Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 231
Filter
1.
Nat Immunol ; 16(5): 458-66, 2015 May.
Article in English | MEDLINE | ID: mdl-25799126

ABSTRACT

Mitophagy is essential for cellular homeostasis, but how mitophagy is regulated is largely unknown. Here we found that the kinase Jnk2 was required for stress-induced mitophagy. Jnk2 promoted ubiquitination and proteasomal degradation of the small mitochondrial form of the tumor suppressor ARF (smARF). Loss of Jnk2 led to the accumulation of smARF, which induced excessive autophagy that resulted in lysosomal degradation of the mitophagy adaptor p62 at steady state. Depletion of p62 prevented Jnk2-deficient cells from mounting mitophagy upon stress. Jnk2-deficient mice displayed defective mitophagy, which resulted in tissue damage under hypoxic stress, as well as hyperactivation of inflammasomes and increased mortality in sepsis. Our findings define a unique mechanism of maintaining immunological homeostasis that protects the host from tissue damage and mortality.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Hypoxia/immunology , Mitogen-Activated Protein Kinase 9/metabolism , Proteasome Endopeptidase Complex/metabolism , Sepsis/immunology , Animals , Cells, Cultured , DNA Damage/physiology , Female , Inflammasomes/metabolism , Lipopolysaccharides/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 9/genetics , Mitophagy/genetics , Proteolysis , Proto-Oncogene Proteins c-myc/metabolism , Sepsis/chemically induced , Ubiquitination
2.
Cell ; 136(2): 249-60, 2009 Jan 23.
Article in English | MEDLINE | ID: mdl-19167327

ABSTRACT

The c-Jun NH(2)-terminal kinase (JNK) signaling pathway has been implicated in the development of tumor necrosis factor (TNF)-dependent hepatitis. JNK may play a critical role in hepatocytes during TNF-stimulated cell death in vivo. To test this hypothesis, we examined the phenotype of mice with compound disruption of the Jnk1 and Jnk2 genes. Mice with loss of JNK1/2 expression in hepatocytes exhibited no defects in the development of hepatitis compared with control mice, whereas mice with loss of JNK1/2 in the hematopoietic compartment exhibited a profound defect in hepatitis that was associated with markedly reduced expression of TNF-alpha. These data indicate that JNK is required for TNF-alpha expression but not for TNF-alpha-stimulated death of hepatocytes. Indeed, TNF-alpha induced similar hepatic damage in both mice with hepatocyte-specific JNK1/2 deficiency and control mice. These observations confirm a role for JNK in the development of hepatitis but identify hematopoietic cells as the site of the essential function of JNK.


Subject(s)
Hepatitis/metabolism , Hepatocytes/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/genetics
3.
PLoS Genet ; 17(12): e1009711, 2021 12.
Article in English | MEDLINE | ID: mdl-34962918

ABSTRACT

Polycystic kidney disease is an inherited degenerative disease in which the uriniferous tubules are replaced by expanding fluid-filled cysts that ultimately destroy organ function. Autosomal dominant polycystic kidney disease (ADPKD) is the most common form, afflicting approximately 1 in 1,000 people. It primarily is caused by mutations in the transmembrane proteins polycystin-1 (Pkd1) and polycystin-2 (Pkd2). The most proximal effects of Pkd mutations leading to cyst formation are not known, but pro-proliferative signaling must be involved for the tubule epithelial cells to increase in number over time. The c-Jun N-terminal kinase (JNK) pathway promotes proliferation and is activated in acute and chronic kidney diseases. Using a mouse model of cystic kidney disease caused by Pkd2 loss, we observe JNK activation in cystic kidneys and observe increased nuclear phospho c-Jun in cystic epithelium. Genetic removal of Jnk1 and Jnk2 suppresses the nuclear accumulation of phospho c-Jun, reduces proliferation and reduces the severity of cystic disease. While Jnk1 and Jnk2 are thought to have largely overlapping functions, we find that Jnk1 loss is nearly as effective as the double loss of Jnk1 and Jnk2. Jnk pathway inhibitors are in development for neurodegeneration, cancer, and fibrotic diseases. Our work suggests that the JNK pathway should be explored as a therapeutic target for ADPKD.


Subject(s)
Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/genetics , Polycystic Kidney, Autosomal Dominant/genetics , TRPP Cation Channels/genetics , Cell Proliferation/genetics , Epithelial Cells/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Kidney/metabolism , Kidney/pathology , MAP Kinase Signaling System/genetics , Mutation/genetics , Polycystic Kidney, Autosomal Dominant/pathology , Signal Transduction/genetics
4.
J Virol ; 96(2): e0171521, 2022 01 26.
Article in English | MEDLINE | ID: mdl-34757837

ABSTRACT

Alternative splicing (AS) is a frequent posttranscriptional regulatory event occurring in response to various endogenous and exogenous stimuli in most eukaryotic organisms. However, little is known about the effects of insect-transmitted viruses on AS events in insect vectors. The present study used third-generation sequencing technology and RNA sequencing (RNA-Seq) to evaluate the AS response in the small brown planthopper Laodelphax striatellus to rice stripe virus (RSV). The full-length transcriptome of L. striatellus was obtained using single-molecule real-time sequencing technology (SMRT). Posttranscriptional regulatory events, including AS, alternative polyadenylation, and fusion transcripts, were analyzed. A total of 28,175 nonredundant transcript isoforms included 24,950 transcripts assigned to 8,500 annotated genes of L. striatellus, and 5,000 of these genes (58.8%) had AS events. RNA-Seq of the gut samples of insects infected by RSV for 8 d identified 3,458 differentially expressed transcripts (DETs); 2,185 of these DETs were transcribed from 1,568 genes that had AS events, indicating that 31.4% of alternatively spliced genes responded to RSV infection of the gut. One of the c-Jun N-terminal kinase (JNK) genes, JNK2, experienced exon skipping, resulting in three transcript isoforms. These three isoforms differentially responded to RSV infection during development and in various organs. Injection of double-stranded RNAs targeting all or two isoforms indicated that three or at least two JNK2 isoforms facilitated RSV accumulation in planthoppers. These results implied that AS events could participate in the regulation of complex relationships between viruses and insect vectors. IMPORTANCE Alternative splicing (AS) is a regulatory mechanism that occurs after gene transcription. AS events can enrich protein diversity to promote the reactions of the organisms to various endogenous and exogenous stimulations. It is not known how insect vectors exploit AS events to cope with transmitted viruses. The present study used third-generation sequencing technology to obtain the profile of AS events in the small brown planthopper Laodelphax striatellus, which is an efficient vector for rice stripe virus (RSV). The results indicated that 31.4% of alternatively spliced genes responded to RSV infection in the gut of planthoppers. One of the c-Jun N-terminal kinase (JNK) genes, JNK2, produced three transcript isoforms by AS. These three isoforms showed different responses to RSV infection, and at least two isoforms facilitated viral accumulation in planthoppers. These results implied that AS events could participate in the regulation of complex relationships between viruses and insect vectors.


Subject(s)
Alternative Splicing , Hemiptera/virology , Insect Vectors/virology , Tenuivirus/physiology , Animals , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/virology , Gene Fusion , Hemiptera/genetics , Insect Proteins/genetics , Insect Vectors/genetics , Mitogen-Activated Protein Kinase 9/genetics , Oryza/virology , Plant Diseases/virology , Polyadenylation , Protein Isoforms , Transcriptome/genetics
5.
FASEB J ; 36(4): e22243, 2022 04.
Article in English | MEDLINE | ID: mdl-35224782

ABSTRACT

Osteoarthritis (OA) is a prevalent degenerative disease of the joint, featured by articular cartilage destruction and subchondral bone marrow lesions. Articular cartilage and subchondral bone constitute an osteochondral unit that guarantees joint homeostasis. During OA initiation, activated osteoclasts in subchondral bone ultimately result in impaired capacities of the subchondral bone in response to mechanical stress, followed by the degradation of overlying articular cartilage. Thus, targeting osteoclasts could be a potential therapeutic option for treating OA. Here, we observed that farnesoid X receptor (FXR) expression and osteoclast fusion and activity in subchondral bone were concomitantly changed during early-stage OA in the OA mouse model established by anterior cruciate ligament transection (ACLT). Then, we explored the therapeutic effects of FXR agonist GW4064 on the osteochondral pathologies in ACLT mice. We showed that GW4064 obviously ameliorated subchondral bone deterioration, associated with reduction in tartrate-resistant acid phosphatase (TRAP) positive multinuclear osteoclast number, as well as articular cartilage degradation, which were blocked by the treatment with FXR antagonist Guggulsterone. Mechanistically, GW4064 impeded osteoclastogenesis through inhibiting subchondral bone osteoclast fusion via suppressing c-Jun N-terminal kinase (JNK) 1/2/nuclear factor of activated T-cells 1 (NFATc1) pathway. Taken together, our results present evidence for the protective effects of GW4064 against OA by blunting osteoclast-mediated aberrant subchondral bone loss and subsequent cartilage deterioration. Therefore, GW4064 demonstrates the potential as an alternative therapeutic option against OA for further drug development.


Subject(s)
Bone Resorption/prevention & control , Gene Expression Regulation/drug effects , Isoxazoles/pharmacology , Osteoarthritis/prevention & control , Osteoclasts/drug effects , Osteogenesis , RNA-Binding Proteins/agonists , Animals , Bone Remodeling , Bone Resorption/etiology , Bone Resorption/metabolism , Bone Resorption/pathology , Female , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/genetics , Mitogen-Activated Protein Kinase 9/metabolism , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Osteoarthritis/etiology , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoclasts/metabolism , Osteoclasts/pathology
6.
Proc Natl Acad Sci U S A ; 117(28): 16492-16499, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32601222

ABSTRACT

Metabolic stress causes activation of the cJun NH2-terminal kinase (JNK) signal transduction pathway. It is established that one consequence of JNK activation is the development of insulin resistance and hepatic steatosis through inhibition of the transcription factor PPARα. Indeed, JNK1/2 deficiency in hepatocytes protects against the development of steatosis, suggesting that JNK inhibition represents a possible treatment for this disease. However, the long-term consequences of JNK inhibition have not been evaluated. Here we demonstrate that hepatic JNK controls bile acid production. We found that hepatic JNK deficiency alters cholesterol metabolism and bile acid synthesis, conjugation, and transport, resulting in cholestasis, increased cholangiocyte proliferation, and intrahepatic cholangiocarcinoma. Gene ablation studies confirmed that PPARα mediated these effects of JNK in hepatocytes. This analysis highlights potential consequences of long-term use of JNK inhibitors for the treatment of metabolic syndrome.


Subject(s)
Bile Acids and Salts/metabolism , Cholangiocarcinoma/enzymology , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Animals , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/physiopathology , Homeostasis , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/genetics , PPAR alpha/genetics , PPAR alpha/metabolism
7.
PLoS Pathog ; 16(8): e1008730, 2020 08.
Article in English | MEDLINE | ID: mdl-32776977

ABSTRACT

Kaposi's sarcoma (KS), caused by Kaposi's sarcoma-associated herpesvirus (KSHV), is a highly angioproliferative disseminated tumor of endothelial cells commonly found in AIDS patients. We have recently shown that KSHV-encoded viral interferon regulatory factor 1 (vIRF1) mediates KSHV-induced cell motility (PLoS Pathog. 2019 Jan 30;15(1):e1007578). However, the role of vIRF1 in KSHV-induced cellular transformation and angiogenesis remains unknown. Here, we show that vIRF1 promotes angiogenesis by upregulating sperm associated antigen 9 (SPAG9) using two in vivo angiogenesis models including the chick chorioallantoic membrane assay (CAM) and the matrigel plug angiogenesis assay in mice. Mechanistically, vIRF1 interacts with transcription factor Lef1 to promote SPAG9 transcription. vIRF1-induced SPAG9 promotes the interaction of mitogen-activated protein kinase kinase 4 (MKK4) with JNK1/2 to increase their phosphorylation, resulting in enhanced VEGFA expression, angiogenesis, cell proliferation and migration. Finally, genetic deletion of ORF-K9 from KSHV genome abolishes KSHV-induced cellular transformation and impairs angiogenesis. Our results reveal that vIRF1 transcriptionally activates SPAG9 expression to promote angiogenesis and tumorigenesis via activating JNK/VEGFA signaling. These novel findings define the mechanism of KSHV induction of the SPAG9/JNK/VEGFA pathway and establish the scientific basis for targeting this pathway for treating KSHV-associated cancers.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Herpesvirus 8, Human/metabolism , Interferon Regulatory Factors/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Sarcoma, Kaposi/metabolism , Vascular Endothelial Growth Factor A/metabolism , Viral Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Transformation, Neoplastic , Herpesvirus 8, Human/genetics , Host-Pathogen Interactions , Humans , Interferon Regulatory Factors/genetics , Male , Mice , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/physiopathology , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/physiopathology , Sarcoma, Kaposi/virology , Vascular Endothelial Growth Factor A/genetics , Viral Proteins/genetics
8.
Hepatology ; 74(5): 2561-2579, 2021 11.
Article in English | MEDLINE | ID: mdl-34048060

ABSTRACT

BACKGROUND AND AIMS: Intrahepatic cholangiocarcinoma (ICC) is a highly aggressive type of liver cancer in urgent need of treatment options. Aberrant activation of the c-Jun N-terminal kinase (JNK) pathway is a key feature in ICC and an attractive candidate target for its treatment. However, the mechanisms by which constitutive JNK activation promotes ICC growth, and therefore the key downstream effectors of this pathway, remain unknown for their applicability as therapeutic targets. Our aim was to obtain a better mechanistic understanding of the role of JNK signaling in ICC that could open up therapeutic opportunities. APPROACH AND RESULTS: Using loss-of-function and gain-of-function studies in vitro and in vivo, we show that activation of the JNK pathway promotes ICC cell proliferation by affecting the protein stability of peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (PIN1), a key driver of tumorigenesis. PIN1 is highly expressed in ICC primary tumors, and its expression positively correlates with active JNK. Mechanistically, the JNK kinases directly bind to and phosphorylate PIN1 at Ser115, and this phosphorylation prevents PIN1 mono-ubiquitination at Lys117 and its proteasomal degradation. Moreover, pharmacological inhibition of PIN1 through all-trans retinoic acid, a Food and Drug Administration-approved drug, impairs the growth of both cultured and xenografted ICC cells. CONCLUSIONS: Our findings implicate the JNK-PIN1 regulatory axis as a functionally important determinant for ICC growth, and provide a rationale for therapeutic targeting of JNK activation through PIN1 inhibition.


Subject(s)
Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/metabolism , Carcinogenesis/metabolism , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/metabolism , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Animals , Antineoplastic Agents/administration & dosage , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Line, Tumor , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Female , Gene Knockdown Techniques , Humans , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred NOD , Mice, SCID , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/genetics , NIMA-Interacting Peptidylprolyl Isomerase/antagonists & inhibitors , NIMA-Interacting Peptidylprolyl Isomerase/genetics , Phosphorylation/drug effects , Phosphorylation/genetics , RNA, Small Interfering/genetics , Tretinoin/administration & dosage , Tumor Burden/drug effects , Tumor Burden/genetics , Xenograft Model Antitumor Assays
9.
J Biol Chem ; 295(20): 6958-6971, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32265302

ABSTRACT

Disrupted clearance of all-trans-retinal (atRAL), a component of the visual (retinoid) cycle in the retina, may cause photoreceptor atrophy in autosomal recessive Stargardt disease (STGD1) and dry age-related macular degeneration (AMD). However, the mechanisms underlying atRAL-induced photoreceptor loss remain elusive. Here, we report that atRAL activates c-Jun N-terminal kinase (JNK) signaling at least partially through reactive oxygen species production, which promoted mitochondria-mediated caspase- and DNA damage-dependent apoptosis in photoreceptor cells. Damage to mitochondria in atRAL-exposed photoreceptor cells resulted from JNK activation, leading to decreased expression of Bcl2 apoptosis regulator (Bcl2), increased Bcl2 antagonist/killer (Bak) levels, and cytochrome c (Cyt c) release into the cytosol. Cytosolic Cyt c specifically provoked caspase-9 and caspase-3 activation and thereby initiated apoptosis. Phosphorylation of JNK in atRAL-loaded photoreceptor cells induced the appearance of γH2AX, a sensitive marker for DNA damage, and was also associated with apoptosis onset. Suppression of JNK signaling protected photoreceptor cells against atRAL-induced apoptosis. Moreover, photoreceptor cells lacking Jnk1 and Jnk2 genes were more resistant to atRAL-associated cytotoxicity. The Abca4-/-Rdh8-/- mouse model displays defects in atRAL clearance that are characteristic of STGD1 and dry AMD. We found that JNK signaling was activated in the neural retina of light-exposed Abca4-/-Rdh8-/- mice. Of note, intraperitoneal administration of JNK-IN-8, which inhibits JNK signaling, effectively ameliorated photoreceptor degeneration and apoptosis in light-exposed Abca4-/-Rdh8-/- mice. We propose that pharmacological inhibition of JNK signaling may represent a therapeutic strategy for preventing photoreceptor loss in retinopathies arising from atRAL overload.


Subject(s)
Apoptosis/drug effects , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Retinaldehyde/pharmacology , Signal Transduction/drug effects , Stargardt Disease/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Apoptosis/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/genetics , Photoreceptor Cells, Vertebrate/pathology , Signal Transduction/genetics , Stargardt Disease/genetics , Stargardt Disease/pathology
10.
FASEB J ; 34(2): 2126-2146, 2020 02.
Article in English | MEDLINE | ID: mdl-31909533

ABSTRACT

Bladder dysfunction is associated with the overexpression of the intermediate filament (IF) proteins desmin and vimentin in obstructed bladder smooth muscle (BSM). However, the mechanisms by which these proteins contribute to BSM dysfunction are not known. Previous studies have shown that desmin and vimentin directly participate in signal transduction. In this study, we hypothesized that BSM dysfunction associated with overexpression of desmin or vimentin is mediated via c-Jun N-terminal kinase (JNK). We employed a model of murine BSM tissue in which increased expression of desmin or vimentin was induced by adenoviral transduction to examine the sufficiency of increased IF protein expression to reduce BSM contraction. Murine BSM strips overexpressing desmin or vimentin generated less force in response to KCl and carbachol relative to the levels in control murine BSM strips, an effect associated with increased JNK2 phosphorylation and reduced myosin light chain (MLC20 ) phosphorylation. Furthermore, desmin and vimentin overexpressions did not alter BSM contractility and MLC20 phosphorylation in strips isolated from JNK2 knockout mice. Pharmacological JNK2 inhibition produced results qualitatively similar to those caused by JNK2 knockout. These findings suggest that inhibition of JNK2 may improve diminished BSM contractility associated with obstructive bladder disease.


Subject(s)
Desmin/biosynthesis , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 9/metabolism , Muscle Contraction , Muscle, Smooth/metabolism , Urinary Bladder/metabolism , Vimentin/biosynthesis , Animals , Desmin/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 9/genetics , Muscle, Smooth/cytology , Urinary Bladder/cytology , Vimentin/genetics
11.
Circ Res ; 124(9): 1386-1400, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30879399

ABSTRACT

RATIONALE: Exercise training, in addition to reducing cardiovascular risk factors, confers direct protection against myocardial ischemia/reperfusion injury and has been associated with improved heart attack survival in humans. However, the underlying mechanisms of exercise-afforded cardioprotection are still unclear. OBJECTIVE: To investigate the role of exercise-derived circulating exosomes in cardioprotection and the molecular mechanisms involved. METHODS AND RESULTS: Circulating exosomes were isolated from the plasma of volunteers with or without exercise training and rats subjected to 4-week swim exercise or sedentary littermates 24 hours after the last training session. Although the total circulating exosome level did not change significantly in exercised subjects 24 hours post-exercise compared with the sedentary control, the isolated plasma exosomes from exercised rats afforded remarkable protection against myocardial ischemia/reperfusion injury. miRNA sequencing combined with quantitative reverse transcription polymerase chain reaction validation identified 12 differentially expressed miRNAs from the circulating exosomes of exercised rats, among which miR-342-5p stood out as the most potent cardioprotective molecule. Importantly, the cardioprotective effects and the elevation of exosomal miR-342-5p were also observed in exercise-trained human volunteers. Moreover, inhibition of miR-342-5p significantly blunted the protective effects of exercise-derived circulating exosomes in hypoxia/reoxygenation cardiomyocytes; in vivo cardiac-specific inhibition of miR-342-5p through serotype 9 adeno-associated virus-mediated gene delivery attenuated exercise-afforded cardioprotection in myocardial ischemia/reperfusion rats. Mechanistically, miR-342-5p inhibited hypoxia/reoxygenation-induced cardiomyocyte apoptosis via targeting Caspase 9 and Jnk2; it also enhanced survival signaling (p-Akt) via targeting phosphatase gene Ppm1f. Of note, exercise training or laminar shear stress directly enhanced the synthesis of miR-342-5p in endothelial cells. CONCLUSIONS: Our findings reveal a novel endogenous cardioprotective mechanism that long-term exercise-derived circulating exosomes protect the heart against myocardial ischemia/reperfusion injury via exosomal miR-342-5p.


Subject(s)
Exercise/physiology , Exosomes/genetics , MicroRNAs/genetics , Animals , Apoptosis/genetics , Caspase 9/genetics , Caspase 9/metabolism , Cells, Cultured , Humans , Male , Mitogen-Activated Protein Kinase 9/genetics , Mitogen-Activated Protein Kinase 9/metabolism , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Physical Conditioning, Animal/physiology , Rats, Sprague-Dawley , Young Adult
12.
Pharmacol Res ; 164: 105375, 2021 02.
Article in English | MEDLINE | ID: mdl-33316384

ABSTRACT

Excessive binge alcohol intake is a common drinking pattern in humans, especially during holidays. Cessation of the binge drinking often leads to aberrant withdrawal behaviors, as well as serious heart rhythm abnormalities (clinically diagnosed as Holiday Heart Syndrome (HHS)). In our HHS mouse model with well-characterized binge alcohol withdrawal (BAW)-induced heart phenotypes, BAW leads to anxiety-like behaviors and cognitive impairment. We have previously reported that stress-activated c-Jun NH(2)-terminal kinase (JNK) plays a causal role in BAW-induced heart phenotypes. In the HHS brain, we found that activation of JNK2 (but not JNK1 and JNK3) in the prefrontal cortex (PFC), but not hippocampus and amygdala, led to anxiety-like behaviors and impaired cognition. DNA methylation mediated by a crucial DNA methylation enzyme, DNA methyltransferase1 (DNMT1), is known to be critical in alcohol-associated behavioral deficits. In HHS mice, JNK2 in the PFC (but not hippocampus and amygdala) causally enhanced total genomic DNA methylation via increased DNMT1 expression, which was regulated by enhanced binding of JNK downstream transcriptional factor c-JUN to the DNMT1 promoter. JNK2-specific inhibition either by an inhibitor JNK2I or JNK2 knockout completely offset c-JUN-regulated DNMT1 upregulation and restored the level of DNA methylation in HHS PFC to the baseline levels seen in sham controls. Strikingly, either JNK2-specific inhibition or genetic JNK2 depletion or DNMT1 inhibition (by an inhibitor 5-Azacytidine) completely abolished BAW-evoked behavioral deficits. In conclusion, our studies revealed a novel mechanism by which JNK2 drives BAW-evoked behavioral deficits through a DNMT1-regulated DNA hypermethylation. JNK2 could be a novel therapeutic target for alcohol withdrawal treatment and/or prevention.


Subject(s)
Behavior, Animal , Binge Drinking , DNA Methylation , Mitogen-Activated Protein Kinase 9 , Substance Withdrawal Syndrome , Amygdala/metabolism , Animals , Anxiety/enzymology , Anxiety/genetics , Binge Drinking/enzymology , Binge Drinking/genetics , Cognition , DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Hippocampus/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 9/antagonists & inhibitors , Mitogen-Activated Protein Kinase 9/genetics , Prefrontal Cortex/metabolism , Substance Withdrawal Syndrome/enzymology , Substance Withdrawal Syndrome/genetics
13.
Eur J Immunol ; 49(2): 255-265, 2019 02.
Article in English | MEDLINE | ID: mdl-30467836

ABSTRACT

Invariant natural killer T (iNKT) cells play critical roles in autoimmune, anti-tumor, and anti-microbial immune responses, and are activated by glycolipids presented by the MHC class I-like molecule, CD1d. How the activation of signaling pathways impacts antigen (Ag)-dependent iNKT cell activation is not well-known. In the current study, we found that the MAPK JNK2 not only negatively regulates CD1d-mediated Ag presentation in APCs, but also contributes to CD1d-independent iNKT cell activation. A deficiency in the JNK2 (but not JNK1) isoform enhanced Ag presentation by CD1d. Using a vaccinia virus (VV) infection model known to cause a loss in iNKT cells in a CD1d-independent, but IL-12-dependent manner, we found the virus-induced loss of iNKT cells in JNK2 KO mice was substantially lower than that observed in JNK1 KO or wild-type (WT) mice. Importantly, compared to WT mice, JNK2 KO mouse iNKT cells were found to express less surface IL-12 receptors. As with a VV infection, an IL-12 injection also resulted in a smaller decrease in JNK2 KO iNKT cells as compared to WT mice. Overall, our work strongly suggests JNK2 is a negative regulator of CD1d-mediated Ag presentation and contributes to IL-12-induced iNKT cell activation and loss during viral infections.


Subject(s)
Antigens, CD1d/immunology , Lymphocyte Activation , Mitogen-Activated Protein Kinase 9/immunology , Natural Killer T-Cells/immunology , Animals , Antigens, CD1d/genetics , Female , Interleukin-12/genetics , Interleukin-12/immunology , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 9/genetics , Receptors, Interleukin-12/genetics , Receptors, Interleukin-12/immunology , Virus Diseases/genetics , Virus Diseases/immunology
14.
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
15.
Mol Cell ; 45(6): 777-89, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22364742

ABSTRACT

Abnormal inflammatory signaling activation occurs commonly in cancer cells. However, how it is initiated and maintained and its roles in early stages of tumorigensis are largely unknown. Here, we report that the monocyte-derived MCP-1-induced transformation of immortal breast epithelial cells is triggered by transient activation of MEK/ERK and IKK/NF-κB pathways and maintained by constitutive activation of a feed-forward inflammatory signaling circuit composed of miR-200c, p65, JNK2, HSF1, and IL6. Suppression of miR-200c by IL6 constitutively activates p65/RelA and JNK2, and the latter phosphorylates and activates HSF1. In turn, HSF1 triggers demethylation of the IL6 promoter that facilitates the binding of p65 and c-Jun, which together drive constitutive IL6 transcription. Importantly, this signaling circuit is manifest in human cancer cells and in a mouse model of ErbB2-driven breast cancer, where IL6 loss significantly impairs tumorigenesis. Therefore, targeting this signaling circuit represents an effective therapeutic avenue for breast cancer prevention and treatment.


Subject(s)
Cell Transformation, Neoplastic/genetics , Inflammation/metabolism , Interleukin-6/metabolism , MicroRNAs/metabolism , Animals , Breast/cytology , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Estrogen Receptor alpha/metabolism , Female , Heat Shock Transcription Factors , Humans , Inflammation/genetics , Interleukin-6/genetics , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 9/genetics , Mitogen-Activated Protein Kinase 9/metabolism , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Promoter Regions, Genetic , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Plant Mol Biol ; 100(4-5): 411-431, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30953279

ABSTRACT

KEY MESSAGE: Physical interaction and phosphorylation by CaMPK9 protects the degradation of CaWRKY40 that induces resistance response in chickpea to Fusarium wilt disease by modulating the transcription of defense responsive genes. WRKY transcription factors (TFs) are the global regulators of plant defense signaling that modulate immune responses in host plants by regulating transcription of downstream target genes upon challenged by pathogens. However, very little is known about immune responsive role of Cicer arietinum L. (Ca) WRKY TFs particularly. Using two contrasting chickpea genotypes with respect to resistance against Fusarium oxysporum f. sp. ciceri Race1 (Foc1), we demonstrate transcript accumulation of different CaWRKYs under multiple stresses and establish that CaWRKY40 triggers defense. CaWRKY40 overexpressing chickpea mounts resistance to Foc1 by positively modulating the defense related gene expression. EMSA, ChIP assay and real-time PCR analyses suggest CaWRKY40 binds at the promoters and positively regulates transcription of CaDefensin and CaWRKY33. Further studies revealed that mitogen Activated Protein Kinase9 (CaMPK9) phosphorylates CaWRKY40 by directly interacting with its two canonical serine residues. Interestingly, CaMPK9 is unable to interact with CaWRKY40 when the relevant two serine residues were replaced by alanine. Overexpression of serine mutated WRKY40 isoform in chickpea fails to provide resistance against Foc1. Mutated WRKY40Ser.224/225 to AA overexpressing chickpea resumes its ability to confer resistance against Foc1 after application of 26S proteasomal inhibitor MG132, suggests that phosphorylation is essential to protect CaWRKY40 from proteasomal degradation. CaMPK9 silencing also led to susceptibility in chickpea to Foc1. Altogether, our results elucidate positive regulatory roles of CaMPK9 and CaWRKY40 in modulating defense response in chickpea upon Foc1 infection.


Subject(s)
Cicer/immunology , Fusarium/physiology , Plant Proteins/physiology , Cicer/metabolism , Cicer/microbiology , Mitogen-Activated Protein Kinase 9/genetics , Mitogen-Activated Protein Kinase 9/metabolism , Mitogen-Activated Protein Kinase 9/physiology , Phosphorylation , Plant Proteins/genetics , Plant Proteins/metabolism , RNA, Messenger/metabolism , Stress, Physiological , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/physiology
17.
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
18.
Biochem Biophys Res Commun ; 516(3): 739-746, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31255288

ABSTRACT

Orosomucoid-like protein 3 (ORMDL3) is a common mutation in many asthma patients and its effects on the specific pathogenesis of asthma are still unclear. Therefore, in this study, we used a mouse that specifically knockout the mouse ORDML3 gene to further study the mechanism. We used ovalbumin (OVA) to induce asthma in wild-type mice and ORMDL3 knockout mice. Lung ventilation resistance, airway inflammation, mucus hypersecretion, collagen deposition, the levels of inflammatory factors and the expression of ORDML3 and JNK1/2-MMP-9 pathway were detected. The results showed that ORMDL3 gene was highly expressed in clinical asthmatic children and mouse asthma model. Knocking down the ORMDL3 gene in the lung tissue of asthmatic mice can reduce airway hyperresponsiveness, airway inflammation, mucus secretion, and collagen deposition around the airway. After knocking down the lung tissue of mice, the IL-4, IL-5 and IL-13 concentrations in broncho alveolar lavage fluid of asthmatic mice were significantly decreased, and the activation of JNK1/2-MMP-9 pathway was inhibited in mouse lung tissue. Collectively, our results demonstrate that the ORMDL3 gene may aggravate asthma symptoms by activating the JNK1/2-MMP-9 pathway, which indicates that the ORMDL3 gene may be the key molecule for the next step of asthma targeted therapy.


Subject(s)
Asthma/genetics , Matrix Metalloproteinase 9/metabolism , Membrane Proteins/genetics , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Respiratory Hypersensitivity/genetics , Airway Remodeling/drug effects , Animals , Asthma/chemically induced , Asthma/physiopathology , Asthma/prevention & control , Bronchoalveolar Lavage Fluid/chemistry , Child , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Interleukin-13/genetics , Interleukin-13/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Interleukin-5/genetics , Interleukin-5/metabolism , Lung/drug effects , Lung/metabolism , Lung/physiopathology , Matrix Metalloproteinase 9/genetics , Membrane Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/genetics , Mucus/chemistry , Mucus/metabolism , Ovalbumin/administration & dosage , Respiratory Function Tests , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/physiopathology , Respiratory Hypersensitivity/prevention & control , Signal Transduction
19.
FASEB J ; 32(5): 2722-2734, 2018 05.
Article in English | MEDLINE | ID: mdl-29298863

ABSTRACT

In the context of diabetes, obesity, and metabolic syndrome, the inflammatory signaling has critical roles in the pathogenesis of nonalcoholic fatty liver disease (NAFLD), but the underlying mechanisms remain poorly delineated. Herein, early and persistently elevated, proinflammatory cytokine HMGB1 expression was detected in a high-fat diet (HFD)-induced NAFLD model in C57BL/6 mice. The expression and extracellular release of HMGB1 was rapidly and dramatically induced by saturated palmitic acid in vitro. HFD-induced inflammatory response and liver function impairment were both mitigated after the inhibition of endogenous HMGB1 by neutralizing antibody in vivo. The up-regulation of HMGB1 was thought to be modified by dual channels: in the transcriptional level, it was regulated by JNK1/JNK2-ATF2 axis; post-transcriptionally, it was regulated by the microRNA (miR)-200 family, especially miR-429. miR-429 liver conditional knockout mice (miR-429Δhep), fed either a normal diet or an HFD, showed severe liver inflammation and dysfunction, accompanied by greater expression of HMGB1. Intriguingly, the up-regulation and release of HMGB1 could in turn self-activate TLR4-JNK1/JNK2-ATF2 signaling, thus forming a positive feedback. Our findings reveal a novel mechanism by which HMGB1 expression was regulated by both the JNK1/2-ATF2 axis and the miR-200 family, which provides a potential new approach for the treatment of NAFLD.-Chen, X., Ling, Y., Wei, Y., Tang, J., Ren, Y., Zhang, B., Jiang, F., Li, H., Wang, R., Wen, W., Lv, G., Wu, M., Chen, L., Li, L., Wang, H. Dual regulation of HMGB1 by combined JNK1/2-ATF2 axis with miR-200 family in nonalcoholic steatohepatitis in mice.


Subject(s)
Activating Transcription Factor 2/metabolism , HMGB1 Protein/biosynthesis , MicroRNAs/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Signal Transduction , Activating Transcription Factor 2/genetics , Animals , Dietary Fats/adverse effects , Dietary Fats/pharmacology , HMGB1 Protein/genetics , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/genetics , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology
20.
Sleep Breath ; 23(1): 259-267, 2019 Mar.
Article in English | MEDLINE | ID: mdl-29992456

ABSTRACT

PURPOSE: Obstructive sleep apnea (OSA) is induced by obstruction of the upper airway, which can raise multiple health risks. This study is designed to reveal the key genes involved in OSA. METHODS: GSE38792 was extracted from Gene Expression Omnibus database, including ten visceral adipose tissues from OSA patients and eight visceral adipose tissues from normal controls. Differential expression analysis was conducted using limma package, and then the functions of the differentially expressed genes (DEGs) were analyzed using DAVID database, followed by protein-protein interaction (PPI) network, and integrated regulatory network analysis was performed using Cytoscape software. RESULTS: A total of 368 DEGs (176 upregulated and 192 downregulated) were identified in OSA samples. Epstein-Barr virus infection (involving IL10RB, MAPK9, and MAPK10) and olfactory transduction were the main pathways separately enriched for the upregulated genes and the downregulated genes. After the PPI network was built, the top ten network nodes (such as TXN) were selected according to node degrees. Two significant PPI network modules were identified. Moreover, the integrated regulatory network was constructed. CONCLUSION: IL10RB, MAPK9, MAPK10, and TXN might function in the pathogenesis of OSA.


Subject(s)
Computational Biology , Sleep Apnea, Obstructive/genetics , Adipose Tissue/metabolism , Down-Regulation/genetics , Epstein-Barr Virus Infections/genetics , Gene Expression Profiling , Gene Regulatory Networks/genetics , Humans , Interleukin-10 Receptor beta Subunit/genetics , Mitogen-Activated Protein Kinase 10/genetics , Mitogen-Activated Protein Kinase 9/genetics , Oligonucleotide Array Sequence Analysis , Protein Interaction Domains and Motifs/genetics , Reference Values , Thioredoxins/genetics , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL