Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.325
Filter
1.
Physiol Res ; 73(2): 295-304, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38710060

ABSTRACT

Aging leads to a decrease in muscle function, mass, and strength in skeletal muscle of animals and humans. The transcriptome identified activation of the JAK/STAT pathway, a pathway that is associated with skeletal muscle atrophy, and endurance training has a significant effect on improving sarcopenia; however, the exact mechanism still requires further study. We investigated the effect of endurance training on sarcopenia. Six-month-old male SAMR1 mice were used as a young control group (group C), and the same month-old male SAMP8 mice were divided into an exercise group (group E) and a model group (group M). A 3-month running exercise intervention was performed on group E, and the other two groups were kept normally. Aging caused significant signs of sarcopenia in the SAMP8 mice, and endurance training effectively improved muscle function, muscle mass, and muscle strength in the SAMP8 mice. The expression of JAK2/STAT3 pathway factor was decreased in group E compared with group M, and the expression of SOCS3, the target gene of STAT3, and NR1D1, an atrophy-related factor, was significantly increased. Endurance training significantly improved the phenotypes associated with sarcopenia, and the JAK2/STAT3 pathway is a possible mechanism for the improvement of sarcopenia by endurance training, while NR1D1 may be its potential target. Keywords: Sarcopenia, Endurance training, Janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3), Nuclear receptor subfamily 1, group D member 1 (Nr1d1).


Subject(s)
Endurance Training , Janus Kinase 2 , Physical Conditioning, Animal , STAT3 Transcription Factor , Sarcopenia , Signal Transduction , Animals , Sarcopenia/metabolism , Sarcopenia/prevention & control , Sarcopenia/therapy , Janus Kinase 2/metabolism , STAT3 Transcription Factor/metabolism , Male , Mice , Physical Conditioning, Animal/physiology , Muscle, Skeletal/metabolism , Aging/metabolism
2.
Nat Commun ; 15(1): 3976, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38729948

ABSTRACT

Bleeding and thrombosis are known as common complications of polycythemia for a long time. However, the role of coagulation system in erythropoiesis is unclear. Here, we discover that an anticoagulant protein tissue factor pathway inhibitor (TFPI) plays an essential role in erythropoiesis via the control of heme biosynthesis in central macrophages. TFPI levels are elevated in erythroblasts of human erythroblastic islands with JAK2V617F mutation and hypoxia condition. Erythroid lineage-specific knockout TFPI results in impaired erythropoiesis through decreasing ferrochelatase expression and heme biosynthesis in central macrophages. Mechanistically, the TFPI interacts with thrombomodulin to promote the downstream ERK1/2-GATA1 signaling pathway to induce heme biosynthesis in central macrophages. Furthermore, TFPI blockade impairs human erythropoiesis in vitro, and normalizes the erythroid compartment in mice with polycythemia. These results show that erythroblast-derived TFPI plays an important role in the regulation of erythropoiesis and reveal an interplay between erythroblasts and central macrophages.


Subject(s)
Erythroblasts , Erythropoiesis , GATA1 Transcription Factor , Heme , Lipoproteins , Macrophages , Polycythemia , Polycythemia/metabolism , Polycythemia/genetics , Polycythemia/pathology , Erythroblasts/metabolism , Heme/metabolism , Humans , Animals , Lipoproteins/metabolism , Macrophages/metabolism , Mice , GATA1 Transcription Factor/metabolism , GATA1 Transcription Factor/genetics , Janus Kinase 2/metabolism , Janus Kinase 2/genetics , Thrombomodulin/metabolism , Thrombomodulin/genetics , Mice, Knockout , Ferrochelatase/metabolism , Ferrochelatase/genetics , Male , MAP Kinase Signaling System , Mice, Inbred C57BL , Female
3.
FASEB J ; 38(10): e23667, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38742812

ABSTRACT

Immunity imbalance of T helper 17 (Th17)/regulatory T (Treg) cells is involved in the pathogenesis of Crohn's disease (CD). Complanatuside A (CA), a flavonol glycoside, exerts anti-inflammatory activities and our study aimed to identify its effect on TNBS-induced colitis and the possible mechanisms. We found that CA alleviated the symptoms of colitis in TNBS mice, as demonstrated by prevented weight loss and colon length shortening, as well as decreased disease activity index scores, inflammatory scores, and levels of proinflammatory factors. Flow cytometry analysis showed that CA markedly reduced the percentage of Th17 cells while increasing the percentage of Treg cells in TNBS mice. Under Th17 cell polarizing conditions, CA inhibited the differentiation of Th17 cells while the Treg cell differentiation was elevated under Treg cell polarizing conditions. Furthermore, it was observed that JAK2 interacted with CA through six hydrogen bonds via molecular docking. The phosphorylation of JAK2/STAT3 was reduced by CA, which might be correlated with the protective effect of CA on colitis. In conclusion, CA reduced the imbalance of Th17/Treg cells by inhibiting the JAK2/STAT3 signaling pathway in TNBS-induced colitis, which may provide novel strategies for CD treatment.


Subject(s)
Colitis , Janus Kinase 2 , STAT3 Transcription Factor , Signal Transduction , T-Lymphocytes, Regulatory , Th17 Cells , Trinitrobenzenesulfonic Acid , Animals , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/metabolism , Janus Kinase 2/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , STAT3 Transcription Factor/metabolism , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Mice , Signal Transduction/drug effects , Trinitrobenzenesulfonic Acid/toxicity , Male , Mice, Inbred BALB C , Cell Differentiation/drug effects
5.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(4): 765-772, 2024 Apr 20.
Article in Chinese | MEDLINE | ID: mdl-38708511

ABSTRACT

OBJECTIVE: To investigate the expression level of Kruppel-like transcription factor family member KLF11 in intestinal mucosal tissues of Crohn's disease (CD) and its regulatory effect on intestinal inflammation in CD-like colitis. METHODS: We examined KLF11 expression levels in diseased and normal colon mucosal tissues from 12 CD patients and 12 patients with colorectal cancer using immunofluorescence staining. KLF11 expression was also detected in the colon mucosal tissues of a mouse model of 2, 4, 6-trinitrobenesulfonic acid (TNBS)-induced colitis. A recombinant adenoviral vector was used to upregulate KLF11 expression in the mouse models and the changes in intestinal inflammation was observed. A Caco-2 cell model with stable KLF11 overexpression was constructed by lentiviral infection. The effect of KLF11 overexpression on expressions of JAK2/STAT3 signaling pathway proteins was investigated using immunoblotting in both the mouse and cell models. The mouse models were treated with coumermycin A1, a JAK2/STAT3 signaling pathway agonist, and the changes in intestinal inflammatory responses were observed. RESULTS: The expression level of KLF11 was significantly lowered in both the clinical specimens of diseased colon mucosal tissues and the colon tissues of mice with TNBS-induced colitis (P < 0.05). Adenovirus-mediated upregulation of KLF11 significantly improved intestinal inflammation and reduced the expression levels of inflammatory factors in the intestinal mucosa of the colitis mouse models (P < 0.05). Overexpression of KLF11 significantly inhibited the expression levels of p-JAK2 and p-STAT3 in intestinal mucosal tissues of the mouse models and in Caco-2 cells (P < 0.05). Treatment with coumermycin A1 obviously inhibited the effect of KLF11 upregulation for improving colitis and significantly increased the expression levels of inflammatory factors in the intestinal mucosa of the mouse models (P < 0.05). CONCLUSION: KLF11 is downregulated in the intestinal mucosa in CD, and upregulation of KLF11 can improve intestinal inflammation and reduce the production of inflammatory factors probably by inhibiting the JAK2/STAT3 signaling pathway.


Subject(s)
Apoptosis Regulatory Proteins , Colitis , Intestinal Mucosa , Janus Kinase 2 , Repressor Proteins , STAT3 Transcription Factor , Signal Transduction , Trinitrobenzenesulfonic Acid , Animals , Mice , Colitis/chemically induced , Colitis/metabolism , Humans , STAT3 Transcription Factor/metabolism , Janus Kinase 2/metabolism , Caco-2 Cells , Intestinal Mucosa/metabolism , Disease Models, Animal , Crohn Disease/metabolism , Inflammation/metabolism , Up-Regulation , Transcription Factors/metabolism , Transcription Factors/genetics
6.
Int J Rheum Dis ; 27(5): e15164, 2024 May.
Article in English | MEDLINE | ID: mdl-38706209

ABSTRACT

BACKGROUND: JAK inhibitors are well known for the treatment of rheumatoid arthritis (RA), but whether they can be used to treat pulmonary fibrosis, a common extra-articular disease of RA, remains to be clarified. METHODS: A jak2 inhibitor, CEP33779 (CEP), was administered to a rat model of RA-associated interstitial lung disease to observe the degree of improvement in both joint swelling and pulmonary fibrosis. HFL1 cells were stimulated with TGF-ß1 to observe the expression of p-JAK2. Then, different concentrations of related gene inhibitors (JAK2, TGFß-R1/2, and p-STAT3) or silencers (STAT3, JAK2) were administered to HFL1 cells, and the expression levels of related proteins were detected to explore the underlying mechanisms of action. RESULTS: CEP not only reduced the degree of joint swelling and inflammation in rats but also improved lung function, inhibited the pro-inflammatory factors IL-1ß and IL-6, reduced lung inflammation and collagen deposition, and alleviated lung fibrosis. CEP decreased the expression levels of TGFß-R2, p-SMAD, p-STAT3, and ECM proteins in rat lung tissues. TGF-ß1 induced HFL1 cells to highly express p-JAK2, with the most pronounced expression at 48 h. The levels of p-STAT3, p-SMAD3, and ECM-related proteins were significantly reduced after inhibition of either JAK2 or STAT3. CONCLUSION: JAK2 inhibitors may be an important and novel immunotherapeutic drug that can improve RA symptoms while also delaying or blocking the development of associated pulmonary fibrotic disease. The mechanism may be related to the downregulation of p-STAT3 protein via inhibition of the JAK2/STAT signaling pathway, which affects the phosphorylation of SMAD3.


Subject(s)
Disease Models, Animal , Down-Regulation , Isoquinolines , Janus Kinase 2 , Lung , Pulmonary Fibrosis , Pyridines , Pyrroles , Signal Transduction , Smad3 Protein , Animals , Smad3 Protein/metabolism , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Janus Kinase 2/metabolism , Janus Kinase 2/antagonists & inhibitors , Phosphorylation , Signal Transduction/drug effects , Lung/drug effects , Lung/metabolism , Lung/pathology , Lung/enzymology , Male , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Humans , Rats, Sprague-Dawley , Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/therapeutic use , Cell Line , Protein Kinase Inhibitors/pharmacology , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Arthritis, Experimental/enzymology , Anti-Inflammatory Agents/pharmacology , Rats
7.
Int Immunopharmacol ; 133: 112133, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38652962

ABSTRACT

There is an increasing tendency for sepsis patients to suffer from diaphragm atrophy as well as mortality. Therefore, reducing diaphragm atrophy could benefit sepsis patients' prognoses. Studies have shown that Anisodamine (Anis) can exert antioxidant effects when blows occur. However, the role of Anisodamine in diaphragm atrophy in sepsis patients has not been reported. Therefore, this study investigated the antioxidant effect of Anisodamine in sepsis-induced diaphragm atrophy and its mechanism. We used cecal ligation aspiration (CLP) to establish a mouse septic mode and stimulated the C2C12 myotube model with lipopolysaccharide (LPS). After treatment with Anisodamine, we measured the mice's bodyweight, diaphragm weight, fiber cross-sectional area and the diameter of C2C12 myotubes. The malondialdehyde (MDA) levels in the diaphragm were detected using the oxidative stress kit. The expression of MuRF1, Atrogin1 and JAK2/STAT3 signaling pathway components in the diaphragm and C2C12 myotubes was measured by RT-qPCR and Western blot. The mean fluorescence intensity of ROS in C2C12 myotubes was measured by flow cytometry. Meanwhile, we also measured the levels of Drp1 and Cytochrome C (Cyt-C) in vivo and in vitro by Western blot. Our study revealed that Anisodamine alleviated the reduction in diaphragmatic mass and the loss of diaphragmatic fiber cross-sectional area and attenuated the atrophy of the C2C12 myotubes by inhibiting the expression of E3 ubiquitin ligases. In addition, we observed that Anisodamine inhibited the JAK2/STAT3 signaling pathway and protects mitochondrial function. In conclusion, Anisodamine alleviates sepsis-induced diaphragm atrophy, and the mechanism may be related to inhibiting the JAK2/STAT3 signaling pathway.


Subject(s)
Diaphragm , Janus Kinase 2 , Muscular Atrophy , STAT3 Transcription Factor , Sepsis , Signal Transduction , Solanaceous Alkaloids , Animals , Janus Kinase 2/metabolism , STAT3 Transcription Factor/metabolism , Sepsis/drug therapy , Sepsis/complications , Solanaceous Alkaloids/therapeutic use , Solanaceous Alkaloids/pharmacology , Mice , Signal Transduction/drug effects , Diaphragm/drug effects , Diaphragm/pathology , Diaphragm/metabolism , Male , Cell Line , Muscular Atrophy/drug therapy , Muscular Atrophy/etiology , Disease Models, Animal , Lipopolysaccharides , Mice, Inbred C57BL , Oxidative Stress/drug effects , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Antioxidants/pharmacology , Antioxidants/therapeutic use , Muscle Proteins/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , SKP Cullin F-Box Protein Ligases/genetics , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/metabolism , Atrophy
8.
Int Immunopharmacol ; 133: 112126, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38669946

ABSTRACT

Type 17 helper T cells (Th17)-dominant neutrophilic airway inflammation is critical in the pathogenesis of steroid-resistant airway inflammation such as severe asthma. Small extracellular vesicles (sEV) derived from human mesenchymal stem cells (MSCs) display extensive therapeutic effects and advantages in many diseases. However, the role of MSC-sEV in Th17-dominant neutrophilic airway inflammation and the related mechanisms are still poorly studied. Here we found that MSC-sEV significantly alleviated the infiltration of inflammatory cells in peribronchial interstitial tissues and reduced levels of inflammatory cells, especially neutrophils, in bronchoalveolar lavage fluids (BALF) of mice with neutrophilic airway inflammation. Consistently, MSC-sEV significantly decreased levels of IL-17A in BALF and Th17 in lung tissues. Furthermore, we found that labelled MSC-sEV were taken up by human CD4+ T cells most obviously at 12 h after incubation, and distributed mostly in mouse lungs. More importantly, potential signaling pathways involved in the MSC-sEV mediated inhibition of Th17 polarization were found using RNA sequencing. Using Western blot, JAK2-STAT3 pathway was identified as an important role in the inhibition of Th17 polarization by MSC-sEV. We found that proteins in MSC-sEV were mostly involved in the therapeutic effects of MSC-sEV. In total, our study suggested that MSC-sEV could be a potential therapeutic strategy for the treatment of neutrophilic airway inflammation.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Neutrophils , STAT3 Transcription Factor , Th17 Cells , Th17 Cells/immunology , Humans , Animals , Extracellular Vesicles/metabolism , Extracellular Vesicles/immunology , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Mice , Neutrophils/immunology , STAT3 Transcription Factor/metabolism , Janus Kinase 2/metabolism , Interleukin-17/metabolism , Lung/immunology , Lung/pathology , Mice, Inbred C57BL , Cells, Cultured , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/cytology , Asthma/immunology , Asthma/therapy , Male , Signal Transduction , Female , Disease Models, Animal
9.
Int J Mol Sci ; 25(8)2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38673975

ABSTRACT

Previously, we reported that epidermal growth factor-like module-containing mucin-like hormone receptor-like 1 (EMR1/ADGRE1) is abnormally expressed in colon cancer (CC) and is a risk factor for lymph node metastasis (LNM) and poor recurrence-free survival in patients with abundant tumor-associated macrophages (TAMs). However, the signaling pathways associated with EMR1 expression in CC progression remain unclear. In this study, we aimed to explore the role of EMR1 and its signaling interactions with macrophages in CC progression. Spatial transcriptomics of pT3 microsatellite unstable CC tissues revealed heightened Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling in EMR1-HL CC with LNM compared to EMR1-N CC without LNM. Through in vitro coculture of CC cells with macrophages, EMR1 expression by CC cells was found to be induced by TAMs, ultimately interacting with upregulated JAK/STAT signaling, increasing cell proliferation, migration, and motility, and reducing apoptosis. JAK2/STAT3 inhibition decreased the levels of EMR1, JAK2, STAT1, and STAT3, significantly impeded the proliferation, migration, and mobility of cells, and increased the apoptosis of EMR1+ CC cells compared to their EMR1KO counterparts. Overall, TAMs-induced EMR1 upregulation in CC cells may promote LNM and CC progression via JAK2/STAT1,3 signaling upregulation. This study provides further insights into the molecular mechanisms involving macrophages and intracellular EMR1 expression in CC progression, suggesting its clinical significance and offering potential interventions to enhance patient outcomes.


Subject(s)
Colonic Neoplasms , Janus Kinase 2 , Signal Transduction , Tumor-Associated Macrophages , Humans , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology , Janus Kinase 2/metabolism , Janus Kinase 2/genetics , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Disease Progression , Up-Regulation , Cell Proliferation , Cell Line, Tumor , Cell Movement/genetics , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Apoptosis/genetics
10.
Int J Biol Macromol ; 267(Pt 2): 131520, 2024 May.
Article in English | MEDLINE | ID: mdl-38615859

ABSTRACT

The adverse microenvironment, including neuroinflammation, hinders the recovery of spinal cord injury (SCI). Regulating microglial polarization to alleviate neuroinflammation at the injury site is an effective strategy for SCI recovery. MG53 protein exerts obvious repair ability on multiple tissues damage, but with short half-life. In this study, we composited an innovative MG53/GMs/HA-Dex neural scaffold using gelatin microspheres (GMs), hyaluronic acid (HA), and dextran (Dex) loaded with MG53 protein. This novel neural scaffold could respond to MMP-2/9 protein and stably release MG53 protein with good physicochemical properties and biocompatibility. In addition, it significantly improved the motor function of SCI mice, suppressed M1 polarization of microglia and neuroinflammation, and promoted neurogenesis and axon regeneration. Further mechanistic experiments demonstrated that MG53/GMs/HA-Dex hydrogel inhibited the JAK2/STAT3 signaling pathway. Thus, this MG53/GMs/HA-Dex neural scaffold promotes the functional recovery of SCI mice by alleviating neuroinflammation, which provides a new intervention strategy for the neural regeneration and functional repair of SCI.


Subject(s)
Gelatin , Hyaluronic Acid , Janus Kinase 2 , Neuroinflammatory Diseases , Recovery of Function , Spinal Cord Injuries , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Animals , Mice , Recovery of Function/drug effects , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Neuroinflammatory Diseases/drug therapy , Gelatin/chemistry , Gelatin/pharmacology , Janus Kinase 2/metabolism , Dextrans/chemistry , Tissue Scaffolds/chemistry , Microspheres , STAT3 Transcription Factor/metabolism , Microglia/drug effects , Microglia/metabolism , Nerve Regeneration/drug effects , Matrix Metalloproteinase 9/metabolism , Disease Models, Animal , Neurogenesis/drug effects , Signal Transduction/drug effects , Matrix Metalloproteinase 2/metabolism , Hydrogels/chemistry , Hydrogels/pharmacology
11.
Pharmacol Res ; 203: 107178, 2024 May.
Article in English | MEDLINE | ID: mdl-38583686

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is one of the most fatal chronic interstitial lung diseases with unknown pathogenesis, current treatments cannot truly reverse the progression of the disease. Pulmonary macrophages, especially bone marrow derived pro-fibrotic macrophages, secrete multiple kinds of profibrotic mediators (SPP1, CD206, CD163, IL-10, CCL18…), thus further promote myofibroblast activation and fibrosis procession. IL20Rb is a cell-surface receptor that belongs to IL-20 family. The role of IL20Rb in macrophage activation and pulmonary fibrosis remains unclear. In this study, we established a bleomycin-induced pulmonary fibrosis model, used IL4/13-inducing THP1 cells to induce profibrotic macrophage (M2-like phenotype) polarization models. We found that IL20Rb is upregulated in the progression of pulmonary fibrosis, and its absence can alleviate the progression of pulmonary fibrosis. In addition, we demonstrated that IL20Rb promote the activation of bone marrow derived profibrotic macrophages by regulating the Jak2/Stat3 and Pi3k/Akt signaling pathways. In terms of therapeutic strategy, we used IL20Rb neutralizing antibodies for animal administration, which was found to alleviate the progression of IPF. Our results suggest that IL20Rb plays a profibrotic role by promoting profibrotic macrophage polarization, and IL20Rb may become a potential therapeutic target for IPF. Neutralizing antibodies against IL20Rb may become a potential drug for the clinical treatment of IPF.


Subject(s)
Bleomycin , Macrophage Activation , Macrophages , Mice, Inbred C57BL , Animals , Humans , Macrophages/metabolism , Macrophages/immunology , Bleomycin/toxicity , Mice , Male , Receptors, Interleukin/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/immunology , Signal Transduction , STAT3 Transcription Factor/metabolism , Janus Kinase 2/metabolism , THP-1 Cells , Lung/pathology , Lung/metabolism , Lung/immunology , Lung/drug effects , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/chemically induced , Mice, Knockout , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism
12.
Oncol Rep ; 51(5)2024 May.
Article in English | MEDLINE | ID: mdl-38577936

ABSTRACT

Cancer cells are characterized by increased glycolysis, known as the Warburg effect, which leads to increased production of cytotoxic methylglyoxal (MGO) and apoptotic cell death. Cancer cells often activate the protective nuclear factor erythroid 2­related factor2 (Nrf2)/glyoxalase1 (Glo1) system to detoxify MGO. The effects of sodium butyrate (NaB), a product of gut microbiota, on Nrf2/Glos/MGO pathway and the underlying mechanisms in prostate cancer (PCa) cells were investigated in the present study. Treatment with NaB induced the cell death and reduced the proliferation of PCa cells (DU145 and LNCap). Moreover, the protein kinase RNA-like endoplasmic reticulum kinase/Nrf2/Glo1 pathway was greatly inhibited by NaB, thereby accumulating MGO-derived adduct hydroimidazolone (MG-H1). In response to a high amount of MGO, the expression of Nrf2 and Glo1 was attenuated, coinciding with an increased cellular death. NaB also markedly inhibited the Janus kinase 2 (JAK2)/Signal transducer and activator of transcription 3 (Stat3) pathway. Conversely, co­treatment with Colivelin, a Stat3 activator, significantly reversed the effects of NaB on Glo1 expression, MG-H1 production, and the cell migration and viability. As expected, overexpression of Stat3 or Glo1 reduced NaB­induced cell death. The activation of calcium/calmodulin dependent protein kinase II gamma and reactive oxygen species production also contributed to the anticancer effect of NaB. The present study, for the first time, demonstrated that NaB greatly increases MGO production through suppression of the JAK2/Stat3/Nrf2/Glo1 pathway in DU145 cells, a cell line mimicking castration­resistant PCa (CRPC), suggesting that NaB may be a potential agent for PCa therapy.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Humans , Male , Butyric Acid/pharmacology , Janus Kinase 2/metabolism , Magnesium Oxide/metabolism , NF-E2-Related Factor 2/metabolism , Pyruvaldehyde/metabolism , STAT3 Transcription Factor/metabolism
13.
Nutrients ; 16(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38674862

ABSTRACT

Leptin is an appetite-regulating adipokine that is reduced in patients with anorexia nervosa (AN), a psychiatric disorder characterized by self-imposed starvation, and has been linked to hyperactivity, a hallmark of AN. However, it remains unknown how leptin receptor (LepR) and its JAK2-STAT3 downstream pathway in extrahypothalamic brain areas, such as the dorsal (dHip) and ventral (vHip) hippocampus, crucial for spatial memory and emotion regulation, may contribute to the maintenance of AN behaviors. Taking advantage of the activity-based anorexia (ABA) model (i.e., the combination of food restriction and physical activity), we observed reduced leptin plasma levels in adolescent female ABA rats at the acute phase of the disorder [post-natal day (PND) 42], while the levels increased over control levels following a 7-day recovery period (PND49). The analysis of the intracellular leptin pathway revealed that ABA rats showed an overall decrease of the LepR/JAK2/STAT3 signaling in dHip at both time points, while in vHip we observed a transition from hypo- (PND42) to hyperactivation (PND49) of the pathway. These changes might add knowledge on starvation-induced fluctuations in leptin levels and in hippocampal leptin signaling as initial drivers of the transition from adaptative mechanisms to starvation toward the maintenance of aberrant behaviors typical of AN patients, such as perpetuating restraint over eating.


Subject(s)
Anorexia , Hippocampus , Janus Kinase 2 , Leptin , Receptors, Leptin , STAT3 Transcription Factor , Signal Transduction , Animals , Female , Janus Kinase 2/metabolism , STAT3 Transcription Factor/metabolism , Hippocampus/metabolism , Leptin/blood , Anorexia/etiology , Anorexia/metabolism , Rats , Receptors, Leptin/metabolism , Anorexia Nervosa/metabolism , Anorexia Nervosa/blood , Disease Models, Animal , Adaptation, Physiological
14.
Molecules ; 29(8)2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38675621

ABSTRACT

Allogeneic hematopoietic cell transplantation (allo-HCT) is a highly effective, well-established treatment for patients with various hematologic malignancies and non-malignant diseases. The therapeutic benefits of allo-HCT are mediated by alloreactive T cells in donor grafts. However, there is a significant risk of graft-versus-host disease (GvHD), in which the donor T cells recognize recipient cells as foreign and attack healthy organs in addition to malignancies. We previously demonstrated that targeting JAK1/JAK2, mediators of interferon-gamma receptor (IFNGR) and IL-6 receptor signaling, in donor T cells using baricitinib and ruxolitinib results in a significant reduction in GvHD after allo-HCT. Furthermore, we showed that balanced inhibition of JAK1/JAK2 while sparing JAK3 is important for the optimal prevention of GvHD. Thus, we have generated novel JAK1/JAK2 inhibitors, termed WU derivatives, by modifying baricitinib. Our results show that WU derivatives have the potential to mitigate GvHD by upregulating regulatory T cells and immune reconstitution while reducing the frequencies of antigen-presenting cells (APCs) and CD80 expression on these APCs in our preclinical mouse model of allo-HCT. In addition, WU derivatives effectively downregulated CXCR3 and T-bet in primary murine T cells. In summary, we have generated novel JAK inhibitors that could serve as alternatives to baricitinib or ruxolitinib.


Subject(s)
Disease Models, Animal , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Janus Kinase 2 , Pyrazoles , Transplantation, Homologous , Animals , Graft vs Host Disease/prevention & control , Graft vs Host Disease/drug therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Mice , Janus Kinase 2/metabolism , Janus Kinase 2/antagonists & inhibitors , Pyrazoles/pharmacology , Purines/pharmacology , Janus Kinase Inhibitors/pharmacology , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Sulfonamides/pharmacology , Azetidines/pharmacology , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/metabolism
15.
Nat Commun ; 15(1): 3415, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649367

ABSTRACT

An important epigenetic component of tyrosine kinase signaling is the phosphorylation of histones, and epigenetic readers, writers, and erasers. Phosphorylation of protein arginine methyltransferases (PRMTs), have been shown to enhance and impair their enzymatic activity. In this study, we show that the hyperactivation of Janus kinase 2 (JAK2) by the V617F mutation phosphorylates tyrosine residues (Y149 and Y334) in coactivator-associated arginine methyltransferase 1 (CARM1), an important target in hematologic malignancies, increasing its methyltransferase activity and altering its target specificity. While non-phosphorylatable CARM1 methylates some established substrates (e.g. BAF155 and PABP1), only phospho-CARM1 methylates the RUNX1 transcription factor, on R223 and R319. Furthermore, cells expressing non-phosphorylatable CARM1 have impaired cell-cycle progression and increased apoptosis, compared to cells expressing phosphorylatable, wild-type CARM1, with reduced expression of genes associated with G2/M cell cycle progression and anti-apoptosis. The presence of the JAK2-V617F mutant kinase renders acute myeloid leukemia (AML) cells less sensitive to CARM1 inhibition, and we show that the dual targeting of JAK2 and CARM1 is more effective than monotherapy in AML cells expressing phospho-CARM1. Thus, the phosphorylation of CARM1 by hyperactivated JAK2 regulates its methyltransferase activity, helps select its substrates, and is required for the maximal proliferation of malignant myeloid cells.


Subject(s)
Apoptosis , Core Binding Factor Alpha 2 Subunit , Janus Kinase 2 , Protein-Arginine N-Methyltransferases , Tyrosine , Humans , Phosphorylation , Janus Kinase 2/metabolism , Janus Kinase 2/genetics , Protein-Arginine N-Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Tyrosine/metabolism , Cell Line, Tumor , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Methylation , Substrate Specificity , HEK293 Cells , Cell Cycle , Mutation
16.
Nat Commun ; 15(1): 2989, 2024 Apr 06.
Article in English | MEDLINE | ID: mdl-38582902

ABSTRACT

Despite the identification of driver mutations leading to the initiation of myeloproliferative neoplasms (MPNs), the molecular pathogenesis of MPNs remains incompletely understood. Here, we demonstrate that growth arrest and DNA damage inducible gamma (GADD45g) is expressed at significantly lower levels in patients with MPNs, and JAK2V617F mutation and histone deacetylation contribute to its reduced expression. Downregulation of GADD45g plays a tumor-promoting role in human MPN cells. Gadd45g insufficiency in the murine hematopoietic system alone leads to significantly enhanced growth and self-renewal capacity of myeloid-biased hematopoietic stem cells, and the development of phenotypes resembling MPNs. Mechanistically, the pathogenic role of GADD45g insufficiency is mediated through a cascade of activations of RAC2, PAK1 and PI3K-AKT signaling pathways. These data characterize GADD45g deficiency as a novel pathogenic factor in MPNs.


Subject(s)
Myeloproliferative Disorders , Neoplasms , Animals , Humans , Mice , Janus Kinase 2/metabolism , Mutation , Myeloproliferative Disorders/pathology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/genetics
17.
Gene ; 916: 148449, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-38588931

ABSTRACT

Germline-specific genes are usually activated in cancer cells and drive cancer progression; such genes are called cancer-germline or cancer-testis genes. The RNA-binding protein DAZL is predominantly expressed in germ cells and plays a role in gametogenesis as a translational activator or repressor. However, its expression and role in non-small cell lung cancer (NSCLC) are unknown. Here, mining of RNA-sequencing data from public resources and immunohistochemical analysis of tissue microarrays showed that DAZL was expressed exclusively in testis among normal human tissues but ectopically expressed in NSCLC tissues. Testis and NSCLC cells expressed the shorter and longer transcript variants of the DAZL gene, respectively. Overexpression of the longer DAZL transcript promoted tumor growth in a mouse xenograft model. Silencing of DAZL suppressed cell proliferation, colony formation, migration, invasion, and cisplatin resistance in vitro and tumor growth in vivo. Quantitative proteomic analysis based on tandem mass tag and Western blot analysis showed that DAZL upregulated the expression of JAK2 and MCM8. RNA-binding protein immunoprecipitation assays showed that DAZL bound to the mRNA of JAK2 and MCM8. The JAK2 inhibitor fedratinib attenuated the oncogenic outcomes induced by DAZL overexpression, whereas silencing MCM8 counteracted the effects of DAZL overexpression on cisplatin-damaged DNA synthesis and half-maximal inhibitory concentration of cisplatin. In conclusion, DAZL was identified as a novel cancer-germline gene that enhances the translation of JAK2 and MCM8 to promote NSCLC progression and resistance to cisplatin, respectively. These findings suggest that DAZL is a potential therapeutic target in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Cisplatin , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Janus Kinase 2 , Lung Neoplasms , RNA-Binding Proteins , Humans , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , Animals , Mice , Lung Neoplasms/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Male , Up-Regulation , Cell Line, Tumor , Cell Proliferation/drug effects , Minichromosome Maintenance Proteins/genetics , Minichromosome Maintenance Proteins/metabolism , Disease Progression , Mice, Nude , Xenograft Model Antitumor Assays , Female , Mice, Inbred BALB C
18.
J Ethnopharmacol ; 330: 118196, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-38631488

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Rosmarinic acid (RA), a natural polyphenol abundant in numerous herbal remedies, has been attracting growing interest owing to its exceptional ability to protect the liver. Toosendanin (TSN), a prominent bioactive compound derived from Melia toosendan Siebold & Zucc., boasts diverse pharmacological properties. Nevertheless, TSN possesses remarkable hepatotoxicity. Intriguingly, the potential of RA to counteract TSN-induced liver damage and its probable mechanisms remain unexplored. AIM OF THE STUDY: This study is aimed at exploring whether RA can alleviate TSN-induced liver injury and the potential mechanisms involved autophagy. MATERIALS AND METHODS: CCK-8 and LDH leakage rate assay were used to evaluate cytotoxicity. Balb/c mice were intraperitoneally administered TSN (20 mg/kg) for 24 h after pretreatment with RA (0, 40, 80 mg/kg) by gavage for 5 days. The autophagic proteins P62 and LC3B expressions were detected using western blot and immunohistochemistry. RFP-GFP-LC3B and transmission electron microscopy were applied to observe the accumulation levels of autophagosomes and autolysosomes. LysoTracker Red and DQ-BSA staining were used to evaluate the lysosomal acidity and degradation ability respectively. Western blot, immunohistochemistry and immunofluorescence staining were employed to measure the expressions of JAK2/STAT3/CTSC pathway proteins. Dual-luciferase reporter gene was used to measure the transcriptional activity of CTSC and RT-PCR was used to detect its mRNA level. H&E staining and serum biochemical assay were employed to determine the degree of damage to the liver. RESULTS: TSN-induced damage to hepatocytes and livers was significantly alleviated by RA. RA markedly diminished the autophagic flux blockade and lysosomal dysfunction caused by TSN. Mechanically, RA alleviated TSN-induced down-regulation of CTSC by activating JAK2/STAT3 signaling pathway. CONCLUSION: RA could protect against TSN-induced liver injury by activating the JAK2/STAT3/CTSC pathway-mediated autophagy and lysosomal function.


Subject(s)
Autophagy , Chemical and Drug Induced Liver Injury , Cinnamates , Depsides , Janus Kinase 2 , Lysosomes , Mice, Inbred BALB C , Rosmarinic Acid , STAT3 Transcription Factor , Signal Transduction , Animals , Depsides/pharmacology , STAT3 Transcription Factor/metabolism , Janus Kinase 2/metabolism , Cinnamates/pharmacology , Autophagy/drug effects , Mice , Lysosomes/drug effects , Lysosomes/metabolism , Male , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Signal Transduction/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Drugs, Chinese Herbal/pharmacology , Humans
19.
J Cell Mol Med ; 28(8): e18332, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38661644

ABSTRACT

The role of KIAA0040 role in glioma development is not yet understood despite its connection to nervous system diseases. In this study, KIAA0040 expression levels were evaluated using qRT-PCR, WB and IHC, and functional assays were conducted to assess its impact on glioma progression, along with animal experiments. Moreover, WB was used to examine the impact of KIAA0040 on the JAK2/STAT3 signalling pathway. Our study found that KIAA0040 was increased in glioma and linked to tumour grade and poor clinical outcomes, serving as an independent prognostic factor. Functional assays showed that KIAA0040 enhances glioma growth, migration and invasion by activating the JAK2/STAT3 pathway. Of course, KIAA0040 enhances glioma growth by preventing tumour cell death and promoting cell cycle advancement. Our findings suggest that targeting KIAA0040 could be an effective treatment for glioma due to its role in promoting aggressive tumour behaviour and poor prognosis.


Subject(s)
Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Glioma , Janus Kinase 2 , STAT3 Transcription Factor , Signal Transduction , Animals , Female , Humans , Male , Mice , Middle Aged , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Glioma/genetics , Glioma/pathology , Glioma/metabolism , Janus Kinase 2/metabolism , Janus Kinase 2/genetics , Mice, Nude , Prognosis , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics
20.
Oncol Res ; 32(5): 943-953, 2024.
Article in English | MEDLINE | ID: mdl-38686052

ABSTRACT

Breast and lung cancers are the leading causes of mortality and most frequently diagnosed cancers in women and men, respectively, worldwide. Although the antitumor activity of chalcones has been extensively studied, the molecular mechanisms of isoliquiritigenin analog 2', 4', 4-trihydroxychalcone (metochalcone; TEC) against carcinomas remain less well understood. In this study, we found that TEC inhibited cell proliferation of breast cancer BT549 cells and lung cancer A549 cells in a concentration-dependent manner. TEC induced cell cycle arrest in the S-phase, cell migration inhibition in vitro, and reduced tumor growth in vivo. Moreover, transcriptomic analysis revealed that TEC modulated the activity of the JAK2/STAT3 and P53 pathways. TEC triggered the senescence-associated secretory phenotype (SASP) by repressing the JAK2/STAT3 axis. The mechanism of metochalcone against breast cancer depended on the induction of SASP via deactivation of the JAK2/STAT3 pathway, highlighting the potential of chalcone in senescence-inducing therapy against carcinomas.


Subject(s)
Breast Neoplasms , Cell Proliferation , Cellular Senescence , Chalcones , Janus Kinase 2 , STAT3 Transcription Factor , Signal Transduction , Humans , STAT3 Transcription Factor/metabolism , Janus Kinase 2/metabolism , Janus Kinase 2/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Female , Chalcones/pharmacology , Cell Proliferation/drug effects , Mice , Signal Transduction/drug effects , Animals , Cellular Senescence/drug effects , Cell Line, Tumor , Xenograft Model Antitumor Assays , Cell Movement/drug effects , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...