1
|
Hu JJ, Deng F, Sun QS, Xiong QM, Min Y, Feng SY, Lin ZB, Chen PH, Hu Z, Wu L, Chen XF, Xie S, Liu WF, Li C, Liu KX. Time-restricted feeding protects against septic liver injury by reshaping gut microbiota and metabolite 3-hydroxybutyrate. Gut Microbes 2025; 17:2486515. [PMID: 40223164 PMCID: PMC12005432 DOI: 10.1080/19490976.2025.2486515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/13/2025] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
Liver injury is an independent risk factor for multiple organ dysfunction and high mortality in patients with sepsis. However, the pathological mechanisms and therapeutic strategies for sepsis-associated liver injury have not been fully elucidated. Time-restricted feeding (TRF) is a promising dietary regime, but its role in septic liver injury remains unknown. Using 16S rRNA gene sequencing, Q200 targeted metabolomics, transcriptomics, germ-free mice, Hmgcs2/Lpin1 gene knockout mice, and Aml12 cells experiments, we revealed that TRF can mitigate septic liver injury by modulating the gut microbiota, particularly by increasing Lactobacillus murinus (L. murinus) abundance, which was significantly reduced in septic mice. Further study revealed that live L. murinus could markedly elevate serum levels of metabolite 3-hydroxybutyrate (3-HB) and alleviate sepsis-related injury, while the knockout of the key enzyme for 3-HB synthesis (3-hydroxy-3-methylglutaryl-CoA synthase 2, Hmgcs2) in the liver negated this protective effect. Additionally, serum 3-HB levels were significantly positively correlated with L. murinus abundance and negatively correlated with liver injury indicators in septic patients, demonstrating a strong predictive value for septic liver injury (AUC = 0.8429). Mechanistically, 3-HB significantly inhibited hepatocyte ferroptosis by activating the PI3K/AKT/mTOR/LPIN1 pathway, reducing ACSL4, MDA, LPO, and Fe2+ levels. This study demonstrates that TRF reduces septic liver injury by modulating gut microbiota to increase L. murinus, which elevates 3-HB to activate PI3K/AKT/mTOR/LPIN1 and inhibit hepatocyte ferroptosis. Overall, this study elucidates the protective mechanism of TRF against septic liver injury and identifies 3-HB as a potential therapeutic target and predictive biomarker, thereby providing new insights into the clinical management and diagnosis of septic liver injury.
Collapse
Affiliation(s)
- Jing-Juan Hu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Fan Deng
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Qi-Shun Sun
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Qing-Ming Xiong
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
- Department of Anesthesiology, The First People’s Hospital of Foshan, Foshan, China
| | - Yue Min
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Si-Yuan Feng
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Ze-Bin Lin
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Peng-Han Chen
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Zhen Hu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Ling Wu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Xiao-Feng Chen
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Sun Xie
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Wei-Feng Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Cai Li
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Ke-Xuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Fang X, Liu H, Liu J, Du Y, Chi Z, Bian Y, Zhao X, Teng T, Shi B. Isobutyrate Confers Resistance to Inflammatory Bowel Disease through Host-Microbiota Interactions in Pigs. RESEARCH (WASHINGTON, D.C.) 2025; 8:0673. [PMID: 40342298 PMCID: PMC12059313 DOI: 10.34133/research.0673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/20/2025] [Accepted: 03/23/2025] [Indexed: 05/11/2025]
Abstract
Supplementation with short-chain fatty acids (SCFAs) is a potential therapeutic approach for inflammatory bowel disease (IBD). However, the therapeutic effects and mechanisms of action of isobutyrate in IBD remain unclear. Clinical data indicate that the fecal levels of isobutyrate are markedly lower in patients with Crohn's disease than in healthy controls. Compared with healthy mice and healthy pigs, mice and pigs with colitis presented significantly lower isobutyrate levels. Furthermore, the level of isobutyrate in pigs was significantly negatively correlated with the disease activity index. We speculate that isobutyrate may play a crucial role in regulating host gut homeostasis. We established a model of dextran sulfate sodium-induced colitis in pigs, which have gastrointestinal structure and function similar to those of humans; we performed multiomic analysis to investigate the therapeutic effects and potential mechanisms of isobutyrate on IBD at both the animal and cellular levels and validated the results. Phenotypically, isobutyrate can significantly alleviate diarrhea, bloody stools, weight loss, and colon shortening caused by colitis in pigs. Mechanistically, isobutyrate can increase the relative abundance of Lactobacillus reuteri, thereby increasing the production of indole-3-lactic acid, regulating aryl hydrocarbon receptor expression and downstream signaling pathways, and regulating Foxp3+ CD4+ T cell recruitment to alleviate colitis. Isobutyrate can directly activate G protein-coupled receptor 109A, promote the expression of Claudin-1, and improve intestinal barrier function. In addition, isobutyrate can increase the production of intestinal SCFAs and 3-hydroxybutyric acid and inhibit the TLR4/MyD88/NF-κB signaling pathway to suppress intestinal inflammation. In conclusion, our findings demonstrate that isobutyrate confers resistance to IBD through host-microbiota interactions, providing a theoretical basis for the use of isobutyrate in alleviating colitis.
Collapse
Affiliation(s)
| | | | - Junling Liu
- College of Animal Science and Technology,
Northeast Agricultural University, Harbin 150030, People’s Republic of China
| | - Yongqing Du
- College of Animal Science and Technology,
Northeast Agricultural University, Harbin 150030, People’s Republic of China
| | - Zihan Chi
- College of Animal Science and Technology,
Northeast Agricultural University, Harbin 150030, People’s Republic of China
| | - Yiqi Bian
- College of Animal Science and Technology,
Northeast Agricultural University, Harbin 150030, People’s Republic of China
| | - Xuan Zhao
- College of Animal Science and Technology,
Northeast Agricultural University, Harbin 150030, People’s Republic of China
| | - Teng Teng
- College of Animal Science and Technology,
Northeast Agricultural University, Harbin 150030, People’s Republic of China
| | - Baoming Shi
- College of Animal Science and Technology,
Northeast Agricultural University, Harbin 150030, People’s Republic of China
| |
Collapse
|
3
|
Sun K, Liu Y, Pan Y, Di D, Li J, Xu F, Li L, Mimata Y, Chen Y, Xie L, Wang S, Qi W, Tang Y, Sheng H, Wang B, Sun R, Tan D, Fu D, Yin Y, Xue A, Shi Y, Shao W, Gong L, Jiang Z, Zhang W, Wu Q, Wang Y, Lang M, Ye W, Xu W, Wei S, Shi W, Xu YJ. Non-invasive micro-test technology and applications. BIOPHYSICS REPORTS 2025; 11:96-111. [PMID: 40308937 PMCID: PMC12035745 DOI: 10.52601/bpr.2024.240009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 10/14/2024] [Indexed: 05/02/2025] Open
Abstract
Non-invasive micro-test technology (NMT) reveals dynamic ionic/molecular concentration gradients by measuring fluxes of ions and small molecules in liquid media in 1D, 2D or 3D fashions with sensitivity up to pico- (10-12) or femto- (10-15) moles per cm2 per second. NMT has been applied to study metabolism, signal transduction, genes and/or proteins physiological functions related to transmembrane ionic/molecular activities with live samples under normal conditions or stress. Data on ion and/or molecule homeostasis (IMH) by NMT in biomedical sciences, plant and crop sciences, environmental sciences, marine and space biology as well as traditional Chinese medicine are reviewed.
Collapse
Affiliation(s)
- Kai Sun
- College of Life Sciences, Nanjing Normal University, Nanjing 210046, China
| | - Yunqi Liu
- Zhongguancun Xuyue NMT Industrial Alliance, Beijing 100080, China
- NMT International Alliance, Amherst, Massachusetts 01002, USA
| | - Yanshu Pan
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Dongwei Di
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing 210008, China
| | - Jianfang Li
- College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Feiyun Xu
- Center for Plant Water-use and Nutrition Regulation and College of Resources and Environment, Joint International Research Laboratory of Water and Nutrient in Crop, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Li Li
- Institute of Agricultural Resources and Environment, Tianjin Academy of Agricultural Sciences, Tianjin 300380, China
| | - Yoshiharu Mimata
- Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences at Weifang, Weifang 261000, Shandong, China
| | - Yingying Chen
- Guangxi Forestry Research Institute, Nanning 530002, China
| | - Lixia Xie
- Institute of Wetland Agriculture and Ecology, Shandong Academy of Agricultural Science, Jinan 250100, China
| | - Siqi Wang
- Key Laboratory of Wastewater Treatment Technology of Liaoning Province, Academy of Environmental and Chemical Engineering, Shenyang Ligong University, Shenyang 110159, China
| | - Wenqian Qi
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Yan Tang
- Technical Institute of Physics and Chemistry, CAS, Beijing 100190, China
| | - Huachun Sheng
- Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu 610225, China
| | - Bing Wang
- Molecular Genetics Key Laboratory of China Tobacco, Guizhou Academy of Tobacco Science, Guiyang 550081, China
| | - Ruixue Sun
- College of Agriculture and Forestry, Hebei North University, Zhangjiakou 075000, Hebei, China
| | - Dingquan Tan
- Smart Health Institute, Chongqing Vocational College of Media, Chongqing 402560, China
| | - Daohong Fu
- Institute of Biology, Humboldt University of Berlin, Berlin 10099, Germany
| | - Ye Yin
- College of Horticulture, Qingdao Agricultural University, Qingdao 266109, Shandong, China
| | - Ao Xue
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Yichao Shi
- Department of Gastroenterology, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, China
| | - Wenjing Shao
- CAS Center for Excellence in Biotic Interactions, College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lei Gong
- State Key Laboratory of Grassland Agro-ecosystems, School of Life Sciences, Lanzhou 730000, China
| | - Zhijian Jiang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Wei Zhang
- Technical Institute of Physics and Chemistry, CAS, Beijing 100190, China
| | - Qiangsheng Wu
- College of Horticulture and Gardening, Yangtze University, Jingzhou 434025, Hubei, China
| | - Yaosheng Wang
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Minglin Lang
- CAS Center for Excellence in Biotic Interactions, College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenxiu Ye
- Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences at Weifang, Weifang 261000, Shandong, China
| | - Weifeng Xu
- Center for Plant Water-use and Nutrition Regulation and College of Resources and Environment, Joint International Research Laboratory of Water and Nutrient in Crop, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Shuhe Wei
- Key Laboratory of Pollution Ecology and Environment Engineering, Institute of Applied Ecology, Shenyang 110016, China
| | - Weiming Shi
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing 210008, China
| | - Yue Jeff Xu
- Zhongguancun Xuyue NMT Industrial Alliance, Beijing 100080, China
- NMT International Alliance, Amherst, Massachusetts 01002, USA
- Xuyue (Beijing) Sci. & Tech. Co., Ltd., Beijing 100080, China
- YoungerUSA LLC, Amherst, Massachusetts 01002, USA
| |
Collapse
|
4
|
Soni S, Skow RJ, Foulkes S, Haykowsky MJ, Dyck JRB. Therapeutic potential of ketone bodies on exercise intolerance in heart failure: looking beyond the heart. Cardiovasc Res 2025; 121:230-240. [PMID: 39825790 PMCID: PMC12012446 DOI: 10.1093/cvr/cvaf004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/13/2024] [Accepted: 12/05/2024] [Indexed: 01/20/2025] Open
Abstract
Recent evidence suggests that ketone bodies have therapeutic potential in many cardiovascular diseases including heart failure (HF). Accordingly, this has led to multiple clinical trials that use ketone esters (KEs) to treat HF patients highlighting the importance of this ketone therapy. KEs, specifically ketone monoesters, are synthetic compounds which, when consumed, are de-esterified into two β-hydroxybutyrate (βOHB) molecules and increase the circulating βOHB concentration. While many studies have primarily focused on the cardiac benefits of ketone therapy in HF, ketones can have numerous favourable effects in other organs such as the vasculature and skeletal muscle. Importantly, vascular and skeletal muscle dysfunction are also heavily implicated in the reduced exercise tolerance, the hallmark feature in HF with reduced ejection fraction and preserved ejection fraction, suggesting that some of the benefits observed in HF in response to ketone therapy may involve these non-cardiac pathways. Thus, we review the evidence suggesting how ketone therapy may be beneficial in improving cardiovascular and skeletal muscle function in HF and identify various potential mechanisms that may be important in the beneficial non-cardiac effects of ketones in HF.
Collapse
Affiliation(s)
- Shubham Soni
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Rachel J Skow
- Integrated Cardiovascular and Exercise Physiology and Rehabilitation (iCARE) Lab, Faculty of Nursing, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Stephen Foulkes
- Integrated Cardiovascular and Exercise Physiology and Rehabilitation (iCARE) Lab, Faculty of Nursing, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada
- Heart, Exercise and Research Trials Lab, St Vincent’s Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Mark J Haykowsky
- Integrated Cardiovascular and Exercise Physiology and Rehabilitation (iCARE) Lab, Faculty of Nursing, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Jason R B Dyck
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
5
|
Qiu X, Wu W, Zhang S, Huang C, Lin D. 3-Hydroxybutyrate Promotes Myoblast Proliferation and Differentiation through Energy Metabolism and GPR109a-Mediated Ca 2+-NFAT Signaling Pathways. J Proteome Res 2025; 24:2063-2080. [PMID: 40099866 DOI: 10.1021/acs.jproteome.4c01150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Skeletal muscle wasting is a critical clinical problem associated with several diseases that significantly impair patient outcomes due to the progressive loss of muscle mass and function. This study explores the potential of 3-hydroxybutyrate (3-HB) as a therapeutic agent to counteract muscle atrophy by promoting the proliferation and differentiation of C2C12 myoblasts. Using nuclear magnetic resonance (NMR)-based metabolomics analysis, we uncover the underlying mechanisms by which 3-HB exerts its effects. Our findings demonstrate that 3-HB exerts its effects through two distinct mechanisms: as a metabolic substrate and as a signaling molecule. As a metabolic substrate, 3-HB enhances myoblast energy efficiency by stimulating the expression of G protein-coupled receptor 109a (GPR109a), which subsequently upregulates the 3-HB transporters MCT1 and CD147, the utilization enzyme OXCT1, and phosphorylated AMPK, thereby increasing ATP production. As a signaling molecule, 3-HB activates GPR109a, promoting calcium influx, improving calcium homeostasis, and increasing the expression of Ca2+-related proteins such as CAMKK2. This signaling cascade activates calcineurin (CaN), facilitating NFAT translocation to the nucleus and gene expression that drives myoblast proliferation and differentiation. By elucidating the dual regulatory roles of 3-HB in energy metabolism and cellular signaling, this study not only advances our understanding of muscle physiology but also highlights the potential of 3-HB as a novel therapeutic approach for the prevention or treatment of skeletal muscle atrophy.
Collapse
Affiliation(s)
- Xu Qiu
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Wenfang Wu
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Shuya Zhang
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Caihua Huang
- Research and Communication Center of Exercise and Health, Xiamen University of Technology, Xiamen 361024, China
| | - Donghai Lin
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
6
|
Cui ZJ. Deep-sea photodynamic vision at low light level - Which is more important, prosthetic retinal or apo-rhodopsin moiety? FASEB J 2025; 39:e70470. [PMID: 40100047 DOI: 10.1096/fj.202500213r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/21/2025] [Accepted: 03/10/2025] [Indexed: 03/20/2025]
Abstract
The special case of far-red vision of mesopelagic dragonfish Malacosteus niger facilitated by the presence in the rod outer segment of photosensitizer chlorin e6 from diet has drawn considerable attention both in vision research and in photodynamic action. Rhodopsin binding of Ce6 from either the extracellular or intracellular loops may exert different effects. Theoretical works predict that the extracellularly bound Ce6 upon absorption of red light produces a singlet oxygen, which could via an oxygen tunnel reach the Lys-tethered 11-cis-retinal, by way of peroxy-dioxetane intermediates, to enhance 11-cis- to all-trans-retinal isomerization, therefore triggering the ultrafast phototransduction process. Recent works on the permanent photodynamic activation of some A-class G protein-coupled receptors suggest that the singlet oxygen generated by Ce6 photodynamic action might also oxidize the scotopsin moiety of rhodopsin, leading to direct oxidative rhodopsin activation. More attention needs to be paid to the latter respects of the far-red vision process of the deep-sea dragonfish, with potential translational values.
Collapse
Affiliation(s)
- Zong Jie Cui
- Department of Biology, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
- Ministry of Education Laboratory of Cell Proliferation and Regulation, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
| |
Collapse
|
7
|
Cui ZJ. To activate a G protein-coupled receptor permanently with cell surface photodynamic action in the gastrointestinal tract. World J Gastroenterol 2025; 31:102423. [PMID: 40182590 PMCID: PMC11962841 DOI: 10.3748/wjg.v31.i12.102423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 03/26/2025] Open
Abstract
Different from reversible agonist-stimulated receptor activation, singlet oxygen oxidation activates permanently G protein-coupled receptor (GPCR) cholecystokinin 1 (CCK1R) in type II photodynamic action, with soluble photosensitizer dyes (sulphonated aluminum phthalocyanine, λmax 675 nm) or genetically encoded protein photosensitizers (KillerRed λmax 585 nm; mini singlet oxygen generator λmax 450 nm), together with a pulse of light (37 mW/cm2, 1-2 minutes). Three lines of evidence shed light on the mechanism of GPCR activated by singlet oxygen (GPCR-ABSO): (1) CCK1R is quantitatively converted from dimer to monomer; (2) Transmembrane domain 3, a pharmacophore for permanent photodynamic CCK1R activation, can be transplanted to non-susceptible M3 acetylcholine receptor; and (3) Larger size of disordered region in intracellular loop 3 correlates with higher sensitivity to photodynamic CCK1R activation. GPCR-ABSO will add to the arsenal of engineered designer GPCR such as receptors activated solely by synthetic ligands and designer receptors exclusively activated by designer drugs, but show some clear advantages: Enhanced selectivity (double selectivity of localized photosensitizer and light illumination), long-lasting activation with no need for repeated drug administration, antagonist-binding site remains intact when needed, ease to apply to multiple GPCR. This type of permanent photodynamic activation may be applied to functional proteins other than GPCR.
Collapse
Affiliation(s)
- Zong-Jie Cui
- Department of Biology, College of Life Sciences, Beijing Normal University, Beijing 100875, China
- The Ministry of Education Laboratory for Cell Proliferation and Regulation, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
8
|
Zheng Z, Lu H, Wang X, Yang Z, Zhang Y, Li K, Shen C, Yin Z, Sha M, Ye J, Zhu L. Integrative analysis of genes provides insights into the molecular and immune characteristics of mitochondria-related genes in atherosclerosis. Genomics 2025; 117:111013. [PMID: 39914597 DOI: 10.1016/j.ygeno.2025.111013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/11/2025] [Accepted: 02/02/2025] [Indexed: 02/09/2025]
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by lipid accumulation in arterial walls. The role of the interplay between mitochondrial dysfunction and immune inflammation in atherosclerosis is still unclear. This study aimed to investigate the molecular characteristics and immune landscape of mitochondrial hub genes involved in atherosclerosis. Based on bioinformatics analysis, three hub Mitochondria-related DEGs (MitoDEGs), including OXCT1, UCP2, and CPT1B, were screened out and showed good diagnostic performance in identifying atherosclerosis patients and controls. Immune analysis demonstrated strong correlations between hub MitoDEGs and immune cells, such as macrophages and T cells. Additionally, the predicted transcription factors of these hub MitoDEGs were significantly enriched in Th17, Th1 and Th2 cell differentiation signaling pathways. Both cell and animal experiments confirmed the expression trends of OXCT1 and CPT1B observed in the bioinformatics analysis. These hub MitoDEGs may play an important role in coordinating mitochondrial metabolism in the immune inflammation of atherosclerosis.
Collapse
Affiliation(s)
- Zhipeng Zheng
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China
| | - Huimin Lu
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China
| | - Xiaowen Wang
- Nanjing University of Chinese Medicine, Nanjing 210023, China
| | | | - Yubin Zhang
- Dalian Medical University, Dalian 116000, China
| | - Kaiyuan Li
- Dalian Medical University, Dalian 116000, China
| | - Cheng Shen
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China
| | - Zhifeng Yin
- Jiangsu Hanjiang Biotechnology Co., LTD, Taizhou 225300, China
| | - Min Sha
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China.
| | - Jun Ye
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China.
| | - Li Zhu
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China; Nanjing University of Chinese Medicine, Nanjing 210023, China; Dalian Medical University, Dalian 116000, China.
| |
Collapse
|
9
|
Ducrest A, San‐Jose LM, Neuenschwander S, Schmid‐Siegert E, Simon C, Pagni M, Iseli C, Richter H, Guex N, Cumer T, Beaudoing E, Dupasquier M, Charruau P, Ducouret P, Xenarios I, Goudet J, Roulin A. Melanin and Neurotransmitter Signalling Genes Are Differentially Co-Expressed in Growing Feathers of White and Rufous Barn Owls. Pigment Cell Melanoma Res 2025; 38:e70001. [PMID: 39910963 PMCID: PMC11799826 DOI: 10.1111/pcmr.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 12/04/2024] [Accepted: 01/12/2025] [Indexed: 02/07/2025]
Abstract
Regulation of melanin-based pigmentation is complex, involving multiple genes. Because different genes can contribute to the same pigmentation phenotype, the genes identified in model organisms may not necessarily apply to wild species. In the barn owl (Tyto alba), ventral plumage colour ranges from white to rufous, with genetic variation in the melanocortin 1 receptor gene (MC1R) accounting for at least a third of this variation. In the present study, we used transcriptomic data to compare the gene expression profiles of growing feathers from nestlings with different MC1R genotypes. We identified 21 differentially expressed genes, nine of which are involved in melanogenesis, while seven are related to neurotransmitter function or synaptic activity. With the exception of CALB1, all of the differentially expressed genes were upregulated in rufous owls compared to white barn owls. To the best of our knowledge, this study is the first to link melanin production with neurotransmitter-related genes, and we discuss possible evolutionary explanations for this connection.
Collapse
Affiliation(s)
- Anne‐Lyse Ducrest
- Department of Ecology and Evolution, BiophoreUniversity of LausanneLausanneSwitzerland
| | - Luis M. San‐Jose
- Centre de Recherche sur la Biodiversité et l'Environnement (CRBE)Université de Toulouse, CNRS, IRD, Toulouse INP, Université Toulouse 3 – Paul Sabatier (UT3)ToulouseFrance
| | - Samuel Neuenschwander
- Vital‐IT, Swiss Institute of BioinformaticLausanneSwitzerland
- Department of Computational BiologyUniversity of LausanneLausanneSwitzerland
| | | | - Céline Simon
- Department of Ecology and Evolution, BiophoreUniversity of LausanneLausanneSwitzerland
| | - Marco Pagni
- Vital‐IT, Swiss Institute of BioinformaticLausanneSwitzerland
| | - Christian Iseli
- Bioinformatics Competence CenterUniversity of LausanneLausanneSwitzerland
- Bioinformatics Competence CenterEcole Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Hannes Richter
- Centre for Integrative Genomics, Genomic Technologies FacilityUniversity of LausanneLausanneSwitzerland
| | - Nicolas Guex
- Bioinformatics Competence CenterUniversity of LausanneLausanneSwitzerland
- Bioinformatics Competence CenterEcole Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Tristan Cumer
- Department of Ecology and Evolution, BiophoreUniversity of LausanneLausanneSwitzerland
| | - Emmanuel Beaudoing
- Centre for Integrative Genomics, Genomic Technologies FacilityUniversity of LausanneLausanneSwitzerland
| | - Mélanie Dupasquier
- Centre for Integrative Genomics, Genomic Technologies FacilityUniversity of LausanneLausanneSwitzerland
| | - Pauline Charruau
- Department of Ecology and Evolution, BiophoreUniversity of LausanneLausanneSwitzerland
| | - Pauline Ducouret
- Department of Ecology and Evolution, BiophoreUniversity of LausanneLausanneSwitzerland
| | - Ioannis Xenarios
- Agora CenterLausanneSwitzerland
- Health2030 Genome CenterGenèveSwitzerland
| | - Jérôme Goudet
- Department of Ecology and Evolution, BiophoreUniversity of LausanneLausanneSwitzerland
| | - Alexandre Roulin
- Department of Ecology and Evolution, BiophoreUniversity of LausanneLausanneSwitzerland
| |
Collapse
|
10
|
Wong A, Sun Q, Latif II, Karwi QG. Macrophage energy metabolism in cardiometabolic disease. Mol Cell Biochem 2025; 480:1763-1783. [PMID: 39198360 PMCID: PMC11842501 DOI: 10.1007/s11010-024-05099-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
In a rapidly expanding body of literature, the major role of energy metabolism in determining the response and polarization status of macrophages has been examined, and it is currently a very active area of research. The metabolic flux through different metabolic pathways in the macrophage is interconnected and complex and could influence the polarization of macrophages. Earlier studies suggested glucose flux through cytosolic glycolysis is a prerequisite to trigger the pro-inflammatory phenotypes of macrophages while proposing that fatty acid oxidation is essential to support anti-inflammatory responses by macrophages. However, recent studies have shown that this understanding is oversimplified and that the metabolic control of macrophage polarization is highly complex and not fully defined yet. In this review, we systematically reviewed and summarized the literature regarding the role of energy metabolism in controlling macrophage activity and how that might be altered in cardiometabolic diseases, namely heart failure, obesity, and diabetes. We critically appraised the experimental studies and methodologies in the published studies. We also highlighted the challenging concepts in macrophage metabolism and identified several research questions yet to be addressed in future investigations.
Collapse
Affiliation(s)
- Angela Wong
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, A1B 3V6, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Qiuyu Sun
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, A1B 3V6, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Ismail I Latif
- Department of Microbiology, College of Medicine, University of Diyala, Baqubaa, Diyala, Iraq
| | - Qutuba G Karwi
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, A1B 3V6, Canada.
| |
Collapse
|
11
|
Sun H, Kim MM, Ong YH, Dimofte A, Singhal S, Busch TM, Cengel KA, Zhu TC. Comprehensive reanalysis of light fluence distribution in pleural photodynamic therapy using standardized anatomical coordinates. Photochem Photobiol 2025. [PMID: 39838651 DOI: 10.1111/php.14063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/29/2024] [Accepted: 12/30/2024] [Indexed: 01/23/2025]
Abstract
Photodynamic therapy (PDT) has shown promise as an adjuvant treatment for malignant pleural mesothelioma when combined with surgical resection. Accurate light dosimetry is critical for treatment efficacy. This study presents an improved method for analyzing light fluence distribution in pleural PDT using a standardized anatomical coordinate system and advanced computational modeling. We utilized an infrared navigation system with an improved treatment delivery wand to track light delivery in real-time. The human chest cavity geometry was reconstructed and the pleura was mapped to a standardized coordinate system, allowing for direct comparisons across patients. Light fluence was calculated using both primary and scattered components, with a novel dual correction method applied to match measured values at detector locations. The standardized approach allowed for statistical analysis of light fluence distribution across anatomical regions in a cohort of 11 patients. Results showed acceptable light fluence uniformity with a standard deviation of 6.6% from the prescribed dose across patients. This comprehensive analysis provides insights for optimizing treatment protocols and lays the groundwork for future studies on singlet oxygen generation and its correlation with treatment outcomes in pleural PDT.
Collapse
Affiliation(s)
- Hongjing Sun
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michele M Kim
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yi Hong Ong
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Andreea Dimofte
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sunil Singhal
- Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Theresa M Busch
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Keith A Cengel
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Timothy C Zhu
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
12
|
Cai A, Shen D, Xiong Q, Ding J, Ding Y, Lin X, Chen L, Yao Q, Lin G, Chen R, Ganapathy V, Kou L. α-methyltryptophan-mediated protection against diabetic nephropathy in db/db mice as studied with a metabolomics approach. Front Pharmacol 2025; 15:1463673. [PMID: 39902076 PMCID: PMC11788373 DOI: 10.3389/fphar.2024.1463673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 12/31/2024] [Indexed: 02/05/2025] Open
Abstract
Introduction Diabetic nephropathy (DN), a major complication of diabetes, presents with poor clinical outcomes and affects patients throughout their lifetime. α-Methyltryptophan (α-MT) is a blocker of the amino acid transporter. SLC6A14 and also an inhibitor of indoleamine 2,3-dioxygenase-1 (IDO1). Methods In this study, we employed a nuclear magnetic resonance-based metabolomic approach to investigate the therapeutic effects of α-MT in a db/db mouse model of DN and explore the underlying molecular mechanisms. Results The results of the study demonstrated that α-MT significantly reduced the urinary excretion of albumin and creatinine, improved kidney function, and decreased renal fibrosis in db/db mice. Metabolomic analyses of kidney tissues and urine samples indicated that db/db mice displayed increased activity of the enzyme IDO1, and alongside pronounced metabolic disturbances. These disturbances are chiefly characterized by alterations in amino acid metabolism, energy production pathways, membrane biochemical features, and nicotinamide metabolism, all of which have been implicated in mTOR signaling and apoptotic pathways. Discussion Administration of α-MT to db/db mice showed evidence of IDO1 inhibition and rectification of metabolic dysfunctions with concurrent suppression of mTOR signaling and apoptosis. These findings highlight the potential of α-MT as a promising therapeutic agent for diabetic nephropathy.
Collapse
Affiliation(s)
- Aimin Cai
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dingchao Shen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qiushuang Xiong
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jie Ding
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yang Ding
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinlu Lin
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lijia Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qing Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Guangyong Lin
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ruijie Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Vadivel Ganapathy
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
13
|
Rai V. High-Fat Diet, Epigenetics, and Atherosclerosis: A Narrative Review. Nutrients 2024; 17:127. [PMID: 39796562 PMCID: PMC11722563 DOI: 10.3390/nu17010127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/29/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES Atherosclerosis is a chronic inflammatory disease developing and progressing in the presence of risk factors including hyperlipidemia, hypercholesterolemia, and chronic inflammation, among others. Atherosclerosis commonly precipitates as ischemic events, transient ischemic attacks, and myocardial infarction. Saturated fatty acids are risk factors; however, their association with epigenetics in the pathophysiology of atherosclerosis is not clearly understood. The preclinical and clinical trials associating atherosclerosis with epigenetics are scarcely documented, and most of the studies reported the use of drugs inhibiting methylation and histone modification to improve atherosclerosis. This narrative review aims to discuss various aspects and the association between a high-fat diet, epigenetic reprogramming, and atherosclerosis. METHODS A literature search with the keywords high-fat diet, epigenetics, and atherosclerosis, alone or in combination, was conducted to search for articles in the English language. Duplicate articles were removed, and articles related to the subject of this review article were included in this review. RESULTS A review of the literature suggests that a high-fat diet with saturated fatty acids is a risk factor for atherosclerosis, but this association is multifactorial, and epigenetics play a critical role. However, the connecting link and the underlying molecular and cellular mechanisms are not clearly understood yet and warrant more research. CONCLUSIONS A high-fat diet rich in saturated fatty acids is a risk factor for atherosclerosis involving epigenetic reprogramming and altered gene expression. The existing preclinical and clinical trials support the role of epigenetics and reversing it using drugs to attenuate atherosclerosis, but definitive evidence warrants larger clinical trials. Further, a high-fat diet in pregnant mothers can manifest as cardiovascular disease in offspring; caution must be taken in pregnant mothers for their diet and nutrients.
Collapse
Affiliation(s)
- Vikrant Rai
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
14
|
Cui ZJ. Special issue on "calcium signaling". BIOPHYSICS REPORTS 2024; 10:349-350. [PMID: 39758421 PMCID: PMC11693498 DOI: 10.52601/bpr.2024.240061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025] Open
Affiliation(s)
- Zong Jie Cui
- College of Life Sciences, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
15
|
Chen Y, You Y, Wang X, Jin Y, Zeng Y, Pan Z, Li D, Ling W. β-Hydroxybutyrate Alleviates Atherosclerotic Calcification by Inhibiting Endoplasmic Reticulum Stress-Mediated Apoptosis via AMPK/Nrf2 Pathway. Nutrients 2024; 17:111. [PMID: 39796543 PMCID: PMC11722964 DOI: 10.3390/nu17010111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/16/2024] [Accepted: 12/21/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Atherosclerotic calcification (AC) is a common feature of atherosclerotic cardiovascular disease. β-Hydroxybutyrate (BHB) has been identified as a molecule that influences cardiovascular disease. However, whether BHB can influence AC is still unknown. METHODS AND RESULTS In this study, ApoE-/- mice, fed a Western diet, were used to examine the effects of BHB on AC. Rat vascular smooth muscle cells (VSMCs) were used to verify the impacts of BHB on AC and to explore the underlying mechanisms. The results show that Western diet-challenged ApoE-/- mice, supplemented with BHB for 24 weeks, exhibited reduced calcified areas, calcium content, and alkaline phosphatase (ALP) activity in the aortas, as well as ameliorated severity of AC. Furthermore, BHB downregulated the expression of glucose-regulated protein 78 (GRP78) and C/EBP homologous protein (CHOP), thereby reducing endoplasmic reticulum stress (ERS) and ERS-mediated apoptosis in the aortas of the mice. Consistently, in vitro studies showed that BHB reduced ALP activity and calcium content in VSMCs, and inhibited VSMC calcification. Additionally, BHB suppressed ERS-mediated apoptosis in VSMCs. CONCLUSIONS In summary, the present results demonstrate that BHB can alleviate atherosclerotic calcification by inhibiting ERS-mediated apoptosis. Therefore, BHB may serve as a viable therapeutic agent for AC.
Collapse
Affiliation(s)
- Yu Chen
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China; (Y.C.); (Y.Y.); (X.W.); (Y.J.); (Y.Z.); (Z.P.); (D.L.)
| | - Yiran You
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China; (Y.C.); (Y.Y.); (X.W.); (Y.J.); (Y.Z.); (Z.P.); (D.L.)
| | - Xin Wang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China; (Y.C.); (Y.Y.); (X.W.); (Y.J.); (Y.Z.); (Z.P.); (D.L.)
| | - Yufeng Jin
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China; (Y.C.); (Y.Y.); (X.W.); (Y.J.); (Y.Z.); (Z.P.); (D.L.)
| | - Yupeng Zeng
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China; (Y.C.); (Y.Y.); (X.W.); (Y.J.); (Y.Z.); (Z.P.); (D.L.)
| | - Zhijun Pan
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China; (Y.C.); (Y.Y.); (X.W.); (Y.J.); (Y.Z.); (Z.P.); (D.L.)
| | - Dan Li
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China; (Y.C.); (Y.Y.); (X.W.); (Y.J.); (Y.Z.); (Z.P.); (D.L.)
| | - Wenhua Ling
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China; (Y.C.); (Y.Y.); (X.W.); (Y.J.); (Y.Z.); (Z.P.); (D.L.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China
- School of Public Health and Management, Ningxia Medical University, Yinchuan 750101, China
| |
Collapse
|
16
|
Xie XB, Shu Y, Cui ZJ. To activate NAD(P)H oxidase with a brief pulse of photodynamic action. FASEB J 2024; 38:e70246. [PMID: 39655710 PMCID: PMC11629461 DOI: 10.1096/fj.202402292r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/14/2024] [Accepted: 11/27/2024] [Indexed: 12/13/2024]
Abstract
Reduced nicotinamide adenine dinucleotide phosphate [NAD(P)H] oxidases (NOX) are a major cellular source of reactive oxygen species, regulating vital physiological functions, whose dys-regulation leads to a plethora of major diseases. Much effort has been made to develop varied types of NOX inhibitors, but biotechnologies for spatially and temporally controlled NOX activation, however, are not readily available. We previously found that ultraviolet A (UVA) irradiation activates NOX2 in rodent mast cells, to elicit persistent calcium spikes. NOX2 is composed of multiple subunits, making studies of its activation rather complicated. Here we show that the single-subunit nonrodent-expressing NOX5, when expressed ectopically in CHO-K1 cells, is activated by UVA irradiation (380 nm, 0.1-12 mW/cm2, 1.5 min) inducing repetitive calcium spikes, as monitored by Fura-2 fluorescent calcium imaging. UVA-elicited calcium oscillations are inhibited by NOX inhibitor diphenyleneiodonium chloride (DPI) and blocked by singlet oxygen (1O2) quencher Trolox-C (300 μM). A brief pulse of photodynamic action (1.5 min) with photosensitizer sulfonated aluminum phthalocyanine (SALPC 2 μM, 675 nm, 85 mW/cm2) in NOX5-CHO-K1 cells, or with genetically encoded protein photosensitizer miniSOG fused to N-terminus of NOX5 (450 nm, 85 mW/cm2) in miniSOG-NOX5-CHO-K1 cells, induces persistent calcium oscillations, which are blocked by DPI. In the presence of Trolox-C, miniSOG photodynamic action no longer induces any calcium increases in miniSOG-NOX5-CHO-K1 cells. DUOX2 in human thyroid follicular cells SW579 and in DUOX2-CHO-K1 cells is similarly activated by UVA irradiation and SALPC photodynamic action. These data together suggest that NOX is activated with a brief pulse of photodynamic action.
Collapse
Affiliation(s)
- Xiao Bing Xie
- College of Life SciencesBeijing Normal UniversityBeijingChina
| | - Yu Shu
- College of Life SciencesBeijing Normal UniversityBeijingChina
| | - Zong Jie Cui
- College of Life SciencesBeijing Normal UniversityBeijingChina
| |
Collapse
|
17
|
Prieto-Montero R, Herrera L, Tejón M, Albaya A, Chiara JL, Fanarraga ML, Martínez-Martínez V. Exploring Gluconamide-Modified Silica Nanoparticles of Different Sizes as Effective Carriers for Antimicrobial Photodynamic Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1982. [PMID: 39728518 PMCID: PMC11728795 DOI: 10.3390/nano14241982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/07/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024]
Abstract
Antimicrobial resistance (AMR), a consequence of the ability of microorganisms, especially bacteria, to develop resistance against conventional antibiotics, hampering the treatment of common infections, is recognized as one of the most imperative health threats of this century. Antibacterial photodynamic therapy (aPDT) has emerged as a promising alternative strategy, utilizing photosensitizers activated by light to generate reactive oxygen species (ROS) that kill pathogens without inducing resistance. In this work, we synthesized silica nanoparticles (NPs) of different sizes (20 nm, 80 nm, and 250 nm) functionalized with the photosensitizer Rose Bengal (RB) and a gluconamide ligand, which targets Gram-negative bacteria, to assess their potential in aPDT. Comprehensive characterization, including dynamic light scattering (DLS) and photophysical analysis, confirmed the stability and effective singlet oxygen production of the functionalized nanoparticles. Although the surface loading density of Rose Bengal was constant at the nanoparticle external surface, RB loading (in mg/g nanoparticle) was size-dependent, decreasing with increasing nanoparticle diameter. Further, the spherical geometry of nanoparticles favored smaller nanoparticles for antibacterial PDT, as this maximizes the surface contact area with the bacteria wall, with the smallest (20 nm) and intermediate (80 nm) particles being more promising. Bacterial assays in E. coli revealed minimal dark toxicity and significant light-activated phototoxicity for the RB-loaded nanoparticles. The addition of gluconamide notably enhanced phototoxic activity, particularly in the smallest nanoparticles (RB-G-20@SiNP), which demonstrated the highest phototoxicity-to-cytotoxicity ratio. These findings indicate that small, gluconamide-functionalized silica nanoparticles are highly effective for targeted aPDT, offering a robust strategy to combat AMR.
Collapse
Affiliation(s)
- Ruth Prieto-Montero
- Departamento de Química Física, Facultad de Ciencia y Tecnología, Universidad del País Vasco, UPV-EHU, Apartado 644, 48080 Bilbao, Spain; (L.H.); (M.T.); (A.A.)
| | - Lucia Herrera
- Departamento de Química Física, Facultad de Ciencia y Tecnología, Universidad del País Vasco, UPV-EHU, Apartado 644, 48080 Bilbao, Spain; (L.H.); (M.T.); (A.A.)
| | - Maite Tejón
- Departamento de Química Física, Facultad de Ciencia y Tecnología, Universidad del País Vasco, UPV-EHU, Apartado 644, 48080 Bilbao, Spain; (L.H.); (M.T.); (A.A.)
| | - Andrea Albaya
- Departamento de Química Física, Facultad de Ciencia y Tecnología, Universidad del País Vasco, UPV-EHU, Apartado 644, 48080 Bilbao, Spain; (L.H.); (M.T.); (A.A.)
- Instituto de Química Orgánica General (IQOG-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain;
| | - Jose Luis Chiara
- Instituto de Química Orgánica General (IQOG-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain;
| | - Mónica L. Fanarraga
- Grupo de Nanomedicina Instituto de Investigación Valdecilla-IDIVAL, Universidad de Cantabria, Herrera Oria s/n, 39011 Santander, Spain;
| | - Virginia Martínez-Martínez
- Departamento de Química Física, Facultad de Ciencia y Tecnología, Universidad del País Vasco, UPV-EHU, Apartado 644, 48080 Bilbao, Spain; (L.H.); (M.T.); (A.A.)
| |
Collapse
|
18
|
Bai YP, Xing YJ, Ma T, Li K, Zhang T, Wang DG, Wan SJ, Zhang CW, Sun Y, Wang MY, Wang GD, Pei WJ, Lv K, Zhang Y, Kong X. β-Hydroxybutyrate suppresses M1 macrophage polarization through β-hydroxybutyrylation of the STAT1 protein. Cell Death Dis 2024; 15:874. [PMID: 39627223 PMCID: PMC11615246 DOI: 10.1038/s41419-024-07268-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 11/16/2024] [Accepted: 11/27/2024] [Indexed: 12/06/2024]
Abstract
β-Hydroxybutyrate (β-OHB), the primary ketone body, is a bioactive metabolite that acts as both an energy substrate and a signaling molecule. Recent studies found that β-OHB inhibits the production of pro-inflammatory cytokines in macrophages, but its underlying molecular mechanisms have not yet been fully elucidated. Lysine β-hydroxybutyrylation (Kbhb), a post-translational modification mediated by β-OHB, plays a key role in regulating the expression and activity of modified proteins. However, whether macrophages undergo protein Kbhb and whether Kbhb modification regulates macrophage polarization remains largely unknown. In this study, treatment with β-OHB and ketone ester significantly decreased the lipopolysaccharide (LPS)-induced enhancement of the M1 phenotype of mouse bone marrow-derived macrophages (BMDMs), RAW264.7 cells, and peritoneal macrophages (PMs) in vitro and in vivo. Moreover, β-OHB treatment induced global protein Kbhb, which is associated with the regulation of macrophage M1 polarization. Proteome-wide Kbhb analysis in β-OHB-treated BMDMs revealed 3469 Kbhb modification sites within 1549 proteins, among which interleukin-12-responding proteins were significantly upregulated. Our results indicated that β-OHB regulated M1 macrophage polarization by inducing Kbhb modification of the signal transducer and activator of transcription 1 (STAT1) K679 site, which inhibited its LPS-induced phosphorylation and transcription. Altogether, our study demonstrated the presence of a widespread Kbhb landscape in the β-OHB-treated macrophages and provided novel insights into the anti-inflammatory effects of β-OHB.
Collapse
Affiliation(s)
- Ya-Ping Bai
- Anhui Provincial Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wannan Medical College, Wuhu, China
- College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Yu-Jie Xing
- Anhui Provincial Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wannan Medical College, Wuhu, China
| | - Tao Ma
- Anhui Provincial Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wannan Medical College, Wuhu, China
| | - Kai Li
- Anhui Provincial Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wannan Medical College, Wuhu, China
| | - Teng Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - De-Guo Wang
- Department of Gerontology, Geriatric Endocrinology unit, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
- National Clinical Research Center for Geriatric Diseases, Anhui Provincial Sub-center, Wuhu, China
| | - Shu-Jun Wan
- Anhui Provincial Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wannan Medical College, Wuhu, China
| | - Cui-Wei Zhang
- Department of Gerontology, Geriatric Endocrinology unit, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
- National Clinical Research Center for Geriatric Diseases, Anhui Provincial Sub-center, Wuhu, China
| | - Yue Sun
- Department of Gastroenterology, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Meng-Yan Wang
- School of Pharmacy, Wannan Medical College, Wuhu, China
| | - Guo-Dong Wang
- School of Pharmacy, Wannan Medical College, Wuhu, China
| | - Wen-Jun Pei
- Anhui Province Key Laboratory of Biological Macro-molecules Research, Wannan Medical College, Wuhu, China
| | - Kun Lv
- Anhui Provincial Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wannan Medical College, Wuhu, China.
| | - Yan Zhang
- Anhui Provincial Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wannan Medical College, Wuhu, China.
- Department of Gastroenterology, The First Affiliated Hospital of Wannan Medical College, Wuhu, China.
| | - Xiang Kong
- Anhui Provincial Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wannan Medical College, Wuhu, China.
- College of Life Sciences, Anhui Normal University, Wuhu, China.
- Department of Gerontology, Geriatric Endocrinology unit, The First Affiliated Hospital of Wannan Medical College, Wuhu, China.
- National Clinical Research Center for Geriatric Diseases, Anhui Provincial Sub-center, Wuhu, China.
| |
Collapse
|
19
|
Yu B, Yuan C, Chen J, Zhou Z, Zhang Y, Su M, Wei D, Wu P. TMAO induces pyroptosis of vascular endothelial cells and atherosclerosis in ApoE -/- mice via MBOAT2-mediated endoplasmic reticulum stress. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159559. [PMID: 39179098 DOI: 10.1016/j.bbalip.2024.159559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/12/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
Trimethylamine N-oxide (TMAO), a metabolite produced by intestinal flora, is recognized as an independent risk factor for atherosclerosis and atherosclerotic cardiovascular diseases. However, the underlying mechanism remains poorly understood. Here, we showed that dietary TMAO supplementation accelerates atherosclerosis in ApoE-/- mice. Pyroptosis and the expression of phospholipid-modifying enzyme MBOAT2 were increased in endothelial cells within atherosclerotic lesions. Genetic upregulation of MBOAT2 via adeno-associated virus with endothelium-specific promoter results in increased atherosclerotic lesions in ApoE-/- mice. Mechanistically, the overexpression of MBOAT2 disrupted glycerophospholipid metabolism and induced endothelial cell pyroptosis in an Endoplasmic reticulum stress-dependent manner. These data reveal that TMAO promotes endothelial cell pyroptosis and the progression of atherosclerotic lesions through the upregulation of MBOAT2, indicating that MBOAT2 is a promising therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Bo Yu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Chuchu Yuan
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Jinna Chen
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zhixiang Zhou
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yile Zhang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Ming Su
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Dangheng Wei
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| | - Peng Wu
- Hengyang Maternal and Child Health Hospital, Hengyang 421001, Hunan Province, China.
| |
Collapse
|
20
|
Bhuvaneshwari S, Venkataraman K, Sankaranarayanan K. Exploring potential ion channel targets for rheumatoid arthritis: combination of network analysis and gene expression analysis. Biotechnol Appl Biochem 2024; 71:1405-1427. [PMID: 39049164 DOI: 10.1002/bab.2638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 06/29/2024] [Indexed: 07/27/2024]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic inflammation of the synovial membrane that leads to the destruction of cartilage and bone. Currently, pharmacological targeting of ion channels is being increasingly recognized as an attractive and feasible strategy for the treatment of RA. The present work employs a network analysis approach to predict the most promising ion channel target for potential RA-treating drugs. A protein-protein interaction map was generated for 343 genes associated with inflammation in RA and ion channel genes using Search Tool for the Retrieval of Interacting Genes and visualized using Cytoscape. Based on the betweenness centrality and traffic values as key topological parameters, 17 hub nodes were identified, including FOS (9800.85), tumor necrosis factor (3654.60), TGFB1 (3305.75), and VEGFA (3052.88). The backbone network constructed with these 17 hub genes was intensely analyzed to identify the most promising ion channel target using network analyzer. Calcium permeating ion channels, especially store-operated calcium entry channels, and their associated regulatory proteins were found to highly interact with RA inflammatory hub genes. This significant ion channel target for RA identified by theoretical and statistical studies was further validated by a pilot case-control gene expression study. Experimental verification of the above findings in 75 RA cases and 25 controls showed increased ORAI1 expression. Thus, with a combination of network analysis approach and gene expression studies, we have explored potential targets for RA treatment.
Collapse
Affiliation(s)
- Sampath Bhuvaneshwari
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology, Anna University, Chennai, India
| | | | - Kavitha Sankaranarayanan
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology, Anna University, Chennai, India
| |
Collapse
|
21
|
Zhuang H, Ren X, Zhang Y, Li H, Zhou P. β-Hydroxybutyrate enhances chondrocyte mitophagy and reduces cartilage degeneration in osteoarthritis via the HCAR2/AMPK/PINK1/Parkin pathway. Aging Cell 2024; 23:e14294. [PMID: 39126207 PMCID: PMC11561673 DOI: 10.1111/acel.14294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/28/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
Osteoarthritis (OA) is widely recognized as the prevailing joint disease associated with aging. The ketogenic diet (KD) has been postulated to impede the advancement of various inflammatory ailments. β-Hydroxybutyrate (βOHB), a prominent constituent of ketone bodies, has recently been proposed to possess crucial signaling capabilities. In this study, we propose to explore the role and mechanism of βOHB in OA. Tissue staining and inflammatory factor assay were employed to evaluate the impacts of KD and βOHB on OA rats. The oxidative stress conditions in chondrocytes were induced using tert-butyl hydroperoxide (TBHP). The mechanisms were determined using the siRNA of hydroxycarboxylic acid receptor 2 (HCAR2), the antagonist of adenosine monophosphate-activated protein kinase (AMPK), and the inhibitor of mitophagy. The administration of KD demonstrated a reduction in pathological damage to cartilage, as well as a decrease in plasma levels of inflammatory factors. Furthermore, it resulted in an increase in the concentration of βOHB in the blood and synovial fluid. In vitro experiments showed that βOHB facilitated mitophagy and adenosine triphosphate production. Besides, βOHB mitigated chondrocyte senescence, inflammatory factors secretion, extracellular matrix degradation, and apoptosis induced by TBHP. Subsequent investigations indicated that the protective effects of βOHB were no longer observed following the knockdown of HCAR2, the antagonist of AMPK, or the inhibitor of mitophagy. Moreover, in vivo studies suggested that βOHB played a protective role by targeting the HCAR2-AMPK-PINK1 axis. In conclusion, βOHB enhanced chondrocyte mitophagy through the HCAR2/AMPK/PINK1/Parkin pathway, offering a potential therapeutic approach for the treatment of OA.
Collapse
Affiliation(s)
- Huangming Zhuang
- Department of OrthopedicsRenmin Hospital of Wuhan UniversityWuhanChina
| | - Xunshan Ren
- Department of OrthopedicsRenmin Hospital of Wuhan UniversityWuhanChina
| | - Yuelong Zhang
- Department of OrthopedicsRenmin Hospital of Wuhan UniversityWuhanChina
| | - Huajie Li
- Department of OrthopedicsRenmin Hospital of Wuhan UniversityWuhanChina
| | - Panghu Zhou
- Department of OrthopedicsRenmin Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
22
|
Pavlicev M, DiFrisco J, Love AC, Wagner GP. Metabolic complementation between cells drives the evolution of tissues and organs. Biol Lett 2024; 20:20240490. [PMID: 39561800 PMCID: PMC11583983 DOI: 10.1098/rsbl.2024.0490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/04/2024] [Accepted: 10/11/2024] [Indexed: 11/21/2024] Open
Abstract
Although evolutionary transitions of individuality have been extensively theorized, little attention has been paid to the origin of levels of organization within organisms. How and why do specialized cells become organized into specialized tissues or organs? What spurs a transition in organizational level in cases where the function is already present in constituent cell types? We propose a hypothesis for this kind of evolutionary transition based on two features of cellular metabolism: metabolic constraints on functional performance and the capacity for metabolic complementation between parenchymal and supporting cells. These features suggest a scenario whereby pre-existing specialized cell types are integrated into tissues when changes to the internal or external environment favour offloading metabolic burdens from a primary specialized cell type onto supporting cells. We illustrate this process of 'supra-functionalization' using the nervous system and pancreas.
Collapse
Affiliation(s)
- Mihaela Pavlicev
- Department of Evolutionary Biology, University of Vienna, Vienna, Austria
- Complexity Science Hub, Vienna, Austria
- Konrad Lorenz Institute of Evolution and Cognition Research, Klosterneuburg, Austria
| | - J DiFrisco
- Theoretical Biology Lab, The Francis Crick Institute, London, UK
| | - Alan C Love
- Department of Philosophy & Minnesota Center for Philosophy of Science, University of Minnesota, Minneapolis, MN, USA
| | - Günter P Wagner
- Department of Evolutionary Biology, University of Vienna, Vienna, Austria
- Konrad Lorenz Institute of Evolution and Cognition Research, Klosterneuburg, Austria
- Yale University, New Haven, CT, USA
- Texas A&M, Hagler Institute for Advanced Study, College Station, TX, USA
| |
Collapse
|
23
|
Cui ZJ. The Golgi Ca 2+ stores, and original contributions by Prof. Shao Bai Xue. BIOPHYSICS REPORTS 2024; 10:266-273. [PMID: 39539285 PMCID: PMC11554580 DOI: 10.52601/bpr.2023.230015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/06/2023] [Indexed: 11/16/2024] Open
Abstract
The Golgi apparatus serves as a distinct part of intracellular Ca2+ stores. Here, the early discovery by Professor Shao Bai Xue is reviewed, and the recent progress in the field is outlined. Golgi Ca2+ stores-related functional proteins, such as secretory pathway Ca2+ ATPases (SPCA1/2) and the Golgi-specific Ca2+ releasing channel Golgi anti-apoptotic protein (GAAP), as well as the recently defined Golgi-specific Ca2+ release agent emetine, collectively corroborate the concept of the Golgi apparatus as unique internal Ca2+ stores.
Collapse
Affiliation(s)
- Zong Jie Cui
- College of Life Sciences, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
24
|
Wang SQ. Ca 2+ sparks and beyond. BIOPHYSICS REPORTS 2024; 10:257-258. [PMID: 39539280 PMCID: PMC11554576 DOI: 10.52601/bpr.2024.240909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Affiliation(s)
- Shi-Qiang Wang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
25
|
Liu K, Yang Y, Yang JH. Underlying mechanisms of ketotherapy in heart failure: current evidence for clinical implementations. Front Pharmacol 2024; 15:1463381. [PMID: 39512825 PMCID: PMC11540999 DOI: 10.3389/fphar.2024.1463381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/15/2024] [Indexed: 11/15/2024] Open
Abstract
Heart failure (HF) is a life-threatening cardiac syndrome characterized by high morbidity and mortality, but current anti-heart failure therapies have limited efficacy, necessitating the urgent development of new treatment drugs. Exogenous ketone supplementation helps prevent heart failure development in HF models, but therapeutic ketosis in failing hearts has not been systematically elucidated, limiting the use of ketones to treat HF. Here, we summarize current evidence supporting ketotherapy in HF, emphasizing ketone metabolism in the failing heart, metabolic and non-metabolic therapeutic effects, and mechanisms of ketotherapy in HF, involving the dynamics within the mitochondria. We also discuss clinical strategies for therapeutic ketosis, aiming to deepen the understanding of the characteristics of ketone metabolism, including mitochondrial involvement, and its clinical therapeutic potential in HF.
Collapse
Affiliation(s)
| | | | - Jing-Hua Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
26
|
Zhang Y, Jiang ZT, Wang Y, Wang HY, Hong S, Li W, Guo DS, Zhang X. A Supramolecular Nanoformulation with Adaptive Photothermal/Photodynamic Transformation for Preventing Dental Caries. ACS NANO 2024; 18:27340-27357. [PMID: 39316824 DOI: 10.1021/acsnano.4c06051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
In the context of an increasingly escalating antibiotics crisis, phototherapy has emerged as a promising therapeutic approach due to its inherent advantages, including high selectivity, noninvasiveness, and low drug resistance. Photothermal therapy (PTT) and photodynamic therapy (PDT) are two complementary and promising phototherapies albeit with inherent limitations, noted as the challenges in achieving precise heat confinement and the associated risk of off-target damage for PTT, while the constraints due to the hypoxic microenvironment are prevalent in biofilms faced by PDT. Herein, we have designed a supramolecular nanoformulation that leverages the complexation-induced quenching of guanidinium-modified calix[5]arene grafted with fluorocarbon chains (GC5AF5), the efficient recognition of adenosine triphosphate (ATP), and the oxygen-carrying capacity of the fluorocarbon chain. This intelligent nanoformulation enables the adaptive enhancement of both photothermal therapy (PTT) and photodynamic therapy (PDT), allowing for on-demand switching between the two modalities. Our nanoformulation utilizes ATP released by dead bacteria to accelerate the elimination of biofilms, rendering bacteria unable to resist while minimizing harm to healthy tissues. This research highlights the particular recognition and assembly capabilities of macrocycles, offering a promising strategy for creating potent, combined antibiofilm therapies.
Collapse
Affiliation(s)
- Yufei Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of functional polymer materials, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Ze-Tao Jiang
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials of Ministry of Education, Frontiers Science Center for New Organic Matter. Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China
| | - Yuxia Wang
- Department of Cariology and Endodontics, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300041, China
| | - Huan-Yu Wang
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials of Ministry of Education, Frontiers Science Center for New Organic Matter. Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China
| | - Shihao Hong
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of functional polymer materials, College of Chemistry, Nankai University, Tianjin 300071, China
| | - WenBo Li
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials of Ministry of Education, Frontiers Science Center for New Organic Matter. Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China
| | - Dong-Sheng Guo
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials of Ministry of Education, Frontiers Science Center for New Organic Matter. Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China
| | - Xinge Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of functional polymer materials, College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
27
|
Li M, Ma S. A review of healthy role of dietary fiber in modulating chronic diseases. Food Res Int 2024; 191:114682. [PMID: 39059940 DOI: 10.1016/j.foodres.2024.114682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/14/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024]
Abstract
Dietary fiber (DF) is considered an interventional diet beneficial for human health. High DF intake effectively reduces the incidence of three major chronic diseases, type 2 diabetes (T2DM), cardiovascular disease (CVD), and colorectal cancer (CRC). The health benefits of DF are closely related to their physicochemical properties with major positive roles in human digestion and intestinal health. However, mechanisms linking DF with diseases remain unclear. The development of genomics, metabolomics, and immunology, and the powerful combination of animal models and clinical trials, have facilitated a better understanding of the relationships between DF and diseases. Accumulating evidence suggests that the physical existence of DF and DF-microbiota interaction are the key parameters controlling the action mechanisms of DF in chronic diseases. Therefore, this review discusses the potential mechanism of DF modulating T2DM, CVD, and CRC, therefore providing a theoretical basis for more effective use of DF to intervene in chronic diseases.
Collapse
Affiliation(s)
- Mengyuan Li
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou, China
| | - Sen Ma
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou, China.
| |
Collapse
|
28
|
Cheng H, Kong CHT, James AF, Cannell MB, Hancox JC. Modulation of Spontaneous Action Potential Rate by Inositol Trisphosphate in Myocytes from the Rabbit Atrioventricular Node. Cells 2024; 13:1455. [PMID: 39273026 PMCID: PMC11394215 DOI: 10.3390/cells13171455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/08/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
The atrioventricular node (AVN) is a key component of the cardiac conduction system and takes over pacemaking of the ventricles if the sinoatrial node fails. IP3 (inositol 1,4,5 trisphosphate) can modulate excitability of myocytes from other regions of the heart, but it is not known whether IP3 receptor (IP3-R) activation modulates AVN cell pacemaking. Consequently, this study investigated effects of IP3 on spontaneous action potentials (APs) from AVN cells isolated from rabbit hearts. Immunohistochemistry and confocal imaging demonstrated the presence of IP3-R2 in isolated AVN cells, with partial overlap with RyR2 ryanodine receptors seen in co-labelling experiments. In whole-cell recordings at physiological temperature, application of 10 µM membrane-permeant Bt3-(1,4,5)IP3-AM accelerated spontaneous AP rate and increased diastolic depolarization rate, without direct effects on ICa,L, IKr, If or INCX. By contrast, application via the patch pipette of 5 µM of the IP3-R inhibitor xestospongin C led to a slowing in spontaneous AP rate and prevented 10 µM Bt3-(1,4,5)IP3-AM application from increasing the AP rate. UV excitation of AVN cells loaded with caged-IP3 led to an acceleration in AP rate, the magnitude of which increased with the extent of UV excitation. 2-APB slowed spontaneous AP rate, consistent with a role for constitutive IP3-R activity; however, it was also found to inhibit ICa,L and IKr, confounding its use for studying IP3-R. Under AP voltage clamp, UV excitation of AVN cells loaded with caged IP3 activated an inward current during diastolic depolarization. Collectively, these results demonstrate that IP3 can modulate AVN cell pacemaking rate.
Collapse
Affiliation(s)
| | | | | | | | - Jules C. Hancox
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK; (H.C.); (C.H.T.K.); (A.F.J.); (M.B.C.)
| |
Collapse
|
29
|
Vertiprakhov V, Grozina A, Fisinin V. Editorial: The role of the pancreas in poultry. Front Physiol 2024; 15:1463203. [PMID: 39263624 PMCID: PMC11387553 DOI: 10.3389/fphys.2024.1463203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 08/20/2024] [Indexed: 09/13/2024] Open
Affiliation(s)
| | - Alena Grozina
- All-Russian Scientific Research and Technological Institute of Poultry, Russian Academy of Agricultural Sciences, Moscow, Russia
| | - Vladimir Fisinin
- All-Russian Scientific Research and Technological Institute of Poultry, Russian Academy of Agricultural Sciences, Moscow, Russia
| |
Collapse
|
30
|
Petusseau AF, Ochoa M, Reed M, Doyley MM, Hasan T, Bruza P, Pogue BW. Pressure-enhanced sensing of tissue oxygenation via endogenous porphyrin: Implications for dynamic visualization of cancer in surgery. Proc Natl Acad Sci U S A 2024; 121:e2405628121. [PMID: 39141355 PMCID: PMC11348300 DOI: 10.1073/pnas.2405628121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/21/2024] [Indexed: 08/15/2024] Open
Abstract
Fluorescence guidance is routinely used in surgery to enhance perfusion contrast in multiple types of diseases. Pressure-enhanced sensing of tissue oxygenation (PRESTO) via fluorescence is a technique extensively analyzed here, that uses an FDA-approved human precursor molecule, 5-aminolevulinic acid (ALA), to stimulate a unique delayed fluorescence signal that is representative of tissue hypoxia. The ALA precontrast agent is metabolized in most tissues into a red fluorescent molecule, protoporphyrin IX (PpIX), which has both prompt fluorescence, indicative of the concentration, and a delayed fluorescence, that is amplified in low tissue oxygen situations. Applied pressure from palpation induces transient capillary stasis and a resulting transient PRESTO contrast, dominant when there is near hypoxia. This study examined the kinetics and behavior of this effect in both normal and tumor tissues, with a prolonged high PRESTO contrast (contrast to background of 7.3) across 5 tumor models, due to sluggish capillaries and inhibited vasodynamics. This tissue function imaging approach is a fundamentally unique tool for real-time palpation-induced tissue response in vivo, relevant for chronic hypoxia, such as vascular diseases or oncologic surgery.
Collapse
Affiliation(s)
| | - Marien Ochoa
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI53705
| | - Matthew Reed
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI53705
| | - Marvin M. Doyley
- Department of Electrical and Computer Engineering, University of Rochester, Rochester, NY14627
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Petr Bruza
- Thayer School of Engineering, Dartmouth College, Hanover, NH03755
| | - Brian W. Pogue
- Thayer School of Engineering, Dartmouth College, Hanover, NH03755
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI53705
| |
Collapse
|
31
|
Jiang L, Xie XB, Zhang L, Tang Y, Zhu X, Huang Y, Hong Y, Hansson BS, Cui ZJ, Han Q. Activation of the G protein-coupled sulfakinin receptor inhibits blood meal intake in the mosquito Aedes aegypti. FASEB J 2024; 38:e23864. [PMID: 39109513 PMCID: PMC11607638 DOI: 10.1096/fj.202401165r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/04/2024] [Accepted: 07/23/2024] [Indexed: 12/02/2024]
Abstract
Little is known about the blood-feeding physiology of arbovirus vector Aedes aegypti although this type of mosquito is known to transmit infectious diseases dengue, Zika, yellow fever, and chikungunya. Blood feeding in the female A. aegypti mosquito is essential for egg maturation and for transmission of disease agents between human subjects. Here, we identify the A. aegypti sulfakinin receptor gene SKR from the A. aegypti genome and show that SKR is expressed at different developmental stages and in varied anatomical localizations in the adult mosquito (at three days after eclosion), with particularly high expression in the CNS. Knockingdown sulfakinin and sulfakinin receptor gene expression in the female A. aegypti results in increased blood meal intake, but microinjection in the thorax of the sulfakinin peptide 1 and 2 both inhibits dose dependently blood meal intake (and delays the time course of blood intake), which is reversible with receptor antagonist. Sulfakinin receptor expressed ectopically in mammalian cells CHO-K1 responds to sulfakinin stimulation with persistent calcium spikes, blockable with receptor antagonist. These data together suggest that activation of the Gq protein-coupled (i.e., calcium-mobilizing) sulfakinin receptor inhibits blood meal intake in female A. aegypti mosquitoes and could serve as a strategic node for the future control of A. aegypti mosquito reproduction/population and disease transmission.
Collapse
Affiliation(s)
- Linlong Jiang
- Laboratory of Tropical Veterinary Medicine and Vector Biology, School of Life SciencesHainan UniversityHaikouHainanChina
- Hainan Province Key Laboratory of One HealthCollaborative Innovation Center of One HealthHainan UniversityHaikouHainanChina
| | - Xiao Bing Xie
- College of Life SciencesBeijing Normal UniversityBeijingChina
| | - Lei Zhang
- Laboratory of Tropical Veterinary Medicine and Vector Biology, School of Life SciencesHainan UniversityHaikouHainanChina
- Hainan Province Key Laboratory of One HealthCollaborative Innovation Center of One HealthHainan UniversityHaikouHainanChina
- Hainan International One Health InstituteHainan UniversityHaikouHainanChina
| | - Yu Tang
- Laboratory of Tropical Veterinary Medicine and Vector Biology, School of Life SciencesHainan UniversityHaikouHainanChina
- Hainan Province Key Laboratory of One HealthCollaborative Innovation Center of One HealthHainan UniversityHaikouHainanChina
| | - Xiaojing Zhu
- Laboratory of Tropical Veterinary Medicine and Vector Biology, School of Life SciencesHainan UniversityHaikouHainanChina
- Hainan Province Key Laboratory of One HealthCollaborative Innovation Center of One HealthHainan UniversityHaikouHainanChina
| | - Yuqi Huang
- Laboratory of Tropical Veterinary Medicine and Vector Biology, School of Life SciencesHainan UniversityHaikouHainanChina
- Hainan Province Key Laboratory of One HealthCollaborative Innovation Center of One HealthHainan UniversityHaikouHainanChina
| | - Yue Hong
- Hainan Province Key Laboratory of One HealthCollaborative Innovation Center of One HealthHainan UniversityHaikouHainanChina
| | - Bill S. Hansson
- Department of Evolutionary NeuroethologyMax Planck Institute for Chemical EcologyJenaGermany
| | - Zong Jie Cui
- College of Life SciencesBeijing Normal UniversityBeijingChina
| | - Qian Han
- Laboratory of Tropical Veterinary Medicine and Vector Biology, School of Life SciencesHainan UniversityHaikouHainanChina
- Hainan Province Key Laboratory of One HealthCollaborative Innovation Center of One HealthHainan UniversityHaikouHainanChina
- Hainan International One Health InstituteHainan UniversityHaikouHainanChina
| |
Collapse
|
32
|
Lun H, Li P, Li J, Liu F. The effect of intestinal flora metabolites on macrophage polarization. Heliyon 2024; 10:e35755. [PMID: 39170251 PMCID: PMC11337042 DOI: 10.1016/j.heliyon.2024.e35755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/28/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024] Open
Abstract
Intestinal flora metabolites played a crucial role in immunomodulation by influencing host immune responses through various pathways. Macrophages, as a type of innate immune cell, were essential in chemotaxis, phagocytosis, inflammatory responses, and microbial elimination. Different macrophage phenotypes had distinct biological functions, regulated by diverse factors and mechanisms. Advances in intestinal flora sequencing and metabolomics have enhanced understanding of how intestinal flora metabolites affect macrophage phenotypes and functions. These metabolites had varying effects on macrophage polarization and different mechanisms of influence. This study summarized the impact of gut microbiota metabolites on macrophage phenotype and function, along with the underlying mechanisms associated with different metabolites produced by intestinal flora.
Collapse
Affiliation(s)
- Hengzhong Lun
- Department of Clinical Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| | - Peilong Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Juan Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Fenfen Liu
- Department of Nephrology, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| |
Collapse
|
33
|
Yang T, Peng J, Zhang Z, Chen Y, Liu Z, Jiang L, Jin L, Han M, Su B, Li Y. Emerging therapeutic strategies targeting extracellular histones for critical and inflammatory diseases: an updated narrative review. Front Immunol 2024; 15:1438984. [PMID: 39206200 PMCID: PMC11349558 DOI: 10.3389/fimmu.2024.1438984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Extracellular histones are crucial damage-associated molecular patterns involved in the development and progression of multiple critical and inflammatory diseases, such as sepsis, pancreatitis, trauma, acute liver failure, acute respiratory distress syndrome, vasculitis and arthritis. During the past decade, the physiopathologic mechanisms of histone-mediated hyperinflammation, endothelial dysfunction, coagulation activation, neuroimmune injury and organ dysfunction in diseases have been systematically elucidated. Emerging preclinical evidence further shows that anti-histone strategies with either their neutralizers (heparin, heparinoids, nature plasma proteins, small anion molecules and nanomedicines, etc.) or extracorporeal blood purification techniques can significantly alleviate histone-induced deleterious effects, and thus improve the outcomes of histone-related critical and inflammatory animal models. However, a systemic evaluation of the efficacy and safety of these histone-targeting therapeutic strategies is currently lacking. In this review, we first update our latest understanding of the underlying molecular mechanisms of histone-induced hyperinflammation, endothelial dysfunction, coagulopathy, and organ dysfunction. Then, we summarize the latest advances in histone-targeting therapy strategies with heparin, anti-histone antibodies, histone-binding proteins or molecules, and histone-affinity hemoadsorption in pre-clinical studies. Finally, challenges and future perspectives for improving the clinical translation of histone-targeting therapeutic strategies are also discussed to promote better management of patients with histone-related diseases.
Collapse
Affiliation(s)
- Tinghang Yang
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Jing Peng
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Zhuyun Zhang
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Yu Chen
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu, China
| | - Zhihui Liu
- Department of Rheumatology and Immunology, West China Hospital of Sichuan University, Chengdu, China
| | - Luojia Jiang
- Jiujiang City Key Laboratory of Cell Therapy, Department of Nephrology, Jiujiang No. 1 People’s Hospital, Jiujiang, China
| | - Lunqiang Jin
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Mei Han
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Baihai Su
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
- Med+ Biomaterial Institute of West China Hospital/West China School of Medicine, Sichuan University, Chengdu, China
- Med-X Center for Materials, Sichuan University, Chengdu, China
| | - Yupei Li
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
34
|
Feng G, Wu Z, Yang L, Wang K, Wang H. β-hydroxybutyrate and ischemic stroke: roles and mechanisms. Mol Brain 2024; 17:48. [PMID: 39075604 PMCID: PMC11287974 DOI: 10.1186/s13041-024-01119-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/14/2024] [Indexed: 07/31/2024] Open
Abstract
Stroke is a significant global burden, causing extensive morbidity and mortality. In metabolic states where glucose is limited, ketone bodies, predominantly β-hydroxybutyrate (BHB), act as alternative fuel sources. Elevated levels of BHB have been found in the ischemic hemispheres of animal models of stroke, supporting its role in the pathophysiology of cerebral ischemia. Clinically, higher serum and urinary BHB concentrations have been associated with adverse outcomes in ischemic stroke, highlighting its potential utility as a prognostic biomarker. In both animal and cellular models, exogenous BHB administration has exhibited neuroprotective effects, reduction of infarct size, and improvement of neurological outcomes. In this review, we focus on the role of BHB before and after ischemic stroke, with an emphasis on the therapeutic potential and mechanisms of ketone administration after ischemic stroke.
Collapse
Affiliation(s)
- Ge Feng
- Graduate School of Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Neurology, Hebei General Hospital, No. 348 21 Heping West Road, Shijiazhuang, 050051, Hebei, China
| | - Zongkai Wu
- Department of Neurology, Hebei General Hospital, No. 348 21 Heping West Road, Shijiazhuang, 050051, Hebei, China
| | - Leyi Yang
- Graduate School of Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Neurology, Hebei General Hospital, No. 348 21 Heping West Road, Shijiazhuang, 050051, Hebei, China
| | - Kaimeng Wang
- Graduate School of Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Neurology, Hebei General Hospital, No. 348 21 Heping West Road, Shijiazhuang, 050051, Hebei, China
| | - Hebo Wang
- Department of Neurology, Hebei General Hospital, No. 348 21 Heping West Road, Shijiazhuang, 050051, Hebei, China.
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Shijiazhuang, Hebei, China.
| |
Collapse
|
35
|
Yu X, Xu R, Huang X, Chen H, Zhang Z, Wong I, Chen Z, Deng F. Size-Dependent Effect of Titania Nanotubes on Endoplasmic Reticulum Stress to Re-establish Diabetic Macrophages Homeostasis. ACS Biomater Sci Eng 2024; 10:4323-4335. [PMID: 38860558 DOI: 10.1021/acsbiomaterials.4c00549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
In patients with diabetes, endoplasmic reticulum stress (ERS) is a crucial disrupting factor of macrophage homeostasis surrounding implants, which remains an obstacle to oral implantation success. Notably, the ERS might be modulated by the implant surface morphology. Titania nanotubes (TNTs) may enhance diabetic osseointegration. However, a consensus has not been achieved regarding the tube-size-dependent effect and the underlying mechanism of TNTs on diabetic macrophage ERS. We manufactured TNTs with small (30 nm) and large diameters (100 nm). Next, we assessed how the different titanium surfaces affected diabetic macrophages and regulated ERS and Ca2+ homeostasis. TNTs alleviated the inflammatory response, oxidative stress, and ERS in diabetic macrophages. Furthermore, TNT30 was superior to TNT100. Inhibiting ERS abolished the positive effect of TNT30. Mechanistically, topography-induced extracellular Ca2+ influx might mitigate excessive ERS in macrophages by alleviating ER Ca2+ depletion and IP3R activation. Furthermore, TNT30 attenuated the peri-implant inflammatory response and promoted osseointegration in diabetic rats. TNTs with small nanodiameters attenuated ERS and re-established diabetic macrophage hemostasis by inhibiting IP3R-induced ER Ca2+ depletion.
Collapse
Affiliation(s)
- Xiaoran Yu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, PR China
| | - Ruogu Xu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, PR China
| | - Xiaoqiong Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, PR China
| | - Hongcheng Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, PR China
| | - Zhengchuan Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, PR China
| | - Iohong Wong
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, PR China
| | - Zetao Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, PR China
| | - Feilong Deng
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, PR China
| |
Collapse
|
36
|
Liang L, Deng Y, Ao Z, Liao C, Tian J, Li C, Yu X. Recent progress in biomimetic nanomedicines based on versatile targeting strategy for atherosclerosis therapy. J Drug Target 2024; 32:606-623. [PMID: 38656224 DOI: 10.1080/1061186x.2024.2347353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/18/2024] [Indexed: 04/26/2024]
Abstract
Atherosclerosis (AS) is considered to be one of the major causes of cardiovascular disease. Its pathological microenvironment is characterised by increased production of reactive oxygen species, lipid oxides, and excessive inflammatory factors, which accumulate at the monolayer endothelial cells in the vascular wall to form AS plaques. Therefore, intervention in the pathological microenvironment would be beneficial in delaying AS. Researchers have designed biomimetic nanomedicines with excellent biocompatibility and the ability to avoid being cleared by the immune system through different therapeutic strategies to achieve better therapeutic effects for the characteristics of AS. Biomimetic nanomedicines can further enhance delivery efficiency and improve treatment efficacy due to their good biocompatibility and ability to evade clearance by the immune system. Biomimetic nanomedicines based on therapeutic strategies such as neutralising inflammatory factors, ROS scavengers, lipid clearance and integration of diagnosis and treatment are versatile approaches for effective treatment of AS. The review firstly summarises the targeting therapeutic strategy of biomimetic nanomedicine for AS in recent 5 years. Biomimetic nanomedicines using cell membranes, proteins, and extracellular vesicles as carriers have been developed for AS.
Collapse
Affiliation(s)
- Lijuan Liang
- Department of Pharmacy, Hejiang County People's Hospital, Luzhou, Sichuan, China
| | - Yiping Deng
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Zuojin Ao
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Changli Liao
- Science and Technology Department, Southwest Medical University, Luzhou, Sichuan, China
| | - Ji Tian
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xin Yu
- Chinese Pharmacy Laboratory, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
37
|
Benítez-King G, Argueta J, Miranda-Riestra A, Muñoz-Delgado J, Estrada-Reyes R. Interaction of the Melatonin/Ca 2+-CaM Complex with Calmodulin Kinase II: Physiological Importance. Mol Pharmacol 2024; 106:3-12. [PMID: 38811168 DOI: 10.1124/molpharm.123.000812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 04/04/2024] [Accepted: 04/15/2024] [Indexed: 05/31/2024] Open
Abstract
Melatonin N-acetyl-5-methoxytriptamine is an ancient molecule which synchronizes the internal biologic activity with the environmental photoperiod. It is synthesized by the pineal gland during the night and released to the general circulation, where it reaches nanomolar concentrations. The indolamine acts through melatonin receptors and binds to different proteins such as calmodulin: a phylogenetically conserved protein which is the main transductor of the calcium signaling. In this review, we will describe evidence supporting that melatonin binds to calmodulin in presence of calcium, and we discuss the effects of this indolamine on the activity of calmodulin kinase II as an inhibitor and as stimulator of calmodulin-dependent protein kinase II activity. We also provide a literature review supporting the relevance of melatonin binding to calmodulin in the regulation of circadian rhythms in unicellular organisms, as well as in neuronal development in mammals as an ancient, conserved mechanism. Finally, we highlight the importance of antioxidant effects of melatonin on calmodulin preservation. SIGNIFICANCE STATEMENT: This review compiled evidence supporting that melatonin binds to calmodulin. We discuss the dual effect of melatonin on the activity of calmodulin kinase II, the possible mechanisms involved, and the relevance on regulation of circadian rhythms and neurodevelopment. Finally, we describe evidence supporting that the binding of melatonin to calmodulin hydrophobic pockets may prevent the oxidation of methionine species with a shielding effect that preserves the functionality of calmodulin.
Collapse
Affiliation(s)
- Gloria Benítez-King
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, México City, México (G.B.-K., J.A., A.M.-R.); Laboratorio de Cronoecología y Etología Humana, Departamento de Etología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, México City, México (J.M.-D.); and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, México City, México (R.E-R.)
| | - Jesús Argueta
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, México City, México (G.B.-K., J.A., A.M.-R.); Laboratorio de Cronoecología y Etología Humana, Departamento de Etología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, México City, México (J.M.-D.); and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, México City, México (R.E-R.)
| | - Armida Miranda-Riestra
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, México City, México (G.B.-K., J.A., A.M.-R.); Laboratorio de Cronoecología y Etología Humana, Departamento de Etología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, México City, México (J.M.-D.); and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, México City, México (R.E-R.)
| | - Jairo Muñoz-Delgado
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, México City, México (G.B.-K., J.A., A.M.-R.); Laboratorio de Cronoecología y Etología Humana, Departamento de Etología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, México City, México (J.M.-D.); and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, México City, México (R.E-R.)
| | - Rosa Estrada-Reyes
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, México City, México (G.B.-K., J.A., A.M.-R.); Laboratorio de Cronoecología y Etología Humana, Departamento de Etología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, México City, México (J.M.-D.); and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, México City, México (R.E-R.)
| |
Collapse
|
38
|
Zhang T, Luo X, Jing L, Mo C, Guo H, Yang S, Wang Y, Zhao K, Lai Y, Liu Y. Toosendanin inhibits T-cell proliferation through the P38 MAPK signalling pathway. Eur J Pharmacol 2024; 973:176562. [PMID: 38588767 DOI: 10.1016/j.ejphar.2024.176562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 03/20/2024] [Accepted: 04/03/2024] [Indexed: 04/10/2024]
Abstract
In recent years, immunosuppressants have shown significant success in the treatment of autoimmune diseases. Therefore, there is an urgent need to develop additional immunosuppressants that offer more options for patients. Toosendanin has been shown to have immunosuppressive activity in vitro as well as effects on autoimmune hepatitis (AIH) in vivo. Toosendanin did not induce apoptosis in activated T-cells and affect the survival rate of naive T-cells. Toosendanin did not affect the expression of CD25 or secretion of IL-2 by activated T-cells, and not affect the expression of IL-4 and INF-γ. Toosendanin did not affect the phosphorylation of STAT5, ERK, AKT, P70S6K. However, toosendanin inhibited proliferation of anti-CD3/anti-CD28 mAbs-activated T-cells with IC50 of (10 ± 2.02) nM. Toosendanin arrested the cell cycle in the G0/G1 phase, significantly inhibited IL-6 and IL-17A secretion, promoted IL-10 expression, and inhibited the P38 MAPK pathway. Finally, toosendanin significantly alleviated ConA-induced AIH in mice. In Summary, toosendanin exhibited immunosuppressive activity in vivo and in vitro. Toosendanin inhibits the proliferation of activated T-cells through the P38 MAPK signalling pathway, significantly suppresses the expression of inflammatory factors, enhances the expression of anti-inflammatory factors, and effectively alleviates ConA-induced AIH in mice, suggesting that toosendanin may be a lead compound for the development of novel immunomodulatory agents with improved efficacy and reduced toxicity.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China; School of Pharmacy, Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China
| | - Xingyan Luo
- Research Center, Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China; Development and Regeneration Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China
| | - Lin Jing
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, 530004, Guangxi, People's Republic of China
| | - Chunfen Mo
- Research Center, Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China
| | - Huijie Guo
- Research Center, Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China
| | - Shuxia Yang
- Research Center, Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China
| | - Yantang Wang
- Research Center, Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China
| | - Ketian Zhao
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China; Research Center, Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China
| | - Yi Lai
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China.
| | - Yang Liu
- Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China; School of Pharmacy, Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China; Development and Regeneration Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China.
| |
Collapse
|
39
|
Xu S, Lu F, Gao J, Yuan Y. Inflammation-mediated metabolic regulation in adipose tissue. Obes Rev 2024; 25:e13724. [PMID: 38408757 DOI: 10.1111/obr.13724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 11/04/2023] [Accepted: 01/17/2024] [Indexed: 02/28/2024]
Abstract
Chronic inflammation of adipose tissue is a prominent characteristic of many metabolic diseases. Lipid metabolism in adipose tissue is consistently dysregulated during inflammation, which is characterized by substantial infiltration by proinflammatory cells and high cytokine concentrations. Adipose tissue inflammation is caused by a variety of endogenous factors, such as mitochondrial dysfunction, reactive oxygen species (ROS) production, endoplasmic reticulum (ER) stress, cellular senescence, ceramides biosynthesis and mediators of lipopolysaccharides (LPS) signaling. Additionally, the gut microbiota also plays a crucial role in regulating adipose tissue inflammation. Essentially, adipose tissue inflammation arises from an imbalance in adipocyte metabolism and the regulation of immune cells. Specific inflammatory signals, including nuclear factor-κB (NF-κB) signaling, inflammasome signaling and inflammation-mediated autophagy, have been shown to be involved in the metabolic regulation. The pathogenesis of metabolic diseases characterized by chronic inflammation (obesity, insulin resistance, atherosclerosis and nonalcoholic fatty liver disease [NAFLD]) and recent research regarding potential therapeutic targets for these conditions are also discussed in this review.
Collapse
Affiliation(s)
- Shujie Xu
- Department of Plastic and Reconstructive Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Feng Lu
- Department of Plastic and Reconstructive Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianhua Gao
- Department of Plastic and Reconstructive Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yi Yuan
- Department of Plastic and Reconstructive Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
40
|
Ren T, He J, Zhang T, Niu A, Yuan Y, Zuo Y, Miao Y, Zhang H, Zang L, Qiao C, Cao X, Yang X, Zheng Z, Xu Y, Wu D, Zheng H. Exercise activates interferon response of the liver via Gpld1 to enhance antiviral innate immunity. SCIENCE ADVANCES 2024; 10:eadk5011. [PMID: 38809975 PMCID: PMC11804790 DOI: 10.1126/sciadv.adk5011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 04/24/2024] [Indexed: 05/31/2024]
Abstract
Healthy behavioral patterns could modulate organ functions to enhance the body's immunity. However, how exercise regulates antiviral innate immunity remains elusive. Here, we found that exercise promotes type I interferon (IFN-I) production in the liver and enhances IFN-I immune activity of the body. Despite the possibility that many exercise-induced factors could affect IFN-I production, we identified Gpld1 as a crucial molecule, and the liver as the major organ to promote IFN-I production after exercise. Exercise largely loses the efficiency to induce IFN-I in Gpld1-/- mice. Further studies demonstrated that exercise-produced 3-hydroxybutanoic acid (3-HB) critically induces Gpld1 expression in the liver. Gpld1 blocks the PP2A-IRF3 interaction, thus enhancing IRF3 activation and IFN-I production, and eventually improving the body's antiviral ability. This study reveals that exercise improves antiviral innate immunity by linking the liver metabolism to systemic IFN-I activity and uncovers an unknown function of liver cells in innate immunity.
Collapse
Affiliation(s)
- Tengfei Ren
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, 215123 Suzhou, Jiangsu, China
- Department/Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jiuyi He
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, 215123 Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, China
| | - Tingting Zhang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, 215123 Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, China
| | - Anxing Niu
- Department of Infectious Diseases, The Affiliated Infectious Diseases Hospital, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, China
| | - Yukang Yuan
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, 215123 Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yibo Zuo
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, 215123 Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, China
| | - Ying Miao
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, 215123 Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, China
| | - Hongguang Zhang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, 215123 Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, China
| | - Lichao Zang
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Caixia Qiao
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, 215123 Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xinhua Cao
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, 215123 Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xinyu Yang
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Zhijin Zheng
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, 215123 Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yang Xu
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Depei Wu
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Hui Zheng
- Department/Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
41
|
Mo S, Wu W, Luo K, Huang C, Wang Y, Qin H, Cai H. Identification and analysis of chemokine-related and NETosis-related genes in acute pancreatitis to develop a predictive model. Front Genet 2024; 15:1389936. [PMID: 38784040 PMCID: PMC11112067 DOI: 10.3389/fgene.2024.1389936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/17/2024] [Indexed: 05/25/2024] Open
Abstract
Background: Chemokines and NETosis are significant contributors to the inflammatory response, yet there still needs to be a more comprehensive understanding regarding the specific molecular characteristics and interactions of NETosis and chemokines in the context of acute pancreatitis (AP) and severe AP (SAP). Methods: To address this gap, the mRNA expression profile dataset GSE194331 was utilized for analysis, comprising 87 AP samples (77 non-SAP and 10 SAP) and 32 healthy control samples. Enrichment analyses were conducted for differentially expressed chemokine-related genes (DECRGs) and NETosis-related genes (DENRGs). Three machine-learning algorithms were used for the identification of signature genes, which were subsequently utilized in the development and validation of nomogram diagnostic models for the prediction of AP and SAP. Furthermore, single-gene Gene Set Enrichment Analysis (GSEA) and Gene Set Variation Analysis (GSVA) were performed. Lastly, an interaction network for the identified signature genes was constructed. Results: We identified 12 DECRGs and 7 DENRGs, and enrichment analyses indicated they were primarily enriched in cytokine-cytokine receptor interaction, chemokine signaling pathway, TNF signaling pathway, and T cell receptor signaling pathway. Moreover, these machine learning algorithms finally recognized three signature genes (S100A8, AIF1, and IL18). Utilizing the identified signature genes, we developed nomogram models with high predictive accuracy for AP and differentiation of SAP from non-SAP, as demonstrated by area under the curve (AUC) values of 0.968 (95% CI 0.937-0.990) and 0.862 (95% CI 0.742-0.955), respectively, in receiver operating characteristic (ROC) curve analysis. Subsequent single-gene GESA and GSVA indicated a significant positive correlation between these signature genes and the proteasome complex. At the same time, a negative association was observed with the Th1 and Th2 cell differentiation signaling pathways. Conclusion: We have identified three genes (S100A8, AIF1, and IL18) related to chemokines and NETosis, and have developed accurate diagnostic models that might provide a novel method for diagnosing AP and differentiating between severe and non-severe cases.
Collapse
Affiliation(s)
- Shuangyang Mo
- Gastroenterology Department, Liuzhou People’s Hospital Affiliated to Guangxi Medical University, Liuzhou, China
| | - Wenhong Wu
- Gastroenterology Department, Liuzhou People’s Hospital Affiliated to Guangxi Medical University, Liuzhou, China
| | - Kai Luo
- Department of Critical Care Medicine, Liuzhou People’s Hospital Affiliated to Guangxi Medical University, Liuzhou, China
| | - Cheng Huang
- Oncology Department, Liuzhou People’s Hospital Affiliated to Guangxi Medical University, Liuzhou, China
| | - Yingwei Wang
- Gastroenterology Department, Liuzhou People’s Hospital Affiliated to Guangxi Medical University, Liuzhou, China
| | - Heping Qin
- Gastroenterology Department, Liuzhou People’s Hospital Affiliated to Guangxi Medical University, Liuzhou, China
| | - Huaiyang Cai
- Gastroenterology Department, Liuzhou People’s Hospital Affiliated to Guangxi Medical University, Liuzhou, China
| |
Collapse
|
42
|
Voronina MV, Frolova AS, Kolesova EP, Kuldyushev NA, Parodi A, Zamyatnin AA. The Intricate Balance between Life and Death: ROS, Cathepsins, and Their Interplay in Cell Death and Autophagy. Int J Mol Sci 2024; 25:4087. [PMID: 38612897 PMCID: PMC11012956 DOI: 10.3390/ijms25074087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Cellular survival hinges on a delicate balance between accumulating damages and repair mechanisms. In this intricate equilibrium, oxidants, currently considered physiological molecules, can compromise vital cellular components, ultimately triggering cell death. On the other hand, cells possess countermeasures, such as autophagy, which degrades and recycles damaged molecules and organelles, restoring homeostasis. Lysosomes and their enzymatic arsenal, including cathepsins, play critical roles in this balance, influencing the cell's fate toward either apoptosis and other mechanisms of regulated cell death or autophagy. However, the interplay between reactive oxygen species (ROS) and cathepsins in these life-or-death pathways transcends a simple cause-and-effect relationship. These elements directly and indirectly influence each other's activities, creating a complex web of interactions. This review delves into the inner workings of regulated cell death and autophagy, highlighting the pivotal role of ROS and cathepsins in these pathways and their intricate interplay.
Collapse
Affiliation(s)
- Maya V. Voronina
- Research Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (M.V.V.); (A.S.F.); (E.P.K.); (N.A.K.); (A.P.)
| | - Anastasia S. Frolova
- Research Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (M.V.V.); (A.S.F.); (E.P.K.); (N.A.K.); (A.P.)
- Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Ekaterina P. Kolesova
- Research Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (M.V.V.); (A.S.F.); (E.P.K.); (N.A.K.); (A.P.)
| | - Nikita A. Kuldyushev
- Research Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (M.V.V.); (A.S.F.); (E.P.K.); (N.A.K.); (A.P.)
| | - Alessandro Parodi
- Research Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (M.V.V.); (A.S.F.); (E.P.K.); (N.A.K.); (A.P.)
| | - Andrey A. Zamyatnin
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- Department of Biological Chemistry, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| |
Collapse
|
43
|
Huang H, Lu W, Zhang X, Pan J, Cao F, Wen L. Fibroblast subtypes in pancreatic cancer and pancreatitis: from mechanisms to therapeutic strategies. Cell Oncol (Dordr) 2024; 47:383-396. [PMID: 37721678 DOI: 10.1007/s13402-023-00874-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 09/19/2023] Open
Abstract
Excessive fibrosis is a predominant feature of pancreatic stroma and plays a crucial role in the development and progression of pancreatic ductal adenocarcinoma (PDAC) and chronic pancreatitis (CP). Emerging evidence showed diversity and heterogeneity of fibroblasts play crucial and somewhat contradictory roles, the interactions between fibroblasts and pancreatic cells or infiltrating immune cells are of great importance during PDAC and CP progression, with some promising therapeutic strategies being tested. Therefore, in this review, we describe the classification of fibroblasts and their functions in PDAC and pancreatitis, the mechanisms by which fibroblasts mediate the development and progression of PDAC and CP through direct or indirect interaction between fibroblast and pancreatic parenchymal cells, or by remodeling the pancreatic immune microenvironment mediates the development and progression of PDAC and CP. Finally, we summarized the current therapeutic strategies and agents that directly target subtypes of fibroblasts or interfere with their essential functions.
Collapse
Affiliation(s)
- Huizhen Huang
- Department of Gastroenterology, Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Nanjing Medical University, Shanghai, China
| | - Wanyi Lu
- Center for Biomarker Discovery and Validation, National Infrastructures for Translational Medicine (PUMCH), Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Xiuli Zhang
- Center for Biomarker Discovery and Validation, National Infrastructures for Translational Medicine (PUMCH), Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Jiachun Pan
- Center for Biomarker Discovery and Validation, National Infrastructures for Translational Medicine (PUMCH), Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Feng Cao
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Li Wen
- Center for Biomarker Discovery and Validation, National Infrastructures for Translational Medicine (PUMCH), Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China.
- State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China.
| |
Collapse
|
44
|
Németh K, Sterczer Á, Kiss DS, Lányi RK, Hemző V, Vámos K, Bartha T, Buzás A, Lányi K. Determination of Bile Acids in Canine Biological Samples: Diagnostic Significance. Metabolites 2024; 14:178. [PMID: 38668306 PMCID: PMC11052161 DOI: 10.3390/metabo14040178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
The comprehensive examination of bile acids is of paramount importance across various fields of health sciences, influencing physiology, microbiology, internal medicine, and pharmacology. While enzymatic reaction-based photometric methods remain fundamental for total BA measurements, there is a burgeoning demand for more sophisticated techniques such as liquid chromatography-tandem mass spectrometry (LC-MS/MS) for comprehensive BA profiling. This evolution reflects a need for nuanced diagnostic assessments in clinical practice. In canines, a BA assessment involves considering factors, such as food composition, transit times, and breed-specific variations. Multiple matrices, including blood, feces, urine, liver tissue, and gallbladder bile, offer insights into BA profiles, yet interpretations remain complex, particularly in fecal analysis due to sampling challenges and breed-specific differences. Despite ongoing efforts, a consensus regarding optimal matrices and diagnostic thresholds remains elusive, highlighting the need for further research. Emphasizing the scarcity of systematic animal studies and underscoring the importance of ap-propriate sampling methodologies, our review advocates for targeted investigations into BA alterations in canine pathology, promising insights into pathomechanisms, early disease detection, and therapeutic avenues.
Collapse
Affiliation(s)
- Krisztián Németh
- Department of Physiology and Biochemistry, University of Veterinary Medicine, István u. 2, H-1078 Budapest, Hungary; (K.N.); (D.S.K.); (V.H.); (T.B.)
| | - Ágnes Sterczer
- Department of Internal Medicine, University of Veterinary Medicine, István u. 2, H-1078 Budapest, Hungary;
| | - Dávid Sándor Kiss
- Department of Physiology and Biochemistry, University of Veterinary Medicine, István u. 2, H-1078 Budapest, Hungary; (K.N.); (D.S.K.); (V.H.); (T.B.)
| | - Réka Katalin Lányi
- Faculty of Pharmacy, University of Szeged, Zrínyi u. 9, H-6720 Szeged, Hungary;
| | - Vivien Hemző
- Department of Physiology and Biochemistry, University of Veterinary Medicine, István u. 2, H-1078 Budapest, Hungary; (K.N.); (D.S.K.); (V.H.); (T.B.)
| | - Kriszta Vámos
- Department of Internal Medicine, University of Veterinary Medicine, István u. 2, H-1078 Budapest, Hungary;
| | - Tibor Bartha
- Department of Physiology and Biochemistry, University of Veterinary Medicine, István u. 2, H-1078 Budapest, Hungary; (K.N.); (D.S.K.); (V.H.); (T.B.)
| | - Anna Buzás
- Institute of Food Chain Science, University of Veterinary Medicine, István u. 2, H-1078 Budapest, Hungary; (A.B.); (K.L.)
| | - Katalin Lányi
- Institute of Food Chain Science, University of Veterinary Medicine, István u. 2, H-1078 Budapest, Hungary; (A.B.); (K.L.)
| |
Collapse
|
45
|
Sirichoat A, Dornlakorn O, Saenno R, Aranarochana A, Sritawan N, Pannangrong W, Wigmore P, Welbat JU. Caffeic acid protects against l-methionine induced reduction in neurogenesis and cognitive impairment in a rat model. Heliyon 2024; 10:e26919. [PMID: 38455532 PMCID: PMC10918208 DOI: 10.1016/j.heliyon.2024.e26919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 02/21/2024] [Accepted: 02/21/2024] [Indexed: 03/09/2024] Open
Abstract
l-methionine (L-met) is a substantial non-polar amino acid for normal development. L-met is converted to homocysteine that leads to hyperhomocysteinemia and subsequent excessive homocysteine in serum resulting in stimulating oxidative stress and vascular dementia. Several studies have found that hyperhomocysteine causes neuronal cell damage, which leads to memory impairment. Caffeic acid is a substrate in phenolic compound discovered in plant biosynthesis. Caffeic acid contains biological antioxidant and neuroprotective properties. The neuroprotective reaction of caffeic acid can protect against the brain disruption from hydrogen peroxide produced by oxidative stress. It also enhances GSH and superoxide dismutase activities, which protect against neuron cell loss caused by oxidative stress in the hippocampus. Hence, we investigated the protective role of caffeic acid in hippocampal neurogenesis and cognitive impairment induced by L-met in rats. Six groups of Sprague Dawley rats were assigned including control, L-met (1.7 g/kg/day), caffeic acid (20, 40 mg/kg), and L-met + caffeic acid (20, 40 mg/kg) groups. Spatial and recognition memories were subsequently examined using novel object location (NOL) and novel object recognition (NOR) tests. Moreover, the immunofluorescence technique was performed to detect Ki-67/RECA-1, bromodeoxyuridine (BrdU)/NeuN and p21 markers to represent hippocampal neurogenesis changes. The results revealed decreases in vasculature related cell proliferation and neuronal cell survival. By contrast, cell cycle arrest was increased in the L-met group. These results showed the association of the spatial and recognition memory impairments. However, the deterioration can be restored by co-administration with caffeic acid.
Collapse
Affiliation(s)
- Apiwat Sirichoat
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Neurogenesis Research Group, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Oabnithi Dornlakorn
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Neurogenesis Research Group, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Rasa Saenno
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Neurogenesis Research Group, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Anusara Aranarochana
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Neurogenesis Research Group, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Nataya Sritawan
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Neurogenesis Research Group, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Wanassanun Pannangrong
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Peter Wigmore
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, United Kingdom
| | - Jariya Umka Welbat
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Neurogenesis Research Group, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| |
Collapse
|
46
|
Shcholok T, Eftekharpour E. Insights into the Multifaceted Roles of Thioredoxin-1 System: Exploring Knockout Murine Models. BIOLOGY 2024; 13:180. [PMID: 38534450 DOI: 10.3390/biology13030180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 03/28/2024]
Abstract
Redox balance is increasingly identified as a major player in cellular signaling. A fundamentally simple reaction of oxidation and reduction of cysteine residues in cellular proteins is the central concept in this complex regulatory mode of protein function. Oxidation of key cysteine residues occurs at the physiological levels of reactive oxygen species (ROS), but they are reduced by a supply of thiol antioxidant molecules including glutathione, glutaredoxin, and thioredoxin. While these molecules show complex compensatory roles in experimental conditions, transgenic animal models provide a comprehensive picture to pinpoint the role of each antioxidant. In this review, we have specifically focused on the available literature on thioredoxin-1 system transgenic models that include thioredoxin and thioredoxin reductase proteins. As the identification of thioredoxin protein targets is technically challenging, the true contribution of this system in maintaining cellular balance remains unidentified, including the role of this system in the brain.
Collapse
Affiliation(s)
- Tetiana Shcholok
- Department of Physiology and Pathophysiology, University of Manitoba, 631-BMSB, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada
| | - Eftekhar Eftekharpour
- Department of Physiology and Pathophysiology, University of Manitoba, 631-BMSB, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
47
|
Vertiprakhov VG, Trukhachev VI, Sergeenkova NA. Reflex phase of regulation of pancreatic secretion in poultry after feed intake is associated with gustatory sensations, and neurohumoral-with nutritional value. Front Physiol 2024; 15:1341132. [PMID: 38532844 PMCID: PMC10963493 DOI: 10.3389/fphys.2024.1341132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 02/21/2024] [Indexed: 03/28/2024] Open
|
48
|
Ji R, Chang L, An C, Zhang J. Proton-sensing ion channels, GPCRs and calcium signaling regulated by them: implications for cancer. Front Cell Dev Biol 2024; 12:1326231. [PMID: 38505262 PMCID: PMC10949864 DOI: 10.3389/fcell.2024.1326231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/14/2024] [Indexed: 03/21/2024] Open
Abstract
Extracellular acidification of tumors is common. Through proton-sensing ion channels or proton-sensing G protein-coupled receptors (GPCRs), tumor cells sense extracellular acidification to stimulate a variety of intracellular signaling pathways including the calcium signaling, which consequently exerts global impacts on tumor cells. Proton-sensing ion channels, and proton-sensing GPCRs have natural advantages as drug targets of anticancer therapy. However, they and the calcium signaling regulated by them attracted limited attention as potential targets of anticancer drugs. In the present review, we discuss the progress in studies on proton-sensing ion channels, and proton-sensing GPCRs, especially emphasizing the effects of calcium signaling activated by them on the characteristics of tumors, including proliferation, migration, invasion, metastasis, drug resistance, angiogenesis. In addition, we review the drugs targeting proton-sensing channels or GPCRs that are currently in clinical trials, as well as the relevant potential drugs for cancer treatments, and discuss their future prospects. The present review aims to elucidate the important role of proton-sensing ion channels, GPCRs and calcium signaling regulated by them in cancer initiation and development. This review will promote the development of drugs targeting proton-sensing channels or GPCRs for cancer treatments, effectively taking their unique advantage as anti-cancer drug targets.
Collapse
Affiliation(s)
- Renhui Ji
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
- Department of Pathophysiology, Basic Medicine College of Inner Mongolia Medical University, Hohhot, China
| | - Li Chang
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
- Department of Pathophysiology, Basic Medicine College of Inner Mongolia Medical University, Hohhot, China
| | - Caiyan An
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
| | - Junjing Zhang
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
| |
Collapse
|
49
|
Yu L, Liu Z, Xu W, Jin K, Liu J, Zhu X, Zhang Y, Wu Y. Towards overcoming obstacles of type II photodynamic therapy: Endogenous production of light, photosensitizer, and oxygen. Acta Pharm Sin B 2024; 14:1111-1131. [PMID: 38486983 PMCID: PMC10935104 DOI: 10.1016/j.apsb.2023.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/20/2023] [Accepted: 10/28/2023] [Indexed: 03/17/2024] Open
Abstract
Conventional photodynamic therapy (PDT) approaches face challenges including limited light penetration, low uptake of photosensitizers by tumors, and lack of oxygen in tumor microenvironments. One promising solution is to internally generate light, photosensitizers, and oxygen. This can be accomplished through endogenous production, such as using bioluminescence as an endogenous light source, synthesizing genetically encodable photosensitizers in situ, and modifying cells genetically to express catalase enzymes. Furthermore, these strategies have been reinforced by the recent rapid advancements in synthetic biology. In this review, we summarize and discuss the approaches to overcome PDT obstacles by means of endogenous production of excitation light, photosensitizers, and oxygen. We envision that as synthetic biology advances, genetically engineered cells could act as precise and targeted "living factories" to produce PDT components, leading to enhanced performance of PDT.
Collapse
Affiliation(s)
- Lin Yu
- Department of Chemical and Environmental Engineering, Shanghai University, Shanghai 200433, China
- School of Medicine, Shanghai University, Shanghai 200433, China
| | - Zhen Liu
- Department of Chemical and Environmental Engineering, Shanghai University, Shanghai 200433, China
| | - Wei Xu
- Department of Chemical and Environmental Engineering, Shanghai University, Shanghai 200433, China
| | - Kai Jin
- Department of Chemical and Environmental Engineering, Shanghai University, Shanghai 200433, China
| | - Jinliang Liu
- Department of Chemical and Environmental Engineering, Shanghai University, Shanghai 200433, China
| | - Xiaohui Zhu
- Department of Chemical and Environmental Engineering, Shanghai University, Shanghai 200433, China
| | - Yong Zhang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Yihan Wu
- Department of Chemical and Environmental Engineering, Shanghai University, Shanghai 200433, China
| |
Collapse
|
50
|
Wang N, Yang A, Tian X, Liao J, Yang Z, Pan Y, Guo Y, He S. Label-free analysis of the β-hydroxybutyricacid drug on mitochondrial redox states repairment in type 2 diabetic mice by resonance raman scattering. Biomed Pharmacother 2024; 172:116320. [PMID: 38387134 DOI: 10.1016/j.biopha.2024.116320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND Mitochondrial redox imbalance underlies the pathophysiology of type2 diabetes mellitus (T2DM), and is closely related to tissue damage and dysfunction. Studies have shown the beneficial effects of dietary strategies that elevate β-hydroxybutyrate (BHB) levels in alleviating T2DM. Nevertheless, the role of BHB has not been clearly elucidated. METHODS We performed a spectral study to visualize the preventive effects of BHB on blood and multiorgan mitochondrial redox imbalance in T2DM mice via using label-free resonance Raman spectroscopy (RRS), and further explored the impact of BHB therapy on the pathology of T2DM mice by histological and biochemical analyses. FINDINGS Our data revealed that RRS-based mitochondrial redox states assay enabled clear and reliable identification of the improvement of mitochondrial redox imbalance by BHB, evidenced by the reduction of Raman peak intensity at 750 cm-1, 1128 cm-1 and 1585 cm-1 in blood, tissue as well as purified mitochondria of db/db mice and the increase of tissue mitochondrial succinic dehydrogenase (SDH) staining after BHB treatment. Exogenous supplementation of BHB was also found to attenuate T2DM pathology related to mitochondrial redox states, involving organ injury, blood glucose control, insulin resistance and systemic inflammation. INTERPRETATION Our findings provide strong evidence for BHB as a potential therapeutic strategy targeting mitochondria for T2DM.
Collapse
Affiliation(s)
- Na Wang
- Taizhou Hospital, Zhejiang University School of Medicine, Linhai, China; Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Anqi Yang
- Centre for Optical and Electromagnetic Research, National Engineering Research Center for Optical Instruments, Zhejiang Provincial Key Laboratory for Sensing Technologies, Zhejiang University, Hangzhou 310058, China
| | - Xiong Tian
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Jiaqi Liao
- Centre for Optical and Electromagnetic Research, National Engineering Research Center for Optical Instruments, Zhejiang Provincial Key Laboratory for Sensing Technologies, Zhejiang University, Hangzhou 310058, China
| | - Zhenyu Yang
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Yixiao Pan
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Yiqing Guo
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Sailing He
- Taizhou Hospital, Zhejiang University School of Medicine, Linhai, China; Centre for Optical and Electromagnetic Research, National Engineering Research Center for Optical Instruments, Zhejiang Provincial Key Laboratory for Sensing Technologies, Zhejiang University, Hangzhou 310058, China; School of Electrical Engineering, Royal Institute of Technology, Stockholm S-100 44, Sweden.
| |
Collapse
|