Pub Date : 2024-10-01DOI: 10.1038/s12276-024-01323-x
Chris E. Cooper, Michelle Simons, Alex Dyson, Nélida Leiva Eriksson, Gary G. A. Silkstone, Natalie Syrett, Victoria Allen-Baume, Leif Bülow, Luca Ronda, Andrea Mozzarelli, Mervyn Singer, Brandon J. Reeder
The clinical utility of hemoglobin-based oxygen carriers (HBOC) is limited by adverse heme oxidative chemistry. A variety of tyrosine residues were inserted on the surface of the γ subunit of recombinant fetal hemoglobin to create novel electron transport pathways. This enhanced the ability of the physiological antioxidant ascorbate to reduce ferryl heme and decrease lipid peroxidation. The γL96Y mutation presented the best profile of oxidative protection unaccompanied by loss of protein stability and function. N-terminal deletions were constructed to facilitate the production of recombinant hemoglobin by fermentation and phenylalanine insertions in the heme pocket to decrease the rate of NO dioxygenation. The resultant mutant (αV1del. αL29F, γG1del. γV67F, γL96Y) significantly decreased NO scavenging and lipid peroxidation in vitro. Unlike native hemoglobin or a recombinant control (αV1del, γG1del), this mutation showed no increase in blood pressure immediately following infusion in a rat model of reperfusion injury, suggesting that it was also able to prevent NO scavenging in vivo. Infusion of the mutant also resulted in no meaningful adverse physiological effects apart from diuresis, and no increase in oxidative stress, as measured by urinary isoprostane levels. Following PEGylation via the Euro-PEG-Hb method to increase vascular retention, this novel protein construct was compared with saline in a severe rat reperfusion injury model (45% blood volume removal for 90 minutes followed by reinfusion to twice the volume of shed blood). Blood pressure and survival were followed for 4 h post-reperfusion. While there was no difference in blood pressure, the PEGylated Hb mutant significantly increased survival. Hemoglobin-based oxygen carriers are modified hemoglobin molecules that can be infused as blood substitutes to replace red blood cell transfusions or as oxygen therapeutics to deliver oxygen to damaged tissues not readily accessible by red cells. However, their clinical use has been limited by adverse side effects caused by free radical production and nitric oxide scavenging by extracellular hemoglobin. The researchers used genetic engineering to insert tyrosine residues into fetal human hemoglobin to decrease radical production and phenylalanine residues to decrease nitric oxide scavenging. The resulting novel hemoglobin was tested in rat models to observe the effects on blood pressure and survival rates. The research offers hope for improved treatment for patients in critical need of blood transfusions or with an otherwise compromised oxygen delivery system, such as in sickle cell disease, stroke or sepsis. This summary was written by the author.
基于血红蛋白的氧载体(HBOC)的临床应用受到血红素氧化化学性质不良的限制。在重组胎儿血红蛋白的γ亚基表面插入了多种酪氨酸残基,以创建新的电子传递途径。这增强了生理抗氧化剂抗坏血酸还原摆渡血红素和减少脂质过氧化的能力。γL96Y突变具有最佳的氧化保护能力,且不会伴随蛋白质稳定性和功能的丧失。为了便于通过发酵生产重组血红蛋白,对 N 端进行了缺失,并在血红素袋中插入了苯丙氨酸,以降低 NO 的二氧化速率。由此产生的突变体(αV1del. αL29F、γG1del. γV67F、γL96Y)显著降低了体外清除 NO 和脂质过氧化的能力。与原生血红蛋白或重组对照(αV1del、γG1del)不同,该突变体在大鼠再灌注损伤模型中输注后血压没有立即升高,这表明它在体内也能阻止 NO 清除。除了利尿之外,输注该突变体也不会导致明显的不良生理效应,而且根据尿液中异前列腺素水平的测量,也不会增加氧化应激。通过 Euro-PEG-Hb 方法进行 PEG 化以增加血管保留率后,在严重的大鼠再灌注损伤模型中将这种新型蛋白质构建物与生理盐水进行了比较(90 分钟内去除 45% 的血容量,然后再灌注两倍于脱落血容量的血液)。对再灌注后 4 小时的血压和存活率进行了跟踪。虽然血压没有差异,但 PEG 化 Hb 突变体显著提高了存活率。
{"title":"Taming hemoglobin chemistry—a new hemoglobin-based oxygen carrier engineered with both decreased rates of nitric oxide scavenging and lipid oxidation","authors":"Chris E. Cooper, Michelle Simons, Alex Dyson, Nélida Leiva Eriksson, Gary G. A. Silkstone, Natalie Syrett, Victoria Allen-Baume, Leif Bülow, Luca Ronda, Andrea Mozzarelli, Mervyn Singer, Brandon J. Reeder","doi":"10.1038/s12276-024-01323-x","DOIUrl":"10.1038/s12276-024-01323-x","url":null,"abstract":"The clinical utility of hemoglobin-based oxygen carriers (HBOC) is limited by adverse heme oxidative chemistry. A variety of tyrosine residues were inserted on the surface of the γ subunit of recombinant fetal hemoglobin to create novel electron transport pathways. This enhanced the ability of the physiological antioxidant ascorbate to reduce ferryl heme and decrease lipid peroxidation. The γL96Y mutation presented the best profile of oxidative protection unaccompanied by loss of protein stability and function. N-terminal deletions were constructed to facilitate the production of recombinant hemoglobin by fermentation and phenylalanine insertions in the heme pocket to decrease the rate of NO dioxygenation. The resultant mutant (αV1del. αL29F, γG1del. γV67F, γL96Y) significantly decreased NO scavenging and lipid peroxidation in vitro. Unlike native hemoglobin or a recombinant control (αV1del, γG1del), this mutation showed no increase in blood pressure immediately following infusion in a rat model of reperfusion injury, suggesting that it was also able to prevent NO scavenging in vivo. Infusion of the mutant also resulted in no meaningful adverse physiological effects apart from diuresis, and no increase in oxidative stress, as measured by urinary isoprostane levels. Following PEGylation via the Euro-PEG-Hb method to increase vascular retention, this novel protein construct was compared with saline in a severe rat reperfusion injury model (45% blood volume removal for 90 minutes followed by reinfusion to twice the volume of shed blood). Blood pressure and survival were followed for 4 h post-reperfusion. While there was no difference in blood pressure, the PEGylated Hb mutant significantly increased survival. Hemoglobin-based oxygen carriers are modified hemoglobin molecules that can be infused as blood substitutes to replace red blood cell transfusions or as oxygen therapeutics to deliver oxygen to damaged tissues not readily accessible by red cells. However, their clinical use has been limited by adverse side effects caused by free radical production and nitric oxide scavenging by extracellular hemoglobin. The researchers used genetic engineering to insert tyrosine residues into fetal human hemoglobin to decrease radical production and phenylalanine residues to decrease nitric oxide scavenging. The resulting novel hemoglobin was tested in rat models to observe the effects on blood pressure and survival rates. The research offers hope for improved treatment for patients in critical need of blood transfusions or with an otherwise compromised oxygen delivery system, such as in sickle cell disease, stroke or sepsis. This summary was written by the author.","PeriodicalId":50466,"journal":{"name":"Experimental and Molecular Medicine","volume":"56 10","pages":"2260-2270"},"PeriodicalIF":9.5,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s12276-024-01323-x.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142331625","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-01DOI: 10.1038/s12276-024-01312-0
Mahn Jae Lee, Jaehyeok Lee, Jeongmin Ha, Geon Kim, Hye-Jin Kim, Sumin Lee, Bon-Kyoung Koo, YongKeun Park
Organoids, which are miniature in vitro versions of organs, possess significant potential for studying human diseases and elucidating their underlying mechanisms. Live imaging techniques play a crucial role in organoid research and contribute to elucidating the complex structure and dynamic biological phenomena of organoids. However, live, unlabeled high-resolution imaging of native organoids is challenging, primarily owing to the complexities of sample handling and optical scattering inherent in three-dimensional (3D) structures. Additionally, conventional imaging methods fail to capture the real-time dynamic processes of growing organoids. In this study, we introduce low-coherence holotomography as an advanced, label-free, quantitative imaging modality designed to overcome several technical obstacles for long-term live imaging of 3D organoids. We demonstrate the efficacy of low-coherence holotomography by capturing high-resolution morphological details and dynamic activities within mouse small intestinal organoids at subcellular resolution. Moreover, our approach facilitates the distinction between viable and nonviable organoids, significantly enhancing its utility in organoid-based research. This advancement underscores the critical role of live imaging in organoid studies, offering a more comprehensive understanding of these complex systems. Organoids, miniature 3D structures that imitate real organs, are grown in labs to study human biology and diseases. However, their complex structures and behaviors are hard to understand due to imaging technology limitations. In this study, researchers used a method called low-coherence holotomography to study mouse small intestinal organoids. This method let them observe organoids’ growth and drug responses in real-time, without altering their natural state. They conducted an experiment involving over 120 hours of continuous imaging, providing new insights into organoid development, cell dynamics, and drug responses. The study shows that low-coherence HT can reveal detailed 3D structures and changes within organoids, such as cell division and death, with high resolution. This research could revolutionize drug development and testing and provide new insights into human biology and diseases. This summary was initially drafted using artificial intelligence, then revised and fact-checked by the author.
{"title":"Long-term three-dimensional high-resolution imaging of live unlabeled small intestinal organoids via low-coherence holotomography","authors":"Mahn Jae Lee, Jaehyeok Lee, Jeongmin Ha, Geon Kim, Hye-Jin Kim, Sumin Lee, Bon-Kyoung Koo, YongKeun Park","doi":"10.1038/s12276-024-01312-0","DOIUrl":"10.1038/s12276-024-01312-0","url":null,"abstract":"Organoids, which are miniature in vitro versions of organs, possess significant potential for studying human diseases and elucidating their underlying mechanisms. Live imaging techniques play a crucial role in organoid research and contribute to elucidating the complex structure and dynamic biological phenomena of organoids. However, live, unlabeled high-resolution imaging of native organoids is challenging, primarily owing to the complexities of sample handling and optical scattering inherent in three-dimensional (3D) structures. Additionally, conventional imaging methods fail to capture the real-time dynamic processes of growing organoids. In this study, we introduce low-coherence holotomography as an advanced, label-free, quantitative imaging modality designed to overcome several technical obstacles for long-term live imaging of 3D organoids. We demonstrate the efficacy of low-coherence holotomography by capturing high-resolution morphological details and dynamic activities within mouse small intestinal organoids at subcellular resolution. Moreover, our approach facilitates the distinction between viable and nonviable organoids, significantly enhancing its utility in organoid-based research. This advancement underscores the critical role of live imaging in organoid studies, offering a more comprehensive understanding of these complex systems. Organoids, miniature 3D structures that imitate real organs, are grown in labs to study human biology and diseases. However, their complex structures and behaviors are hard to understand due to imaging technology limitations. In this study, researchers used a method called low-coherence holotomography to study mouse small intestinal organoids. This method let them observe organoids’ growth and drug responses in real-time, without altering their natural state. They conducted an experiment involving over 120 hours of continuous imaging, providing new insights into organoid development, cell dynamics, and drug responses. The study shows that low-coherence HT can reveal detailed 3D structures and changes within organoids, such as cell division and death, with high resolution. This research could revolutionize drug development and testing and provide new insights into human biology and diseases. This summary was initially drafted using artificial intelligence, then revised and fact-checked by the author.","PeriodicalId":50466,"journal":{"name":"Experimental and Molecular Medicine","volume":"56 10","pages":"2162-2170"},"PeriodicalIF":9.5,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s12276-024-01312-0.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142331621","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-01DOI: 10.1038/s12276-024-01322-y
Duk-Hwa Kwon, Sera Shin, Yoon Seok Nam, Nakwon Choe, Yongwoon Lim, Anna Jeong, Yun-Gyeong Lee, Young-Kook Kim, Hyun Kook
Vascular calcification (VC) refers to the accumulation of mineral deposits on the walls of arteries and veins, and it is closely associated with increased mortality in cardiovascular disease patients, particularly among high-risk patients with diabetes and chronic kidney disease (CKD). Neuronal precursor cell-expressed developmentally downregulated protein 8 (NEDD8) is a ubiquitin-like protein that plays a pivotal role in various cellular functions, primarily through its conjugation to target proteins and subsequent relay of biological signals. However, the role of NEDDylation in VC has not been investigated. In our study, we observed that MLN4924, an inhibitor of the NEDD8-activating E1 enzyme, effectively impedes the progression of VC. LC‒MS/MS analysis revealed that poly(ADP‒ribose) polymerase 1 (PARP-1) is subjected to NEDD8 conjugation, leading to an increase in PARP-1 activity during VC. We subsequently revealed that PARP-1 NEDDylation is mediated by the E3 ligase CBL proto-oncogene B (CBL-b) and is reversed by NEDD8-specific protease 1 (NEDP-1) during VC. Furthermore, the CBL-b C373 peptide effectively mitigated the inactive form of the E3 ligase activity of CBL-b, ultimately preventing VC. These findings provide compelling evidence that the NEDD8-dependent activation of PARP-1 represents a novel mechanism underlying vascular calcification and suggests a promising new therapeutic target for VC. Vascular calcification, a condition where calcium deposits in blood vessels, can increase heart disease risk. Researchers studied a process called neddylation, which modifies proteins, to see if it affects this calcium buildup. They experimented on cells and mice using various techniques like chemical treatments and gene alteration. They found that a protein, NEDD8, when joined to another protein, PARP-1, encourages calcium deposition in blood vessels. By preventing NEDD8 from joining to PARP-1 with a specific inhibitor, they reduced calcium buildup in cells and mice. They also found that an enzyme, Cbl-b, helps NEDD8 attach to PARP-1, suggesting a new way to prevent vascular calcification. Results suggest that stopping neddylation, particularly the joining of NEDD8 to PARP-1, can significantly reduce vascular calcification. This discovery could lead to new treatments for vascular calcification and related heart conditions. This summary was initially drafted using artificial intelligence, then revised and fact-checked by the author.
{"title":"CBL-b E3 ligase-mediated neddylation and activation of PARP-1 induce vascular calcification","authors":"Duk-Hwa Kwon, Sera Shin, Yoon Seok Nam, Nakwon Choe, Yongwoon Lim, Anna Jeong, Yun-Gyeong Lee, Young-Kook Kim, Hyun Kook","doi":"10.1038/s12276-024-01322-y","DOIUrl":"10.1038/s12276-024-01322-y","url":null,"abstract":"Vascular calcification (VC) refers to the accumulation of mineral deposits on the walls of arteries and veins, and it is closely associated with increased mortality in cardiovascular disease patients, particularly among high-risk patients with diabetes and chronic kidney disease (CKD). Neuronal precursor cell-expressed developmentally downregulated protein 8 (NEDD8) is a ubiquitin-like protein that plays a pivotal role in various cellular functions, primarily through its conjugation to target proteins and subsequent relay of biological signals. However, the role of NEDDylation in VC has not been investigated. In our study, we observed that MLN4924, an inhibitor of the NEDD8-activating E1 enzyme, effectively impedes the progression of VC. LC‒MS/MS analysis revealed that poly(ADP‒ribose) polymerase 1 (PARP-1) is subjected to NEDD8 conjugation, leading to an increase in PARP-1 activity during VC. We subsequently revealed that PARP-1 NEDDylation is mediated by the E3 ligase CBL proto-oncogene B (CBL-b) and is reversed by NEDD8-specific protease 1 (NEDP-1) during VC. Furthermore, the CBL-b C373 peptide effectively mitigated the inactive form of the E3 ligase activity of CBL-b, ultimately preventing VC. These findings provide compelling evidence that the NEDD8-dependent activation of PARP-1 represents a novel mechanism underlying vascular calcification and suggests a promising new therapeutic target for VC. Vascular calcification, a condition where calcium deposits in blood vessels, can increase heart disease risk. Researchers studied a process called neddylation, which modifies proteins, to see if it affects this calcium buildup. They experimented on cells and mice using various techniques like chemical treatments and gene alteration. They found that a protein, NEDD8, when joined to another protein, PARP-1, encourages calcium deposition in blood vessels. By preventing NEDD8 from joining to PARP-1 with a specific inhibitor, they reduced calcium buildup in cells and mice. They also found that an enzyme, Cbl-b, helps NEDD8 attach to PARP-1, suggesting a new way to prevent vascular calcification. Results suggest that stopping neddylation, particularly the joining of NEDD8 to PARP-1, can significantly reduce vascular calcification. This discovery could lead to new treatments for vascular calcification and related heart conditions. This summary was initially drafted using artificial intelligence, then revised and fact-checked by the author.","PeriodicalId":50466,"journal":{"name":"Experimental and Molecular Medicine","volume":"56 10","pages":"2246-2259"},"PeriodicalIF":9.5,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s12276-024-01322-y.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142331615","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-01DOI: 10.1038/s12276-024-01314-y
Dashdulam Davaanyam, Song-I Seol, Sang-A Oh, Hahnbi Lee, Ja-Kyeong Lee
We previously reported that high mobility group box 1 (HMGB1), a danger-associated molecular pattern (DAMP), increases intracellular iron levels in the postischemic brain by upregulating hepcidin, a key regulator of iron homeostasis, triggering ferroptosis. Since hepatocytes are the primary cells that produce hepcidin and control systemic iron levels, we investigated whether cerebral ischemia induces hepcidin upregulation in hepatocytes. Following middle cerebral artery occlusion (MCAO) in a rodent model, significant liver injury was observed. This injury was evidenced by significantly elevated Eckhoff’s scores and increased serum levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST). Additionally, total iron levels were significantly elevated in the liver, with intracellular iron accumulation detected in hepatocytes. Hepcidin expression in the liver, which is primarily localized in hepatocytes, increased significantly starting at 3 h after MCAO and continued to increase rapidly, reaching a peak at 24 h. Interestingly, HMGB1 levels in the liver were also significantly elevated after MCAO, with the disulfide form of HMGB1 being the major subtype. In vitro experiments using AML12 hepatocytes showed that recombinant disulfide HMGB1 significantly upregulated hepcidin expression in a Toll-like receptor 4 (TLR4)- and RAGE-dependent manner. Furthermore, treatment with a ROS scavenger and a peptide HMGB1 antagonist revealed that both ROS generation and HMGB1 induction contributed to hepatocyte activation and liver damage following MCAO–reperfusion. In conclusion, this study revealed that cerebral ischemia triggers hepatocyte activation and liver injury. HMGB1 potently induces hepcidin not only in the brain but also in the liver, thereby influencing systemic iron homeostasis following ischemic stroke. Iron is vital for many body functions, but its quantity needs careful monitoring to avoid harm. The processes controlling iron, particularly after brain injuries like strokes, are not entirely known. Researchers studied how strokes affect liver function and iron control. They experimented on rats, causing strokes and then observing the impact on liver damage, iron quantities, and the production of hepcidin, a hormone crucial for iron control. This research used animal models to better comprehend these biological processes. The findings reveal that strokes can harm the liver and disrupt the body’s iron control by increasing hepcidin quantities. The researchers conclude that understanding these processes could help create treatments to manage iron quantities after a stroke, potentially improving stroke patients. This summary was initially drafted using artificial intelligence, then revised and fact-checked by the author.
{"title":"Hepatocyte activation and liver injury following cerebral ischemia promote HMGB1-mediated hepcidin upregulation in hepatocytes and regulation of systemic iron levels","authors":"Dashdulam Davaanyam, Song-I Seol, Sang-A Oh, Hahnbi Lee, Ja-Kyeong Lee","doi":"10.1038/s12276-024-01314-y","DOIUrl":"10.1038/s12276-024-01314-y","url":null,"abstract":"We previously reported that high mobility group box 1 (HMGB1), a danger-associated molecular pattern (DAMP), increases intracellular iron levels in the postischemic brain by upregulating hepcidin, a key regulator of iron homeostasis, triggering ferroptosis. Since hepatocytes are the primary cells that produce hepcidin and control systemic iron levels, we investigated whether cerebral ischemia induces hepcidin upregulation in hepatocytes. Following middle cerebral artery occlusion (MCAO) in a rodent model, significant liver injury was observed. This injury was evidenced by significantly elevated Eckhoff’s scores and increased serum levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST). Additionally, total iron levels were significantly elevated in the liver, with intracellular iron accumulation detected in hepatocytes. Hepcidin expression in the liver, which is primarily localized in hepatocytes, increased significantly starting at 3 h after MCAO and continued to increase rapidly, reaching a peak at 24 h. Interestingly, HMGB1 levels in the liver were also significantly elevated after MCAO, with the disulfide form of HMGB1 being the major subtype. In vitro experiments using AML12 hepatocytes showed that recombinant disulfide HMGB1 significantly upregulated hepcidin expression in a Toll-like receptor 4 (TLR4)- and RAGE-dependent manner. Furthermore, treatment with a ROS scavenger and a peptide HMGB1 antagonist revealed that both ROS generation and HMGB1 induction contributed to hepatocyte activation and liver damage following MCAO–reperfusion. In conclusion, this study revealed that cerebral ischemia triggers hepatocyte activation and liver injury. HMGB1 potently induces hepcidin not only in the brain but also in the liver, thereby influencing systemic iron homeostasis following ischemic stroke. Iron is vital for many body functions, but its quantity needs careful monitoring to avoid harm. The processes controlling iron, particularly after brain injuries like strokes, are not entirely known. Researchers studied how strokes affect liver function and iron control. They experimented on rats, causing strokes and then observing the impact on liver damage, iron quantities, and the production of hepcidin, a hormone crucial for iron control. This research used animal models to better comprehend these biological processes. The findings reveal that strokes can harm the liver and disrupt the body’s iron control by increasing hepcidin quantities. The researchers conclude that understanding these processes could help create treatments to manage iron quantities after a stroke, potentially improving stroke patients. This summary was initially drafted using artificial intelligence, then revised and fact-checked by the author.","PeriodicalId":50466,"journal":{"name":"Experimental and Molecular Medicine","volume":"56 10","pages":"2171-2183"},"PeriodicalIF":9.5,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s12276-024-01314-y.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142331620","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-01DOI: 10.1038/s12276-024-01289-w
Jiao Sun, Jin-Young Kim, Semo Jun, Meeyeon Park, Ebbing de Jong, Jae-Woong Chang, Sze Cheng, Deliang Fan, Yue Chen, Timothy J. Griffin, Jung-Hee Lee, Ho Jin You, Wei Zhang, Jeongsik Yong
Alternative cleavage and polyadenylation within introns (intronic APA) generate shorter mRNA isoforms; however, their physiological significance remains elusive. In this study, we developed a comprehensive workflow to analyze intronic APA profiles using the mammalian target of rapamycin (mTOR)-regulated transcriptome as a model system. Our investigation revealed two contrasting effects within the transcriptome in response to fluctuations in cellular mTOR activity: an increase in intronic APA for a subset of genes and a decrease for another subset of genes. The application of this workflow to RNA-seq data from The Cancer Genome Atlas demonstrated that this dichotomous intronic APA pattern is a consistent feature in transcriptomes across both normal tissues and various cancer types. Notably, our analyses of protein length changes resulting from intronic APA events revealed two distinct phenomena in proteome programming: a loss of functional domains due to significant changes in protein length or minimal alterations in C-terminal protein sequences within unstructured regions. Focusing on conserved intronic APA events across 10 different cancer types highlighted the prevalence of the latter cases in cancer transcriptomes, whereas the former cases were relatively enriched in normal tissue transcriptomes. These observations suggest potential, yet distinct, roles for intronic APA events during pathogenic processes and emphasize the abundance of protein isoforms with similar lengths in the cancer proteome. Furthermore, our investigation into the isoform-specific functions of JMJD6 intronic APA events supported the hypothesis that alterations in unstructured C-terminal protein regions lead to functional differences. Collectively, our findings underscore intronic APA events as a discrete molecular signature present in both normal tissues and cancer transcriptomes, highlighting the contribution of APA to the multifaceted functionality of the cancer proteome. Understanding our genes is vital for combating diseases like cancer. A crucial gene expression process is alternative polyadenylation. These versions can influence cell behavior and are associated with various diseases, including cancer. The role of a specific APA type, intronic APA, in cancer was unclear. This study examined intronic APA’s effect on cancer by analyzing cancer patient data. They found that intronic APA profiles vary greatly between normal and tumor tissues across different cancer types, indicating that intronic APA plays a complex role in cancer biology. The results showed that intronic APA contributes to the diversity of mRNA endings in cancer, affecting gene expression. This could lead to new diagnosis or treatment approaches. The researchers concluded that intronic APA is a key factor in cancer’s molecular landscape, providing new insights into cancer development and progression. This summary was initially drafted using artificial intelligence, then revised and fact-checked by the au
内含子内的替代性裂解和多腺苷酸化(内含子 APA)会产生较短的 mRNA 异构体;然而,它们的生理意义仍然难以捉摸。在这项研究中,我们以哺乳动物雷帕霉素靶标(mTOR)调控的转录组为模型系统,开发了一套全面的工作流程来分析内含子 APA 图谱。我们的研究揭示了转录组对细胞 mTOR 活性波动的两种截然不同的反应:一部分基因的内含子 APA 增加,另一部分基因的内含子 APA 减少。将这一工作流程应用于癌症基因组图谱的 RNA-seq 数据表明,这种二分的内含子 APA 模式是正常组织和各种癌症类型转录组的一致特征。值得注意的是,我们对内含子 APA 事件导致的蛋白质长度变化的分析表明,在蛋白质组编程中存在两种截然不同的现象:由于蛋白质长度的显著变化而导致功能域的缺失,或者在非结构化区域内 C 端蛋白质序列发生极小的变化。重点研究 10 种不同癌症类型中的保守内含子 APA 事件突出表明,后一种情况在癌症转录组中普遍存在,而前一种情况在正常组织转录组中相对较多。这些观察结果表明,内含子 APA 事件在致病过程中具有潜在但不同的作用,并强调了癌症蛋白质组中具有相似长度的蛋白质同工型的丰富性。此外,我们对 JMJD6 内含子 APA 事件的异构体特异性功能的研究支持了非结构化 C 端蛋白区域的改变导致功能差异的假设。总之,我们的研究结果强调了内含子 APA 事件是存在于正常组织和癌症转录组中的一种离散分子特征,突出了 APA 对癌症蛋白质组多方面功能的贡献。
{"title":"Dichotomous intronic polyadenylation profiles reveal multifaceted gene functions in the pan-cancer transcriptome","authors":"Jiao Sun, Jin-Young Kim, Semo Jun, Meeyeon Park, Ebbing de Jong, Jae-Woong Chang, Sze Cheng, Deliang Fan, Yue Chen, Timothy J. Griffin, Jung-Hee Lee, Ho Jin You, Wei Zhang, Jeongsik Yong","doi":"10.1038/s12276-024-01289-w","DOIUrl":"10.1038/s12276-024-01289-w","url":null,"abstract":"Alternative cleavage and polyadenylation within introns (intronic APA) generate shorter mRNA isoforms; however, their physiological significance remains elusive. In this study, we developed a comprehensive workflow to analyze intronic APA profiles using the mammalian target of rapamycin (mTOR)-regulated transcriptome as a model system. Our investigation revealed two contrasting effects within the transcriptome in response to fluctuations in cellular mTOR activity: an increase in intronic APA for a subset of genes and a decrease for another subset of genes. The application of this workflow to RNA-seq data from The Cancer Genome Atlas demonstrated that this dichotomous intronic APA pattern is a consistent feature in transcriptomes across both normal tissues and various cancer types. Notably, our analyses of protein length changes resulting from intronic APA events revealed two distinct phenomena in proteome programming: a loss of functional domains due to significant changes in protein length or minimal alterations in C-terminal protein sequences within unstructured regions. Focusing on conserved intronic APA events across 10 different cancer types highlighted the prevalence of the latter cases in cancer transcriptomes, whereas the former cases were relatively enriched in normal tissue transcriptomes. These observations suggest potential, yet distinct, roles for intronic APA events during pathogenic processes and emphasize the abundance of protein isoforms with similar lengths in the cancer proteome. Furthermore, our investigation into the isoform-specific functions of JMJD6 intronic APA events supported the hypothesis that alterations in unstructured C-terminal protein regions lead to functional differences. Collectively, our findings underscore intronic APA events as a discrete molecular signature present in both normal tissues and cancer transcriptomes, highlighting the contribution of APA to the multifaceted functionality of the cancer proteome. Understanding our genes is vital for combating diseases like cancer. A crucial gene expression process is alternative polyadenylation. These versions can influence cell behavior and are associated with various diseases, including cancer. The role of a specific APA type, intronic APA, in cancer was unclear. This study examined intronic APA’s effect on cancer by analyzing cancer patient data. They found that intronic APA profiles vary greatly between normal and tumor tissues across different cancer types, indicating that intronic APA plays a complex role in cancer biology. The results showed that intronic APA contributes to the diversity of mRNA endings in cancer, affecting gene expression. This could lead to new diagnosis or treatment approaches. The researchers concluded that intronic APA is a key factor in cancer’s molecular landscape, providing new insights into cancer development and progression. This summary was initially drafted using artificial intelligence, then revised and fact-checked by the au","PeriodicalId":50466,"journal":{"name":"Experimental and Molecular Medicine","volume":"56 10","pages":"2145-2161"},"PeriodicalIF":9.5,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s12276-024-01289-w.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142331617","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-01DOI: 10.1038/s12276-024-01311-1
Hye Ji Cha
Erythropoiesis, the process underlying the production of red blood cells, which are essential for oxygen transport, involves the development of hematopoietic stem cells into mature red blood cells. This review focuses on the critical roles of transcription factors and epigenetic mechanisms in modulating gene expression critical for erythroid differentiation. It emphasizes the significance of chromatin remodeling in ensuring gene accessibility, a key factor for the orderly progression of erythropoiesis. This review also discusses how dysregulation of these processes can lead to erythroid disorders and examines the promise of genome editing and gene therapy as innovative therapeutic approaches. By shedding light on the genomic regulation of erythropoiesis, this review suggests avenues for novel treatments for hematological conditions, underscoring the need for continued molecular studies to improve human health. Understanding how our bodies produce red blood cells, a process called erythropoiesis, is vital for treating blood disorders. Researchers have reviewed the stages of erythropoiesis, focusing on the genetic and molecular factors that influence the development of red blood cells. Erythropoiesis starts with stem cells in the bone marrow, which transform into several stages of red blood cell precursors before becoming mature, oxygen-carrying cells. The review emphasizes the role of specific growth factors, like erythropoietin, and transcription factors, proteins that activate or deactivate specific genes, in guiding these developmental stages. It also discusses how manipulating these factors in the lab can produce red blood cells outside of the body, a technique that could have therapeutic uses. By targeting the specific genes and pathways involved in red blood cell development, researchers can develop more effective treatments. This summary was initially drafted using artificial intelligence, then revised and fact-checked by the author.
{"title":"Erythropoiesis: insights from a genomic perspective","authors":"Hye Ji Cha","doi":"10.1038/s12276-024-01311-1","DOIUrl":"10.1038/s12276-024-01311-1","url":null,"abstract":"Erythropoiesis, the process underlying the production of red blood cells, which are essential for oxygen transport, involves the development of hematopoietic stem cells into mature red blood cells. This review focuses on the critical roles of transcription factors and epigenetic mechanisms in modulating gene expression critical for erythroid differentiation. It emphasizes the significance of chromatin remodeling in ensuring gene accessibility, a key factor for the orderly progression of erythropoiesis. This review also discusses how dysregulation of these processes can lead to erythroid disorders and examines the promise of genome editing and gene therapy as innovative therapeutic approaches. By shedding light on the genomic regulation of erythropoiesis, this review suggests avenues for novel treatments for hematological conditions, underscoring the need for continued molecular studies to improve human health. Understanding how our bodies produce red blood cells, a process called erythropoiesis, is vital for treating blood disorders. Researchers have reviewed the stages of erythropoiesis, focusing on the genetic and molecular factors that influence the development of red blood cells. Erythropoiesis starts with stem cells in the bone marrow, which transform into several stages of red blood cell precursors before becoming mature, oxygen-carrying cells. The review emphasizes the role of specific growth factors, like erythropoietin, and transcription factors, proteins that activate or deactivate specific genes, in guiding these developmental stages. It also discusses how manipulating these factors in the lab can produce red blood cells outside of the body, a technique that could have therapeutic uses. By targeting the specific genes and pathways involved in red blood cell development, researchers can develop more effective treatments. This summary was initially drafted using artificial intelligence, then revised and fact-checked by the author.","PeriodicalId":50466,"journal":{"name":"Experimental and Molecular Medicine","volume":"56 10","pages":"2099-2104"},"PeriodicalIF":9.5,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s12276-024-01311-1.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142331618","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-19DOI: 10.1038/s12276-024-01320-0
Hyondeog Kim, Wonyeop Lee, Youngwook Kim, Sang-Jin Lee, Wonyoung Choi, Geon Kook Lee, Seung-Jin Park, Shinyeong Ju, Seon-Young Kim, Cheolju Lee, Ji-Youn Han
Patients with lung adenocarcinoma who have never smoked (NSLA) and lack key driver mutations, such as those in the EGFR and ALK genes, face limited options for targeted therapies. They also tend to have poorer outcomes with immune checkpoint inhibitors than lung cancer patients who have a history of smoking. The proteogenomic profile of nonsmoking lung adenocarcinoma patients without these oncogenic driver mutations is poorly understood, which complicates the precise molecular classification of these cancers and highlights a significant area of unmet clinical need. This study analyzed the genome, transcriptome, and LC‒MS/MS-TMT-driven proteome data of tumors obtained from 99 Korean never-smoker lung adenocarcinoma patients. NSLA tumors without EGFR or ALK driver oncogenes were classified into four proteogenomic subgroups: proliferation, angiogenesis, immune, and metabolism subgroups. These 4 molecular subgroups were strongly associated with distinct clinical outcomes. The proliferation and angiogenesis subtypes were associated with a poorer prognosis, while the immune subtype was associated with the most favorable outcome, which was validated in an external lung cancer dataset. Genomic-wide impacts were analyzed, and significant correlations were found between copy number alterations and both the transcriptome and proteome for several genes, with enrichment in the ERBB, neurotrophin, insulin, and MAPK signaling pathways. Proteogenomic analyses suggested several targetable genes and proteins, including CDKs and ATR, as potential therapeutic targets in the proliferation subgroup. Upregulated cytokines, such as CCL5 and CXCL13, in the immune subgroup may serve as potential targets for combination immunotherapy. Our comprehensive proteogenomic analysis revealed the molecular subtypes of EGFR- and ALK-wild-type NSLA with significant unmet clinical needs. Lung cancer is the leading cause of cancer deaths worldwide, with increasing cases in non-smokers, particularly Asian women. This research investigates lung adenocarcinoma in non-smokers who don’t have common genetic changes, using a multi-omics approach. The study involved 99 patients, specifically those without typical EGFR or ALK mutations, to better understand the disease at a molecular level and find new treatments. The study shows the variety within non-smoker lung cancers and suggests that different groups may need specific treatments. Understanding the molecular types of lung adenocarcinoma in non-smokers can lead to better, personalized treatments and improved health outcomes. This research could lead to more effective treatments for non-smoker lung cancer, potentially improving survival and quality of life for this growing patient group. This summary was initially drafted using artificial intelligence, then revised and fact-checked by the author.
{"title":"Proteogenomic characterization identifies clinical subgroups in EGFR and ALK wild-type never-smoker lung adenocarcinoma","authors":"Hyondeog Kim, Wonyeop Lee, Youngwook Kim, Sang-Jin Lee, Wonyoung Choi, Geon Kook Lee, Seung-Jin Park, Shinyeong Ju, Seon-Young Kim, Cheolju Lee, Ji-Youn Han","doi":"10.1038/s12276-024-01320-0","DOIUrl":"10.1038/s12276-024-01320-0","url":null,"abstract":"Patients with lung adenocarcinoma who have never smoked (NSLA) and lack key driver mutations, such as those in the EGFR and ALK genes, face limited options for targeted therapies. They also tend to have poorer outcomes with immune checkpoint inhibitors than lung cancer patients who have a history of smoking. The proteogenomic profile of nonsmoking lung adenocarcinoma patients without these oncogenic driver mutations is poorly understood, which complicates the precise molecular classification of these cancers and highlights a significant area of unmet clinical need. This study analyzed the genome, transcriptome, and LC‒MS/MS-TMT-driven proteome data of tumors obtained from 99 Korean never-smoker lung adenocarcinoma patients. NSLA tumors without EGFR or ALK driver oncogenes were classified into four proteogenomic subgroups: proliferation, angiogenesis, immune, and metabolism subgroups. These 4 molecular subgroups were strongly associated with distinct clinical outcomes. The proliferation and angiogenesis subtypes were associated with a poorer prognosis, while the immune subtype was associated with the most favorable outcome, which was validated in an external lung cancer dataset. Genomic-wide impacts were analyzed, and significant correlations were found between copy number alterations and both the transcriptome and proteome for several genes, with enrichment in the ERBB, neurotrophin, insulin, and MAPK signaling pathways. Proteogenomic analyses suggested several targetable genes and proteins, including CDKs and ATR, as potential therapeutic targets in the proliferation subgroup. Upregulated cytokines, such as CCL5 and CXCL13, in the immune subgroup may serve as potential targets for combination immunotherapy. Our comprehensive proteogenomic analysis revealed the molecular subtypes of EGFR- and ALK-wild-type NSLA with significant unmet clinical needs. Lung cancer is the leading cause of cancer deaths worldwide, with increasing cases in non-smokers, particularly Asian women. This research investigates lung adenocarcinoma in non-smokers who don’t have common genetic changes, using a multi-omics approach. The study involved 99 patients, specifically those without typical EGFR or ALK mutations, to better understand the disease at a molecular level and find new treatments. The study shows the variety within non-smoker lung cancers and suggests that different groups may need specific treatments. Understanding the molecular types of lung adenocarcinoma in non-smokers can lead to better, personalized treatments and improved health outcomes. This research could lead to more effective treatments for non-smoker lung cancer, potentially improving survival and quality of life for this growing patient group. This summary was initially drafted using artificial intelligence, then revised and fact-checked by the author.","PeriodicalId":50466,"journal":{"name":"Experimental and Molecular Medicine","volume":"56 9","pages":"2082-2095"},"PeriodicalIF":9.5,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s12276-024-01320-0.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142267994","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-18DOI: 10.1038/s12276-024-01300-4
Ji Min Park, Yen-Hao Su, Chi-Shuan Fan, Hsin-Hua Chen, Yuan-Kai Qiu, Li-Li Chen, Hsin-An Chen, Thamil Selvee Ramasamy, Jung-Su Chang, Shih-Yi Huang, Wun-Shaing Wayne Chang, Alan Yueh-Luen Lee, Tze-Sing Huang, Cheng-Chin Kuo, Ching-Feng Chiu
Ferritin, comprising heavy (FTH1) and light (FTL) chains, is the main iron storage protein, and pancreatic cancer patients exhibit elevated serum ferritin levels. Specifically, higher ferritin levels are correlated with poorer pancreatic ductal adenocarcinoma (PDAC) prognosis; however, the underlying mechanism and metabolic programming of ferritin involved in KRAS-mutant PDAC progression remain unclear. Here, we observed a direct correlation between FTH1 expression and cell viability and clonogenicity in KRAS-mutant PDAC cell lines as well as with in vivo tumor growth through the control of proline metabolism. Our investigation highlights the intricate relationship between FTH1 and pyrroline-5-carboxylate reductase 1 (PYCR1), a crucial mitochondrial enzyme facilitating the glutamate-to-proline conversion, underscoring its impact on proline metabolic imbalance in KRAS-mutant PDAC. This regulation is further reversed by miR-5000-3p, whose dysregulation results in the disruption of proline metabolism, thereby accentuating the progression of KRAS-mutant PDAC. Additionally, our study demonstrated that deferasirox, an oral iron chelator, significantly diminishes cell viability and tumor growth in KRAS-mutant PDAC by targeting FTH1-mediated pathways and altering the PYCR1/PRODH expression ratio. These findings underscore the novel role of FTH1 in proline metabolism and its potential as a target for PDAC therapy development. Iron storage in our body is mainly controlled by a protein named ferritin, which reflects the amount of stored iron through its blood levels. Low ferritin levels usually suggest iron-deficiency anemia, while high levels can indicate inflammation and hinting at ferritin’s potential as a cancer indicator. In this study, scientists focused on pancreatic cancer, notorious for its low survival rates and limited treatment options. They examined the expression of different ferritin components and their link with the KRAS mutation, a common characteristic in pancreatic cancer that promotes tumor growth. The main discovery is that high FTH1 expression is associated with worse survival in pancreatic cancer patients, suggesting that targeting FTH1 could be a promising treatment for this aggressive cancer. This study enhances our knowledge of the molecular processes driving pancreatic cancer and opens new paths for targeted treatments. This summary was initially drafted using artificial intelligence, then revised and fact-checked by the author.
{"title":"Crosstalk between FTH1 and PYCR1 dysregulates proline metabolism and mediates cell growth in KRAS-mutant pancreatic cancer cells","authors":"Ji Min Park, Yen-Hao Su, Chi-Shuan Fan, Hsin-Hua Chen, Yuan-Kai Qiu, Li-Li Chen, Hsin-An Chen, Thamil Selvee Ramasamy, Jung-Su Chang, Shih-Yi Huang, Wun-Shaing Wayne Chang, Alan Yueh-Luen Lee, Tze-Sing Huang, Cheng-Chin Kuo, Ching-Feng Chiu","doi":"10.1038/s12276-024-01300-4","DOIUrl":"10.1038/s12276-024-01300-4","url":null,"abstract":"Ferritin, comprising heavy (FTH1) and light (FTL) chains, is the main iron storage protein, and pancreatic cancer patients exhibit elevated serum ferritin levels. Specifically, higher ferritin levels are correlated with poorer pancreatic ductal adenocarcinoma (PDAC) prognosis; however, the underlying mechanism and metabolic programming of ferritin involved in KRAS-mutant PDAC progression remain unclear. Here, we observed a direct correlation between FTH1 expression and cell viability and clonogenicity in KRAS-mutant PDAC cell lines as well as with in vivo tumor growth through the control of proline metabolism. Our investigation highlights the intricate relationship between FTH1 and pyrroline-5-carboxylate reductase 1 (PYCR1), a crucial mitochondrial enzyme facilitating the glutamate-to-proline conversion, underscoring its impact on proline metabolic imbalance in KRAS-mutant PDAC. This regulation is further reversed by miR-5000-3p, whose dysregulation results in the disruption of proline metabolism, thereby accentuating the progression of KRAS-mutant PDAC. Additionally, our study demonstrated that deferasirox, an oral iron chelator, significantly diminishes cell viability and tumor growth in KRAS-mutant PDAC by targeting FTH1-mediated pathways and altering the PYCR1/PRODH expression ratio. These findings underscore the novel role of FTH1 in proline metabolism and its potential as a target for PDAC therapy development. Iron storage in our body is mainly controlled by a protein named ferritin, which reflects the amount of stored iron through its blood levels. Low ferritin levels usually suggest iron-deficiency anemia, while high levels can indicate inflammation and hinting at ferritin’s potential as a cancer indicator. In this study, scientists focused on pancreatic cancer, notorious for its low survival rates and limited treatment options. They examined the expression of different ferritin components and their link with the KRAS mutation, a common characteristic in pancreatic cancer that promotes tumor growth. The main discovery is that high FTH1 expression is associated with worse survival in pancreatic cancer patients, suggesting that targeting FTH1 could be a promising treatment for this aggressive cancer. This study enhances our knowledge of the molecular processes driving pancreatic cancer and opens new paths for targeted treatments. This summary was initially drafted using artificial intelligence, then revised and fact-checked by the author.","PeriodicalId":50466,"journal":{"name":"Experimental and Molecular Medicine","volume":"56 9","pages":"2065-2081"},"PeriodicalIF":9.5,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s12276-024-01300-4.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142267995","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-12DOI: 10.1038/s12276-024-01304-0
Hyang Hee Cho, Siyeon Rhee, Dong Im Cho, Ju Hee Jun, HyoJung Heo, Su Han Cho, Dohyup Kim, Mingqiang Wang, Bo Gyeong Kang, Soo Ji Yoo, Meeyoung Cho, Soo yeon Lim, Jae Yeong Cho, In Seok Jeong, Yong Sook Kim, Youngkeun Ahn
The regulatory role of the inhibitor of NF-kB kinase ε (IKKε) in postmyocardial infarction (MI) inflammation remains uncertain. Using an MI mouse model, we examined the cardiac outcomes of IKKε knockout (KO) mice and wild-type mice. We employed single-cell RNA sequencing (scRNA-seq) and phosphorylated protein array techniques to profile cardiac macrophages. IKKε KO mice exhibited compromised survival, heightened inflammation, pronounced cardiac fibrosis, and a reduced ejection fraction. A distinct cardiac macrophage subset in IKKε KO mice exhibited increased fibrotic marker expression and decreased phosphorylated p38 (p-p38) levels, indicating an enhanced macrophage–myofibroblast transition (MMT) post-MI. While cardiac inflammation is crucial for initiating compensatory pathways, the timely resolution of inflammation was impaired in the IKKε KO group, while the MMT in macrophages accelerated post-MI, leading to cardiac failure. Additionally, our study highlighted the potential of 5-azacytidine (5-Aza), known for its anti-inflammatory and cardioprotective effects, in restoring p-p38 levels in stimulated macrophages. The administration of 5-Aza significantly reduced the MMT in cardiac macrophages from the IKKε KO group. These findings underscore the regulation of the inflammatory response and macrophage transition by the IKKε-p38 axis, indicating that the MMT is a promising therapeutic target for ischemic heart disease. Heart attacks can result in heart failure due to ongoing inflammation and scarring. This research investigates how a specific protein, IKKε, affects inflammation in heart cells. Researchers used genetically altered mice and sophisticated genetic methods to study IKKε‘s role in heart disease. They analyzed heart tissue after inducing heart attacks in the mice, focusing on how cells involved in inflammation and scarring behave differently when the IKKε protein is changed. Macrophage-myofibroblast transition (MMT) is a process in which macrophages acquire characteristics similar to myofibroblasts, potentially contributing to tissue fibrosis. The main discovery is that blocking IKKε causes more scarring by encouraging excessive MMT, suggesting it could be a target for heart disease treatment. The researchers believe that controlling the MMT process could be a new method to enhance heart health after a heart attack by reducing harmful scarring. This summary was initially drafted using artificial intelligence, then revised and fact-checked by the author.
NF-kB激酶ε抑制剂(IKKε)在心肌梗死(MI)后炎症中的调节作用仍不确定。我们利用心肌梗死小鼠模型,研究了IKKε基因敲除(KO)小鼠和野生型小鼠的心脏预后。我们采用了单细胞 RNA 测序(scRNA-seq)和磷酸化蛋白阵列技术来分析心脏巨噬细胞。IKKε KO小鼠表现出存活率下降、炎症加剧、明显的心脏纤维化和射血分数降低。IKKε KO 小鼠中一个独特的心脏巨噬细胞亚群表现出纤维化标志物表达增加和磷酸化 p38(p-p38)水平降低,表明心肌梗死后巨噬细胞-肌成纤维细胞转化(MMT)增强。虽然心脏炎症是启动代偿途径的关键,但在 IKKε KO 组中,炎症的及时解决受到了影响,而巨噬细胞的 MMT 在心肌梗死后加速,从而导致心力衰竭。此外,我们的研究还强调了 5-氮杂胞苷(5-Aza)在恢复受刺激巨噬细胞中 p-p38 水平方面的潜力,5-氮杂胞苷具有抗炎和保护心脏的作用。给予 5-Aza 能明显降低 IKKε KO 组心脏巨噬细胞中的 MMT。这些发现强调了IKKε-p38轴对炎症反应和巨噬细胞转化的调控作用,表明MMT是治疗缺血性心脏病的一个很有前景的靶点。
{"title":"IKKε-deficient macrophages impede cardiac repair after myocardial infarction by enhancing the macrophage–myofibroblast transition","authors":"Hyang Hee Cho, Siyeon Rhee, Dong Im Cho, Ju Hee Jun, HyoJung Heo, Su Han Cho, Dohyup Kim, Mingqiang Wang, Bo Gyeong Kang, Soo Ji Yoo, Meeyoung Cho, Soo yeon Lim, Jae Yeong Cho, In Seok Jeong, Yong Sook Kim, Youngkeun Ahn","doi":"10.1038/s12276-024-01304-0","DOIUrl":"10.1038/s12276-024-01304-0","url":null,"abstract":"The regulatory role of the inhibitor of NF-kB kinase ε (IKKε) in postmyocardial infarction (MI) inflammation remains uncertain. Using an MI mouse model, we examined the cardiac outcomes of IKKε knockout (KO) mice and wild-type mice. We employed single-cell RNA sequencing (scRNA-seq) and phosphorylated protein array techniques to profile cardiac macrophages. IKKε KO mice exhibited compromised survival, heightened inflammation, pronounced cardiac fibrosis, and a reduced ejection fraction. A distinct cardiac macrophage subset in IKKε KO mice exhibited increased fibrotic marker expression and decreased phosphorylated p38 (p-p38) levels, indicating an enhanced macrophage–myofibroblast transition (MMT) post-MI. While cardiac inflammation is crucial for initiating compensatory pathways, the timely resolution of inflammation was impaired in the IKKε KO group, while the MMT in macrophages accelerated post-MI, leading to cardiac failure. Additionally, our study highlighted the potential of 5-azacytidine (5-Aza), known for its anti-inflammatory and cardioprotective effects, in restoring p-p38 levels in stimulated macrophages. The administration of 5-Aza significantly reduced the MMT in cardiac macrophages from the IKKε KO group. These findings underscore the regulation of the inflammatory response and macrophage transition by the IKKε-p38 axis, indicating that the MMT is a promising therapeutic target for ischemic heart disease. Heart attacks can result in heart failure due to ongoing inflammation and scarring. This research investigates how a specific protein, IKKε, affects inflammation in heart cells. Researchers used genetically altered mice and sophisticated genetic methods to study IKKε‘s role in heart disease. They analyzed heart tissue after inducing heart attacks in the mice, focusing on how cells involved in inflammation and scarring behave differently when the IKKε protein is changed. Macrophage-myofibroblast transition (MMT) is a process in which macrophages acquire characteristics similar to myofibroblasts, potentially contributing to tissue fibrosis. The main discovery is that blocking IKKε causes more scarring by encouraging excessive MMT, suggesting it could be a target for heart disease treatment. The researchers believe that controlling the MMT process could be a new method to enhance heart health after a heart attack by reducing harmful scarring. This summary was initially drafted using artificial intelligence, then revised and fact-checked by the author.","PeriodicalId":50466,"journal":{"name":"Experimental and Molecular Medicine","volume":"56 9","pages":"2052-2064"},"PeriodicalIF":9.5,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s12276-024-01304-0.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142191252","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}