling and crystae disarray were evident by electron microscopy. Jnk1(-/-) and Tlr4(-/-) hepatocytes were refractory to FC lipotoxicity; JNK inhibitors (1-2 µM CC-401, CC-930) blocked apoptosis and necrosis. Cyclosporine A and caspase-3 inhibitors protected FC-loaded hepatocytes, confirming mitochondrial cell death pathways; in contrast, 4-phenylbutyric acid, which improves ER folding capacity did not protect FC-loaded hepatocytes. HMGB1 was released into the culture medium of FC-loaded wild type (WT) but not Jnk1(-/-) or Tlr4(-/-) hepatocytes, while anti-HMGB1 anti-serum prevented JNK activation and FC lipotoxicity in WT hepatocytes. CONCLUSIONS: These novel findings show that mitochondrial FC deposition causes hepatocyte apoptosis and necrosis by activating JNK1; inhibition of which could be a novel therapeutic approach in NASH. Further, there is a tight link between JNK1-dependent HMGB1 secretion from lipotoxic hepatocytes and a paracrine cytolytic effect on neighbouring cholesterol-loaded hepatocytes operating via TLR4.


Subject(s)
Cholesterol/metabolism , HMGB1 Protein/metabolism , Hepatocytes/metabolism , Mitochondria, Liver/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Toll-Like Receptor 4/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis/physiology , Cells, Cultured , Cytochromes c/metabolism , Disease Models, Animal , Female , Hepatocytes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mitogen-Activated Protein Kinase 8/deficiency , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Mitogen-Activated Protein Kinase 9/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/physiopathology , Oxidative Stress/physiology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
11.
Cell Mol Life Sci ; 71(1): 143-63, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23784308

ABSTRACT

Glucocorticoids (GCs) block inflammation via interference of the liganded glucocorticoid receptor (GR) with the activity of pro-inflammatory transcription factors NF-κB and AP-1, a mechanism known as transrepression. This mechanism is believed to involve the activity of GR monomers. Here, we explored how the GR monomer-favoring Compound A (CpdA) affects AP-1 activation and activity. Our results demonstrate that non-steroidal CpdA, unlike classic steroidal GCs, blocks NF-κB- but not AP-1-driven gene expression. CpdA rather sustains AP-1-driven gene expression, a result which could mechanistically be explained by the failure of CpdA to block upstream JNK kinase activation and concomitantly also phosphorylation of c-Jun. In concordance and in contrast to DEX, CpdA maintained the expression of the activated AP-1 target gene c-jun, as well as the production of the c-Jun protein. As for the underlying mechanism, GR is a necessary intermediate in the CpdA-mediated gene expression of AP-1-regulated genes, but seems to be superfluous to CpdA-mediated JNK phosphorylation prolongation. The latter phenomenon concurs with the inability of CpdA to stimulate DUSP1 gene expression. ChIP analysis demonstrates that DEX-activated GR, but not CpdA-activated GR, is recruited to AP-1-driven promoters. Furthermore, in mice we observed that CpdA instigates a strong enhancement of TNF-induced AP-1-driven gene expression. Finally, we demonstrate that this phenomenon coincides with an increased sensitivity towards TNF lethality, and implicate again a role for JNK2. In conclusion, our data support the hypothesis that a ligand-induced differential conformation of GR yields a different transcription factor cross-talk profile.


Subject(s)
NF-kappa B/metabolism , Receptors, Glucocorticoid/metabolism , Transcription Factor AP-1/metabolism , Animals , Cell Line, Tumor , Dexamethasone/pharmacology , Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 1/metabolism , Gene Expression/drug effects , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Mitogen-Activated Protein Kinase 9/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/genetics , Staurosporine/pharmacology , Transcription Factor AP-1/genetics , Transcriptional Activation/drug effects , Triazoles/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
12.
Science ; 339(6116): 218-22, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-23223452

ABSTRACT

The cJun NH(2)-terminal kinase (JNK) signaling pathway contributes to inflammation and plays a key role in the metabolic response to obesity, including insulin resistance. Macrophages are implicated in this process. To test the role of JNK, we established mice with selective JNK deficiency in macrophages. We report that feeding a high-fat diet to control and JNK-deficient mice caused similar obesity, but only mice with JNK-deficient macrophages remained insulin-sensitive. The protection of mice with macrophage-specific JNK deficiency against insulin resistance was associated with reduced tissue infiltration by macrophages. Immunophenotyping demonstrated that JNK was required for pro-inflammatory macrophage polarization. These studies demonstrate that JNK in macrophages is required for the establishment of obesity-induced insulin resistance and inflammation.


Subject(s)
Inflammation/physiopathology , Insulin Resistance , Macrophages/enzymology , Macrophages/immunology , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Obesity/physiopathology , Adipose Tissue/immunology , Adipose Tissue/pathology , Animals , Diet, High-Fat , Glucose Clamp Technique , Immunophenotyping , Inflammation/immunology , Islets of Langerhans/pathology , MAP Kinase Signaling System , Macrophage Activation , Macrophages/physiology , Mice , Mitogen-Activated Protein Kinase 8/deficiency , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Obesity/immunology
13.
Free Radic Biol Med ; 53(5): 1170-80, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22749809

ABSTRACT

Binge alcohol drinking induces hepatic steatosis. Recent studies showed that chronic ethanol-induced fatty liver was, at least in part, CYP2E1 dependent. The mechanism of acute alcohol-induced steatosis and whether CYP2E1 plays any role are still unclear. Increasing oxidative stress by alcohol can activate the JNK MAP kinase signaling pathway, suggesting that JNK might be a target for prevention of alcohol-induced steatosis. We used CYP2E1 knockout (KO) mice, a JNK inhibitor, and JNK1 or JNK2 knockout mice to test the role of CYP2E1, JNK, and the individual role of JNK1 and JNK2 in acute alcohol-induced steatosis. In wild-type (WT) mice, acute alcohol activates CYP2E1 and increases oxidative stress, which reciprocally increases activation of the JNK signaling pathway. Acute alcohol-induced fatty liver and oxidative stress were blunted in CYP2E1 KO mice and by the JNK inhibitor in WT mice. The antioxidant N-acetylcysteine decreased the acute alcohol-induced oxidative stress, the activation of JNK, and the steatosis but not the activation of CYP2E1. Acute alcohol decreased autophagy and increased expression of SREBP, effects blocked by the JNK inhibitor. Acute alcohol-induced fatty liver was the same in JNK1 and JNK2 KO mice as in WT mice; thus either JNK1 or JNK2 per se is sufficient for induction of steatosis by acute alcohol. The results show that acute alcohol elevation of CYP2E1, oxidative stress, and activation of JNK interact to lower autophagy and increase lipogenic SREBP resulting in fatty liver.


Subject(s)
Autophagy , Cytochrome P-450 CYP2E1/metabolism , Fatty Liver, Alcoholic/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Oxidative Stress , Animals , Cytochrome P-450 CYP2E1/deficiency , Ethanol/administration & dosage , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 8/deficiency , Mitogen-Activated Protein Kinase 9/deficiency
14.
Am J Physiol Gastrointest Liver Physiol ; 303(4): G498-506, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22700824

ABSTRACT

Chronic liver disease promotes hepatocellular injury involving apoptosis and triggers compensatory regeneration that leads to the activation of quiescent stellate cells in the liver. The deposition of extracellular matrix from activated myofibroblasts promotes hepatic fibrosis and the progression to cirrhosis with deleterious effects on liver physiology. The role of apoptosis signaling pathways in the development of fibrosis remains undefined. The aim of the current study was to determine the involvement of the caspase-8 homologue cellular FLICE-inhibitory protein (cFLIP) during the initiation and progression of fibrosis. Liver injury and fibrosis from carbon tetrachloride (CCl(4)) and thioacetamide (TAA) were examined in mice exhibiting a hepatocyte-specific deletion of cFLIP (flip(-/-)). Acute liver injury from CCl(4) and TAA were enhanced in flip(-/-) mice. This was accompanied by increased activation of caspase-3 and -9, pronounced phosphorylation of JNK, and decreased phosphorylation of Erk. Deletion of the cJun NH(2)-terminal kinase 2 (JNK2) in flip(-/-) mice protected from injury. Hepatic fibrosis was increased at baseline in 12-wk-old flip(-/-) mice, and progression of fibrosis from TAA was accelerated compared with the wild type. In conclusion, deletion of cFLIP in hepatocytes leads to increased fibrosis and accelerated fibrosis progression. This is accompanied by increased injury involving the activation of caspases and JNK2. Thus predisposition to liver injury involving increased hepatocellular apoptosis is a critical mediator of accelerated fibrogenesis, and prevention of liver injury will be a most important measure for patients with chronic liver disease.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/deficiency , Chemical and Drug Induced Liver Injury/etiology , Hepatocytes/enzymology , Liver Cirrhosis/etiology , Liver/enzymology , Mitogen-Activated Protein Kinase 9/metabolism , Animals , Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Carbon Tetrachloride , Caspase 3/metabolism , Caspase 9/metabolism , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Disease Progression , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Genotype , Hepatocytes/pathology , Liver/pathology , Liver Cirrhosis/enzymology , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Phenotype , Phosphorylation , Signal Transduction , Thioacetamide , Time Factors
15.
Diabetes ; 61(8): 2084-92, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22688338

ABSTRACT

Oxidative stress and apoptosis are implicated in the pathogenesis of diabetic embryopathy. The proapoptotic c-Jun NH(2)-terminal kinases (JNK)1/2 activation is associated with diabetic embryopathy. We sought to determine whether 1) hyperglycemia-induced oxidative stress is responsible for the activation of JNK1/2 signaling, 2) JNK1 contributes to the teratogenicity of hyperglycemia, and 3) both JNK1 and JNK2 activation cause activation of downstream transcription factors, caspase activation, and apoptosis, resulting in neural tube defects (NTDs). Wild-type (WT) embryos from nondiabetic WT dams and WT, superoxide dismutase (SOD)1-overexpressing, jnk1(+/-), jnk1(-/-), and jnk2(-/-) embryos exposed to maternal hyperglycemia were used to assess JNK1/2 activation, NTDs, activation of transcription factors downstream of JNK1/2, caspase cascade, and apoptosis. SOD1 overexpression abolished diabetes-induced activation of JNK1/2 and their downstream effectors: phosphorylation of c-Jun, activating transcription factor 2, and E twenty-six-like transcription factor 1 and dephosphorylation of forkhead box class O3a. jnk1(-/-) embryos had significantly lower incidences of NTDs than those of WT or jnk1(+/-) embryos. Either jnk1 or jnk2 gene deletion blocked diabetes-induced activation of JNK1/2 signaling, caspases 3 and 8, and apoptosis in Sox1(+) neural progenitors of the developing neural tube. Our results show that JNK1 and JNK2 are equally involved in diabetic embryopathy and that the oxidative stress-JNK1/2-caspase pathway mediates the proapoptotic signals and the teratogenicity of maternal diabetes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/physiology , Diabetes Mellitus/physiopathology , Fetal Diseases/etiology , MAP Kinase Kinase 7/metabolism , MAP Kinase Signaling System/physiology , Nerve Tissue Proteins/metabolism , Oxidative Stress/physiology , Animals , Caspases/metabolism , Enzyme Activation , Female , Gene Deletion , Hyperglycemia/physiopathology , Mice , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/deficiency , Neural Tube Defects/etiology , Pregnancy , Pregnancy Complications/etiology , Superoxide Dismutase/biosynthesis , Superoxide Dismutase-1
16.
Biol Chem ; 393(6): 525-34, 2012 May.
Article in English | MEDLINE | ID: mdl-22628315

ABSTRACT

C-Jun N-terminal kinases (JNK) are activated in course of many viral infections. Here we analyzed the activity of JNK inhibitors on influenza A virus (IAV) amplification. Human lung epithelial cells were infected with either the highly pathogenic avian virus strain A/FPV/Bratislava/79 (H7N7) or the pandemic swine-origin influenza virus A/Hamburg/4/09 (H1N1v). The application of the JNK inhibitors SP600125 and AS601245 reduced IAV amplification by suppressing viral protein and RNA synthesis. Although AS601245 appeared to generally block the transcription of newly introduced genes, SP600125 specifically affected viral RNA synthesis. Overexpression of a dominant negative mutant of SEK/MKK4 and siRNA-mediated suppression of JNK2 expression confirmed that specific manipulation of the JNK pathway attenuates virus propagation. An IAV minigenome replication assay revealed that SP600125 did not directly affect the activity of the viral RNA polymerase complex but seems to suppress an anti-influenza nonstructural protein 1-mediated virus supportive function. Finally, when H7N7- or H1N1v-infected mice were treated with SP600125, the viral load is reduced in lungs of treated compared with untreated mice. Our data suggest that this class of ATP competitive inhibitors once optimized for antiviral action potentially represent novel drugs for antiviral intervention.


Subject(s)
Antiviral Agents/pharmacology , Influenza A virus/drug effects , Influenza A virus/pathogenicity , Influenza in Birds/virology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Pandemics , Protein Kinase Inhibitors/pharmacology , Animals , Antiviral Agents/chemistry , Birds , Cell Line , DNA-Directed RNA Polymerases/metabolism , Dogs , Enzyme Activation/drug effects , Gene Knockdown Techniques , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H7N7 Subtype/drug effects , Influenza A Virus, H7N7 Subtype/genetics , Influenza A Virus, H7N7 Subtype/pathogenicity , Influenza A Virus, H7N7 Subtype/physiology , Influenza A virus/genetics , Influenza A virus/physiology , JNK Mitogen-Activated Protein Kinases/metabolism , Lung/drug effects , Lung/virology , MAP Kinase Kinase 4/genetics , Mice , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Mutation , Protein Biosynthesis/drug effects , Protein Kinase Inhibitors/chemistry , RNA, Small Interfering/genetics , Transcription, Genetic/drug effects , Viral Load/drug effects , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects
17.
J Clin Invest ; 122(4): 1529-40, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22378044

ABSTRACT

Integrin signaling critically contributes to the progression, growth, and therapy resistance of malignant tumors. Here, we show that targeting of ß1 integrins with inhibitory antibodies enhances the sensitivity to ionizing radiation and delays the growth of human head and neck squamous cell carcinoma cell lines in 3D cell culture and in xenografted mice. Mechanistically, dephosphorylation of focal adhesion kinase (FAK) upon inhibition of ß1 integrin resulted in dissociation of a FAK/cortactin protein complex. This, in turn, downregulated JNK signaling and induced cell rounding, leading to radiosensitization. Thus, these findings suggest that robust and selective pharmacological targeting of ß1 integrins may provide therapeutic benefit to overcome tumor cell resistance to radiotherapy.


Subject(s)
Carcinoma, Squamous Cell/radiotherapy , Cortactin/physiology , Focal Adhesion Kinase 1/physiology , Head and Neck Neoplasms/radiotherapy , Integrin beta1/physiology , Neoplasm Proteins/physiology , Radiation Tolerance/physiology , Amino Acid Motifs , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cortactin/chemistry , Female , Focal Adhesion Kinase 1/chemistry , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Integrin beta1/immunology , Male , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 8/deficiency , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/physiology , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Mitogen-Activated Protein Kinase 9/physiology , Multiprotein Complexes , Neoplasm Proteins/chemistry , Neoplasm Transplantation , Phosphorylation , Protein Interaction Mapping , Protein Processing, Post-Translational , RNA, Small Interfering/pharmacology , Radiation-Sensitizing Agents/pharmacology , Signal Transduction , Tumor Cells, Cultured/radiation effects
18.
Cancer Res ; 72(2): 472-81, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22127926

ABSTRACT

cJun NH(2)-terminal kinase (JNK) signaling has been implicated in the developmental morphogenesis of epithelial organs. In this study, we employed a compound deletion of the murine Jnk1 and Jnk2 genes in the mammary gland to evaluate the requirement for these ubiquitously expressed genes in breast development and tumorigenesis. JNK1/2 was not required for breast epithelial cell proliferation or motility. However, JNK1/2 deficiency caused increased branching morphogenesis and defects in the clearance of lumenal epithelial cells. In the setting of breast cancer development, JNK1/2 deficiency significantly increased tumor formation. Together, these findings established that JNK signaling is required for normal mammary gland development and that it has a suppressive role in mammary tumorigenesis.


Subject(s)
MAP Kinase Signaling System/physiology , Mammary Glands, Animal/enzymology , Mammary Glands, Animal/growth & development , Mammary Neoplasms, Experimental/enzymology , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Animals , Cell Growth Processes/physiology , Cell Movement/physiology , Female , Gene Expression , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Mitogen-Activated Protein Kinase 8/deficiency , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics
19.
Am J Pathol ; 179(4): 1884-93, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21839715

ABSTRACT

The roles of the c-Jun N-terminal kinases (JNKs) in inflammatory arthritis have been investigated; however, the roles of each isotype (ie, JNK1 and JNK2) in rheumatoid arthritis and conclusions about whether inhibition of one or both is necessary for amelioration of disease are unclear. By using JNK1- or JNK2-deficient mice in the collagen-induced arthritis and the KRN T-cell receptor transgenic mouse on C57BL/6 nonobese diabetic (K/BxN) serum transfer arthritis models, we demonstrate that JNK1 deficiency results in protection from arthritis, as judged by clinical score and histological evaluation in both models of inflammatory arthritis. In contrast, abrogation of JNK2 exacerbates disease. In collagen-induced arthritis, the distinct roles of the JNK isotypes can, at least in part, be explained by altered regulation of CD86 expression in JNK1- or JNK2-deficient macrophages in response to microbial products, thereby affecting T-cell-mediated immunity. The protection from K/BxN serum-induced arthritis in Jnk1(-/-) mice can also be explained by inept macrophage function because adoptive transfer of wild-type macrophages to Jnk1(-/-) mice restored disease susceptibility. Thus, our results provide a possible explanation for the modest therapeutic effects of broad JNK inhibitors and suggest that future therapies should selectively target the JNK1 isoform.


Subject(s)
Arthritis, Experimental/enzymology , Arthritis, Experimental/pathology , Inflammation/enzymology , Inflammation/pathology , Mitogen-Activated Protein Kinase 8/metabolism , Animals , Arthritis, Experimental/complications , Arthritis, Experimental/immunology , B7-2 Antigen/metabolism , Collagen Type II/immunology , Disease Models, Animal , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Inflammation/complications , Joints/enzymology , Joints/immunology , Joints/pathology , Macrophages/enzymology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 8/deficiency , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/metabolism , Serum , Up-Regulation
20.
Hear Res ; 278(1-2): 77-85, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21554942

ABSTRACT

Jun N-terminal kinase (JNK) is a multifunctional protein kinase crucial for neuronal apoptosis as well as neurite growth. We have previously shown that JNK activity is correlated with spiral ganglion neuron (SGN) apoptosis following hair cell loss in rats (Alam et al., 2007) implying that JNK inhibition may have therapeutic potential to protect SGNs in deaf individuals. Here we investigated the role of JNK in neurite outgrowth from cultured neonatal rat and mouse SGNs. We show that JNK is required for initial growth of neurites and for continued extension of already established neurites. The effect of JNK inhibition on neurite growth is rapid and is also rapidly reversible after washout of the inhibitor. Using phosphoJNK immunoreactivity as an indicator, we show that JNK is activated in growth cones within 30 min after transfer to medium lacking neurotrophic stimuli (5 K medium) but activation in the nucleus and soma requires hours. By transfecting epitope-tagged JNK1, JNK2, or JNK3 isoforms into SGNs, we found that all are present in the nucleus and cytoplasm and that there is no preferential redistribution to the nucleus after transfer to 5 K medium. Cotransfection of dominant-negative (dn) JNK1 and JNK2 into SGNs reduced neurite growth, although transfection of dnJNK1 or dnJNK2 alone had no significant effect. SGNs cultured from JNK3(-/-) mice showed reduced neurite growth that was further reduced by transfection of dnJNK1 and dnJNK2. This indicates that all three JNK isoforms promote SGN neurite growth although there may be functional redundancy between JNK1 and JNK2.


Subject(s)
MAP Kinase Signaling System , Neurites/enzymology , Neurites/ultrastructure , Spiral Ganglion/enzymology , Spiral Ganglion/innervation , Animals , Cells, Cultured , Enzyme Activation , Kinetics , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 10/deficiency , Mitogen-Activated Protein Kinase 10/genetics , Mitogen-Activated Protein Kinase 10/metabolism , Mitogen-Activated Protein Kinase 8/deficiency , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Mitogen-Activated Protein Kinase 9/metabolism , Neurons/enzymology , Neurons/ultrastructure , Phosphorylation , Rats , Spiral Ganglion/ultrastructure , Subcellular Fractions/enzymology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...