首页 > 最新文献

American journal of physiology. Cell physiology最新文献

英文 中文
An Endogenous Aryl Hydrocarbon Receptor Ligand Dysregulates Endothelial Functions, Transcriptome, and Phosphoproteome.
IF 5 2区 生物学 Q2 CELL BIOLOGY Pub Date : 2025-02-05 DOI: 10.1152/ajpcell.00849.2024
Ying-Jie Zhao, Si-Yan Zhang, Ying-Ying Wei, Hui-Hui Li, Wei Lei, Kai Wang, Sathish Kumar, Chi Zhou, Jing Zheng

We have reported that an endogenous aryl hydrocarbon receptor (AhR) ligand, 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE), inhibits functions of human umbilical vein endothelial cells (HUVECs) and induces preeclampsia (PE)-like symptoms in rats. Herein, we tested the hypothesis that ITE impairs endothelial functions via disturbing transcriptome and phosphoproteome in HUVECs. We measured AhR activity in human maternal and umbilical vein sera from PE and normotensive (NT) pregnancies. The serum-induced changes in CYP1A1/B1 mRNA (indexes of AhR activation) in HUVECs were quantified using RT-qPCR. ITE's effects on endothelial proliferation and monolayer integrity in female and male HUVECs were determined. We profiled ITE-induced changes in transcriptome and phosphoproteome in HUVECs using RNA-seq and bottom-up phosphoproteomics, respectively. After 12 hr of treatment, umbilical vein sera from PE increased CYP1A1 mRNA (1.7-fold of NT) HUVECs, which was blocked by CH223191, an AhR antagonist. ITE dose-dependently inhibited endothelial proliferation (76%-87% of control) and time-dependently reduced endothelial integrity with a maximum inhibition (~ 10%) at 40 hr. ITE induced 140 and 80 differentially expressed genes in female and male HUVECs, respectively. ITE altered phosphorylation of 92 and 105 proteins at 4 and 24 hr, respectively, in HUVECs. These ITE-dysregulated genes and phosphoproteins were enriched in biological functions and pathways are relevant to heart, liver, and kidney diseases, vascular functions, and inflammatory responses. Thus, endogenous AhR ligands may impair endothelial functions by disturbing transcriptome and phosphoproteome. These AhR ligand-dysregulated genes and phosphoproteins may be therapeutic and cell sex-specific targets for PE-induced endothelial dysfunction.

{"title":"An Endogenous Aryl Hydrocarbon Receptor Ligand Dysregulates Endothelial Functions, Transcriptome, and Phosphoproteome.","authors":"Ying-Jie Zhao, Si-Yan Zhang, Ying-Ying Wei, Hui-Hui Li, Wei Lei, Kai Wang, Sathish Kumar, Chi Zhou, Jing Zheng","doi":"10.1152/ajpcell.00849.2024","DOIUrl":"10.1152/ajpcell.00849.2024","url":null,"abstract":"<p><p>We have reported that an endogenous aryl hydrocarbon receptor (AhR) ligand, 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE), inhibits functions of human umbilical vein endothelial cells (HUVECs) and induces preeclampsia (PE)-like symptoms in rats. Herein, we tested the hypothesis that ITE impairs endothelial functions via disturbing transcriptome and phosphoproteome in HUVECs. We measured AhR activity in human maternal and umbilical vein sera from PE and normotensive (NT) pregnancies. The serum-induced changes in CYP1A1/B1 mRNA (indexes of AhR activation) in HUVECs were quantified using RT-qPCR. ITE's effects on endothelial proliferation and monolayer integrity in female and male HUVECs were determined. We profiled ITE-induced changes in transcriptome and phosphoproteome in HUVECs using RNA-seq and bottom-up phosphoproteomics, respectively. After 12 hr of treatment, umbilical vein sera from PE increased CYP1A1 mRNA (1.7-fold of NT) HUVECs, which was blocked by CH223191, an AhR antagonist. ITE dose-dependently inhibited endothelial proliferation (76%-87% of control) and time-dependently reduced endothelial integrity with a maximum inhibition (~ 10%) at 40 hr. ITE induced 140 and 80 differentially expressed genes in female and male HUVECs, respectively. ITE altered phosphorylation of 92 and 105 proteins at 4 and 24 hr, respectively, in HUVECs. These ITE-dysregulated genes and phosphoproteins were enriched in biological functions and pathways are relevant to heart, liver, and kidney diseases, vascular functions, and inflammatory responses. Thus, endogenous AhR ligands may impair endothelial functions by disturbing transcriptome and phosphoproteome. These AhR ligand-dysregulated genes and phosphoproteins may be therapeutic and cell sex-specific targets for PE-induced endothelial dysfunction.</p>","PeriodicalId":7585,"journal":{"name":"American journal of physiology. Cell physiology","volume":" ","pages":""},"PeriodicalIF":5.0,"publicationDate":"2025-02-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143187659","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Effects of oral γ-aminobutyric acid intake on muscle regeneration in diabetic mice.
IF 5 2区 生物学 Q2 CELL BIOLOGY Pub Date : 2025-02-05 DOI: 10.1152/ajpcell.00963.2024
Mayu Horii, Chanikan Bumrungkit, Noriyuki Yanaka, Thomas J Hawke, Irena A Rebalka, Thanutchaporn Kumrungsee

Though γ-aminobutyric acid (GABA) serves as the primary inhibitory neurotransmitter in the brain, its numerous biological activities in the periphery, including anti-inflammatory and anti-diabetic functions, have been documented. Additionally, GABA may be a mediator underlying effects of ketone bodies/ketogenic diets on muscle regeneration. Here, we investigated effects of GABA on muscle regeneration in Type 1 Diabetes mouse models. Akita and wild-type (WT) mice were treated with GABA in drinking water for 6 weeks, followed by cardiotoxin (CTX)-induced muscle injury. At 5 days post-injury, GABA treatment exhibited no effects on regenerating myofiber size in both WT and Akita mice. Unexpectedly, regenerating GABA-treated Akita muscles exhibited significantly increased embryonic myosin heavy chain (eMHC) expression and higher intramuscular-macrophage content, suggesting delays in muscle regeneration and in elevated macrophage infiltration in diabetic muscles. Next, we determined if GABA treatment delayed the inflammatory process during muscle regeneration. Providing GABA in the drinking water during the peak inflammatory period (days 0 to 5 post-injury) resulted in a significantly greater amount of small regenerating myofibers and higher expressions of TNFα and eMHC in regenerating streptozotocin (STZ)-diabetic muscles, indicating delays in inflammation process and muscle regeneration in diabetes. Plasma GABA levels were found higher in GABA-treated STZ mice than WT mice and negatively correlated with regenerating myofiber size. This delay in muscle regeneration in STZ-diabetic mice was abolished by a lower dose of GABA water that did not raise plasma GABA levels. Together, high doses of GABA intake during the early phases of muscle repair may delay regeneration.

{"title":"Effects of oral γ-aminobutyric acid intake on muscle regeneration in diabetic mice.","authors":"Mayu Horii, Chanikan Bumrungkit, Noriyuki Yanaka, Thomas J Hawke, Irena A Rebalka, Thanutchaporn Kumrungsee","doi":"10.1152/ajpcell.00963.2024","DOIUrl":"https://doi.org/10.1152/ajpcell.00963.2024","url":null,"abstract":"<p><p>Though γ-aminobutyric acid (GABA) serves as the primary inhibitory neurotransmitter in the brain, its numerous biological activities in the periphery, including anti-inflammatory and anti-diabetic functions, have been documented. Additionally, GABA may be a mediator underlying effects of ketone bodies/ketogenic diets on muscle regeneration. Here, we investigated effects of GABA on muscle regeneration in Type 1 Diabetes mouse models. Akita and wild-type (WT) mice were treated with GABA in drinking water for 6 weeks, followed by cardiotoxin (CTX)-induced muscle injury. At 5 days post-injury, GABA treatment exhibited no effects on regenerating myofiber size in both WT and Akita mice. Unexpectedly, regenerating GABA-treated Akita muscles exhibited significantly increased embryonic myosin heavy chain (eMHC) expression and higher intramuscular-macrophage content, suggesting delays in muscle regeneration and in elevated macrophage infiltration in diabetic muscles. Next, we determined if GABA treatment delayed the inflammatory process during muscle regeneration. Providing GABA in the drinking water during the peak inflammatory period (days 0 to 5 post-injury) resulted in a significantly greater amount of small regenerating myofibers and higher expressions of TNFα and eMHC in regenerating streptozotocin (STZ)-diabetic muscles, indicating delays in inflammation process and muscle regeneration in diabetes. Plasma GABA levels were found higher in GABA-treated STZ mice than WT mice and negatively correlated with regenerating myofiber size. This delay in muscle regeneration in STZ-diabetic mice was abolished by a lower dose of GABA water that did not raise plasma GABA levels. Together, high doses of GABA intake during the early phases of muscle repair may delay regeneration.</p>","PeriodicalId":7585,"journal":{"name":"American journal of physiology. Cell physiology","volume":" ","pages":""},"PeriodicalIF":5.0,"publicationDate":"2025-02-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143187661","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Inflammation induced PFKFB3-mediated glycolysis promoting myometrium contraction through the PI3K-Akt-mTOR pathway in preterm birth mice.
IF 5 2区 生物学 Q2 CELL BIOLOGY Pub Date : 2025-02-05 DOI: 10.1152/ajpcell.00704.2024
Jing He, Xuan Li, Huihui Yu, Chenyi Xu, Ruixian Tian, Ping Zhou, Zongzhi Yin

Inflammation is a significant risk factor for preterm birth. Inflammation enhances glycolytic processes in various cell types and contributes to the development of myometrial contractions. However, the potential of inflammation to activate glycolysis in pregnant murine uterine smooth muscle cells (mUSMCs) and its role in promoting inflammatory preterm birth remain unexplored. In this study, lipopolysaccharide was employed to establish both cell and animal inflammation models. We found that inflammation of mUSMCs during late pregnancy could initiate glycolysis and promoted cell contraction. Subsequently, the inhibition of glycolysis using the glycolysis inhibitor 2-deoxyglucose can reverse inflammation-induced cell contraction. The expression of 6-phosphofructokinase 2 kinase (PFKFB3) was significantly upregulated in mUSMCs following lipopolysaccharide stimulation. Additionally, lactate accumulation and enhanced contraction were observed. Inhibition of PFKFB3 reversed the lactate accumulation and enhanced contraction induced by inflammation. We also found that inflammation activated the phosphatidylinositol 3-kinase (PI3K) - protein kinase B (Akt) - mammalian target of rapamycin (mTOR) pathway, leading to the upregulation of PFKFB3 expression. The PI3K-Akt pathway inhibitor LY294002 and the mTOR pathway inhibitor Rapamycin effectively inhibited the upregulation of PFKFB3 protein expression, lactate production, and the enhancement of cell contraction induced by lipopolysaccharide. This study indicates that inflammation regulates PFKFB3 through the PI3K-Akt-mTOR pathway, which enhances the glycolytic process in pregnant mUSMCs, ultimately leading to myometrial contraction.

{"title":"Inflammation induced PFKFB3-mediated glycolysis promoting myometrium contraction through the PI3K-Akt-mTOR pathway in preterm birth mice.","authors":"Jing He, Xuan Li, Huihui Yu, Chenyi Xu, Ruixian Tian, Ping Zhou, Zongzhi Yin","doi":"10.1152/ajpcell.00704.2024","DOIUrl":"https://doi.org/10.1152/ajpcell.00704.2024","url":null,"abstract":"<p><p>Inflammation is a significant risk factor for preterm birth. Inflammation enhances glycolytic processes in various cell types and contributes to the development of myometrial contractions. However, the potential of inflammation to activate glycolysis in pregnant murine uterine smooth muscle cells (mUSMCs) and its role in promoting inflammatory preterm birth remain unexplored. In this study, lipopolysaccharide was employed to establish both cell and animal inflammation models. We found that inflammation of mUSMCs during late pregnancy could initiate glycolysis and promoted cell contraction. Subsequently, the inhibition of glycolysis using the glycolysis inhibitor 2-deoxyglucose can reverse inflammation-induced cell contraction. The expression of 6-phosphofructokinase 2 kinase (PFKFB3) was significantly upregulated in mUSMCs following lipopolysaccharide stimulation. Additionally, lactate accumulation and enhanced contraction were observed. Inhibition of PFKFB3 reversed the lactate accumulation and enhanced contraction induced by inflammation. We also found that inflammation activated the phosphatidylinositol 3-kinase (PI3K) - protein kinase B (Akt) - mammalian target of rapamycin (mTOR) pathway, leading to the upregulation of PFKFB3 expression. The PI3K-Akt pathway inhibitor LY294002 and the mTOR pathway inhibitor Rapamycin effectively inhibited the upregulation of PFKFB3 protein expression, lactate production, and the enhancement of cell contraction induced by lipopolysaccharide. This study indicates that inflammation regulates PFKFB3 through the PI3K-Akt-mTOR pathway, which enhances the glycolytic process in pregnant mUSMCs, ultimately leading to myometrial contraction.</p>","PeriodicalId":7585,"journal":{"name":"American journal of physiology. Cell physiology","volume":" ","pages":""},"PeriodicalIF":5.0,"publicationDate":"2025-02-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143187663","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cellular mechanisms underlying overreaching in skeletal muscle following excessive high-intensity interval training.
IF 5 2区 生物学 Q2 CELL BIOLOGY Pub Date : 2025-02-04 DOI: 10.1152/ajpcell.00623.2024
Daiki Watanabe, Masanobu Wada

Overreaching (OR) can be defined as a decline in physical performance resulting from excessive exercise training, necessitating days to weeks recovery. Impairments in the contractile function of skeletal muscle are believed to be a primary factor contributing to OR. However, the cellular mechanism triggering OR remains unclear. The purpose of this study was to elu idate the mechanisms underlying OR. Rats' plantar flexor muscles were subjected to repeated electrical stimulations mimicking excessive high-intensity interval training (HIIT) daily for 13 consecutive days, and isometric torques were monitored. The torque was measured one day after HIIT, and subsequently, the physiological function of type II fibers was analyzed by using mechanically-skinned-fiber technique. Eleven out of 17 rats exhibited torque decline, while others did not. Thus, the rats were divided into OR and non-overreaching (NOR) groups. Skinned fibers from the gastrocnemius (GAS) muscles of both groups showed decreased depolarization-induced force and increased myofibrillar Ca2+ sensitivity.However, the fibers from the OR group, but not the NOR group, exhibited a decrease in myofibrillar maximal force. Biochemical analyses of a superficial region of GAS muscle revealed that α-actinin 2 content was increased in the NOR group, but not the OR group, whereas calpain-3 autolysis was increased in the OR group, but not the NOR group. These findings shed light on the cellular mechanism underlying OR: OR following excessive HIIT was induced by a decreased myofibrillar maximal force, while Ca2+ sensitivity was increased.

{"title":"Cellular mechanisms underlying overreaching in skeletal muscle following excessive high-intensity interval training.","authors":"Daiki Watanabe, Masanobu Wada","doi":"10.1152/ajpcell.00623.2024","DOIUrl":"https://doi.org/10.1152/ajpcell.00623.2024","url":null,"abstract":"<p><p>Overreaching (OR) can be defined as a decline in physical performance resulting from excessive exercise training, necessitating days to weeks recovery. Impairments in the contractile function of skeletal muscle are believed to be a primary factor contributing to OR. However, the cellular mechanism triggering OR remains unclear. The purpose of this study was to elu idate the mechanisms underlying OR. Rats' plantar flexor muscles were subjected to repeated electrical stimulations mimicking excessive high-intensity interval training (HIIT) daily for 13 consecutive days, and isometric torques were monitored. The torque was measured one day after HIIT, and subsequently, the physiological function of type II fibers was analyzed by using mechanically-skinned-fiber technique. Eleven out of 17 rats exhibited torque decline, while others did not. Thus, the rats were divided into OR and non-overreaching (NOR) groups. Skinned fibers from the gastrocnemius (GAS) muscles of both groups showed decreased depolarization-induced force and increased myofibrillar Ca<sup>2+</sup> sensitivity.However, the fibers from the OR group, but not the NOR group, exhibited a decrease in myofibrillar maximal force. Biochemical analyses of a superficial region of GAS muscle revealed that α-actinin 2 content was increased in the NOR group, but not the OR group, whereas calpain-3 autolysis was increased in the OR group, but not the NOR group. These findings shed light on the cellular mechanism underlying OR: OR following excessive HIIT was induced by a decreased myofibrillar maximal force, while Ca<sup>2+</sup> sensitivity was increased.</p>","PeriodicalId":7585,"journal":{"name":"American journal of physiology. Cell physiology","volume":" ","pages":""},"PeriodicalIF":5.0,"publicationDate":"2025-02-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143187660","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Runx2-NLRP3 axis orchestrates matrix stiffness-evoked vascular smooth muscle cell inflammation.
IF 5 2区 生物学 Q2 CELL BIOLOGY Pub Date : 2025-02-01 Epub Date: 2025-01-06 DOI: 10.1152/ajpcell.00448.2024
Zhiqing Li, Hao Wu, Fang Yao, Yiran Li, Yanjie Li, Si-An Xie, Fang Yu, Yi Fu, Li Wang, Jing Zhou, Wei Kong

Arterial stiffening is a hallmark of chronic kidney disease (CKD)-related cardiovascular events and is primarily attributed to the elevated matrix stiffness. Stiffened arteries are accompanied by low-grade inflammation, but the causal effects of matrix stiffness on inflammation remain unknown. For analysis of the relationship between arterial stiffness and vascular inflammation, pulse-wave velocity (PWV) and aortic inflammatory markers were analyzed in an adenine-induced mouse model of CKD in chronological order. Compared with their control littermates, mice with CKD showed elevated arterial stiffness at the early stage of disease progression, which preceded the onset of vascular inflammation. Correspondingly, the increase of matrix stiffness induced vascular smooth muscle cells (VSMCs) to transdifferentiate into an inflammatory phenotype, as indicated by the increased expression and secretion of MCP-1, IL-6, IL-1β, and IL-18. RNA-sequencing analysis of stiff matrix-cultured VSMCs and bioinformatics analysis with the ChIP-Atlas database revealed the potential involvement of the transcription factor Runx2. The expression and the nuclear localization of Runx2 were significantly increased in stiff matrix-cultured VSMCs. High-throughput ChIP-sequencing and promoter luciferase assays further revealed that NLRP3 was directly transcriptionally regulated by Runx2. The inhibition of Runx2 or NLRP3 inflammasome abrogated the proinflammatory effect of matrix stiffening on VSMCs. Together, these data revealed that arterial stiffness precedes vascular inflammatory responses in CKD mice and that the Runx2-NLRP3 axis orchestrates matrix stiffness and the VSMC inflammatory phenotype, which may contribute to the pathogenic role in arterial stiffness-related vascular inflammation and CKD-related cardiovascular complications.NEW & NOTEWORTHY As a hallmark of chronic kidney disease (CKD), arterial stiffening is related to increased vascular inflammation and cardiovascular morbidity, whereas the underlying mechanism is unclear. The study demonstrates that increased arterial stiffness precedes the onset of vascular inflammation, and matrix stiffness stimulates the transdifferentiation of vascular smooth muscle cells (VSMCs) to an inflammatory phenotype via activating Runx2-NLRP3 signaling, which provides novel insights into CKD-related cardiovascular disorder treatment.

{"title":"Runx2-NLRP3 axis orchestrates matrix stiffness-evoked vascular smooth muscle cell inflammation.","authors":"Zhiqing Li, Hao Wu, Fang Yao, Yiran Li, Yanjie Li, Si-An Xie, Fang Yu, Yi Fu, Li Wang, Jing Zhou, Wei Kong","doi":"10.1152/ajpcell.00448.2024","DOIUrl":"https://doi.org/10.1152/ajpcell.00448.2024","url":null,"abstract":"<p><p>Arterial stiffening is a hallmark of chronic kidney disease (CKD)-related cardiovascular events and is primarily attributed to the elevated matrix stiffness. Stiffened arteries are accompanied by low-grade inflammation, but the causal effects of matrix stiffness on inflammation remain unknown. For analysis of the relationship between arterial stiffness and vascular inflammation, pulse-wave velocity (PWV) and aortic inflammatory markers were analyzed in an adenine-induced mouse model of CKD in chronological order. Compared with their control littermates, mice with CKD showed elevated arterial stiffness at the early stage of disease progression, which preceded the onset of vascular inflammation. Correspondingly, the increase of matrix stiffness induced vascular smooth muscle cells (VSMCs) to transdifferentiate into an inflammatory phenotype, as indicated by the increased expression and secretion of MCP-1, IL-6, IL-1β, and IL-18. RNA-sequencing analysis of stiff matrix-cultured VSMCs and bioinformatics analysis with the ChIP-Atlas database revealed the potential involvement of the transcription factor Runx2. The expression and the nuclear localization of Runx2 were significantly increased in stiff matrix-cultured VSMCs. High-throughput ChIP-sequencing and promoter luciferase assays further revealed that <i>NLRP3</i> was directly transcriptionally regulated by Runx2. The inhibition of Runx2 or NLRP3 inflammasome abrogated the proinflammatory effect of matrix stiffening on VSMCs. Together, these data revealed that arterial stiffness precedes vascular inflammatory responses in CKD mice and that the Runx2-NLRP3 axis orchestrates matrix stiffness and the VSMC inflammatory phenotype, which may contribute to the pathogenic role in arterial stiffness-related vascular inflammation and CKD-related cardiovascular complications.<b>NEW & NOTEWORTHY</b> As a hallmark of chronic kidney disease (CKD), arterial stiffening is related to increased vascular inflammation and cardiovascular morbidity, whereas the underlying mechanism is unclear. The study demonstrates that increased arterial stiffness precedes the onset of vascular inflammation, and matrix stiffness stimulates the transdifferentiation of vascular smooth muscle cells (VSMCs) to an inflammatory phenotype via activating Runx2-NLRP3 signaling, which provides novel insights into CKD-related cardiovascular disorder treatment.</p>","PeriodicalId":7585,"journal":{"name":"American journal of physiology. Cell physiology","volume":"328 2","pages":"C467-C482"},"PeriodicalIF":5.0,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143021508","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Histone lactylation-mediated overexpression of RASD2 promotes endometriosis progression via upregulating the SUMOylation of CTPS1. 组蛋白乳酸化介导的RASD2过表达通过上调CTPS1的SUMOylation促进子宫内膜异位症的进展。
IF 5 2区 生物学 Q2 CELL BIOLOGY Pub Date : 2025-02-01 Epub Date: 2024-12-13 DOI: 10.1152/ajpcell.00493.2024
Ziwei Wang, Yanhong Mao, Zihan Wang, Shuwei Li, Zhidan Hong, Rong Zhou, Shaoyuan Xu, Yao Xiong, Yuanzhen Zhang

Histone lactylation is crucial in a variety of physiopathological processes; however, the function and mechanism of histone lactylation in endometriosis remain poorly understood. Therefore, the objective of this investigation was to illuminate the function and mechanism of histone lactylation in endometriosis. Immunohistochemistry was used to investigate the expression of histone lactylation. Cell Counting Kit-8 assay (CCK8), Transwell assay, and endometriosis mouse models were used to investigate the effects of histone lactylation in vitro and in vivo. Transcriptomics and immunoprecipitation-mass spectrometry (IP-MS), Western blot, co-immunoprecipitation (Co-IP), quantitative reverse transcription polymerase chain reaction (qRT-PCR), and chromatin immunoprecipitation-qPCR (ChIP-qPCR) were used to explore the intrinsic mechanisms. In this study, we found that histone lactylation was upregulated in endometriosis and could promote endometriosis progression both in vivo and in vitro. Mechanistically, histone lactylation H3K18la promoted the transcription of Ras homolog enriched in striatum (RASD2), and RASD2, in turn, increased the stability of CTP synthase 1 (CTPS1) by promoting the SUMOylation and inhibiting the ubiquitination of CTPS1, thereby promoting endometriosis progression. Overall, our findings indicated that histone lactylation could promote the progression of endometriosis through the RASD2/CTPS1 axis. This investigation uncovered a novel mechanism and identified prospective targets for endometriosis diagnosis and therapy.NEW & NOTEWORTHY Our finding reveals a novel mechanism that promotes the progression of endometriosis, namely the histone lactylation/RASD2/CTPS1 axis. This finding suggests that inhibiting histone lactylation or inhibiting RASD2 and CTPS1 might be a potential therapeutic strategy to inhibit endometriosis lesion growth.

背景:组蛋白乳酸化在多种生理病理过程中起着至关重要的作用,然而,组蛋白乳酸化在子宫内膜异位症中的作用和机制仍然知之甚少。因此,本研究的目的是阐明组蛋白乳酸化在子宫内膜异位症中的作用和机制。方法:采用免疫组化方法检测组蛋白乳酸化的表达。采用细胞计数试剂盒-8 (CCK8)法、Transwell法和子宫内膜异位症小鼠模型研究组蛋白乳酸化对体外和体内的影响。利用转录组学和免疫沉淀-质谱(IP-MS)、Western Blot、共免疫沉淀(Co-IP)、定量逆转录聚合酶链反应(qRT-PCR)和染色质免疫沉淀- qpcr (ChIP-qPCR)等方法探讨其内在机制。结果:在本研究中,我们发现组蛋白乳酸化在子宫内膜异位症中上调,并在体内和体外促进子宫内膜异位症的进展。机制上,组蛋白乳酸化H3K18la促进纹状体富集Ras同源物(RASD2)的转录,而RASD2又通过促进CTPS1的sumo化和抑制泛素化,增加CTP合成酶1 (CTPS1)的稳定性,从而促进子宫内膜异位症的进展。结论:总体而言,我们的研究结果表明,组蛋白乳酸化可通过RASD2/CTPS1轴促进子宫内膜异位症的进展。本研究揭示了子宫内膜异位症的新机制,并确定了子宫内膜异位症诊断和治疗的潜在靶点。
{"title":"Histone lactylation-mediated overexpression of RASD2 promotes endometriosis progression via upregulating the SUMOylation of CTPS1.","authors":"Ziwei Wang, Yanhong Mao, Zihan Wang, Shuwei Li, Zhidan Hong, Rong Zhou, Shaoyuan Xu, Yao Xiong, Yuanzhen Zhang","doi":"10.1152/ajpcell.00493.2024","DOIUrl":"10.1152/ajpcell.00493.2024","url":null,"abstract":"<p><p>Histone lactylation is crucial in a variety of physiopathological processes; however, the function and mechanism of histone lactylation in endometriosis remain poorly understood. Therefore, the objective of this investigation was to illuminate the function and mechanism of histone lactylation in endometriosis. Immunohistochemistry was used to investigate the expression of histone lactylation. Cell Counting Kit-8 assay (CCK8), Transwell assay, and endometriosis mouse models were used to investigate the effects of histone lactylation in vitro and in vivo. Transcriptomics and immunoprecipitation-mass spectrometry (IP-MS), Western blot, co-immunoprecipitation (Co-IP), quantitative reverse transcription polymerase chain reaction (qRT-PCR), and chromatin immunoprecipitation-qPCR (ChIP-qPCR) were used to explore the intrinsic mechanisms. In this study, we found that histone lactylation was upregulated in endometriosis and could promote endometriosis progression both in vivo and in vitro. Mechanistically, histone lactylation H3K18la promoted the transcription of Ras homolog enriched in striatum (RASD2), and RASD2, in turn, increased the stability of CTP synthase 1 (CTPS1) by promoting the SUMOylation and inhibiting the ubiquitination of CTPS1, thereby promoting endometriosis progression. Overall, our findings indicated that histone lactylation could promote the progression of endometriosis through the RASD2/CTPS1 axis. This investigation uncovered a novel mechanism and identified prospective targets for endometriosis diagnosis and therapy.<b>NEW & NOTEWORTHY</b> Our finding reveals a novel mechanism that promotes the progression of endometriosis, namely the histone lactylation/RASD2/CTPS1 axis. This finding suggests that inhibiting histone lactylation or inhibiting RASD2 and CTPS1 might be a potential therapeutic strategy to inhibit endometriosis lesion growth.</p>","PeriodicalId":7585,"journal":{"name":"American journal of physiology. Cell physiology","volume":" ","pages":"C500-C513"},"PeriodicalIF":5.0,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142821823","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
dEREGulated pathways: unraveling the role of epiregulin in skin, kidney, and lung fibrosis. 解除管制的途径:揭示表调节蛋白在皮肤、肾脏和肺纤维化中的作用。
IF 5 2区 生物学 Q2 CELL BIOLOGY Pub Date : 2025-02-01 Epub Date: 2025-01-03 DOI: 10.1152/ajpcell.00813.2024
Aysan Ezaddoustdar, Daniel Kalina, Maximilian Bielohuby, Mario Boehm, Malgorzata Wygrecka

The epidermal growth factor receptor (EGFR) signaling pathway is an evolutionary conserved mechanism to control cell behavior during tissue development and homeostasis. Deregulation of this pathway has been associated with abnormal cell behavior, including hyperproliferation, senescence, and an inflammatory cell phenotype, thereby contributing to pathologies across a variety of organs, including the kidneys, skin, and lungs. To date, there are seven distinct EGFR ligands described. Although binding of these ligands to the receptor is cell type-specific and spatio-temporally controlled with distinct affinities and kinetics, epiregulin (EREG) stands out as a long-acting EGFR ligand that emerges under pathological conditions, particularly in tissue fibrosis. Although EREG has been extensively studied in cancer, its contribution to the maladaptive remodeling of tissue is elusive. The aim of this review is to highlight the role of EREG in skin, kidney, and lung fibrosis and to discuss opportunities for therapeutic intervention.

表皮生长因子受体(EGFR)信号通路是一种进化保守的机制,在组织发育和稳态过程中控制细胞行为。该通路的解除与异常细胞行为相关,包括过度增生、衰老和炎症细胞表型,从而导致多种器官的病理,包括肾脏、皮肤和肺。迄今为止,已经描述了七种不同的EGFR配体。虽然这些配体与受体的结合具有细胞类型特异性和时空控制,具有不同的亲和力和动力学,但表调节蛋白(EREG)作为一种长效EGFR配体在病理条件下出现,特别是在组织纤维化中。尽管EREG在癌症中已经被广泛研究,但其对组织不适应重塑的贡献尚不明确。这篇综述的目的是强调EREG在皮肤、肾脏和肺纤维化中的作用,并讨论治疗干预的机会。
{"title":"dEREGulated pathways: unraveling the role of epiregulin in skin, kidney, and lung fibrosis.","authors":"Aysan Ezaddoustdar, Daniel Kalina, Maximilian Bielohuby, Mario Boehm, Malgorzata Wygrecka","doi":"10.1152/ajpcell.00813.2024","DOIUrl":"10.1152/ajpcell.00813.2024","url":null,"abstract":"<p><p>The epidermal growth factor receptor (EGFR) signaling pathway is an evolutionary conserved mechanism to control cell behavior during tissue development and homeostasis. Deregulation of this pathway has been associated with abnormal cell behavior, including hyperproliferation, senescence, and an inflammatory cell phenotype, thereby contributing to pathologies across a variety of organs, including the kidneys, skin, and lungs. To date, there are seven distinct EGFR ligands described. Although binding of these ligands to the receptor is cell type-specific and spatio-temporally controlled with distinct affinities and kinetics, epiregulin (EREG) stands out as a long-acting EGFR ligand that emerges under pathological conditions, particularly in tissue fibrosis. Although EREG has been extensively studied in cancer, its contribution to the maladaptive remodeling of tissue is elusive. The aim of this review is to highlight the role of EREG in skin, kidney, and lung fibrosis and to discuss opportunities for therapeutic intervention.</p>","PeriodicalId":7585,"journal":{"name":"American journal of physiology. Cell physiology","volume":" ","pages":"C617-C626"},"PeriodicalIF":5.0,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142920397","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
METTL14 promotes ferroptosis in smooth muscle cells during thoracic aortic aneurysm by stabilizing the m6A modification of ACSL4. METTL14通过稳定ACSL4的m6A修饰促进胸主动脉瘤期间平滑肌细胞的铁下垂。
IF 5 2区 生物学 Q2 CELL BIOLOGY Pub Date : 2025-02-01 Epub Date: 2024-12-13 DOI: 10.1152/ajpcell.00577.2024
Wenjun Wang, Jiayi Chen, Songqing Lai, Ruiyuan Zeng, Ming Fang, Li Wan, Yiying Li

Thoracic aortic aneurysm (TAA) is a vascular disease associated with high mortality rates. Ferroptosis has been shown to mediate the transformation of vascular smooth muscle cells (VSMCs). However, the regulatory mechanisms by which ferroptosis influences TAA remain unclear. In this study, we induced TAA mouse models using angiotensin II (Ang II) and evaluated the impact of ferroptosis on the pathological changes observed in TAA mice, employing liproxstatin-1 as a treatment. In addition, we assessed the regulatory effect of METTL14 on the ferroptosis of VSMCs after treating them with a ferroptosis activator (imidazole ketone erastin, IKE). RNA binding protein immunoprecipitation (RIP) and RNA pull-down assays were conducted to investigate the interaction between acyl-CoA synthase long-chain family member 4 (ACSL4) mRNA and the proteins METTL14 or IGF2BP2. The results indicated that the level of ferroptosis was elevated in the thoracic aorta of TAA mice, and METTL14 was upregulated in TAA models and IKE-induced VSMCs. Knockdown of METTL14 was found to inhibit the progression of TAA by reducing the ferroptosis of VSMCs. Furthermore, IGF2BP2 recognized METTL14-modified ACSL4 mRNA and regulated its stability, thereby mediating the ferroptosis of VSMCs. Collectively, the effects of METTL14 on VSMC ferroptosis present therapeutic potential for the treatment of TAA.NEW & NOTEWORTHY Our study confirmed that METTL14 can induce ferroptosis in vascular smooth muscle cells during the progression of thoracic aortic aneurysm by mediating the m6A modification of ACSL4 mRNA.

胸主动脉瘤(TAA)是一种高死亡率的血管性疾病。铁下垂已被证明介导血管平滑肌细胞(VSMCs)的转化。然而,铁下垂影响TAA的调节机制尚不清楚。本研究采用血管紧张素II (angiotensin II, Ang II)诱导TAA小鼠模型,采用利普司他汀-1治疗,评估铁下垂对TAA小鼠病理改变的影响。此外,我们评估了METTL14在使用铁下垂激活剂(IKE)处理VSMCs后对其铁下垂的调节作用。采用RNA结合蛋白免疫沉淀(RIP)和RNA拉下法研究ACSL4 mRNA与METTL14或IGF2BP2蛋白的相互作用。结果表明,TAA小鼠胸主动脉铁下垂水平升高,METTL14在TAA模型和ike诱导的VSMCs中表达上调。敲低METTL14可通过减少VSMCs的铁下垂来抑制TAA的进展。此外,IGF2BP2识别mettl14修饰的ACSL4 mRNA并调节其稳定性,从而介导VSMCs的铁凋亡。总的来说,METTL14对VSMC铁下垂的作用为TAA的治疗提供了治疗潜力。
{"title":"METTL14 promotes ferroptosis in smooth muscle cells during thoracic aortic aneurysm by stabilizing the m<sup>6</sup>A modification of ACSL4.","authors":"Wenjun Wang, Jiayi Chen, Songqing Lai, Ruiyuan Zeng, Ming Fang, Li Wan, Yiying Li","doi":"10.1152/ajpcell.00577.2024","DOIUrl":"10.1152/ajpcell.00577.2024","url":null,"abstract":"<p><p>Thoracic aortic aneurysm (TAA) is a vascular disease associated with high mortality rates. Ferroptosis has been shown to mediate the transformation of vascular smooth muscle cells (VSMCs). However, the regulatory mechanisms by which ferroptosis influences TAA remain unclear. In this study, we induced TAA mouse models using angiotensin II (Ang II) and evaluated the impact of ferroptosis on the pathological changes observed in TAA mice, employing liproxstatin-1 as a treatment. In addition, we assessed the regulatory effect of METTL14 on the ferroptosis of VSMCs after treating them with a ferroptosis activator (imidazole ketone erastin, IKE). RNA binding protein immunoprecipitation (RIP) and RNA pull-down assays were conducted to investigate the interaction between acyl-CoA synthase long-chain family member 4 (<i>ACSL4</i>) mRNA and the proteins METTL14 or IGF2BP2. The results indicated that the level of ferroptosis was elevated in the thoracic aorta of TAA mice, and METTL14 was upregulated in TAA models and IKE-induced VSMCs. Knockdown of METTL14 was found to inhibit the progression of TAA by reducing the ferroptosis of VSMCs. Furthermore, IGF2BP2 recognized METTL14-modified <i>ACSL4</i> mRNA and regulated its stability, thereby mediating the ferroptosis of VSMCs. Collectively, the effects of METTL14 on VSMC ferroptosis present therapeutic potential for the treatment of TAA.<b>NEW & NOTEWORTHY</b> Our study confirmed that METTL14 can induce ferroptosis in vascular smooth muscle cells during the progression of thoracic aortic aneurysm by mediating the m<sup>6</sup>A modification of <i>ACSL4</i> mRNA.</p>","PeriodicalId":7585,"journal":{"name":"American journal of physiology. Cell physiology","volume":" ","pages":"C387-C399"},"PeriodicalIF":5.0,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142821824","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Gut-kidney interaction reinforces dapagliflozin-mediated alleviation in diabetic nephropathy. 肠肾相互作用加强了达格列净介导的糖尿病肾病缓解。
IF 5 2区 生物学 Q2 CELL BIOLOGY Pub Date : 2025-02-01 Epub Date: 2024-12-31 DOI: 10.1152/ajpcell.00651.2024
Yinhua Ni, Haimei Du, Lehui Ke, Liujie Zheng, Sujie Nan, Liyang Ni, Yuxiang Pan, Zhengwei Fu, Qiang He, Juan Jin

Intestinal microbiota are pathophysiologically involved in diabetic nephropathy (DN). Dapagliflozin, recognized for its blood glucose-lowering effect, has demonstrated efficacy in improving DN. However, the mechanisms beyond glycemic control that mediate the impact of dapagliflozin on DN remain unclear. Here, we investigated the effects of dapagliflozin on DN and gut microbiota, elucidating how it mitigates DN via the gut-kidney axis. Low-dose dapagliflozin markedly ameliorated renal inflammation and fibrosis and improved gut barrier function in high-fat diet (HFD)/streptozotocin (STZ)-induced DN mice and db/db mice without affecting blood glucose levels. These effects were associated with altered gut microbial composition and function. Eradication of the resident microbiota abolished the protective effects of dapagliflozin against kidney injury in DN mice. Moreover, dapagliflozin significantly altered microbial metabolites in DN mice, decreasing argininosuccinic acid (ASA) and palmitic acid (PA), while increasing S-allylcysteine (SAC) levels. ASA and PA increased the expression of renal inflammation- and fibrosis-related markers in HK-2 cells, whereas SAC ameliorated renal damage and altered the microbial composition in a manner similar to dapagliflozin in DN mice. Notably, Muribaculaceae and Desulfovibrionaceae were correlated with the alleviation of DN-associated renal dysfunction by low- and high-dose dapagliflozin treatments in DN mice. These findings demonstrate a potential application of dapagliflozin in managing DN by targeting the gut microbiota.NEW & NOTEWORTHY We demonstrated that dapagliflozin administration alleviated renal inflammation and fibrosis in vivo and in vitro, along with reshaping the gut microbiota composition and altering levels of key microbial metabolites, including argininosuccinic acid (ASA) and palmitic acid (PA), while increasing S-allylcysteine (SAC). Importantly, the genera Muribaculaceae and Desulfovibrionaceae emerged as pivotal microbial genera mediating the protective effects of dapagliflozin against diabetic nephropathy.

肠道微生物群与糖尿病肾病(DN)的病理生理相关。达格列净以其降血糖作用而闻名,已证明对DN有改善作用。然而,除血糖控制外,达格列净对DN的影响介导机制尚不清楚。在这里,我们研究了达格列净对DN和肠道微生物群的影响,阐明了它是如何通过肠肾轴减轻DN的。低剂量达格列净显著改善高脂饮食(HFD)/链脲佐菌素(STZ)诱导的DN小鼠和db/db小鼠的肾脏炎症和纤维化,改善肠道屏障功能,而不影响血糖水平。这些影响与肠道微生物组成和功能的改变有关。根除常驻微生物群可消除达格列净对DN小鼠肾损伤的保护作用。此外,达格列净显著改变了DN小鼠的微生物代谢物,降低了精氨酸琥珀酸(ASA)和棕榈酸(PA),同时增加了s -烯丙基半胱氨酸(SAC)水平。ASA和PA增加了HK-2细胞中肾脏炎症和纤维化相关标志物的表达,而SAC改善了肾损伤,并以类似于达格列净的方式改变了DN小鼠的微生物组成。值得注意的是,Muribaculaceae和Desulfovibrionaceae与低剂量和高剂量达格列净治疗DN小鼠DN相关性肾功能障碍的缓解相关。这些发现证明了达格列净通过靶向肠道微生物群来管理DN的潜在应用。
{"title":"Gut-kidney interaction reinforces dapagliflozin-mediated alleviation in diabetic nephropathy.","authors":"Yinhua Ni, Haimei Du, Lehui Ke, Liujie Zheng, Sujie Nan, Liyang Ni, Yuxiang Pan, Zhengwei Fu, Qiang He, Juan Jin","doi":"10.1152/ajpcell.00651.2024","DOIUrl":"10.1152/ajpcell.00651.2024","url":null,"abstract":"<p><p>Intestinal microbiota are pathophysiologically involved in diabetic nephropathy (DN). Dapagliflozin, recognized for its blood glucose-lowering effect, has demonstrated efficacy in improving DN. However, the mechanisms beyond glycemic control that mediate the impact of dapagliflozin on DN remain unclear. Here, we investigated the effects of dapagliflozin on DN and gut microbiota, elucidating how it mitigates DN via the gut-kidney axis. Low-dose dapagliflozin markedly ameliorated renal inflammation and fibrosis and improved gut barrier function in high-fat diet (HFD)/streptozotocin (STZ)-induced DN mice and <i>db</i>/<i>db</i> mice without affecting blood glucose levels. These effects were associated with altered gut microbial composition and function. Eradication of the resident microbiota abolished the protective effects of dapagliflozin against kidney injury in DN mice. Moreover, dapagliflozin significantly altered microbial metabolites in DN mice, decreasing argininosuccinic acid (ASA) and palmitic acid (PA), while increasing <i>S</i>-allylcysteine (SAC) levels. ASA and PA increased the expression of renal inflammation- and fibrosis-related markers in HK-2 cells, whereas SAC ameliorated renal damage and altered the microbial composition in a manner similar to dapagliflozin in DN mice. Notably, <i>Muribaculaceae</i> and <i>Desulfovibrionaceae</i> were correlated with the alleviation of DN-associated renal dysfunction by low- and high-dose dapagliflozin treatments in DN mice. These findings demonstrate a potential application of dapagliflozin in managing DN by targeting the gut microbiota.<b>NEW & NOTEWORTHY</b> We demonstrated that dapagliflozin administration alleviated renal inflammation and fibrosis in vivo and in vitro, along with reshaping the gut microbiota composition and altering levels of key microbial metabolites, including argininosuccinic acid (ASA) and palmitic acid (PA), while increasing <i>S</i>-allylcysteine (SAC). Importantly, the genera <i>Muribaculaceae</i> and <i>Desulfovibrionaceae</i> emerged as pivotal microbial genera mediating the protective effects of dapagliflozin against diabetic nephropathy.</p>","PeriodicalId":7585,"journal":{"name":"American journal of physiology. Cell physiology","volume":" ","pages":"C452-C466"},"PeriodicalIF":5.0,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142909085","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Sulforaphane treatment mimics contractile activity-induced mitochondrial adaptations in muscle myotubes. 草甘膦处理可模仿肌肉肌管中收缩活动诱导的线粒体适应性。
IF 5 2区 生物学 Q2 CELL BIOLOGY Pub Date : 2025-02-01 Epub Date: 2024-12-13 DOI: 10.1152/ajpcell.00669.2024
Sabrina Champsi, David A Hood

Mitochondria are metabolic hubs that govern skeletal muscle health. Although exercise has been established as a powerful inducer of quality control processes that ultimately enhance mitochondrial function, there are currently limited pharmaceutical interventions available that emulate exercise-induced mitochondrial adaptations. To investigate a novel candidate for this role, we examined sulforaphane (SFN), a naturally occurring compound found in cruciferous vegetables. SFN has been documented as a potent antioxidant inducer through its activation of the nuclear factor erythroid 2-related factor 2 (Nrf-2) antioxidant response pathway. However, its effects on muscle health have been underexplored. To investigate the interplay between chronic exercise and SFN, C2C12 myotubes were electrically stimulated to model chronic contractile activity (CCA) in the presence or absence of SFN. SFN promoted Nrf-2 nuclear translocation, enhanced mitochondrial respiration, and upregulated key antioxidant proteins including catalase and glutathione reductase. These adaptations were accompanied by reductions in cellular and mitochondrial reactive oxygen species (ROS) emission. Signaling toward biogenesis was enhanced, demonstrated by increases in mitochondrial transcription factor A (TFAM), peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1α nuclear translocation, PGC-1α promoter activity, mitochondrial content, and organelle branching, suggestive of a larger, more interconnected mitochondrial pool. These mitochondrial adaptations were accompanied by an increase in lysosomal proteins, suggesting coordinated regulation. There was no difference in mitochondrial and antioxidant-related proteins between CCA and non-CCA SFN-treated cells. Our data suggest that SFN activates signaling cascades that are common to those produced by contractile activity, indicating that SFN-centered therapeutic strategies may improve the mitochondrial phenotype in skeletal muscle.NEW & NOTEWORTHY Nrf-2 is a transcription factor that has been implicated in mitigating oxidative stress and regulating mitochondrial homeostasis. However, limited research has demonstrated how Nrf-2-mediated adaptations compare with those produced by exercise. To investigate this, we treated myotubes with Sulforaphane, a well-established Nrf-2 activator, and combined this with stimulation-induced chronic contractile activity to model exercise training. Our work is the first to establish that sulforaphane mimics training-induced mitochondrial adaptations, including enhancements in respiration, biogenesis, and dynamics.

线粒体是控制骨骼肌健康的代谢中枢。虽然运动已被确定为质量控制过程的强大诱导剂,最终增强线粒体功能,但目前可用于模拟运动诱导的线粒体适应的药物干预有限。为了研究这一作用的新候选物,我们研究了萝卜硫素(SFN),一种在十字花科蔬菜中发现的天然化合物。SFN通过激活核因子红系2相关因子2 (Nrf-2)抗氧化反应途径,已被证明是一种有效的抗氧化诱导剂。然而,它对肌肉健康的影响尚未得到充分研究。为了研究慢性运动与SFN之间的相互作用,C2C12肌管在SFN存在或不存在的情况下被电刺激来模拟慢性收缩活动(CCA)。SFN促进Nrf-2核易位,增强线粒体呼吸,上调过氧化氢酶和谷胱甘肽还原酶等关键抗氧化蛋白。这些适应伴随着细胞和线粒体ROS释放的减少。通过线粒体转录因子A (TFAM)、过氧化物酶体增殖体激活受体- γ辅助激活因子(PGC)-1α核易位、PGC-1α启动子活性、线粒体含量和细胞器分支的增加,证明了生物发生信号的增强,这表明线粒体池更大、更相互关联。这些线粒体适应伴随着溶酶体蛋白的增加,表明协调调节。在CCA和非CCA sfn处理的细胞中,线粒体和抗氧化相关蛋白没有差异。我们的数据表明,SFN激活了与收缩活动产生的信号级联反应,表明以SFN为中心的治疗策略可能改善骨骼肌的线粒体表型。
{"title":"Sulforaphane treatment mimics contractile activity-induced mitochondrial adaptations in muscle myotubes.","authors":"Sabrina Champsi, David A Hood","doi":"10.1152/ajpcell.00669.2024","DOIUrl":"10.1152/ajpcell.00669.2024","url":null,"abstract":"<p><p>Mitochondria are metabolic hubs that govern skeletal muscle health. Although exercise has been established as a powerful inducer of quality control processes that ultimately enhance mitochondrial function, there are currently limited pharmaceutical interventions available that emulate exercise-induced mitochondrial adaptations. To investigate a novel candidate for this role, we examined sulforaphane (SFN), a naturally occurring compound found in cruciferous vegetables. SFN has been documented as a potent antioxidant inducer through its activation of the nuclear factor erythroid 2-related factor 2 (Nrf-2) antioxidant response pathway. However, its effects on muscle health have been underexplored. To investigate the interplay between chronic exercise and SFN, C2C12 myotubes were electrically stimulated to model chronic contractile activity (CCA) in the presence or absence of SFN. SFN promoted Nrf-2 nuclear translocation, enhanced mitochondrial respiration, and upregulated key antioxidant proteins including catalase and glutathione reductase. These adaptations were accompanied by reductions in cellular and mitochondrial reactive oxygen species (ROS) emission. Signaling toward biogenesis was enhanced, demonstrated by increases in mitochondrial transcription factor A (TFAM), peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1α nuclear translocation, PGC-1α promoter activity, mitochondrial content, and organelle branching, suggestive of a larger, more interconnected mitochondrial pool. These mitochondrial adaptations were accompanied by an increase in lysosomal proteins, suggesting coordinated regulation. There was no difference in mitochondrial and antioxidant-related proteins between CCA and non-CCA SFN-treated cells. Our data suggest that SFN activates signaling cascades that are common to those produced by contractile activity, indicating that SFN-centered therapeutic strategies may improve the mitochondrial phenotype in skeletal muscle.<b>NEW & NOTEWORTHY</b> Nrf-2 is a transcription factor that has been implicated in mitigating oxidative stress and regulating mitochondrial homeostasis. However, limited research has demonstrated how Nrf-2-mediated adaptations compare with those produced by exercise. To investigate this, we treated myotubes with Sulforaphane, a well-established Nrf-2 activator, and combined this with stimulation-induced chronic contractile activity to model exercise training. Our work is the first to establish that sulforaphane mimics training-induced mitochondrial adaptations, including enhancements in respiration, biogenesis, and dynamics.</p>","PeriodicalId":7585,"journal":{"name":"American journal of physiology. Cell physiology","volume":" ","pages":"C335-C354"},"PeriodicalIF":5.0,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142821827","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","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
American journal of physiology. Cell physiology
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