首页 > 最新文献

Cellular and Molecular Gastroenterology and Hepatology最新文献

英文 中文
The Glutamine Metabolic Switch Influences the Response of Neonatal Intestinal Macrophages to Breast Milk 谷氨酰胺代谢开关影响新生儿肠巨噬细胞对母乳的反应。
IF 7.1 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2026-01-01 DOI: 10.1016/j.jcmgh.2025.101683
Xu Han , Yutong Hou , Taoying Chen , Zhenyu Jia , Fuqiang Yuan , Mingxin Wang , Yinling Su , Ru Yan , Xiaolei Wang , Weilai Jin , Yahui Zhou , Yun Li , Le Zhang

Background & Aims

Breast milk contains abundant glutamine and glutamate, yet their roles in neonatal gut health remain controversial. We aimed to investigate how these amino acids influence neonatal enteritis and the underlying mechanisms.

Methods

We used a neonatal rat necrotizing enterocolitis (NEC) model to test the effects of glutamine and glutamate given either before disease onset or during NEC progression. Previously published human neonatal NEC single-cell RNA psequencing (scRNA-seq) data were reanalyzed to assess immune cell composition and pathway activity. Bone marrow-derived macrophages (BMDMs) were used to examine inflammatory responses in vitro. Flow cytometry and transcriptomics were applied to analyze metabolic reprogramming of ileal macrophages.

Results

Glutamine and glutamate prevented NEC when administered prophylactically but worsened disease when given during NEC progression. scRNA-seq revealed enrichment of macrophages with activated inflammatory pathways in NEC ileum. In vitro, pretreatment with glutamine or glutamate reduced lipopolysaccharide-induced cytokine expression in BMDMs, whereas administration after stimulation had no benefit. In vivo, glutamine pretreatment decreased CD45+F4/80+CD11b/c+TNFα+ macrophages, whereas treatment during NEC increased this subset. Integrated analyses showed NEC upregulated glutaminase (GLS) and downregulated glutamate dehydrogenase (GLUD1) in ileal macrophages. Pretreatment with glutamine or glutamate restored GLUD1 expression, favoring α-ketoglutarate (α-KG) rather than succinate metabolism. Supplementation with α-KG reversed glutamine-induced macrophage activation during NEC, whereas succinate abolished the protective effect of glutamine pretreatment.

Conclusions

Glutamine and glutamate exert dual, context-dependent effects in neonatal enteritis by modulating macrophage glutamine metabolism. These findings provide mechanistic insight and suggest a basis for personalized glutamine supplementation strategies in neonatal gut disorders.
背景与目的:母乳中含有丰富的谷氨酰胺和谷氨酸,但它们在新生儿肠道健康中的作用仍然存在争议。我们的目的是研究这些氨基酸如何影响新生儿肠炎和潜在的机制。方法:我们使用新生大鼠坏死性小肠结肠炎(NEC)模型来测试谷氨酰胺和谷氨酸在疾病发作前或NEC进展期间给予的影响。重新分析先前发表的人类新生儿NEC scRNA-seq数据,以评估免疫细胞组成和途径活性。骨髓源性巨噬细胞(bmmdms)用于体外检测炎症反应。应用流式细胞术和转录组学分析回肠巨噬细胞的代谢重编程。结果:谷氨酰胺和谷氨酸可预防NEC的发生,但在NEC进展过程中使病情恶化。scRNA-seq显示,NEC回肠中巨噬细胞富集,炎症通路活化。在体外实验中,谷氨酰胺或谷氨酸预处理可降低脂多糖诱导的BMDMs细胞因子表达,而刺激后给药则没有任何益处。在体内,谷氨酰胺预处理降低了CD45+F4/80+CD11b/c+TNFα+巨噬细胞,而NEC期间的治疗增加了这一亚群。综合分析显示,NEC上调了回肠巨噬细胞的谷氨酰胺酶(GLS),下调了谷氨酸脱氢酶(GLUD1)。谷氨酰胺或谷氨酸预处理恢复GLUD1表达,有利于α-酮戊二酸(α-KG)而不是琥珀酸代谢。补充α-KG可逆转NEC期间谷氨酰胺诱导的巨噬细胞活化,而琥珀酸可消除谷氨酰胺预处理的保护作用。结论:谷氨酰胺和谷氨酸通过调节巨噬细胞谷氨酰胺代谢在新生儿肠炎中发挥双重作用。这些发现提供了机制的见解,并为新生儿肠道疾病的个性化谷氨酰胺补充策略提供了基础。
{"title":"The Glutamine Metabolic Switch Influences the Response of Neonatal Intestinal Macrophages to Breast Milk","authors":"Xu Han ,&nbsp;Yutong Hou ,&nbsp;Taoying Chen ,&nbsp;Zhenyu Jia ,&nbsp;Fuqiang Yuan ,&nbsp;Mingxin Wang ,&nbsp;Yinling Su ,&nbsp;Ru Yan ,&nbsp;Xiaolei Wang ,&nbsp;Weilai Jin ,&nbsp;Yahui Zhou ,&nbsp;Yun Li ,&nbsp;Le Zhang","doi":"10.1016/j.jcmgh.2025.101683","DOIUrl":"10.1016/j.jcmgh.2025.101683","url":null,"abstract":"<div><h3>Background &amp; Aims</h3><div>Breast milk contains abundant glutamine and glutamate, yet their roles in neonatal gut health remain controversial. We aimed to investigate how these amino acids influence neonatal enteritis and the underlying mechanisms.</div></div><div><h3>Methods</h3><div>We used a neonatal rat necrotizing enterocolitis (NEC) model to test the effects of glutamine and glutamate given either before disease onset or during NEC progression. Previously published human neonatal NEC single-cell RNA psequencing (scRNA-seq) data were reanalyzed to assess immune cell composition and pathway activity. Bone marrow-derived macrophages (BMDMs) were used to examine inflammatory responses in vitro. Flow cytometry and transcriptomics were applied to analyze metabolic reprogramming of ileal macrophages.</div></div><div><h3>Results</h3><div>Glutamine and glutamate prevented NEC when administered prophylactically but worsened disease when given during NEC progression. scRNA-seq revealed enrichment of macrophages with activated inflammatory pathways in NEC ileum. In vitro, pretreatment with glutamine or glutamate reduced lipopolysaccharide-induced cytokine expression in BMDMs, whereas administration after stimulation had no benefit. In vivo, glutamine pretreatment decreased CD45<sup>+</sup>F4/80<sup>+</sup>CD11b/c<sup>+</sup>TNFα<sup>+</sup> macrophages, whereas treatment during NEC increased this subset. Integrated analyses showed NEC upregulated glutaminase (GLS) and downregulated glutamate dehydrogenase (GLUD1) in ileal macrophages. Pretreatment with glutamine or glutamate restored GLUD1 expression, favoring α-ketoglutarate (α-KG) rather than succinate metabolism. Supplementation with α-KG reversed glutamine-induced macrophage activation during NEC, whereas succinate abolished the protective effect of glutamine pretreatment.</div></div><div><h3>Conclusions</h3><div>Glutamine and glutamate exert dual, context-dependent effects in neonatal enteritis by modulating macrophage glutamine metabolism. These findings provide mechanistic insight and suggest a basis for personalized glutamine supplementation strategies in neonatal gut disorders.</div></div>","PeriodicalId":55974,"journal":{"name":"Cellular and Molecular Gastroenterology and Hepatology","volume":"20 3","pages":"Article 101683"},"PeriodicalIF":7.1,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145650222","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Loss of NKCC1 Activates the NLRP3 Inflammasome in Intestinal Epithelia NKCC1缺失激活肠上皮NLRP3炎性体。
IF 7.1 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2026-01-01 DOI: 10.1016/j.jcmgh.2025.101681
Rainelli B. Koumangoye, Mohammed Z. Ferdaus, Xenia Davis, Julia K. Bohannon, Eric Delpire

Background & Aims

Potassium and chloride efflux have been reported to regulate nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3) inflammasome activation. Aberrant activation of NLRP3 inflammasome causes autoimmune and chronic inflammatory diseases. The Na+-K+-2Cl- cotransporter (NKCC1) maintains the intracellular concentrations of sodium, potassium, and chloride. Here we ask whether NKCC1 modulates NLRP3 inflammasome activation.

Methods

Mice with intestinal epithelial-specific deletion of NKCC1, CRISPR-Cas9 NKCC1-deleted Caco-2 and HT-29 cells, and human fibroblasts expressing mutant NKCC1 were used to evaluate NLRP3 inflammasome activation.

Results

We found that deletion of NKCC1 in established intestinal epithelial cells (IECs) in culture causes increased pyroptosis and interleukin (IL)-1β and IL-18 secretion upon NLRP3 inflammasome activation. Similarly, organoids derived from mice with conditional NKCC1 knockout in IECs also exhibit increased IL-1β secretion when stimulated with adenosine triphosphate (ATP). Moreover, fibroblasts from a patient with NKCC1 mutant also showed increased pyroptosis and IL-1β and IL-18 secretion. Loss of NKCC1 sensitizes IECs to changes in intracellular concentration of K+ and decreases the threshold required for NLRP3 inflammasome activation. Finally, we showed that NKCC1ΔIEC mice have increased infiltration of innate immune cells in the colon mucosa and peritoneal cavity.

Conclusions

NKCC1 functions as a negative regulator of NLRP3 inflammasome activation and this may explain why patients of loss-of-function mutations in NKCC1 are susceptible to inflammatory diseases.
背景:钾和氯的外排已被报道调节NLRP3炎性体的激活。NLRP3炎性小体的异常激活导致自身免疫性和慢性炎症性疾病。Na+- k +- 2cl -共转运体(NKCC1)维持细胞内钠、钾和氯化物的浓度。在这里,我们询问NKCC1是否调节NLRP3炎性体的激活。方法:采用肠上皮特异性缺失NKCC1的小鼠、CRISPR-Cas9缺失NKCC1的caco2和HT-29细胞以及表达突变型NKCC1的人成纤维细胞来评估NLRP3炎性体的激活。结果:我们发现在培养的小肠上皮细胞中缺失NKCC1导致NLRP3炎性体激活后的焦亡、IL-1β和IL-18分泌增加。同样,在肠上皮细胞(IECs)中NKCC1条件敲除小鼠的类器官在ATP刺激下也表现出IL-1β分泌增加。此外,NKCC1突变患者的成纤维细胞也显示出焦亡,IL-1β和IL-18分泌增加。NKCC1的缺失使IECs对细胞内K+浓度的变化敏感,并降低NLRP3炎性体激活所需的阈值。最后,我们发现NKCC1ΔIEC小鼠在结肠黏膜和腹膜腔中增加了先天免疫细胞的浸润。结论:NKCC1作为NLRP3炎性小体激活的负调节因子,这可能解释了为什么NKCC1功能缺失突变的患者易患炎性疾病。
{"title":"Loss of NKCC1 Activates the NLRP3 Inflammasome in Intestinal Epithelia","authors":"Rainelli B. Koumangoye,&nbsp;Mohammed Z. Ferdaus,&nbsp;Xenia Davis,&nbsp;Julia K. Bohannon,&nbsp;Eric Delpire","doi":"10.1016/j.jcmgh.2025.101681","DOIUrl":"10.1016/j.jcmgh.2025.101681","url":null,"abstract":"<div><h3>Background &amp; Aims</h3><div>Potassium and chloride efflux have been reported to regulate nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3) inflammasome activation. Aberrant activation of NLRP3 inflammasome causes autoimmune and chronic inflammatory diseases. The Na<sup>+</sup>-K<sup>+</sup>-2Cl<sup>-</sup> cotransporter (NKCC1) maintains the intracellular concentrations of sodium, potassium, and chloride. Here we ask whether NKCC1 modulates NLRP3 inflammasome activation.</div></div><div><h3>Methods</h3><div>Mice with intestinal epithelial-specific deletion of NKCC1, CRISPR-Cas9 NKCC1-deleted Caco-2 and HT-29 cells, and human fibroblasts expressing mutant NKCC1 were used to evaluate NLRP3 inflammasome activation.</div></div><div><h3>Results</h3><div>We found that deletion of NKCC1 in established intestinal epithelial cells (IECs) in culture causes increased pyroptosis and interleukin (IL)-1β and IL-18 secretion upon NLRP3 inflammasome activation. Similarly, organoids derived from mice with conditional NKCC1 <em>knockout</em> in IECs also exhibit increased IL-1β secretion when stimulated with adenosine triphosphate (ATP). Moreover, fibroblasts from a patient with NKCC1 mutant also showed increased pyroptosis and IL-1β and IL-18 secretion. Loss of NKCC1 sensitizes IECs to changes in intracellular concentration of K<sup>+</sup> and decreases the threshold required for NLRP3 inflammasome activation. Finally, we showed that NKCC1<sup>ΔIEC</sup> mice have increased infiltration of innate immune cells in the colon mucosa and peritoneal cavity.</div></div><div><h3>Conclusions</h3><div>NKCC1 functions as a negative regulator of NLRP3 inflammasome activation and this may explain why patients of loss-of-function mutations in NKCC1 are susceptible to inflammatory diseases.</div></div>","PeriodicalId":55974,"journal":{"name":"Cellular and Molecular Gastroenterology and Hepatology","volume":"20 3","pages":"Article 101681"},"PeriodicalIF":7.1,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145558291","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Inhibition of Adipocyte Lipolysis Reduces Liver Injury in a Mouse Model of Ischemia Reperfusion Injury 抑制脂肪细胞脂解可减轻小鼠缺血再灌注损伤模型中的肝损伤。
IF 7.1 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2026-01-01 DOI: 10.1016/j.jcmgh.2025.101679
Kim H.H. Liss , Samantha Goldman , Mai He , Trevor M. Shew , Daniel Ferguson , Brian N. Finck

Background & Aims

Hepatic ischemia reperfusion injury (IRI) is unavoidable in most liver operations and is associated with poor patient and graft outcomes in the setting of liver transplantation. However, there are no pharmacological interventions available for treatment of IRI. Prior work has demonstrated that liver IRI leads to hepatic lipid accumulation, suggesting that increased adipocyte lipolytic rates may contribute to hepatic steatosis. Inhibition of adipose triglyceride lipase (ATGL), the rate-limiting enzyme involved in triglyceride hydrolysis, may be beneficial in cardiac injury and alcoholic liver disease, but its role in liver IRI has not been investigated. Our objective was to assess the effects of inhibition of adipose tissue lipolysis in the setting of liver IRI.

Methods

Wild-type mice were treated with Atglistatin, a small molecule inhibitor of ATGL, prior to IRI. Mice with hepatocyte- or adipocyte-specific deletion of Pnpla2, the gene encoding ATGL, were generated and subjected to a mouse model of IRI. Mouse hepatocytes were cultured with fatty acids in an in vitro model of IRI.

Results

We demonstrated that experimental IRI was associated with increased adipocyte lipolysis. Pharmacological and genetic inhibition of adipocyte lipolysis reduced plasma and hepatic free fatty acids and decreased circulating transaminases and liver inflammation following hepatic IRI. Furthermore, exogenous fatty acids were sufficient to increase cell death and the expression of inflammatory cytokines in in vitro IRI.

Conclusions

These data suggest that targeting adipocyte lipolysis may represent a novel therapeutic approach in the prevention of hepatic IRI, which could improve patient and graft outcomes following liver transplantation.
背景和目的:肝脏缺血再灌注损伤(IRI)在大多数肝脏手术中是不可避免的,并且与肝移植患者和移植物预后差有关。然而,目前还没有治疗IRI的药物干预措施。先前的研究表明,肝脏IRI导致肝脏脂质积累,这表明脂肪细胞脂质分解率的增加可能导致肝脏脂肪变性。脂肪甘油三酯脂肪酶(ATGL)是参与甘油三酯水解的限速酶,抑制ATGL可能对心脏损伤和酒精性肝病有益,但其在肝脏IRI中的作用尚未被研究。我们的目的是评估在肝脏IRI的情况下抑制脂肪组织脂解的效果。方法:野生型小鼠在IRI前用Atglistatin(一种ATGL小分子抑制剂)治疗。产生肝细胞或脂肪细胞特异性缺失Pnpla2(编码ATGL的基因)的小鼠,并进行小鼠IRI模型。用脂肪酸培养小鼠肝细胞,建立体外IRI模型。结果:我们证明了实验性IRI与脂肪细胞脂解增加有关。脂肪细胞脂解的药理学和遗传学抑制降低了血浆和肝脏游离脂肪酸,减少了肝脏IRI后的循环转氨酶和肝脏炎症。此外,外源性脂肪酸足以增加体外IRI中细胞死亡和炎症细胞因子的表达。结论:这些数据表明,靶向脂肪细胞脂解可能是预防肝脏IRI的一种新的治疗方法,可以改善肝移植后患者和移植物的预后。
{"title":"Inhibition of Adipocyte Lipolysis Reduces Liver Injury in a Mouse Model of Ischemia Reperfusion Injury","authors":"Kim H.H. Liss ,&nbsp;Samantha Goldman ,&nbsp;Mai He ,&nbsp;Trevor M. Shew ,&nbsp;Daniel Ferguson ,&nbsp;Brian N. Finck","doi":"10.1016/j.jcmgh.2025.101679","DOIUrl":"10.1016/j.jcmgh.2025.101679","url":null,"abstract":"<div><h3>Background &amp; Aims</h3><div>Hepatic ischemia reperfusion injury (IRI) is unavoidable in most liver operations and is associated with poor patient and graft outcomes in the setting of liver transplantation. However, there are no pharmacological interventions available for treatment of IRI. Prior work has demonstrated that liver IRI leads to hepatic lipid accumulation, suggesting that increased adipocyte lipolytic rates may contribute to hepatic steatosis. Inhibition of adipose triglyceride lipase (ATGL), the rate-limiting enzyme involved in triglyceride hydrolysis, may be beneficial in cardiac injury and alcoholic liver disease, but its role in liver IRI has not been investigated. Our objective was to assess the effects of inhibition of adipose tissue lipolysis in the setting of liver IRI.</div></div><div><h3>Methods</h3><div>Wild-type mice were treated with Atglistatin, a small molecule inhibitor of ATGL, prior to IRI. Mice with hepatocyte- or adipocyte-specific deletion of <em>Pnpla2</em>, the gene encoding ATGL, were generated and subjected to a mouse model of IRI. Mouse hepatocytes were cultured with fatty acids in an in vitro model of IRI.</div></div><div><h3>Results</h3><div>We demonstrated that experimental IRI was associated with increased adipocyte lipolysis. Pharmacological and genetic inhibition of adipocyte lipolysis reduced plasma and hepatic free fatty acids and decreased circulating transaminases and liver inflammation following hepatic IRI. Furthermore, exogenous fatty acids were sufficient to increase cell death and the expression of inflammatory cytokines in in vitro IRI.</div></div><div><h3>Conclusions</h3><div>These data suggest that targeting adipocyte lipolysis may represent a novel therapeutic approach in the prevention of hepatic IRI, which could improve patient and graft outcomes following liver transplantation.</div></div>","PeriodicalId":55974,"journal":{"name":"Cellular and Molecular Gastroenterology and Hepatology","volume":"20 3","pages":"Article 101679"},"PeriodicalIF":7.1,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145566188","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
The P2X7 receptor promotes intestinal fibrosis by modulating the gut microbiota and the inflammasome. P2X7受体通过调节肠道微生物群和炎性体促进肠道纤维化。
IF 7.1 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-30 DOI: 10.1016/j.jcmgh.2025.101718
Beatriz Elias Ribeiro, Isadora Schmukler de Lima, Karen Cristina da Silva E Souza, Siane Lopes Bittencourt Rosas, Patrícia Teixeira Santana, Gilda Amaral, João Carlos Machado, Rodrigo Pereira de Oliveira, Camille Leal, Cristiane Thompson, Fabiano Thompson, Morgana Teixeira Lima Castelo-Branco, Robson Coutinho-Silva, Heitor Siffert Pereira de Souza

Background and aims: Considering the role of the P2X7 receptor in intestinal inflammation, we examined its potential involvement in fibrosis development.

Methods: Colonic biopsies from patients with inflammatory bowel disease (IBD) were analyzed via double immunofluorescence under confocal microscopy. Colon fibroblasts were used to analyze P2X7 receptor modulation and chemotaxis. Experimental chronic colitis was induced with three cycles of oral dextran sodium sulfate (DSS) treatment in P2X7+/+ and P2X7-/- mice. The mice were evaluated via follow-up video endoscopy with an endoluminal ultrasound biomicroscopic (eUBM) system, histological scoring, immunohistochemistry, cytokine measurement in colon explants, gene expression analysis of P2X7 signaling targets via qRT‒PCR, and microbiome composition analysis.

Results: Colocalization studies revealed a greater density of P2X7+/α-SMA+ cells in colon sections from patients than in those from controls, especially in patients with Crohn's disease (p<0.05). Activation of the ATP-P2X7 pathway in human fibroblasts increased cell migration, calcium influx, and collagen production. Video colonoscopy with the eUBM system revealed significantly more inflammation, with greater wall thickness and stiffness, in P2X7+/+- mice than in P2X7-/- and P2X7+/+ mice treated with A740003 (a P2X7-selective inhibitor). P2X7+/+ mice exhibited increased caspase-1 and NLRP3 expression, as well as NF-κB and ERK activation, accompanied by decreased PPARγ expression. In the supernatants of colon explants, TNF-α, IL-1β, IFN-γ, TGF-β, IL-10, and collagen production were increased in P2X7+/+ mice. Various microbial changes were observed in P2X7-/- and P2X7+/+ mice.

Conclusion: Regulatory mechanisms downstream of P2X7, combined with signals from a dysbiotic microbiota, activate intracellular signaling pathways and the inflammasome, leading to intestinal inflammation and promoting fibrogenesis.

Clinical trial registration:

背景和目的:考虑到P2X7受体在肠道炎症中的作用,我们研究了其在纤维化发展中的潜在参与。方法:采用双免疫荧光共聚焦显微镜对炎症性肠病(IBD)患者结肠活检进行分析。用结肠成纤维细胞分析P2X7受体的调节和趋化性。采用三周期口服葡聚糖硫酸钠(DSS)治疗P2X7+/+和P2X7-/-小鼠,诱导实验性慢性结肠炎。通过腔内超声生物显微镜(eUBM)系统的随访视频内窥镜、组织学评分、免疫组织化学、结肠外植体细胞因子测量、qRT-PCR分析P2X7信号靶点的基因表达以及微生物组组成分析对小鼠进行评估。结果:共定位研究显示,患者结肠切片中的P2X7+/α-SMA+细胞密度高于对照组,尤其是克罗恩病患者(p+/+-小鼠比使用A740003(一种P2X7选择性抑制剂)治疗的P2X7-/-和P2X7+/+小鼠密度更高)。P2X7+/+小鼠caspase-1和NLRP3表达增加,NF-κB和ERK活化,PPARγ表达降低。在结肠外植体上清液中,P2X7+/+小鼠的TNF-α、IL-1β、IFN-γ、TGF-β、IL-10和胶原生成均增加。在P2X7-/-和P2X7+/+小鼠中观察到各种微生物变化。结论:P2X7下游的调控机制,结合来自益生菌群的信号,激活细胞内信号通路和炎性小体,导致肠道炎症,促进纤维生成。临床试验注册:
{"title":"The P2X7 receptor promotes intestinal fibrosis by modulating the gut microbiota and the inflammasome.","authors":"Beatriz Elias Ribeiro, Isadora Schmukler de Lima, Karen Cristina da Silva E Souza, Siane Lopes Bittencourt Rosas, Patrícia Teixeira Santana, Gilda Amaral, João Carlos Machado, Rodrigo Pereira de Oliveira, Camille Leal, Cristiane Thompson, Fabiano Thompson, Morgana Teixeira Lima Castelo-Branco, Robson Coutinho-Silva, Heitor Siffert Pereira de Souza","doi":"10.1016/j.jcmgh.2025.101718","DOIUrl":"https://doi.org/10.1016/j.jcmgh.2025.101718","url":null,"abstract":"<p><strong>Background and aims: </strong>Considering the role of the P2X7 receptor in intestinal inflammation, we examined its potential involvement in fibrosis development.</p><p><strong>Methods: </strong>Colonic biopsies from patients with inflammatory bowel disease (IBD) were analyzed via double immunofluorescence under confocal microscopy. Colon fibroblasts were used to analyze P2X7 receptor modulation and chemotaxis. Experimental chronic colitis was induced with three cycles of oral dextran sodium sulfate (DSS) treatment in P2X7<sup>+/+</sup> and P2X7<sup>-/-</sup> mice. The mice were evaluated via follow-up video endoscopy with an endoluminal ultrasound biomicroscopic (eUBM) system, histological scoring, immunohistochemistry, cytokine measurement in colon explants, gene expression analysis of P2X7 signaling targets via qRT‒PCR, and microbiome composition analysis.</p><p><strong>Results: </strong>Colocalization studies revealed a greater density of P2X7<sup>+</sup>/α-SMA<sup>+</sup> cells in colon sections from patients than in those from controls, especially in patients with Crohn's disease (p<0.05). Activation of the ATP-P2X7 pathway in human fibroblasts increased cell migration, calcium influx, and collagen production. Video colonoscopy with the eUBM system revealed significantly more inflammation, with greater wall thickness and stiffness, in P2X7<sup>+/+</sup>- mice than in P2X7<sup>-/-</sup> and P2X7<sup>+/+</sup> mice treated with A740003 (a P2X7-selective inhibitor). P2X7<sup>+/+</sup> mice exhibited increased caspase-1 and NLRP3 expression, as well as NF-κB and ERK activation, accompanied by decreased PPARγ expression. In the supernatants of colon explants, TNF-α, IL-1β, IFN-γ, TGF-β, IL-10, and collagen production were increased in P2X7<sup>+/+</sup> mice. Various microbial changes were observed in P2X7<sup>-/-</sup> and P2X7<sup>+/+</sup> mice.</p><p><strong>Conclusion: </strong>Regulatory mechanisms downstream of P2X7, combined with signals from a dysbiotic microbiota, activate intracellular signaling pathways and the inflammasome, leading to intestinal inflammation and promoting fibrogenesis.</p><p><strong>Clinical trial registration: </strong></p>","PeriodicalId":55974,"journal":{"name":"Cellular and Molecular Gastroenterology and Hepatology","volume":" ","pages":"101718"},"PeriodicalIF":7.1,"publicationDate":"2025-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145890609","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
ΔNP63 defines an exocrine-committed subset of murine pancreatic progenitor cells. ΔNP63定义了小鼠胰腺祖细胞的外分泌亚群。
IF 7.1 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-24 DOI: 10.1016/j.jcmgh.2025.101715
Katarina Coolens, Melissa Van der Vliet, Jente Van Campenhout, Natalia Del Pozo, Alejo Torres-Cano, Catharina Olson, Jianming Xu, Heiko Lickert, Meritxell Rovira, Isabelle Houbracken, Jonathan Baldan, Francisco X Real, Francesca M Spagnoli, Ilse Rooman
{"title":"ΔNP63 defines an exocrine-committed subset of murine pancreatic progenitor cells.","authors":"Katarina Coolens, Melissa Van der Vliet, Jente Van Campenhout, Natalia Del Pozo, Alejo Torres-Cano, Catharina Olson, Jianming Xu, Heiko Lickert, Meritxell Rovira, Isabelle Houbracken, Jonathan Baldan, Francisco X Real, Francesca M Spagnoli, Ilse Rooman","doi":"10.1016/j.jcmgh.2025.101715","DOIUrl":"https://doi.org/10.1016/j.jcmgh.2025.101715","url":null,"abstract":"","PeriodicalId":55974,"journal":{"name":"Cellular and Molecular Gastroenterology and Hepatology","volume":" ","pages":"101715"},"PeriodicalIF":7.1,"publicationDate":"2025-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145844307","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Genetically induced mouse model for colon-specific epithelial cell tumorigenesis driven by loss of K8 and Apc. 由K8和Apc缺失驱动的结肠特异性上皮细胞肿瘤的基因诱导小鼠模型。
IF 7.1 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-24 DOI: 10.1016/j.jcmgh.2025.101716
Mina Tayyab, Mira M E Minkkinen, Carl-Gustaf A Stenvall, Lauri Polari, Victor Nielsen, Yatrik M Shah, Diana M Toivola

Background and aims: Loss of keratin 8 (K8) has been shown to increase susceptibility towards colonocyte hyperproliferation and tumorigenesis. However, most colorectal cancer (CRC) mouse models require carcinogen, develop small intestinal tumors or have a long latency period. The aim was to establish a genetic, colon-specific and more human like CRC model driven by loss of K8 and Apc.

Methods: Colon-specific targeting using CDX2P-CreERT2 mice was used to generate K8flox/flox; CDX2P-CreERT2 and K8flox/flox; CDX2P-CreERT2; Apcflox/+ mice. Disease activity was monitored, and colon was analyzed for tumor burden and histopathology over time. Keratin expression, inflammation, proliferation, cell polarity, colonocyte populations and cell division symmetry were assessed using immunoblotting and immunofluorescence analysis. This data was compared to K8 expression analysis in CRC patients and in UALCAN database.

Results: K8flox/flox; CDX2P-CreERT2 mice develop mild diarrhea and express reduced K8 and partner keratins in a mosaic pattern in the colonic epithelium. K8-negative colon areas display increased crypt loss and more inflammation predominantly in the proximal colon. Increased colonocyte proliferation is observed throughout the colon. Impaired cell polarity and higher number of stem and progenitor cells with a shift towards asymmetric cell division in K8-negative areas of the distal colon highlight a pro-tumorigenic environment. Mice with additional monoallelic Apc inactivation show colon tumorigenesis and epithelial to mesenchymal transition distally. In CRC patients, tumor K8 expression is decreased independent of disease type and stage, age or gender.

Conclusion: Genetic colon-specific mouse model with loss of K8 and Apc adequately resembles human CRC. This study identifies anti-tumorigenic protective roles of colonocyte K8 in the colon.

背景和目的:角蛋白8 (K8)的缺失已被证明会增加对结肠细胞过度增殖和肿瘤发生的易感性。然而,大多数结直肠癌(CRC)小鼠模型需要致癌物,发展为小肠肿瘤或潜伏期长。目的是建立一个由K8和Apc缺失驱动的遗传的、结肠特异性的、更像人类的CRC模型。方法:利用CDX2P-CreERT2小鼠结肠特异性靶向产生K8flox/flox;CDX2P-CreERT2和K8flox/flox;CDX2P-CreERT2;Apcflox / +老鼠。监测疾病活动性,并随时间分析结肠的肿瘤负荷和组织病理学。使用免疫印迹和免疫荧光分析评估角蛋白表达、炎症、增殖、细胞极性、结肠菌群和细胞分裂对称性。将该数据与结直肠癌患者和UALCAN数据库中的K8表达分析进行比较。结果:K8flox /液氧;CDX2P-CreERT2小鼠出现轻度腹泻,并在结肠上皮中以马赛克模式表达减少的K8和伴侣角蛋白。k8阴性结肠区域显示隐窝丢失增加,炎症加重,主要发生在近端结肠。整个结肠可见结肠细胞增殖增加。在远端结肠k8阴性区域,细胞极性受损,干细胞和祖细胞数量增加,细胞分裂向不对称方向转变,突出了促肿瘤发生的环境。单等位基因Apc失活小鼠显示结肠肿瘤发生和上皮向远端间质转化。在结直肠癌患者中,肿瘤K8表达的降低与疾病类型、分期、年龄或性别无关。结论:K8和Apc缺失的基因结肠特异性小鼠模型与人类结直肠癌完全相似。本研究确定了结肠细胞K8在结肠中的抗肿瘤保护作用。
{"title":"Genetically induced mouse model for colon-specific epithelial cell tumorigenesis driven by loss of K8 and Apc.","authors":"Mina Tayyab, Mira M E Minkkinen, Carl-Gustaf A Stenvall, Lauri Polari, Victor Nielsen, Yatrik M Shah, Diana M Toivola","doi":"10.1016/j.jcmgh.2025.101716","DOIUrl":"https://doi.org/10.1016/j.jcmgh.2025.101716","url":null,"abstract":"<p><strong>Background and aims: </strong>Loss of keratin 8 (K8) has been shown to increase susceptibility towards colonocyte hyperproliferation and tumorigenesis. However, most colorectal cancer (CRC) mouse models require carcinogen, develop small intestinal tumors or have a long latency period. The aim was to establish a genetic, colon-specific and more human like CRC model driven by loss of K8 and Apc.</p><p><strong>Methods: </strong>Colon-specific targeting using CDX2P-CreER<sup>T2</sup> mice was used to generate K8<sup>flox/flox</sup>; CDX2P-CreER<sup>T2</sup> and K8<sup>flox/flox</sup>; CDX2P-CreER<sup>T2</sup>; Apc<sup>flox/+</sup> mice. Disease activity was monitored, and colon was analyzed for tumor burden and histopathology over time. Keratin expression, inflammation, proliferation, cell polarity, colonocyte populations and cell division symmetry were assessed using immunoblotting and immunofluorescence analysis. This data was compared to K8 expression analysis in CRC patients and in UALCAN database.</p><p><strong>Results: </strong>K8<sup>flox/flox</sup>; CDX2P-CreER<sup>T2</sup> mice develop mild diarrhea and express reduced K8 and partner keratins in a mosaic pattern in the colonic epithelium. K8-negative colon areas display increased crypt loss and more inflammation predominantly in the proximal colon. Increased colonocyte proliferation is observed throughout the colon. Impaired cell polarity and higher number of stem and progenitor cells with a shift towards asymmetric cell division in K8-negative areas of the distal colon highlight a pro-tumorigenic environment. Mice with additional monoallelic Apc inactivation show colon tumorigenesis and epithelial to mesenchymal transition distally. In CRC patients, tumor K8 expression is decreased independent of disease type and stage, age or gender.</p><p><strong>Conclusion: </strong>Genetic colon-specific mouse model with loss of K8 and Apc adequately resembles human CRC. This study identifies anti-tumorigenic protective roles of colonocyte K8 in the colon.</p>","PeriodicalId":55974,"journal":{"name":"Cellular and Molecular Gastroenterology and Hepatology","volume":" ","pages":"101716"},"PeriodicalIF":7.1,"publicationDate":"2025-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145844547","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Repeated DSS Exposure Elicits Distinct Immune Responses Reflecting Human Ulcerative Colitis. 重复的DSS暴露引起不同的免疫反应,反映人类溃疡性结肠炎。
IF 7.1 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-24 DOI: 10.1016/j.jcmgh.2025.101714
Laura D Manzanares, Zeinab Dehghani, Caroline J Herrnreiter, Xingsheng Ren, Maya E Sumagin, Anastasiia Serdiukova, Ankur Naqib, Stefan Green, Joshua B Wechsler, Ronen Sumagin

Background and aims: Dextran sulfate sodium (DSS)-induced colitis is a widely used model to study ulcerative colitis (UC). However, the extent to which acute versus repeated DSS exposure mimics human disease remains unclear.

Methods: Using histopathology, flow cytometry, single-cell RNA sequencing (scRNA-seq) and 16S rRNA profiling we compared disease outcomes, immune infiltrate, transcriptional programs and changes in the microbiome in mice subjected to a single (acute) versus two (repeated) DSS cycles. We further evaluated which experimental condition better represents key immune cell subtype states in human disease.

Results: While disease activity indices were similar between groups, repeated DSS exposure resulted in greater colon shortening, mucosal remodeling and elevated immune infiltration, particularly by neutrophils (PMNs) in the distal colon. scRNA-seq revealed that PMNs and T cells acquired distinct transcriptional programs in repeated versus acute colitis. Cycle 1 PMNs showed inflammatory and cytotoxic signatures, whereas cycle 2 PMNs were enriched in tissue remodeling and survival pathways. CD4 and CD8 T cells in repeated colitis exhibited migratory, and pathogen-responsive phenotypes, with expanded Treg and exhausted CD8 subsets. In contrast, macrophage numbers decreased with repeated DSS, though remaining cells exhibited pro-resolution gene expression profiles. Microbiome analysis revealed normalization trends with repeated DSS exposure, including reduced pro-inflammatory and increased beneficial genera. Cross-species transcriptomic comparisons indicated cycle-specific overlap with human UC, where cycle 2 activated PMNs and alternatively activated macrophages, closer aligned with active human UC.

Conclusion: Collectively, our data indicate that repeated DSS cycles provide a better experimental colitis model for studying immune cells in UC and identifying distinct immune subtypes relevant to UC therapeutics.

背景与目的:葡聚糖硫酸钠(DSS)诱导结肠炎是研究溃疡性结肠炎(UC)的一种广泛使用的模型。然而,急性与反复接触DSS在多大程度上模拟人类疾病仍不清楚。方法:利用组织病理学、流式细胞术、单细胞RNA测序(scRNA-seq)和16S rRNA分析,我们比较了单次(急性)和两次(重复)DSS周期小鼠的疾病结局、免疫浸润、转录程序和微生物组的变化。我们进一步评估了哪种实验条件更能代表人类疾病中的关键免疫细胞亚型状态。结果:虽然两组之间的疾病活动指数相似,但反复暴露于DSS导致结肠缩短,粘膜重塑和免疫浸润升高,特别是结肠远端中性粒细胞(pmn)。scRNA-seq显示重复性结肠炎和急性结肠炎中pmn和T细胞获得不同的转录程序。周期1 PMNs表现出炎症和细胞毒性特征,而周期2 PMNs在组织重塑和生存途径中富集。重复性结肠炎中的CD4和CD8 T细胞表现出迁移性和病原体应答性表型,Treg扩增和CD8亚群耗尽。相比之下,巨噬细胞数量随着DSS的重复而减少,尽管剩余的细胞表现出支持分辨率的基因表达谱。微生物组分析揭示了重复暴露于DSS后的正常化趋势,包括促炎菌减少和有益菌增多。跨物种转录组学比较表明,周期2激活pmn和选择性激活巨噬细胞,与人类UC活性更接近。结论:总的来说,我们的数据表明,重复的DSS周期为研究UC中的免疫细胞和识别与UC治疗相关的不同免疫亚型提供了更好的实验性结肠炎模型。
{"title":"Repeated DSS Exposure Elicits Distinct Immune Responses Reflecting Human Ulcerative Colitis.","authors":"Laura D Manzanares, Zeinab Dehghani, Caroline J Herrnreiter, Xingsheng Ren, Maya E Sumagin, Anastasiia Serdiukova, Ankur Naqib, Stefan Green, Joshua B Wechsler, Ronen Sumagin","doi":"10.1016/j.jcmgh.2025.101714","DOIUrl":"https://doi.org/10.1016/j.jcmgh.2025.101714","url":null,"abstract":"<p><strong>Background and aims: </strong>Dextran sulfate sodium (DSS)-induced colitis is a widely used model to study ulcerative colitis (UC). However, the extent to which acute versus repeated DSS exposure mimics human disease remains unclear.</p><p><strong>Methods: </strong>Using histopathology, flow cytometry, single-cell RNA sequencing (scRNA-seq) and 16S rRNA profiling we compared disease outcomes, immune infiltrate, transcriptional programs and changes in the microbiome in mice subjected to a single (acute) versus two (repeated) DSS cycles. We further evaluated which experimental condition better represents key immune cell subtype states in human disease.</p><p><strong>Results: </strong>While disease activity indices were similar between groups, repeated DSS exposure resulted in greater colon shortening, mucosal remodeling and elevated immune infiltration, particularly by neutrophils (PMNs) in the distal colon. scRNA-seq revealed that PMNs and T cells acquired distinct transcriptional programs in repeated versus acute colitis. Cycle 1 PMNs showed inflammatory and cytotoxic signatures, whereas cycle 2 PMNs were enriched in tissue remodeling and survival pathways. CD4 and CD8 T cells in repeated colitis exhibited migratory, and pathogen-responsive phenotypes, with expanded Treg and exhausted CD8 subsets. In contrast, macrophage numbers decreased with repeated DSS, though remaining cells exhibited pro-resolution gene expression profiles. Microbiome analysis revealed normalization trends with repeated DSS exposure, including reduced pro-inflammatory and increased beneficial genera. Cross-species transcriptomic comparisons indicated cycle-specific overlap with human UC, where cycle 2 activated PMNs and alternatively activated macrophages, closer aligned with active human UC.</p><p><strong>Conclusion: </strong>Collectively, our data indicate that repeated DSS cycles provide a better experimental colitis model for studying immune cells in UC and identifying distinct immune subtypes relevant to UC therapeutics.</p>","PeriodicalId":55974,"journal":{"name":"Cellular and Molecular Gastroenterology and Hepatology","volume":" ","pages":"101714"},"PeriodicalIF":7.1,"publicationDate":"2025-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145844604","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Endogenous glucocorticoids moderate the gastric inflammatory response to Helicobacter infection and protect from autoimmunity. 内源性糖皮质激素调节胃对幽门螺杆菌感染的炎症反应并保护自身免疫。
IF 7.1 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-24 DOI: 10.1016/j.jcmgh.2025.101699
Sara R Druffner, Benjamin C Duncan, Maeve T Morris, Jordan L Pascoe, Tyler M Abner, Salik Hussain, M Blanca Piazuelo, Richard M Peek, Melody Zhang, Richard J DiPaolo, Jonathan T Busada

Background and aims: Immune responses to infection must balance pathogen clearance with minimizing tissue damage and autoimmunity. Chronic gastric inflammation caused by H. pylori damages the gastric mucosa and promotes carcinogenesis. Glucocorticoids are immunoregulatory hormones that limit immune activation in the stomach. This study aimed to determine how endogenous glucocorticoids regulate the gastric immune response to Helicobacter infection and their impact on preneoplastic lesion development.

Methods: We examined the role of endogenous glucocorticoids in shaping the gastric immune response to Helicobacter felis colonization. Gastric immune cell infiltration, atrophy, metaplasia, and preneoplastic lesion development were evaluated in adrenal-intact and adrenalectomized (ADX) mice. Auto-reactive IgG antibodies were assessed using a mouse self-antigen array and by measuring their binding to healthy gastric tissue.

Results: Loss of endogenous glucocorticoids led to significantly increased H. felis-induced gastric T cell infiltration and proinflammatory cytokine expression compared to intact-infected controls. While all intact mice maintained chronic infection for up to 12 months post-colonization, nearly all ADX mice eradicated H. felis within 2-3 weeks. Despite bacterial clearance, ADX mice continued to exhibit chronic gastric inflammation and developed dysplasia. Autoantibody profiling showed that both intact and ADX groups generated self-reactive IgG during active infection. However, only ADX mice sustained autoantibody production following bacterial eradication.

Conclusions: Endogenous glucocorticoids attenuate gastric inflammation during Helicobacter infection, supporting bacterial persistence while maintaining immune tolerance. These findings suggest that heightened immune responses to H. pylori may trigger autoimmune gastritis (AIG) development, which can persist after H. pylori clearance and continue to drive gastric cancer risk.

背景和目的:对感染的免疫反应必须平衡病原体清除与最小化组织损伤和自身免疫。幽门螺旋杆菌引起的慢性胃炎损害胃黏膜,促进癌变。糖皮质激素是限制胃免疫激活的免疫调节激素。本研究旨在确定内源性糖皮质激素如何调节胃对幽门螺杆菌感染的免疫反应及其对肿瘤前病变发展的影响。方法:我们研究了内源性糖皮质激素在塑造胃对幽门螺杆菌定植的免疫反应中的作用。在肾上腺完整和肾上腺切除(ADX)小鼠中评估胃免疫细胞浸润、萎缩、化生和瘤前病变的发展。使用小鼠自身抗原阵列和通过测量其与健康胃组织的结合来评估自身反应性IgG抗体。结果:与未受感染的对照组相比,内源性糖皮质激素的缺失导致H. felis诱导的胃T细胞浸润和促炎细胞因子表达显著增加。虽然所有完整的小鼠在定植后保持慢性感染长达12个月,但几乎所有ADX小鼠在2-3周内根除了狐猴。尽管细菌清除,ADX小鼠继续表现出慢性胃炎症并发展为发育不良。自身抗体分析显示,在活动性感染期间,完整组和ADX组都产生了自身反应性IgG。然而,只有ADX小鼠在细菌根除后仍能产生自身抗体。结论:内源性糖皮质激素可减轻幽门螺杆菌感染时的胃炎症,在维持免疫耐受的同时支持细菌的持续存在。这些发现表明,对幽门螺杆菌的免疫反应增强可能引发自身免疫性胃炎(AIG)的发展,这种发展可能在幽门螺杆菌清除后持续存在,并继续推动胃癌的风险。
{"title":"Endogenous glucocorticoids moderate the gastric inflammatory response to Helicobacter infection and protect from autoimmunity.","authors":"Sara R Druffner, Benjamin C Duncan, Maeve T Morris, Jordan L Pascoe, Tyler M Abner, Salik Hussain, M Blanca Piazuelo, Richard M Peek, Melody Zhang, Richard J DiPaolo, Jonathan T Busada","doi":"10.1016/j.jcmgh.2025.101699","DOIUrl":"10.1016/j.jcmgh.2025.101699","url":null,"abstract":"<p><strong>Background and aims: </strong>Immune responses to infection must balance pathogen clearance with minimizing tissue damage and autoimmunity. Chronic gastric inflammation caused by H. pylori damages the gastric mucosa and promotes carcinogenesis. Glucocorticoids are immunoregulatory hormones that limit immune activation in the stomach. This study aimed to determine how endogenous glucocorticoids regulate the gastric immune response to Helicobacter infection and their impact on preneoplastic lesion development.</p><p><strong>Methods: </strong>We examined the role of endogenous glucocorticoids in shaping the gastric immune response to Helicobacter felis colonization. Gastric immune cell infiltration, atrophy, metaplasia, and preneoplastic lesion development were evaluated in adrenal-intact and adrenalectomized (ADX) mice. Auto-reactive IgG antibodies were assessed using a mouse self-antigen array and by measuring their binding to healthy gastric tissue.</p><p><strong>Results: </strong>Loss of endogenous glucocorticoids led to significantly increased H. felis-induced gastric T cell infiltration and proinflammatory cytokine expression compared to intact-infected controls. While all intact mice maintained chronic infection for up to 12 months post-colonization, nearly all ADX mice eradicated H. felis within 2-3 weeks. Despite bacterial clearance, ADX mice continued to exhibit chronic gastric inflammation and developed dysplasia. Autoantibody profiling showed that both intact and ADX groups generated self-reactive IgG during active infection. However, only ADX mice sustained autoantibody production following bacterial eradication.</p><p><strong>Conclusions: </strong>Endogenous glucocorticoids attenuate gastric inflammation during Helicobacter infection, supporting bacterial persistence while maintaining immune tolerance. These findings suggest that heightened immune responses to H. pylori may trigger autoimmune gastritis (AIG) development, which can persist after H. pylori clearance and continue to drive gastric cancer risk.</p>","PeriodicalId":55974,"journal":{"name":"Cellular and Molecular Gastroenterology and Hepatology","volume":" ","pages":"101699"},"PeriodicalIF":7.1,"publicationDate":"2025-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145844563","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Novel Acinar Metaplastic States Uncovered in Exocrine Pancreas Disease. 外分泌胰腺疾病新发现的腺泡化生状态。
IF 7.1 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-24 DOI: 10.1016/j.jcmgh.2025.101717
Katherine J Aney, Woo-Jeong Jeong, Pal Koak, Anders W Ohman, Canh Hiep Nguyen, Brian M Wolpin, Jonathan A Nowak, Sahar Nissim

Background & aims: In response to injury, pancreatic acinar cells undergo acinar-to-ductal metaplasia (ADM), marked by loss of acinar identity and acquisition of ductal features. While ADM can resolve to support tissue repair, it may also persist and serve as a precursor to pancreatic cancer. Whether diverse pancreatic stressors drive a shared or context-specific ADM program remains unclear. We sought to comprehensively define metaplastic responses to clinically relevant exocrine pancreas diseases known to increase cancer risk.

Methods: We profiled ADM and the surrounding microenvironment across mouse models of exocrine disease-including acute, recurrent, and chronic pancreatitis, as well as in the setting of oncogenic Kras-capturing over 300,000 single cells. To enable high-quality transcriptomic profiling in enzyme-rich tissue, we leveraged FixNCut, a method that preserves RNA integrity in the exocrine pancreas. Findings were validated in human pancreas tissue using CosMx spatial transcriptomics.

Results: We identify a conserved acinar response across disease contexts that gives rise to previously unrecognized distinct metaplastic states, including a "gateway" ADM population that precedes more advanced metaplastic states marked by complete loss of acinar identity. In pancreatic intraepithelial neoplasia (PanIN) precancerous lesions, we detect classical-like and basal-like states, suggesting that pancreatic cancer subtypes are specified much earlier than previously appreciated. In Kras-mutant tissue, we identify a second wave of inflammation and the emergence of an immunosuppressive niche, coinciding with PanIN formation CONCLUSIONS: Our findings define a conserved program of acinar plasticity across exocrine pancreas diseases. We further link unresolved ADM to immune remodeling during precursor lesion formation and observe the emergence of pancreatic cancer subtypes in early PanIN lesions.

背景与目的:作为对损伤的反应,胰腺腺泡细胞发生腺泡到导管的化生(ADM),其特征是腺泡特征的丧失和导管特征的获得。虽然ADM可以支持组织修复,但它也可能持续存在并作为胰腺癌的前兆。不同的胰腺应激源是否驱动共享的或特定环境的ADM程序尚不清楚。我们试图全面定义已知增加癌症风险的临床相关外分泌胰腺疾病的化生反应。方法:我们在外分泌疾病(包括急性、复发性和慢性胰腺炎)的小鼠模型中,以及在捕获超过30万个单细胞的致癌kras环境中,分析了ADM和周围微环境。为了在富含酶的组织中实现高质量的转录组分析,我们利用了FixNCut,一种在外分泌胰腺中保持RNA完整性的方法。使用CosMx空间转录组学在人类胰腺组织中验证了研究结果。结果:我们确定了一种保守的腺泡反应,在疾病背景下引起以前未被识别的不同的化生状态,包括“入口”ADM人群,其在更高级的化生状态之前,标志着腺泡身份的完全丧失。在胰腺上皮内瘤变(PanIN)癌前病变中,我们检测到经典样和基底样状态,这表明胰腺癌亚型的指定比以前认识的要早得多。在kras突变组织中,我们发现了第二波炎症和免疫抑制生态位的出现,与PanIN的形成相一致。结论:我们的研究结果定义了外分泌胰腺疾病中腺泡可塑性的保守程序。我们进一步将未解决的ADM与前体病变形成过程中的免疫重塑联系起来,并观察了早期PanIN病变中胰腺癌亚型的出现。
{"title":"Novel Acinar Metaplastic States Uncovered in Exocrine Pancreas Disease.","authors":"Katherine J Aney, Woo-Jeong Jeong, Pal Koak, Anders W Ohman, Canh Hiep Nguyen, Brian M Wolpin, Jonathan A Nowak, Sahar Nissim","doi":"10.1016/j.jcmgh.2025.101717","DOIUrl":"https://doi.org/10.1016/j.jcmgh.2025.101717","url":null,"abstract":"<p><strong>Background & aims: </strong>In response to injury, pancreatic acinar cells undergo acinar-to-ductal metaplasia (ADM), marked by loss of acinar identity and acquisition of ductal features. While ADM can resolve to support tissue repair, it may also persist and serve as a precursor to pancreatic cancer. Whether diverse pancreatic stressors drive a shared or context-specific ADM program remains unclear. We sought to comprehensively define metaplastic responses to clinically relevant exocrine pancreas diseases known to increase cancer risk.</p><p><strong>Methods: </strong>We profiled ADM and the surrounding microenvironment across mouse models of exocrine disease-including acute, recurrent, and chronic pancreatitis, as well as in the setting of oncogenic Kras-capturing over 300,000 single cells. To enable high-quality transcriptomic profiling in enzyme-rich tissue, we leveraged FixNCut, a method that preserves RNA integrity in the exocrine pancreas. Findings were validated in human pancreas tissue using CosMx spatial transcriptomics.</p><p><strong>Results: </strong>We identify a conserved acinar response across disease contexts that gives rise to previously unrecognized distinct metaplastic states, including a \"gateway\" ADM population that precedes more advanced metaplastic states marked by complete loss of acinar identity. In pancreatic intraepithelial neoplasia (PanIN) precancerous lesions, we detect classical-like and basal-like states, suggesting that pancreatic cancer subtypes are specified much earlier than previously appreciated. In Kras-mutant tissue, we identify a second wave of inflammation and the emergence of an immunosuppressive niche, coinciding with PanIN formation CONCLUSIONS: Our findings define a conserved program of acinar plasticity across exocrine pancreas diseases. We further link unresolved ADM to immune remodeling during precursor lesion formation and observe the emergence of pancreatic cancer subtypes in early PanIN lesions.</p>","PeriodicalId":55974,"journal":{"name":"Cellular and Molecular Gastroenterology and Hepatology","volume":" ","pages":"101717"},"PeriodicalIF":7.1,"publicationDate":"2025-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145844549","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Role of innate oral immunity and the salivary fluid in inflammatory bowel disease. 先天口腔免疫和唾液在炎症性肠病中的作用。
IF 7.1 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-22 DOI: 10.1016/j.jcmgh.2025.101713
Joydeep Aoun, Ahmed Kabrah, Malini Ahuja, Benjamin Leblanc, Changyu Zhang, Li Li, Yan Wang, Shmuel Muallem

Background & aims: Oral and gut health are tightly connected through their microbiome and immunity, including in disease states. The oral adaptive immunity contributes to the severity of inflammatory bowel disease (IBD). However, the role of oral innate immunity, and more specifically the saliva in gut microbiome and IBD, is poorly understood.

Methods: We used two mouse models with reduced saliva, NOD and Aqp5-/- mice, and recovery of salivation in the NOD mice by treatment with a CFTR corrector to examine the role of salivation in oral and gut microbiome, IBD, and survival.

Results: Analysis of the oral microbiome at various conditions revealed that the saliva has a minimal role in shaping the oral microbiome. However, salivation affected the composition of the gut microbiome. Moreover, the lack of saliva significantly delayed development of DSS-induced colitis, but resulted in a later, age-dependent, rapidly developed weight loss and death. The dual roles of the saliva were caused by two immunomodulatory peptides secreted by salivary glands. Fractionation and mass spectroscopy analysis identified trefoil factor 2 (TFF2) as a protective component and the cytokine macrophage migration inhibitory factor (MIF) as the damaging component of the saliva. The effects of the salivary fluid, TFF2, and MIF were primarily due to control of the gut barrier, rather than the gut microbiome. Scavenging salivary TFF2 and MIF with antibodies resulted in exacerbating and protection, respectively, of IBD.

Conclusions: The oral innate immunity has a major role in shaping the gut microbiome through secretion of MIF and TFF2. Control of MIF and TFF2 can benefit the treatment of colitis.

背景与目的:口腔和肠道健康通过它们的微生物群和免疫紧密相连,包括在疾病状态下。口腔适应性免疫与炎症性肠病(IBD)的严重程度有关。然而,口腔先天免疫,更具体地说是唾液在肠道微生物群和IBD中的作用,人们知之甚少。方法:我们使用两种唾液减少的小鼠模型,NOD和Aqp5-/-小鼠,以及通过CFTR校正剂治疗NOD小鼠的唾液分泌恢复,来研究唾液分泌在口腔和肠道微生物组、IBD和生存中的作用。结果:对不同条件下口腔微生物组的分析表明,唾液在口腔微生物组的形成中起着最小的作用。然而,唾液分泌会影响肠道微生物群的组成。此外,唾液的缺乏显著延缓了dss诱导的结肠炎的发展,但导致了较晚的、年龄依赖性的、迅速发展的体重下降和死亡。唾液的双重作用是由唾液腺分泌的两种免疫调节肽引起的。分离和质谱分析鉴定三叶因子2 (TFF2)为唾液的保护成分,细胞因子巨噬细胞迁移抑制因子(MIF)为唾液的损害成分。唾液液、TFF2和MIF的作用主要是由于肠道屏障的控制,而不是肠道微生物群。用抗体清除唾液TFF2和MIF分别导致IBD的恶化和保护。结论:口腔先天免疫通过分泌MIF和TFF2对肠道微生物群的形成起重要作用。控制MIF和TFF2有利于结肠炎的治疗。
{"title":"Role of innate oral immunity and the salivary fluid in inflammatory bowel disease.","authors":"Joydeep Aoun, Ahmed Kabrah, Malini Ahuja, Benjamin Leblanc, Changyu Zhang, Li Li, Yan Wang, Shmuel Muallem","doi":"10.1016/j.jcmgh.2025.101713","DOIUrl":"https://doi.org/10.1016/j.jcmgh.2025.101713","url":null,"abstract":"<p><strong>Background & aims: </strong>Oral and gut health are tightly connected through their microbiome and immunity, including in disease states. The oral adaptive immunity contributes to the severity of inflammatory bowel disease (IBD). However, the role of oral innate immunity, and more specifically the saliva in gut microbiome and IBD, is poorly understood.</p><p><strong>Methods: </strong>We used two mouse models with reduced saliva, NOD and Aqp5<sup>-/-</sup> mice, and recovery of salivation in the NOD mice by treatment with a CFTR corrector to examine the role of salivation in oral and gut microbiome, IBD, and survival.</p><p><strong>Results: </strong>Analysis of the oral microbiome at various conditions revealed that the saliva has a minimal role in shaping the oral microbiome. However, salivation affected the composition of the gut microbiome. Moreover, the lack of saliva significantly delayed development of DSS-induced colitis, but resulted in a later, age-dependent, rapidly developed weight loss and death. The dual roles of the saliva were caused by two immunomodulatory peptides secreted by salivary glands. Fractionation and mass spectroscopy analysis identified trefoil factor 2 (TFF2) as a protective component and the cytokine macrophage migration inhibitory factor (MIF) as the damaging component of the saliva. The effects of the salivary fluid, TFF2, and MIF were primarily due to control of the gut barrier, rather than the gut microbiome. Scavenging salivary TFF2 and MIF with antibodies resulted in exacerbating and protection, respectively, of IBD.</p><p><strong>Conclusions: </strong>The oral innate immunity has a major role in shaping the gut microbiome through secretion of MIF and TFF2. Control of MIF and TFF2 can benefit the treatment of colitis.</p>","PeriodicalId":55974,"journal":{"name":"Cellular and Molecular Gastroenterology and Hepatology","volume":" ","pages":"101713"},"PeriodicalIF":7.1,"publicationDate":"2025-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145828890","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
期刊
Cellular and Molecular Gastroenterology and Hepatology
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:604180095
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