Experimental and Molecular Medicine最新文献

筛选
英文 中文
IFN-γ reprograms cardiac microvascular endothelial cells to mediate doxorubicin transport and influences the sensitivity of mice to doxorubicin-induced cardiotoxicity
IF 9.5 2区 医学
Experimental and Molecular Medicine Pub Date : 2025-01-22 DOI: 10.1038/s12276-024-01389-7
Haoyu Ji, Wenya Ma, Xu Liu, Hongyang Chen, Yining Liu, Zhongyu Ren, Daohong Yin, Ao Cai, Zizhen Zhang, Xin Wang, Wei Huang, Leping Shi, Yanan Tian, Yang Yu, Xiuxiu Wang, Yang Li, Yu Liu, Benzhi Cai
{"title":"IFN-γ reprograms cardiac microvascular endothelial cells to mediate doxorubicin transport and influences the sensitivity of mice to doxorubicin-induced cardiotoxicity","authors":"Haoyu Ji, Wenya Ma, Xu Liu, Hongyang Chen, Yining Liu, Zhongyu Ren, Daohong Yin, Ao Cai, Zizhen Zhang, Xin Wang, Wei Huang, Leping Shi, Yanan Tian, Yang Yu, Xiuxiu Wang, Yang Li, Yu Liu, Benzhi Cai","doi":"10.1038/s12276-024-01389-7","DOIUrl":"10.1038/s12276-024-01389-7","url":null,"abstract":"Doxorubicin (DOX) is a first-line chemotherapy agent known for its cardiac toxicity. DOX-induced cardiotoxicity (DIC) severely limits the use for treating malignant tumors and is associated with a poor prognosis. The sensitivity to DIC varies among patients, but the precise mechanisms remain elusive. Here we constructed a mouse model of DIC using DOX to investigate potential mechanisms contributing to the differential susceptibility to DIC. Through surface-enhanced Raman spectroscopy and single-cell RNA sequencing, we explored the mechanisms underlying DIC phenotypic variations. In vitro and in vivo studies with small-molecule drugs were conducted. DIC-insensitive mice displayed preserved ejection fractions, lower DOX levels in cardiac tissues and higher levels in the serum. Single-cell RNA sequencing revealed differences of gene expression in cardiac endothelial cells between DIC-insensitive and DIC-sensitive groups. The expression of IFN-γ pathway-related genes was high in DIC-insensitive mice. IFN-γ administration decreased the DOX distribution in cardiac tissues, whereas PPAR-γ activation increased DIC susceptibility. IFN-γ stimulation upregulated P-glycoprotein expression, leading to increased DOX efflux and DIC insensitivity. Our model provides insights into the mechanisms of DIC sensitivity and potential preventive strategies. Doxorubicin is a powerful cancer drug, but it can harm the heart, leading to a condition called doxorubicin-induced cardiotoxicity (DIC). Some people are more affected by DIC than others, and scientists want to understand why. They found that the heterogeneity observed among endothelial cells (ECs) plays a potential role in determining DIC sensitivity. In mice less sensitive to DIC, reprogramming of ECs increases levels of P-glycoprotein (P-gp), which helps to pump drugs out of cells. They discovered that activating a pathway involving IFN-γ increased P-gp levels, reducing heart damage. Conversely, activating another pathway, PPAR-γ, decreased P-gp levels and increased heart damage. These findings provide new insights into DIC pathogenesis and suggest that boosting P-gp in ECs could be a new strategy to protect against DIC. 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":"57 1","pages":"249-263"},"PeriodicalIF":9.5,"publicationDate":"2025-01-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s12276-024-01389-7.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143025110","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}
引用次数: 0
Exosome-based targeted delivery of NF-κB ameliorates age-related neuroinflammation in the aged mouse brain 基于外泌体的NF-κB靶向递送可改善老年小鼠大脑中与年龄相关的神经炎症。
IF 9.5 2区 医学
Experimental and Molecular Medicine Pub Date : 2025-01-20 DOI: 10.1038/s12276-024-01388-8
Chae-Jeong Lee, Seung Hyun Jang, Jiwoo Lim, Hyunju Park, So-Hee Ahn, Seon Young Park, Hyangmi Seo, Soo-Jin Song, Jung-A Shin, Chulhee Choi, Heon Yung Gee, Youn-Hee Choi
{"title":"Exosome-based targeted delivery of NF-κB ameliorates age-related neuroinflammation in the aged mouse brain","authors":"Chae-Jeong Lee, Seung Hyun Jang, Jiwoo Lim, Hyunju Park, So-Hee Ahn, Seon Young Park, Hyangmi Seo, Soo-Jin Song, Jung-A Shin, Chulhee Choi, Heon Yung Gee, Youn-Hee Choi","doi":"10.1038/s12276-024-01388-8","DOIUrl":"10.1038/s12276-024-01388-8","url":null,"abstract":"Neuroinflammation, a significant contributor to various neurodegenerative diseases, is strongly associated with the aging process; however, to date, no efficacious treatments for neuroinflammation have been developed. In aged mouse brains, the number of infiltrating immune cells increases, and the key transcription factor associated with increased chemokine levels is nuclear factor kappa B (NF-κB). Exosomes are potent therapeutics or drug delivery vehicles for various materials, including proteins and regulatory genes, to target cells. In the present study, we evaluated the therapeutic efficacy of exosomes loaded with a nondegradable form of IκB (Exo-srIκB), which inhibits the nuclear translocation of NF-κB to suppress age-related neuroinflammation. Single-cell RNA sequencing revealed that these anti-inflammatory exosomes targeted macrophages and microglia, reducing the expression of inflammation-related genes. Treatment with Exo-srIκB also suppressed the interactions between macrophages/microglia and T and B cells in the aged brain. We demonstrated that Exo-srIκB successfully alleviates neuroinflammation by primarily targeting activated macrophages and partially modulating the functions of age-related interferon-responsive microglia in the brain. Thus, our findings highlight Exo-srIκB as a potential therapeutic agent for treating age-related neuroinflammation. As we age, our bodies undergo changes that can lead to diseases like Alzheimer’s and Parkinson’s. A key factor in this process is inflammation in the brain, driven by a protein called NF-κB. Researcher explored a new way to reduce this inflammation using tiny particles called exosomes. These exosomes were engineered to carry a special form of a protein that blocks NF-κB, called srIκB. In their study, the team injected these exosomes into mice and observed their effects on neuroinflammation. They used advanced techniques to analyze changes in cells of the brain and found that the exosomes reduced inflammation-related genes and altered immune cell behavior. This suggests that the exosomes can help control inflammation in the aging brain. The results indicate that these engineered exosomes could be a promising treatment for age-related brain 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":"57 1","pages":"235-248"},"PeriodicalIF":9.5,"publicationDate":"2025-01-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s12276-024-01388-8.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143015501","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}
引用次数: 0
Author Correction: Influencing immunity: role of extracellular vesicles in tumor immune checkpoint dynamics 作者更正:影响免疫:细胞外囊泡在肿瘤免疫检查点动力学中的作用。
IF 9.5 2区 医学
Experimental and Molecular Medicine Pub Date : 2025-01-20 DOI: 10.1038/s12276-025-01394-4
Ziyang Ye, Genpeng Li, Jianyong Lei
{"title":"Author Correction: Influencing immunity: role of extracellular vesicles in tumor immune checkpoint dynamics","authors":"Ziyang Ye, Genpeng Li, Jianyong Lei","doi":"10.1038/s12276-025-01394-4","DOIUrl":"10.1038/s12276-025-01394-4","url":null,"abstract":"","PeriodicalId":50466,"journal":{"name":"Experimental and Molecular Medicine","volume":"57 1","pages":"283-283"},"PeriodicalIF":9.5,"publicationDate":"2025-01-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s12276-025-01394-4.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143015497","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}
引用次数: 0
mtSTAT3 suppresses rheumatoid arthritis by regulating Th17 and synovial fibroblast inflammatory cell death with IL-17-mediated autophagy dysfunction mtSTAT3通过调节Th17和滑膜成纤维细胞炎症细胞死亡以及il -17介导的自噬功能障碍来抑制类风湿关节炎。
IF 9.5 2区 医学
Experimental and Molecular Medicine Pub Date : 2025-01-17 DOI: 10.1038/s12276-024-01376-y
Seon-Yeong Lee, Jeonghyeon Moon, A Ram Lee, Young-Mee Moon, Jeong Won Choi, Chae Rim Lee, Su Been Jeon, Hee Su Sohn, Jeehee Youn, Dongyun Shin, Sung-Hwan Park, Mi-La Cho
{"title":"mtSTAT3 suppresses rheumatoid arthritis by regulating Th17 and synovial fibroblast inflammatory cell death with IL-17-mediated autophagy dysfunction","authors":"Seon-Yeong Lee, Jeonghyeon Moon, A Ram Lee, Young-Mee Moon, Jeong Won Choi, Chae Rim Lee, Su Been Jeon, Hee Su Sohn, Jeehee Youn, Dongyun Shin, Sung-Hwan Park, Mi-La Cho","doi":"10.1038/s12276-024-01376-y","DOIUrl":"10.1038/s12276-024-01376-y","url":null,"abstract":"Th17 cells are activated by STAT3 factors in the nucleus, and these factors are correlated with the pathologic progression of rheumatoid arthritis (RA). Recent studies have demonstrated the presence of STAT3 in mitochondria, but its function is unclear. We investigated the novel role of mitochondrial STAT3 (mitoSTAT3) in Th17 cells and fibroblast-like synoviocytes (FLSs) and analyzed the correlation of mitoSTAT3 with RA. We used a collagen-induced arthritis (CIA) mouse model to determine the effect of mitochondrial STAT3. We observed changes in the RA mouse model via the use of a mitochondrial STAT3-inducing vector and inhibitor. We observed the accumulation of abnormal autophagosomes, increased inflammatory cell death signaling, and decreased mitoSTAT3 activity in FLSs from both patients with RA and patients with IL-17-treated FLSs. We first discovered that IL-17 increased the accumulation of abnormal autophagosomes and the expression of inflammatory cell death factors in synovial fibroblasts and decreased mitoSTAT3 activation. In a mouse model of CIA, arthritis and joint inflammation were decreased by injection vectors that induced mitoSTAT3 overexpression. The abnormal accumulation of autophagosomes and the expression of inflammatory cell death factors were also decreased in these mice. In mouse and human immune cells, ZnSO4, an inducer of mitochondrial STAT3, decreases the production of reactive oxygen species, the IL-17 concentration, and differentiation into Th17 cells. However, mitoSTAT3 blockade accelerated the development of arthritis, inflammatory cell death, and abnormal autophagosome/autophagolysosome formation. Therefore, this study suggests a novel inhibitory mechanism of RA using mitoSTAT3 via the regulation of autophagy, Th17 differentiation, and inflammatory cell death. Rheumatoid arthritis is a long-term disease where the immune system attacks the joints, causing pain and swelling. Researchers have found that a protein called mitoSTAT3, located in the mitochondria, might help reduce inflammation in RA. Researchers used mice with arthritis and human cells to study mitoSTAT3’s role. They increased mitoSTAT3 levels in mice and observed less joint damage and inflammation and found that mitoSTAT3 helps control a process called autophagy. This is important because poor autophagy can worsen RA. The results showed that boosting mitoSTAT3 reduced inflammation and joint damage in mice. The researchers concluded that mitoSTAT3 could be a new target for RA treatment by improving cell cleanup processes and reducing harmful immune responses. In the future, therapies that increase mitoSTAT3 might help people with RA by reducing inflammation and joint damage. 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":"57 1","pages":"221-234"},"PeriodicalIF":9.5,"publicationDate":"2025-01-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s12276-024-01376-y.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143015513","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}
引用次数: 0
Advancing pancreatic cancer research and therapeutics: the transformative role of organoid technology 推进胰腺癌研究和治疗:类器官技术的变革作用。
IF 9.5 2区 医学
Experimental and Molecular Medicine Pub Date : 2025-01-16 DOI: 10.1038/s12276-024-01378-w
Jihao Xu, Minh Duc Pham, Vincenzo Corbo, Mariano Ponz-Sarvise, Tobiloba Oni, Daniel Öhlund, Chang-Il Hwang
{"title":"Advancing pancreatic cancer research and therapeutics: the transformative role of organoid technology","authors":"Jihao Xu, Minh Duc Pham, Vincenzo Corbo, Mariano Ponz-Sarvise, Tobiloba Oni, Daniel Öhlund, Chang-Il Hwang","doi":"10.1038/s12276-024-01378-w","DOIUrl":"10.1038/s12276-024-01378-w","url":null,"abstract":"Research on pancreatic cancer has transformed with the advent of organoid technology, providing a better platform that closely mimics cancer biology in vivo. This review highlights the critical advancements facilitated by pancreatic organoid models in understanding disease progression, evaluating therapeutic responses, and identifying biomarkers. These three-dimensional cultures enable the proper recapitulation of the cellular architecture and genetic makeup of the original tumors, providing insights into the complex molecular and cellular dynamics at various stages of pancreatic ductal adenocarcinoma (PDAC). We explore the applications of pancreatic organoids in dissecting the tumor microenvironment (TME); elucidating cancer progression, metastasis, and drug resistance mechanisms; and personalizing therapeutic strategies. By overcoming the limitations of traditional 2D cultures and animal models, the use of pancreatic organoids has significantly accelerated translational research, which is promising for improving diagnostic and therapeutic approaches in clinical settings, ultimately aiming to improve the outcomes of patients with pancreatic cancer. Pancreatic cancer is a challenging disease to study and treat. This article discusses how researchers have developed pancreatic organoids to better study this cancer. Organoids are created by growing cells in a specialized 3D matrix, allowing them to form structures that resemble tissues found in the body. This method is more effective than traditional 2D cultures because it better replicates the natural environment of the cells. Researchers use these organoids to study cancer progression, test new drugs, and understand genetic changes in tumors. They can be made from small tissue samples, making them useful for studying advanced cancer stages where tissue is scarce. The findings from organoid studies help identify potential new treatments and improve our understanding of pancreatic cancer biology. In conclusion, pancreatic organoids offer a promising tool for advancing cancer research and developing personalized 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":"57 1","pages":"50-58"},"PeriodicalIF":9.5,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s12276-024-01378-w.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143015493","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}
引用次数: 0
Pancreatic organogenesis mapped through space and time 通过空间和时间绘制胰腺器官发生图。
IF 9.5 2区 医学
Experimental and Molecular Medicine Pub Date : 2025-01-08 DOI: 10.1038/s12276-024-01384-y
Marissa A. Scavuzzo, Wojciech J. Szlachcic, Matthew C. Hill, Natalia M. Ziojla, Jessica Teaw, Jeffrey C. Carlson, Jonathan Tiessen, Jolanta Chmielowiec, James F. Martin, Malgorzata Borowiak
{"title":"Pancreatic organogenesis mapped through space and time","authors":"Marissa A. Scavuzzo, Wojciech J. Szlachcic, Matthew C. Hill, Natalia M. Ziojla, Jessica Teaw, Jeffrey C. Carlson, Jonathan Tiessen, Jolanta Chmielowiec, James F. Martin, Malgorzata Borowiak","doi":"10.1038/s12276-024-01384-y","DOIUrl":"10.1038/s12276-024-01384-y","url":null,"abstract":"The spatial organization of cells within a tissue is dictated throughout dynamic developmental processes. We sought to understand whether cells geometrically coordinate with one another throughout development to achieve their organization. The pancreas is a complex cellular organ with a particular spatial organization. Signals from the mesenchyme, neurons, and endothelial cells instruct epithelial cell differentiation during pancreatic development. To understand the cellular diversity and spatial organization of the developing pancreatic niche, we mapped the spatial relationships between single cells over time. We found that four transcriptionally unique subtypes of mesenchyme in the developing pancreas spatially coordinate throughout development, with each subtype at fixed locations in space and time in relation to other cells, including beta cells, vasculature, and epithelial cells. Our work provides insight into the mechanisms of pancreatic development by showing that cells are organized in a space and time manner. This study explores how different types of cells, called mesenchyme, help form the pancreas during development. Researchers used various techniques to study mouse embryos and human fetal tissue. They identified several subtypes of mesenchyme in the developing pancreas and found that these subtypes are not randomly distributed; instead, they occupy specific locations. The study involved analyzing single-cell RNA sequencing data (a method to study gene expression in individual cells) and using advanced imaging techniques to map the positions of these cells. The researchers discovered that different mesenchyme subtypes have unique roles, such as supporting blood vessel formation or nerve development. These findings suggest that understanding mesenchyme organization could improve regenerative medicine approaches for diseases like diabetes. Future research may explore how these cells interact with other pancreatic components over time. 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":"57 1","pages":"204-220"},"PeriodicalIF":9.5,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s12276-024-01384-y.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142958279","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}
引用次数: 0
Multiomic quantification of the KRAS mutation dosage improves the preoperative prediction of survival and recurrence in patients with pancreatic ductal adenocarcinoma KRAS突变剂量的多组量化提高了胰腺导管腺癌患者术前生存和复发的预测。
IF 9.5 2区 医学
Experimental and Molecular Medicine Pub Date : 2025-01-08 DOI: 10.1038/s12276-024-01382-0
Won-Gun Yun, Daeun Kim, Youngmin Han, Wooil Kwon, Seong-Geun Lee, Jin-Young Jang, Daechan Park
{"title":"Multiomic quantification of the KRAS mutation dosage improves the preoperative prediction of survival and recurrence in patients with pancreatic ductal adenocarcinoma","authors":"Won-Gun Yun, Daeun Kim, Youngmin Han, Wooil Kwon, Seong-Geun Lee, Jin-Young Jang, Daechan Park","doi":"10.1038/s12276-024-01382-0","DOIUrl":"10.1038/s12276-024-01382-0","url":null,"abstract":"Most cancer mutation profiling studies are laboratory-based and lack direct clinical application. For clinical use, it is necessary to focus on key genes and integrate them with relevant clinical variables. We aimed to evaluate the prognostic value of the dosage of the KRAS G12 mutation, a key pancreatic ductal adenocarcinoma (PDAC) variant and to investigate the biological mechanism of the prognosis associated with the dosage of the KRAS G12 mutation. In this retrospective cohort study, we analyzed 193 surgically treated patients with PDAC between 2009 and 2016. RNA, whole-exome, and KRAS-targeted sequencing data were used to estimate the dosage of the KRAS G12 mutant. Our prognostic scoring system included the mutation dosage from targeted sequencing ( > 0.195, 1 point), maximal tumor diameter at preoperative imaging ( > 20 mm, 1 point), and carbohydrate antigen 19-9 levels ( > 150 U/mL, 1 point). The KRAS mutation dosage exhibited comparable performance with clinical variables for survival prediction. High KRAS mutation dosages activated the cell cycle, leading to high mutation rates and poor prognosis. According to prognostic scoring systems that integrate mutation dosage with clinical factors, patients with 0 points had superior median overall survival of 97.0 months and 1-year, 3-year, and 5-year overall survival rates of 95.8%, 70.8%, and 66.4%, respectively. In contrast, patients with 3 points had worse median overall survival of only 16.0 months and 1-year, 3-year, and 5-year overall survival rates of 65.2%, 8.7%, and 8.7%, respectively. The incorporation of the KRAS G12 mutation dosage variable into prognostic scoring systems can improve clinical variable-based survival prediction, highlighting the feasibility of an integrated scoring system with clinical significance. Pancreatic ductal adenocarcinoma (PDAC) is a severe health issue with low survival rates. This study is aimed to improve PDAC treatment by examining the KRAS gene mutation, which is common in these tumors. The study involved 193 patients who had surgery for PDAC. Researchers used different sequencing methods to measure the KRAS mutation levels and compared these with clinical data. They found that higher KRAS mutation levels were linked to faster tumor growth and earlier recurrence after surgery. By combining KRAS mutation data with clinical factors like tumor size and a blood marker, they developed a scoring system to predict patient outcomes. This system could help doctors tailor treatments more effectively. The study suggests that using KRAS mutation levels can improve predictions about PDAC progression and guide personalized treatment plans in the future. 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":"57 1","pages":"193-203"},"PeriodicalIF":9.5,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s12276-024-01382-0.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142958278","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}
引用次数: 0
Activators of the 26S proteasome when protein degradation increases 当蛋白质降解增加时,26S蛋白酶体的激活剂。
IF 9.5 2区 医学
Experimental and Molecular Medicine Pub Date : 2025-01-08 DOI: 10.1038/s12276-024-01385-x
Donghoon Lee
{"title":"Activators of the 26S proteasome when protein degradation increases","authors":"Donghoon Lee","doi":"10.1038/s12276-024-01385-x","DOIUrl":"10.1038/s12276-024-01385-x","url":null,"abstract":"In response to extra- and intracellular stimuli that constantly challenge and disturb the proteome, cells rapidly change their proteolytic capacity to maintain proteostasis. Failure of such efforts often becomes a major cause of diseases or is associated with exacerbation. Increase in protein breakdown occurs at multiple steps in the ubiquitin-proteasome system, and the regulation of ubiquitination has been extensively studied. However, the activities of the 26S proteasome are also stimulated, especially under highly catabolic conditions such as those associated with atrophying skeletal muscle, proteotoxic stress such as heat shock and arsenite, or hormonal cues such as cAMP or cGMP agonists. Among the proteins that enhance proteasomal degradation are the PKA, PKG, UBL-UBA proteins and the Zn finger AN1-type domain (ZFAND) family proteins. ZFAND proteins are of particular interest because of their inducible expression in response to various stimuli and their abilities to control protein quality by stimulating the 26S proteasome and p97/VCP. The regulatory roles of ZFAND proteins appear to be important not only for the control of protein degradation but also for other cellular processes, such as mRNA stability and signaling pathways. This review summarizes the known functions of proteasome activators and discusses their possible roles in regulating proteostasis and other cellular processes. For many years, scientists have studied how cells break down proteins using a proteolytic complex like the proteasome. The proteasome is a complex machine that degrades majority of intracellular proteins. However, its activation mechanism under stresses is not fully understood. Researchers explored proteins called ZFANDs, which can activate the proteasome. The studies used various methods to understand how ZFAND proteins work and found that ZFAND5, as an example, helps the proteasome break down proteins more efficiently. This is important because it can help cells manage stresses by removing unwanted proteins. The results showed that ZFAND5 binds to the proteasome and enhances its activity, especially in stresses causing muscle wasting. This discovery helps us understand how cells adapt to stresses and could lead to new treatments for diseases where protein breakdown is disrupted. 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":"57 1","pages":"41-49"},"PeriodicalIF":9.5,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s12276-024-01385-x.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142958277","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}
引用次数: 0
The WAVE complex in developmental and adulthood brain disorders 发展性和成人期脑障碍的WAVE复合体。
IF 9.5 2区 医学
Experimental and Molecular Medicine Pub Date : 2025-01-07 DOI: 10.1038/s12276-024-01386-w
Hyung-Goo Kim, Clara Berdasco, Angus C. Nairn, Yong Kim
{"title":"The WAVE complex in developmental and adulthood brain disorders","authors":"Hyung-Goo Kim, Clara Berdasco, Angus C. Nairn, Yong Kim","doi":"10.1038/s12276-024-01386-w","DOIUrl":"10.1038/s12276-024-01386-w","url":null,"abstract":"Actin polymerization and depolymerization are fundamental cellular processes required not only for the embryonic and postnatal development of the brain but also for the maintenance of neuronal plasticity and survival in the adult and aging brain. The orchestrated organization of actin filaments is controlled by various actin regulatory proteins. Wiskott‒Aldrich syndrome protein-family verprolin-homologous protein (WAVE) members are key activators of ARP2/3 complex-mediated actin polymerization. WAVE proteins exist as heteropentameric complexes together with regulatory proteins, including CYFIP, NCKAP, ABI and BRK1. The activity of the WAVE complex is tightly regulated by extracellular cues and intracellular signaling to execute its roles in specific intracellular events in brain cells. Notably, dysregulation of the WAVE complex and WAVE complex-mediated cellular processes confers vulnerability to a variety of brain disorders. De novo mutations in WAVE genes and other components of the WAVE complex have been identified in patients with developmental disorders such as intellectual disability, epileptic seizures, schizophrenia, and/or autism spectrum disorder. In addition, alterations in the WAVE complex are implicated in the pathophysiology of Alzheimer’s disease and Parkinson’s disease, as well as in behavioral adaptations to psychostimulants or maladaptive feeding. The WAVE complex is a group of proteins that work together to regulate actin, an essential component of the cell cytoskeleton. This article reviews how changes in the WAVE complex can affect brain function and contribute to various brain disorders. It summarizes how different parts of the WAVE complex interact with other proteins to regulate actin polymerization. This regulation is crucial for programming brain cell growth, migration, and synaptic formation and function. This article also explores recent studies on genetic changes in the WAVE complex and their links to developmental and adult brain disorders. Key findings suggest disruptions in the WAVE complex can lead to conditions like intellectual disabilities, autism, and neurodegenerative diseases such as Alzheimer’s. 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":"57 1","pages":"13-29"},"PeriodicalIF":9.5,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s12276-024-01386-w.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142957149","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}
引用次数: 0
Sociobiology meets oncology: unraveling altruistic cooperation in cancer cells and its implications 社会生物学与肿瘤学:揭示癌细胞中的利他合作及其含义。
IF 9.5 2区 医学
Experimental and Molecular Medicine Pub Date : 2025-01-07 DOI: 10.1038/s12276-024-01387-9
Muhammad Sufyan bin Masroni, Evelyn Siew-Chuan Koay, Victor Kwan Min Lee, Siok Bian Ng, Soo Yong Tan, Karen Meiling Tan, Marco Archetti, Sai Mun Leong
{"title":"Sociobiology meets oncology: unraveling altruistic cooperation in cancer cells and its implications","authors":"Muhammad Sufyan bin Masroni, Evelyn Siew-Chuan Koay, Victor Kwan Min Lee, Siok Bian Ng, Soo Yong Tan, Karen Meiling Tan, Marco Archetti, Sai Mun Leong","doi":"10.1038/s12276-024-01387-9","DOIUrl":"10.1038/s12276-024-01387-9","url":null,"abstract":"Altruism, an act of benefiting others at a cost to the self, challenges our understanding of evolution. This Perspective delves into the importance of altruism in cancer cells and its implications for therapy. Against the backdrop of existing knowledge on various social organisms found in nature, we explore the mechanisms underlying the manifestation of altruism within breast tumors, revealing a complex interplay of seemingly counteracting cancer signaling pathways and processes that orchestrate the delicate balance between cost and benefit underlying altruistic cooperation. We also discuss how evolutionary game theory, coupled with contemporary molecular tools, may shed light on understudied mechanisms governing the dynamics of altruistic cooperation in cancer cells. Finally, we discuss how molecular insights gleaned from these mechanistic dissections may fuel advancements in our comprehension of altruism among cancer cells, with implications across multiple disciplines, offering innovative prospects for therapeutic strategies, molecular discoveries, and evolutionary investigations. Altruism, or selfless behavior, has puzzled scientists for years, especially in the context of evolution. Traditionally, cancer cells are seen as selfish, growing uncontrollably. However, recent research suggests some cancer cells might act altruistically. They found that certain breast cancer cells produce substances that help neighboring cells survive chemotherapy, even though this slows their own growth. The study used breast cancer cell lines to observe these interactions. Researchers identified a subpopulation of cells with high levels of a molecule called miR-125b. These cells secrete proteins that protect other cells but grow more slowly themselves. This behavior fits the definition of biological altruism, where one organism incurs a cost to benefit others. The findings suggest that understanding these altruistic behaviors in cancer could lead to new treatment strategies. 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":"57 1","pages":"30-40"},"PeriodicalIF":9.5,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s12276-024-01387-9.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142958280","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}
引用次数: 0
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信