首页 > 最新文献

Cellular and Molecular Gastroenterology and Hepatology最新文献

英文 中文
A Dual-compartment Scaffolding Role for Receptor for Activate C Kinase 1 in Hepatic Glucagon Signaling and Gluconeogenesis RACK1在肝胰高血糖素信号传导和糖异生中的双室支架作用。
IF 7.1 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-10-20 DOI: 10.1016/j.jcmgh.2025.101666
Cancan Lyu , Ling Yang , Songhai Chen

Background & Aims

The hepatic glucagon–protein kinase A (PKA)–cAMP response element-binding protein (CREB) signaling axis plays a central role in regulating gluconeogenesis and maintaining glucose homeostasis during fasting. However, the mechanisms that govern the spatial coordination and substrate specificity of this pathway remain incompletely understood. This study determines the role of the scaffolding protein RACK1 (Receptor for Activated C Kinase 1) in orchestrating glucagon signaling to regulate hepatic gluconeogenesis.

Methods

RACK1 was acutely deleted in mouse liver and primary hepatocytes. Metabolic phenotypes were assessed by glucose, pyruvate, glucagon and insulin tolerance tests, as well as hepatocyte glucose production assays. Protein interactions were examined by coimmunoprecipitation, glutathione S-transferase (GST) pulldown, and miniTurbo-ID-mediated proximity labeling. Subcellular localization and signaling events were assessed by Western blotting, confocal microscopy, and cellular fractionation. Functional rescue was achieved by hepatic expression of a constitutively active PKA catalytic subunit (PKAcαW196R).

Results

Acute hepatic RACK1 deficiency caused fasting hypoglycemia, impaired gluconeogenesis, and improved glucose, pyruvate, and glucagon tolerance without affecting insulin signaling. RACK1 directly bound glucagon receptor (GCGR) and PKA regulatory (RIIα) and catalytic (PKAcα) subunits, as well as CREB, functioning as a dual-compartment scaffold assembling GCGR–PKA complexes at the plasma membrane and PKAcα–CREB complexes in the nucleus. Loss of RACK1 impaired PKAcα translocation, CREB phosphorylation, and gluconeogenic gene expression. These defects were rescued by PKAcαW196R expression. Overexpression of RACK1 WD1–2 and WD3–4 domains, which mediate PKA, GCGR, and CREB interactions, similarly disrupted PKA signaling and gluconeogenesis.

Conclusions

RACK1 functions as a dual-compartment scaffold, assembling GCGR–PKA at the plasma membrane and PKAcα–CREB in the nucleus, enabling precise glucagon signaling and gluconeogenesis while sparing insulin pathways, thereby ensuring compartmentalized regulation of hepatic glucose homeostasis.
背景与目的:胰高血糖素- pka - creb信号轴在空腹期间调节糖异生和维持葡萄糖稳态中起核心作用。然而,控制这一途径的空间协调和底物特异性的机制仍然不完全清楚。本研究确定了支架蛋白RACK1(活化C激酶1受体)在胰高血糖素信号传导调节肝脏糖异生中的作用。方法:在小鼠肝脏和原代肝细胞中急性缺失RACK1。代谢表型通过葡萄糖、丙酮酸、胰高血糖素和胰岛素耐量试验以及肝细胞葡萄糖生成测定来评估。通过共免疫沉淀、GST下拉和miniturbo - id介导的接近标记来检测蛋白质相互作用。亚细胞定位和信号事件通过Western blotting、共聚焦显微镜和细胞分离进行评估。功能恢复是通过肝脏表达组成活性PKA催化亚基(PKAcαW196R)实现的。结果:急性肝脏RACK1缺乏导致空腹低血糖,糖异生受损,改善葡萄糖、丙酮酸和胰高血糖素耐量,但不影响胰岛素信号传导。RACK1直接结合GCGR、PKA调控亚基(RIIα)和催化亚基(PKAcα)以及CREB,作为双室支架在质膜上组装GCGR-PKA复合物和细胞核内组装PKAcα-CREB复合物。RACK1缺失会损害PKAcα易位、CREB磷酸化和糖异生基因表达。这些缺陷通过表达PKAcαW196R得以修复。介导PKA、GCGR和CREB相互作用的RACK1 WD1-2和WD3-4结构域的过表达同样会破坏PKA信号传导和糖异生。结论:RACK1作为一个双室支架,在质膜上组装GCGR-PKA和细胞核内组装PKAcα-CREB,在保留胰岛素通路的同时,实现精确的胰高血糖素信号传导和糖异生,从而确保肝糖稳态的区室化调节。
{"title":"A Dual-compartment Scaffolding Role for Receptor for Activate C Kinase 1 in Hepatic Glucagon Signaling and Gluconeogenesis","authors":"Cancan Lyu ,&nbsp;Ling Yang ,&nbsp;Songhai Chen","doi":"10.1016/j.jcmgh.2025.101666","DOIUrl":"10.1016/j.jcmgh.2025.101666","url":null,"abstract":"<div><h3>Background &amp; Aims</h3><div>The hepatic glucagon–protein kinase A (PKA)–cAMP response element-binding protein (CREB) signaling axis plays a central role in regulating gluconeogenesis and maintaining glucose homeostasis during fasting. However, the mechanisms that govern the spatial coordination and substrate specificity of this pathway remain incompletely understood. This study determines the role of the scaffolding protein RACK1 (Receptor for Activated C Kinase 1) in orchestrating glucagon signaling to regulate hepatic gluconeogenesis.</div></div><div><h3>Methods</h3><div>RACK1 was acutely deleted in mouse liver and primary hepatocytes. Metabolic phenotypes were assessed by glucose, pyruvate, glucagon and insulin tolerance tests, as well as hepatocyte glucose production assays. Protein interactions were examined by coimmunoprecipitation, glutathione S-transferase (GST) pulldown, and miniTurbo-ID-mediated proximity labeling. Subcellular localization and signaling events were assessed by Western blotting, confocal microscopy, and cellular fractionation. Functional rescue was achieved by hepatic expression of a constitutively active PKA catalytic subunit (PKAcα<sup>W196R</sup>).</div></div><div><h3>Results</h3><div>Acute hepatic RACK1 deficiency caused fasting hypoglycemia, impaired gluconeogenesis, and improved glucose, pyruvate, and glucagon tolerance without affecting insulin signaling. RACK1 directly bound glucagon receptor (GCGR) and PKA regulatory (RIIα) and catalytic (PKAcα) subunits, as well as CREB, functioning as a dual-compartment scaffold assembling GCGR–PKA complexes at the plasma membrane and PKAcα–CREB complexes in the nucleus. Loss of RACK1 impaired PKAcα translocation, CREB phosphorylation, and gluconeogenic gene expression. These defects were rescued by PKAcα<sup>W196R</sup> expression. Overexpression of RACK1 WD1–2 and WD3–4 domains, which mediate PKA, GCGR, and CREB interactions, similarly disrupted PKA signaling and gluconeogenesis.</div></div><div><h3>Conclusions</h3><div>RACK1 functions as a dual-compartment scaffold, assembling GCGR–PKA at the plasma membrane and PKAcα–CREB in the nucleus, enabling precise glucagon signaling and gluconeogenesis while sparing insulin pathways, thereby ensuring compartmentalized regulation of hepatic glucose homeostasis.</div></div>","PeriodicalId":55974,"journal":{"name":"Cellular and Molecular Gastroenterology and Hepatology","volume":"20 2","pages":"Article 101666"},"PeriodicalIF":7.1,"publicationDate":"2025-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145338320","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
Mitochondrial Metabolism Meets LncRNA Biology: The Role of uc.173 in Intestinal Homeostasis. 线粒体代谢与LncRNA生物学:uc的作用173肠内稳态。
IF 7.1 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-10-18 DOI: 10.1016/j.jcmgh.2025.101662
Hua Geng, Joann Romano-Keeler, Xiao-Di Tan
{"title":"Mitochondrial Metabolism Meets LncRNA Biology: The Role of uc.173 in Intestinal Homeostasis.","authors":"Hua Geng, Joann Romano-Keeler, Xiao-Di Tan","doi":"10.1016/j.jcmgh.2025.101662","DOIUrl":"10.1016/j.jcmgh.2025.101662","url":null,"abstract":"","PeriodicalId":55974,"journal":{"name":"Cellular and Molecular Gastroenterology and Hepatology","volume":" ","pages":"101662"},"PeriodicalIF":7.1,"publicationDate":"2025-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145338340","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 Intestinal Cues Promoting Serotonin Release From the Human Gut. 肠道信号促进人体肠道释放血清素。
IF 7.1 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-10-18 DOI: 10.1016/j.jcmgh.2025.101664
Sara C Di Rienzi
{"title":"The Intestinal Cues Promoting Serotonin Release From the Human Gut.","authors":"Sara C Di Rienzi","doi":"10.1016/j.jcmgh.2025.101664","DOIUrl":"10.1016/j.jcmgh.2025.101664","url":null,"abstract":"","PeriodicalId":55974,"journal":{"name":"Cellular and Molecular Gastroenterology and Hepatology","volume":" ","pages":"101664"},"PeriodicalIF":7.1,"publicationDate":"2025-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145338346","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
Probiotics Mitigate High-cholesterol Diet-driven Fatty Liver and Pancreatic Cancer by Restoring Macrophage Homeostasis 益生菌通过恢复巨噬细胞稳态减轻高胆固醇饮食驱动的脂肪肝和胰腺癌。
IF 7.1 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-10-18 DOI: 10.1016/j.jcmgh.2025.101644
Deepti Parida , Swayambara Mishra , Amlan Priyadarshee Mohapatra , Manisha Sethi , Kirti Ranjan Das , Salona Kar , Voddu Suresh , Shrikrishna Jayaram Bhagat , Amruta Mohapatra , Supriya Halder , Shantibhusan Senapati

Background & Aims

High dietary cholesterol is a known risk factor for metabolic dysfunction-associated steatotic liver disease (MASLD) and its associated hepatic carcinogenesis; however, its effect on pancreatic ductal adenocarcinoma (PDAC) is yet to be investigated. The current study explored the mechanistic association between high dietary cholesterol, MASLD, and PDAC. Importantly, we aimed to evaluate the effect of a multi-strain probiotic formulation on hypercholesterolemia-driven MASLD and PDAC.

Methods

In this study, wild-type (C57BL/6) and KC (Pdx-1 Cre; KrasLSL-G12D) mice were fed either with regular diet or high cholesterol and cholic acid diet (HCCD) and received an oral probiotic consortium LR+F15 (Lactobacillus rhamnosus GG and Lactiplantibacillus plantarum ILSF15). Additionally, we also used a syngeneic orthotopic murine PDAC model to evaluate the efficacy of this probiotic consortium. For mechanistic studies, pancreas, liver, intestine, peri-pancreatic fats, peritoneal cells/lavage, and blood were evaluated for metabolic, inflammatory, and malignant changes through histology, enzyme-linked immunosorbent assay, flow cytometry, and quantitative reverse transcription polymerase chain reaction.

Results

HCCD induced nonobese MASLD and PDAC progression, which was eased upon probiotics intervention. Importantly, it also increased the survival of the HCCD-fed KC mice. The probiotic intervention protected against HCCD-induced leaky gut, gut microbiota translocation, and inflammatory milieu in different tissues. Interestingly, HCCD significantly increased the population of pro-inflammatory/pro-tumorigenic peritoneal macrophages, which got normalized upon probiotic administration.

Conclusions

The probiotic formulation LR+F15 significantly suppressed HCCD-induced MASLD and PDAC progression partly through suppressing leaky gut and normalizing peritoneal macrophages’ inflammatory properties. These findings encourage evaluation of the potential benefits of this probiotic consortium in combination with the existing therapies against PDAC in the future.
背景与目的:高膳食胆固醇是代谢功能障碍相关脂肪变性肝病(MASLD)及其相关肝癌发生的已知危险因素;然而,其对胰腺导管腺癌(PDAC)的影响尚未研究。目前的研究探讨了高膳食胆固醇、MASLD和PDAC之间的机制联系。重要的是,我们旨在评估多菌株益生菌制剂对高胆固醇血症驱动的MASLD和PDAC的影响。方法:将野生型(C57BL/6)和KC型(Pdx-1 Cre; KrasLSL-G12D)小鼠分别饲喂常规日粮(RD)和高胆固醇高胆酸日粮(HCCD),并口服益生菌联合剂LR+F15(鼠李糖乳杆菌GG和植物乳杆菌ILSF15)。此外,我们还使用同源原位小鼠PDAC模型来评估该益生菌联盟的功效。对于机制研究,通过组织学、ELISA、流式细胞术和qRT-PCR评估胰腺、肝脏、肠道、胰腺周围脂肪、腹膜细胞/灌洗液和血液的代谢、炎症和恶性变化。结果:HCCD诱导非肥胖型MASLD和PDAC进展,益生菌干预后缓解。重要的是,它还提高了喂食hccd的KC小鼠的存活率。益生菌干预可以防止hccd诱导的肠漏、肠道菌群移位和不同组织的炎症环境。有趣的是,HCCD显著增加了促炎/促肿瘤腹膜巨噬细胞(PM)的数量,并在给予益生菌后恢复正常。结论:益生菌制剂LR+F15通过抑制漏肠和使pm炎症特性正常化,显著抑制hccd诱导的MASLD和PDAC进展。这些发现鼓励对益生菌联合现有治疗PDAC的潜在益处进行评估。
{"title":"Probiotics Mitigate High-cholesterol Diet-driven Fatty Liver and Pancreatic Cancer by Restoring Macrophage Homeostasis","authors":"Deepti Parida ,&nbsp;Swayambara Mishra ,&nbsp;Amlan Priyadarshee Mohapatra ,&nbsp;Manisha Sethi ,&nbsp;Kirti Ranjan Das ,&nbsp;Salona Kar ,&nbsp;Voddu Suresh ,&nbsp;Shrikrishna Jayaram Bhagat ,&nbsp;Amruta Mohapatra ,&nbsp;Supriya Halder ,&nbsp;Shantibhusan Senapati","doi":"10.1016/j.jcmgh.2025.101644","DOIUrl":"10.1016/j.jcmgh.2025.101644","url":null,"abstract":"<div><h3>Background &amp; Aims</h3><div>High dietary cholesterol is a known risk factor for metabolic dysfunction-associated steatotic liver disease (MASLD) and its associated hepatic carcinogenesis; however, its effect on pancreatic ductal adenocarcinoma (PDAC) is yet to be investigated. The current study explored the mechanistic association between high dietary cholesterol, MASLD, and PDAC. Importantly, we aimed to evaluate the effect of a multi-strain probiotic formulation on hypercholesterolemia-driven MASLD and PDAC.</div></div><div><h3>Methods</h3><div>In this study, wild-type (C57BL/6) and KC (Pdx-1 Cre; Kras<sup>LSL-G12D</sup>) mice were fed either with regular diet or high cholesterol and cholic acid diet (HCCD) and received an oral probiotic consortium LR+F15 (<em>Lactobacillus rhamnosus</em> GG and <em>Lactiplantibacillus plantarum</em> ILSF15). Additionally, we also used a syngeneic orthotopic murine PDAC model to evaluate the efficacy of this probiotic consortium. For mechanistic studies, pancreas, liver, intestine, peri-pancreatic fats, peritoneal cells/lavage, and blood were evaluated for metabolic, inflammatory, and malignant changes through histology, enzyme-linked immunosorbent assay, flow cytometry, and quantitative reverse transcription polymerase chain reaction.</div></div><div><h3>Results</h3><div>HCCD induced nonobese MASLD and PDAC progression, which was eased upon probiotics intervention. Importantly, it also increased the survival of the HCCD-fed KC mice. The probiotic intervention protected against HCCD-induced leaky gut, gut microbiota translocation, and inflammatory milieu in different tissues. Interestingly, HCCD significantly increased the population of pro-inflammatory/pro-tumorigenic peritoneal macrophages, which got normalized upon probiotic administration.</div></div><div><h3>Conclusions</h3><div>The probiotic formulation LR+F15 significantly suppressed HCCD-induced MASLD and PDAC progression partly through suppressing leaky gut and normalizing peritoneal macrophages’ inflammatory properties. These findings encourage evaluation of the potential benefits of this probiotic consortium in combination with the existing therapies against PDAC in the future.</div></div>","PeriodicalId":55974,"journal":{"name":"Cellular and Molecular Gastroenterology and Hepatology","volume":"20 3","pages":"Article 101644"},"PeriodicalIF":7.1,"publicationDate":"2025-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145330944","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
CREB Fuels Tumorigenesis in Alcoholic Pancreatitis. CREB促进酒精性胰腺炎的肿瘤发生
IF 7.1 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-10-18 DOI: 10.1016/j.jcmgh.2025.101660
Urvinder Kaur Sardarni, Jennifer M Bailey-Lundberg
{"title":"CREB Fuels Tumorigenesis in Alcoholic Pancreatitis.","authors":"Urvinder Kaur Sardarni, Jennifer M Bailey-Lundberg","doi":"10.1016/j.jcmgh.2025.101660","DOIUrl":"10.1016/j.jcmgh.2025.101660","url":null,"abstract":"","PeriodicalId":55974,"journal":{"name":"Cellular and Molecular Gastroenterology and Hepatology","volume":" ","pages":"101660"},"PeriodicalIF":7.1,"publicationDate":"2025-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145338286","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
When Liver Slices Meet Single-cell Technology. 当肝脏切片与单细胞技术相遇。
IF 7.1 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-10-18 DOI: 10.1016/j.jcmgh.2025.101661
Mi Jeong Heo, Kang Ho Kim
{"title":"When Liver Slices Meet Single-cell Technology.","authors":"Mi Jeong Heo, Kang Ho Kim","doi":"10.1016/j.jcmgh.2025.101661","DOIUrl":"10.1016/j.jcmgh.2025.101661","url":null,"abstract":"","PeriodicalId":55974,"journal":{"name":"Cellular and Molecular Gastroenterology and Hepatology","volume":" ","pages":"101661"},"PeriodicalIF":7.1,"publicationDate":"2025-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145338266","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
Barrington’s Nucleus: A Pontine Defecation Brain Area Exhibiting Prompt and Delayed Defecation Responses 巴林顿核:脑桥排便脑区,表现出迅速和延迟的排便反应。
IF 7.1 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-10-10 DOI: 10.1016/j.jcmgh.2025.101635
Kota Bussaka , Yoshimasa Tanaka , Kunio Kondoh , Ken-ichiro Nakajima , Takatoshi Chinen , Xiaopeng Bai , Yosuke Minoda , Hiroko Ikeda , Kazuki Inamura , Tsubasa Takeshima , Haruei Ogino , Eikichi Ihara , Yasuhiko Minokoshi , Yoshihiro Ogawa

Background & Aims

Chronic constipation has attracted considerable attention because of its negative impact on quality of life. Although defecation depends on local anorectal motility coordinated by the central nervous system, how it is regulated by the brain remains unclear.

Methods

Brain areas responsible for defecation, known as the defecation brain area (DBA), were identified using a trans-synaptic tracing virus, pseudorabies virus (PRV). Candidate DBAs were assessed using opto- and chemogenetic methods and in vivo monitoring of neural activity.

Results

A significant number of PRV-infected cells were observed in the Barrington’s nucleus (Bar), locus coeruleus (LC), ventrolateral periaqueductal gray (vlPAG), and paraventricular hypothalamic nucleus (PVH) following virus infection in the distal colon. Opto- and chemogenetic activation studies revealed that vesicular glutamate transporter 2 (VGluT2) neurons in the Bar and LC, and corticotropin-releasing hormone (CRH) neurons in the Bar exhibit prompt (short-acting) and delayed (long-lasting) contractions in the distal colon, respectively. Their neural activities increased and peaked during spontaneous defecation. In contrast, activation of tyrosine hydroxylase neurons in the LC, which co-express VGluT2, exhibited no response. PRV experiments revealed that PVHVGluT2 and vlPAGCRH neurons are upstream neurons that connect to BarVGluT2 neurons, and their optogenetic activation resulted in a contraction of the distal colon.

Conclusions

The study is the first to show that the Bar works as the pontine DBA, where BarVGluT2 and BarCRH neurons exert prompt and delayed defecation activity, respectively. PVHVGluT2 and vlPAGCRH neurons are candidates for upstream neurons that regulate defecation through BarVGluT2 neurons.
背景与目的:慢性便秘因其对生活质量的负面影响而受到广泛关注。虽然排便依赖于由中枢神经系统协调的局部肛肠运动,但它是如何由大脑调节的尚不清楚。方法:利用假狂犬病毒(PRV)的跨突触追踪病毒,对负责排便的脑区,即排便脑区(DBA)进行鉴定。候选dba使用光学和化学发生方法以及体内神经活动监测进行评估。结果:在远端结肠病毒感染后,在巴林顿核(Bar)、蓝斑核(LC)、腹外侧导水管周围灰核(vlPAG)和室旁下丘脑核(PVH)中观察到大量prv感染细胞。光学和化学激活研究显示,Bar和LC中的水疱性谷氨酸转运蛋白2 (VGluT2)神经元以及Bar中的促肾上腺皮质激素释放激素(CRH)神经元分别在结肠远端表现出提示性(短效)和延迟性(持久)收缩。他们的神经活动增加,并在自然排便时达到高峰。相比之下,LC中共同表达VGluT2的酪氨酸羟化酶神经元的激活没有反应。PRV实验显示PVHVGluT2和vlPAGCRH神经元是连接BarVGluT2神经元的上游神经元,它们的光遗传激活导致远端结肠收缩。结论:该研究首次表明,Bar作为脑桥的DBA,其中BarVGluT2和BarCRH神经元分别发挥及时和延迟排便活动。PVHVGluT2和vlPAGCRH神经元是通过BarVGluT2神经元调节排便的上游神经元的候选细胞。
{"title":"Barrington’s Nucleus: A Pontine Defecation Brain Area Exhibiting Prompt and Delayed Defecation Responses","authors":"Kota Bussaka ,&nbsp;Yoshimasa Tanaka ,&nbsp;Kunio Kondoh ,&nbsp;Ken-ichiro Nakajima ,&nbsp;Takatoshi Chinen ,&nbsp;Xiaopeng Bai ,&nbsp;Yosuke Minoda ,&nbsp;Hiroko Ikeda ,&nbsp;Kazuki Inamura ,&nbsp;Tsubasa Takeshima ,&nbsp;Haruei Ogino ,&nbsp;Eikichi Ihara ,&nbsp;Yasuhiko Minokoshi ,&nbsp;Yoshihiro Ogawa","doi":"10.1016/j.jcmgh.2025.101635","DOIUrl":"10.1016/j.jcmgh.2025.101635","url":null,"abstract":"<div><h3>Background &amp; Aims</h3><div>Chronic constipation has attracted considerable attention because of its negative impact on quality of life. Although defecation depends on local anorectal motility coordinated by the central nervous system, how it is regulated by the brain remains unclear.</div></div><div><h3>Methods</h3><div>Brain areas responsible for defecation, known as the defecation brain area (DBA), were identified using a trans-synaptic tracing virus, pseudorabies virus (PRV). Candidate DBAs were assessed using opto- and chemogenetic methods and in vivo monitoring of neural activity.</div></div><div><h3>Results</h3><div>A significant number of PRV-infected cells were observed in the Barrington’s nucleus (Bar), locus coeruleus (LC), ventrolateral periaqueductal gray (vlPAG), and paraventricular hypothalamic nucleus (PVH) following virus infection in the distal colon. Opto- and chemogenetic activation studies revealed that vesicular glutamate transporter 2 (VGluT2) neurons in the Bar and LC, and corticotropin-releasing hormone (CRH) neurons in the Bar exhibit prompt (short-acting) and delayed (long-lasting) contractions in the distal colon, respectively. Their neural activities increased and peaked during spontaneous defecation. In contrast, activation of tyrosine hydroxylase neurons in the LC, which co-express VGluT2, exhibited no response. PRV experiments revealed that PVH<sup>VGluT2</sup> and vlPAG<sup>CRH</sup> neurons are upstream neurons that connect to Bar<sup>VGluT2</sup> neurons, and their optogenetic activation resulted in a contraction of the distal colon.</div></div><div><h3>Conclusions</h3><div>The study is the first to show that the Bar works as the pontine DBA, where Bar<sup>VGluT2</sup> and Bar<sup>CRH</sup> neurons exert prompt and delayed defecation activity, respectively. PVH<sup>VGluT2</sup> and vlPAG<sup>CRH</sup> neurons are candidates for upstream neurons that regulate defecation through Bar<sup>VGluT2</sup> neurons.</div></div>","PeriodicalId":55974,"journal":{"name":"Cellular and Molecular Gastroenterology and Hepatology","volume":"20 1","pages":"Article 101635"},"PeriodicalIF":7.1,"publicationDate":"2025-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145281942","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
Sympathetic Overactivation Drives Colonic Eosinophil Infiltration Linked to Visceral Hypersensitivity in Irritable Bowel Syndrome 交感神经过度激活驱动结肠嗜酸性粒细胞浸润与肠易激综合征的内脏超敏反应有关。
IF 7.1 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-10-09 DOI: 10.1016/j.jcmgh.2025.101658
Shaoqi Duan , Hirosato Kanda , Feng Zhu , Masamichi Okubo , Taro Koike , Yoshiya Ohno , Toshiyuki Tanaka , Yukiko Harima , Kazunari Miyamichi , Hirokazu Fukui , Shinichiro Shinzaki , Yilong Cui , Koichi Noguchi , Yi Dai

Background & Aims

Mucosal immune alteration is a characteristic clinical manifestation of irritable bowel syndrome (IBS), and its symptoms are often triggered by psychological stress. The present study aimed to investigate the impact of early life stress-associated dysfunction of the sympathetic nervous system (SNS) on mucosal immune changes in the gastrointestinal tract (GI) and its contribution to visceral hypersensitivity of IBS.

Methods

We utilized a traditional animal model of IBS with maternal separation (MS) and evaluated colorectal hypersensitivity, immune alteration, and SNS activity in adult rats with MS. We conducted a series of experiments to manipulate peripheral SNS activity pharmacologically and chemogenetically to explore the interaction between SNS activity and GI events.

Results

The MS-induced IBS model exhibited visceral hypersensitivity and eosinophilic infiltration in the colonic mucosa, along with SNS overactivation. Degeneration of the SNS using 6-OHDA neurotoxin decreased eosinophil infiltration and visceral hypersensitivity in the MS model. Notably, specific chemogenetic activation of the peripheral SNS induced eosinophil infiltration in the intestinal mucosa through the noradrenergic signaling-mediated release of eotaxin-1 from mesenchymal cells.

Conclusions

This study highlights the critical role of SNS overactivation in eotaxin-1-driven eosinophil infiltration in the colon, leading to the development of visceral hypersensitivity in IBS. The results provide important insights into the mechanistic links among increased sympathetic activity, mucosal immune alteration, and visceral hypersensitivity in individuals with IBS, suggesting potential therapeutic approaches.
背景与目的:黏膜免疫改变是肠易激综合征(IBS)的特征性临床表现,其症状常由心理应激引发。本研究旨在探讨早期应激相关交感神经系统(SNS)功能障碍对胃肠道(GI)粘膜免疫变化的影响及其对肠易激综合征内脏超敏反应的影响。方法:采用传统的IBS伴母分离(MS)动物模型,对成年MS大鼠的结肠超敏反应、免疫改变和SNS活性进行了评估。我们通过一系列实验,从药理学和化学遗传学角度操纵外周SNS活性,探讨SNS活性与GI事件之间的相互作用。结果:ms诱导的IBS模型表现为内脏超敏,结肠粘膜嗜酸性粒细胞浸润,SNS过度激活。在MS模型中,使用6-OHDA神经毒素使SNS变性可减少嗜酸性粒细胞浸润和内脏过敏。值得注意的是,外周SNS的特异性化学发生激活通过间充质细胞释放eotaxin-1的去甲肾上腺素能信号介导的肠粘膜嗜酸性粒细胞浸润。结论:本研究强调了SNS过度激活在eotaxin-1驱动的结肠嗜酸性粒细胞浸润中起关键作用,导致肠易激综合征内脏超敏反应的发生。该结果为IBS患者交感神经活动增加、粘膜免疫改变和内脏过敏之间的机制联系提供了重要见解,并提出了潜在的治疗方法。
{"title":"Sympathetic Overactivation Drives Colonic Eosinophil Infiltration Linked to Visceral Hypersensitivity in Irritable Bowel Syndrome","authors":"Shaoqi Duan ,&nbsp;Hirosato Kanda ,&nbsp;Feng Zhu ,&nbsp;Masamichi Okubo ,&nbsp;Taro Koike ,&nbsp;Yoshiya Ohno ,&nbsp;Toshiyuki Tanaka ,&nbsp;Yukiko Harima ,&nbsp;Kazunari Miyamichi ,&nbsp;Hirokazu Fukui ,&nbsp;Shinichiro Shinzaki ,&nbsp;Yilong Cui ,&nbsp;Koichi Noguchi ,&nbsp;Yi Dai","doi":"10.1016/j.jcmgh.2025.101658","DOIUrl":"10.1016/j.jcmgh.2025.101658","url":null,"abstract":"<div><h3>Background &amp; Aims</h3><div>Mucosal immune alteration is a characteristic clinical manifestation of irritable bowel syndrome (IBS), and its symptoms are often triggered by psychological stress. The present study aimed to investigate the impact of early life stress-associated dysfunction of the sympathetic nervous system (SNS) on mucosal immune changes in the gastrointestinal tract (GI) and its contribution to visceral hypersensitivity of IBS.</div></div><div><h3>Methods</h3><div>We utilized a traditional animal model of IBS with maternal separation (MS) and evaluated colorectal hypersensitivity, immune alteration, and SNS activity in adult rats with MS. We conducted a series of experiments to manipulate peripheral SNS activity pharmacologically and chemogenetically to explore the interaction between SNS activity and GI events.</div></div><div><h3>Results</h3><div>The MS-induced IBS model exhibited visceral hypersensitivity and eosinophilic infiltration in the colonic mucosa, along with SNS overactivation. Degeneration of the SNS using 6-OHDA neurotoxin decreased eosinophil infiltration and visceral hypersensitivity in the MS model. Notably, specific chemogenetic activation of the peripheral SNS induced eosinophil infiltration in the intestinal mucosa through the noradrenergic signaling-mediated release of eotaxin-1 from mesenchymal cells.</div></div><div><h3>Conclusions</h3><div>This study highlights the critical role of SNS overactivation in eotaxin-1-driven eosinophil infiltration in the colon, leading to the development of visceral hypersensitivity in IBS. The results provide important insights into the mechanistic links among increased sympathetic activity, mucosal immune alteration, and visceral hypersensitivity in individuals with IBS, suggesting potential therapeutic approaches.</div></div>","PeriodicalId":55974,"journal":{"name":"Cellular and Molecular Gastroenterology and Hepatology","volume":"20 3","pages":"Article 101658"},"PeriodicalIF":7.1,"publicationDate":"2025-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145260063","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
Pathogenic OTUD3 Mutations Predispose to Ulcerative Colitis Due to Barrier Dysfunction 由于屏障功能障碍,致病性OTUD3突变易导致溃疡性结肠炎。
IF 7.1 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-10-09 DOI: 10.1016/j.jcmgh.2025.101659
Rabina Giri , Minyi Lee , Graham W. Magor , Anne-Sophie Bergot , Yaowu He , Thomas Kryza , Tashbib Khan , Veronika Schreiber , Robert J. Gordon , Rachid Zagani , Sumaira Z. Hasnain , Rohan Lourie , Adam Ewing , John D. Hooper , Ranjeny Thomas , Timothy H. Florin , Andrew Perkins , Manish Gala , Jakob Begun

Background & Aims

The contribution of common genetic polymorphisms to ulcerative colitis (UC) pathogenesis is modest; however, families with severe colitis may harbor rare variants with large effect sizes that highlight unrecognized pathways.

Methods

A multigenerational family with UC necessitating colectomy was identified. Whole exome sequencing of this kindred was performed, implicating a rare variant in OTUD3. Constitutive knock-out and intestinal specific Otud3 deficient and heterozygous mice were generated. OTUD3 expression in human colonic biopsies and intestinal organoids was assessed using quantitative reverse transcription polymerase chain reaction and immunofluorescence. Prevalence of rare, damaging variants were compared in distinct patient cohorts. Plasmids containing OTUD3 missense variants were introduced into cell lines where OTUD3 was disrupted to determine their effects on cellular response to cytokine stimulation.

Results

Constitutive disruption or heterozygosity of Otud3 in mice, or intestinal-specific deletion, resulted in impaired barrier integrity, tight-junction dysregulation, increased endoplasmic reticulum stress, and penetration of luminal bacteria deep into the colonic crypts that preceded a spontaneous progressive colitis. Analysis of distinct UC cohorts demonstrated enrichment of rare, damaging variants in OTUD3. Introduction of OTUD3 variants in intestinal cell lines phenocopied the epithelial immune dysregulation observed in knockout mice. Finally, OTUD3 mRNA and epithelial protein expression were decreased in the quiescent colonic epithelial tissue of genotype-unselected individuals with UC compared with matched non-UC controls.

Conclusions

Our results demonstrate that OTUD3 is required for colonic epithelial barrier function, and plays a role in the pathogenesis of UC.
背景和目的:常见遗传多态性对溃疡性结肠炎(UC)发病机制的贡献不大,然而,严重结肠炎的家庭可能存在具有大效应量的罕见变异,这些变异突出了未被识别的途径。方法:确定了一个多代人患有UC需要结肠切除术的家庭。对该亲缘进行了全外显子组测序,发现OTUD3存在罕见变异。产生了组成型敲除小鼠和肠道特异性Otud3缺陷小鼠和杂合小鼠。采用qRT-PCR和免疫荧光技术评估OTUD3在人结肠活检和肠道类器官中的表达。在不同的患者队列中比较罕见的、破坏性变异的患病率。将含有OTUD3错义变体的质粒引入细胞系,破坏OTUD3,以确定其对细胞对细胞因子刺激的反应的影响。结果:小鼠中Otud3的组成性破坏或杂合性,或肠道特异性缺失,导致屏障完整性受损,紧密连接失调,内质网应激增加,以及肠道细菌深入结肠隐窝,导致自发性进行性结肠炎。对不同UC队列的分析表明,OTUD3中存在罕见的破坏性变异。在肠细胞系中引入OTUD3变异体,表型化了在基因敲除小鼠中观察到的上皮免疫失调。最后,与匹配的非UC对照相比,未选择基因型的UC患者的静止结肠上皮组织中OTUD3 mRNA和上皮蛋白的表达降低。结论:我们的研究结果表明OTUD3是结肠上皮屏障功能所必需的,并在UC的发病机制中发挥作用。
{"title":"Pathogenic OTUD3 Mutations Predispose to Ulcerative Colitis Due to Barrier Dysfunction","authors":"Rabina Giri ,&nbsp;Minyi Lee ,&nbsp;Graham W. Magor ,&nbsp;Anne-Sophie Bergot ,&nbsp;Yaowu He ,&nbsp;Thomas Kryza ,&nbsp;Tashbib Khan ,&nbsp;Veronika Schreiber ,&nbsp;Robert J. Gordon ,&nbsp;Rachid Zagani ,&nbsp;Sumaira Z. Hasnain ,&nbsp;Rohan Lourie ,&nbsp;Adam Ewing ,&nbsp;John D. Hooper ,&nbsp;Ranjeny Thomas ,&nbsp;Timothy H. Florin ,&nbsp;Andrew Perkins ,&nbsp;Manish Gala ,&nbsp;Jakob Begun","doi":"10.1016/j.jcmgh.2025.101659","DOIUrl":"10.1016/j.jcmgh.2025.101659","url":null,"abstract":"<div><h3>Background &amp; Aims</h3><div>The contribution of common genetic polymorphisms to ulcerative colitis (UC) pathogenesis is modest; however, families with severe colitis may harbor rare variants with large effect sizes that highlight unrecognized pathways.</div></div><div><h3>Methods</h3><div>A multigenerational family with UC necessitating colectomy was identified. Whole exome sequencing of this kindred was performed, implicating a rare variant in <em>OTUD3</em>. Constitutive knock-out and intestinal specific <em>Otud3</em> deficient and heterozygous mice were generated. OTUD3 expression in human colonic biopsies and intestinal organoids was assessed using quantitative reverse transcription polymerase chain reaction and immunofluorescence. Prevalence of rare, damaging variants were compared in distinct patient cohorts. Plasmids containing <em>OTUD3</em> missense variants were introduced into cell lines where <em>OTUD3</em> was disrupted to determine their effects on cellular response to cytokine stimulation.</div></div><div><h3>Results</h3><div>Constitutive disruption or heterozygosity of <em>Otud3</em> in mice, or intestinal-specific deletion, resulted in impaired barrier integrity, tight-junction dysregulation, increased endoplasmic reticulum stress, and penetration of luminal bacteria deep into the colonic crypts that preceded a spontaneous progressive colitis. Analysis of distinct UC cohorts demonstrated enrichment of rare, damaging variants in <em>OTUD3</em>. Introduction of <em>OTUD3</em> variants in intestinal cell lines phenocopied the epithelial immune dysregulation observed in knockout mice. Finally, <em>OTUD3</em> mRNA and epithelial protein expression were decreased in the quiescent colonic epithelial tissue of genotype-unselected individuals with UC compared with matched non-UC controls.</div></div><div><h3>Conclusions</h3><div>Our results demonstrate that OTUD3 is required for colonic epithelial barrier function, and plays a role in the pathogenesis of UC.</div></div>","PeriodicalId":55974,"journal":{"name":"Cellular and Molecular Gastroenterology and Hepatology","volume":"20 2","pages":"Article 101659"},"PeriodicalIF":7.1,"publicationDate":"2025-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145259984","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 Placode Lineage Contributes to the Enteric Nervous System: A Caution for Cell Transplantation Therapy for Hirschsprung Disease 基板谱系有助于肠神经系统:巨结肠疾病细胞移植治疗的一个警告。
IF 7.1 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-10-03 DOI: 10.1016/j.jcmgh.2025.101657
Shigeru Sato, Henry M. Sucov, Takako Makita
{"title":"The Placode Lineage Contributes to the Enteric Nervous System: A Caution for Cell Transplantation Therapy for Hirschsprung Disease","authors":"Shigeru Sato,&nbsp;Henry M. Sucov,&nbsp;Takako Makita","doi":"10.1016/j.jcmgh.2025.101657","DOIUrl":"10.1016/j.jcmgh.2025.101657","url":null,"abstract":"","PeriodicalId":55974,"journal":{"name":"Cellular and Molecular Gastroenterology and Hepatology","volume":"20 2","pages":"Article 101657"},"PeriodicalIF":7.1,"publicationDate":"2025-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145234038","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