1
|
Liao Y, Niu L, Ling J, Cui Y, Huang Z, Xu J, Jiang Y, Yu P, Liu X. Turning sour into sweet: Lactylation modification as a promising target in cardiovascular health. Metabolism 2025; 168:156234. [PMID: 40113080 DOI: 10.1016/j.metabol.2025.156234] [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: 07/08/2024] [Revised: 02/26/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
Lactylation, a recently identified posttranslational modification (PTM), has emerged as a critical regulatory mechanism in cardiovascular diseases (CVDs). This PTM involves the addition of lactyl groups to lysine residues on histones and nonhistone proteins, influencing gene expression and cellular metabolism. The discovery of lactylation has revealed new directions for understanding metabolic and immune processes, particularly in the context of CVDs. This review describes the intricate roles of specific lactylated proteins and enzymes, such as H3K18, HMGB1, MCT1/4, and LDH, in the regulation of cardiovascular pathology. This study also highlights the unique impact of lactylation on myocardial hypertrophy and distinguishes it from other PTMs, such as SUMOylation and acetylation, underscoring its potential as a therapeutic target. Emerging drugs targeting lactate transporters and critical enzymes involved in lactylation offer promising avenues for novel CVD therapies. This review calls for further research to elucidate the mechanisms linking lactylation to CVDs, emphasizing the need for comprehensive studies at the molecular, cellular, and organismal levels to pave the way for innovative preventive, diagnostic, and treatment strategies in cardiovascular medicine.
Collapse
Affiliation(s)
- Yajie Liao
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China; Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China; Queen Mary School, Nanchang University, Nanchang, Jiangxi, China
| | - Liyan Niu
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jitao Ling
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yuzhen Cui
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China; Queen Mary School, Nanchang University, Nanchang, Jiangxi, China
| | - Zixuan Huang
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China; Queen Mary School, Nanchang University, Nanchang, Jiangxi, China
| | - Jingdong Xu
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China; Queen Mary School, Nanchang University, Nanchang, Jiangxi, China
| | - Yuan Jiang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| | - Peng Yu
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China; Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| | - Xiao Liu
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China; Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Xie X, Wen C, Peng Q, Xiang D, Pan X, Lan X, Chen Z, Hua X, Wang G, Huang C. H3K9/18 lactylation regulates DNA damage due to nickel exposure in human bronchial epithelial cells. Toxicol Appl Pharmacol 2025; 499:117347. [PMID: 40250488 DOI: 10.1016/j.taap.2025.117347] [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: 12/30/2024] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
Nickel, a well-known heavy metal with lung carcinogenic properties, is recognized for its effects on cellular metabolism, oxidative stress, and gene expression. While these cellular alterations have prompted investigations into its potential impact on histone modifications, specific associations with histone lactylation remain under exploration. In the present study, we demonstrate that nickel exposure induces lactylation of histone H3 at lysines 9 (H3K9) and 18 (H3K18), accompanied by reactive oxygen species (ROS) accumulation and DNA damage in human bronchial epithelial Beas-2B cells. Inhibition of H3K9 and H3K18 lactylation, achieved by overexpressing mutated H3K9R and H3K18R, respectively, markedly abolishes ROS generation and DNA damage caused by nickel exposure. This highlights the novel biological effects of H3K9 and H3K18 lactylation in nickel-induced lung toxicity. Mechanistic investigations show that nickel-induced lactylation of H3K9 and H3K18 is mediated by elevated LDHA expression, leading to lactate accumulation, which results from the upregulation of LDHA mRNA transcription through HIF-1α/c-Jun axis and enhanced LDHA protein stability via TNF-α-mediated induction of HSP70, respectively. Our findings uncover a novel effect of nickel exposure on histone H3 lactylation and its biological impact on ROS accumulation and DNA damage through the HIF-1α/c-Jun/LDHA and TNF-α/HSP70/LDHA pathways. These results provide significant insights into the role of histone lactylation in heavy metal-induced lung toxicity.
Collapse
Affiliation(s)
- Xiaomin Xie
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou 325000, Zhejiang, China
| | - Chaowei Wen
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou 325000, Zhejiang, China
| | - Quekun Peng
- School of Bioscience and Technology, Chengdu Medical College, Chengdu 610500, Sichuan, China
| | - Daimin Xiang
- The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200433, China
| | - Xinyi Pan
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Xinmei Lan
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Zijian Chen
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Xiaohui Hua
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, China
| | - Guiying Wang
- Department of General Surgery, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Chuanshu Huang
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou 325000, Zhejiang, China.
| |
Collapse
|
3
|
Wang P, Lin K, Huang D, Jiang Z, Liao L, Wang X. The regulatory role of protein lactylation in various diseases: Special focus on the regulatory role of non-histone lactylation. Gene 2025; 963:149595. [PMID: 40441322 DOI: 10.1016/j.gene.2025.149595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 04/18/2025] [Accepted: 05/22/2025] [Indexed: 06/02/2025]
Abstract
Lactylation, an emerging form of post-translational modification derived from lactate, plays a pivotal role in numerous cellular processes such as tumor proliferation, metabolism, inflammation, and embryonic development. However, the precise molecular mechanisms by which lactylation controls these biological functions in both physiological and pathological contexts remain elusive. This review summarizes the latest reported regulatory mechanisms of protein lactylation in various diseases since 2024, introducing the latest research progress regarding the regulatory functions of protein lactylation in pathological processes, with particular attention to the regulatory mechanisms of non-histone lactylation modification in diseases. Finally, it outlines the potential of targeted lactylation therapy, proposes the main directions for future research, and emphasizes its scientific significance for future studies.
Collapse
Affiliation(s)
- Peipei Wang
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Kexin Lin
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Degao Huang
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Zihan Jiang
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Linchuan Liao
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China.
| | - Xia Wang
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China.
| |
Collapse
|
4
|
Chen C, Wang J, Zhu X, Zhang S, Yuan X, Hu J, Liu C, Liu L, Zhang Z, Li J. Lactylation as a metabolic epigenetic modification: Mechanistic insights and regulatory pathways from cells to organs and diseases. Metabolism 2025; 169:156289. [PMID: 40324589 DOI: 10.1016/j.metabol.2025.156289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/20/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
In recent years, lactylation, a novel post-translational modification, has demonstrated a unique role in bridging cellular metabolism and epigenetic regulation. This modification exerts a dual-edged effect in both cancer and non-cancer diseases by dynamically integrating the supply of metabolic substrates and the activity of modifying enzymes: on one hand, it promotes tissue homeostasis and repair through the activation of repair genes; on the other, it exacerbates pathological progression by driving malignant phenotypes. In the field of oncology, lactylation regulates key processes such as metabolic reprogramming, immune evasion, and therapeutic resistance, thereby shaping the heterogeneity of the tumor microenvironment. In non-cancerous diseases, including neurodegeneration and cardiovascular disorders, its aberrant activation can lead to mitochondrial dysfunction, fibrosis, and chronic inflammation. Existing studies have revealed a dynamic regulatory network formed by the cooperation of modifying and demodifying enzymes, and have identified mechanisms such as subcellular localization and RNA metabolism intervention that influence disease progression. Nevertheless, several challenges remain in the field. This article comprehensively summarizes the disease-specific regulatory mechanisms of lactylation, with the aim of providing a theoretical foundation for its targeted therapeutic application.
Collapse
Affiliation(s)
- Cong Chen
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Jie Wang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China.
| | - Xueying Zhu
- Department of Anatomy, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shan Zhang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xiandun Yuan
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing 100096, China
| | - Jun Hu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Chao Liu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Lanchun Liu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Zhenpeng Zhang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China.
| | - Jun Li
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China.
| |
Collapse
|
5
|
Ren H, Tang Y, Zhang D. The emerging role of protein L-lactylation in metabolic regulation and cell signalling. Nat Metab 2025; 7:647-664. [PMID: 40175761 DOI: 10.1038/s42255-025-01259-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 03/03/2025] [Indexed: 04/04/2025]
Abstract
L-Lactate has emerged as a crucial metabolic intermediate, moving beyond its traditional view as a mere waste product. The recent discovery of L-lactate-driven protein lactylation as a post-translational modification has unveiled a pathway that highlights the role of lactate in cellular signalling. In this Perspective, we explore the enzymatic and metabolic mechanisms underlying protein lactylation and its impacts on both histone and non-histone proteins in the contexts of physiology and diseases. We discuss growing evidence suggesting that this modification regulates a wide range of cellular functions and is involved in various physiological and pathological processes, such as cell-fate determination, development, cardiovascular diseases, cancer and autoimmune disorders. We propose that protein lactylation acts as a pivotal mechanism, integrating metabolic and signalling pathways to enable cellular adaptation, and highlight its potential as a therapeutic target in various diseases.
Collapse
Affiliation(s)
- Haowen Ren
- State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
| | - Yuwei Tang
- State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
- Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Di Zhang
- State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| |
Collapse
|
6
|
Zhang S, Xie X, Zhao J, Jiang Y, Huang C, Li Q, Xia B, Yin L, Yuan X, You Q. Andrographolide and its Derivatives in Cardiovascular Disease: A Comprehensive Review. PLANTA MEDICA 2025; 91:259-270. [PMID: 40054492 DOI: 10.1055/a-2542-0756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Cardiovascular disease is one of the main causes of mortality worldwide. Andrographolide represents an important category of natural phytochemicals that has significant therapeutic potential in various conditions such as acute lung injury, heart disease, and viral infections due to its anti-oxidative, anti-inflammatory, and anti-apoptotic properties. This compound plays a protective role in human pathophysiology. This review provides a comprehensive overview of the effects of andrographolide on cardiovascular disease and examines its essential roles and mechanisms in cardiovascular disease and other vascular dysfunctions. The data collected in this review serve as a comprehensive reference for the role of andrographolide in cardiovascular disease and provide valuable insights for further research and the development of andrographolide as a novel therapeutic approach for cardiovascular disease.
Collapse
Affiliation(s)
- Shenjie Zhang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaokai Xie
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Juan Zhao
- Department of Cardiology, Tongzhou People's Hospital, Nantong, China
| | - Yilong Jiang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| | - Qi Li
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Boyu Xia
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Le Yin
- Department of Cardiology, Tongzhou People's Hospital, Nantong, China
| | - Xiaomei Yuan
- Department of Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Qingsheng You
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
7
|
Peng TY, Lu JM, Zheng XL, Zeng C, He YH. The role of lactate metabolism and lactylation in pulmonary arterial hypertension. Respir Res 2025; 26:99. [PMID: 40075458 PMCID: PMC11905457 DOI: 10.1186/s12931-025-03163-3] [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: 01/14/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Pulmonary arterial hypertension (PAH) is a complex and progressive disease characterized by elevated pulmonary artery pressure and vascular remodeling. Recent studies have underscored the pivotal role of metabolic dysregulation and epigenetic modifications in the pathogenesis of PAH. Lactate, a byproduct of glycolysis, is now recognized as a key molecule that links cellular metabolism with activity regulation. Recent findings indicate that, in addition to altered glycolytic activity and dysregulated. Lactate homeostasis and lactylation-a novel epigenetic modification-also play a significant role in the development of PAH. This review synthesizes current knowledge regarding the relationship between altered glycolytic activity and PAH, with a particular focus on the cumulative effects of lactate in pulmonary vascular cells. Furthermore, lactylation, an emerging epigenetic modification, is discussed in the context of PAH. By elucidating the complex interplay between lactate metabolism and lactylation in PAH, this review aims to provide insights into potential therapeutic targets. Understanding these metabolic pathways may lead to innovative strategies for managing PAH and improving patient outcomes. Future research should focus on the underlying mechanisms through which lactylation influences the pathophysiology of PAH, thereby aiding in the development of targeted interventions.
Collapse
Affiliation(s)
- Tong-Yu Peng
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jun-Mi Lu
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xia-Lei Zheng
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Cheng Zeng
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yu-Hu He
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
8
|
Llibre A, Kucuk S, Gope A, Certo M, Mauro C. Lactate: A key regulator of the immune response. Immunity 2025; 58:535-554. [PMID: 40073846 DOI: 10.1016/j.immuni.2025.02.008] [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: 11/22/2024] [Revised: 01/22/2025] [Accepted: 02/06/2025] [Indexed: 03/14/2025]
Abstract
Lactate, the end product of both anaerobic and aerobic glycolysis in proliferating and growing cells-with the latter process known as the Warburg effect-is historically considered a mere waste product of cell and tissue metabolism. However, research over the past ten years has unveiled multifaceted functions of lactate that critically shape and impact cellular biology. Beyond serving as a fuel source, lactate is now known to influence gene expression through histone modification and to function as a signaling molecule that impacts a wide range of cellular activities. These properties have been particularly studied in the context of both adaptive and innate immune responses. Here, we review the diverse roles of lactate in the regulation of the immune system during homeostasis and disease pathogenesis (including cancer, infection, cardiovascular diseases, and autoimmunity). Furthermore, we describe recently proposed therapeutic interventions for manipulating lactate metabolism in human diseases.
Collapse
Affiliation(s)
- Alba Llibre
- College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Salih Kucuk
- College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Atrayee Gope
- College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Michelangelo Certo
- College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Claudio Mauro
- College of Medicine and Health, University of Birmingham, Birmingham, UK.
| |
Collapse
|
9
|
Hejazi J, Ghobadian B, Ghasemi N, Sadeh H, Abedimanesh N, Rahimlou M. Relationship of serum irisin levels, physical activity, and metabolic syndrome biomarkers in obese individuals with low-calorie intake and non-obese individuals with high-calorie intake. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2025; 44:2. [PMID: 39748434 PMCID: PMC11697921 DOI: 10.1186/s41043-024-00730-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Despite all the advances in our knowledge regarding obesity, our understanding of its etiology is still far from complete. This study aimed to evaluate the association of serum irisin levels with physical activity and some of the metabolic syndrome-related biomarkers among obese people with low-calorie intake and non-obese people with high-calorie intake. METHODS Obese and non-obese healthy individuals with respectively low and high-calorie intakes were recruited. Irisin and other biomarkers were measured using standard biochemical methods. Participants' physical activity was evaluated by administering the International Physical Activity Questionnaire (IPAQ). To analyze the body composition of the participants, a standard body composition device (ioi 353) was applied. Logistic regression was used to calculate the odds ratio (OR) and to examine the effect of confounders such as age, sex, genetics, and activity. RESULTS Data from the seventy-seven participants were included in the final analysis. The mean age of the participants in the obese and non-obese groups was 38.33 ± 14.88 and 30.24 ± 13.37 years, respectively. Participants in the obese group had lower physical activity compared to the non-obese group (3395.38 ± 2801 MET-min/week vs. 6015.18 ± 3178 MET-min/week; p < 0.001). The Irisin concentration in the obese and non-obese groups was 7.84 ± 2.49 ng/ml and 8.06 ± 1.89 ng/ml, respectively, which wasn't significantly different (p = 0.66). We observed a noteworthy and favorable association between irisin concentration and total body water (TBW), lean body mass (LBM), and soft lean mass (SLM) in the non-obese group. CONCLUSIONS These data indicated that although obese participants were relatively inactive compared to non-obese individuals, circulating irisin level wasn't significantly different between the two groups.
Collapse
Affiliation(s)
- Jalal Hejazi
- Social Determinants of Health Research Center, Health and Metabolic Diseases Research Institute , Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Nutrition, School of Public Health, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Bijan Ghobadian
- Metabolic Diseases Research Center, Health and Metabolic Research Institute , Zanjan University of Medical Science , Zanjan, Iran.
| | - Nasrin Ghasemi
- Zanjan Health and Treatment Center, Health Deputy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hossein Sadeh
- Department of Nutrition, School of Public Health, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Nasim Abedimanesh
- Department of Nutrition, School of Public Health, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehran Rahimlou
- Department of Nutrition, School of Public Health, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
10
|
Pan Y, Wang S, Ming K, Liu X, Yu H, Du Q, Deng C, Chi Q, Liu X, Wang C, Xu K. Leveraging AI technology for distinguishing Eucommiae Cortex processing levels and evaluating anti-fatigue potential. Comput Biol Med 2025; 184:109408. [PMID: 39550909 DOI: 10.1016/j.compbiomed.2024.109408] [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: 05/28/2024] [Revised: 10/14/2024] [Accepted: 11/08/2024] [Indexed: 11/19/2024]
Abstract
Eucommiae Cortex (ECO) is a traditional medicinal and edible plant endemic to China, highly prized for its numerous health benefits. It typically undergoes special processing before application. The efficacy of ECO is influenced by processing techniques, necessitating the assurance of stability and consistency in its effects. However, existing methods for identifying ECO are cumbersome, thus, there is an urgent need to develop an accurate, rapid, and non-invasive assessment method. Deep learning techniques employing ResNet and Vision Transformer (ViT) models were employed to classify ECO images at various processing levels. Concurrently, the anti-fatigue properties of ECO were assessed through swimming time, pole climbing experiments, and biochemical analyses including SDH, LDH, ATP content, Na+-K+-ATPase, and Ca2+-Mg2+-ATPase indices. We demonstrated the efficacy of using image analysis to automatically classify ECO with a high degree of accuracy. The results indicated that the Vision Transformer model performed exceptionally well, achieving an accuracy rate exceeding 95 % in grading ECO images. Additionally, our study revealed that mice treated with moderately processed ECO displayed enhanced fatigue mitigation compared to other processing levels, as evidenced by multiple assessments.
Collapse
Affiliation(s)
- Yijing Pan
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430065, China
| | - Shunshun Wang
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430065, China
| | - Kehong Ming
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430065, China
| | - Xinyue Liu
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430065, China
| | - Huiming Yu
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430065, China
| | - Qianqian Du
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430065, China
| | - Chenxi Deng
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430065, China
| | - Qingjia Chi
- Department of Engineering Structure and Mechanics, School of Science, Wuhan University of Technology, Wuhan, 430070, China
| | - Xianqiong Liu
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430065, China
| | - Chunli Wang
- Hubei Shizhen Laboratory, Wuhan, 430065, China; School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Kang Xu
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430065, China.
| |
Collapse
|
11
|
Wang S, Han J, Wang Z, Liu X, Wang C, Nisar MF, Pan L, Xu K. Targeted Therapy of Tumors and Cancer Stem Cells based on Oxidant-regulated Redox Pathway and its Mechanism. Curr Comput Aided Drug Des 2025; 21:425-440. [PMID: 38818918 DOI: 10.2174/0115734099299174240522115944] [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: 12/17/2023] [Revised: 03/18/2024] [Accepted: 03/28/2024] [Indexed: 06/01/2024]
Abstract
A malignant tumor is a frequent and common disease that severely threatens human health. Many mechanisms, such as cell signaling pathway, anti-apoptosis mechanism, cell stemness, metabolism, and cell phenotype, have been studied to explain the reasons for chemotherapy, radioresistance, and tumor recurrences in antitumor treatment. Cancer stem cells (CSCs) are important tumor cell subclasses that can potentially organize and regulate stem cell properties. Growing evidence suggests that CSCs can initiate tumors and constitute a significant factor in metastasis, recurrence, and treatment resistance. The inability to completely target and remove CSCs is a considerable obstacle in tumor treatment. Therefore, drugs and therapeutic strategies that can effectively intervene with CSCs are essential for the treatment of different tumor types. However, the current strategies and efficacy of targeted elimination of CSCs are very limited. Oxidative stress has been recognized to play a crucial role in cancer pathophysiology. Moreover, reactive oxygen species (ROS) production and imbalance of the built-in cellular antioxidant defense system are hallmarks of tumor and cancer etiology. The current paper will focus on the regulation and mechanism behind oxidative stress in tumors and cancer stem cells and its tumor therapy applications. Additionally, the article discusses the role of CSCs in causing tumor treatment resistance and recurrence based on a redox perspective. The study also emphasizes that targeted modulation of oxidative stress in CSCs has great potential in tumor therapy, providing novel prospects for tumor therapy.
Collapse
Affiliation(s)
- Shunshun Wang
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Juanjuan Han
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Zijun Wang
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xianqiong Liu
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Chunli Wang
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Muhammad Farrukh Nisar
- Department of Physiology and Biochemistry, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur, 63100, Pakistan
| | - Lianhong Pan
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing Three Gorges Medical College, Chongqing, 400030, China
| | - Kang Xu
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| |
Collapse
|
12
|
Pan Y, Ming K, Guo D, Liu X, Deng C, Chi Q, Liu X, Wang C, Xu K. Non-targeted metabolomics and explainable artificial intelligence: Effects of processing and color on coniferyl aldehyde levels in Eucommiae cortex. Food Chem 2024; 460:140564. [PMID: 39089015 DOI: 10.1016/j.foodchem.2024.140564] [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: 05/14/2024] [Revised: 07/15/2024] [Accepted: 07/20/2024] [Indexed: 08/03/2024]
Abstract
Eucommia ulmoides, a plant native to China, is valued for its medicinal properties and has applications in food, health products, and traditional Chinese medicine. Processed Eucommiae Cortex (EC) has historically been a highly valued medicine. Ancient doctors had ample experience processing EC, especially with ginger juice, as documented in traditional Chinese medical texts. The combination of EC and ginger juice helps release and transform the active ingredients, strengthening the medicine's effectiveness and improving its taste and shelf life. However, the lack of quality control standards for Ginger-Eucommiae Cortex (G-EC), processed from EC and ginger, presents challenges for its industrial and clinical use. This study optimized G-EC processing using the CRITIC and Box-Behnken methods. Metabolomics showed 517 chemical changes between raw and processed G-EC, particularly an increase in coniferyl aldehyde (CFA). Explainable artificial intelligence techniques revealed the feasibility of using color to CFA content, providing insights into quality indicators.
Collapse
Affiliation(s)
- Yijing Pan
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430065, China
| | - Kehong Ming
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430065, China
| | - Dongmei Guo
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430065, China
| | - Xinyue Liu
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430065, China
| | - Chenxi Deng
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430065, China
| | - Qingjia Chi
- Hubei Key Laboratory of Theory and Application of Advanced Materials Mechanics, Department of Mechanics and Engineering Structure, Wuhan University of Technology, China.
| | - Xianqiong Liu
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430065, China.
| | - Chunli Wang
- Hubei Shizhen Laboratory, Wuhan 430065, China; School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan 430065, China.
| | - Kang Xu
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430065, China; Center of Traditional Chinese Medicine Modernization for Liver Diseases, Hubei University of Chinese Medicine, Wuhan 430065, China.
| |
Collapse
|
13
|
Zhang H, Zhao J, Yu J, Zhang X, Ran S, Wang S, Ye W, Luo Z, Li X, Hao Y, Zong J, Li R, Lai L, Zheng K, Huang P, Zhou C, Wu J, Li Y, Xia J. Lactate metabolism and lactylation in cardiovascular disease: novel mechanisms and therapeutic targets. Front Cardiovasc Med 2024; 11:1489438. [PMID: 39664763 PMCID: PMC11631895 DOI: 10.3389/fcvm.2024.1489438] [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: 09/01/2024] [Accepted: 11/11/2024] [Indexed: 12/13/2024] Open
Abstract
Cardiovascular disease (CVD) is responsible for approximately 30% of annual global mortality rates, yet existing treatments for this condition are considered less than ideal. Despite being previously overlooked, lactate, a byproduct of glycolysis, is now acknowledged for its crucial role in the cellular functions of the cardiovascular system. Recent studies have shown that lactate influences the proliferation, differentiation, and activation of immune cells through its modulation of post-translational protein modifications, thereby affecting the development and prognosis of cardiovascular disease. Consequently, there has been a notable increase in interest towards drug targets targeting lactylation in immune cells, prompting further exploration. In light of the swift advancements in this domain, this review article is dedicated to examining lactylation in cardiovascular disease and potential drug targets for regulating lactylation, with the aim of enhancing comprehension of this intricate field.
Collapse
Affiliation(s)
- Han Zhang
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiulu Zhao
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jizhang Yu
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xi Zhang
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuan Ran
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Song Wang
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Weicong Ye
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zilong Luo
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaohan Li
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yanglin Hao
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Junjie Zong
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ran Li
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Longyong Lai
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kexiao Zheng
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Pinyan Huang
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Cheng Zhou
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jie Wu
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, ChineseAcademy of Medical Sciences, Wuhan, Hubei, China
| | - Yuan Li
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, ChineseAcademy of Medical Sciences, Wuhan, Hubei, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, ChineseAcademy of Medical Sciences, Wuhan, Hubei, China
| |
Collapse
|
14
|
Zhang S, Ye Y, Li Q, Zhao J, Song R, Huang C, Lu X, Huang C, Yin L, You Q. Andrographolide Attenuates Myocardial Ischemia-Reperfusion Injury in Mice by Up-Regulating PPAR-α. Inflammation 2024:10.1007/s10753-024-02193-1. [PMID: 39585583 DOI: 10.1007/s10753-024-02193-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/06/2024] [Accepted: 11/13/2024] [Indexed: 11/26/2024]
Abstract
Andrographolide (AGP), a bioactive diterpene lactone, is an active constituent extracted from Andrographis paniculata. It has many biological activities, such as antioxidant, antitumor, antivirus, anti-inflammation, hepatoprotection, and cardioprotection. The aim of the present study is to investigate the cardioprotective effects of AGP in a mouse model of myocardial ischemia-reperfusion injury (MIRI). Adult male C57BL/6 J mice were pre-treated orally with AGP (25 mg/kg) for six days. After 30 min of the left anterior descending coronary artery occlusion followed by 24 h of reperfusion, mice received an additional dose of AGP. The results showed that: (i) AGP pretreatment significantly reduced myocardial infarct size and cardiac injury biomarkers in MIRI mice and improved left ventricular ejection fraction (EF) and fractional shortening (FS); (ii) AGP pretreatment attenuated MIRI-induced oxidative stress imbalance in MIRI mice by increasing total antioxidant capacity (T-AOC) and reducing the levels of hydrogen peroxide (H2O2), nitric oxide (NO), malondialdehyde (MDA), and dihydroethidium (DHE); (iii) AGP pretreatment increased Bcl-2 expression and decreased caspase-3 and Bax expression in ischemic myocardial tissue, along with a reduction in TUNEL-positive cells. Further analysis showed that stimulation by I/R decreased peroxisome proliferator-activated receptor-α (PPAR-α) expression in ischemic cardiac tissue, which was prevented by AGP administration. Moreover, administration of the PPAR-α antagonist GW6471 (1 mg/kg) abolished the protective effect of AGP on oxidative stress and apoptosis in the ischemic heart tissue of mice stimulated by ischemia-reperfusion. Taken together, these results suggest that AGP attenuates MIRI-induced cardiac injury by up-regulating PPAR-α expression, thereby preventing oxidative stress and cellular apoptosis.
Collapse
Affiliation(s)
- Shenjie Zhang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Ying Ye
- Department of Ultrasound, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Qi Li
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Juan Zhao
- Department of Cardiology, Tongzhou People's Hospital, 999 Jianshe Road, Nantong, 226300, Jiangsu Province, China
| | - Rongrong Song
- Department of Emergency and Critical Care Medicine, Tongzhou People's Hospital, 999 Jianshe Road, Nantong, 226300, Jiangsu Province, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Chen Huang
- Department of Vascular Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China.
| | - Le Yin
- Department of Cardiology, Tongzhou People's Hospital, 999 Jianshe Road, Nantong, 226300, Jiangsu Province, China.
| | - Qingsheng You
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China.
| |
Collapse
|
15
|
Zhao C, Jin H, Lei Y, Li Q, Zhang Y, Lu Q. The dual effects of Benzo(a)pyrene/Benzo(a)pyrene-7,8-dihydrodiol-9,10-epoxide on DNA Methylation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 950:175042. [PMID: 39084379 DOI: 10.1016/j.scitotenv.2024.175042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/04/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
Benzo(a)pyrene (BaP) is one of the most thoroughly studied polycyclic aromatic hydrocarbons(PAHs) and a widespread organic pollutant in various areas of human life. Its teratogenic, immunotoxic and carcinogenic effects on organisms are well documented and widely recognized by researchers. In the body, BaP is enzymatically converted to form a more active benzo(a)pyrene-7,8-dihydrodiol-9,10-epoxide (BPDE). BaP/BPDE has the potential to trigger gene mutations, influence epigenetic modifications and cause damage to cellular structures, ultimately contributing to disease onset and progression. However, there are different points of view when studying epigenetics using BaP/BPDE. On the one hand, it is claimed in cancer research that BaP/BPDE contributes to gene hypermethylation and, in particular, induces the hypermethylation of tumor's suppressor gene promoters, leading to gene silencing and subsequent cancer development. Conversely, studies in human and animal populations suggest that exposure to BaP results in genome-wide DNA hypomethylation, potentially leading to adverse outcomes in inflammatory diseases. This apparent contradiction has not been summarized in research for almost four decades. This article presents a comprehensive review of the current literature on the influence of BaP/BPDE on DNA methylation regulation. It demonstrates that BaP/BPDE exerts a dual-phase regulatory effect on methylation, which is influenced by factors such as the concentration and duration of BaP/BPDE exposure, experimental models and detection methods used in various studies. Acute/high concentration exposure to BaP/BPDE often results in global demethylation of DNA, which is associated with inhibition of DNA methyltransferase 1 (DNMT1) after exposure. At certain specific gene loci (e.g., RAR-β), BPDE can form DNA adducts, recruiting DNMT3 and leading to hypermethylation at specific sites. By integrating these different mechanisms, our goal is to unravel the patterns and regulations of BaP/BPDE-induced DNA methylation changes and provide insights into future precision therapies targeting epigenetics.
Collapse
Affiliation(s)
- Cheng Zhao
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Central South University Hunan Key Laboratory of Medical Epigenomics Changsha, China; Research Unit of Key Technologies of Immune-related Skin Diseases Diagnosis and Treatment, Chinese Academy of Medical Sciences Institute of Dermatology, Nanjing, China; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China; Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Hui Jin
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China.
| | - Yu Lei
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Central South University Hunan Key Laboratory of Medical Epigenomics Changsha, China; Research Unit of Key Technologies of Immune-related Skin Diseases Diagnosis and Treatment, Chinese Academy of Medical Sciences Institute of Dermatology, Nanjing, China; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China; Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qilin Li
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Central South University Hunan Key Laboratory of Medical Epigenomics Changsha, China; Research Unit of Key Technologies of Immune-related Skin Diseases Diagnosis and Treatment, Chinese Academy of Medical Sciences Institute of Dermatology, Nanjing, China; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China; Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ying Zhang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China; Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qianjin Lu
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Central South University Hunan Key Laboratory of Medical Epigenomics Changsha, China; Research Unit of Key Technologies of Immune-related Skin Diseases Diagnosis and Treatment, Chinese Academy of Medical Sciences Institute of Dermatology, Nanjing, China; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China.
| |
Collapse
|
16
|
Abreu H, Lallukka M, Raineri D, Leigheb M, Ronga M, Cappellano G, Spriano S, Chiocchetti A. Evaluation of the immune response of peripheral blood mononuclear cells cultured on Ti6Al4V-ELI polished or etched surfaces. Front Bioeng Biotechnol 2024; 12:1458091. [PMID: 39439551 PMCID: PMC11493608 DOI: 10.3389/fbioe.2024.1458091] [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: 07/01/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
INTRODUCTION While titanium and its alloys exhibit excellent biocompatibility and corrosion resistance, their polished surfaces can hinder fast and effective osseointegration and other biological processes, such as angiogenesis, due to their inert and hydrophobic properties. Despite being commonly used for orthopedic implants, research focuses on developing surface treatments to improve osseointegration, promoting cell adhesion and proliferation, as well as increasing protein adsorption capacity. This study explores a chemical treatment intended for titanium-based implants that enhances tissue integration without compromising the mechanical properties of the Ti6Al4V substrate. However, recognizing that inflammation contributes to nearly half of early implant failures, we assessed the impact of this treatment on T-cell viability, cytokine production, and phenotype. METHODS Ti6Al4V with extra low interstitial (ELI) content discs were treated with hydrofluoric acid followed by a controlled oxidation step in hydrogen peroxide that creates a complex surface topography with micro- and nano-texture and modifies the chemistry of the surface oxide layer. The acid etched surface contains an abundance of hydroxyl groups, crucial for promoting bone growth and apatite precipitation, while also enabling further functionalization with biomolecules. RESULTS While cell viability remained high in both groups, untreated discs triggered an increase in Th2 cells and a decrease of the Th17 subset. Furthermore, peripheral blood mononuclear cells exposed to untreated discs displayed a rise in various pro-inflammatory and anti-inflammatory cytokines compared to the control and treated groups. Conversely, the treated discs showed a similar profile to the control, both in terms of immune cell subset frequencies and cytokine secretion. DISCUSSION The dysregulation of the cytokine profile upon contact with untreated Ti6Al4V-ELI discs, namely upregulation of IL-2 could be responsible for the decrease in Th17 frequency, and thus might contribute to implant-associated bacterial infection. Interestingly, the chemical treatment restores the immune response to levels comparable to the control condition, suggesting the treatment's potential to mitigate inflammation by enhancing biocompatibility.
Collapse
Affiliation(s)
- Hugo Abreu
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Università del Piemonte Orientale, Novara, Italy
| | - Mari Lallukka
- Applied Science and Technology Department, Politecnico di Torino, Torino, Italy
| | - Davide Raineri
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Università del Piemonte Orientale, Novara, Italy
| | - Massimiliano Leigheb
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
- Orthopaedics and Traumatology Unit, “Maggiore della Carità” Hospital, Novara, Italy
| | - Mario Ronga
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
- Orthopaedics and Traumatology Unit, “Maggiore della Carità” Hospital, Novara, Italy
| | - Giuseppe Cappellano
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Università del Piemonte Orientale, Novara, Italy
| | - Silvia Spriano
- Applied Science and Technology Department, Politecnico di Torino, Torino, Italy
| | - Annalisa Chiocchetti
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
17
|
Huang Y, Wang C, Zhou T, Xie F, Liu Z, Xu H, Liu M, Wang S, Li L, Chi Q, Shi J, Dong N, Xu K. Lumican promotes calcific aortic valve disease through H3 histone lactylation. Eur Heart J 2024; 45:3871-3885. [PMID: 38976370 DOI: 10.1093/eurheartj/ehae407] [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: 09/11/2023] [Revised: 12/06/2023] [Accepted: 06/18/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND AND AIMS Valve interstitial cells (VICs) undergo a transition to intermediate state cells before ultimately transforming into the osteogenic cell population, which is a pivotal cellular process in calcific aortic valve disease (CAVD). Herein, this study successfully delineated the stages of VIC osteogenic transformation and elucidated a novel key regulatory role of lumican (LUM) in this process. METHODS Single-cell RNA-sequencing (scRNA-seq) from nine human aortic valves was used to characterize the pathological switch process and identify key regulatory factors. The in vitro, ex vivo, in vivo, and double knockout mice were constructed to further unravel the calcification-promoting effect of LUM. Moreover, the multi-omic approaches were employed to analyse the molecular mechanism of LUM in CAVD. RESULTS ScRNA-seq successfully delineated the process of VIC pathological transformation and highlighted the significance of LUM as a novel molecule in this process. The pro-calcification role of LUM is confirmed on the in vitro, ex vivo, in vivo level, and ApoE-/-//LUM-/- double knockout mice. The LUM induces osteogenesis in VICs via activation of inflammatory pathways and augmentation of cellular glycolysis, resulting in the accumulation of lactate. Subsequent investigation has unveiled a novel LUM driving histone modification, lactylation, which plays a role in facilitating valve calcification. More importantly, this study has identified two specific sites of histone lactylation, namely, H3K14la and H3K9la, which have been found to facilitate the process of calcification. The confirmation of these modification sites' association with the expression of calcific genes Runx2 and BMP2 has been achieved through ChIP-PCR analysis. CONCLUSIONS The study presents novel findings, being the first to establish the involvement of lumican in mediating H3 histone lactylation, thus facilitating the development of aortic valve calcification. Consequently, lumican would be a promising therapeutic target for intervention in the treatment of CAVD.
Collapse
Affiliation(s)
- Yuming Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chunli Wang
- Hubei Shizhen Laboratory, Wuhan 430065, China
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Tingwen Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Fei Xie
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zongtao Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Haiying Xu
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Ming Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shunshun Wang
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Lanqing Li
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Qingjia Chi
- Department of Engineering Structure and Mechanics, School of Science, Wuhan University of Technology, China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Kang Xu
- Hubei Shizhen Laboratory, Wuhan 430065, China
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| |
Collapse
|
18
|
Feng F, Wu J, Chi Q, Wang S, Liu W, Yang L, Song G, Pan L, Xu K, Wang C. Lactylome Analysis Unveils Lactylation-Dependent Mechanisms of Stemness Remodeling in the Liver Cancer Stem Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405975. [PMID: 39099416 PMCID: PMC11481176 DOI: 10.1002/advs.202405975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/23/2024] [Indexed: 08/06/2024]
Abstract
Lactate plays a critical role as an energy substrate, metabolite, and signaling molecule in hepatocellular carcinoma (HCC). Intracellular lactate-derived protein lysine lactylation (Kla) is identified as a contributor to the progression of HCC. Liver cancer stem cells (LCSCs) are believed to be the root cause of phenotypic and functional heterogeneity in HCC. However, the impact of Kla on the biological processes of LCSCs remains poorly understood. Here enhanced glycolytic metabolism, lactate accumulation, and elevated levels of lactylation are observed in LCSCs compared to HCC cells. H3K56la was found to be closely associated with tumourigenesis and stemness of LCSCs. Notably, a comprehensive examination of the lactylome and proteome of LCSCs and HCC cells identified the ALDOA K230/322 lactylation, which plays a critical role in promoting the stemness of LCSCs. Furthermore, this study demonstrated the tight binding between aldolase A (ALDOA) and dead box deconjugate enzyme 17 (DDX17), which is attenuated by ALDOA lactylation, ultimately enhancing the regulatory function of DDX17 in maintaining the stemness of LCSCs. This investigation highlights the significance of Kla in modulating the stemness of LCSCs and its impact on the progression of HCC. Targeting lactylation in LCSCs may offer a promising therapeutic approach for treating HCC.
Collapse
Affiliation(s)
- Fan Feng
- Hubei Shizhen LaboratoryWuhan430065China
- School of PharmacyHubei University of Chinese MedicineWuhan430065China
| | - Jiaqin Wu
- School of Laboratory MedicineHubei University of Chinese MedicineWuhan430065China
- National Innovation and Attracting Talents “111” baseKey Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqing400000China
| | - Qingjia Chi
- Department of Engineering Structure and MechanicsSchool of ScienceWuhan University of TechnologyWuhan430070China
| | - Shunshun Wang
- Hubei Shizhen LaboratoryWuhan430065China
- School of PharmacyHubei University of Chinese MedicineWuhan430065China
| | - Wanqian Liu
- National Innovation and Attracting Talents “111” baseKey Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqing400000China
| | - Li Yang
- National Innovation and Attracting Talents “111” baseKey Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqing400000China
| | - Guanbin Song
- National Innovation and Attracting Talents “111” baseKey Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqing400000China
| | - Lianhong Pan
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir AreaChongqing Engineering Research Center of Antitumor Natural DrugsChongqing Three Gorges Medical CollegeChongqing400030China
| | - Kang Xu
- Hubei Shizhen LaboratoryWuhan430065China
- School of PharmacyHubei University of Chinese MedicineWuhan430065China
- Center of Traditional Chinese Medicine Modernization for Liver DiseasesHubei University of Chinese MedicineWuhan430065China
| | - Chunli Wang
- Hubei Shizhen LaboratoryWuhan430065China
- School of Laboratory MedicineHubei University of Chinese MedicineWuhan430065China
| |
Collapse
|
19
|
Jiang R, Ren WJ, Wang LY, Zhang W, Jiang ZH, Zhu GY. Targeting Lactate: An Emerging Strategy for Macrophage Regulation in Chronic Inflammation and Cancer. Biomolecules 2024; 14:1202. [PMID: 39456135 PMCID: PMC11505598 DOI: 10.3390/biom14101202] [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: 08/27/2024] [Revised: 09/16/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
Lactate accumulation and macrophage infiltration are pivotal features of both chronic inflammation and cancer. Lactate, once regarded merely as an aftereffect of glucose metabolism, is now gaining recognition for its burgeoning spectrum of biological roles and immunomodulatory significance. Recent studies have evidenced that macrophages display divergent immunophenotypes in different diseases, which play a pivotal role in disease management by modulating macrophage polarization within the disease microenvironment. The specific polarization patterns of macrophages in a high-lactate environment and their contribution to the progression of chronic inflammation and cancer remain contentious. This review presents current evidence on the crosstalk of lactate and macrophage in chronic inflammation and cancer. Additionally, we provide an in-depth exploration of the pivotal yet enigmatic mechanisms through which lactate orchestrates disease pathogenesis, thereby offering novel perspectives to the development of targeted therapeutic interventions for chronic inflammation and cancer.
Collapse
Affiliation(s)
| | | | | | | | - Zhi-Hong Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, Macau Institute for Applied Research in Medicine and Health, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; (R.J.); (W.-J.R.); (L.-Y.W.); (W.Z.)
| | - Guo-Yuan Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, Macau Institute for Applied Research in Medicine and Health, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; (R.J.); (W.-J.R.); (L.-Y.W.); (W.Z.)
| |
Collapse
|
20
|
Bukhari HA, Afzal M, Al-Abbasi FA, Sheikh RA, Alqurashi MM, Bawadood AS, Alzarea SI, Alamri A, Sayyed N, Kazmi I. In vivo and computational investigation of butin against alloxan-induced diabetes via biochemical, histopathological, and molecular interactions. Sci Rep 2024; 14:20633. [PMID: 39232184 PMCID: PMC11374895 DOI: 10.1038/s41598-024-71577-y] [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: 01/23/2024] [Accepted: 08/29/2024] [Indexed: 09/06/2024] Open
Abstract
Herbs have been used as medicines since antiquity, and it has been discovered that the human body responds well to herbal remedies. Research on the effect of butin was conducted in the current study in the alloxan-induced diabetic rat paradigm. A total of 30 Wistar rats were randomly assigned into the following groups (n = 6): I-Normal; II-Alloxan-induced (50 mg/kg); III-Alloxan + butin 25 mg/kg; IV-Alloxan + butin 50 mg/kg; V-Butin per se 50 mg/kg. Various diabetic parameters (blood glucose, insulin, HbA1c), lipid profile, inflammatory (TNF-α, IL-1β, IL-6 and NF-κB), antioxidant enzymes (CAT, SOD and GSH), oxidative stress indicators (MDA), apoptosis marker (caspase-3), hepatic markers (ALT and AST), and histopathological changes were assessed. Additionally, molecular docking and dynamics were performed to evaluate the interaction of butin with target proteins. Butin treatment, at both doses, significantly restored biochemical parameters and preserved pancreatic histopathology in diabetic rats. It effectively modulated blood parameters, lipid profiles, inflammatory markers, apoptosis, antioxidant enzyme activity, oxidative stress, and hepatic markers. Molecular docking revealed that butin binds to proteins such as caspase-3 (1NME), NF-κB (1SVC), and serum insulin (4IBM) with binding affinities of - 7.4, - 6.5, and - 8.2 kcal/mol, respectively. Molecular dynamics simulations further suggested that butin induces significant conformational changes in these proteins. Butin exhibits potential effects against alloxan-induced diabetic rats by restoring biochemical balance, reducing inflammation, and protecting pancreatic tissue. Its binding to key proteins involved in apoptosis and inflammation highlights its therapeutic potential in diabetes management.
Collapse
Affiliation(s)
- Hussam A Bukhari
- Department of Pathology, Faculty of Medicine, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
- King Abdulaziz University Hospital, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Muhammad Afzal
- Pharmacy Program, Department of Pharmaceutical Sciences, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Fahad A Al-Abbasi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Ryan A Sheikh
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - May M Alqurashi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Azizah Salim Bawadood
- Basic Medical Sciences Department, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Aljouf, Sakaka, Saudi Arabia
| | - Abdulaziz Alamri
- Department of Biochemistry, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Nadeem Sayyed
- School of Pharmacy, Glocal University, Saharanpur, 247121, India
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, 21589, Jeddah, Saudi Arabia.
| |
Collapse
|
21
|
Ur Rahman S, Alam A, Parveen Z, Zainab, Assad M, Adnan Ali Shah S, Rafiq H, Ayaz M, Latif A, Naveed Umar M, Ali M, Ahmad M. Novel acyl hydrazide derivatives of polyhydroquinoline as potent anti-diabetic and anti-glycating agents: Synthesis, in vitro α-amylase, α-glucosidase inhibition and anti-glycating activity with molecular docking insights. Bioorg Chem 2024; 150:107501. [PMID: 38865858 DOI: 10.1016/j.bioorg.2024.107501] [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: 04/02/2024] [Revised: 05/24/2024] [Accepted: 05/25/2024] [Indexed: 06/14/2024]
Abstract
In this study, eleven novel acyl hydrazides derivative of polyhydroquinoline were synthesized, characterized and screened for their in vitro anti-diabetic and anti-glycating activities. Seven compounds 2a, 2d, 2i, 2 h, 2j, 2f, and 2 g exhibited notable α-amylase inhibitory activity having IC50 values from 3.51 ± 2.13 to 11.92 ± 2.30 µM. Similarly, six compounds 2d, 2f, 2 h, 2i, 2j, and 2 g displayed potent α-glucosidase inhibitory activity compared to the standard acarbose. Moreover, eight derivatives 2d, 2 g, 2f, 2j, 2a, 2i, 2 g, and 2e showed excellent anti-glycating activity with IC50 values from 6.91 ± 2.66 to 15.80 ± 1.87 µM when compared them with the standard rutin (IC50 = 22.5 ± 0.90 µM). Molecular docking was carried out to predict the binding modes of all the compounds with α-amylase and α-glucosidase. The docking analysis revealed that most of the compounds established strong interactions with α-amylase and α-glucosidase. All compounds fitted well into the binding pockets of α-amylase and α-glucosidase. Among all compounds 2a and 2f were most potent based on docking score -8.2515 and -7.3949 against α-amylase and α-glucosidase respectively. These results hold promise for the development of novel candidates targeted at controlling postprandial glucose levels in individuals with diabetes.
Collapse
Affiliation(s)
- Sajjad Ur Rahman
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Khyber Pakhtunkhwa, Pakistan
| | - Aftab Alam
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Khyber Pakhtunkhwa, Pakistan
| | - Zahida Parveen
- Department of Biochemistry, Abdul Wali Khan University Mardan, Khyber Pakhtunkhwa 23200, Pakistan
| | - Zainab
- College of Chemistry and Materials Science, Hebei Normal University, Shijiazhuang 050024, China
| | - Mohammad Assad
- Department of Biochemistry, Abdul Wali Khan University Mardan, Khyber Pakhtunkhwa 23200, Pakistan
| | - Syed Adnan Ali Shah
- Faculty of Pharmacy, Universiti Teknologi MARA Cawangan Selangor Kampus Puncak Alam, Bandar Puncak Alam, Selangor 42300, Malaysia; Atta-ur-Rahman Institute for Natural Products Discovery (AuRIns), Universiti Teknologi MARA Cawangan Selangor Kampus Puncak Alam, Bandar Puncak Alam, Selangor 42300, Malaysia
| | - Huma Rafiq
- Department of Biochemistry, Abdul Wali Khan University Mardan, Khyber Pakhtunkhwa 23200, Pakistan
| | - Muhammad Ayaz
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Khyber Pakhtunkhwa, Pakistan
| | - Abdul Latif
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Khyber Pakhtunkhwa, Pakistan
| | - Muhammad Naveed Umar
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Khyber Pakhtunkhwa, Pakistan.
| | - Mumtaz Ali
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Khyber Pakhtunkhwa, Pakistan.
| | - Manzoor Ahmad
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Khyber Pakhtunkhwa, Pakistan.
| |
Collapse
|
22
|
Singh G, Singh R, Monga V, Mehan S. Thiazolidine-2,4-dione hybrids as dual alpha-amylase and alpha-glucosidase inhibitors: design, synthesis, in vitro and in vivo anti-diabetic evaluation. RSC Med Chem 2024; 15:2826-2854. [PMID: 39149094 PMCID: PMC11324062 DOI: 10.1039/d4md00199k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/17/2024] [Indexed: 08/17/2024] Open
Abstract
Twelve 3,5-disubstituted-thiazolidine-2,4-dione (TZD) hybrids were synthesized using solution phase chemistry. Continuing our previous work, nine O-modified ethyl vanillin (8a-i) derivatives were synthesized and reacted with the TZD core via Knoevenagel condensation under primary reaction conditions to obtain final derivatives 9a-i. Additionally, three isatin-TZD hybrids (11a-c) were synthesized. The intermediates and final derivatives were characterized using 1H and 13C NMR spectroscopy, and the observed chemical shifts agreed with the proposed structures. The in vitro alpha-amylase and alpha-glucosidase inhibitory evaluation of newly synthesized derivatives revealed compounds 9F and 9G as the best dual inhibitors, with IC50 values of 9.8 ± 0.047 μM for alpha-glucosidase (9F) and 5.15 ± 0.0017 μM for alpha-glucosidase (9G), 17.10 ± 0.015 μM for alpha-amylase (9F), and 9.2 ± 0.092 μM for alpha-amylase (9G). The docking analysis of synthesized compounds indicated that compounds have a higher binding affinity for alpha-glucosidase as compared to alpha-amylase, as seen from docking scores ranging from -1.202 to -5.467 (for alpha-amylase) and -4.373 to -7.300 (for alpha-glucosidase). Further, the molecules possess a high LD50 value, typically ranging from 1000 to 1600 mg kg-1 of body weight, and exhibit non-toxic properties. The in vitro cytotoxicity assay results on PANC-1 and INS-1 cells demonstrated that the compounds were devoid of significant toxicity against the tested cells. Compounds 9F and 9G showed high oral absorption, i.e., oral absorption >96%, and their molecular dynamics simulation yielded results closely aligned with the observed docking outcomes. Finally, compounds 9F and 9G were evaluated for in vivo antidiabetic assessment by the induction of diabetes in Wistar rats using streptozotocin. Molecule 9G has been identified as the most effective anti-diabetic molecule due to its ability to modulate several biochemical markers in blood plasma and tissue homogenates. The results were further confirmed by histology investigations conducted on isolated pancreas, liver, and kidney.
Collapse
Affiliation(s)
- Gurpreet Singh
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, (Affiliated to IK Gujral Punjab Technical University, Kapurthala) GT Road, Ghal Kalan Moga-142001 Punjab India
- Research Scholar, IK Gujral Punjab Technical University Kapurthala Punjab India
| | - Rajveer Singh
- Department of Pharmacognosy, ISF College of Pharmacy GT Road, Ghal Kalan Moga Punjab India
| | - Vikramdeep Monga
- Drug Design and Molecular Synthesis Laboratory, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab VPO-Ghudda Bathinda Punjab India
| | - Sidharth Mehan
- Department of Pharmacology, ISF College of Pharmacy, (Affiliated to IK Gujral Punjab Technical University, Kapurthala) GT Road, Ghal Kalan Moga Punjab India
| |
Collapse
|
23
|
Jetti R, Vaca Cárdenas ML, Al-Saedi HFS, Hussein SA, Abdulridui HA, Al-Abdeen SHZ, Radi UK, Abdulkadhim AH, Hussein SB, Alawadi A, Alsalamy A. Ultrasonic synthesis of green lipid nanocarriers loaded with Scutellaria barbata extract: a sustainable approach for enhanced anticancer and antibacterial therapy. Bioprocess Biosyst Eng 2024; 47:1321-1334. [PMID: 38647679 DOI: 10.1007/s00449-024-03021-4] [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: 01/20/2024] [Accepted: 04/10/2024] [Indexed: 04/25/2024]
Abstract
Ultrasonic manufacturing has emerged as a promising eco-friendly approach to synthesize lipid-based nanocarriers for targeted drug delivery. This study presents the novel ultrasonic preparation of lipid nanocarriers loaded with Scutellaria barbata extract, repurposed for anticancer and antibacterial use. High-frequency ultrasonic waves enabled the precise self-assembly of DSPE-PEG, Span 40, and cholesterol to form nanocarriers encapsulating the therapeutic extract without the use of toxic solvents, exemplifying green nanotechnology. Leveraging the inherent anticancer and antibacterial properties of Scutellaria barbata, the study demonstrates that lipid encapsulation enhances the bioavailability and controlled release of the extract, which is vital for its therapeutic efficacy. Dynamic light scattering and transmission electron microscopy analyses confirmed the increase in size and successful encapsulation post-loading, along with an augmented negative zeta potential indicating enhanced stability. A high encapsulation efficiency of 91.93% was achieved, and in vitro assays revealed the loaded nanocarriers' optimized release kinetics and improved antimicrobial potency against Pseudomonas aeruginosa, compared to the free extract. The combination of ultrasonic synthesis and Scutellaria barbata in an eco-friendly manufacturing process not only advances green nanotechnology but also contributes to sustainable practices in pharmaceutical manufacturing. The data suggest that this innovative nanocarrier system could provide a robust platform for the development of nanotechnology-based therapeutics, enhancing drug delivery efficacy while aligning with environmental sustainability.
Collapse
Affiliation(s)
- Raghu Jetti
- Department of Basic Medical Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Maritza Lucia Vaca Cárdenas
- Facultad de Ciencias Pecuarias, Escuela Superior Politécnica de Chimborazo (ESPOCH), Panamericana Sur Km 1½, Riobamba, 060155, Ecuador
| | | | | | | | | | - Usama Kadem Radi
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Adnan Hashim Abdulkadhim
- Department of Computer Engineering, Technical Engineering College, Al-Ayen University, Dhi Qar, Iraq
| | | | - Ahmed Alawadi
- College of Technical Engineering, The Islamic University, Najaf, Iraq.
- College of Technical Engineering, The Islamic University of Al-Diwaniyah, Al-Diwaniyah, Iraq.
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq.
| | - Ali Alsalamy
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna, 66002, Iraq
| |
Collapse
|
24
|
Ghasemi P, Jafari M, Maskouni SJ, Hosseini SA, Amiri R, Hejazi J, Chambari M, Tavasolian R, Rahimlou M. Impact of very low carbohydrate ketogenic diets on cardiovascular risk factors among patients with type 2 diabetes; GRADE-assessed systematic review and meta-analysis of clinical trials. Nutr Metab (Lond) 2024; 21:50. [PMID: 39030553 PMCID: PMC11264514 DOI: 10.1186/s12986-024-00824-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 07/08/2024] [Indexed: 07/21/2024] Open
Abstract
OBJECTIVE This study was designed to evaluate the impact of VLCKD on cardiovascular risk factors in patients with T2DM. METHODS Until March 2024, extensive searches were conducted on PubMed, Scopus, Web of Science, Embase, and other relevant databases. The purpose was to identify clinical trials examining the impact of VLCKD on glycemic control, lipid profile, and blood pressure. The GRADE (Grading of Recommendations Assessment, Development, and Evaluation) method was used to assess the evidence's degree of certainty. RESULTS Our initial search found a total of 2568 records and finally 29 trials were included in final analysis. Our results showed that adherence from VLCKD led to significant reduction in fasting blood sugar (WMD= -11.68 mg/dl; 95% CI: -18.79, -4.56; P = 0.001), HbA1c (WMD= -0.29; 95% CI: -0.44, -0.14; P < 0.001), HOMA-IR(WMD= -0.71; 95% CI: -1.14, -0.29; P = 0.001), insulin (WMD= -1.45; 95% CI: -2.54, -0.36; P = 0.009), triglyceride (WMD= -17.95; 95% CI: -26.82, -9.07; P < 0.001), systolic blood pressure (WMD= -2.85, 95% CI: -4.99, -0.71; P = 0.009) and diastolic blood pressure (WMD= -1.40; 95% CI: -2.66, -0.13; P = 0.03). We also found a significant increase in high-density lipoprotein (HDL) level after adherence from VLCKD diet (WMD = 3.93, 95% CI: 2.03, 5.84; P = 0.000). We couldn't find any significant differences between groups in term of LDL and total cholesterol levels. CONCLUSION People following a VLCKD experience a more significant improvement in cardiovascular risk factors when compared to individuals on control diets.
Collapse
Affiliation(s)
- Parisa Ghasemi
- Research Committee, Medical School, Arak University of Medical Sciences, Arak, Iran
| | - Malihe Jafari
- Department of Exercise physiology, Faculty of Physical Education and Sports Sciences, Allameh Tabataba'i University, Tehran, Iran
| | - Saber Jafari Maskouni
- Department of nutrition, School of Public health, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Seyed Ahmad Hosseini
- Department of Nutrition, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Roksaneh Amiri
- Mofid Childrens Hospital Clinical Research Development Unit, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Jalal Hejazi
- Department of Nutrition, School of Public Health, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mahla Chambari
- Department of Food Science and Nutrition, Faculty of Applied Sciences, UCSI University, 56000 Cheras, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Ronia Tavasolian
- Department of Nutrition, School of Public Health, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehran Rahimlou
- Department of Nutrition, School of Public Health, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
25
|
Wei SJ, Xiong Q, Yao H, He QM, Yu PL. Is systemic lupus erythematosus linked to Immunoglobulin G4 Autoantibodies? Hum Immunol 2024; 85:110826. [PMID: 38954949 DOI: 10.1016/j.humimm.2024.110826] [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: 08/26/2023] [Revised: 04/26/2024] [Accepted: 05/21/2024] [Indexed: 07/04/2024]
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disorder characterized by a hyperactive immune system with multiple abnormalities in B-cell proliferation, antibody production, T-cell regulation, and immune complex (IC) formation. In humans, Immunoglobulin (Ig) G is found in four subclasses. IgG1-IgG4, which are distinguished by both structural and biological differences. Fab-arm Exchange (FAE), specific biases in the IgG4 response repertoire, and a decreased capacity to induce effector functions mediated by interactions in the fragment crystallizable (Fc) region are just a few of the distinctive characteristics of IgG4. The recent finding of the presence of double-stranded DNA (dsDNA) and antinuclear antibody (ANA)-IgG4 has raised attention to this IgG subclass and its possible role in SLE. IgG4 was previously believed to just have anti-inflammatory effects by inhibiting immune responses, but recent studies have shown that these antibodies can also play a role in the onset and development of some clinical disorders. To consider the clinical effects of IgG4 presence, it is necessary to discuss its characteristics, which could underlie the potential role it can play in SLE. Therefore, this study aimed to comprehensively review the role of IgG4 in SLE to elucidate the collective incidence of high IgG4 levels reported in some SLE patients.
Collapse
Affiliation(s)
- Shu-Jun Wei
- Sichuan Police College, Longtouguan Road, Jiangyang District, Luzhou City, Sichuan Province, China
| | - Qian Xiong
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing Three Gorges Medical College, China
| | - Huan Yao
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan, China; Sichuan Provincial Engineering Research Center of Innovative Re-development of Famous Classical Formulas, Pengzhou 611930, China
| | - Qing-Man He
- Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peng-Long Yu
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing Three Gorges Medical College, China.
| |
Collapse
|
26
|
Bhaumik S, Sarkar A, Debnath S, Debnath B, Ghosh R, Zaki ME, Al-Hussain SA. α-Glucosidase inhibitory potential of Oroxylum indicum using molecular docking, molecular dynamics, and in vitro evaluation. Saudi Pharm J 2024; 32:102095. [PMID: 38766274 PMCID: PMC11101736 DOI: 10.1016/j.jsps.2024.102095] [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: 02/19/2024] [Accepted: 05/01/2024] [Indexed: 05/22/2024] Open
Abstract
Background According to the International Diabetes Federation, there will be 578 million individuals worldwide with diabetes by 2030 and 700 million by 2045. One of the promising drug targets to fight diabetes is α-glucosidase (AG), and its inhibitors may be used to manage diabetes by reducing the breakdown of complex carbohydrates into simple sugars. The study aims to identify and validate potential AG inhibitors in natural sources to combat diabetes. Methods Computational techniques such as structure-based virtual screening and molecular dyncamic simulation were employed to predict potential AG inhibitors from compounds of Oroxylum indicum. Finally, in silico results were validated by in vitro analysis using n-butanol fraction of crude methanol extracts. Results The XP glide scores of top seven hits OI_13, OI_66, OI_16, OI_44, OI_43, OI_20, OI_78 and acarbose were -14.261, -13.475, -13.074, -13.045, -12.978, -12.659, -12.354 and -12.296 kcal/mol, respectively. These hits demonstrated excellent binding affinity towards AG, surpassing the known AG inhibitor acarbose. The MM-GBSA dG binding energies of OI_13, OI_66, and acarbose were -69.093, -62.950, and -53.055 kcal/mol, respectively. Most of the top hits were glycosides, indicating that active compounds lie in the n-butanol fraction of the extract. The IC50 value for AG inhibition by n-butanol fraction was 248.1 μg/ml, and for that of pure acarbose it was 89.16 μg/ml. The predicted oral absorption rate in humans for the top seven hits was low like acarbose, which favors the use of these compounds as anti-diabetes in the small intestine. Conclusion In summary, the study provides promising insights into the use of natural compounds derived from O. indicum as potential AG inhibitors to manage diabetes. However, further research, including clinical trials and pharmacological studies, would be necessary to validate their efficacy and safety before clinical use.
Collapse
Affiliation(s)
- Samhita Bhaumik
- Department of Chemistry, Women’s College, Agartala, Tripura 799001, India
| | - Alekhya Sarkar
- Department of Forestry and Biodiversity, Tripura University, Suryamaninagar, Tripura, India
| | - Sudhan Debnath
- Department of Chemistry, Netaji Subhash Mahavidyalaya, Udaipur, Tripura 799 114, India
| | - Bimal Debnath
- Department of Forestry and Biodiversity, Tripura University, Suryamaninagar, Tripura, India
| | - Rajat Ghosh
- In Silico Drug Design Lab., Department of Pharmacy, Tripura University, Suryamaninagar, Tripura, India
| | - Magdi E.A. Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh 11623, Saudi Arabia
| | - Sami A. Al-Hussain
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh 11623, Saudi Arabia
| |
Collapse
|
27
|
Li X, Cai P, Tang X, Wu Y, Zhang Y, Rong X. Lactylation Modification in Cardiometabolic Disorders: Function and Mechanism. Metabolites 2024; 14:217. [PMID: 38668345 PMCID: PMC11052226 DOI: 10.3390/metabo14040217] [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: 03/12/2024] [Revised: 04/01/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
Cardiovascular disease (CVD) is recognized as the primary cause of mortality and morbidity on a global scale, and developing a clear treatment is an important tool for improving it. Cardiometabolic disorder (CMD) is a syndrome resulting from the combination of cardiovascular, endocrine, pro-thrombotic, and inflammatory health hazards. Due to their complex pathological mechanisms, there is a lack of effective diagnostic and treatment methods for cardiac metabolic disorders. Lactylation is a type of post-translational modification (PTM) that plays a regulatory role in various cellular physiological processes by inducing changes in the spatial conformation of proteins. Numerous studies have reported that lactylation modification plays a crucial role in post-translational modifications and is closely related to cardiac metabolic diseases. This article discusses the molecular biology of lactylation modifications and outlines the roles and mechanisms of lactylation modifications in cardiometabolic disorders, offering valuable insights for the diagnosis and treatment of such conditions.
Collapse
Affiliation(s)
- Xu Li
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; (X.L.); (P.C.); (X.T.); (Y.W.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Guangzhou 510006, China
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Guangzhou 510006, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Pingdong Cai
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; (X.L.); (P.C.); (X.T.); (Y.W.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Guangzhou 510006, China
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Guangzhou 510006, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xinyuan Tang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; (X.L.); (P.C.); (X.T.); (Y.W.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Guangzhou 510006, China
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Guangzhou 510006, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yingzi Wu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; (X.L.); (P.C.); (X.T.); (Y.W.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Guangzhou 510006, China
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Guangzhou 510006, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yue Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; (X.L.); (P.C.); (X.T.); (Y.W.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Guangzhou 510006, China
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Guangzhou 510006, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xianglu Rong
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; (X.L.); (P.C.); (X.T.); (Y.W.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Guangzhou 510006, China
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Guangzhou 510006, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|