Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.350
Filter
1.
Front Immunol ; 15: 1385473, 2024.
Article in English | MEDLINE | ID: mdl-38720890

ABSTRACT

Interferons (IFNs) are a family of cytokines that activate the JAK-STAT signaling pathway to induce an antiviral state in cells. Interleukin 27 (IL-27) is a member of the IL-6 and/or IL-12 family that elicits both pro- and anti-inflammatory responses. Recent studies have reported that IL-27 also induces a robust antiviral response against diverse viruses, both in vitro and in vivo, suggesting that IFNs and IL-27 share many similarities at the functional level. However, it is still unknown how similar or different IFN- and IL-27-dependent signaling pathways are. To address this question, we conducted a comparative analysis of the transcriptomic profiles of human monocyte-derived macrophages (MDMs) exposed to IL-27 and those exposed to recombinant human IFN-α, IFN-γ, and IFN-λ. We utilized bioinformatics approaches to identify common differentially expressed genes between the different transcriptomes. To verify the accuracy of this approach, we used RT-qPCR, ELISA, flow cytometry, and microarrays data. We found that IFNs and IL-27 induce transcriptional changes in several genes, including those involved in JAK-STAT signaling, and induce shared pro-inflammatory and antiviral pathways in MDMs, leading to the common and unique expression of inflammatory factors and IFN-stimulated genes (ISGs)Importantly, the ability of IL-27 to induce those responses is independent of IFN induction and cellular lineage. Additionally, functional analysis demonstrated that like IFNs, IL-27-mediated response reduced chikungunya and dengue viruses replication in MDMs. In summary, IL-27 exhibits properties similar to those of all three types of human IFN, including the ability to stimulate a protective antiviral response. Given this similarity, we propose that IL-27 could be classified as a distinct type of IFN, possibly categorized as IFN-pi (IFN-π), the type V IFN (IFN-V).


Subject(s)
Chikungunya virus , Dengue Virus , Dengue , Interferons , Janus Kinases , Macrophages , STAT Transcription Factors , Signal Transduction , Virus Replication , Humans , Chikungunya virus/physiology , Chikungunya virus/immunology , Dengue Virus/physiology , Dengue Virus/immunology , Janus Kinases/metabolism , Virus Replication/drug effects , STAT Transcription Factors/metabolism , Macrophages/immunology , Macrophages/virology , Macrophages/metabolism , Interferons/metabolism , Dengue/immunology , Dengue/virology , Chikungunya Fever/immunology , Chikungunya Fever/virology , Interleukin-27/metabolism , Interleukins/metabolism , Interleukins/pharmacology , Interleukins/immunology , Transcriptome , Cells, Cultured
2.
Front Immunol ; 15: 1385190, 2024.
Article in English | MEDLINE | ID: mdl-38711523

ABSTRACT

The discovery of Suppressor of Cytokine Signaling 1 (SOCS1) in 1997 marked a significant milestone in understanding the regulation of Janus kinase/Signal transducer and activator of transcription (JAK/STAT) signaling pathways. Subsequent research deciphered its cellular functions, and recent insights into SOCS1 deficiencies in humans underscored its critical role in immune regulation. In humans, SOCS-haploinsufficiency (SOCS1-HI) presents a diverse clinical spectrum, encompassing autoimmune diseases, infection susceptibility, and cancer. Variability in disease manifestation, even within families sharing the same genetic variant, raises questions about clinical penetrance and the need for individualized treatments. Current therapeutic strategies include JAK inhibition, with promising results in controlling inflammation in SOCS1-HI patients. Hematopoietic stem cell transplantation and gene therapy emerge as promising avenues for curative treatments. The evolving landscape of SOCS1 research, emphasizes the need for a nuanced understanding of genetic variants and their functional consequences.


Subject(s)
Signal Transduction , Suppressor of Cytokine Signaling 1 Protein , Humans , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/metabolism , Animals , Janus Kinases/metabolism , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/therapy , Haploinsufficiency , STAT Transcription Factors/metabolism , STAT Transcription Factors/genetics , Genetic Therapy
3.
Int J Mol Sci ; 25(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38731900

ABSTRACT

Psoriasis is a highly prevalent dermatological disease associated with an increased systemic inflammatory response. In addition, joint involvement is also present in around 20% of patients. Therefore, treatment modalities used in this condition should be simultaneously effective at improving skin manifestations, reducing inflammation, and addressing psoriatic arthritis when present. Twenty years ago, the introduction of biologic treatments for psoriasis was a turning point in the management of this condition, offering an effective and reasonably safe option for patients whose disease could not be adequately controlled with conventional therapies. At the moment, Janus Kinase inhibitors (JAKis) are a new class of promising molecules in the management of psoriasis. They are orally administered and can show benefits in patients who failed biologic therapy. We conducted a scoping review in order to identify randomized-controlled trials that investigated different JAKis in patients with plaque psoriasis and psoriatic arthritis, with an emphasis on molecules that have been approved by the European Medicines Agency and the Food and Drug Administration. The added value of this study is that it collected information about JAKis approved for two different indications, plaque psoriasis and psoriatic arthritis, in order to provide an integrated understanding of the range of effects that JAKis have on the whole spectrum of psoriasis manifestations.


Subject(s)
Janus Kinase Inhibitors , Janus Kinases , Psoriasis , STAT Transcription Factors , Signal Transduction , Humans , Psoriasis/drug therapy , Psoriasis/metabolism , Janus Kinase Inhibitors/therapeutic use , Janus Kinase Inhibitors/pharmacology , Janus Kinases/metabolism , Janus Kinases/antagonists & inhibitors , Signal Transduction/drug effects , STAT Transcription Factors/metabolism , Arthritis, Psoriatic/drug therapy , Arthritis, Psoriatic/metabolism
4.
Yakugaku Zasshi ; 144(5): 489-496, 2024.
Article in Japanese | MEDLINE | ID: mdl-38692922

ABSTRACT

The tumor necrosis factor receptor (TNFR)-associated factor (TRAF) family of molecules are intracellular adaptors that regulate cellular signaling through members of the TNFR and Toll-like receptor superfamily. Mammals have seven TRAF molecules numbered sequentially from TRAF1 to TRAF7. Although TRAF5 was identified as a potential regulator of TNFR superfamily members, the in vivo function of TRAF5 has not yet been fully elucidated. We identified an unconventional role of TRAF5 in interleukin-6 (IL-6) receptor signaling involving CD4+ T cells. Moreover, TRAF5 binds to the signal-transducing glycoprotein 130 (gp130) receptor for IL-6 and inhibits the activity of the janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling pathway. In addition, Traf5-deficient CD4+ T cells exhibit significantly enhanced IL-6-driven differentiation of T helper 17 (Th17) cells, which exacerbates neuroinflammation in experimental autoimmune encephalomyelitis. Furthermore, TRAF5 demonstrates a similar activity to gp130 for IL-27, another cytokine of the IL-6 family. Additionally, Traf5-deficient CD4+ T cells display significantly increased IL-27-mediated differentiation of Th1 cells, which increases footpad swelling in delayed-type hypersensitivity response. Thus, TRAF5 functions as a negative regulator of gp130 in CD4+ T cells. This review aimed to explain how TRAF5 controls the differentiation of CD4+ T cells and discuss how the expression of TRAF5 in T cells and other cell types can influence the development and progression of autoimmune and inflammatory diseases.


Subject(s)
CD4-Positive T-Lymphocytes , Encephalomyelitis, Autoimmune, Experimental , Signal Transduction , TNF Receptor-Associated Factor 5 , Humans , Animals , TNF Receptor-Associated Factor 5/genetics , TNF Receptor-Associated Factor 5/metabolism , TNF Receptor-Associated Factor 5/physiology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Cytokine Receptor gp130/physiology , Cytokine Receptor gp130/metabolism , Th17 Cells/immunology , Interleukin-6/metabolism , Interleukin-6/physiology , Cell Differentiation , Receptors, Interleukin-6/physiology , Receptors, Interleukin-6/metabolism , Janus Kinases/metabolism , Janus Kinases/physiology , STAT Transcription Factors/physiology , STAT Transcription Factors/metabolism , Mice
5.
Cardiovasc Toxicol ; 24(6): 576-586, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38691302

ABSTRACT

Hypertension is a pathological state of the metabolic syndrome that increases the risk of cardiovascular disease. Managing hypertension is challenging, and we aimed to identify the pathogenic factors and discern therapeutic targets for metabolic hypertension (MHR). An MHR rat model was established with the combined treatment of a high-sugar, high-fat diet and ethanol. Histopathological observations were performed using hematoxylin-eosin and Sirius Red staining. Transcriptome sequencing was performed to screen differentially expressed genes. The role of ubiquitin-specific protease 18 (USP18) in the proliferation, apoptosis, and oxidative stress of HUVECs was explored using Cell Counting Kit-8, flow cytometry, and enzyme-linked immunosorbent assays. Moreover, USP18 downstream signaling pathways in MHR were screened, and the effects of USP18 on these signaling pathways were investigated by western blotting. In the MHR model, total cholesterol and low-density lipoprotein levels increased, while high-density lipoprotein levels decreased. Moreover, high vessel thickness and percentage of collagen were noted along with increased malondialdehyde, decreased superoxide dismutase and catalase levels. The staining results showed that the MHR model exhibited an irregular aortic intima and disordered smooth muscle cells. There were 78 differentially expressed genes in the MHR model, and seven hub genes, including USP18, were identified. USP18 overexpression facilitated proliferation and reduced apoptosis and oxidative stress in HUVECs treated with Ang in vitro. In addition, the JAK/STAT pathway was identified as a USP18 downstream signaling pathway, and USP18 overexpression inhibited the expression of JAK/STAT pathway-related proteins. Conclusively, USP18 restrained MHR progression by promoting cell proliferation, reversing apoptosis and oxidative stress, and suppressing the JAK/STAT pathway.


Subject(s)
Apoptosis , Cell Proliferation , Disease Models, Animal , Human Umbilical Vein Endothelial Cells , Hypertension , Janus Kinases , Metabolic Syndrome , Oxidative Stress , Signal Transduction , Ubiquitin Thiolesterase , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Animals , Humans , Cell Proliferation/drug effects , Apoptosis/drug effects , Oxidative Stress/drug effects , Janus Kinases/metabolism , Male , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Hypertension/metabolism , Hypertension/physiopathology , Hypertension/pathology , Hypertension/enzymology , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Metabolic Syndrome/enzymology , Blood Pressure/drug effects , Disease Progression , Vascular Remodeling/drug effects , STAT Transcription Factors/metabolism , Cells, Cultured , Rats, Sprague-Dawley , Gene Expression Regulation , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Rats
6.
Cells ; 13(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38727296

ABSTRACT

Derangement of the epidermal barrier lipids and dysregulated immune responses are key pathogenic features of atopic dermatitis (AD). The Th2-type cytokines interleukin IL-4 and IL-13 play a prominent role in AD by activating the Janus Kinase/Signal Transduction and Activator of Transcription (JAK/STAT) intracellular signaling axis. This study aimed to investigate the role of JAK/STAT in the lipid perturbations induced by Th2 signaling in 3D epidermal equivalents. Tofacitinib, a low-molecular-mass JAK inhibitor, was used to screen for JAK/STAT-mediated deregulation of lipid metabolism. Th2 cytokines decreased the expression of elongases 1, 3, and 4 and serine-palmitoyl-transferase and increased that of sphingolipid delta(4)-desaturase and carbonic anhydrase 2. Th2 cytokines inhibited the synthesis of palmitoleic acid and caused depletion of triglycerides, in association with altered phosphatidylcholine profiles and fatty acid (FA) metabolism. Overall, the ceramide profiles were minimally affected. Except for most sphingolipids and very-long-chain FAs, the effects of Th2 on lipid pathways were reversed by co-treatment with tofacitinib. An increase in the mRNA levels of CPT1A and ACAT1, reduced by tofacitinib, suggests that Th2 cytokines promote FA beta-oxidation. In conclusion, pharmacological inhibition of JAK/STAT activation prevents the lipid disruption caused by the halted homeostasis of FA metabolism.


Subject(s)
Cytokines , Janus Kinases , Lipid Metabolism , STAT Transcription Factors , Th2 Cells , Humans , Th2 Cells/metabolism , Th2 Cells/drug effects , STAT Transcription Factors/metabolism , Janus Kinases/metabolism , Cytokines/metabolism , Lipid Metabolism/drug effects , Epidermis/metabolism , Epidermis/drug effects , Signal Transduction/drug effects , Piperidines/pharmacology , Pyrimidines/pharmacology , Janus Kinase Inhibitors/pharmacology , Interleukin-4/metabolism , Fatty Acids/metabolism
7.
Cell Commun Signal ; 22(1): 203, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38566182

ABSTRACT

BACKGROUND: The metabolically demanding nature of immune response requires nutrients to be preferentially directed towards the immune system at the expense of peripheral tissues. We study the mechanisms by which this metabolic reprograming occurs using the parasitoid infection of Drosophila larvae. To overcome such an immune challenge hemocytes differentiate into lamellocytes, which encapsulate and melanize the parasitoid egg. Hemocytes acquire the energy for this process by expressing JAK/STAT ligands upd2 and upd3, which activates JAK/STAT signaling in muscles and redirects carbohydrates away from muscles in favor of immune cells. METHODS: Immune response of Drosophila larvae was induced by parasitoid wasp infestation. Carbohydrate levels, larval locomotion and gene expression of key proteins were compared between control and infected animals. Efficacy of lamellocyte production and resistance to wasp infection was observed for RNAi and mutant animals. RESULTS: Absence of upd/JAK/STAT signaling leads to an impaired immune response and increased mortality. We demonstrate how JAK/STAT signaling in muscles leads to suppression of insulin signaling through activation of ImpL2, the inhibitor of Drosophila insulin like peptides. CONCLUSIONS: Our findings reveal cross-talk between immune cells and muscles mediates a metabolic shift, redirecting carbohydrates towards immune cells. We emphasize the crucial function of muscles during immune response and show the benefits of insulin resistance as an adaptive mechanism that is necessary for survival.


Subject(s)
Drosophila Proteins , Insulin Resistance , Wasps , Animals , Transcription Factors/metabolism , Drosophila Proteins/metabolism , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Drosophila/genetics , Muscles , Wasps/metabolism , Larva/metabolism , Immunity , Carbohydrates , Insulin-Like Growth Factor Binding Proteins/metabolism
8.
Sci Rep ; 14(1): 8762, 2024 04 16.
Article in English | MEDLINE | ID: mdl-38627442

ABSTRACT

Metastatic colorectal cancer (CRC) is still in need of effective treatments. This study applies a holistic approach to propose new targets for treatment of primary and liver metastatic CRC and investigates their therapeutic potential in-vitro. An integrative analysis of primary and metastatic CRC samples was implemented for alternative target and treatment proposals. Integrated microarray samples were grouped based on a co-expression network analysis. Significant gene modules correlated with primary CRC and metastatic phenotypes were identified. Network clustering and pathway enrichments were applied to gene modules to prioritize potential targets, which were shortlisted by independent validation. Finally, drug-target interaction search led to three agents for primary and liver metastatic CRC phenotypes. Hesperadin and BAY-1217389 suppress colony formation over a 14-day period, with Hesperadin showing additional efficacy in reducing cell viability within 48 h. As both candidates target the G2/M phase proteins NEK2 or TTK, we confirmed their anti-proliferative properties by Ki-67 staining. Hesperadinin particular arrested the cell cycle at the G2/M phase. IL-29A treatment reduced migration and invasion capacities of TGF-ß induced metastatic cell lines. In addition, this anti-metastatic treatment attenuated TGF-ß dependent mesenchymal transition. Network analysis suggests IL-29A induces the JAK/STAT pathway in a preventive manner.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Indoles , Liver Neoplasms , Rectal Neoplasms , Sulfonamides , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Transcriptome , Janus Kinases/metabolism , Signal Transduction , STAT Transcription Factors/metabolism , Colonic Neoplasms/genetics , Rectal Neoplasms/genetics , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Transforming Growth Factor beta/metabolism , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , NIMA-Related Kinases/genetics
9.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(3): 541-552, 2024 Mar 20.
Article in Chinese | MEDLINE | ID: mdl-38597446

ABSTRACT

OBJECTIVE: To investigate the role of JAK1/STAT3/KHSRP axis in mediating the regulatory effect of LINC00626 on progression of esophagogastric junction adenocarcinoma. METHODS: We collected surgical tumor and adjacent tissue specimens from 64 patients with esophagogastric junction adenocarcinoma and examined the expression levels of LINC00626 and KHSRP. qRT-PCR was used to detect the expressions of LINC00626 and KHSRP in 6 esophageal adenocarcinoma cell lines (OE-19, TE-7, Bic-1, Flo-1, SK-GT-4, and BE-3) and a normal esophageal epithelial cell line (HET-1A). OE-19 and TE-7 cell lines with stable LINC00626 knockdown and FLO-1 and SK-GT-4 cells stably overexpressing LINC00626 were constructed by lentiviral transfection, and the changes in proliferation, migration and invasion of the cells were evaluated using Cell Counting Kit-8 (CCK-8) assay and Transwell migration/invasion assay. The expressions of KHSRP and JAK/STAT pathway proteins in the transfected cells were detected with Western blotting. The effects of LINC006266 knockdown and overexpression on subcutaneous tumor formation and lung metastasis of OE-19 and FLO-1 cell xenografts were tested in nude mice. RESULTS: The expression levels of LINC00626 and KHSRP were significantly increased in esophagogastric junction adenocarcinoma tissues and in esophageal adenocarcinoma cells. LINC00626 knockdown obviously inhibited the proliferation, migration and invasion of esophageal adenocarcinoma cells in vitro and decreased their tumor formation and lung metastasis abilities in nude mice, while overexpression of LINC00626 produced the opposite effects. In esophageal adenocarcinoma cells, LINC0626 knockdown significantly decreased and LINC00626 overexpression strongly enhanced the phosphorylation of JAK1 and STAT3. CONCLUSION: High LINC00626 expression promotes esophageal-gastric junction adenocarcinoma metastasis by activating the JAK1/STAT3/KHSRP signal axis.


Subject(s)
Adenocarcinoma , Esophageal Neoplasms , Janus Kinase 1 , Lung Neoplasms , RNA-Binding Proteins , Animals , Humans , Mice , Adenocarcinoma/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Esophagogastric Junction/metabolism , Esophagogastric Junction/pathology , Gene Expression Regulation, Neoplastic , Janus Kinases/metabolism , Lung Neoplasms/metabolism , Mice, Nude , Signal Transduction , STAT Transcription Factors/metabolism , STAT3 Transcription Factor/metabolism , Trans-Activators , RNA, Long Noncoding/genetics
10.
Nat Immunol ; 25(5): 847-859, 2024 May.
Article in English | MEDLINE | ID: mdl-38658806

ABSTRACT

Immune cells need to sustain a state of constant alertness over a lifetime. Yet, little is known about the regulatory processes that control the fluent and fragile balance that is called homeostasis. Here we demonstrate that JAK-STAT signaling, beyond its role in immune responses, is a major regulator of immune cell homeostasis. We investigated JAK-STAT-mediated transcription and chromatin accessibility across 12 mouse models, including knockouts of all STAT transcription factors and of the TYK2 kinase. Baseline JAK-STAT signaling was detected in CD8+ T cells and macrophages of unperturbed mice-but abrogated in the knockouts and in unstimulated immune cells deprived of their normal tissue context. We observed diverse gene-regulatory programs, including effects of STAT2 and IRF9 that were independent of STAT1. In summary, our large-scale dataset and integrative analysis of JAK-STAT mutant and wild-type mice uncovered a crucial role of JAK-STAT signaling in unstimulated immune cells, where it contributes to a poised epigenetic and transcriptional state and helps prepare these cells for rapid response to immune stimuli.


Subject(s)
Homeostasis , Janus Kinases , Macrophages , Mice, Knockout , STAT Transcription Factors , Signal Transduction , Animals , Mice , Macrophages/immunology , Macrophages/metabolism , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , Mice, Inbred C57BL , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , TYK2 Kinase/metabolism , TYK2 Kinase/genetics , Gene Expression Regulation
12.
Fish Shellfish Immunol ; 149: 109589, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38685444

ABSTRACT

Members of the Signal Transducer and Activator of Transcription (STAT) family function pivotally as transcriptional activators integral to the modulation of inflammatory responses. The aquaculture of silver pomfret is frequently compromised by the imposition of exogenous stressors, which include thermal fluctuations, notably low-temperatures, diminished oxygen levels, and the onslaught of bacterial pathogens. Notwithstanding the critical impact of these stressors, the scientific literature presents a notable gap in our understanding of the STAT pathway's role in the silver pomfret's adaptive response mechanisms. To address this lacuna, we identified stat genes in the silver pomfret-denominated as Pastat1, Pastat2, Pastat3, Pastat4, and Pastat5-through a thorough and systematic bioinformatics analysis. Further scrutiny of the gene configurations and constituent motifs has elucidated that STAT proteins possess analogous structural frameworks and exhibit significant evolutionary preservation. Subsequently, the expression patterns of five stat genes were verified by RT-qPCR in twelve different tissues and four growth periods in healthy fish, showing that the expression of Pastat genes was temporally and spatially specific, with most of the stat genes expressed at higher levels in the spleen, following muscle, gill, and liver. Transcriptomic analysis of exposure to exogenous stressors, specifically formaldehyde and low-temperature conditions, elucidated that Pastat1 and Pastat2 genes exhibited a heightened sensitivity to these environmental challenges. RT-qPCR assays demonstrated a marked alteration in the expression profiles of jak1 and Pastat gene suites in PaS upon prolonged bacterial infection subsequent to these exogenous insults. Moreover, the gene expression of the downstream effectors involved in innate immunity and apoptosis displayed marked deviations. This study additionally elucidated the Pastat gene family's role in modulating the innate immune response and apoptotic regulation within the silver pomfret during exogenous stressors and subsequent pathogenic incursions.


Subject(s)
Fish Diseases , Fish Proteins , Immunity, Innate , Perciformes , STAT Transcription Factors , Stress, Physiological , Animals , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/chemistry , Fish Diseases/immunology , Perciformes/immunology , Perciformes/genetics , Immunity, Innate/genetics , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Gene Expression Regulation/immunology , Gene Expression Regulation/drug effects , Gene Expression Profiling/veterinary , Phylogeny , Sequence Alignment/veterinary , Vibrio Infections/immunology , Vibrio Infections/veterinary , Amino Acid Sequence
13.
Fish Shellfish Immunol ; 149: 109560, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38615702

ABSTRACT

The JAK (Janus kinase)-STAT (Signal transducer and activator of transcription) is a well-known functional signaling pathway that plays a key role in several important biological activities such as apoptosis, cell proliferation, differentiation, and immunity. However, limited studies have explored the functions of STAT genes in invertebrates. In the present study, the gene sequences of two STAT genes from the Pacific oyster (Crassostrea gigas), termed CgSTAT-Like-1 (CgSTAT-L1) and CgSTAT-Like-2 (CgSTAT-L2), were obtained using polymerase chain reaction (PCR) amplification and cloning. Multiple sequence comparisons revealed that the sequences of crucial domains of these proteins were conserved, and the similarity with the protein sequence of other molluscan STAT is close to 90 %. The phylogenetic analyses indicated that CgSTAT-L1 and CgSTAT-L2 are novel members of the mollusk STAT family. Quantitative real-time PCR results implied that CgSTAT-L1 and CgSTAT-L2 mRNA expression was found in all tissues, and significantly induced after challenge with lipopolysaccharide (LPS), peptidoglycan (PGN), or poly(I:C). After that, dual-luciferase reporter assays denoted that overexpression of CgSTAT-L1 and CgSTAT-L2 significantly activated the NF-κB signaling, and, interestingly, the overexpressed CgSTAT proteins potentiated LPS-induced NF-κB activation. These results contributed a preliminary analysis of the immune-related function of STAT genes in oysters, laying the foundation for deeper understanding of the function of invertebrate STAT genes.


Subject(s)
Amino Acid Sequence , Crassostrea , Phylogeny , STAT Transcription Factors , Sequence Alignment , Animals , Crassostrea/genetics , Crassostrea/immunology , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Sequence Alignment/veterinary , Lipopolysaccharides/pharmacology , Immunity, Innate/genetics , Peptidoglycan/pharmacology , Poly I-C/pharmacology , Base Sequence , Gene Expression Regulation/immunology , Gene Expression Regulation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , DNA, Complementary/genetics , Cloning, Molecular , Signal Transduction
14.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(3): 465-473, 2024 Mar 20.
Article in Chinese | MEDLINE | ID: mdl-38597437

ABSTRACT

OBJECTIVE: To explore the therapeutic mechanism of Jianpi Zishen (JPZS) granules for systemic lupus erythematosus(SLE) in light of podocyte autophagy regulation. METHODS: TCMSP, GeneCards, OMIM, and TTD databases were used to obtain the targets of JPZS granules, SLE, and podocyte autophagy. The protein-protein interaction network was constructed using Cytoscape, and the key active ingredients and targets were screened for molecular docking. In the clinical study, 46 patients with SLE were randomized into two groups to receive baseline treatment with prednisone acetate and mycophenolate mofetil (control group) and additional treatment with JPZS granules (observation group) for 12 weeks, with 10 healthy volunteers as the healthy control group. Urinary levels of nephrin and synaptopodin of the patients were detected with ELISA. Western blotting was performed to determine peripheral blood levels of p-JAK1/JAK1, p-STAT1/STAT1, LC3II/LC3I, and p62 proteins of the participants. RESULTS: Four key active ingredients and 5 core target genes (STAT1, PIK3CG, MAPK1, PRKCA, and CJA1) were obtained, and enrichment analysis identified the potentially involved signaling pathways including AGE-RAGE, JAK/STAT, EGFR, and PI3K/Akt. Molecular docking analysis showed that STAT1 was the most promising target protein with the highest binding activity, suggesting its role as an important mediator for signal transduction after JPZS granule treatment. In the 43 SLE patients available for analysis, treatment with JPZS granule significantly reduced serum levels of p-JAK1/JAK1, p-STAT1/STAT1, and LC3II/LC3I (P < 0.05 or 0.01), increased the protein level of P62 (P < 0.05), and reduced urinary levels of nephrin and synaptopodin (P < 0.05). CONCLUSION: The therapeutic effect of JPZS granules on SLE is mediated probably by coordinated actions of quercetin, kaempferol, ß-sitosterol, and isorhamnetin on their target gene STAT1 to inhibit the JAK/STAT pathway, thus suppressing autophagy and alleviating podocyte injuries in SLE.


Subject(s)
Drugs, Chinese Herbal , Lupus Erythematosus, Systemic , Podocytes , Humans , Autophagy , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Janus Kinases/metabolism , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/metabolism , Molecular Docking Simulation , Network Pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Podocytes/metabolism , Signal Transduction , STAT Transcription Factors/metabolism
15.
Environ Pollut ; 349: 123966, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38621451

ABSTRACT

Polybrominated diphenyl ethers (PBDEs) are widely present in water ecosystems where they pose a significant threat to aquatic life, but our knowledge about how PBDEs affect feeding is limited. Therefore, this study explored the effects of continuous dietary exposure to 2,2',4,4'-tetrabromodiphenyl ether (BDE-47) (40 and 4000 ng/g) on the feeding in common carp (Cyprinus carpio) and the underlying mechanism. BDE-47 significantly decreased the food intake of carp. Transcriptome analysis of brain tissue showed that BDE-47 mainly affected the nervous, immune, and endocrine systems. Further examination of the expression levels of appetite factors in the brain revealed that BDE-47 caused dysregulation of appetite factors expressions such as agrp, pomc, cart, etc. In addition, the JAK-STAT signaling pathway was activated under BDE-47 exposure. It can be concluded from these findings that BDE-47 activated the JAK-STAT signaling pathway, causing imbalanced expression of appetite factors, leading to disordered feeding behavior and decreased food intake in carp. These results provide an important reference for a more comprehensive understanding of the hazards posed by BDE-47 on animal feeding and the associated mechanisms.


Subject(s)
Carps , Dietary Exposure , Halogenated Diphenyl Ethers , Janus Kinases , Signal Transduction , Water Pollutants, Chemical , Animals , Halogenated Diphenyl Ethers/toxicity , Carps/metabolism , Carps/physiology , Signal Transduction/drug effects , Water Pollutants, Chemical/toxicity , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Feeding Behavior/drug effects
16.
Nature ; 629(8012): 688-696, 2024 May.
Article in English | MEDLINE | ID: mdl-38658752

ABSTRACT

Although cancer initiation and progression are generally associated with the accumulation of somatic mutations1,2, substantial epigenomic alterations underlie many aspects of tumorigenesis and cancer susceptibility3-6, suggesting that genetic mechanisms might not be the only drivers of malignant transformation7. However, whether purely non-genetic mechanisms are sufficient to initiate tumorigenesis irrespective of mutations has been unknown. Here, we show that a transient perturbation of transcriptional silencing mediated by Polycomb group proteins is sufficient to induce an irreversible switch to a cancer cell fate in Drosophila. This is linked to the irreversible derepression of genes that can drive tumorigenesis, including members of the JAK-STAT signalling pathway and zfh1, the fly homologue of the ZEB1 oncogene, whose aberrant activation is required for Polycomb perturbation-induced tumorigenesis. These data show that a reversible depletion of Polycomb proteins can induce cancer in the absence of driver mutations, suggesting that tumours can emerge through epigenetic dysregulation leading to inheritance of altered cell fates.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Epigenesis, Genetic , Janus Kinases , Neoplasms , Polycomb-Group Proteins , STAT Transcription Factors , Animals , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Polycomb-Group Proteins/metabolism , Polycomb-Group Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , STAT Transcription Factors/metabolism , STAT Transcription Factors/genetics , Janus Kinases/metabolism , Janus Kinases/genetics , Female , Carcinogenesis/genetics , Male , Signal Transduction/genetics , Gene Silencing , Cell Transformation, Neoplastic/genetics , Cell Lineage/genetics , Gene Expression Regulation, Neoplastic
17.
J Cardiovasc Pharmacol Ther ; 29: 10742484241248046, 2024.
Article in English | MEDLINE | ID: mdl-38656132

ABSTRACT

Atherosclerosis is now widely considered to be a chronic inflammatory disease, with increasing evidence suggesting that lipid alone is not the main factor contributing to its development. Rather, atherosclerotic plaques contain a significant amount of inflammatory cells, characterized by the accumulation of monocytes and lymphocytes on the vessel wall. This suggests that inflammation may play a crucial role in the occurrence and progression of atherosclerosis. As research deepens, other pathological factors have also been found to influence the development of the disease. The Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway is a recently discovered target of inflammation that has gained attention in recent years. Numerous studies have provided evidence for the causal role of this pathway in atherosclerosis, and its downstream signaling factors play a significant role in this process. This brief review aims to explore the crucial role of the JAK/STAT pathway and its representative downstream signaling factors in the development of atherosclerosis. It provides a new theoretical basis for clinically affecting the development of atherosclerosis by interfering with the JAK/STAT signaling pathway.


Subject(s)
Atherosclerosis , Janus Kinases , STAT Transcription Factors , Signal Transduction , Humans , Atherosclerosis/metabolism , Atherosclerosis/drug therapy , STAT Transcription Factors/metabolism , Janus Kinases/metabolism , Animals , Janus Kinase Inhibitors/therapeutic use , Janus Kinase Inhibitors/pharmacology , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Inflammation Mediators/metabolism
18.
Sci Rep ; 14(1): 7292, 2024 03 27.
Article in English | MEDLINE | ID: mdl-38538691

ABSTRACT

Glioblastoma multiforme (GBM) IDH-wildtype is the most prevalent brain malignancy in adults. However, molecular mechanisms, which leads to GBM have not been completely elucidated. Granulocyte colony-stimulating factor (GCSF), Granulocyte colony-stimulating factor receptor GCSFR, and Signal transducers and activators of transcription 3 (STAT3) have been involved in the occurrence and development of various cancers, but their role in GBM is little known. Herein, we have investigated the gene and protein expression of GCSF, GCSFR, and STAT3 in 21 tissue biopsy samples and also in tumor associated normal tissue (TANT) samples derived from glioblastoma patients, which revealed significantly differential expression of these genes. To validate our findings, we performed a comprehensive integrated analysis of transcriptomic and proteomic profiling of respective genes by retrieving GBM RNA-sequence data from Genome Atlas Databases. GO and KEGG analysis revealed enrichment in disease-related pathways, such as JAK/STAT pathway activation, which were associated with GBM progression. We further performed computational docking analysis of potential drug candidate Nisin against GCSF, and the results were validated in vitro through cytotoxic activity assay using a human glioblastoma cell line SF-767 in a dose-dependent manner. Our comprehensive analysis reveals that GCSF augments glioma progression, and its blockade with anticancer bacteriocin peptide Nisin can potentially inhibit the growth and metastasis of GBM.


Subject(s)
Brain Neoplasms , Glioblastoma , Nisin , Adult , Humans , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Nisin/metabolism , Janus Kinases/metabolism , Proteomics , Signal Transduction , STAT Transcription Factors/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Granulocyte Colony-Stimulating Factor/metabolism , Gene Expression Regulation, Neoplastic
19.
Breast Cancer Res ; 26(1): 54, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38553760

ABSTRACT

Fibroblast growth factors (FGFs) control various cellular functions through fibroblast growth factor receptor (FGFR) activation, including proliferation, differentiation, migration, and survival. FGFR amplification in ER + breast cancer patients correlate with poor prognosis, and FGFR inhibitors are currently being tested in clinical trials. By comparing three-dimensional spheroid growth of ER + breast cancer cells with and without FGFR1 amplification, our research discovered that FGF2 treatment can paradoxically decrease proliferation in cells with FGFR1 amplification or overexpression. In contrast, FGF2 treatment in cells without FGFR1 amplification promotes classical FGFR proliferative signaling through the MAPK cascade. The growth inhibitory effect of FGF2 in FGFR1 amplified cells aligned with an increase in p21, a cell cycle inhibitor that hinders the G1 to S phase transition in the cell cycle. Additionally, FGF2 addition in FGFR1 amplified cells activated JAK-STAT signaling and promoted a stem cell-like state. FGF2-induced paradoxical effects were reversed by inhibiting p21 or the JAK-STAT pathway and with pan-FGFR inhibitors. Analysis of patient ER + breast tumor transcriptomes from the TCGA and METABRIC datasets demonstrated a strong positive association between expression of FGF2 and stemness signatures, which was further enhanced in tumors with high FGFR1 expression. Overall, our findings reveal a divergence in FGFR signaling, transitioning from a proliferative to stemness state driven by activation of JAK-STAT signaling and modulation of p21 levels. Activation of these divergent signaling pathways in FGFR amplified cancer cells and paradoxical growth effects highlight a challenge in the use of FGFR inhibitors in cancer treatment.


Subject(s)
Breast Neoplasms , Signal Transduction , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 2/therapeutic use , Janus Kinases/metabolism , Janus Kinases/pharmacology , Janus Kinases/therapeutic use , STAT Transcription Factors/metabolism , STAT Transcription Factors/pharmacology , STAT Transcription Factors/therapeutic use , Receptor, Fibroblast Growth Factor, Type 1 , Cell Proliferation , Fibroblast Growth Factors/pharmacology , Cell Line, Tumor
20.
Front Immunol ; 15: 1341981, 2024.
Article in English | MEDLINE | ID: mdl-38464510

ABSTRACT

Axial spondyloarthritis (axSpA) is a chronic inflammatory disease that is characterized by new bone formation in the axial musculoskeletal system, with X-ray discriminating between radiographic and non-radiographic forms. Current therapeutic options include non-steroidal anti-inflammatory drugs in addition to biological disease-modifying anti-rheumatic drugs that specifically target tumor necrosis factor-alpha (TNFα) or interleukin (IL)-17. Pain is the most critical symptom for axSpA patients, significantly contributing to the burden of disease and impacting daily life. While the inflammatory process exerts a major role in determining pain in the early phases of the disease, the symptom may also result from mechanical and neuromuscular causes that require complex, multi-faceted pharmacologic and non-pharmacologic treatment, especially in the later phases. In clinical practice, pain often persists and does not respond further despite the absence of inflammatory disease activity. Cytokines involved in axSpA pathogenesis interact directly/indirectly with the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling cascade, a fundamental component in the origin and development of spondyloarthropathies. The JAK/STAT pathway also plays an important role in nociception, and new-generation JAK inhibitors have demonstrated rapid pain relief. We provide a comprehensive review of the different pain types observed in axSpA and the potential role of JAK/STAT signaling in this context, with specific focus on data from preclinical studies and data from clinical trials with JAK inhibitors.


Subject(s)
Axial Spondyloarthritis , Janus Kinase Inhibitors , Humans , Janus Kinases/metabolism , Signal Transduction , Janus Kinase Inhibitors/therapeutic use , STAT Transcription Factors/metabolism , Pain
SELECTION OF CITATIONS
SEARCH DETAIL
...