首页 > 最新文献

Metabolism: clinical and experimental最新文献

英文 中文
CHK1 attenuates cardiac dysfunction via suppressing SIRT1-ubiquitination CHK1 通过抑制 SIRT1 泛素化减轻心脏功能障碍
IF 10.8 1区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-10-24 DOI: 10.1016/j.metabol.2024.156048
Tong-Tong Yang , Liu-Hua Zhou , Ling-Feng Gu , Ling-Ling Qian , Yu-Lin Bao , Peng Jing , Jia-Teng Sun , Chong Du , Tian-Kai Shan , Si-Bo Wang , Wen-Jing Wang , Jia-Yi Chen , Ze-Mu Wang , Hao Wang , Qi-Ming Wang , Ru-Xing Wang , Lian-Sheng Wang

Background

Mitochondrial dysfunction is linked to myocardial ischemia-reperfusion (I/R) injury. Checkpoint kinase 1 (CHK1) could facilitate cardiomyocyte proliferation, however, its role on mitochondrial function in I/R injury remains unknown.

Methods

To investigate the role of CHK1 on mitochondrial function following I/R injury, cardiomyocyte-specific knockout/overexpression mouse models were generated. Adult mouse cardiomyocytes (AMCMs) were isolated for in vitro study. Mass spectrometry-proteomics analysis and protein co-immunoprecipitation assays were conducted to dissect the molecular mechanism.

Results

CHK1 was downregulated in myocardium post I/R and AMCMs post oxygen-glucose deprivation/re‑oxygenation (OGD/R). In vivo, CHK1 overexpression protected against I/R induced cardiac dysfunction, while heterogenous CHK1 knockout exacerbated cardiomyopathy. In vitro, CHK1 inhibited OGD/R-induced cardiomyocyte apoptosis and bolstered cardiomyocyte survival. Mechanistically, CHK1 attenuated oxidative stress and preserved mitochondrial metabolism in cardiomyocytes under I/R. Moreover, disrupted mitochondrial homeostasis in I/R myocardium was restored by CHK1 through the promotion of mitochondrial biogenesis and mitophagy. Through mass spectrometry analysis following co-immunoprecipitation, SIRT1 was identified as a direct target of CHK1. The 266–390 domain of CHK1 interacted with the 160–583 domain of SIRT1. Importantly, CHK1 phosphorylated SIRT1 at Thr530 residue, thereby inhibiting SMURF2-mediated degradation of SIRT1. The role of CHK1 in maintaining mitochondrial dynamics control and myocardial protection is abolished by SIRT1 inhibition, while inactivated mutation of SIRT1 Thr530 fails to reverse the impaired mitochondrial dynamics following CHK1 knockdown. CHK1 Δ390 amino acids (aa) mutant functioned similarly to full-length CHK1 in scavenging ROS and maintaining mitochondrial dynamics. Consistently, cardiac-specific SIRT1 knockdown attenuated the protective role of CHK1 in I/R injury.

Conclusions

Our findings revealed that CHK1 mitigates I/R injury and restores mitochondrial dynamics in cardiomyocytes through a SIRT1-dependent mechanism.
背景:线粒体功能障碍与心肌缺血再灌注(I/R)损伤有关。检查点激酶 1(CHK1)可促进心肌细胞增殖,但它在 I/R 损伤中对线粒体功能的作用仍不清楚:方法:为了研究 I/R 损伤后 CHK1 对线粒体功能的作用,我们建立了心肌细胞特异性基因敲除/表达小鼠模型。分离成年小鼠心肌细胞(AMCMs)进行体外研究。通过质谱-蛋白质组学分析和蛋白质共沉淀实验来揭示其分子机制:结果:CHK1在I/R后的心肌和氧-葡萄糖剥夺/再氧合(OGD/R)后的AMCMs中下调。在体内,CHK1 的过表达可防止 I/R 引起的心脏功能障碍,而异源 CHK1 敲除会加重心肌病。在体外,CHK1 可抑制 OGD/R 诱导的心肌细胞凋亡,提高心肌细胞存活率。从机理上讲,CHK1 可减轻氧化应激,保护 I/R 条件下心肌细胞的线粒体代谢。此外,CHK1 还通过促进线粒体生物生成和有丝分裂来恢复 I/R 心肌中被破坏的线粒体平衡。通过共免疫沉淀后的质谱分析,SIRT1 被确定为 CHK1 的直接靶标。CHK1 的 266-390 结构域与 SIRT1 的 160-583 结构域相互作用。重要的是,CHK1 在 Thr530 残基上磷酸化了 SIRT1,从而抑制了 SMURF2 介导的 SIRT1 降解。抑制 SIRT1 会取消 CHK1 在维持线粒体动力学控制和心肌保护方面的作用,而 SIRT1 Thr530 的失活突变无法逆转 CHK1 敲除后线粒体动力学受损的情况。CHK1 Δ390氨基酸(aa)突变体在清除ROS和维持线粒体动力学方面的功能与全长CHK1相似。同样,心脏特异性 SIRT1 基因敲除削弱了 CHK1 在 I/R 损伤中的保护作用:我们的研究结果表明,CHK1 可通过 SIRT1 依赖性机制减轻 I/R 损伤并恢复心肌细胞线粒体的活力。
{"title":"CHK1 attenuates cardiac dysfunction via suppressing SIRT1-ubiquitination","authors":"Tong-Tong Yang ,&nbsp;Liu-Hua Zhou ,&nbsp;Ling-Feng Gu ,&nbsp;Ling-Ling Qian ,&nbsp;Yu-Lin Bao ,&nbsp;Peng Jing ,&nbsp;Jia-Teng Sun ,&nbsp;Chong Du ,&nbsp;Tian-Kai Shan ,&nbsp;Si-Bo Wang ,&nbsp;Wen-Jing Wang ,&nbsp;Jia-Yi Chen ,&nbsp;Ze-Mu Wang ,&nbsp;Hao Wang ,&nbsp;Qi-Ming Wang ,&nbsp;Ru-Xing Wang ,&nbsp;Lian-Sheng Wang","doi":"10.1016/j.metabol.2024.156048","DOIUrl":"10.1016/j.metabol.2024.156048","url":null,"abstract":"<div><h3>Background</h3><div>Mitochondrial dysfunction is linked to myocardial ischemia-reperfusion (I/R) injury. Checkpoint kinase 1 (CHK1) could facilitate cardiomyocyte proliferation, however, its role on mitochondrial function in I/R injury remains unknown.</div></div><div><h3>Methods</h3><div>To investigate the role of CHK1 on mitochondrial function following I/R injury, cardiomyocyte-specific knockout/overexpression mouse models were generated. Adult mouse cardiomyocytes (AMCMs) were isolated for <em>in vitro</em> study. Mass spectrometry-proteomics analysis and protein co-immunoprecipitation assays were conducted to dissect the molecular mechanism.</div></div><div><h3>Results</h3><div>CHK1 was downregulated in myocardium post I/R and AMCMs post oxygen-glucose deprivation/re‑oxygenation (OGD/R). <em>In vivo</em>, CHK1 overexpression protected against I/R induced cardiac dysfunction, while heterogenous CHK1 knockout exacerbated cardiomyopathy. <em>In vitro</em>, CHK1 inhibited OGD/R-induced cardiomyocyte apoptosis and bolstered cardiomyocyte survival. Mechanistically, CHK1 attenuated oxidative stress and preserved mitochondrial metabolism in cardiomyocytes under I/R. Moreover, disrupted mitochondrial homeostasis in I/R myocardium was restored by CHK1 through the promotion of mitochondrial biogenesis and mitophagy. Through mass spectrometry analysis following co-immunoprecipitation, SIRT1 was identified as a direct target of CHK1. The 266–390 domain of CHK1 interacted with the 160–583 domain of SIRT1. Importantly, CHK1 phosphorylated SIRT1 at Thr530 residue, thereby inhibiting SMURF2-mediated degradation of SIRT1. The role of CHK1 in maintaining mitochondrial dynamics control and myocardial protection is abolished by SIRT1 inhibition, while inactivated mutation of SIRT1 Thr530 fails to reverse the impaired mitochondrial dynamics following CHK1 knockdown. CHK1 Δ390 amino acids (aa) mutant functioned similarly to full-length CHK1 in scavenging ROS and maintaining mitochondrial dynamics. Consistently, cardiac-specific SIRT1 knockdown attenuated the protective role of CHK1 in I/R injury.</div></div><div><h3>Conclusions</h3><div>Our findings revealed that CHK1 mitigates I/R injury and restores mitochondrial dynamics in cardiomyocytes through a SIRT1-dependent mechanism.</div></div>","PeriodicalId":18694,"journal":{"name":"Metabolism: clinical and experimental","volume":"162 ","pages":"Article 156048"},"PeriodicalIF":10.8,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142504210","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
ZBED3 exacerbates hyperglycemia by promoting hepatic gluconeogenesis through CREB signaling ZBED3 通过 CREB 信号促进肝脏葡萄糖生成,从而加剧高血糖。
IF 10.8 1区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-10-24 DOI: 10.1016/j.metabol.2024.156049
Yuan-yuan Luo , Chang-shun Ruan , Fu-zhen Zhao , Min Yang , Wei Cui , Xi Cheng , Xiao-he Luo , Xian-xiang Zhang , Cheng Zhang

Background

Elevated hepatic glucose production (HGP) is a prominent manifestation of impaired hepatic glucose metabolism in individuals with diabetes. Increased hepatic gluconeogenesis plays a pivotal role in the dysregulation of hepatic glucose metabolism and contributes significantly to fasting hyperglycemia in diabetes. Previous studies have identified zinc-finger BED domain-containing 3 (ZBED3) as a risk gene for type 2 diabetes (T2DM), and its single nucleotide polymorphism (SNPs) is closely associated with the fasting blood glucose level, suggesting a potential correlation between ZBED3 and the onset of diabetes. This study primarily explores the effect of ZBED3 on hepatic gluconeogenesis and analyzes the relevant signaling pathways that regulate hepatic gluconeogenesis.

Methods

The expression level of ZBED3 was assessed in the liver of insulin-resistant (IR)-related disease. RNA-seq and bioinformatics analyses were employed to examine the ZBED3-related pathway that modulated HGP. To investigate the role of ZBED3 in hepatic gluconeogenesis, the expression of ZBED3 was manipulated by upregulation or silencing using adeno-associated virus (AAV) in mouse primary hepatocytes (MPHs) and HHL-5 cells. In vivo, hepatocyte-specific ZBED3 knockout mice were generated. Moreover, AAV8 was employed to achieve hepatocyte-specific overexpression and knockdown of ZBED3 in C57BL/6 and db/db mice. Immunoprecipitation and mass spectrometry (IP-MS) analyses were employed to identify proteins that interacted with ZBED3. Co-immunoprecipitation (co-IP), glutathione S-transferase (GST) - pulldown, and dual-luciferase reporter assays were conducted to further elucidate the underlying mechanism of ZBED3 in regulating hepatic gluconeogenesis.

Results

The expression of ZBED3 in the liver of IR-related disease models was found to be increased. Under the stimulation of glucagon, ZBED3 promoted the expression of hepatic gluconeogenesis-related genes PGC1A, PCK1, G6PC, thereby increasing HGP. Consistently, the rate of hepatic gluconeogenesis was found to be elevated in mice with hepatocyte-specific overexpression of ZBED3 and decreased in those with ZBED3 knockout. Additionally, the knockdown of ZBED3 in the liver of db/db mice resulted in a reduction in hepatic gluconeogenesis. Moreover, the study revealed that ZBED3 facilitated the nuclear translocation of protein arginine methyltransferases 5 (PRMT5) to influence the regulation of PRMT5-mediated symmetrical dimethylation of arginine (s-DMA) of cyclic adenosine monophosphate (cAMP) response element binding protein (CREB), which in turn affects the phosphorylation of CREB and ultimately promotes HGP.

Conclusions

This study indicates that ZBED3 promotes hepatic gluconeogenesis and serves as a critical regulator of the progression of diabetes.
背景:肝糖生成(HGP)升高是糖尿病患者肝糖代谢受损的一个突出表现。肝糖生成增加在肝糖代谢失调中起着关键作用,是导致糖尿病患者空腹高血糖的重要原因。以往的研究发现,含锌指BED结构域3(ZBED3)是2型糖尿病(T2DM)的风险基因,其单核苷酸多态性(SNPs)与空腹血糖水平密切相关,提示ZBED3与糖尿病发病之间存在潜在的相关性。本研究主要探讨ZBED3对肝糖原生成的影响,并分析调控肝糖原生成的相关信号通路:方法:评估胰岛素抵抗(IR)相关疾病患者肝脏中ZBED3的表达水平。方法:评估 ZBED3 在胰岛素抵抗(IR)相关疾病的肝脏中的表达水平,采用 RNA-seq 和生物信息学分析来研究调节 HGP 的 ZBED3 相关通路。为了研究ZBED3在肝糖原生成中的作用,研究人员使用腺相关病毒(AAV)通过上调或沉默ZBED3在小鼠原代肝细胞(MPHs)和HHL-5细胞中的表达。在体内,产生了肝细胞特异性 ZBED3 基因敲除小鼠。此外,还利用 AAV8 在 C57BL/6 和 db/db 小鼠中实现了肝细胞特异性 ZBED3 的过表达和基因敲除。免疫沉淀和质谱分析(IP-MS)被用来鉴定与ZBED3相互作用的蛋白质。通过共免疫沉淀(co-IP)、谷胱甘肽 S-转移酶(GST)下拉和双荧光素酶报告实验进一步阐明了 ZBED3 调节肝糖原生成的内在机制:结果:发现ZBED3在红外相关疾病模型肝脏中的表达增加。在胰高血糖素的刺激下,ZBED3能促进肝糖生成相关基因PGC1A、PCK1、G6PC的表达,从而增加HGP。同样,研究发现肝细胞特异性过表达 ZBED3 的小鼠肝糖原生成率升高,而 ZBED3 基因敲除的小鼠肝糖原生成率降低。此外,在 db/db 小鼠肝脏中敲除 ZBED3 会导致肝糖生成减少。此外,研究还发现,ZBED3促进了蛋白精氨酸甲基转移酶5(PRMT5)的核转位,从而影响了PRMT5介导的环磷酸腺苷(cAMP)反应元件结合蛋白(CREB)的精氨酸对称二甲基化(s-DMA)的调控,进而影响了CREB的磷酸化,最终促进了HGP的发生:本研究表明,ZBED3 促进肝糖原生成,是糖尿病进展的关键调节因子。
{"title":"ZBED3 exacerbates hyperglycemia by promoting hepatic gluconeogenesis through CREB signaling","authors":"Yuan-yuan Luo ,&nbsp;Chang-shun Ruan ,&nbsp;Fu-zhen Zhao ,&nbsp;Min Yang ,&nbsp;Wei Cui ,&nbsp;Xi Cheng ,&nbsp;Xiao-he Luo ,&nbsp;Xian-xiang Zhang ,&nbsp;Cheng Zhang","doi":"10.1016/j.metabol.2024.156049","DOIUrl":"10.1016/j.metabol.2024.156049","url":null,"abstract":"<div><h3>Background</h3><div>Elevated hepatic glucose production (HGP) is a prominent manifestation of impaired hepatic glucose metabolism in individuals with diabetes. Increased hepatic gluconeogenesis plays a pivotal role in the dysregulation of hepatic glucose metabolism and contributes significantly to fasting hyperglycemia in diabetes. Previous studies have identified zinc-finger BED domain-containing 3 (<em>ZBED3</em>) as a risk gene for type 2 diabetes (T2DM), and its single nucleotide polymorphism (SNPs) is closely associated with the fasting blood glucose level, suggesting a potential correlation between ZBED3 and the onset of diabetes. This study primarily explores the effect of ZBED3 on hepatic gluconeogenesis and analyzes the relevant signaling pathways that regulate hepatic gluconeogenesis.</div></div><div><h3>Methods</h3><div>The expression level of ZBED3 was assessed in the liver of insulin-resistant (IR)-related disease. RNA-seq and bioinformatics analyses were employed to examine the ZBED3-related pathway that modulated HGP. To investigate the role of ZBED3 in hepatic gluconeogenesis, the expression of ZBED3 was manipulated by upregulation or silencing using adeno-associated virus (AAV) in mouse primary hepatocytes (MPHs) and HHL-5 cells. In vivo, hepatocyte-specific ZBED3 knockout mice were generated. Moreover, AAV8 was employed to achieve hepatocyte-specific overexpression and knockdown of ZBED3 in C57BL/6 and db/db mice. Immunoprecipitation and mass spectrometry (IP-MS) analyses were employed to identify proteins that interacted with ZBED3. Co-immunoprecipitation (co-IP), glutathione S-transferase (GST) - pulldown, and dual-luciferase reporter assays were conducted to further elucidate the underlying mechanism of ZBED3 in regulating hepatic gluconeogenesis.</div></div><div><h3>Results</h3><div>The expression of ZBED3 in the liver of IR-related disease models was found to be increased. Under the stimulation of glucagon, ZBED3 promoted the expression of hepatic gluconeogenesis-related genes <em>PGC1A</em>, <em>PCK1</em>, <em>G6PC</em>, thereby increasing HGP. Consistently, the rate of hepatic gluconeogenesis was found to be elevated in mice with hepatocyte-specific overexpression of ZBED3 and decreased in those with ZBED3 knockout. Additionally, the knockdown of ZBED3 in the liver of db/db mice resulted in a reduction in hepatic gluconeogenesis. Moreover, the study revealed that ZBED3 facilitated the nuclear translocation of protein arginine methyltransferases 5 (PRMT5) to influence the regulation of PRMT5-mediated symmetrical dimethylation of arginine (s-DMA) of cyclic adenosine monophosphate (cAMP) response element binding protein (CREB), which in turn affects the phosphorylation of CREB and ultimately promotes HGP.</div></div><div><h3>Conclusions</h3><div>This study indicates that ZBED3 promotes hepatic gluconeogenesis and serves as a critical regulator of the progression of diabetes.</div></div>","PeriodicalId":18694,"journal":{"name":"Metabolism: clinical and experimental","volume":"162 ","pages":"Article 156049"},"PeriodicalIF":10.8,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142504212","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
Impaired unsaturated fatty acid elongation alters mitochondrial function and accelerates metabolic dysfunction-associated steatohepatitis progression 不饱和脂肪酸伸长受损会改变线粒体功能,加速代谢功能障碍相关性脂肪性肝炎的进展。
IF 10.8 1区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-10-23 DOI: 10.1016/j.metabol.2024.156051
Adrien Vouilloz , Thibaut Bourgeois , Marc Diedisheim , Thomas Pilot , Antoine Jalil , Naig Le Guern , Victoria Bergas , Noéline Rohmer , Florence Castelli , Damien Leleu , Alexis Varin , Jean-Paul Pais de Barros , Pascal Degrace , Mickael Rialland , Camille Blériot , Nicolas Venteclef , Charles Thomas , David Masson

Background and aims

Although qualitative and quantitative alterations in liver Polyunsaturated Fatty Acids (PUFAs) are observed in MASH in humans, a causal relationship of PUFAs biosynthetic pathways is yet to be clarified. ELOVL5, an essential enzyme in PUFA elongation regulates hepatic triglyceride metabolism. Nonetheless, the long-term consequences of elongase disruption, particularly in murine models of MASH, have not been evaluated.

Approach & results

In humans, transcriptomic data indicated that PUFAs biosynthesis enzymes and notably ELOVL5 were induced during MASH progression. Moreover, gene module association determination revealed that ELOVL5 expression was associated with mitochondrial function in both humans and mice. WT and Elovl5-deficient mice were fed a high-fat, high-sucrose (HF/HS) diet for four months. Elovl5 deficiency led to limited systemic metabolic alterations but significant hepatic phenotype was observed in Elovl5−/− mice after the HF/HS diet, including hepatomegaly, pronounced macrovesicular and microvesicular steatosis, hepatocyte ballooning, immune cell infiltration, and fibrosis. Lipid analysis confirmed hepatic triglyceride accumulation and a reshaping of FA profile. Transcriptomic analysis indicated significant upregulation of genes involved in immune cell recruitment and fibrosis, and downregulation of genes involved in oxidative phosphorylation in Elovl5−/− mice. Alterations of FA oxidation and energy metabolism were confirmed by non-targeted metabolomic approach. Analysis of mitochondrial function in Elovl5−/− mice showed morphological alterations, qualitative cardiolipin changes with an enrichment in species containing shorter unsaturated FAs, and decreased activity of I and III respiratory chain complexes.

Conclusion

Enhanced susceptibility to diet-induced MASH and fibrosis in Elovl5−/− mice is intricately associated with disruptions in mitochondrial homeostasis, stemming from a profound reshaping of mitochondrial lipids, notably cardiolipins.
背景和目的:尽管在人类 MASH 中观察到肝脏多不饱和脂肪酸(PUFAs)的定性和定量改变,但多不饱和脂肪酸生物合成途径的因果关系尚未明确。ELOVL5 是 PUFA 延长过程中的一个重要酶,它调节肝脏甘油三酯的代谢。然而,人们尚未评估破坏伸长酶的长期后果,尤其是在小鼠 MASH 模型中:在人体中,转录组数据表明,在 MASH 进展过程中,PUFAs 生物合成酶,特别是 ELOVL5 被诱导。此外,基因模块关联测定显示,在人类和小鼠中,ELOVL5 的表达与线粒体功能有关。用高脂肪、高蔗糖(HF/HS)饮食喂养 WT 小鼠和 Elovl5 缺乏小鼠四个月。Elovl5 缺乏导致的全身代谢改变有限,但在 Elovl5-/- 小鼠中观察到高脂/高糖饮食后出现了显著的肝脏表型,包括肝肿大、明显的大泡和小泡脂肪变性、肝细胞气球化、免疫细胞浸润和纤维化。脂质分析证实了肝脏甘油三酯的积累和脂肪酸谱的重塑。转录组分析表明,在Elovl5-/-小鼠体内,参与免疫细胞招募和纤维化的基因明显上调,参与氧化磷酸化的基因下调。非靶向代谢组学方法证实了脂肪酸氧化和能量代谢的改变。对Elovl5-/-小鼠线粒体功能的分析表明,线粒体形态发生了改变,心磷脂发生了定性变化,含有较短不饱和脂肪酸的种类增多,I和III呼吸链复合物的活性降低:结论:Elovl5-/-小鼠对饮食诱导的MASH和纤维化的易感性增强与线粒体稳态的破坏密切相关,而线粒体脂质(尤其是心磷脂)的深刻变化则是线粒体稳态破坏的根源。
{"title":"Impaired unsaturated fatty acid elongation alters mitochondrial function and accelerates metabolic dysfunction-associated steatohepatitis progression","authors":"Adrien Vouilloz ,&nbsp;Thibaut Bourgeois ,&nbsp;Marc Diedisheim ,&nbsp;Thomas Pilot ,&nbsp;Antoine Jalil ,&nbsp;Naig Le Guern ,&nbsp;Victoria Bergas ,&nbsp;Noéline Rohmer ,&nbsp;Florence Castelli ,&nbsp;Damien Leleu ,&nbsp;Alexis Varin ,&nbsp;Jean-Paul Pais de Barros ,&nbsp;Pascal Degrace ,&nbsp;Mickael Rialland ,&nbsp;Camille Blériot ,&nbsp;Nicolas Venteclef ,&nbsp;Charles Thomas ,&nbsp;David Masson","doi":"10.1016/j.metabol.2024.156051","DOIUrl":"10.1016/j.metabol.2024.156051","url":null,"abstract":"<div><h3>Background and aims</h3><div>Although qualitative and quantitative alterations in liver Polyunsaturated Fatty Acids (PUFAs) are observed in MASH in humans, a causal relationship of PUFAs biosynthetic pathways is yet to be clarified. ELOVL5, an essential enzyme in PUFA elongation regulates hepatic triglyceride metabolism. Nonetheless, the long-term consequences of elongase disruption, particularly in murine models of MASH, have not been evaluated.</div></div><div><h3>Approach &amp; results</h3><div>In humans, transcriptomic data indicated that PUFAs biosynthesis enzymes and notably ELOVL5 were induced during MASH progression. Moreover, gene module association determination revealed that ELOVL5 expression was associated with mitochondrial function in both humans and mice. WT and <em>Elovl5</em>-deficient mice were fed a high-fat, high-sucrose (HF/HS) diet for four months. <em>Elovl5</em> deficiency led to limited systemic metabolic alterations but significant hepatic phenotype was observed in <em>Elovl5</em>−/− mice after the HF/HS diet, including hepatomegaly, pronounced macrovesicular and microvesicular steatosis, hepatocyte ballooning, immune cell infiltration, and fibrosis. Lipid analysis confirmed hepatic triglyceride accumulation and a reshaping of FA profile. Transcriptomic analysis indicated significant upregulation of genes involved in immune cell recruitment and fibrosis, and downregulation of genes involved in oxidative phosphorylation in <em>Elovl5</em>−/− mice. Alterations of FA oxidation and energy metabolism were confirmed by non-targeted metabolomic approach. Analysis of mitochondrial function in <em>Elovl5</em>−/− mice showed morphological alterations, qualitative cardiolipin changes with an enrichment in species containing shorter unsaturated FAs, and decreased activity of I and III respiratory chain complexes.</div></div><div><h3>Conclusion</h3><div>Enhanced susceptibility to diet-induced MASH and fibrosis in <em>Elovl5</em>−/− mice is intricately associated with disruptions in mitochondrial homeostasis, stemming from a profound reshaping of mitochondrial lipids, notably cardiolipins.</div></div>","PeriodicalId":18694,"journal":{"name":"Metabolism: clinical and experimental","volume":"162 ","pages":"Article 156051"},"PeriodicalIF":10.8,"publicationDate":"2024-10-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142504211","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
NPRC promotes hepatic steatosis via USP30-mediated deubiquitination of C/EBPβ NPRC 通过 USP30 介导的 C/EBPβ 去泛素化促进肝脏脂肪变性。
IF 10.8 1区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-10-19 DOI: 10.1016/j.metabol.2024.156050
Feng Jiang, Xinmiao Li, Lifan Lin, Mengyuan Li, Jianjian Zheng

Background and aims

Metabolic dysfunction-associated fatty liver disease (MAFLD) is a prevalent chronic liver condition characterised by dysregulated lipid metabolism. The role of Natriuretic Peptide Receptor C (NPRC), a receptor responsible for clearing natriuretic peptides, in MAFLD remains elusive. Therefore, the aim of the present study was to elucidate the role of NPRC in MAFLD progression.

Approach and results

This study demonstrated that NPRC enhanced lipid metabolism reprogramming and accelerated MAFLD progression. Mechanistic investigations, including proteomic and ubiquitination analyses, revealed that elevated NPRC levels stabilized the C/EBPβ protein, leading to excessive lipid accumulation. The DNA-binding domain (DBD) of C/EBPβ interacted with the deubiquitinase USP30, a key regulator that inhibited K149-specific K48-linked polyubiquitination of C/EBPβ. Importantly, the ANPR region of NPRC bound to USP30, facilitating the deubiquitination of C/EBPβ. Furthermore, virtual screening identified punicalin, a natural compound, as a potential inhibitor of NPRC expression, which may reduce hepatic lipid accumulation, inflammation and fibrosis.

Conclusions

Our findings indicate that NPRC recruits USP30 to mediate the deubiquitination of C/EBPβ, driving lipid metabolism reprogramming. Targeting NPRC could represent a promising therapeutic approach for MAFLD.
背景和目的:代谢功能障碍相关性脂肪肝(MAFLD)是一种普遍存在的慢性肝病,其特点是脂质代谢紊乱。钠尿肽受体 C(Natriuretic Peptide Receptor C,NPRC)是一种负责清除钠尿肽的受体,它在 MAFLD 中的作用仍然难以捉摸。因此,本研究旨在阐明 NPRC 在 MAFLD 进展中的作用:本研究表明,NPRC可促进脂质代谢重编程并加速MAFLD的进展。包括蛋白质组学和泛素化分析在内的机理研究表明,NPRC水平升高会稳定C/EBPβ蛋白,从而导致脂质过度积累。C/EBPβ的DNA结合域(DBD)与去泛素化酶USP30相互作用,后者是抑制C/EBPβ的K149特异性K48连锁多泛素化的关键调节因子。重要的是,NPRC 的 ANPR 区域与 USP30 结合,促进了 C/EBPβ 的去泛素化。此外,通过虚拟筛选发现,天然化合物 punicalin 是一种潜在的 NPRC 表达抑制剂,可减少肝脏脂质积累、炎症和纤维化:我们的研究结果表明,NPRC会招募USP30来介导C/EBPβ的去泛素化,从而推动脂质代谢的重编程。以 NPRC 为靶点可能是治疗 MAFLD 的一种有前景的方法。
{"title":"NPRC promotes hepatic steatosis via USP30-mediated deubiquitination of C/EBPβ","authors":"Feng Jiang,&nbsp;Xinmiao Li,&nbsp;Lifan Lin,&nbsp;Mengyuan Li,&nbsp;Jianjian Zheng","doi":"10.1016/j.metabol.2024.156050","DOIUrl":"10.1016/j.metabol.2024.156050","url":null,"abstract":"<div><h3>Background and aims</h3><div>Metabolic dysfunction-associated fatty liver disease (MAFLD) is a prevalent chronic liver condition characterised by dysregulated lipid metabolism. The role of Natriuretic Peptide Receptor C (NPRC), a receptor responsible for clearing natriuretic peptides, in MAFLD remains elusive. Therefore, the aim of the present study was to elucidate the role of NPRC in MAFLD progression.</div></div><div><h3>Approach and results</h3><div>This study demonstrated that NPRC enhanced lipid metabolism reprogramming and accelerated MAFLD progression. Mechanistic investigations, including proteomic and ubiquitination analyses, revealed that elevated NPRC levels stabilized the C/EBPβ protein, leading to excessive lipid accumulation. The DNA-binding domain (DBD) of C/EBPβ interacted with the deubiquitinase USP30, a key regulator that inhibited K149-specific K48-linked polyubiquitination of C/EBPβ. Importantly, the ANPR region of NPRC bound to USP30, facilitating the deubiquitination of C/EBPβ. Furthermore, virtual screening identified punicalin, a natural compound, as a potential inhibitor of NPRC expression, which may reduce hepatic lipid accumulation, inflammation and fibrosis.</div></div><div><h3>Conclusions</h3><div>Our findings indicate that NPRC recruits USP30 to mediate the deubiquitination of C/EBPβ, driving lipid metabolism reprogramming. Targeting NPRC could represent a promising therapeutic approach for MAFLD.</div></div>","PeriodicalId":18694,"journal":{"name":"Metabolism: clinical and experimental","volume":"162 ","pages":"Article 156050"},"PeriodicalIF":10.8,"publicationDate":"2024-10-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142470017","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
GDF-15 improves the predictive capacity of Steatotic liver disease non-invasive tests for incident morbidity and mortality risk for cardio-renal-metabolic diseases and malignancies. GDF-15 提高了脂肪肝无创检测对心肾代谢疾病和恶性肿瘤发病率和死亡率的预测能力。
IF 10.8 1区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-10-11 DOI: 10.1016/j.metabol.2024.156047
Michail Kokkorakis, Pytrik Folkertsma, José Castela Forte, Bruce H R Wolffenbuttel, Sipko van Dam, Christos S Mantzoros

Background & aims: Noninvasive tools (NITs) are currently used to stratify the risk of having or developing hepatic steatosis or fibrosis. Their performance and a proteomic-enabled improvement in forecasting long-term cardio-renal-metabolic morbidity, malignancies, as well as cause-specific and all-cause mortality, are lacking. Therefore, the performance of established NITs needs to be investigated in identifying cardio-renal-metabolic morbidity, malignancies, cause-specific and overall mortality and improve their performance with novel, proteomic-enabled NITs, including growth differentiation factor 15 (GDF-15), allowing multipurpose utilization.

Methods: 502,359 UK Biobank participants free of the study outcomes at baseline with a 14-year median follow-up were grouped into three categories: a) general population, b) potentially metabolic dysfunction-associated steatotic liver disease (MASLD) population, c) individuals with type 2 diabetes mellitus. The investigated NITs include Aspartate aminotransferase to Platelet Ratio Index (APRI), Fibrosis 4 Index (FIB-4), Fatty Liver Index (FLI), Hepatic Steatosis Index (HSI), Lipid Accumulation Product (LAP), and metabolic dysfunction-associated fibrosis (MAF-5) score.

Results: Adding GDF-15 to the existing NITs led to significantly increased prognostic performance compared to the traditional NITs in almost all instances, reaching substantially high C-indices, ranging between 0.601 and 0.808, with an overall >0.2 improvement in C-index. Overall, with the GDF-15 enhanced NITs, up to more than seven times fewer individuals need to be screened to identify more incident cases of adverse outcomes compared to the traditional NITs. The cumulative incidence of all outcomes, based on the continuous value percentiles of NITs, is increasing exponentially in the upper quintile of the GDF-15 enhanced NITs.

Conclusions: The herein-developed GDF-15 enhanced indices demonstrate higher screening effectiveness and significantly improved prognostic abilities, which are reduced to practice through an easy-to-use web-based calculator tool (https://clinicalpredictor.shinyapps.io/multimorbidity-mortality-risk/).

背景和目的:无创工具(NIT)目前用于对肝脏脂肪变性或肝纤维化的风险进行分层。这些工具在预测长期心肾代谢疾病、恶性肿瘤以及特定病因和全因死亡率方面的性能和蛋白质组学改进方面尚有欠缺。因此,需要研究已建立的 NITs 在识别心肾代谢疾病、恶性肿瘤、特定病因和全因死亡率方面的性能,并利用新型蛋白质组 NITs(包括生长分化因子 15 (GDF-15))提高其性能,从而实现多用途利用。方法:502,359 名英国生物库参与者在基线时未出现研究结果,中位随访期为 14 年,他们被分为三类:a)普通人群;b)潜在的代谢功能障碍相关性脂肪性肝病(MASLD)人群;c)2 型糖尿病患者。研究的NIT包括天冬氨酸氨基转移酶与血小板比率指数(APRI)、纤维化4指数(FIB-4)、脂肪肝指数(FLI)、肝脏脂肪变性指数(HSI)、脂质累积产物(LAP)和代谢功能障碍相关纤维化(MAF-5)评分:与传统的NIT相比,在现有的NIT中添加GDF-15几乎在所有情况下都能显著提高预后效果,C指数大幅提高,在0.601到0.808之间,C指数总体提高了0.2以上。总体而言,与传统的 NIT 相比,GDF-15 增强型 NIT 所需的筛查人数要少七倍以上,才能发现更多的不良后果事件。根据 NIT 的连续值百分位数,所有结果的累积发生率在 GDF-15 增强型 NIT 的上五分位数呈指数增长:本文开发的 GDF-15 增强指数显示出更高的筛查效果和显著改善的预后能力,并通过一个易于使用的网络计算工具 (https://clinicalpredictor.shinyapps.io/multimorbidity-mortality-risk/) 将其应用到实践中。
{"title":"GDF-15 improves the predictive capacity of Steatotic liver disease non-invasive tests for incident morbidity and mortality risk for cardio-renal-metabolic diseases and malignancies.","authors":"Michail Kokkorakis, Pytrik Folkertsma, José Castela Forte, Bruce H R Wolffenbuttel, Sipko van Dam, Christos S Mantzoros","doi":"10.1016/j.metabol.2024.156047","DOIUrl":"https://doi.org/10.1016/j.metabol.2024.156047","url":null,"abstract":"<p><strong>Background & aims: </strong>Noninvasive tools (NITs) are currently used to stratify the risk of having or developing hepatic steatosis or fibrosis. Their performance and a proteomic-enabled improvement in forecasting long-term cardio-renal-metabolic morbidity, malignancies, as well as cause-specific and all-cause mortality, are lacking. Therefore, the performance of established NITs needs to be investigated in identifying cardio-renal-metabolic morbidity, malignancies, cause-specific and overall mortality and improve their performance with novel, proteomic-enabled NITs, including growth differentiation factor 15 (GDF-15), allowing multipurpose utilization.</p><p><strong>Methods: </strong>502,359 UK Biobank participants free of the study outcomes at baseline with a 14-year median follow-up were grouped into three categories: a) general population, b) potentially metabolic dysfunction-associated steatotic liver disease (MASLD) population, c) individuals with type 2 diabetes mellitus. The investigated NITs include Aspartate aminotransferase to Platelet Ratio Index (APRI), Fibrosis 4 Index (FIB-4), Fatty Liver Index (FLI), Hepatic Steatosis Index (HSI), Lipid Accumulation Product (LAP), and metabolic dysfunction-associated fibrosis (MAF-5) score.</p><p><strong>Results: </strong>Adding GDF-15 to the existing NITs led to significantly increased prognostic performance compared to the traditional NITs in almost all instances, reaching substantially high C-indices, ranging between 0.601 and 0.808, with an overall >0.2 improvement in C-index. Overall, with the GDF-15 enhanced NITs, up to more than seven times fewer individuals need to be screened to identify more incident cases of adverse outcomes compared to the traditional NITs. The cumulative incidence of all outcomes, based on the continuous value percentiles of NITs, is increasing exponentially in the upper quintile of the GDF-15 enhanced NITs.</p><p><strong>Conclusions: </strong>The herein-developed GDF-15 enhanced indices demonstrate higher screening effectiveness and significantly improved prognostic abilities, which are reduced to practice through an easy-to-use web-based calculator tool (https://clinicalpredictor.shinyapps.io/multimorbidity-mortality-risk/).</p>","PeriodicalId":18694,"journal":{"name":"Metabolism: clinical and experimental","volume":" ","pages":"156047"},"PeriodicalIF":10.8,"publicationDate":"2024-10-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142470016","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
A partial loss-of-function variant (Ile191Val) of the TAS1R2 glucose receptor is associated with enhanced responses to exercise training in older adults with obesity: A translational study. TAS1R2葡萄糖受体的部分功能缺失变体(Ile191Val)与肥胖症老年人对运动训练的反应增强有关:一项转化研究。
IF 10.8 1区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-10-10 DOI: 10.1016/j.metabol.2024.156045
Joan Serrano, Saki Kondo, Grace M Link, Ian S Brown, Richard E Pratley, Kedryn K Baskin, Bret H Goodpaster, Paul M Coen, George A Kyriazis

Background: The TAS1R2 receptor, known for its role in taste perception, has also emerged as a key regulator of muscle physiology. Previous studies have shown that genetic ablation of TAS1R2 in mice enhances muscle fitness mimicking responses to endurance exercise training. However, the translational relevance of these findings to humans remains uncertain.

Methods: We explored responses to endurance exercise training in mice and humans with genetic deficiency of TAS1R2. First, we assessed the effects of muscle-specific deletion of TAS1R2 in mice (mKO) or wild type controls (mWT) following 4 weeks of voluntary wheel running (VWR). Next, we investigated the effects of the TAS1R2-Ile191Val (rs35874116) partial loss-of-function variant on responses to a 6-month diet-induced weight loss with exercise training (WLEX), weight loss alone (WL), or education control (CON) interventions in older individuals with obesity. Participants were retrospectively genotyped for the TAS1R2-Ile191Val polymorphism and classified as conventional function (Ile/Ile) or partial loss-of-function (Val carriers: Ile/Val and Val/Val). Body composition, cardiorespiratory fitness, and skeletal muscle mitochondrial function were assessed before and after the intervention.

Results: In response to VWR, mKO mice demonstrated enhanced running endurance and mitochondrial protein content. Similarly, TAS1R2 Val carriers exhibited distinctive improvements in body composition, including increased muscle mass, along with enhanced cardiorespiratory fitness and mitochondrial function in skeletal muscle following the WLEX intervention compared to Ile/Ile counterparts. Notably, every Val carrier demonstrated substantial responses to exercise training and weight loss, surpassing all Ile/Ile participants in overall performance metrics.

Conclusions: Our findings suggest that TAS1R2 partial loss-of-function confers beneficial effects on muscle function and metabolism in humans in response to exercise training, akin to observations in TAS1R2 muscle-deficient mice. Targeting TAS1R2 may help enhancing exercise training adaptations in individuals with compromised exercise tolerance or metabolic disorders, presenting a potential avenue for personalized exercise interventions.

背景:TAS1R2 受体因其在味觉感知中的作用而闻名,它也已成为肌肉生理的一个关键调节因子。先前的研究表明,在小鼠体内对 TAS1R2 进行基因消减,可增强肌肉体能,模拟对耐力运动训练的反应。然而,这些发现对人类的转化意义仍不确定:方法:我们研究了基因缺乏 TAS1R2 的小鼠和人类对耐力运动训练的反应。首先,我们评估了肌肉特异性缺失 TAS1R2 的小鼠(mKO)或野生型对照组(mWT)在 4 周自愿轮跑(VWR)后的影响。接下来,我们研究了 TAS1R2-Ile191Val (rs35874116) 部分功能缺失变异对老年肥胖症患者进行为期 6 个月的饮食诱导减肥与运动训练 (WLEX)、单纯减肥 (WL) 或教育控制 (CON) 干预的影响。研究人员对参与者进行了TAS1R2-Ile191Val多态性基因分型,并将其分为传统功能型(Ile/Ile)或部分功能缺失型(Val携带者:Ile/Val和Val/Val)。干预前后对身体成分、心肺功能和骨骼肌线粒体功能进行了评估:结果:针对 VWR,mKO 小鼠的跑步耐力和线粒体蛋白含量均有所提高。同样,TAS1R2 Val 基因携带者与 Ile/Ile 基因携带者相比,在接受 WLEX 干预后,身体成分有明显改善,包括肌肉质量增加,心肺功能和骨骼肌线粒体功能增强。值得注意的是,每个 Val 基因携带者都对运动训练和体重减轻表现出实质性的反应,在总体表现指标上超过了所有 Ile/Ile 参与者:我们的研究结果表明,TAS1R2部分功能缺失会对人类的肌肉功能和新陈代谢产生有益影响,从而对运动训练做出反应,这与在TAS1R2肌肉缺陷小鼠身上观察到的结果类似。以TAS1R2为靶标可能有助于增强运动耐受性受损或代谢紊乱个体的运动训练适应性,为个性化运动干预提供了潜在的途径。
{"title":"A partial loss-of-function variant (Ile191Val) of the TAS1R2 glucose receptor is associated with enhanced responses to exercise training in older adults with obesity: A translational study.","authors":"Joan Serrano, Saki Kondo, Grace M Link, Ian S Brown, Richard E Pratley, Kedryn K Baskin, Bret H Goodpaster, Paul M Coen, George A Kyriazis","doi":"10.1016/j.metabol.2024.156045","DOIUrl":"10.1016/j.metabol.2024.156045","url":null,"abstract":"<p><strong>Background: </strong>The TAS1R2 receptor, known for its role in taste perception, has also emerged as a key regulator of muscle physiology. Previous studies have shown that genetic ablation of TAS1R2 in mice enhances muscle fitness mimicking responses to endurance exercise training. However, the translational relevance of these findings to humans remains uncertain.</p><p><strong>Methods: </strong>We explored responses to endurance exercise training in mice and humans with genetic deficiency of TAS1R2. First, we assessed the effects of muscle-specific deletion of TAS1R2 in mice (mKO) or wild type controls (mWT) following 4 weeks of voluntary wheel running (VWR). Next, we investigated the effects of the TAS1R2<sup>-Ile191Val</sup> (rs35874116) partial loss-of-function variant on responses to a 6-month diet-induced weight loss with exercise training (WLEX), weight loss alone (WL), or education control (CON) interventions in older individuals with obesity. Participants were retrospectively genotyped for the TAS1R2<sup>-Ile191Val</sup> polymorphism and classified as conventional function (Ile/Ile) or partial loss-of-function (Val carriers: Ile/Val and Val/Val). Body composition, cardiorespiratory fitness, and skeletal muscle mitochondrial function were assessed before and after the intervention.</p><p><strong>Results: </strong>In response to VWR, mKO mice demonstrated enhanced running endurance and mitochondrial protein content. Similarly, TAS1R2 Val carriers exhibited distinctive improvements in body composition, including increased muscle mass, along with enhanced cardiorespiratory fitness and mitochondrial function in skeletal muscle following the WLEX intervention compared to Ile/Ile counterparts. Notably, every Val carrier demonstrated substantial responses to exercise training and weight loss, surpassing all Ile/Ile participants in overall performance metrics.</p><p><strong>Conclusions: </strong>Our findings suggest that TAS1R2 partial loss-of-function confers beneficial effects on muscle function and metabolism in humans in response to exercise training, akin to observations in TAS1R2 muscle-deficient mice. Targeting TAS1R2 may help enhancing exercise training adaptations in individuals with compromised exercise tolerance or metabolic disorders, presenting a potential avenue for personalized exercise interventions.</p>","PeriodicalId":18694,"journal":{"name":"Metabolism: clinical and experimental","volume":" ","pages":"156045"},"PeriodicalIF":10.8,"publicationDate":"2024-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142406618","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
Central NUCB2/nesfatin-1 signaling ameliorates liver steatosis through suppression of endoplasmic reticulum stress in the hypothalamus. 中枢 NUCB2/nesfatin-1 信号通过抑制下丘脑内质网应激改善肝脏脂肪变性。
IF 10.8 1区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-10-09 DOI: 10.1016/j.metabol.2024.156046
Yirui He, Cheng Zhang, Shaobo Wu, Ke Li, Siliang Zhang, Mingyuan Tian, Chen Chen, Dongfang Liu, Gangyi Yang, Ling Li, Mengliu Yang

Background & aims: Nucleobindin-2 (NUCB2)/nesfatin-1, a signal with recognized anorexigenic and insulin-sensitizing properties in peripheral tissues, is expressed within the hypothalamus. However, the potential involvement of central nesfatin-1 signaling in the pathophysiology of hepatic steatosis remains unknown. This study aimed to determine whether and how central NUCB2/nesfatin-1 plays a role in liver steatosis.

Methods: We generated Nucb2 knockout (Nucb2-/-) rats and administered continuous intracerebroventricular (ICV) nesfatin-1 infusion, while observing its effect on liver steatosis. The molecular mechanism of action of nesfatin-1 was elucidated via proteomics, phosphoproteomics and molecular biology methods.

Results: Herein, we present compelling evidence indicating diminished NUCB2 expression in the hypothalamus of obese rodents. We demonstrated that chronic ICV infusion of nesfatin-1 mitigated both diet-induced obesity and liver steatosis in high-fat diet (HFD)-fed Nucb2-/- rats by regulating hypothalamic endoplasmic reticulum (ER) stress and Akt phosphorylation. Furthermore, we revealed that the increase in hypothalamic insulin resistance (IR) and ER stress induced by tunicamycin infusion or Ero1α overexpression exacerbated hepatic steatosis and offset the favorable influence of central nesfatin-1 on hepatic steatosis. The metabolic action of central nesfatin-1 is contingent upon vagal nerve transmission to the liver. Mechanistically, nesfatin-1 impedes ER stress and interacts with Ero1α to repress its Ser106 phosphorylation. This leads to the enhancement of Akt activity in the hypothalamus, culminating in the inhibition of hepatic lipogenesis.

Conclusions: These findings underscore the importance of hypothalamic NUCB2/nesfatin-1 as a key mediator in the top-down neural mechanism that combats diet-induced liver steatosis.

背景和目的:核吲哚-2(NUCB2)/nesfatin-1是一种在外周组织中具有公认厌食和胰岛素敏感特性的信号,在下丘脑中也有表达。然而,中枢内司蛋白-1 信号在肝脏脂肪变性的病理生理学中的潜在参与仍然未知。本研究旨在确定中枢 NUCB2/nesfatin-1 是否以及如何在肝脏脂肪变性中发挥作用:方法:我们培育了Nucb2基因敲除(Nucb2-/-)大鼠,并连续脑室内注射(ICV)nesfatin-1,同时观察其对肝脏脂肪变性的影响。通过蛋白质组学、磷酸化蛋白质组学和分子生物学方法阐明了内司法亭-1的分子作用机制:在此,我们提出了令人信服的证据,表明肥胖啮齿动物下丘脑中的 NUCB2 表达减少。我们证明,通过调节下丘脑内质网(ER)应激和Akt磷酸化,长期ICV输注nesfatin-1可减轻高脂饮食(HFD)喂养的Nucb2-/-大鼠饮食诱发的肥胖和肝脏脂肪变性。此外,我们还发现,输注曲安奈德霉素或Ero1α过表达诱导的下丘脑胰岛素抵抗(IR)和ER应激的增加加剧了肝脂肪变性,并抵消了中枢nesfatin-1对肝脂肪变性的有利影响。中枢nesfatin-1的代谢作用取决于迷走神经对肝脏的传导。从机理上讲,nesfatin-1 会阻碍 ER 应激,并与 Ero1α 相互作用,抑制其 Ser106 磷酸化。这导致下丘脑中Akt活性增强,最终抑制了肝脏脂肪生成:这些发现强调了下丘脑NUCB2/nesfatin-1作为自上而下的神经机制中的一个关键介质的重要性,该机制可对抗饮食诱导的肝脏脂肪变性。
{"title":"Central NUCB2/nesfatin-1 signaling ameliorates liver steatosis through suppression of endoplasmic reticulum stress in the hypothalamus.","authors":"Yirui He, Cheng Zhang, Shaobo Wu, Ke Li, Siliang Zhang, Mingyuan Tian, Chen Chen, Dongfang Liu, Gangyi Yang, Ling Li, Mengliu Yang","doi":"10.1016/j.metabol.2024.156046","DOIUrl":"10.1016/j.metabol.2024.156046","url":null,"abstract":"<p><strong>Background & aims: </strong>Nucleobindin-2 (NUCB2)/nesfatin-1, a signal with recognized anorexigenic and insulin-sensitizing properties in peripheral tissues, is expressed within the hypothalamus. However, the potential involvement of central nesfatin-1 signaling in the pathophysiology of hepatic steatosis remains unknown. This study aimed to determine whether and how central NUCB2/nesfatin-1 plays a role in liver steatosis.</p><p><strong>Methods: </strong>We generated Nucb2 knockout (Nucb2<sup>-/-</sup>) rats and administered continuous intracerebroventricular (ICV) nesfatin-1 infusion, while observing its effect on liver steatosis. The molecular mechanism of action of nesfatin-1 was elucidated via proteomics, phosphoproteomics and molecular biology methods.</p><p><strong>Results: </strong>Herein, we present compelling evidence indicating diminished NUCB2 expression in the hypothalamus of obese rodents. We demonstrated that chronic ICV infusion of nesfatin-1 mitigated both diet-induced obesity and liver steatosis in high-fat diet (HFD)-fed Nucb2<sup>-/-</sup> rats by regulating hypothalamic endoplasmic reticulum (ER) stress and Akt phosphorylation. Furthermore, we revealed that the increase in hypothalamic insulin resistance (IR) and ER stress induced by tunicamycin infusion or Ero1α overexpression exacerbated hepatic steatosis and offset the favorable influence of central nesfatin-1 on hepatic steatosis. The metabolic action of central nesfatin-1 is contingent upon vagal nerve transmission to the liver. Mechanistically, nesfatin-1 impedes ER stress and interacts with Ero1α to repress its Ser106 phosphorylation. This leads to the enhancement of Akt activity in the hypothalamus, culminating in the inhibition of hepatic lipogenesis.</p><p><strong>Conclusions: </strong>These findings underscore the importance of hypothalamic NUCB2/nesfatin-1 as a key mediator in the top-down neural mechanism that combats diet-induced liver steatosis.</p>","PeriodicalId":18694,"journal":{"name":"Metabolism: clinical and experimental","volume":" ","pages":"156046"},"PeriodicalIF":10.8,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142400753","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
Corrigendum to “Increased adrenal steroidogenesis and suppressed corticosteroid responsiveness in critical COVID-19” [Metabolism volume 160 (2024) 155980] 对 "临界 COVID-19 中肾上腺类固醇生成增加和皮质类固醇反应受抑制 "的更正[《新陈代谢》第 160 卷(2024 年)第 155980 期]。
IF 10.8 1区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-10-04 DOI: 10.1016/j.metabol.2024.156033
Tian-Zi Wen , Tian-Ran Li , Xin-Yu Chen , He-Yuan Chen , Shuai Wang , Wen-Juan Fu , Shi-Qi Xiao , Jie Luo , Jia-Feng Huang , Rui Tang , Zhi-Cheng He , Tao Luo , Hong-Liang Zhao , Cong Chen , Jing-Ya Miao , Qin Niu , Yan Wang , Xiu-Wu Bian , Xiao-Hong Yao
{"title":"Corrigendum to “Increased adrenal steroidogenesis and suppressed corticosteroid responsiveness in critical COVID-19” [Metabolism volume 160 (2024) 155980]","authors":"Tian-Zi Wen ,&nbsp;Tian-Ran Li ,&nbsp;Xin-Yu Chen ,&nbsp;He-Yuan Chen ,&nbsp;Shuai Wang ,&nbsp;Wen-Juan Fu ,&nbsp;Shi-Qi Xiao ,&nbsp;Jie Luo ,&nbsp;Jia-Feng Huang ,&nbsp;Rui Tang ,&nbsp;Zhi-Cheng He ,&nbsp;Tao Luo ,&nbsp;Hong-Liang Zhao ,&nbsp;Cong Chen ,&nbsp;Jing-Ya Miao ,&nbsp;Qin Niu ,&nbsp;Yan Wang ,&nbsp;Xiu-Wu Bian ,&nbsp;Xiao-Hong Yao","doi":"10.1016/j.metabol.2024.156033","DOIUrl":"10.1016/j.metabol.2024.156033","url":null,"abstract":"","PeriodicalId":18694,"journal":{"name":"Metabolism: clinical and experimental","volume":"161 ","pages":"Article 156033"},"PeriodicalIF":10.8,"publicationDate":"2024-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142378097","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 ACSS2 triggers glycolysis inhibition and nuclear translocation to activate SIRT1/ATG5/ATG2B deacetylation axis, promoting autophagy and reducing malignancy and chemoresistance in ovarian cancer. 抑制 ACSS2 会引发糖酵解抑制和核转运,从而激活 SIRT1/ATG5/ATG2B 去乙酰化轴,促进自噬,降低卵巢癌的恶性程度和化疗耐药性。
IF 10.8 1区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-10-02 DOI: 10.1016/j.metabol.2024.156041
Jiang Yang, Haoyu Wang, Bingshu Li, Jingchun Liu, Xiaoyi Zhang, Ying Wang, Jiaxin Peng, Likun Gao, Xinqi Wang, Siyuan Hu, Wenyi Zhang, Li Hong
<p><strong>Background: </strong>Metabolic reprogramming is a hallmark of cancer, characterized by a high dependence on glycolysis and an enhanced utilization of acetate as an alternative carbon source. ACSS2 is a critical regulator of acetate metabolism, playing a significant role in the development and progression of various malignancies. ACSS2 facilitates the conversion of acetate to acetyl-CoA, which participates in multiple metabolic pathways and functions as an epigenetic regulator of protein acetylation, thereby modulating key cellular processes such as autophagy. However, the roles and intrinsic connections of ACSS2, glycolysis, protein acetylation, and autophagy in ovarian cancer (OC) remain to be elucidated.</p><p><strong>Basic procedures: </strong>Utilizing clinical specimens and online databases, we analysed the expression of ACSS2 in OC and its relationship with clinical prognosis. By knocking down ACSS2, we evaluated its effects on the malignant phenotype, acetate metabolism, glycolysis, and autophagy. The metabolic alterations in OC cells were comprehensively analysed using Seahorse assays, transmission electron microscopy, membrane potential measurements, and stable-isotope labeling techniques. CUT&TAG and co-immunoprecipitation techniques were employed to explore the deacetylation of autophagy-related proteins mediated by ACSS2 via SIRT1. Additionally, through molecular docking, transcriptome sequencing, and metabolomics analyses, we validated the pharmacological effects of paeonol on ACSS2 and the glycolytic process in OC cells. Finally, both in vitro and in vivo experiments were performed to investigate the impact of paeonol on autophagy and its anti-OC effects mediated through the ACSS2/SIRT1 deacetylation axis.</p><p><strong>Main findings: </strong>ACSS2 is significantly upregulated in OC and is associated with poor prognosis. Knockdown of ACSS2 inhibits OC cells proliferation, migration, invasion, angiogenesis, and platinum resistance, while reducing tumour burden in vivo. Mechanistically, inhibiting ACSS2 reduces acetate metabolism and suppresses glycolysis by targeting HXK2. This glycolytic reduction promotes the translocation of ACSS2 from the cytoplasm to the nucleus, leading to increased expression of the deacetylase SIRT1. SIRT1 mediates the deacetylation of autophagy-related proteins, such as ATG5 and ATG2B, thereby significantly activating autophagy in OC cells and exerting antitumor effects. Paeonol inhibits acetate metabolism and glycolysis in OC cells by targeting ACSS2. Paeonol activates autophagy through the ACSS2/SIRT1/ATG5/ATG2B deacetylation axis, demonstrating inhibition of OC in vitro and in vivo.</p><p><strong>Principal conclusions: </strong>Pae can serve as an effective, low-toxicity, multi-targeted drug targeting ACSS2 and glycolysis. It activates autophagy through the ACSS2/SIRT1/ATG5/ATG2B deacetylation signalling cascade, thereby exerting anti-OC effects. Our study provides new insights into the malign
背景:代谢重编程是癌症的一个特征,其特点是高度依赖糖酵解,并加强利用醋酸盐作为替代碳源。ACSS2 是乙酸代谢的关键调节因子,在各种恶性肿瘤的发生和发展过程中发挥着重要作用。ACSS2 促进乙酸酯向乙酰-CoA 的转化,乙酰-CoA 参与多种代谢途径,并充当蛋白质乙酰化的表观遗传调节因子,从而调节自噬等关键细胞过程。然而,ACSS2、糖酵解、蛋白质乙酰化和自噬在卵巢癌(OC)中的作用和内在联系仍有待阐明:利用临床标本和在线数据库,我们分析了ACSS2在卵巢癌中的表达及其与临床预后的关系。通过敲除 ACSS2,我们评估了其对恶性表型、醋酸代谢、糖酵解和自噬的影响。我们使用海马测定法、透射电子显微镜、膜电位测量和稳定同位素标记技术全面分析了OC细胞的代谢变化。利用CUT&TAG和共免疫沉淀技术探讨了ACSS2通过SIRT1介导的自噬相关蛋白的去乙酰化。此外,通过分子对接、转录组测序和代谢组学分析,我们验证了芍药酚对 ACSS2 和 OC 细胞糖酵解过程的药理作用。最后,我们进行了体外和体内实验,研究芍药酚对自噬的影响及其通过 ACSS2/SIRT1 去乙酰化轴介导的抗 OC 作用:主要发现:ACSS2在OC中明显上调,并与预后不良有关。敲除 ACSS2 可抑制 OC 细胞的增殖、迁移、侵袭、血管生成和铂类抗性,同时减轻体内肿瘤负荷。从机理上讲,抑制 ACSS2 可减少醋酸代谢,并通过靶向 HXK2 抑制糖酵解。糖酵解的减少促进 ACSS2 从细胞质转位到细胞核,导致去乙酰化酶 SIRT1 的表达增加。SIRT1 可介导 ATG5 和 ATG2B 等自噬相关蛋白的去乙酰化,从而显著激活 OC 细胞的自噬,发挥抗肿瘤作用。芍药酚通过靶向 ACSS2 抑制 OC 细胞的乙酸代谢和糖酵解。芍药酚通过ACSS2/SIRT1/ATG5/ATG2B去乙酰化轴激活自噬,对体外和体内的OC均有抑制作用:主要结论:Pae 可作为一种有效、低毒、多靶点的药物,靶向 ACSS2 和糖酵解。它通过 ACSS2/SIRT1/ATG5/ATG2B 去乙酰化信号级联激活自噬,从而发挥抗肿瘤作用。我们的研究为了解 OC 的恶性机制提供了新的视角,并为其治疗提供了新的策略。
{"title":"Inhibition of ACSS2 triggers glycolysis inhibition and nuclear translocation to activate SIRT1/ATG5/ATG2B deacetylation axis, promoting autophagy and reducing malignancy and chemoresistance in ovarian cancer.","authors":"Jiang Yang, Haoyu Wang, Bingshu Li, Jingchun Liu, Xiaoyi Zhang, Ying Wang, Jiaxin Peng, Likun Gao, Xinqi Wang, Siyuan Hu, Wenyi Zhang, Li Hong","doi":"10.1016/j.metabol.2024.156041","DOIUrl":"10.1016/j.metabol.2024.156041","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Background: &lt;/strong&gt;Metabolic reprogramming is a hallmark of cancer, characterized by a high dependence on glycolysis and an enhanced utilization of acetate as an alternative carbon source. ACSS2 is a critical regulator of acetate metabolism, playing a significant role in the development and progression of various malignancies. ACSS2 facilitates the conversion of acetate to acetyl-CoA, which participates in multiple metabolic pathways and functions as an epigenetic regulator of protein acetylation, thereby modulating key cellular processes such as autophagy. However, the roles and intrinsic connections of ACSS2, glycolysis, protein acetylation, and autophagy in ovarian cancer (OC) remain to be elucidated.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Basic procedures: &lt;/strong&gt;Utilizing clinical specimens and online databases, we analysed the expression of ACSS2 in OC and its relationship with clinical prognosis. By knocking down ACSS2, we evaluated its effects on the malignant phenotype, acetate metabolism, glycolysis, and autophagy. The metabolic alterations in OC cells were comprehensively analysed using Seahorse assays, transmission electron microscopy, membrane potential measurements, and stable-isotope labeling techniques. CUT&TAG and co-immunoprecipitation techniques were employed to explore the deacetylation of autophagy-related proteins mediated by ACSS2 via SIRT1. Additionally, through molecular docking, transcriptome sequencing, and metabolomics analyses, we validated the pharmacological effects of paeonol on ACSS2 and the glycolytic process in OC cells. Finally, both in vitro and in vivo experiments were performed to investigate the impact of paeonol on autophagy and its anti-OC effects mediated through the ACSS2/SIRT1 deacetylation axis.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Main findings: &lt;/strong&gt;ACSS2 is significantly upregulated in OC and is associated with poor prognosis. Knockdown of ACSS2 inhibits OC cells proliferation, migration, invasion, angiogenesis, and platinum resistance, while reducing tumour burden in vivo. Mechanistically, inhibiting ACSS2 reduces acetate metabolism and suppresses glycolysis by targeting HXK2. This glycolytic reduction promotes the translocation of ACSS2 from the cytoplasm to the nucleus, leading to increased expression of the deacetylase SIRT1. SIRT1 mediates the deacetylation of autophagy-related proteins, such as ATG5 and ATG2B, thereby significantly activating autophagy in OC cells and exerting antitumor effects. Paeonol inhibits acetate metabolism and glycolysis in OC cells by targeting ACSS2. Paeonol activates autophagy through the ACSS2/SIRT1/ATG5/ATG2B deacetylation axis, demonstrating inhibition of OC in vitro and in vivo.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Principal conclusions: &lt;/strong&gt;Pae can serve as an effective, low-toxicity, multi-targeted drug targeting ACSS2 and glycolysis. It activates autophagy through the ACSS2/SIRT1/ATG5/ATG2B deacetylation signalling cascade, thereby exerting anti-OC effects. Our study provides new insights into the malign","PeriodicalId":18694,"journal":{"name":"Metabolism: clinical and experimental","volume":" ","pages":"156041"},"PeriodicalIF":10.8,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142372342","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
Fam3a-mediated prohormone convertase switch in α-cells regulates pancreatic GLP-1 production in an Nr4a2-Foxa2-dependent manner. α细胞中 Fam3a 介导的原激素转换酶转换以 Nr4a2-Foxa2 依赖性方式调节胰腺 GLP-1 的产生。
IF 10.8 1区 医学 Q1 ENDOCRINOLOGY & METABOLISM Pub Date : 2024-10-01 DOI: 10.1016/j.metabol.2024.156042
Dandan Wang, Tianjiao Wei, Xiaona Cui, Li Xia, Yafei Jiang, Deshan Yin, Xinyue Liao, Fei Li, Jian Li, Qi Wu, Xiafang Lin, Shan Lang, Yunyi Le, Jichun Yang, Jin Yang, Rui Wei, Tianpei Hong
<p><strong>Background: </strong>Fam3a has been demonstrated to regulate pancreatic β-cell function and glucose homeostasis. However, the role and mechanism of Fam3a in regulating α-cell function remain unexplored.</p><p><strong>Methods: </strong>Glucagon and glucagon-like peptide-1 (GLP-1) levels in pancreas and plasma were measured in global Fam3a knockout (Fam3a<sup>-/-</sup>) mice. Human islet single-cell RNA sequencing (scRNA-seq) datasets were utilized to analyze gene expression correlations between FAM3A and PCSK1 (encoding PC1/3, which processes proglucagon into GLP-1). Mouse pancreatic α-cell line αTC1.9 cells were transfected with Fam3a siRNA or plasmid for Fam3a knockdown or overexpression to explore the effects of Fam3a on PC1/3 expression and GLP-1 production. The downstream mediator (including Nr4a2) was identified by transcriptomic analysis, and its role was confirmed by Fam3a knockdown or overexpression in αTC1.9 cells. Based on the interacted protein of Nr4a2 and the direct binding to Pcsk1 promoter, the transcription factor Foxa2 was selected for further verification. Nuclear translocation assay and dual-luciferase reporter assay were used to clarify the involvement of Fam3a-Nr4a2-Foxa2 pathway in PC1/3 expression and GLP-1 production. Moreover, α-cell-specific Fam3a knockout (Fam3a<sup>α-/-</sup>) mice were constructed to evaluate the metabolic variables and hormone levels under normoglycemic, high-fat diet (HFD)-fed and streptozotocin (STZ)-induced diabetic conditions. Exendin 9-39 (Ex9), a GLP-1 receptor antagonist, was used to investigate GLP-1 paracrine effects in Fam3a<sup>α-/-</sup> mice and in their primary islets.</p><p><strong>Results: </strong>Compared with wild-type mice, pancreatic and plasma active GLP-1 levels were increased in Fam3a<sup>-/-</sup> mice. Analysis of human islet scRNA-seq datasets showed a significant negative correction between FAM3A and PCSK1 in α-cells. Fam3a knockdown upregulated PC1/3 expression and GLP-1 production in αTC1.9 cells, while Fam3a overexpression displayed inverse effects. Transcriptomic analysis identified Nr4a2 as a key downstream mediator of Fam3a, and Nr4a2 expression in αTC1.9 cells was downregulated and upregulated by Fam3a knockdown and overexpression, respectively. Nr4a2 silencing increased PC1/3 expression, albeit Nr4a2 did not directly bind to Pcsk1 promoter. Instead, Nr4a2 formed a complex with Foxa2 to facilitate Fam3a-mediated Foxa2 nuclear translocation. Foxa2 negatively regulated PC1/3 expression and GLP-1 production. Besides, Foxa2 inhibited the transcriptional activity of Pcsk1 promoter at specific binding sites 10 and 6, and this inhibition was intensified by Nr4a2 in αTC1.9 cells. Compared with Flox/cre littermates, improved glucose tolerance, increased active GLP-1 level in pancreas and plasma, upregulated plasma insulin level in response to glucose, and decreased plasma glucagon level were observed in Fam3a<sup>α-/-</sup> mice. Primary islets isolated from Fam3a
背景:Fam3a已被证实能调节胰岛β细胞功能和葡萄糖稳态。然而,Fam3a在调节α细胞功能方面的作用和机制仍有待探索:方法:测定了全基因Fam3a基因敲除(Fam3a-/-)小鼠胰腺和血浆中的胰高血糖素和胰高血糖素样肽-1(GLP-1)水平。利用人体胰岛单细胞 RNA 测序(scRNA-seq)数据集分析 FAM3A 和 PCSK1(编码 PC1/3,将胰高血糖素转化为 GLP-1)之间的基因表达相关性。用Fam3a siRNA或质粒转染小鼠胰腺α细胞系αTC1.9细胞,以敲除或过表达Fam3a,从而探讨Fam3a对PC1/3表达和GLP-1生成的影响。通过转录组分析确定了下游介质(包括Nr4a2),并通过Fam3a在αTC1.9细胞中的敲除或过表达证实了其作用。根据 Nr4a2 的相互作用蛋白和与 Pcsk1 启动子的直接结合,转录因子 Foxa2 被选中进行进一步验证。核转位实验和双荧光素酶报告实验明确了Fam3a-Nr4a2-Foxa2通路参与了PC1/3的表达和GLP-1的产生。此外,还构建了α细胞特异性Fam3a基因敲除(Fam3aα-/-)小鼠,以评估正常血糖、高脂饮食(HFD)和链脲佐菌素(STZ)诱导糖尿病条件下的代谢变量和激素水平。GLP-1受体拮抗剂Exendin 9-39(Ex9)被用来研究GLP-1在Fam3aα-/-小鼠及其原代胰岛中的旁分泌效应:结果:与野生型小鼠相比,Fam3a-/-小鼠的胰腺和血浆活性GLP-1水平均有所增加。对人类胰岛 scRNA-seq 数据集的分析表明,α 细胞中的 FAM3A 和 PCSK1 之间存在显著的负校正。Fam3a敲除会上调αTC1.9细胞中PC1/3的表达和GLP-1的产生,而Fam3a过表达则会产生反作用。转录组分析发现,Nr4a2是Fam3a的一个关键下游介质,Fam3a敲除和过表达分别下调和上调了Nr4a2在αTC1.9细胞中的表达。Nr4a2 沉默会增加 PC1/3 的表达,尽管 Nr4a2 并不直接与 Pcsk1 启动子结合。相反,Nr4a2与Foxa2形成复合物,促进Fam3a介导的Foxa2核转位。Foxa2 负向调节 PC1/3 的表达和 GLP-1 的产生。此外,在αTC1.9细胞中,Foxa2在特定结合位点10和6抑制了Pcsk1启动子的转录活性,Nr4a2加强了这种抑制作用。与Flox/cre同窝小鼠相比,Fam3aα-/-小鼠的糖耐量得到改善,胰腺和血浆中活性GLP-1水平升高,血浆胰岛素水平对葡萄糖的反应上调,血浆胰高血糖素水平降低。从 Fam3aα-/- 小鼠体内分离的原代胰岛也显示出活性 GLP-1 和胰岛素释放的增加。此外,在Fam3aα-/-小鼠及其原代胰岛中,胰岛内GLP-1的促胰岛素作用被Ex9阻断。同样,HFD喂养的Fam3aα-/-小鼠的糖耐量也有所改善。HFD喂养和STZ诱导的糖尿病Fam3aα-/-小鼠均表现出胰腺活性GLP-1水平升高、血浆胰岛素水平升高和血浆胰高血糖素水平降低:结论:α细胞中 Fam3a 的缺乏可通过 Nr4a2-Foxa2-PC1/3 依赖性旁分泌信号增强胰腺 GLP-1 的产生,从而改善β细胞的功能。我们的研究揭示了一种将α细胞胰高血糖素加工输出从胰高血糖素重编程为GLP-1的新策略,并加深了对α细胞和β细胞之间串联的理解。
{"title":"Fam3a-mediated prohormone convertase switch in α-cells regulates pancreatic GLP-1 production in an Nr4a2-Foxa2-dependent manner.","authors":"Dandan Wang, Tianjiao Wei, Xiaona Cui, Li Xia, Yafei Jiang, Deshan Yin, Xinyue Liao, Fei Li, Jian Li, Qi Wu, Xiafang Lin, Shan Lang, Yunyi Le, Jichun Yang, Jin Yang, Rui Wei, Tianpei Hong","doi":"10.1016/j.metabol.2024.156042","DOIUrl":"10.1016/j.metabol.2024.156042","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Background: &lt;/strong&gt;Fam3a has been demonstrated to regulate pancreatic β-cell function and glucose homeostasis. However, the role and mechanism of Fam3a in regulating α-cell function remain unexplored.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;Glucagon and glucagon-like peptide-1 (GLP-1) levels in pancreas and plasma were measured in global Fam3a knockout (Fam3a&lt;sup&gt;-/-&lt;/sup&gt;) mice. Human islet single-cell RNA sequencing (scRNA-seq) datasets were utilized to analyze gene expression correlations between FAM3A and PCSK1 (encoding PC1/3, which processes proglucagon into GLP-1). Mouse pancreatic α-cell line αTC1.9 cells were transfected with Fam3a siRNA or plasmid for Fam3a knockdown or overexpression to explore the effects of Fam3a on PC1/3 expression and GLP-1 production. The downstream mediator (including Nr4a2) was identified by transcriptomic analysis, and its role was confirmed by Fam3a knockdown or overexpression in αTC1.9 cells. Based on the interacted protein of Nr4a2 and the direct binding to Pcsk1 promoter, the transcription factor Foxa2 was selected for further verification. Nuclear translocation assay and dual-luciferase reporter assay were used to clarify the involvement of Fam3a-Nr4a2-Foxa2 pathway in PC1/3 expression and GLP-1 production. Moreover, α-cell-specific Fam3a knockout (Fam3a&lt;sup&gt;α-/-&lt;/sup&gt;) mice were constructed to evaluate the metabolic variables and hormone levels under normoglycemic, high-fat diet (HFD)-fed and streptozotocin (STZ)-induced diabetic conditions. Exendin 9-39 (Ex9), a GLP-1 receptor antagonist, was used to investigate GLP-1 paracrine effects in Fam3a&lt;sup&gt;α-/-&lt;/sup&gt; mice and in their primary islets.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;Compared with wild-type mice, pancreatic and plasma active GLP-1 levels were increased in Fam3a&lt;sup&gt;-/-&lt;/sup&gt; mice. Analysis of human islet scRNA-seq datasets showed a significant negative correction between FAM3A and PCSK1 in α-cells. Fam3a knockdown upregulated PC1/3 expression and GLP-1 production in αTC1.9 cells, while Fam3a overexpression displayed inverse effects. Transcriptomic analysis identified Nr4a2 as a key downstream mediator of Fam3a, and Nr4a2 expression in αTC1.9 cells was downregulated and upregulated by Fam3a knockdown and overexpression, respectively. Nr4a2 silencing increased PC1/3 expression, albeit Nr4a2 did not directly bind to Pcsk1 promoter. Instead, Nr4a2 formed a complex with Foxa2 to facilitate Fam3a-mediated Foxa2 nuclear translocation. Foxa2 negatively regulated PC1/3 expression and GLP-1 production. Besides, Foxa2 inhibited the transcriptional activity of Pcsk1 promoter at specific binding sites 10 and 6, and this inhibition was intensified by Nr4a2 in αTC1.9 cells. Compared with Flox/cre littermates, improved glucose tolerance, increased active GLP-1 level in pancreas and plasma, upregulated plasma insulin level in response to glucose, and decreased plasma glucagon level were observed in Fam3a&lt;sup&gt;α-/-&lt;/sup&gt; mice. Primary islets isolated from Fam3a","PeriodicalId":18694,"journal":{"name":"Metabolism: clinical and experimental","volume":" ","pages":"156042"},"PeriodicalIF":10.8,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142372341","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
Metabolism: clinical and experimental
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1