Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.203
Filtrer
1.
Front Mol Biosci ; 11: 1419072, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948079

RÉSUMÉ

Low-grade glioma (LGG) is a prevalent and lethal primary brain malignancy, with most patients succumbing to recurrence and progression. The signal transducer and activator of transcription (STAT) family has long been implicated in tumor initiation and progression. However, a comprehensive evaluation of the expression status and overall function of STAT genes in LGG remains largely unreported. In this study, we investigated the association between the expression of STAT family genes and the progression of LGG. Through a comprehensive analysis that combined bioinformatics screening and validation assays, we determined that STAT1, STAT3, and STAT5A were upregulated and contributed to the malignant progression of LGG. Notably, our findings suggest that STAT3 is a critical prognostic marker that regulates the progression of LGG. STAT3 emerged as the most significant prognostic indicator governing the advancement of LGG. Additionally, our inquiry into the STAT3-binding proteins and differentially expressed-correlated genes (DEGs) revealed that STAT3 played a pivotal role in the progression of LGG by stimulating the expression of STAT1, FOXO1, and MYC. In summary, our recent study conducted a thorough analysis of the STAT family genes and revealed that directing therapeutic interventions towards STAT3 holds potential as a viable strategy for treating patients with LGG.

2.
Biomol Ther (Seoul) ; 32(4): 467-473, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38844804

RÉSUMÉ

In this study, we investigated the potential protective effects of (+)-afzelechin (AZC), a natural compound that is derived from Bergenia ligulata, on lipopolysaccharide (LPS)-induced inflammatory responses. AZC is known to have antioxidant, anticancer, antimicrobial, and cardiovascular protective properties. However, knowledge regarding the therapeutic potential of AZC against LPS-induced inflammatory responses is limited. Thus, we investigated the protective attributes of AZC against inflammatory damage caused by LPS exposure. We examined the effects of AZC on heme oxygenase (HO)-1, cyclooxygenase (COX)-2, and inducible nitric oxide synthase (iNOS) in LPS-activated human umbilical vein endothelial cells (HUVECs). In addition, the effects of AZC on the expression of iNOS, tumor necrosis factor (TNF)-α, and interleukin (IL)-1ß were analyzed in the lung tissues of LPS-injected mice. Data revealed that AZC promoted the production of HO-1, inhibited the interaction between luciferase and nuclear factor (NF)-κB, and reduced the levels of COX-2/PGE2 and iNOS/NO, thereby leading to a decrease in the signal transducer and activator of transcription (STAT)-1 phosphorylation. Moreover, AZC facilitated the nuclear translocation of Nrf2, increased the binding activity between Nrf2 and the antioxidant response elements (AREs), and lowered the expression of IL-1ß in the LPS-treated HUVECs. In the animal model, AZC significantly reduced the expression of iNOS in the lung tissue structure and the TNF-α level in the bronchoalveolar lavage fluid. These findings demonstrate that AZC possesses anti-inflammatory properties that regulate iNOS through the inhibition of both NF-κB expression and p-STAT-1. Consequently, AZC has potential as a future candidate for the development of new clinical substances for the treatment of pathological inflammation.

3.
BMC Pediatr ; 24(1): 408, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38918745

RÉSUMÉ

Hypereosinophilia is a rare presentation in all age groups, particularly when it is severe, persistent, and progressive. We describe the clinical characteristics and course of severe hypereosinophilia in a full-term Saudi female neonate. A febrile respiratory illness evolved with a progressive increase in peripheral blood leukocyte and eosinophil counts, reaching 44.9% of leukocytes and an absolute value of 57,000 cells/µl. Different etiological examinations (for viral, bacterial, immunodeficiency, hyper IgE syndrome, gene mutations) revealed extremely high CMV antigenemia and a homozygous mutation in the STAT1 gene. Anhelation was relieved by oxygen and anti-viral treatment. Steroids brought a dramatic response in peripheral blood counts within 24 h. After a 6-week course of antiviral and steroid treatment at home, she had an excellent general condition. Conclusion: Although a rare pathology, it is important to consider genetic disorders when there is an atypical immune response to viral infections.


Sujet(s)
Infections à cytomégalovirus , Mutation , Facteur de transcription STAT-1 , Humains , Femelle , Facteur de transcription STAT-1/génétique , Nouveau-né , Infections à cytomégalovirus/complications , Éosinophilie/génétique
4.
Cells ; 13(12)2024 Jun 16.
Article de Anglais | MEDLINE | ID: mdl-38920670

RÉSUMÉ

Proinflammatory T-lymphocytes recruited into the brain and spinal cord mediate multiple sclerosis (MS) and currently there is no cure for MS. IFN-γ-producing Th1 cells induce ascending paralysis in the spinal cord while IL-17-producing Th17 cells mediate cerebellar ataxia. STAT1 and STAT3 are required for Th1 and Th17 development, respectively, and the simultaneous targeting of STAT1 and STAT3 pathways is therefore a potential therapeutic strategy for suppressing disease in the spinal cord and brain. However, the pharmacological targeting of STAT1 and STAT3 presents significant challenges because of their intracellular localization. We have developed a STAT-specific single-domain nanobody (SBT-100) derived from camelids that targets conserved residues in Src homolog 2 (SH2) domains of STAT1 and STAT3. This study investigated whether SBT-100 could suppress experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. We show that SBT-100 ameliorates encephalomyelitis through suppressing the expansion of Th17 and Th1 cells in the brain and spinal cord. Adoptive transfer experiments revealed that lymphocytes from SBT-100-treated EAE mice have reduced capacity to induce EAE, indicating that the immunosuppressive effects derived from the direct suppression of encephalitogenic T-cells. The small size of SBT-100 makes this STAT-specific nanobody a promising immunotherapy for CNS autoimmune diseases, including multiple sclerosis.


Sujet(s)
Encéphalomyélite auto-immune expérimentale , Souris de lignée C57BL , Anticorps à domaine unique , Cellules Th17 , Animaux , Encéphalomyélite auto-immune expérimentale/immunologie , Encéphalomyélite auto-immune expérimentale/thérapie , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Anticorps à domaine unique/pharmacologie , Anticorps à domaine unique/immunologie , Anticorps à domaine unique/usage thérapeutique , Souris , Cellules Th17/immunologie , Cellules Th17/effets des médicaments et des substances chimiques , Femelle , Camélidés du Nouveau Monde , Facteur de transcription STAT-3/métabolisme , Lymphocytes auxiliaires Th1/immunologie , Lymphocytes auxiliaires Th1/effets des médicaments et des substances chimiques , Maladies neuro-inflammatoires/immunologie , Maladies neuro-inflammatoires/traitement médicamenteux , Facteur de transcription STAT-1/métabolisme , Moelle spinale/anatomopathologie , Moelle spinale/effets des médicaments et des substances chimiques , Moelle spinale/immunologie
5.
Immun Inflamm Dis ; 12(6): e1317, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38869352

RÉSUMÉ

BACKGROUND: Numerous studies have demonstrated that Absent in Melanoma 2 (AIM2) is upregulated in aortic plaques, especially in Vascular Smooth Muscle Cells in Coronary Artery Disease (CAD), and is related to inflammasome-induced inflammation. However, the underlying mechanism of this phenomenon and the role of AIM2 in atherosclerosis remained unclear. METHODS: This study enrolled 133 CAD patients and 123 controls. We isolated Peripheral Blood Leukocytes (PBLs) and the mRNA expression of AIM2 inflammasome and its downstream genes (ASC, Caspase-1, IL-1ß, and IL-18) were detected by real-time quantitative PCR (qPCR). We assessed correlations between AIM2 expressions and clinical characteristics by multiple linear regression and spearman's correlation. The THP-1 cells cultured in poly(dA:dT), A151, interferon-gamma (IFN-γ), AG490, or JC2-11. And then the mRNA and protein levels of AIM2, ASC, Caspase-1, IL-1ß, IL-18, GSDMD, and STAT1 were analyzed by qPCR and Western blot analysis, respectively. The migration and adhesive capacity of THP-1 cells was assessed using an inverted microscope and an inverted fluorescence microscope, respectively. RESULTS: In this study, we found that expressions of components of AIM2 inflammasome and its downstream genes (ASC, Caspase-1, IL-1ß, and IL-18), were all increased in PBLs of CAD patients, which indicated the inflammasome activation. AIM2 inflammasome activation further induced pyroptosis, and stimulated migration and adhesion in monocyte cell lines, which was regulated by IFN-γ probably through JAK2/STAT1 pathway. In addition, AIM2 expressions were positively correlated with systemic inflammatory indicators as an independent risk factor for CAD. CONCLUSIONS: In conclusion, increased AIM2 expression, induced by the IFN-γ/JAK2/STAT1 signal, orientates monocytes to inflammatory status or even pyroptosis through AIM2 inflammasome activation, which is involved in the development of CAD.


Sujet(s)
Maladie des artères coronaires , Protéines de liaison à l'ADN , Inflammasomes , Interféron gamma , Kinase Janus-2 , Monocytes , Pyroptose , Facteur de transcription STAT-1 , Transduction du signal , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Maladie des artères coronaires/immunologie , Maladie des artères coronaires/métabolisme , Maladie des artères coronaires/anatomopathologie , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Inflammasomes/métabolisme , Interféron gamma/métabolisme , Kinase Janus-2/métabolisme , Kinase Janus-2/génétique , Monocytes/métabolisme , Monocytes/immunologie , Facteur de transcription STAT-1/métabolisme , Cellules THP-1
6.
Ecotoxicol Environ Saf ; 280: 116534, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38823345

RÉSUMÉ

The correlation between formaldehyde (FA) exposure and prevalence of asthma has been widely reported. However, the underlying mechanism is still not fully understood. FA exposure at 2.0 mg/m3 was found to exacerbate asthma in OVA-induced murine models. IFN-γ, the cytokine produced by T helper 1 (Th1) cells, was significantly induced by FA in serum and bronchoalveolar lavage fluid (BALF) of asthmatic mice, which was different from cytokines secreted by other Th cells. The observation was also confirmed by mRNA levels of Th marker genes in CD4+ T cells isolated from BALF. In addition, increased production of IFN-γ and expression of T-bet in Jurkat T cells primed with phorbol ester and phytohaemagglutinin were also observed with 100 µM FA treatment in vitro. Upregulated STAT1 phosphorylation, T-bet expression and IFN-γ production induced by FA was found to be restrained by STAT1 inhibitor fludarabine, indicating that FA promoted Th1 commitment through the autocrine IFN-γ/STAT1/T-bet pathway in asthma. This work not only revealed that FA could bias Th lineage commitment to exacerbate allergic asthma, but also identified the signaling mechanism of FA-induced Th1 differentiation, which may be utilized as the target for development of interfering strategies against FA-induced immune disorders.


Sujet(s)
Asthme , Formaldéhyde , Interféron gamma , Facteur de transcription STAT-1 , Protéines à domaine boîte-T , Asthme/induit chimiquement , Animaux , Facteur de transcription STAT-1/métabolisme , Interféron gamma/métabolisme , Souris , Protéines à domaine boîte-T/génétique , Protéines à domaine boîte-T/métabolisme , Formaldéhyde/toxicité , Inflammation/induit chimiquement , Souris de lignée BALB C , Humains , Femelle , Liquide de lavage bronchoalvéolaire/cytologie , Liquide de lavage bronchoalvéolaire/composition chimique , Lymphocytes T auxiliaires/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Lymphocytes auxiliaires Th1/effets des médicaments et des substances chimiques , Lymphocytes auxiliaires Th1/immunologie , Cellules Jurkat
7.
Mucosal Immunol ; 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38838816

RÉSUMÉ

The complement system is an evolutionarily conserved arm of innate immunity, which forms one of the first lines of host response to pathogens and assists in the clearance of debris. A deficiency in key activators/amplifiers of the cascade results in recurrent infection, whereas a deficiency in regulating the cascade predisposes to accelerated organ failure, as observed in colitis and transplant rejection. Given that there are over 60 proteins in this system, it has become an attractive target for immunotherapeutics, many of which are United States Food and Drug Administration-approved or in multiple phase 2/3 clinical trials. Moreover, there have been key advances in the last few years in the understanding of how the complement system operates locally in tissues, independent of its activities in circulation. In this review, we will put into perspective the abovementioned discoveries to optimally modulate the spatiotemporal nature of complement activation and regulation at mucosal surfaces.

8.
Biochem Pharmacol ; 226: 116367, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38876258

RÉSUMÉ

Gastric cancer (GC) is a common form of cancer and the leading cause of cancer-related deaths worldwide. Chemotherapy is the primary treatment for patients with unresectable or partially resectable GC. However, its adverse effects and chemoresistance greatly restrict its applicability and efficacy. Although HER2-targeted therapy and immunotherapy have been successfully used for GC treatment, their beneficial population is limited. To expand the range of cancer treatments, drug repurposing has emerged as a promising strategy. In this study, we evaluated the potential of Metformin, an oral anti-hyperglycemic agent, to suppress GC progression both in vivo and in vitro. Functional investigations showed that Metformin significantly inhibits GC proliferation and migration. Furthermore, we discovered that Metformin bound and disrupted STAT1 phosphorylation, inhibiting PRMT1 expression and consequently GC progression. In conclusion, our study not only provides further evidence for the anti-GC role of Metformin but also identifies the direct target mediating the tumor-inhibitory effects of Metformin in GC.

9.
J Mol Histol ; 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38856930

RÉSUMÉ

Systemic lupus erythematosus (SLE) is a persistent autoimmune disorder that can culminate in lupus nephritis (LN), an intricate renal complication. In pursuit of unraveling the intricate molecular underpinnings governing LN progression, we conducted bioinformatics analysis employing gene expression data sourced from the GSE32591 dataset. Our scrutiny revealed a panoply of differentially expressed genes (DEGs) within the glomerulus and tubulointerstitial compartments of LN patients. Enrichment analysis for DEGs engaged in diverse processes, encompassing virus defense, viral life cycle, cell adhesion molecules, and the NOD-like receptor signaling pathway. Notably, STAT1 emerged as an eminent central hub gene intrinsically tied to NOD-like receptor signaling. To explore the functional significance of STAT1 in the context of LN, MRL-lpr mice model was used to knockout STAT1. The results unveiled that STAT1 silencing yielded a migratory effect on kidney injury, concurrently curbing inflammatory markers. Meanwhile, knockout STAT1 also reduced NLRP3 expression and Cleaved caspase-1 expression. These findings offer tantalizing prospects for targeting STAT1 as a potential therapeutic conduit in the management of LN.

10.
J Virol ; : e0033424, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38829137

RÉSUMÉ

Porcine deltacoronavirus (PDCoV) is an enteric pathogenic coronavirus that causes acute and severe watery diarrhea in piglets and has the ability of cross-species transmission, posing a great threat to swine production and public health. The interferon (IFN)-mediated signal transduction represents an important component of virus-host interactions and plays an essential role in regulating viral infection. Previous studies have suggested that multifunctional viral proteins encoded by coronaviruses antagonize the production of IFN via various means. However, the function of these viral proteins in regulating IFN-mediated signaling pathways is largely unknown. In this study, we demonstrated that PDCoV and its encoded nucleocapsid (N) protein antagonize type I IFN-mediated JAK-STAT signaling pathway. We identified that PDCoV infection stimulated but delayed the production of IFN-stimulated genes (ISGs). In addition, PDCoV inhibited JAK-STAT signal transduction by targeting the nuclear translocation of STAT1 and ISGF3 formation. Further evidence showed that PDCoV N is the essential protein involved in the inhibition of type I IFN signaling by targeting STAT1 nuclear translocation via its C-terminal domain. Mechanistically, PDCoV N targets STAT1 by interacting with it and subsequently inhibiting its nuclear translocation. Furthermore, PDCoV N inhibits STAT1 nuclear translocation by specifically targeting KPNA2 degradation through the lysosomal pathway, thereby inhibiting the activation of downstream sensors in the JAK-STAT signaling pathway. Taken together, our results reveal a novel mechanism by which PDCoV N interferes with the host antiviral response.IMPORTANCEPorcine deltacoronavirus (PDCoV) is a novel enteropathogenic coronavirus that receives increased attention and seriously threatens the pig industry and public health. Understanding the underlying mechanism of PDCoV evading the host defense during infection is essential for developing targeted drugs and effective vaccines against PDCoV. This study demonstrated that PDCoV and its encoded nucleocapsid (N) protein antagonize type I interferon signaling by targeting STAT1, which is a crucial signal sensor in the JAK-STAT signaling pathway. Further experiments suggested that PDCoV N-mediated inhibition of the STAT1 nuclear translocation involves the degradation of KPNA2, and the lysosome plays a role in KPNA2 degradation. This study provides new insights into the regulation of PDCoV N in the JAK-STAT signaling pathway and reveals a novel mechanism by which PDCoV evades the host antiviral response. The novel findings may guide us to discover new therapeutic targets and develop live attenuated vaccines for PDCoV infection.

11.
Redox Biol ; 74: 103236, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38875958

RÉSUMÉ

The pathogenesis of epilepsy remains unclear; however, a prevailing hypothesis suggests that the primary underlying cause is an imbalance between neuronal excitability and inhibition. Glucose-6-phosphate dehydrogenase (G6PD) is a key enzyme in the pentose phosphate pathway, which is primarily involved in deoxynucleic acid synthesis and antioxidant defense mechanisms and exhibits increased expression during the chronic phase of epilepsy, predominantly colocalizing with neurons. G6PD overexpression significantly reduces the frequency and duration of spontaneous recurrent seizures. Furthermore, G6PD overexpression enhances signal transducer and activator of transcription 1 (STAT1) expression, thus influencing N-methyl-d-aspartic acid receptors expression, and subsequently affecting seizure activity. Importantly, the regulation of STAT1 by G6PD appears to be mediated primarily through reactive oxygen species signaling pathways. Collectively, our findings highlight the pivotal role of G6PD in modulating epileptogenesis, and suggest its potential as a therapeutic target for epilepsy.


Sujet(s)
Glucose 6-phosphate dehydrogenase , Espèces réactives de l'oxygène , Récepteurs du N-méthyl-D-aspartate , Facteur de transcription STAT-1 , Crises épileptiques , Glucose 6-phosphate dehydrogenase/métabolisme , Glucose 6-phosphate dehydrogenase/antagonistes et inhibiteurs , Glucose 6-phosphate dehydrogenase/génétique , Espèces réactives de l'oxygène/métabolisme , Animaux , Récepteurs du N-méthyl-D-aspartate/métabolisme , Récepteurs du N-méthyl-D-aspartate/génétique , Crises épileptiques/métabolisme , Crises épileptiques/traitement médicamenteux , Facteur de transcription STAT-1/métabolisme , Épilepsie/métabolisme , Épilepsie/traitement médicamenteux , Épilepsie/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Souris , Humains , Neurones/métabolisme , Mâle , Rats , Modèles animaux de maladie humaine
12.
Int J Mol Sci ; 25(9)2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38731828

RÉSUMÉ

HLA-DR-positive NK cells, found in both healthy individuals and patients with different inflammatory diseases, are characterized as activated cells. However, data on their capacity for IFNγ production or cytotoxic response vary between studies. Thus, more precise investigation is needed of the mechanisms related to the induction of HLA-DR expression in NK cells, their associations with NK cell differentiation stage, and functional or metabolic state. In this work, HLA-DR-expressing NK cell subsets were investigated using transcriptomic analysis, metabolic activity assays, and analysis of intercellular signaling cascades. We demonstrated that HLA-DR+CD56bright NK cells were characterized by a proliferative phenotype, while HLA-DR+CD56dim NK cells exhibited features of adaptive cells and loss of inhibitory receptors with increased expression of MHC class II trans-activator CIITA. The activated state of HLA-DR-expressing NK cells was confirmed by higher levels of ATP and mitochondrial mass observed in this subset compared to HLA-DR- cells, both ex vivo and after stimulation in culture. We showed that HLA-DR expression in NK cells in vitro can be induced both through stimulation by exogenous IL-2 and IL-21, as well as through auto-stimulation by NK-cell-produced IFNγ. At the intracellular level, HLA-DR expression depended on the activation of STAT3- and ERK1/2-mediated pathways, with subsequent activation of isoform 3 of the transcription factor CIITA. The obtained results broaden the knowledge about HLA-DR-positive NK cell appearance, diversity, and functions, which might be useful in terms of understanding the role of this subset in innate immunity and assessing their possible implications in NK cell-based therapy.


Sujet(s)
Différenciation cellulaire , Antigènes HLA-DR , Interféron gamma , Cellules tueuses naturelles , Cellules tueuses naturelles/métabolisme , Cellules tueuses naturelles/immunologie , Humains , Antigènes HLA-DR/métabolisme , Antigènes HLA-DR/génétique , Interféron gamma/métabolisme , Antigènes CD56/métabolisme , Activation des lymphocytes/immunologie , Interleukine-2/métabolisme , Interleukine-2/pharmacologie , Cellules cultivées , Protéines nucléaires , Transactivateurs
13.
Biochem Biophys Res Commun ; 722: 150156, 2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-38797155

RÉSUMÉ

Osteosarcoma, considered as the primary cause of malignant bone tumors in children, necessitates novel therapeutic strategies to enhance overall survival rates. KAT7, a histone acetyltransferase, exerts pivotal functions in gene transcription and immune modulation. In light of this, our study identified a significant upregulation of KAT7 in the mRNA and protein levels in human osteosarcoma, boosting cell proliferation in vivo and in vitro. In addition, KAT7-mediated H3K14ac activation induced MMP14 transcription, leading to increased expression and facilitation of osteosarcoma cell metastasis. Subsequent bioinformatics analyses highlighted a correlation between KAT7 and adaptive immune responses, indicating CCL3 as a downstream target of KAT7. Mechanistically, STAT1 was found to transcriptionally upregulate CCL3 expression. Furthermore, overexpression of KAT7 suppressed CCL3 secretions, whereas knockdown of KAT7 enhanced its release. Overall, these findings underscore the oncogenic role of KAT7 in regulating immune responses for osteosarcoma treatment.


Sujet(s)
Tumeurs osseuses , Chimiokine CCL3 , Régulation de l'expression des gènes tumoraux , Histone acetyltransferases , Ostéosarcome , Facteur de transcription STAT-1 , Transduction du signal , Animaux , Humains , Souris , Tumeurs osseuses/génétique , Tumeurs osseuses/métabolisme , Tumeurs osseuses/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Chimiokine CCL3/métabolisme , Chimiokine CCL3/génétique , Histone acetyltransferases/métabolisme , Histone acetyltransferases/génétique , Souris nude , Ostéosarcome/génétique , Ostéosarcome/métabolisme , Ostéosarcome/anatomopathologie , Facteur de transcription STAT-1/métabolisme , Facteur de transcription STAT-1/génétique
14.
J Clin Immunol ; 44(5): 124, 2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38758476

RÉSUMÉ

PURPOSES: STAT1 is a transduction and transcriptional regulator that functions within the classical JAK/STAT pathway. In addition to chronic mucocutaneous candidiasis, bacterial infections are a common occurrence in patients with STAT1 gain-of-function (GOF) mutations. These patients often exhibit skewing of B cell subsets; however, the impact of STAT1-GOF mutations on B cell-mediated humoral immunity remains largely unexplored. It is also unclear whether these patients with IgG within normal range require regular intravenous immunoglobulin (IVIG) therapy. METHODS: Eleven patients (harboring nine different STAT1-GOF mutations) were enrolled. Reporter assays and immunoblot analyses were performed to confirm STAT1 mutations. Flow cytometry, deep sequencing, ELISA, and ELISpot were conducted to assess the impact of STAT1-GOF on humoral immunity. RESULTS: All patients exhibited increased levels of phospho-STAT1 and total STAT1 protein, with two patients carrying novel mutations. In vitro assays showed that these two novel mutations were GOF mutations. Three patients with normal total IgG levels received regular IVIG infusions, resulting in effective control of bacterial infections. Four cases showed impaired affinity and specificity of pertussis toxin-specific antibodies, accompanied by reduced generation of class-switched memory B cells. Patients also had a disrupted immunoglobulin heavy chain (IGH) repertoire, coupled with a marked reduction in the somatic hypermutation frequency of switched Ig transcripts. CONCLUSION: STAT1-GOF mutations disrupt B cell compartments and skew IGH characteristics, resulting in impaired affinity and antigen-specificity of antibodies and recurrent bacterial infections. Regular IVIG therapy can control these infections in patients, even those with normal total IgG levels.


Sujet(s)
Lymphocytes B , Infections bactériennes , Mutation gain de fonction , Immunoglobulines par voie veineuse , Facteur de transcription STAT-1 , Humains , Facteur de transcription STAT-1/génétique , Infections bactériennes/immunologie , Infections bactériennes/génétique , Femelle , Mâle , Enfant , Immunoglobulines par voie veineuse/usage thérapeutique , Lymphocytes B/immunologie , Adulte , Immunoglobuline G/immunologie , Immunoglobuline G/sang , Enfant d'âge préscolaire , Adolescent , Jeune adulte , Immunité humorale
15.
J Transl Med ; 22(1): 460, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38750462

RÉSUMÉ

BACKGROUND: Chaperonin Containing TCP1 Subunit 6 A (CCT6A) is a prominent protein involved in the folding and stabilization of newly synthesized proteins. However, its roles and underlying mechanisms in lung adenocarcinoma (LUAD), one of the most aggressive cancers, remain elusive. METHODS: Our study utilized in vitro cell phenotype experiments to assess CCT6A's impact on the proliferation and invasion capabilities of LUAD cell lines. To delve into CCT6A's intrinsic mechanisms affecting glycolysis and proliferation in lung adenocarcinoma, we employed transcriptomic sequencing and liquid chromatography-mass spectrometry analysis. Co-immunoprecipitation (Co-IP) and chromatin immunoprecipitation (CHIP) assays were also conducted to substantiate the mechanism. RESULTS: CCT6A was found to be significantly overexpressed in LUAD and associated with a poorer prognosis. The silencing of CCT6A inhibited the proliferation and migration of LUAD cells and elevated apoptosis rates. Mechanistically, CCT6A interacted with STAT1 protein, forming a complex that enhances the stability of STAT1 by protecting it from ubiquitin-mediated degradation. This, in turn, facilitated the transcription of hexokinase 2 (HK2), a critical enzyme in aerobic glycolysis, thereby stimulating LUAD's aerobic glycolysis and progression. CONCLUSION: Our findings reveal that the CCT6A/STAT1/HK2 axis orchestrated a reprogramming of glucose metabolism and thus promoted LUAD progression. These insights position CCT6A as a promising candidate for therapeutic intervention in LUAD treatment.


Sujet(s)
Adénocarcinome pulmonaire , Prolifération cellulaire , Chaperonine contenant TCP-1 , Évolution de la maladie , Glycolyse , Hexokinase , Tumeurs du poumon , Facteur de transcription STAT-1 , Humains , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Adénocarcinome pulmonaire/génétique , Hexokinase/métabolisme , Facteur de transcription STAT-1/métabolisme , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Chaperonine contenant TCP-1/métabolisme , Lignée cellulaire tumorale , Mouvement cellulaire , Régulation de l'expression des gènes tumoraux , Apoptose , Transduction du signal , Invasion tumorale
16.
Cell Commun Signal ; 22(1): 272, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38750472

RÉSUMÉ

BACKGROUND: In the tumor immune microenvironment (TIME), triggering receptor expressed on myeloid cells 2 (trem2) is widely considered to be a crucial molecule on tumor-associated macrophages(TAMs). Multiple studies have shown that trem2 may function as an immune checkpoint in various malignant tumors, mediating tumor immune evasion. However, its specific molecular mechanisms, especially in glioma, remain elusive. METHODS: Lentivirus was transfected to establish cells with stable knockdown of trem2. A Transwell system was used for segregated coculture of glioma cells and microglia. Western blotting, quantitative real-time polymerase chain reaction (qRT‒PCR), and immunofluorescence (IF) were used to measure the expression levels of target proteins. The proliferation, invasion, and migration of cells were detected by colony formation, cell counting kit-8 (CCK8), 5-ethynyl-2'-deoxyuridine (EdU) and transwell assays. The cell cycle, apoptosis rate and reactive oxygen species (ROS) level of cells were assessed using flow cytometry assays. The comet assay and tube formation assay were used to detect DNA damage in glioma cells and angiogenesis activity, respectively. Gl261 cell lines and C57BL/6 mice were used to construct the glioma orthotopic transplantation tumor model. RESULTS: Trem2 was highly overexpressed in glioma TAMs. Knocking down trem2 in microglia suppressed the growth and angiogenesis activity of glioma cells in vivo and in vitro. Mechanistically, knockdown of trem2 in microglia promoted proinflammatory microglia and inhibited anti-inflammatory microglia by activating jak2/stat1 and inhibiting the NF-κB p50 signaling pathway. The proinflammatory microglia produced high concentrations of nitric oxide (NO) and high levels of the proinflammatory cytokines TNF-α, IL-6, and IL-1ß, and caused further DNA damage and promoted the apoptosis rate of tumor cells. CONCLUSIONS: Our findings revealed that trem2 in microglia plays a significant role in the TIME of gliomas. Knockdown of trem2 in microglia might help to improve the efficiency of inhibiting glioma growth and delaying tumor progression and provide new ideas for further treatment of glioma.


Sujet(s)
Gliome , Kinase Janus-2 , Glycoprotéines membranaires , Microglie , Facteur de transcription NF-kappa B , Récepteurs immunologiques , Facteur de transcription STAT-3 , Transduction du signal , Gliome/génétique , Gliome/anatomopathologie , Gliome/métabolisme , Kinase Janus-2/génétique , Kinase Janus-2/métabolisme , Microglie/métabolisme , Microglie/anatomopathologie , Animaux , Récepteurs immunologiques/génétique , Récepteurs immunologiques/métabolisme , Glycoprotéines membranaires/génétique , Glycoprotéines membranaires/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Souris , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique , Transduction du signal/génétique , Lignée cellulaire tumorale , Souris de lignée C57BL , Techniques de knock-down de gènes , Prolifération cellulaire/génétique , Humains , Inflammation/génétique , Inflammation/anatomopathologie , Apoptose/génétique , Évolution de la maladie , Mouvement cellulaire/génétique
17.
J Autoimmun ; 147: 103243, 2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38788537

RÉSUMÉ

OBJECTIVES: Autoreactive B cells and interferon (IFN) signature are hallmarks of primary sjögren's syndrome (pSS), but how IFN signaling pathways influence autoantibody production and clinical manifestations remain unclear. More detailed studies hold promise for improved diagnostic methodologies and personalized treatment. METHODS: We analyzed peripheral blood T and B cell subsets from 34 pSS patients and 38 healthy donors (HDs) at baseline and upon stimulation regarding their expression levels of type I and II IFN signaling molecules (STAT1/2, IRF1, IRF9). Additionally, we investigated how the levels of these molecules correlated with serological and clinical characteristics and performed ROC analysis. RESULTS: Patients showed elevated IFN pathway molecules, including STAT1, STAT2 and IRF9 among most T and B cell subsets. We found a reduced ratio of phosphorylated STAT1 and STAT2 in patients in comparison to HDs, although B cells from patients were highly responsive by increased phosphorylation upon IFN stimulation. Correlation matrices showed further interrelations between STAT1, IRF1 and IRF9 in pSS. Levels of STAT1 and IRF9 in T and B cells correlated with the IFN type I marker Siglec-1 (CD169) on monocytes. High levels of STAT1 and IRF9 within pSS B cells were significantly associated with hypergammaglobulinemia as well as anti-SSA/anti-SSB autoantibodies. Elevated STAT1 levels were found in patients with extraglandular disease and could serve as a biomarker for this subgroup (p < 0.01). Notably, IRF9 levels in T and B cells correlated with EULAR Sjögren's syndrome disease activity index (ESSDAI). CONCLUSION: Here, we provide evidence that in active pSS patients, enhanced IFN signaling incl. unphosphorylated STAT1 and STAT2 with IRFs entertain chronic T and B cell activation. Furthermore, increased STAT1 levels candidate as biomarker of extraglandular disease, while IRF9 levels can serve as biomarker for disease activity.

18.
Int Immunopharmacol ; 134: 112191, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38759369

RÉSUMÉ

Social behavior is inextricably linked to the immune system. Although IFN-γ is known to be involved in social behavior, yet whether and how it encodes social memory remains unclear. In the current study, we injected with IFN-γ into the lateral ventricle of male C57BL/6J mice, and three-chamber social test was used to examine the effects of IFN-γ on their social preference and social memory. The morphology of microglia in the hippocampus, prelimbic cortex and amygdala was examined using immunohistochemistry, and the phenotype of microglia were examined using immunohistochemistry and enzyme-linked immunosorbent assays. The IFN-γ-injected mice were treated with lipopolysaccharide, and effects of IFN-γ on behavior and microglial responses were evaluated. STAT1 pathway and microglia-neuron interactions were examined in vivo or in vitro using western blotting and immunohistochemistry. Finally, we use STAT1 inhibitor or minocycline to evaluated the role of STAT1 in mediating the microglial priming and effects of primed microglia in IFN-γ-induced social dysfunction. We demonstrated that 500 ng of IFN-γ injection results in significant decrease in social index and social novelty recognition index, and induces microglial priming in hippocampus, characterized by enlarged cell bodies, shortened branches, increased expression of CD68, CD86, CD74, CD11b, CD11c, CD47, IL-33, IL-1ß, IL-6 and iNOS, and decreased expression of MCR1, Arg-1, IGF-1 and BDNF. This microglia subpopulation is more sensitive to LPS challenge, which characterized by more significant morphological changes and inflammatory responses, as well as induced increased sickness behaviors in mice. IFN-γ upregulated pSTAT1 and STAT1 and promoted the nuclear translocation of STAT1 in the hippocampal microglia and in the primary microglia. Giving minocycline or STAT1 inhibitor fludarabin blocked the priming of hippocampal microglia induced by IFN-γ, ameliorated the dysfunction in hippocampal microglia-neuron interactions and synapse pruning by microglia, thereby improving social memory deficits in IFN-γ injected mice. IFN-γ initiates STAT1 pathway to induce priming of hippocampal microglia, thereby disrupts hippocampal microglia-neuron interactions and neural circuit link to social memory. Blocking STAT1 pathway or inhibiting microglial priming may be strategies to reduce the effects of IFN-γ on social behavior.


Sujet(s)
Hippocampe , Interféron gamma , Souris de lignée C57BL , Microglie , Facteur de transcription STAT-1 , Transduction du signal , Comportement social , Animaux , Microglie/effets des médicaments et des substances chimiques , Microglie/immunologie , Microglie/métabolisme , Facteur de transcription STAT-1/métabolisme , Mâle , Interféron gamma/métabolisme , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Hippocampe/immunologie , Souris , Transduction du signal/effets des médicaments et des substances chimiques , Lipopolysaccharides , Mémoire/effets des médicaments et des substances chimiques , Cellules cultivées , Neurones/effets des médicaments et des substances chimiques , Neurones/immunologie , Neurones/métabolisme
19.
J Exp Clin Cancer Res ; 43(1): 152, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38812060

RÉSUMÉ

BACKGROUND: Intrahepatic cholangiocarcinoma (ICCA) is a heterogeneous group of malignant tumors characterized by high recurrence rate and poor prognosis. Heterochromatin Protein 1α (HP1α) is one of the most important nonhistone chromosomal proteins involved in transcriptional silencing via heterochromatin formation and structural maintenance. The effect of HP1α on the progression of ICCA remained unclear. METHODS: The effect on the proliferation of ICCA was detected by experiments in two cell lines and two ICCA mouse models. The interaction between HP1α and Histone Deacetylase 1 (HDAC1) was determined using Electrospray Ionization Mass Spectrometry (ESI-MS) and the binding mechanism was studied using immunoprecipitation assays (co-IP). The target gene was screened out by RNA sequencing (RNA-seq). The occupation of DNA binding proteins and histone modifications were predicted by bioinformatic methods and evaluated by Cleavage Under Targets and Tagmentation (CUT & Tag) and Chromatin immunoprecipitation (ChIP). RESULTS: HP1α was upregulated in intrahepatic cholangiocarcinoma (ICCA) tissues and regulated the proliferation of ICCA cells by inhibiting the interferon pathway in a Signal Transducer and Activator of Transcription 1 (STAT1)-dependent manner. Mechanistically, STAT1 is transcriptionally regulated by the HP1α-HDAC1 complex directly and epigenetically via promoter binding and changes in different histone modifications, as validated by high-throughput sequencing. Broad-spectrum HDAC inhibitor (HDACi) activates the interferon pathway and inhibits the proliferation of ICCA cells by downregulating HP1α and targeting the heterodimer. Broad-spectrum HDACi plus interferon preparation regimen was found to improve the antiproliferative effects and delay ICCA development in vivo and in vitro, which took advantage of basal activation as well as direct activation of the interferon pathway. HP1α participates in mediating the cellular resistance to both agents. CONCLUSIONS: HP1α-HDAC1 complex influences interferon pathway activation by directly and epigenetically regulating STAT1 in transcriptional level. The broad-spectrum HDACi plus interferon preparation regimen inhibits ICCA development, providing feasible strategies for ICCA treatment. Targeting the HP1α-HDAC1-STAT1 axis is a possible strategy for treating ICCA, especially HP1α-positive cases.


Sujet(s)
Tumeurs des canaux biliaires , Cholangiocarcinome , Homologue-5 de la protéine chromobox , Histone Deacetylase 1 , Facteur de transcription STAT-1 , Animaux , Femelle , Humains , Mâle , Souris , Tumeurs des canaux biliaires/métabolisme , Tumeurs des canaux biliaires/traitement médicamenteux , Tumeurs des canaux biliaires/anatomopathologie , Tumeurs des canaux biliaires/génétique , Lignée cellulaire tumorale , Prolifération cellulaire , Cholangiocarcinome/métabolisme , Cholangiocarcinome/traitement médicamenteux , Cholangiocarcinome/anatomopathologie , Cholangiocarcinome/génétique , Homologue-5 de la protéine chromobox/métabolisme , Protéines chromosomiques nonhistones/métabolisme , Protéines chromosomiques nonhistones/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Histone Deacetylase 1/métabolisme , Facteur de transcription STAT-1/métabolisme
20.
Immun Inflamm Dis ; 12(4): e1234, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38578001

RÉSUMÉ

BACKGROUND AND OBJECTIVE: Long noncoding RNAs (lncRNAs) are crucial in regulating various physiological and pathological processes, including immune responses. LINC01686 is a lncRNA with previously uncharacterized functions in immune regulation. This study aims to investigate the function of LINC01686 in lipopolysaccharide (LPS)-induced inflammatory responses in the human monocytic leukemia cell line THP-1 and its potential regulatory mechanisms involving miR-18a-5p and the anti-inflammatory protein A20. METHOD: THP-1 cells were stimulated with LPS to induce inflammatory responses, followed by analysis of LINC01686 expression levels. The role of LINC01686 in regulating the expression of interleukin (IL)-6, IL-8, A20, and signal transducer and activator of transcription 1 (STAT1) was examined using small interfering RNA-mediated knockdown. Additionally, the involvement of miR-18a-5p in LINC01686-mediated regulatory pathways was assessed by transfection with decoy RNAs mimicking the miR-18a-5p binding sites of LINC01686 or A20 messenger RNA. RESULTS: LINC01686 expression was upregulated in THP-1 cells following LPS stimulation. Suppression of LINC01686 enhanced LPS-induced expression of IL-6 and IL-8, mediated through increased production of reactive oxygen species. Moreover, LINC01686 knockdown upregulated the expression and activation of IκB-ζ, STAT1, and downregulated A20 expression. Transfection with decoy RNAs reversed the effects of LINC01686 suppression on A20, STAT1, IL-6, and IL-8 expression, highlighting the role of LINC01686 in sponging miR-18a-5p and regulating A20 expression. CONCLUSION: This study provides the first evidence that LINC01686 plays a critical role in modulating LPS-induced inflammatory responses in THP-1 cells by sponging miR-18a-5p, thereby regulating the expression and activation of A20 and STAT1. These findings shed light on the complex regulatory mechanisms involving lncRNAs in immune responses and offer potential therapeutic targets for inflammatory diseases.


Sujet(s)
Cytokines , microARN , ARN long non codant , Humains , Cytokines/génétique , Cytokines/métabolisme , Interleukine-6 , Interleukine-8/métabolisme , Lipopolysaccharides , microARN/génétique , ARN long non codant/génétique , ARN long non codant/métabolisme , Facteur de transcription STAT-1/génétique , Facteur de transcription STAT-1/métabolisme , Cellules THP-1
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...