首页 > 最新文献

Inflammation最新文献

英文 中文
REGγ-mediated Barrier Disruption and NF-κB Activation Aggravates Intestinal Inflammation in Necrotizing Enterocolitis. regγ介导的屏障破坏和NF-κB激活加重坏死性小肠结肠炎的肠道炎症。
IF 4.5 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-12-10 DOI: 10.1007/s10753-024-02203-2
Ya Li, Deng Pan, Huifeng Liu, Wenya Xie, Xiaoyong Li, Xin Mu, Li Chen, Min Yang, Xianliang Wang, Xianxiao Li, Jianhui Li, Bianhong Zhang, Fangxia Guan, Faisal Aziz, Jingyu Cao, Xiangzhan Zhu

Necrotizing enterocolitis (NEC) stands as the most prevalent and severe gastrointestinal ailment affecting premature newborns, particularly those with extremely low birth weights. Intestinal barrier dysfunction emerges as a crucial factor in NEC's pathogenesis. REGγ predominantly manifests in the intestinal epithelium and has shown to regulate experimental colitis. However, the potential correlation between REGγ and NEC remains ambiguous. This study reveals that REGγ is notably overexpressed in both human and murine NEC samples. REGγ-deficient mice displayed improvements in intestinal mucosal inflammation and reduced NEC severity. Additionally, REGγ deficiency notably lowered the expression of phosphorylated transcription factor p65 and inflammatory factors in intestinal epithelial cells of NEC-afflicted mice. REGγ exacerbates the local inflammation by triggering the degradation of IκBɛ, a negative regulator of NFκB. Clinically, REGγ and p65 expression levels exhibit a positive correlation in NEC specimens. Moreover, pre-treatment of mice with a p65 inhibitor effectively suppressed the expression of inflammatory mediators and alleviated REGγ-mediated NEC development. These findings underscore that abnormal upregulation of REGγ triggers NEC development by enhancing NF-κB activity and exacerbating epithelial barrier dysfunction. Thus, novel therapeutic approaches targeting REGγ inhibition may offer enhanced treatment for NEC.

坏死性小肠结肠炎(NEC)是影响早产儿的最普遍和严重的胃肠道疾病,特别是那些出生体重极低的新生儿。肠屏障功能障碍是NEC发病的重要因素。REGγ主要表现在肠上皮中,并已被证明调节实验性结肠炎。然而,REGγ和NEC之间的潜在相关性仍然不明确。这项研究表明REGγ在人和小鼠NEC样本中都明显过表达。regγ缺乏小鼠表现出肠黏膜炎症改善和NEC严重程度降低。此外,REGγ缺乏显著降低了nec小鼠肠上皮细胞中磷酸化转录因子p65和炎症因子的表达。REGγ通过触发nf - κ b的负调节因子i - κ b的降解而加剧局部炎症。在临床上,REGγ和p65的表达水平在NEC标本中呈正相关。此外,用p65抑制剂预处理小鼠可有效抑制炎症介质的表达,减轻regγ介导的NEC发展。这些发现强调REGγ的异常上调通过增强NF-κB活性和加剧上皮屏障功能障碍来触发NEC的发展。因此,针对REGγ抑制的新治疗方法可能为NEC提供增强治疗。
{"title":"REGγ-mediated Barrier Disruption and NF-κB Activation Aggravates Intestinal Inflammation in Necrotizing Enterocolitis.","authors":"Ya Li, Deng Pan, Huifeng Liu, Wenya Xie, Xiaoyong Li, Xin Mu, Li Chen, Min Yang, Xianliang Wang, Xianxiao Li, Jianhui Li, Bianhong Zhang, Fangxia Guan, Faisal Aziz, Jingyu Cao, Xiangzhan Zhu","doi":"10.1007/s10753-024-02203-2","DOIUrl":"https://doi.org/10.1007/s10753-024-02203-2","url":null,"abstract":"<p><p>Necrotizing enterocolitis (NEC) stands as the most prevalent and severe gastrointestinal ailment affecting premature newborns, particularly those with extremely low birth weights. Intestinal barrier dysfunction emerges as a crucial factor in NEC's pathogenesis. REGγ predominantly manifests in the intestinal epithelium and has shown to regulate experimental colitis. However, the potential correlation between REGγ and NEC remains ambiguous. This study reveals that REGγ is notably overexpressed in both human and murine NEC samples. REGγ-deficient mice displayed improvements in intestinal mucosal inflammation and reduced NEC severity. Additionally, REGγ deficiency notably lowered the expression of phosphorylated transcription factor p65 and inflammatory factors in intestinal epithelial cells of NEC-afflicted mice. REGγ exacerbates the local inflammation by triggering the degradation of IκBɛ, a negative regulator of NFκB. Clinically, REGγ and p65 expression levels exhibit a positive correlation in NEC specimens. Moreover, pre-treatment of mice with a p65 inhibitor effectively suppressed the expression of inflammatory mediators and alleviated REGγ-mediated NEC development. These findings underscore that abnormal upregulation of REGγ triggers NEC development by enhancing NF-κB activity and exacerbating epithelial barrier dysfunction. Thus, novel therapeutic approaches targeting REGγ inhibition may offer enhanced treatment for NEC.</p>","PeriodicalId":13524,"journal":{"name":"Inflammation","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142800611","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Complement Inhibition in Chronic Subdural Hematoma Fluid. 慢性硬膜下血肿液的补体抑制
IF 4.5 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-12-09 DOI: 10.1007/s10753-024-02210-3
Niklas Marklund, Shaian Zolfaghari, Gustaf Westerberg, Karsten Ruscher, Elisabet Englund, Henrietta Nittby Redebrandt

Background: Emerging data suggest a complex pathophysiology of chronic subdural hematoma (CSDH) to which an inflammatory response might contribute. The complement system is activated in acute traumatic setting, although its role in CSDH is unknown. To investigate the complement system in CSDH pathophysiology, we analyzed blood and hematoma fluid biomarkers, as well as immunohistochemistry of the CSDH membrane and dura.

Materials and methods: We simultaneously collected CSDH fluid and peripheral blood from 20 CSDH patients at the time of surgery. Biopsies of the dura mater and the CSDH capsule were obtained and analyzed by immunohistochemistry for C5b-C9 or C5a deposition. Biomarkers of inflammation and complement activation were analyzed by a 21-multiplex assay, including Adiponectin, Clusterin, Complement factor C9 and CRP. Complement factor C5a was analyzed separately by a commercial R-plex electrochemiluminescence assay.

Results: Ten biomarkers differed significantly between peripheral blood and paired CSDH of which two were significantly increased in CSDH fluid (Clusterin and Cystatin C). Eight of the significantly altered biomarkers were significantly decreased in CSDH fluid, including C5a, Complement 9 and Adiponectin. There was no immunoreactivity for C5a or the C5b-C9 membrane attack complex in the dura or CSDH membrane.

Conclusions: In CSDH levels of the complement inhibitor Clusterin were increased, whereas levels of C5a and C9 were decreased. Membrane attack complex C5b-C9 was not detected in the membrane or dura surrounding the CSDH. Inhibition of complement could lead to reduced clearance of debris in the CSDH as well as secondary inflammatory reactions.

背景:新出现的数据表明慢性硬膜下血肿(CSDH)的复杂病理生理可能与炎症反应有关。补体系统在急性创伤环境中被激活,尽管其在CSDH中的作用尚不清楚。为了研究补体系统在CSDH病理生理中的作用,我们分析了血液和血肿液生物标志物,以及CSDH膜和硬脑膜的免疫组织化学。材料和方法:我们在20例CSDH患者手术时同时采集CSDH液和外周血。取硬脑膜及CSDH囊组织切片,免疫组化检测C5b-C9或C5a沉积。炎症和补体激活的生物标志物通过21-多重试验进行分析,包括脂联素、Clusterin、补体因子C9和CRP。补体因子C5a通过商业R-plex电化学发光试验单独分析。结果:外周血和配对CSDH之间有10项生物标志物显著差异,其中CSDH液中有2项显著升高(Clusterin和Cystatin C), CSDH液中有8项显著改变的生物标志物显著降低,包括C5a、补体9和脂联素。硬脑膜和CSDH膜的C5a、C5b-C9膜攻击复合物无免疫反应性。结论:CSDH中补体抑制剂Clusterin水平升高,C5a和C9水平降低。膜攻击复合物C5b-C9未在CSDH周围的膜或硬脑膜中检测到。抑制补体可导致CSDH中碎片清除减少以及继发性炎症反应。
{"title":"Complement Inhibition in Chronic Subdural Hematoma Fluid.","authors":"Niklas Marklund, Shaian Zolfaghari, Gustaf Westerberg, Karsten Ruscher, Elisabet Englund, Henrietta Nittby Redebrandt","doi":"10.1007/s10753-024-02210-3","DOIUrl":"https://doi.org/10.1007/s10753-024-02210-3","url":null,"abstract":"<p><strong>Background: </strong>Emerging data suggest a complex pathophysiology of chronic subdural hematoma (CSDH) to which an inflammatory response might contribute. The complement system is activated in acute traumatic setting, although its role in CSDH is unknown. To investigate the complement system in CSDH pathophysiology, we analyzed blood and hematoma fluid biomarkers, as well as immunohistochemistry of the CSDH membrane and dura.</p><p><strong>Materials and methods: </strong>We simultaneously collected CSDH fluid and peripheral blood from 20 CSDH patients at the time of surgery. Biopsies of the dura mater and the CSDH capsule were obtained and analyzed by immunohistochemistry for C5b-C9 or C5a deposition. Biomarkers of inflammation and complement activation were analyzed by a 21-multiplex assay, including Adiponectin, Clusterin, Complement factor C9 and CRP. Complement factor C5a was analyzed separately by a commercial R-plex electrochemiluminescence assay.</p><p><strong>Results: </strong>Ten biomarkers differed significantly between peripheral blood and paired CSDH of which two were significantly increased in CSDH fluid (Clusterin and Cystatin C). Eight of the significantly altered biomarkers were significantly decreased in CSDH fluid, including C5a, Complement 9 and Adiponectin. There was no immunoreactivity for C5a or the C5b-C9 membrane attack complex in the dura or CSDH membrane.</p><p><strong>Conclusions: </strong>In CSDH levels of the complement inhibitor Clusterin were increased, whereas levels of C5a and C9 were decreased. Membrane attack complex C5b-C9 was not detected in the membrane or dura surrounding the CSDH. Inhibition of complement could lead to reduced clearance of debris in the CSDH as well as secondary inflammatory reactions.</p>","PeriodicalId":13524,"journal":{"name":"Inflammation","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142800610","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Lentiviral Injection of Inter-α-Trypsin Inhibitor Heavy Chain 4 Promotes Female Spinal Cord Injury Mice Recuperation by Diminishing Peripheral and Central Inflammation. 慢病毒注射间α-胰蛋白酶抑制剂重链4通过减少外周和中枢炎症促进雌性脊髓损伤小鼠恢复。
IF 4.5 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-12-09 DOI: 10.1007/s10753-024-02196-y
Jiaqi Li, Gang Liu

Inter-alpha-trypsin inhibitor heavy chain H4 (ITIH4) acts as a mediator of inflammation and extracellular matrix stabilization. The current study intended to delve into the impact of ITIH4 on locomotor performance, nerve injury, neuroinflammation, systemic inflammation, and the downstream pathway in spinal cord injury (SCI) mice. Overexpression lentivirus of ITIH4 (LV-ITIH4) and negative control lentivirus (LV-NC) were intravenously injected into adult C57BL/6 mice on 7 days before SCI surgery. All mice were euthanized on day 28 after SCI surgery, and their blood samples and spinal cord tissues were collected. Decreased relative gene expression and protein levels of ITIH4 were observed in SCI mice. LV-ITIH4 improved the locomotor performance compared to LV-NC in SCI mice. In spinal cord of SCI mice, LV-ITIH4 reduced apoptosis and increased survival of neurons compared to LV-NC. By comparison with LV-NC, LV-ITIH4 also reduced relative gene expressions of interleukin (IL)-6 and tumor necrosis factor-α in spinal cord of SCI mice. Moreover, LV-ITIH4 reduced microglia M1 polarization compared with LV-NC in spinal cord of SCI mice. In the serum, LV-ITIH4 decreased the protein levels of IL-6 and IL-1β compared to LV-NC in SCI mice. LV-ITIH4 also inhibited the nuclear factor kappa-B (NF-κB) pathway compared to LV-NC in spinal cord of SCI mice. ITIH4 enhances locomotor performance in SCI mice, and it inhibits nerve injury, neuroinflammation, systemic inflammation, and the NF-κB pathway in SCI mice.

胰蛋白酶间抑制剂重链H4 (ITIH4)作为炎症和细胞外基质稳定的介质。本研究旨在探讨ITIH4对脊髓损伤(SCI)小鼠运动性能、神经损伤、神经炎症、全身炎症及下游通路的影响。在脊髓损伤手术前7天,将ITIH4过表达慢病毒(LV-ITIH4)和阴性对照慢病毒(LV-NC)静脉注射到成年C57BL/6小鼠体内。所有小鼠均于脊髓损伤术后第28天实施安乐死,并采集其血液和脊髓组织。在脊髓损伤小鼠中,ITIH4的相对基因表达和蛋白水平均下降。与LV-NC相比,LV-ITIH4改善了脊髓损伤小鼠的运动能力。在脊髓损伤小鼠中,与LV-NC相比,LV-ITIH4减少了神经元的凋亡,提高了神经元的存活率。与LV-NC相比,LV-ITIH4还能降低脊髓损伤小鼠中白细胞介素(IL)-6和肿瘤坏死因子-α的相对基因表达。此外,与LV-NC相比,LV-ITIH4可降低脊髓小胶质细胞M1极化。与LV-NC相比,LV-ITIH4降低了脊髓损伤小鼠血清中IL-6和IL-1β的蛋白水平。与LV-NC相比,LV-ITIH4对脊髓核因子κ b (NF-κB)通路也有抑制作用。ITIH4可增强脊髓损伤小鼠的运动表现,抑制脊髓损伤小鼠的神经损伤、神经炎症、全身炎症和NF-κB通路。
{"title":"Lentiviral Injection of Inter-α-Trypsin Inhibitor Heavy Chain 4 Promotes Female Spinal Cord Injury Mice Recuperation by Diminishing Peripheral and Central Inflammation.","authors":"Jiaqi Li, Gang Liu","doi":"10.1007/s10753-024-02196-y","DOIUrl":"https://doi.org/10.1007/s10753-024-02196-y","url":null,"abstract":"<p><p>Inter-alpha-trypsin inhibitor heavy chain H4 (ITIH4) acts as a mediator of inflammation and extracellular matrix stabilization. The current study intended to delve into the impact of ITIH4 on locomotor performance, nerve injury, neuroinflammation, systemic inflammation, and the downstream pathway in spinal cord injury (SCI) mice. Overexpression lentivirus of ITIH4 (LV-ITIH4) and negative control lentivirus (LV-NC) were intravenously injected into adult C57BL/6 mice on 7 days before SCI surgery. All mice were euthanized on day 28 after SCI surgery, and their blood samples and spinal cord tissues were collected. Decreased relative gene expression and protein levels of ITIH4 were observed in SCI mice. LV-ITIH4 improved the locomotor performance compared to LV-NC in SCI mice. In spinal cord of SCI mice, LV-ITIH4 reduced apoptosis and increased survival of neurons compared to LV-NC. By comparison with LV-NC, LV-ITIH4 also reduced relative gene expressions of interleukin (IL)-6 and tumor necrosis factor-α in spinal cord of SCI mice. Moreover, LV-ITIH4 reduced microglia M1 polarization compared with LV-NC in spinal cord of SCI mice. In the serum, LV-ITIH4 decreased the protein levels of IL-6 and IL-1β compared to LV-NC in SCI mice. LV-ITIH4 also inhibited the nuclear factor kappa-B (NF-κB) pathway compared to LV-NC in spinal cord of SCI mice. ITIH4 enhances locomotor performance in SCI mice, and it inhibits nerve injury, neuroinflammation, systemic inflammation, and the NF-κB pathway in SCI mice.</p>","PeriodicalId":13524,"journal":{"name":"Inflammation","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142794686","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
TWEAK and TNFɑ Pro-inflammatory Soluble Cytokines and their Specific Autoantibodies Secretion in Multiple Sclerosis Patients. 多发性硬化症患者促炎可溶性细胞因子及其特异性自身抗体分泌。
IF 4.5 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-12-06 DOI: 10.1007/s10753-024-02205-0
Sylvie Carmona, Jehanne Aghzadi, Thierry Vincent, Pierre Labauge, Clarisse Carra-Dallière, Sylvain Lehmann, Sophie Desplat-Jégo, Xavier Ayrignac

Multiple sclerosis (MS) is a complex, chronic inflammatory disease of the central nervous system, where immune dysregulation plays a critical role. We sought to explore the modulation of the pro-inflammatory cytokines tumor necrosis factor-alpha (TNFɑ) and TNF-like weak inducer of apoptosis (TWEAK), along with their respective autoantibodies, TNAb and TWAb, and to decipher potential associations between these and clinical characteristics which could assist personalized therapy in MS. We also assessed the complementarity to leading candidate biomarkers in MS patient monitoring, namely, glial fibrillary acidic protein (GFAP) and neurofilament light chain (NfL). Serum levels of these cytokines and their autoantibodies were measured in 150 MS patients and 186 healthy controls (HC) by ELISA. We found that sTWEAK levels were significantly higher, while sTNFɑ levels were lower in MS patients compared to HC. Additionally, we detected TWAb in 10% of MS patients, a prevalence significantly higher than in HC (4%) and TNAb in only one patient. TWAb-positive patients were significantly younger, had lower EDSS scores, a shorter disease duration, and predominantly presented with the relapsing-remitting form of MS. Together, these results provide new actors to be considered in the development of a biomarker profiling panel to be used in MS patient management. Further research is warranted to elucidate the clinical significance of these autoantibodies and their role in MS neuroinflammatory modulation.

多发性硬化症(MS)是一种复杂的中枢神经系统慢性炎症性疾病,其中免疫失调起着关键作用。我们试图探索促炎细胞因子肿瘤坏死因子- α (TNF])和TNF样细胞凋亡弱诱导剂(TWEAK)的调节,以及它们各自的自身抗体TNAb和TWAb,并解读它们与临床特征之间的潜在关联,这些特征可能有助于MS的个性化治疗。我们还评估了MS患者监测中主要候选生物标志物的互补性,即:胶质原纤维酸性蛋白(GFAP)和神经丝轻链(NfL)。用ELISA法测定了150例MS患者和186例健康对照(HC)血清中这些细胞因子及其自身抗体的水平。我们发现,与HC相比,MS患者的sTWEAK水平明显较高,而sTNF _水平较低。此外,我们在10%的MS患者中检测到TWAb,患病率明显高于HC (4%), TNAb仅在1例患者中检测到。twab阳性患者明显更年轻,EDSS评分较低,病程较短,主要表现为MS复发缓解型,这些结果为开发用于MS患者管理的生物标志物分析小组提供了新的因素。需要进一步的研究来阐明这些自身抗体的临床意义及其在多发性硬化症神经炎症调节中的作用。
{"title":"TWEAK and TNFɑ Pro-inflammatory Soluble Cytokines and their Specific Autoantibodies Secretion in Multiple Sclerosis Patients.","authors":"Sylvie Carmona, Jehanne Aghzadi, Thierry Vincent, Pierre Labauge, Clarisse Carra-Dallière, Sylvain Lehmann, Sophie Desplat-Jégo, Xavier Ayrignac","doi":"10.1007/s10753-024-02205-0","DOIUrl":"https://doi.org/10.1007/s10753-024-02205-0","url":null,"abstract":"<p><p>Multiple sclerosis (MS) is a complex, chronic inflammatory disease of the central nervous system, where immune dysregulation plays a critical role. We sought to explore the modulation of the pro-inflammatory cytokines tumor necrosis factor-alpha (TNFɑ) and TNF-like weak inducer of apoptosis (TWEAK), along with their respective autoantibodies, TNAb and TWAb, and to decipher potential associations between these and clinical characteristics which could assist personalized therapy in MS. We also assessed the complementarity to leading candidate biomarkers in MS patient monitoring, namely, glial fibrillary acidic protein (GFAP) and neurofilament light chain (NfL). Serum levels of these cytokines and their autoantibodies were measured in 150 MS patients and 186 healthy controls (HC) by ELISA. We found that sTWEAK levels were significantly higher, while sTNFɑ levels were lower in MS patients compared to HC. Additionally, we detected TWAb in 10% of MS patients, a prevalence significantly higher than in HC (4%) and TNAb in only one patient. TWAb-positive patients were significantly younger, had lower EDSS scores, a shorter disease duration, and predominantly presented with the relapsing-remitting form of MS. Together, these results provide new actors to be considered in the development of a biomarker profiling panel to be used in MS patient management. Further research is warranted to elucidate the clinical significance of these autoantibodies and their role in MS neuroinflammatory modulation.</p>","PeriodicalId":13524,"journal":{"name":"Inflammation","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142785320","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
IRF-1 Regulates Mitochondrial Respiration and Intrinsic Apoptosis Under Metabolic Stress through ATP Synthase Ancillary Factor TMEM70. IRF-1通过ATP合酶辅助因子TMEM70调控代谢应激下线粒体呼吸和内在凋亡
IF 4.5 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-12-06 DOI: 10.1007/s10753-024-02209-w
ChongXiu Sun, Haotian Sun, Jiahao Wei, Xing Fan, Scott I Simon, Anthony G Passerini

Mitochondrial dysfunction, which can be caused by metabolic stressors such as oxidized low-density lipoprotein (oxLDL), sensitizes the endothelium to pathological changes. The transcription factor interferon regulatory factor 1 (IRF-1) is a master regulator of inflammation, previously shown to promote oxLDL-induced inflammatory pyroptosis in human aortic endothelial cells (HAEC). However, a presumed role for IRF-1 in regulating the intrinsic apoptotic pathway in response to metabolic stress has not been demonstrated. Here targeted deletion of IRF-1 by siRNA in HAEC aggravated oxLDL-induced, mitochondria-mediated intrinsic apoptosis, as evidenced by increased Caspase-3 and Caspase-9 activation, and chromosomal DNA breakage. The increased apoptosis was concomitant with accumulation of mitochondrial ROS, decrease in intracellular ATP production and respiratory oxygen consumption, and abnormal mitochondrial structure. RNA profiling of endothelial cells isolated from wild type and Irf1 knockout mice, followed by quantitative PCR, luciferase activity assay and chromatin immunoprecipitation (ChIP), revealed that IRF-1 directly regulated the expression of transmembrane protein 70 (TMEM70), an ancillary factor required for the assembly of ATP synthase and conversion of an electrochemical gradient to ATP synthesis. Mirroring the effect of IRF1 knockdown, depletion of TMEM70 in HAEC resulted in impaired mitochondrial function and enhanced cell apoptosis. In contrast, overexpression of TMEM70 rescued ATP biosynthesis and suppressed apoptosis in oxLDL-treated, IRF-1-deficient HAEC. These results reveal a novel homeostatic role for IRF-1 in the regulation of mitochondrial function and associated stress-induced apoptosis.

线粒体功能障碍可由代谢应激源(如氧化低密度脂蛋白(oxLDL))引起,使内皮细胞对病理变化敏感。转录因子干扰素调节因子1 (IRF-1)是炎症的主要调节因子,先前显示可促进人主动脉内皮细胞(HAEC)中氧化低密度脂蛋白诱导的炎症性焦亡。然而,IRF-1在代谢应激下调节固有凋亡通路中的作用尚未得到证实。在HAEC中,siRNA靶向缺失IRF-1会加重氧化低密度脂蛋白诱导的线粒体介导的内在凋亡,Caspase-3和Caspase-9激活增加,染色体DNA断裂。细胞凋亡的增加伴随着线粒体ROS的积累、细胞内ATP生成和呼吸耗氧量的减少以及线粒体结构的异常。对野生型和Irf1基因敲除小鼠内皮细胞的RNA谱分析、定量PCR、荧光素酶活性测定和染色质免疫沉淀(ChIP)分析显示,IRF-1直接调节跨膜蛋白70 (TMEM70)的表达,TMEM70是ATP合成酶组装和电化学梯度转化为ATP合成所需的辅助因子。与IRF1敲低的作用类似,在HAEC中TMEM70的缺失导致线粒体功能受损和细胞凋亡增强。相比之下,在氧化ldl处理的irf -1缺陷HAEC中,TMEM70的过表达挽救了ATP的生物合成并抑制了细胞凋亡。这些结果揭示了IRF-1在调节线粒体功能和相关应激诱导的细胞凋亡中的一种新的稳态作用。
{"title":"IRF-1 Regulates Mitochondrial Respiration and Intrinsic Apoptosis Under Metabolic Stress through ATP Synthase Ancillary Factor TMEM70.","authors":"ChongXiu Sun, Haotian Sun, Jiahao Wei, Xing Fan, Scott I Simon, Anthony G Passerini","doi":"10.1007/s10753-024-02209-w","DOIUrl":"https://doi.org/10.1007/s10753-024-02209-w","url":null,"abstract":"<p><p>Mitochondrial dysfunction, which can be caused by metabolic stressors such as oxidized low-density lipoprotein (oxLDL), sensitizes the endothelium to pathological changes. The transcription factor interferon regulatory factor 1 (IRF-1) is a master regulator of inflammation, previously shown to promote oxLDL-induced inflammatory pyroptosis in human aortic endothelial cells (HAEC). However, a presumed role for IRF-1 in regulating the intrinsic apoptotic pathway in response to metabolic stress has not been demonstrated. Here targeted deletion of IRF-1 by siRNA in HAEC aggravated oxLDL-induced, mitochondria-mediated intrinsic apoptosis, as evidenced by increased Caspase-3 and Caspase-9 activation, and chromosomal DNA breakage. The increased apoptosis was concomitant with accumulation of mitochondrial ROS, decrease in intracellular ATP production and respiratory oxygen consumption, and abnormal mitochondrial structure. RNA profiling of endothelial cells isolated from wild type and Irf1 knockout mice, followed by quantitative PCR, luciferase activity assay and chromatin immunoprecipitation (ChIP), revealed that IRF-1 directly regulated the expression of transmembrane protein 70 (TMEM70), an ancillary factor required for the assembly of ATP synthase and conversion of an electrochemical gradient to ATP synthesis. Mirroring the effect of IRF1 knockdown, depletion of TMEM70 in HAEC resulted in impaired mitochondrial function and enhanced cell apoptosis. In contrast, overexpression of TMEM70 rescued ATP biosynthesis and suppressed apoptosis in oxLDL-treated, IRF-1-deficient HAEC. These results reveal a novel homeostatic role for IRF-1 in the regulation of mitochondrial function and associated stress-induced apoptosis.</p>","PeriodicalId":13524,"journal":{"name":"Inflammation","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142785404","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
NOD3 Reduces Sepsis-Induced Acute Lung Injury by Regulating the Activation of NLRP3 Inflammasome and the Polarization of Alveolar Macrophages. NOD3通过调节NLRP3炎性体的激活和肺泡巨噬细胞的极化,减轻脓毒症诱导的急性肺损伤。
IF 4.5 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-12-02 DOI: 10.1007/s10753-024-02197-x
Yue Zhao, Yongran Wu, Lianlian Qu, Yingying Hu, Shengwen Sun, Ruishan Yao, Ruiting Li

The pathogenesis of sepsis-induced Acute lung injury (ALI) progresses rapidly, and no effective treatment drugs are known, resulting in a high mortality rate. NLR family pyrin domain containing 3 (NLRP3) inflammasome activation plays an important role in the pathological progression of ALI, and often coincide with the inflammatory activation and polarization of macrophages. NLR family CARD domain-containing protein 3 (NOD3) was reported protecting against sepsis-induced pulmonary pathological injury and inhibiting the inflammatory response in lung tissue. NOD3 can also inhibit NLRP3 inflammasome activation by competitively inhibiting the binding of pro-caspase-1 to apoptosis-related ASC or reducing NLRP3/cryopyrin-induced ASC speckle formation. In this study, we aimed to explore whether NOD3 decrease sepsis-induced lung injury by interfering with NLRP3 inflammasome activation and regulating alveolar macrophages (AMs) polarization. To investigate whether NOD3 reduce sepsis-induced ALI by inhibiting the activation of NLRP3 inflammasome to regulate the polarization of AMs. Sepsis-induced WT (C57BL/6) and NLRC3-/--C57BL/6 mice ALI models were established by intraperitoneal injection of lipopolysaccharide (LPS). In vitro experiments, AMs and bone marrow-derived macrophages (BMDMs) were isolated from WT and NLRC3-/- mice. Using in vivo and in vitro experiments, we found that NOD3 knockout promoted the sepsis-induced inflammatory response in lung tissue. In addition, NOD3 knockout promoted the activation of the TRAF6-NF-κB signaling pathway and the NLRP3 inflammasome in AMs, enhanced the M1-type polarization of AMs and decreased the M2-type polarization of AMs in sepsis-induced lung injury model mice. NOD3 interfered with NLRP3 inflammasome activation by inhibiting NLRP3 inflammasome assembly or negatively regulating the TRAF6-NF-κB signaling pathway, and regulating the polarization of AMs, thereby alleviating sepsis-induced lung injury.

脓毒症引起的急性肺损伤(ALI)的发病机制进展迅速,目前尚无有效的治疗药物,死亡率高。NLR家族pyrin domain containing 3 (NLRP3)炎性小体激活在ALI的病理进展中起重要作用,且常与巨噬细胞的炎症激活和极化相吻合。据报道,NLR家族CARD结构域蛋白3 (NOD3)对脓毒症诱导的肺病理损伤具有保护作用,并抑制肺组织的炎症反应。NOD3还可以通过竞争性抑制前caspase-1与凋亡相关的ASC结合或减少NLRP3/cryopyrin诱导的ASC斑点形成来抑制NLRP3炎性小体的激活。在本研究中,我们旨在探讨NOD3是否通过干扰NLRP3炎性体激活和调节肺泡巨噬细胞(AMs)极化来减轻败血症诱导的肺损伤。探讨NOD3是否通过抑制NLRP3炎性小体的激活来调节am的极化,从而减少败血症诱导的ALI。通过腹腔注射脂多糖(LPS)建立脓毒症诱导的WT (C57BL/6)和NLRC3-/—C57BL/6小鼠ALI模型。在体外实验中,从WT和NLRC3-/-小鼠中分离出AMs和骨髓源性巨噬细胞(bmdm)。通过体内和体外实验,我们发现NOD3敲除可促进脓毒症诱导的肺组织炎症反应。此外,敲除NOD3可促进脓毒症肺损伤模型小鼠AMs中TRAF6-NF-κB信号通路和NLRP3炎性小体的激活,增强AMs的m1型极化,降低AMs的m2型极化。NOD3通过抑制NLRP3炎性小体组装或负调控TRAF6-NF-κB信号通路,调控AMs的极化,从而干扰NLRP3炎性小体的激活,从而减轻败血症诱导的肺损伤。
{"title":"NOD3 Reduces Sepsis-Induced Acute Lung Injury by Regulating the Activation of NLRP3 Inflammasome and the Polarization of Alveolar Macrophages.","authors":"Yue Zhao, Yongran Wu, Lianlian Qu, Yingying Hu, Shengwen Sun, Ruishan Yao, Ruiting Li","doi":"10.1007/s10753-024-02197-x","DOIUrl":"https://doi.org/10.1007/s10753-024-02197-x","url":null,"abstract":"<p><p>The pathogenesis of sepsis-induced Acute lung injury (ALI) progresses rapidly, and no effective treatment drugs are known, resulting in a high mortality rate. NLR family pyrin domain containing 3 (NLRP3) inflammasome activation plays an important role in the pathological progression of ALI, and often coincide with the inflammatory activation and polarization of macrophages. NLR family CARD domain-containing protein 3 (NOD3) was reported protecting against sepsis-induced pulmonary pathological injury and inhibiting the inflammatory response in lung tissue. NOD3 can also inhibit NLRP3 inflammasome activation by competitively inhibiting the binding of pro-caspase-1 to apoptosis-related ASC or reducing NLRP3/cryopyrin-induced ASC speckle formation. In this study, we aimed to explore whether NOD3 decrease sepsis-induced lung injury by interfering with NLRP3 inflammasome activation and regulating alveolar macrophages (AMs) polarization. To investigate whether NOD3 reduce sepsis-induced ALI by inhibiting the activation of NLRP3 inflammasome to regulate the polarization of AMs. Sepsis-induced WT (C57BL/6) and NLRC3<sup>-</sup><sup>/</sup><sup>-</sup>-C57BL/6 mice ALI models were established by intraperitoneal injection of lipopolysaccharide (LPS). In vitro experiments, AMs and bone marrow-derived macrophages (BMDMs) were isolated from WT and NLRC3<sup>-</sup><sup>/</sup><sup>-</sup> mice. Using in vivo and in vitro experiments, we found that NOD3 knockout promoted the sepsis-induced inflammatory response in lung tissue. In addition, NOD3 knockout promoted the activation of the TRAF6-NF-κB signaling pathway and the NLRP3 inflammasome in AMs, enhanced the M1-type polarization of AMs and decreased the M2-type polarization of AMs in sepsis-induced lung injury model mice. NOD3 interfered with NLRP3 inflammasome activation by inhibiting NLRP3 inflammasome assembly or negatively regulating the TRAF6-NF-κB signaling pathway, and regulating the polarization of AMs, thereby alleviating sepsis-induced lung injury.</p>","PeriodicalId":13524,"journal":{"name":"Inflammation","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142768602","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Oroxylin A-induced Trained Immunity Promotes LC3-associated Phagocytosis in Macrophage in Protecting Mice Against Sepsis. Oroxylin A诱导的训练免疫促进巨噬细胞中与LC3相关的吞噬作用,保护小鼠免受败血症影响
IF 4.5 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-12-01 Epub Date: 2024-05-13 DOI: 10.1007/s10753-024-02033-2
Lijie Yin, Ziqian Bing, Yaojun Zheng, Yuchen Pan, Yue Dong, Jiali Wang, Renjie Luo, Yue Zhao, Huan Dou, Yayi Hou

Sepsis is defined as a dysregulated host response to infection that leads to multiorgan failure. Innate immune memory, i.e., "trained immunity", can result in stronger immune responses and provide protection against various infections. Many biological agents, including β-glucan, can induce trained immunity, but these stimuli may cause uncontrolled inflammation. Oroxylin A (OA) is an active flavonoid compound that is derived from Scutellaria baicalensis. OA is an agonist for inducing trained immunity in vivo and in vitro, and β-glucan was used as a positive control. The protective effects of OA-induced trained immunity were evaluated in mouse models that were established by either lipopolysaccharide (LPS) administration or caecal ligation and puncture (CLP). The expression of inflammatory factors and signaling pathway components involved in trained immunity was evaluated in vitro using qRT‒PCR, western blotting (WB) and enzyme-linked immunosorbent assay (ELISA). Flow cytometry and confocal microscopy were used to examine reactive oxygen species (ROS) levels and phagocytosis in trained macrophages. A PCR array was used to screen genes that were differentially expressed in trained macrophages. Here, we revealed that OA alleviated sepsis via trained immunity. OA-treated macrophages displayed increased glycolysis and mTOR phosphorylation, and mTOR inhibitors suppressed OA-induced trained immunity by effectively reprogramming macrophages. The PCR array revealed key genes in the mTOR signaling pathway in OA-treated macrophages. Furthermore, OA targeted the Dectin-1-syk axis to promote LC3-associated phagocytosis (LAP) by trained macrophages, thereby enhancing the ability of these macrophages to protect against infection. This ability could be transferred to a new host via the adoptive transfer of peritoneal macrophages. This study is the first to provide new insights into the potential of OA-induced trained immunity to be used as a strategy to protect mice against sepsis by promoting LAP by macrophages.

败血症的定义是宿主对感染的反应失调,导致多器官功能衰竭。先天性免疫记忆,即 "训练有素的免疫",可产生更强的免疫反应,并提供抵御各种感染的保护。包括β-葡聚糖在内的许多生物制剂都能诱导训练有素的免疫,但这些刺激可能会导致炎症失控。Oroxylin A(OA)是从黄芩中提取的一种活性黄酮化合物。OA 是诱导体内和体外训练免疫的激动剂,β-葡聚糖被用作阳性对照。在脂多糖(LPS)给药或盲肠结扎和穿刺(CLP)建立的小鼠模型中,评估了 OA 诱导训练免疫的保护作用。使用 qRT-PCR、Western 印迹(WB)和酶联免疫吸附试验(ELISA)在体外评估了参与训练免疫的炎症因子和信号通路成分的表达。流式细胞术和共聚焦显微镜用于检测活性氧(ROS)水平和训练巨噬细胞的吞噬能力。PCR 阵列用于筛选训练巨噬细胞中差异表达的基因。在这里,我们发现 OA 可通过训练免疫缓解败血症。经OA处理的巨噬细胞显示糖酵解和mTOR磷酸化增加,而mTOR抑制剂通过有效重编程巨噬细胞抑制了OA诱导的训练免疫。PCR 阵列揭示了经 OA 处理的巨噬细胞中 mTOR 信号通路的关键基因。此外,OA靶向Dectin-1-syk轴,促进训练有素的巨噬细胞的LC3相关吞噬(LAP),从而增强这些巨噬细胞抵御感染的能力。这种能力可以通过腹腔巨噬细胞的领养转移转移到新的宿主身上。这项研究首次对OA诱导的训练型免疫通过促进巨噬细胞的LAP作为一种保护小鼠免受败血症的策略的潜力提供了新的见解。
{"title":"Oroxylin A-induced Trained Immunity Promotes LC3-associated Phagocytosis in Macrophage in Protecting Mice Against Sepsis.","authors":"Lijie Yin, Ziqian Bing, Yaojun Zheng, Yuchen Pan, Yue Dong, Jiali Wang, Renjie Luo, Yue Zhao, Huan Dou, Yayi Hou","doi":"10.1007/s10753-024-02033-2","DOIUrl":"10.1007/s10753-024-02033-2","url":null,"abstract":"<p><p>Sepsis is defined as a dysregulated host response to infection that leads to multiorgan failure. Innate immune memory, i.e., \"trained immunity\", can result in stronger immune responses and provide protection against various infections. Many biological agents, including β-glucan, can induce trained immunity, but these stimuli may cause uncontrolled inflammation. Oroxylin A (OA) is an active flavonoid compound that is derived from Scutellaria baicalensis. OA is an agonist for inducing trained immunity in vivo and in vitro, and β-glucan was used as a positive control. The protective effects of OA-induced trained immunity were evaluated in mouse models that were established by either lipopolysaccharide (LPS) administration or caecal ligation and puncture (CLP). The expression of inflammatory factors and signaling pathway components involved in trained immunity was evaluated in vitro using qRT‒PCR, western blotting (WB) and enzyme-linked immunosorbent assay (ELISA). Flow cytometry and confocal microscopy were used to examine reactive oxygen species (ROS) levels and phagocytosis in trained macrophages. A PCR array was used to screen genes that were differentially expressed in trained macrophages. Here, we revealed that OA alleviated sepsis via trained immunity. OA-treated macrophages displayed increased glycolysis and mTOR phosphorylation, and mTOR inhibitors suppressed OA-induced trained immunity by effectively reprogramming macrophages. The PCR array revealed key genes in the mTOR signaling pathway in OA-treated macrophages. Furthermore, OA targeted the Dectin-1-syk axis to promote LC3-associated phagocytosis (LAP) by trained macrophages, thereby enhancing the ability of these macrophages to protect against infection. This ability could be transferred to a new host via the adoptive transfer of peritoneal macrophages. This study is the first to provide new insights into the potential of OA-induced trained immunity to be used as a strategy to protect mice against sepsis by promoting LAP by macrophages.</p>","PeriodicalId":13524,"journal":{"name":"Inflammation","volume":" ","pages":"2196-2214"},"PeriodicalIF":4.5,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140912102","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Kidney Injury in a Murine Hemorrhagic Shock/Resuscitation Model Is Alleviated by sulforaphane's Anti-Inflammatory and Antioxidant Action. 红景天的抗炎和抗氧化作用可缓解小鼠失血性休克/复苏模型中的肾损伤
IF 4.5 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-12-01 Epub Date: 2024-07-18 DOI: 10.1007/s10753-024-02106-2
You Li, Kang Qin, Weiqiang Liang, Weining Yan, Athanassios Fragoulis, Thomas Pufe, Eva Miriam Buhl, Qun Zhao, Johannes Greven

Hemorrhagic shock/resuscitation (HS/R) can lead to acute kidney injury, mainly manifested as oxidative stress and inflammatory injury in the renal tubular epithelial cells, as well as abnormal autophagy and apoptosis. Sulforaphane (SFN), an agonist of the nuclear factor-erythroid factor 2-related factor 2 (Nrf2) signaling pathway, is involved in multiple biological activities, such as anti-inflammatory, antioxidant, autophagy, and apoptosis regulation. This study investigated the effect of SFN on acute kidney injury after HS/R in mice. Hemorrhagic shock was induced in mice by controlling the arterial blood pressure at a range of 35-45 mmHg for 90 min within arterial blood withdrawal. Fluid resuscitation was carried out by reintroducing withdrawn blood and 0.9% NaCl. We found that SFN suppressed the elevation of urea nitrogen and serum creatinine levels in the blood induced by HS/R. SFN mitigated pathological alterations including swollen renal tubules and renal casts in kidney tissue of HS/R mice. Inflammation levels and oxidative stress were significantly downregulated in mouse kidney tissue after SFN administration. In addition, the kidney tissue of HS/R mice showed high levels of autophagosomes as observed by electron microscopy. However, SFN can further enhance the formation of autophagosomes in the HS/R + SFN group. SFN also increased autophagy-related proteins Beclin1 expression and suppressed P62 expression, while increasing the ratio of microtubule-associated protein 1 light chain 3 (LC3)-II and LC3-I (LC3-II/LC3-I). SFN also effectively decreased cleaved caspase-3 level and enhanced the ratio of anti-apoptotic protein B cell lymphoma 2 and Bcl2-associated X protein (Bcl2/Bax). Collectively, SFN effectively inhibited inflammation and oxidative stress, enhanced autophagy, thereby reducing HS/R-induced kidney injury and apoptosis levels in mouse kidneys.

失血性休克/复苏(HS/R)可导致急性肾损伤,主要表现为肾小管上皮细胞的氧化应激和炎症损伤,以及自噬和细胞凋亡异常。菔素(SFN)是核因子-红细胞生成因子2相关因子2(Nrf2)信号通路的激动剂,参与多种生物活性,如抗炎、抗氧化、自噬和细胞凋亡调节。本研究探讨了 SFN 对小鼠 HS/R 后急性肾损伤的影响。通过控制动脉血压在 35-45 mmHg 范围内 90 分钟,在动脉血抽取过程中诱发小鼠失血性休克。通过重新输入抽取的血液和0.9%氯化钠进行液体复苏。我们发现,SFN 可抑制 HS/R 引起的血液中尿素氮和血清肌酐水平的升高。SFN减轻了HS/R小鼠肾组织的病理改变,包括肾小管肿胀和肾铸型。服用 SFN 后,小鼠肾脏组织中的炎症水平和氧化应激明显降低。此外,通过电子显微镜观察,HS/R 小鼠的肾组织显示出高水平的自噬体。然而,在 HS/R + SFN 组中,SFN 可进一步促进自噬体的形成。SFN 还能增加自噬相关蛋白 Beclin1 的表达,抑制 P62 的表达,同时增加微管相关蛋白 1 轻链 3(LC3)-II 和 LC3-I 的比例(LC3-II/LC3-I)。SFN还能有效降低裂解的Caspase-3水平,提高抗凋亡蛋白B细胞淋巴瘤2和Bcl2相关X蛋白(Bcl2/Bax)的比例。总之,SFN能有效抑制炎症和氧化应激,增强自噬作用,从而减轻HS/R引起的肾损伤,降低小鼠肾脏的细胞凋亡水平。
{"title":"Kidney Injury in a Murine Hemorrhagic Shock/Resuscitation Model Is Alleviated by sulforaphane's Anti-Inflammatory and Antioxidant Action.","authors":"You Li, Kang Qin, Weiqiang Liang, Weining Yan, Athanassios Fragoulis, Thomas Pufe, Eva Miriam Buhl, Qun Zhao, Johannes Greven","doi":"10.1007/s10753-024-02106-2","DOIUrl":"10.1007/s10753-024-02106-2","url":null,"abstract":"<p><p>Hemorrhagic shock/resuscitation (HS/R) can lead to acute kidney injury, mainly manifested as oxidative stress and inflammatory injury in the renal tubular epithelial cells, as well as abnormal autophagy and apoptosis. Sulforaphane (SFN), an agonist of the nuclear factor-erythroid factor 2-related factor 2 (Nrf2) signaling pathway, is involved in multiple biological activities, such as anti-inflammatory, antioxidant, autophagy, and apoptosis regulation. This study investigated the effect of SFN on acute kidney injury after HS/R in mice. Hemorrhagic shock was induced in mice by controlling the arterial blood pressure at a range of 35-45 mmHg for 90 min within arterial blood withdrawal. Fluid resuscitation was carried out by reintroducing withdrawn blood and 0.9% NaCl. We found that SFN suppressed the elevation of urea nitrogen and serum creatinine levels in the blood induced by HS/R. SFN mitigated pathological alterations including swollen renal tubules and renal casts in kidney tissue of HS/R mice. Inflammation levels and oxidative stress were significantly downregulated in mouse kidney tissue after SFN administration. In addition, the kidney tissue of HS/R mice showed high levels of autophagosomes as observed by electron microscopy. However, SFN can further enhance the formation of autophagosomes in the HS/R + SFN group. SFN also increased autophagy-related proteins Beclin1 expression and suppressed P62 expression, while increasing the ratio of microtubule-associated protein 1 light chain 3 (LC3)-II and LC3-I (LC3-II/LC3-I). SFN also effectively decreased cleaved caspase-3 level and enhanced the ratio of anti-apoptotic protein B cell lymphoma 2 and Bcl2-associated X protein (Bcl2/Bax). Collectively, SFN effectively inhibited inflammation and oxidative stress, enhanced autophagy, thereby reducing HS/R-induced kidney injury and apoptosis levels in mouse kidneys.</p>","PeriodicalId":13524,"journal":{"name":"Inflammation","volume":" ","pages":"2215-2227"},"PeriodicalIF":4.5,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141633423","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Esaxerenone Inhibits Interferon-γ Induced Pyroptosis of Macrophages in the Lungs of Aldosterone-treated Mice. 依沙硒酮抑制干扰素-γ诱导的醛固酮治疗小鼠肺部巨噬细胞的嗜热现象
IF 4.5 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-12-01 Epub Date: 2024-05-07 DOI: 10.1007/s10753-024-02030-5
Jingyue Chang, Xiaomeng Gao, Fan Yang, Panpan Qiang, Lili Fan, Ziqian Liu, Tatsuo Shimosawa, Qingyou Xu, Yi Chang

Lung immune cells such as lymphocytes and macrophages can induce an inflammatory response due to the activation of mineralocorticoid receptor (MR), which is manifested by the infiltration of inflammatory cells and the secretion of inflammatory cytokines and subsequent apoptosis, pyroptosis and necrosis of intrinsic lung cells and immune cells. Macrophages are immune cells that are abundant in the lung and act as the first line of defense against pathogens but are also aggravating factors of infection. The activation of the renin-angiotensin-aldosterone system (RAAS), especially aldosterone-stimulated MR activation, can induce macrophage and CD8+ T cell aggregation and the secretion of cytokines such as tumor necrosis factor-α (TNF-α) and interferon-gamma (IFN-γ). Increased IFN-γ secretion can induce macrophage pyroptosis and the release of interleukin 1-β (IL-1β), aggravating lung injury. In this study, lung injury in C57BL/6 mice was induced by subcutaneous micro-osmotic pump infusion of aldosterone. After 12 weeks of administration, the kidney, heart, blood vessels and lungs all showed obvious inflammatory injury, which manifested as rapid accumulation of macrophages. The overexpression of IFN-γ in the lungs of aldosterone-treated mice and the stimulation of MH-S and RAW264.7 alveolar macrophages (AMs) with aldosterone in vitro showed that IFN-γ induced pyroptosis of macrophages via the activation of the inflammasome, and the MR blocker esaxerenone effectively inhibited this effect and alleviated lung injury. In addition, IFN-γ secreted by CD8+ T cells is associated with macrophage pyroptosis. In conclusion, the inhibition of macrophage pyroptosis can effectively alleviate lung injury.

淋巴细胞和巨噬细胞等肺部免疫细胞可因矿质皮质激素受体(MR)的激活而诱发炎症反应,表现为炎症细胞的浸润和炎症细胞因子的分泌,以及随之而来的肺固有细胞和免疫细胞的凋亡、热解和坏死。巨噬细胞是肺部大量存在的免疫细胞,是抵御病原体的第一道防线,但也是加重感染的因素。肾素-血管紧张素-醛固酮系统(RAAS)的激活,尤其是醛固酮刺激的 MR 激活,可诱导巨噬细胞和 CD8+ T 细胞聚集,并分泌肿瘤坏死因子-α(TNF-α)和γ 干扰素(IFN-γ)等细胞因子。IFN-γ 分泌增加可诱导巨噬细胞脓毒症和白细胞介素 1-β (IL-1β)的释放,从而加重肺损伤。本研究通过皮下微渗透泵输注醛固酮诱导 C57BL/6 小鼠肺损伤。给药 12 周后,肾脏、心脏、血管和肺部均出现明显的炎症损伤,表现为巨噬细胞的快速聚集。IFN-γ 在醛固酮治疗小鼠肺部的过表达以及体外醛固酮对 MH-S 和 RAW264.7 肺泡巨噬细胞(AMs)的刺激表明,IFN-γ 通过激活炎症小体诱导巨噬细胞的热凋亡,而磁共振阻断剂埃沙塞酮能有效抑制这种效应并减轻肺损伤。此外,CD8+ T 细胞分泌的 IFN-γ 与巨噬细胞的脓毒症有关。总之,抑制巨噬细胞脓毒症可有效缓解肺损伤。
{"title":"Esaxerenone Inhibits Interferon-γ Induced Pyroptosis of Macrophages in the Lungs of Aldosterone-treated Mice.","authors":"Jingyue Chang, Xiaomeng Gao, Fan Yang, Panpan Qiang, Lili Fan, Ziqian Liu, Tatsuo Shimosawa, Qingyou Xu, Yi Chang","doi":"10.1007/s10753-024-02030-5","DOIUrl":"10.1007/s10753-024-02030-5","url":null,"abstract":"<p><p>Lung immune cells such as lymphocytes and macrophages can induce an inflammatory response due to the activation of mineralocorticoid receptor (MR), which is manifested by the infiltration of inflammatory cells and the secretion of inflammatory cytokines and subsequent apoptosis, pyroptosis and necrosis of intrinsic lung cells and immune cells. Macrophages are immune cells that are abundant in the lung and act as the first line of defense against pathogens but are also aggravating factors of infection. The activation of the renin-angiotensin-aldosterone system (RAAS), especially aldosterone-stimulated MR activation, can induce macrophage and CD8<sup>+</sup> T cell aggregation and the secretion of cytokines such as tumor necrosis factor-α (TNF-α) and interferon-gamma (IFN-γ). Increased IFN-γ secretion can induce macrophage pyroptosis and the release of interleukin 1-β (IL-1β), aggravating lung injury. In this study, lung injury in C57BL/6 mice was induced by subcutaneous micro-osmotic pump infusion of aldosterone. After 12 weeks of administration, the kidney, heart, blood vessels and lungs all showed obvious inflammatory injury, which manifested as rapid accumulation of macrophages. The overexpression of IFN-γ in the lungs of aldosterone-treated mice and the stimulation of MH-S and RAW264.7 alveolar macrophages (AMs) with aldosterone in vitro showed that IFN-γ induced pyroptosis of macrophages via the activation of the inflammasome, and the MR blocker esaxerenone effectively inhibited this effect and alleviated lung injury. In addition, IFN-γ secreted by CD8<sup>+</sup> T cells is associated with macrophage pyroptosis. In conclusion, the inhibition of macrophage pyroptosis can effectively alleviate lung injury.</p>","PeriodicalId":13524,"journal":{"name":"Inflammation","volume":" ","pages":"2145-2158"},"PeriodicalIF":4.5,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140851478","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeted Demethylation of FOXP3-TSDR Enhances the Suppressive Capacity of STAT6-deficient Inducible T Regulatory Cells. FOXP3-TSDR的靶向去甲基化增强了STAT6缺陷诱导性T调节细胞的抑制能力
IF 4.5 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-12-01 Epub Date: 2024-05-03 DOI: 10.1007/s10753-024-02031-4
Rubén D Arroyo-Olarte, Juan C Flores-Castelán, Leonel Armas-López, Galileo Escobedo, Luis I Terrazas, Federico Ávila-Moreno, Sonia Leon-Cabrera

In vitro induced T regulatory cells (iTregs) are promising for addressing inflammation-driven diseases. However, current protocols for the generation and expansion of iTregs fail to induce extensive demethylation of the Treg-specific demethylated region (TSDR) within the FOXP3 gene, recognized as the master regulator for regulatory T cells (Tregs). This deficiency results in the rapid loss of Foxp3 expression and an unstable regulatory phenotype. Nevertheless, inhibition of STAT6 signaling effectively stabilizes Foxp3 expression in iTregs. Thus, this study aimed to develop a protocol combining epigenetic editing with STAT6 deficiency to improve iTregs' ability to maintain stable suppressive function and a functional phenotype. Our findings demonstrate that the combination of STAT6 deficiency (STAT6-/-) with targeted demethylation of the TSDR using a CRISPR-TET1 tool leads to extensive demethylation of FOXP3-TSDR. Demethylation in STAT6-/- iTregs was associated with enhanced expression of Foxp3 and suppressive markers such as CTLA-4, PD-1, IL-10, and TGF-β. Furthermore, the edited STAT6-/- iTregs exhibited an increased capacity to suppress CD8+ and CD4+ lymphocytes and could more efficiently impair Th1-signature gene expression compared to conventional iTregs. In conclusion, the deactivation of STAT6 and TSDR-targeted demethylation via CRISPR-TET1 is sufficient to induce iTregs with heightened stability and increased suppressive capacity, offering potential applications against inflammatory and autoimmune diseases.

体外诱导 T 调节细胞(iTregs)有望用于治疗炎症驱动的疾病。然而,目前生成和扩增 iTregs 的方案未能诱导 FOXP3 基因中 Treg 特异性去甲基化区域(TSDR)的广泛去甲基化,而 FOXP3 基因被认为是调节性 T 细胞(Tregs)的主调节因子。这种缺陷会导致 Foxp3 表达的快速丧失和不稳定的调节表型。然而,抑制 STAT6 信号传导可有效稳定 iTregs 中 Foxp3 的表达。因此,本研究旨在开发一种将表观遗传编辑与 STAT6 缺乏相结合的方案,以提高 iTregs 保持稳定抑制功能和功能表型的能力。我们的研究结果表明,STAT6 缺乏(STAT6-/-)与使用 CRISPR-TET1 工具对 TSDR 进行靶向去甲基化相结合,会导致 FOXP3-TSDR 的广泛去甲基化。STAT6-/- iTregs 中的去甲基化与 Foxp3 和 CTLA-4、PD-1、IL-10 和 TGF-β 等抑制性标记物的表达增强有关。此外,与传统的 iTregs 相比,经过编辑的 STAT6-/- iTregs 表现出更强的抑制 CD8+ 和 CD4+ 淋巴细胞的能力,并能更有效地损害 Th1 标志基因的表达。总之,通过 CRISPR-TET1 使 STAT6 失活和 TSDR 靶向去甲基化足以诱导出具有更高的稳定性和更强的抑制能力的 iTregs,为治疗炎症和自身免疫性疾病提供了潜在的应用前景。
{"title":"Targeted Demethylation of FOXP3-TSDR Enhances the Suppressive Capacity of STAT6-deficient Inducible T Regulatory Cells.","authors":"Rubén D Arroyo-Olarte, Juan C Flores-Castelán, Leonel Armas-López, Galileo Escobedo, Luis I Terrazas, Federico Ávila-Moreno, Sonia Leon-Cabrera","doi":"10.1007/s10753-024-02031-4","DOIUrl":"10.1007/s10753-024-02031-4","url":null,"abstract":"<p><p>In vitro induced T regulatory cells (iTregs) are promising for addressing inflammation-driven diseases. However, current protocols for the generation and expansion of iTregs fail to induce extensive demethylation of the Treg-specific demethylated region (TSDR) within the FOXP3 gene, recognized as the master regulator for regulatory T cells (Tregs). This deficiency results in the rapid loss of Foxp3 expression and an unstable regulatory phenotype. Nevertheless, inhibition of STAT6 signaling effectively stabilizes Foxp3 expression in iTregs. Thus, this study aimed to develop a protocol combining epigenetic editing with STAT6 deficiency to improve iTregs' ability to maintain stable suppressive function and a functional phenotype. Our findings demonstrate that the combination of STAT6 deficiency (STAT6-/-) with targeted demethylation of the TSDR using a CRISPR-TET1 tool leads to extensive demethylation of FOXP3-TSDR. Demethylation in STAT6-/- iTregs was associated with enhanced expression of Foxp3 and suppressive markers such as CTLA-4, PD-1, IL-10, and TGF-β. Furthermore, the edited STAT6-/- iTregs exhibited an increased capacity to suppress CD8+ and CD4+ lymphocytes and could more efficiently impair Th1-signature gene expression compared to conventional iTregs. In conclusion, the deactivation of STAT6 and TSDR-targeted demethylation via CRISPR-TET1 is sufficient to induce iTregs with heightened stability and increased suppressive capacity, offering potential applications against inflammatory and autoimmune diseases.</p>","PeriodicalId":13524,"journal":{"name":"Inflammation","volume":" ","pages":"2159-2172"},"PeriodicalIF":4.5,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11606997/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140864247","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Inflammation
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1