Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.449
Filtrer
1.
Front Immunol ; 15: 1430938, 2024.
Article de Anglais | MEDLINE | ID: mdl-39114664

RÉSUMÉ

Severe aplastic anemia (SAA) is a life-threatening bone marrow failure syndrome whose development can be triggered by environmental, autoimmune, and/or genetic factors. The latter comprises germ line pathogenic variants in genes that bring about habitually predisposing syndromes as well as immune deficiencies that do so only occasionally. One of these disorders is the autosomal dominant form of chronic mucocutaneous candidiasis (CMC), which is defined by germ line STAT1 gain-of-function (GOF) pathogenic variants. The resultant overexpression and constitutive activation of STAT1 dysregulate the Janus kinase/signal transducer and activator of transcription 1 (STAT) signaling pathway, which normally organizes the development and proper interaction of different components of the immunologic and hematopoietic system. Although SAA is an extremely rare complication in this disorder, it gained a more widespread interest when it became clear that the underlying causative pathomechanism may, in a similar fashion, also be instrumental in at least some of the idiopathic SAA cases. Based on these premises, we present herein what is the historically most likely first cord blood-transplanted SAA case in a CMC family with a documented STAT1 GOF pathogenic variant. In addition, we recapitulate the characteristics of the six CMC SAA cases that have been reported so far and discuss the significance of STAT1 GOF pathogenic variants and other STAT1 signaling derangements in the context of these specific types of bone marrow failure syndromes. Because a constitutively activated STAT1 signaling, be it driven by STAT1 GOF germ line pathogenic variants or any other pathogenic variant-independent events, is apparently important for initiating and maintaining the SAA disease process, we propose to acknowledge that SAA is one of the definite disease manifestations in STAT1-mutated CMC cases. For the same reason, we deem it necessary to also incorporate molecular and functional analyses of STAT1 into the diagnostic work-up of SAA cases.


Sujet(s)
Anémie aplasique , Candidose mucocutanée chronique , Facteur de transcription STAT-1 , Adulte , Femelle , Humains , Mâle , Anémie aplasique/génétique , Candidose mucocutanée chronique/génétique , Transplantation de cellules souches de sang du cordon , Pedigree , Études rétrospectives , Facteur de transcription STAT-1/génétique , Facteur de transcription STAT-1/métabolisme
2.
Skin Res Technol ; 30(8): e13888, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39099447

RÉSUMÉ

BACKGROUND: Incontinence-associated dermatitis (IAD) is a tough problem in clinical settings, not only increasing the risk of complications like catheter-related urinary tract infections and pressure ulcers in elderly and critically ill patients, but also prolonging hospital stays, raising hospital costs, and possibly leading to medical disputes. This study is aimed to evaluate the therapeutic effect of silicone dressing combined with topical oxygen therapy on IAD in a rat model. METHODS: An IAD rat model induced by synthetic urine with trypsin was established. Hematoxylin & eosin staining was carried out to examine skin histology. Using immunofluorescence, the microvessel density in the affected skin tissues was determined. ELISA was performed to measure the concentrations of inflammatory cytokines and angiogenic factors in serum. The mRNA expression of EGF, PDGF, and VEGF was detected via qRT-PCR. Western blotting was employed to determine NF-κB p65/STAT1 pathway-related protein levels. RESULTS: Compared to single therapy, silicone dressing combined with topical oxygen therapy could significantly reduce the severity of IAD, improve skin histology, inhibit inflammation, and promote angiogenesis in IAD rat models. Additionally, the results showed that relatively speaking, the combined therapy suppressed the NF-κB p65/STAT1 signaling pathway more effectively. CONCLUSION: These findings indicated that silicone dressing combined with topical oxygen therapy can alleviate IAD through promoting wound healing and inhibiting inflammation via NF-κB p65/STAT1 signaling pathway in a rat model, which provided a theoretical basis for the prevention and treatment of IAD in clinic.


Sujet(s)
Bandages , Dermatite , Modèles animaux de maladie humaine , Oxygène , Rat Sprague-Dawley , Facteur de transcription STAT-1 , Transduction du signal , Silicone , Facteur de transcription RelA , Incontinence urinaire , Animaux , Rats , Transduction du signal/effets des médicaments et des substances chimiques , Oxygène/administration et posologie , Facteur de transcription STAT-1/métabolisme , Dermatite/thérapie , Dermatite/étiologie , Facteur de transcription RelA/métabolisme , Incontinence urinaire/thérapie , Incontinence urinaire/étiologie , Mâle
3.
FASEB J ; 38(16): e70001, 2024 Aug 31.
Article de Anglais | MEDLINE | ID: mdl-39139033

RÉSUMÉ

Interferon-gamma (IFNγ) is traditionally recognized for its pro-inflammatory role during intestinal inflammation. Here, we demonstrate that IFNγ also functions as a pro-repair molecule by increasing TNFα receptor 2 (TNFR2 protein/TNFRSF1B gene) expression on intestinal epithelial cells (IEC) following injury in vitro and in vivo. In silico analyses identified binding sites for the IFNγ signaling transcription factor STAT1 in the promoter region of TNFRSF1B. Scratch-wounded IEC exposed to IFNγ exhibited a STAT1-dependent increase in TNFR2 expression. In situ hybridization revealed elevated Tnfrsf1b mRNA levels in biopsy-induced colonic mucosal wounds, while intraperitoneal administration of IFNγ neutralizing antibodies following mucosal injury resulted in impaired IEC Tnfrsf1b mRNA and inhibited colonic mucosal repair. These findings challenge conventional notions that "pro-inflammatory" mediators solely exacerbate damage by highlighting latent pro-repair functions. Moreover, these results emphasize the critical importance of timing and amount in the synthesis and release of IFNγ and TNFα during the inflammatory process, as they are pivotal in restoring tissue homeostasis.


Sujet(s)
Côlon , Interféron gamma , Muqueuse intestinale , Récepteur au facteur de nécrose tumorale de type II , Facteur de transcription STAT-1 , Transduction du signal , Interféron gamma/métabolisme , Récepteur au facteur de nécrose tumorale de type II/métabolisme , Récepteur au facteur de nécrose tumorale de type II/génétique , Animaux , Humains , Côlon/métabolisme , Côlon/anatomopathologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Facteur de transcription STAT-1/métabolisme , Souris , Cicatrisation de plaie/physiologie , Souris de lignée C57BL , Mâle , Cellules épithéliales/métabolisme , Facteur de nécrose tumorale alpha/métabolisme
4.
Front Biosci (Landmark Ed) ; 29(7): 247, 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-39082331

RÉSUMÉ

BACKGROUND: Retinal pigment epithelial (RPE) cells have a pivotal function in preserving the equilibrium of the retina and moderating the immunological interaction between the choroid and the retina. This study primarily focuses on delineating the protective effect offered by Kaempferol (Kae) against RPE cell damage. METHODS: Bioinformatics analysis was performed on the GSE30719 dataset to identify hub genes associated with RPE. Subsequently, we analyzed the impact of Kae on RPE apoptosis, cell viability, and inflammatory response through cell experiments, and explored the interaction between hub genes and Kae. RESULTS: Based on the GSE30719 dataset, nine hub genes (ISG15, IFIT1, IFIT3, STAT1, OASL, RSAD2, IRF7, MX2, and MX1) were identified, all of which were highly expressed in the GSE30719 case group. Kae could boost the proliferative activity of RPE cells caused by lipopolysaccharide (LPS), as well as reduce apoptosis and the generation of inflammatory factors (tumor necrosis factor receptor (TNFR), interleukin-1beta (IL-1ß)) and cytokines (IL-1, IL-6, IL-12). STAT1 was shown to inhibit cell proliferation, promote apoptosis, and secrete IL-1/IL-6/IL-12 in LPS-induced RPE cells. Moreover, IRF7 was found to interact with STAT1 in LPS-induced RPE cells, and STAT1 could maintain IRF7 levels through deubiquitination. In addition, we also found that the protective effect of Kae on LPS-induced RPE cell injury was mediated through STAT1/IRF7 axis. CONCLUSION: This study provided evidence that Kae protects RPE cells via regulating the STAT1/IRF7 signaling pathways, indicating its potential therapeutic relevance in the diagnosis and management of retinal disorders linked with RPE cell damage.


Sujet(s)
Apoptose , Facteur-7 de régulation d'interféron , Kaempférols , Épithélium pigmentaire de la rétine , Facteur de transcription STAT-1 , Ubiquitination , Humains , Épithélium pigmentaire de la rétine/métabolisme , Épithélium pigmentaire de la rétine/effets des médicaments et des substances chimiques , Facteur de transcription STAT-1/métabolisme , Facteur-7 de régulation d'interféron/métabolisme , Facteur-7 de régulation d'interféron/génétique , Ubiquitination/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Kaempférols/pharmacologie , Lignée cellulaire , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Lipopolysaccharides , Protéolyse/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Cytokines/génétique
5.
J Infect Public Health ; 17(8): 102468, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38991411

RÉSUMÉ

Mendelian susceptibility to mycobacterial disease (MSMD) is caused by approximately 21 genetic defects, including a mutation in Interferon-Gamma Receptor 1 (IFNGR1). IFNGR1 deficiency leads to a loss of cellular responsiveness to type II Interferon (IFN-γ), which plays a significant role in controlling intracellular bacteria. This study explored the response of IFN-ß therapy in a patient with partial IFNGR1 deficiency to treat invasive mycobacterial infection. The biological therapy was used successfully as an adjuvant to anti-mycobacterial medications to treat a 17-year-old girl with partial IFNGR1 deficiency who presented with a recurrent mycobacterial infection that extended to her central nervous system, which resulted in clinical and radiological improvement. This report suggests that activation of type I IFN through Signal Transducers and Activators of Transcription1 (STAT1) could bypass the early IFN-γ signaling defects and activate IFN-γ production. For that reason, IFN-ß might be used as a beneficial adjuvant therapy for managing extensive central nervous system mycobacterial infection, especially in patients with IFNGR1 deficiency.


Sujet(s)
, Interféron bêta , Infections à Mycobacterium , Récepteur interféron , Humains , Femelle , Adolescent , Récepteur interféron/déficit , Récepteur interféron/génétique , Interféron bêta/usage thérapeutique , Infections à Mycobacterium/traitement médicamenteux , Résultat thérapeutique , Interféron gamma/génétique , Facteur de transcription STAT-1/génétique , Facteur de transcription STAT-1/métabolisme
6.
Int Immunopharmacol ; 138: 112651, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-38986303

RÉSUMÉ

Peripheral blood mononuclear cells (PBMC), sourced autologously, offer numerous advantages when procured: easier acquisition process, no in vitro amplification needed, decreased intervention and overall increased acceptability make PBMC an attractive candidate for cell therapy treatment. However, the exact mechanism by which PBMC treat diseases remains poorly understood. Immune imbalance is the pathological basis of many diseases, with macrophages playing a crucial role in this process. However, research on the role and mechanisms of PBMC in regulating macrophages remains scarce. This study employed an in vitro co-culture model of PBMC and RAW264.7 macrophages to explore the role and mechanisms of PBMC in regulating macrophages. The results showed that the co-culturing led to decreased expression of inflammatory cytokines and increased expression of anti-inflammatory cytokines in RAW264.7 or in the culture supernatant. Additionally, the pro-inflammatory, tissue matrix-degrading M1 macrophages decreased, while the anti-inflammatory, matrix-synthesizing, regenerative M2 macrophages increased in both RAW264.7 and monocytes within PBMC. Moreover, co-cultured macrophages exhibited a significantly decreased p-STAT1/STAT1 ratio, while the p-STAT6/STAT6 ratio significantly increased. This suggests that PBMC may inhibit M1 macrophage polarization by blocking STAT1 signaling cascades and may promote M2 macrophage polarization through the activation of STAT6 signaling cascades. Overall, this study sheds light on the role and mechanism of PBMC in regulating macrophages. Moreover, it was found that monocytes within co-cultured PBMC differentiated into M2 macrophages in the presence of macrophages. This finding provides experimental evidence for the use of PBMC in treating inflammatory diseases, especially macrophage-depleting inflammatory diseases such as osteoarthritis.


Sujet(s)
Techniques de coculture , Agranulocytes , Macrophages , Facteur de transcription STAT-1 , Facteur de transcription STAT-6 , Transduction du signal , Animaux , Souris , Cytokines/métabolisme , Agranulocytes/immunologie , Agranulocytes/métabolisme , Activation des macrophages , Macrophages/immunologie , Macrophages/métabolisme , Cellules RAW 264.7 , Facteur de transcription STAT-1/métabolisme , Facteur de transcription STAT-6/métabolisme
7.
J Neuroinflammation ; 21(1): 178, 2024 Jul 21.
Article de Anglais | MEDLINE | ID: mdl-39034417

RÉSUMÉ

BACKGROUND: Reactive astrocytes participate in various pathophysiology after subarachnoid hemorrhage (SAH), including neuroinflammation, glymphatic-lymphatic system dysfunction, brain edema, BBB disruption, and cell death. Astrocytes transform into two new reactive phenotypes with changed morphology, altered gene expression, and secretion profiles, termed detrimental A1 and beneficial A2. This study investigates the effect of 67LR activation by PEDF-34, a PEDF peptide, on neuroinflammation and astrocyte polarization after the experimental SAH. METHODS: A total of 318 male adult Sprague-Dawley rats were used in experiments in vivo, of which 272 rats were subjected to the endovascular perforation model of SAH and 46 rats underwent sham surgery. 67LR agonist (PEDF-34) was administrated intranasally 1 h after SAH. 67LR-specific inhibitor (NSC-47924) and STAT1 transcriptional activator (2-NP) were injected intracerebroventricularly 48 h before SAH. Short- and long-term neurological tests, brain water content, immunostaining, Nissl staining, western blot, and ELISA assay were performed. In experiments in vitro, primary astrocyte culture with hemoglobin (Hb) stimulation was used to mimic SAH. The expression of the PEDF-34/67LR signaling pathway and neuro-inflammatory cytokines were assessed using Western blot, ELISA, and immunohistochemistry assays both in vivo and in vitro. RESULTS: Endogenous PEDF and 67LR expressions were significantly reduced at 6 h after SAH. 67LR was expressed in astrocytes and neurons. Intranasal administration of PEDF-34 significantly reduced brain water content, pro-inflammatory cytokines, and short-term and long-term neurological deficits after SAH. The ratio of p-JNK/JNK and p-STAT1/STAT1 and the expression of CFB and C3 (A1 astrocytes marker), significantly decreased after PEDF-34 treatment, along with fewer expression of TNF-α and IL-1ß at 24 h after SAH. However, 2-NP (STAT1 transcriptional activator) and NSC-47924 (67LR inhibitor) reversed the protective effects of PEDF-34 in vivo and in vitro by promoting A1 astrocyte polarization with increased inflammatory cytokines. CONCLUSION: PEDF-34 activated 67LR, attenuating neuroinflammation and inhibiting astrocyte A1 polarization partly via the JNK/STAT1 pathway, suggesting that PEDF-34 might be a potential treatment for SAH patients.


Sujet(s)
Astrocytes , Facteurs de croissance nerveuse , Maladies neuro-inflammatoires , Facteur de transcription STAT-1 , Serpines , Hémorragie meningée , Animaux , Mâle , Rats , Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Polarité de la cellule , Cellules cultivées , Système de signalisation des MAP kinases , Facteurs de croissance nerveuse/métabolisme , Maladies neuro-inflammatoires/traitement médicamenteux , Maladies neuro-inflammatoires/métabolisme , Rat Sprague-Dawley , Serpines/métabolisme , Transduction du signal , Facteur de transcription STAT-1/métabolisme , Hémorragie meningée/traitement médicamenteux , Hémorragie meningée/métabolisme
8.
Front Immunol ; 15: 1397117, 2024.
Article de Anglais | MEDLINE | ID: mdl-39040107

RÉSUMÉ

Intestinal epithelial cells possess the requisite molecular machinery to initiate cell-intrinsic defensive responses against intracellular pathogens, including intracellular parasites. Interferons(IFNs) have been identified as cornerstones of epithelial cell-intrinsic defense against such pathogens in the gastrointestinal tract. Long non-coding RNAs (lncRNAs) are RNA transcripts (>200 nt) not translated into protein and represent a critical regulatory component of mucosal defense. We report here that lncRNA Nostrill facilitates IFN-γ-stimulated intestinal epithelial cell-intrinsic defense against infection by Cryptosporidium, an important opportunistic pathogen in AIDS patients and a common cause of diarrhea in young children. Nostrill promotes transcription of a panel of genes controlled by IFN-γ through facilitating Stat1 chromatin recruitment and thus, enhances expression of several genes associated with cell-intrinsic defense in intestinal epithelial cells in response to IFN-γ stimulation, including Igtp, iNos, and Gadd45g. Induction of Nostrill enhances IFN-γ-stimulated intestinal epithelial defense against Cryptosporidium infection, which is associated with an enhanced autophagy in intestinal epithelial cells. Our findings reveal that Nostrill enhances the transcription of a set of genes regulated by IFN-γ in intestinal epithelial cells. Moreover, induction of Nostrill facilitates the IFN-γ-mediated epithelial cell-intrinsic defense against cryptosporidial infections.


Sujet(s)
Cryptosporidiose , Interféron gamma , Muqueuse intestinale , ARN long non codant , Interféron gamma/métabolisme , ARN long non codant/génétique , Cryptosporidiose/immunologie , Muqueuse intestinale/immunologie , Muqueuse intestinale/parasitologie , Muqueuse intestinale/métabolisme , Animaux , Humains , Transcription génétique , Cellules épithéliales/immunologie , Cellules épithéliales/métabolisme , Cellules épithéliales/parasitologie , Souris , Facteur de transcription STAT-1/métabolisme , Facteur de transcription STAT-1/génétique , Cryptosporidium/génétique , Cryptosporidium/immunologie , Régulation de l'expression des gènes , Autophagie/immunologie
9.
J Neuroinflammation ; 21(1): 166, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956653

RÉSUMÉ

BACKGROUND: Type 2 diabetes mellitus (T2DM) and obstructive sleep apnea (OSA) are mutual risk factors, with both conditions inducing cognitive impairment and anxiety. However, whether OSA exacerbates cognitive impairment and anxiety in patients with T2DM remains unclear. Moreover, TREM2 upregulation has been suggested to play a protective role in attenuating microglia activation and improving synaptic function in T2DM mice. The aim of this study was to explore the regulatory mechanisms of TREM2 and the cognitive and anxiety-like behavioral changes in mice with OSA combined with T2DM. METHODS: A T2DM with OSA model was developed by treating mice with a 60% kcal high-fat diet (HFD) combined with intermittent hypoxia (IH). Spatial learning memory capacity and anxiety in mice were investigated. Neuronal damage in the brain was determined by the quantity of synapses density, the number and morphology of brain microglia, and pro-inflammatory factors. For mechanism exploration, an in vitro model of T2DM combined with OSA was generated by co-treating microglia with high glucose (HG) and IH. Regulation of TREM2 on IFNAR1-STAT1 pathway was determined by RNA sequencing and qRT-PCR. RESULTS: Our results showed that HFD mice exhibited significant cognitive dysfunction and anxiety-like behavior, accompanied by significant synaptic loss. Furthermore, significant activation of brain microglia and enhanced microglial phagocytosis of synapses were observed. Moreover, IH was found to significantly aggravate anxiety in the HFD mice. The mechanism of HG treatment may potentially involve the promotion of TREM2 upregulation, which in turn attenuates the proinflammatory microglia by inhibiting the IFNAR1-STAT1 pathway. Conversely, a significant reduction in TREM2 in IH-co-treated HFD mice and HG-treated microglia resulted in the further activation of the IFNAR1-STAT1 pathway and consequently increased proinflammatory microglial activation. CONCLUSIONS: HFD upregulated the IFNAR1-STAT1 pathway and induced proinflammatory microglia, leading to synaptic damage and causing anxiety and cognitive deficits. The upregulated TREM2 inT2DM mice brain exerted a negative regulation of the IFNAR1-STAT1 pathway. Mice with T2DM combined with OSA exacerbated anxiety via the downregulation of TREM2, causing heightened IFNAR1-STAT1 pathway activation and consequently increasing proinflammatory microglia.


Sujet(s)
Anxiété , Diabète de type 2 , Alimentation riche en graisse , Hypoxie , Glycoprotéines membranaires , Souris de lignée C57BL , Récepteur à l'interféron alpha-bêta , Récepteurs immunologiques , Transduction du signal , Animaux , Souris , Alimentation riche en graisse/effets indésirables , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/génétique , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/génétique , Anxiété/étiologie , Anxiété/métabolisme , Transduction du signal/physiologie , Transduction du signal/effets des médicaments et des substances chimiques , Hypoxie/métabolisme , Hypoxie/complications , Mâle , Diabète de type 2/complications , Diabète de type 2/métabolisme , Diabète de type 2/psychologie , Récepteur à l'interféron alpha-bêta/métabolisme , Récepteur à l'interféron alpha-bêta/génétique , Diabète expérimental/complications , Diabète expérimental/métabolisme , Microglie/métabolisme , Facteur de transcription STAT-1/métabolisme , Syndrome d'apnées obstructives du sommeil/complications , Syndrome d'apnées obstructives du sommeil/métabolisme , Syndrome d'apnées obstructives du sommeil/psychologie
10.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(6): 1098-1108, 2024 Jun 20.
Article de Chinois | MEDLINE | ID: mdl-38977339

RÉSUMÉ

OBJECTIVE: To identify the biomarkers for early rheumatoid arthritis (RA) diagnosis and explore the possible immune regulatory mechanisms. METHODS: The differentially expressed genesin RA were screened and functionally annotated using the limma, RRA, batch correction, and clusterProfiler. The protein-protein interaction network was retrieved from the STRING database, and Cytoscape 3.8.0 and GeneMANIA were used to select the key genes and predicting their interaction mechanisms. ROC curves was used to validate the accuracy of diagnostic models based on the key genes. The disease-specific immune cells were selected via machine learning, and their correlation with the key genes were analyzed using Corrplot package. Biological functions of the key genes were explored using GSEA method. The expression of STAT1 was investigated in the synovial tissue of rats with collagen-induced arthritis (CIA). RESULTS: We identified 9 core key genes in RA (CD3G, CD8A, SYK, LCK, IL2RG, STAT1, CCR5, ITGB2, and ITGAL), which regulate synovial inflammation primarily through cytokines-related pathways. ROC curve analysis showed a high predictive accuracy of the 9 core genes, among which STAT1 had the highest AUC (0.909). Correlation analysis revealed strong correlations of CD3G, ITGAL, LCK, CD8A, and STAT1 with disease-specific immune cells, and STAT1 showed the strongest correlation with M1-type macrophages (R=0.68, P=2.9e-08). The synovial tissues of the ankle joints of CIA rats showed high expressions of STAT1 and p-STAT1 with significant differential expression of STAT1 between the nucleus and the cytoplasm of the synovial fibroblasts. The protein expressions of p-STAT1 and STAT1 in the cell nuclei were significantly reduced after treatment. CONCLUSION: CD3G, CD8A, SYK, LCK, IL2RG, STAT1, CCR5, ITGB2, and ITGAL may serve as biomarkers for early diagnosis of RA. Gene-immune cell pathways such as CD3G/CD8A/LCK-γδ T cells, ITGAL-Tfh cells, and STAT1-M1-type macrophages may be closely related with the development of RA.


Sujet(s)
Polyarthrite rhumatoïde , Marqueurs biologiques , Cartes d'interactions protéiques , Facteur de transcription STAT-1 , Membrane synoviale , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/métabolisme , Animaux , Rats , Facteur de transcription STAT-1/métabolisme , Marqueurs biologiques/métabolisme , Membrane synoviale/métabolisme , Arthrite expérimentale/immunologie , Arthrite expérimentale/métabolisme , Protéine tyrosine kinase p56(lck) spécifique des lymphocytes/métabolisme , Protéine tyrosine kinase p56(lck) spécifique des lymphocytes/génétique , Analyse de profil d'expression de gènes , Bases de données génétiques , Humains , Antigènes CD8/métabolisme , Récepteurs CCR5/métabolisme , Récepteurs CCR5/génétique , Syk kinase/métabolisme , Syk kinase/génétique , Courbe ROC
11.
Int J Biol Sci ; 20(9): 3530-3543, 2024.
Article de Anglais | MEDLINE | ID: mdl-38993551

RÉSUMÉ

During muscle regeneration, interferon-gamma (IFN-γ) coordinates inflammatory responses critical for activation of quiescent muscle stem cells upon injury via the Janus kinase (JAK) - signal transducer and activator of transcription 1 (STAT1) pathway. Dysregulation of JAK-STAT1 signaling results in impaired muscle regeneration, leading to muscle dysfunction or muscle atrophy. Until now, the underlying molecular mechanism of how JAK-STAT1 signaling resolves during muscle regeneration remains largely elusive. Here, we demonstrate that epithelial-stromal interaction 1 (Epsti1), an interferon response gene, has a crucial role in regulating the IFN-γ-JAK-STAT1 signaling at early stage of muscle regeneration. Epsti1-deficient mice exhibit impaired muscle regeneration with elevated inflammation response. In addition, Epsti1-deficient myoblasts display aberrant interferon responses. Epsti1 interacts with valosin-containing protein (VCP) and mediates the proteasomal degradation of IFN-γ-activated STAT1, likely contributing to dampening STAT1-mediated inflammation. In line with the notion, mice lacking Epsti1 exhibit exacerbated muscle atrophy accompanied by increased inflammatory response in cancer cachexia model. Our study suggests a crucial function of Epsti1 in the resolution of IFN-γ-JAK-STAT1 signaling through interaction with VCP which provides insights into the unexplored mechanism of crosstalk between inflammatory response and muscle regeneration.


Sujet(s)
Interféron gamma , Régénération , Facteur de transcription STAT-1 , Facteur de transcription STAT-1/métabolisme , Animaux , Souris , Régénération/physiologie , Interféron gamma/métabolisme , Transduction du signal , Inflammation/métabolisme , Muscles squelettiques/métabolisme , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Souris de lignée C57BL , Souris knockout
12.
Cells ; 13(13)2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38995014

RÉSUMÉ

PD-1 (Programmed cell death protein 1) regulates the metabolic reprogramming of myeloid-derived suppressor cells and myeloid cell differentiation, as well as the type I interferon (IFN-I) signaling pathway in myeloid cells in the tumor microenvironment. PD-1, therefore, is a key inhibitory receptor in myeloid cells. However, the regulation of PD-1 expression in myeloid cells is unknown. We report that the expression level of PDCD1, the gene that encodes the PD-1 protein, is positively correlated with the levels of IFNB1 and IFNAR1 in myeloid cells in human colorectal cancer. Treatment of mouse myeloid cell lines with recombinant IFNß protein elevated PD-1 expression in myeloid cells in vitro. Knocking out IFNAR1, the gene that encodes the IFN-I-specific receptor, diminished the inductive effect of IFNß on PD-1 expression in myeloid cells in vitro. Treatment of tumor-bearing mice with a lipid nanoparticle-encapsulated IFNß-encoding plasmid (IFNBCOL01) increased IFNß expression, resulting in elevated PD-1 expression in tumor-infiltrating myeloid cells. At the molecular level, we determined that IFNß activates STAT1 (signal transducer and activator of transcription 1) and IRFs (interferon regulatory factors) in myeloid cells. Analysis of the cd279 promoter identified IRF2-binding consensus sequence elements. ChIP (chromatin immunoprecipitation) analysis determined that the pSTAT1 directly binds to the irf2 promoter and that IRF2 directly binds to the cd279 promoter in myeloid cells in vitro and in vivo. In colon cancer patients, the expression levels of STAT1, IRF2 and PDCD1 are positively correlated in tumor-infiltrating myeloid cells. Our findings determine that IFNß activates PD-1 expression at least in part by an autocrine mechanism via the stimulation of the pSTAT1-IRF2 axis in myeloid cells.


Sujet(s)
Facteur-2 de régulation d'interféron , Cellules myéloïdes , Récepteur-1 de mort cellulaire programmée , Facteur de transcription STAT-1 , Transduction du signal , Cellules myéloïdes/métabolisme , Cellules myéloïdes/effets des médicaments et des substances chimiques , Animaux , Humains , Facteur de transcription STAT-1/métabolisme , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur-1 de mort cellulaire programmée/génétique , Souris , Facteur-2 de régulation d'interféron/métabolisme , Facteur-2 de régulation d'interféron/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Interféron de type I/métabolisme , Récepteur à l'interféron alpha-bêta/métabolisme , Récepteur à l'interféron alpha-bêta/génétique , Interféron bêta/métabolisme , Lignée cellulaire tumorale , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Souris de lignée C57BL
13.
BMC Immunol ; 25(1): 53, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39080525

RÉSUMÉ

PURPOSE: Primary Sjogren's syndrome (pSS) is a prevalent autoimmune disease. The immune dysregulation it causes often leads to the development of diffuse large B-cell lymphoma (DLBCL) in clinical practice. However, how it contributes to these two disorders at the molecular level is not yet known. This study explored the potential molecular mechanisms associated with the differences between DLBCL and pSS. PATIENTS AND METHODS: Gene expression matrices from discovery cohort 1, discovery cohort 2, and the validation cohort were downloaded from the GEO and TCGA databases. Weighted gene coexpression network analysis (WGCNA) was performed to identify the coexpression modules of DLBCL and pSS in discovery cohort 1 and obtain shared genes. GO and KEGG enrichment analyses and PPI network analysis were performed on the shared genes. Immune-related genes (IRGs) were intersected with shared genes to obtain common genes. Afterward, common genes were identified via machine learning methods. The immune infiltration analysis, miRNA-TF-hub gene regulatory chart, gene interactions of the hub genes, and gene‒drug target analysis were performed. Finally, STAT1 was identified as a possible essential gene by the above analysis, and immune infiltration and GSEA pathway analyses were performed in the high- and low-expression groups in discovery cohort 2. The diagnostic efficacy of the hub genes was assessed in the validation cohort, and clinical samples were collected for validation. RESULTS: By WGCNA, one modular gene in each group was considered highly associated with the disease, and we obtained 28 shared genes. Enrichment analysis revealed shared genes involved in the viral response and regulation. We obtained four hub genes (ISG20, STAT1, TLR7, and RSAD2) via the algorithm. Hub genes and similar genes are primarily involved in regulating type I IFNs. The construction of a miRNA-TF-hub gene regulatory chart revealed that hsa-mir-155-5p, hsa-mir-146b-5p, hsa-mir-21-3p, and hsa-mir-126-3p play essential roles in both diseases. Hub genes were differentially expressed in B-cell memory according to immune infiltration analysis. Hub genes had a strong diagnostic effect on both diseases. STAT1 plays an essential role in immune cells in both diseases. CONCLUSION: We identified hub susceptibility genes for DLBCL and pSS and identified hub genes and potential therapeutic targets that may act as biomarkers.


Sujet(s)
Analyse de profil d'expression de gènes , Réseaux de régulation génique , Lymphome B diffus à grandes cellules , Syndrome de Gougerot-Sjögren , Transcriptome , Syndrome de Gougerot-Sjögren/génétique , Syndrome de Gougerot-Sjögren/immunologie , Humains , Lymphome B diffus à grandes cellules/génétique , Lymphome B diffus à grandes cellules/immunologie , microARN/génétique , Facteur de transcription STAT-1/génétique , Facteur de transcription STAT-1/métabolisme , Régulation de l'expression des gènes tumoraux , Lymphocytes B/immunologie , Biologie informatique/méthodes , Cartes d'interactions protéiques
14.
Mol Med ; 30(1): 110, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39080527

RÉSUMÉ

BACKGROUND AND AIMS: Inflammation is initiates the propagation phase of aortic valve calcification. The activation of NLRP3 signaling in macrophages plays a crucial role in the progression of calcific aortic valve stenosis (CAVS). IFN-γ regulates NLRP3 activity in macrophages. This study aimed to explore the mechanism of IFN-γ regulation and its impact on CAVS progression and valve interstitial cell transdifferentiation. METHODS AND RESULTS: The number of Th1 cells and the expression of IFN-γ and STAT1 in the aortic valve, spleen and peripheral blood increased significantly as CAVS progressed. To explore the mechanisms underlying the roles of Th1 cells and IFN-γ, we treated CAVS mice with IFN-γ-AAV9 or an anti-IFN-γ neutralizing antibody. While IFN-γ promoted aortic valve calcification and dysfunction, it significantly decreased NLRP3 signaling in splenic macrophages and Ly6C+ monocytes. In vitro coculture showed that Th1 cells inhibited NLPR3 activation in ox-LDL-treated macrophages through the IFN-γR1/IFN-γR2-STAT1 pathway. Compared with untreated medium, conditioned medium from Th1-treated bone marrow-derived macrophages reduced the osteogenic calcification of valvular interstitial cells. CONCLUSION: Inhibition of the NLRP3 inflammasome by Th1 cells protects against valvular interstitial cell calcification as a negative feedback mechanism of adaptive immunity toward innate immunity. This study provides a precision medicine strategy for CAVS based on the targeting of anti-inflammatory mechanisms.


Sujet(s)
Sténose aortique , Valve aortique , Calcinose , Inflammasomes , Interféron gamma , Macrophages , Protéine-3 de la famille des NLR contenant un domaine pyrine , Ostéoblastes , Lymphocytes auxiliaires Th1 , Animaux , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Valve aortique/métabolisme , Valve aortique/anatomopathologie , Valve aortique/cytologie , Souris , Macrophages/métabolisme , Macrophages/immunologie , Inflammasomes/métabolisme , Lymphocytes auxiliaires Th1/immunologie , Lymphocytes auxiliaires Th1/métabolisme , Sténose aortique/métabolisme , Sténose aortique/anatomopathologie , Ostéoblastes/métabolisme , Calcinose/métabolisme , Calcinose/immunologie , Interféron gamma/métabolisme , Mâle , Modèles animaux de maladie humaine , Phénotype , Transduction du signal , Souris de lignée C57BL , Facteur de transcription STAT-1/métabolisme
15.
BMC Cancer ; 24(1): 922, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39080642

RÉSUMÉ

Lenvatinib, a multitarget kinase inhibitor, has been proven to be effective in the treatment of advanced hepatocellular carcinoma. It has been previously demonstrated that tumour associated macrophages (TAMs) in tumour tissues can promote HCC growth, invasion and metastasis. Furthermore, lenvatinib has certain immunomodulatory effects on the treatment of HCC. However, the role of lenvatinib in macrophage polarization during HCC treatment has not been fully explored. In this study, we used a variety of experimental methods both in vitro and in vivo to investigate the effect of lenvatinib on TAMs during HCC progression. This study is the first to show that lenvatinib can alter macrophage polarization in both humans and mice. Moreover, macrophages treated with lenvatinib in vitro displayed enhanced classically activated macrophages (M1) activity and suppressed liver cancer cell proliferation, invasion, and migration. Furthermore, during the progression of M1 macrophage polarization induced by lenvatinib, STAT-1 was the main target transcription factor, and inhibiting STAT-1 activity reversed the effect of lenvatinib. Overall, the present study provides a theoretical basis for the immunomodulatory function of lenvatinib in the treatment of HCC.


Sujet(s)
Carcinome hépatocellulaire , Prolifération cellulaire , Évolution de la maladie , Tumeurs du foie , Phénylurées , Quinoléines , Facteur de transcription STAT-1 , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/immunologie , Quinoléines/pharmacologie , Quinoléines/usage thérapeutique , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Tumeurs du foie/immunologie , Phénylurées/pharmacologie , Phénylurées/usage thérapeutique , Facteur de transcription STAT-1/métabolisme , Animaux , Souris , Humains , Prolifération cellulaire/effets des médicaments et des substances chimiques , Macrophages associés aux tumeurs/effets des médicaments et des substances chimiques , Macrophages associés aux tumeurs/immunologie , Macrophages associés aux tumeurs/métabolisme , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Activation des macrophages/effets des médicaments et des substances chimiques , Mâle , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Macrophages/immunologie
16.
Sci Rep ; 14(1): 16081, 2024 07 12.
Article de Anglais | MEDLINE | ID: mdl-38992114

RÉSUMÉ

Tumor-associated macrophages play a crucial role in the tumor microenvironment. Tripartite motif 59 (TRIM59), a member of the tripartite motif (TRIM) family, is known to be associated with immunological diseases and macrophage activation. The functional and molecular mechanisms by which TRIM59 affects the occurrence and development of colorectal cancer (CRC) through macrophages are still not well understood. To address this, we generated macrophage-specific TRIM59 conditional knockout mice and utilized these mice to establish colitis-associated cancer and MC38 transplanted CRC models for further investigation. We found that the deficiency of TRIM59 in macrophages inhibited colorectal tumorigenesis in mice. This tumor-suppressive effect was achieved by promoting the activation of M1 macrophages via STAT1 signaling pathway. Further mechanistic studies revealed that TRIM59 could regulate macrophage polarization by ubiquitinating and degrading STAT1. These findings provide evidence that TRIM59 deficiency promotes M1 macrophage activation and inhibits CRC through the STAT1 signaling pathway, suggesting that the TRIM59/STAT1 signaling pathway may be a promising target for CRC.


Sujet(s)
Tumeurs colorectales , Protéines et peptides de signalisation intracellulaire , Activation des macrophages , Macrophages , Souris knockout , Facteur de transcription STAT-1 , Transduction du signal , Protéines à motif tripartite , Animaux , Facteur de transcription STAT-1/métabolisme , Facteur de transcription STAT-1/génétique , Activation des macrophages/génétique , Protéines à motif tripartite/métabolisme , Protéines à motif tripartite/génétique , Souris , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Protéines et peptides de signalisation intracellulaire/déficit , Macrophages/métabolisme , Humains , Souris de lignée C57BL
17.
Fish Shellfish Immunol ; 152: 109776, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39019128

RÉSUMÉ

Type I IFNs are a subset of cytokines exerting their antiviral effects mainly through the JAK-STAT signalling. Immunogenetic studies have shown that fish possess key components of IFN-JAK-STAT cascade, but the information about the distinct responses of STAT1 and STAT2 to different IFNs is rather limited in fish. Here, we identified and cloned STAT1 and STAT2 genes (named as On-STAT1 and On-STAT2) from tilapia, Oreochromis niloticus. On-STAT1 and On-STAT2 genes were detected in all orangs/tissues examined, and were rapidly induced in spleen, head kidney, and liver following the stimulation of poly(I:C). In addition, the stimulation of poly(I:C), poly(A:T), and different subgroups of recombinant IFNs could induce the expression of On-STAT1 and On-STAT2 in TA-02 cells with distinct induction levels. Importantly, On-STAT2 was rapidly phosphorylated by all three subgroups of IFNs, but the phosphorylation of On-STAT1 was only observed in IFNc- and IFNh-treated TA-02 cells, reflecting the distinct activation of STAT by different subgroups of fish IFNs. The present results thus contribute to better understanding of the JAK-STAT signalling mediated by different subgroups of IFNs in fish.


Sujet(s)
Protéines de poisson , Facteur de transcription STAT-1 , Facteur de transcription STAT-2 , Animaux , Facteur de transcription STAT-2/génétique , Facteur de transcription STAT-2/métabolisme , Protéines de poisson/génétique , Protéines de poisson/immunologie , Protéines de poisson/métabolisme , Facteur de transcription STAT-1/génétique , Facteur de transcription STAT-1/métabolisme , Phosphorylation , Interféron de type I/génétique , Interféron de type I/immunologie , Cichlides/immunologie , Cichlides/génétique , Séquence d'acides aminés , Régulation de l'expression des gènes/immunologie , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Phylogenèse , Poly I-C/pharmacologie , Alignement de séquences/médecine vétérinaire , Transduction du signal/effets des médicaments et des substances chimiques
18.
Cancer Immunol Immunother ; 73(9): 175, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38953994

RÉSUMÉ

Tumor immunotherapies targeting PD-(L)1 exhibit anti-tumor efficacy in only 10-30% of patients with various cancers. Literature has demonstrated that a "hot tumor" which contains high T lymphocytes in the tumor microenvironment exhibits a better response to immunotherapies than a "cold tumor." This study aimed to investigate whether tumor-intrinsic IFNα and CXCL10 determine the recruitment and activation of CD8+ T cells to become "hot tumor." In this study, we found that CXCL10 overexpressed in a variety of tumors including lung, colon, and liver tumors with a correlation with PD-L1. High PD-L1 and CXCL10 are associated with better survival rates in tumor patients receiving immunotherapies. IFNs-downstream transcriptional factor IRF-1 and STAT1 were correlated with PD-L1 and CXCL10 expression. We demonstrated that IRF-1 and STAT1 were both bound with the promoters of PD-L1 and CXCL10, sharing the same signaling pathway and determining IFNs-mediated PD-L1 and CXCL10 expression. In addition, IFNα significantly increased activation marker IFNγ in PBMCs, promoting M1 type monocyte differentiation, CD4+ T, and CD8+ T cell activation. Particularly, we found that CD8+ T lymphocytes abundantly expressed CXCR3, a receptor of CXCL10, by flow cytometry, indicating that tumor-intrinsic CXCL10 potentially recruited CD8+ T in tumor microenvironment. To demonstrate the hypothesis, immunotherapy-sensitive CT26 and immunotherapy-resistant LL/2 were used and we found that CT26 cells exhibited higher IFNα, IFNγ, CXCL10, and PD-L1 levels compared to LL/2, leading to higher IFNγ expression in mouse splenocytes. Moreover, we found that CD8+ T cells were recruited by CXCL10 in vitro, whereas SCH546738, an inhibitor of CXCR3, inhibited T cell migration and splenocytes-mediated anti-tumor effect. We then confirmed that CT26-derived tumor was sensitive to αPD-L1 immunotherapy and LL/2-tumor was resistant, whereas αPD-L1 significantly increased T lymphocyte activation marker CD107a in CT26-derived BALB/c mice. In conclusion, this study revealed that CXCL10 expression is correlated with PD-L1 in tumors, sharing the same signaling pathway and associating with better immunotherapeutic efficacy. Further evidence in the syngeneic tumor models demonstrated that immunotherapy-sensitive CT26 intrinsically exhibited higher IFNα and CXCL10 compared to immunotherapy-resistant LL/2 to recruit and activate CD8+ T cells in the tumor microenvironment, exhibiting "hot tumor" characteristic of sensitizing αPD-L1 immunotherapies.


Sujet(s)
Chimiokine CXCL10 , Immunothérapie , Interféron alpha , Microenvironnement tumoral , Chimiokine CXCL10/métabolisme , Chimiokine CXCL10/immunologie , Microenvironnement tumoral/immunologie , Animaux , Souris , Humains , Immunothérapie/méthodes , Tumeurs/immunologie , Tumeurs/thérapie , Activation des lymphocytes/immunologie , Lignée cellulaire tumorale , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Antigène CD274/métabolisme , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/immunologie , Femelle , Facteur de transcription STAT-1/métabolisme
19.
Ecotoxicol Environ Saf ; 282: 116686, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38971100

RÉSUMÉ

Constituents of cigarette smoke are known to be carcinogens. Additionally, there is mounting evidence that the liver is an organ susceptible to tobacco carcinogenicity. Nicotine, the primary constituent of tobacco, plays a role in cancer progression. In our previous study, it was found that nicotine enhances the proliferation of a human normal fetal hepatic (WRL68) cell due to the activation of p53 mutation at Ser249 (p53-RS)/STAT1/CCND1 signaling pathway. Here, we further elucidated the mechanism of regulating this pathway. Firstly, dose-dependent increase of SETDB1 protein level in WRL68 cells upon exposure to nicotine (1.25, 2.5, and 5 µM), significantly enhanced cellular proliferation. In addition, the upregulation of SETDB1 protein was necessary for the nuclear translocation of p53-RS to establish a ternary complex with STAT1 and SETDB1, which facilitated p53-RS di-methylation at K370 (p53-RS/K370me2). After that, the activation of CCND1/PI3K/AKT pathway was initiated when STAT1 stability was enhanced by p53-RS/K370me2, ultimately resulting in cell proliferation. Altogether, the study revealed that the increase in SETDB1 expression could potentially have a significant impact on the activation of CCND1/PI3K/AKT pathway through p53-RS/K370me2, leading to the proliferation of WRL68 cells induced by nicotine, which could contribute to hepatocellular carcinoma for smokers. Besides, the results of this study provided a foundation for the development of anticancer therapies for cancers associated with tobacco use.


Sujet(s)
Prolifération cellulaire , Cycline D1 , Histone-lysine N-methyltransferase , Nicotine , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Protéine p53 suppresseur de tumeur , Humains , Nicotine/toxicité , Cycline D1/métabolisme , Cycline D1/génétique , Histone-lysine N-methyltransferase/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Méthylation/effets des médicaments et des substances chimiques , Lignée cellulaire , Facteur de transcription STAT-1/métabolisme
20.
Front Immunol ; 15: 1398955, 2024.
Article de Anglais | MEDLINE | ID: mdl-38994355

RÉSUMÉ

Introduction: STAT1a is an essential signal transduction protein involved in the interferon pathway, playing a vital role in IFN-alpha/beta and gamma signaling. Limited information is available about the STAT protein in fish, particularly in Indian major carps (IMC). This study aimed to identify and characterize the STAT1a protein in Labeo rohita (LrSTAT1a). Methods: The full-length CDS of LrSTAT1a transcript was identified and sequenced. Phylogenetic analyses were performed based on the nucleotide sequences. The in-vivo immune stimulant poly I: C was used to treat various tissues, and the expression of LrSTAT1a was determined using quantitative real-time polymerase chain reaction (qRT-PCR). A 3D model of the STAT1a protein was generated using close structure homologs available in the database and checked using molecular dynamics (MD) simulations. Results: The full-length CDS of Labeo rohita STAT1a (LrSTAT1a) transcript consisted of 3238 bp that encoded a polypeptide of 721 amino acids sequence was identified. Phylogenetic analyses were performed based on the nucleotide sequences. Based on our findings, other vertebrates share a high degree of conservation with STAT1a. Additionally, we report that the in vivo immune stimulant poly I: C treatment of various tissues resulted in the expression of LrSTAT1a as determined by quantitative real-time polymerase chain reaction (qRT-PCR). In the current investigation, treatment with poly I: C dramatically increased the expression of LrSTAT1a in nearly every organ and tissue, with the brain, muscle, kidney, and intestine showing the highest levels of expression compared to the control. We made a 3D model of the STAT1a protein by using close structure homologs that were already available in the database. The model was then checked using molecular dynamics (MD) simulations. Consistent with previous research, the MD study highlighted the significance of the STAT1a protein, which is responsible for Src homology 2 (SH2) recognition. An important H-bonding that successfully retains SH2 inside the STAT1a binding cavity was determined to be formed by the conserved residues SER107, GLN530, SER583, LYS584, MET103, and ALA106. Discussion: This study provides molecular insights into the STAT1a protein in Rohu (Labeo rohita) and highlights the potential role of STAT1a in the innate immune response in fish. The high degree of conservation of STAT1a among other vertebrates suggests its crucial role in the immune response. The in-vivo immune stimulation results indicate that STAT1a is involved in the immune response in various tissues, with the brain, muscle, kidney, and intestine being the most responsive. The 3D model and MD study provide further evidence of the significance of STAT1a in the immune response, specifically in SH2 recognition. Further research is necessary to understand the specific mechanisms involved in the IFN pathway and the role of STAT1a in the immune response of IMC.


Sujet(s)
Protéines de poisson , Phylogenèse , Poly I-C , Facteur de transcription STAT-1 , Animaux , Poly I-C/immunologie , Facteur de transcription STAT-1/métabolisme , Facteur de transcription STAT-1/génétique , Protéines de poisson/génétique , Protéines de poisson/immunologie , Protéines de poisson/métabolisme , Domaine d'homologie SRC , Liaison aux protéines , Séquence d'acides aminés , Simulation de dynamique moléculaire , Carpes (poisson)/immunologie , Carpes (poisson)/génétique , Carpes (poisson)/métabolisme , Analyse de profil d'expression de gènes , Cyprinidae/immunologie , Cyprinidae/génétique , Cyprinidae/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE