1
|
Gao C, Li J, Shan B. Research progress on the regulatory role of lactate and lactylation in tumor microenvironment. Biochim Biophys Acta Rev Cancer 2025; 1880:189339. [PMID: 40311713 DOI: 10.1016/j.bbcan.2025.189339] [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: 11/27/2024] [Revised: 04/26/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
The tumor microenvironment (TME) arises from the dynamic interactions between tumor cells and the surrounding medium, including a variety of cell types and extracellular components, which have an important impact on the genesis and development of tumors. A key player in TME is lactate, a metabolic byproduct of glycolysis, which serves as a significant energy source. Lactate has direct implications on the survival and differentiation of immune cells, the metabolic reprogramming and progression of tumor cells. Moreover, lactylation, a unique post-translational modification, exerts a regulatory effect on TME by affecting gene transcription via adding lactate groups to both histone and non-histone proteins. This review systematically and comprehensively synthesizes emerging evidence on how the lactate-lactylation axis drives immune evasion, therapy resistance, and TME remodeling, highlighting the therapeutic targets related to lactate and lactylation that dismantle this metabolic-epigenetic crosstalk.
Collapse
Affiliation(s)
- Chunyan Gao
- Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, China; Key Laboratory of Tumor Prevention, Precision Diagnosis and Treatment of Hebei, Clinical Oncology Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, China
| | - Jiali Li
- Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, China; Key Laboratory of Tumor Prevention, Precision Diagnosis and Treatment of Hebei, Clinical Oncology Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, China
| | - Baoen Shan
- Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, China; Key Laboratory of Tumor Prevention, Precision Diagnosis and Treatment of Hebei, Clinical Oncology Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, China.
| |
Collapse
|
2
|
Chen X, Yuan Y, Zhou F, Li L, Pu J, Zeng Y, Jiang X. Lactylation: From Homeostasis to Pathological Implications and Therapeutic Strategies. MedComm (Beijing) 2025; 6:e70226. [PMID: 40443721 PMCID: PMC12122191 DOI: 10.1002/mco2.70226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 04/16/2025] [Accepted: 04/24/2025] [Indexed: 06/02/2025] Open
Abstract
Lactylation, a recently identified post-translational modification, represents a groundbreaking addition to the epigenetic landscape, revealing its pivotal role in gene regulation and metabolic adaptation. Unlike traditional modifications, lactylation directly links metabolic intermediates, such as lactate, to protein function and cellular behavior. Emerging evidence highlights the critical involvement of lactylation in diverse biological processes, including immune response modulation, cellular differentiation, and tumor progression. However, its regulatory mechanisms, biological implications, and disease associations remain poorly understood. This review systematically explores the enzymatic and nonenzymatic mechanisms underlying protein lactylation, shedding light on the interplay between cellular metabolism and epigenetic control. We comprehensively analyze its biological functions in normal physiology, such as immune homeostasis and tissue repair, and its dysregulation in pathological contexts, including cancer, inflammation, and metabolic disorders. Moreover, we discuss advanced detection technologies and potential therapeutic interventions targeting lactylation pathways. By integrating these insights, this review aims to bridge critical knowledge gaps and propose future directions for research. Highlighting lactylation's multifaceted roles in health and disease, this review provides a timely resource for understanding its clinical implications, particularly as a novel target for precision medicine in metabolic and oncological therapies.
Collapse
Affiliation(s)
- Xi Chen
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan ProvinceThe Second Affiliated Hospital of Kunming Medical UniversityKunmingChina
- NHC Key Laboratory of Drug Addiction MedicineKunming Medical UniversityKunmingChina
| | - Yixiao Yuan
- Department of Medicine, UF Health Cancer CenterUniversity of FloridaGainesvilleFloridaUSA
| | - Fan Zhou
- Department of Hematologythe Second Hospital Affiliated to Kunming Medical UniversityKunmingChina
| | - Lihua Li
- NHC Key Laboratory of Drug Addiction MedicineKunming Medical UniversityKunmingChina
| | - Jun Pu
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan ProvinceThe Second Affiliated Hospital of Kunming Medical UniversityKunmingChina
- NHC Key Laboratory of Drug Addiction MedicineKunming Medical UniversityKunmingChina
| | - Yong Zeng
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan ProvinceThe Second Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Xiulin Jiang
- Department of Medicine, UF Health Cancer CenterUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
3
|
Zou W, Huo B, Tu Y, Zhu Y, Hu Y, Li Q, Yu X, Liu B, Tang W, Tan S, Xiao H. Metabolic reprogramming by chemo-gene co-delivery nanoparticles for chemo-immunotherapy in head and neck squamous cell carcinoma. Acta Biomater 2025; 199:361-373. [PMID: 40252747 DOI: 10.1016/j.actbio.2025.04.031] [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: 01/28/2025] [Revised: 03/24/2025] [Accepted: 04/14/2025] [Indexed: 04/21/2025]
Abstract
The therapeutic effects of platinum-based drugs are closely linked to the dysregulation of tumor metabolic-immune microenvironment, particularly aberrant lactate accumulation. Herein, we engineered multifunctional nanoparticles (PPPtIV NPs) through electrostatic self-assembly of poly(β-amino ester) to co-encapsulate a cisplatin prodrug (PtIV) and CRISPR/Cas9-PKM2 plasmids. Mechanistically, PPPtIV NPs efficiently entered cells via endocytosis, followed by escape from lysosomal degradation and cargo release. The reduction of PtIV prodrug to active PtII via GSH depletion induced DNA damage and ROS upregulation, thereby triggering apoptosis. Concurrently, CRISPR/Cas9-mediated PKM2 knockdown suppressed the Warburg effect, resulting in reduced lactate production and downregulated expression of HIF-1α and PD-L1. These alterations drove immune microenvironment remodeling through enhanced dendritic cell maturation, polarized M1 macrophages, and altered cytokine profiles (characterized by upregulation of IFN-γ, TNF-α, and IL-12 alongside suppression of IL-10), ultimately activating T cell-mediated antitumor immunity. Compared to conventional cisplatin, PPPtIV NPs demonstrated superior efficacy against both primary and recurrent tumors while reducing nephrotoxicity through synergistic chemo-immunotherapeutic effects, offering a valuable strategy for HNSCC treatment. STATEMENT OF SIGNIFICANCE: This study engineered an innovative nanoplatform (PPPtIV) that synergistically integrates a PtIV prodrug with a CRISPR/Cas9-PKM2 plasmid for treating head and neck squamous cell carcinoma. By simultaneously enhancing DNA damage and reversing lactate-mediated immunosuppression, PPPtIV nanoplatform achieved chemo-immunotherapy that showed greater suppression of primary and recurrent tumors with reduced renal toxicity compared to cisplatin. This nanotechnology-driven strategy provides valuable insights into the combination of platinum-based drugs with immunometabolic interventions.
Collapse
Affiliation(s)
- Wenqing Zou
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bingyue Huo
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yaqin Tu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuhe Zhu
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuwei Hu
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qianru Li
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xuan Yu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Otorhinolaryngology-Head and Neck Surgery, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Clinical Research Center for Deafness and Vertigo, Wuhan 430022, China
| | - Bo Liu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Otorhinolaryngology-Head and Neck Surgery, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Clinical Research Center for Deafness and Vertigo, Wuhan 430022, China
| | - Wei Tang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Otorhinolaryngology-Head and Neck Surgery, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Clinical Research Center for Deafness and Vertigo, Wuhan 430022, China
| | - Songwei Tan
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Hongjun Xiao
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Otorhinolaryngology-Head and Neck Surgery, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Clinical Research Center for Deafness and Vertigo, Wuhan 430022, China.
| |
Collapse
|
4
|
Li L, Lu J, Fu S, Li W, Wang Y, Wang K, Tao Y, Liu S. Evodiamine induces ferroptosis in prostate cancer cells by inhibiting TRIM26-mediated stabilization of GPX4. Chin Med 2025; 20:71. [PMID: 40420092 PMCID: PMC12105283 DOI: 10.1186/s13020-025-01130-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Accepted: 05/12/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Prostate cancer is a major global health challenge, characterized by high morbidity and mortality rates. Traditional treatment options, including androgen deprivation therapy and chemotherapy, often lead to drug resistance. In recent years, natural compounds have garnered attention for their potential therapeutic effects. Evodiamine, a bioactive alkaloid from Evodia rutaecarpa, has demonstrated promising anti-cancer properties in various malignancies, including oral squamous cell carcinoma, breast, colorectal, and ovarian cancers. This study explores the efficacy of evodiamine in prostate cancer cells and investigates the mechanisms underlying evodiamine-induced cell death. METHODS To investigate the effects of evodiamine on prostate cancer cells, various cell lines, including both castration-sensitive and castration-resistant variants, were treated with different concentrations of evodiamine for various durations. Cell viability, proliferation, invasion ability, and colony formation were assessed using the CCK8 assay, EdU assay, 3D matrigel drop invasion assay, and colony formation assay, respectively. The effects of evodiamine on apoptosis were analyzed using FACS, Hoechst staining, and Western blot. To evaluate its effects on ferroptosis, malondialdehyde (MDA) and glutathione (GSH) assay kits, as well as DCFH-DA and the lipid peroxidation sensor BODIPY™ 581/501 C11 fluorescent probes, were employed. The molecular mechanisms through which evodiamine regulates GPX4 protein instability were investigated using Western blot and TRIM26 ectopic expression. Additionally, a mouse xenograft model derived from DU145 cells was established to evaluate the in vivo effects of evodiamine and its molecular mechanisms, utilizing hematoxylin and eosin (H&E) staining, immunohistochemistry (IHC), and Western blot analysis. RESULTS Evodiamine significantly suppressed cell viability, proliferation, invasion, and colony formation in prostate cancer cells. Importantly, evodiamine-induced cell death in the PC3 and DU145 cell lines was independent of apoptosis pathway. Instead, evodiamine increased reactive oxygen species (ROS) production, lipid ROS levels and MDA levels, while decreasing GSH levels, indicating the induction of ferroptosis. The key role of ROS in evodiamine-induced ferroptosis was further confirmed by the partial reversal of cell death upon treatment with the ROS scavenger N-acetylcysteine (NAC). Mechanistically, evodiamine induced ferroptosis by destabilizing GPX4 protein in a TRIM26-dependent manner. Moreover, in vivo studies demonstrated that evodiamine significantly inhibited tumor growth and induced ferroptosis in tumor cells, highlighting its therapeutic potential. CONCLUSION This study demonstrates that evodiamine exerts potent antitumor effects against prostate cancer through inhibiting TRIM26-mediated stabilization of GPX4 protein and triggering ferroptosis. These findings suggest that evodiamine, a natural product derived from traditional Chinese medicine, could be a promising therapeutic agent for prostate cancer.
Collapse
Affiliation(s)
- Lanlan Li
- Institute of Urology, Gansu Province Clinical Research Center for Urinary System Diseases, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Jianzhong Lu
- Institute of Urology, Gansu Province Clinical Research Center for Urinary System Diseases, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Shengjun Fu
- Institute of Urology, Gansu Province Clinical Research Center for Urinary System Diseases, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Wenyan Li
- Department of Gynaecology and Obstetrics, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Ying Wang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, Gansu, China
| | - Ke Wang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, Gansu, China
| | - Yan Tao
- Institute of Urology, Gansu Province Clinical Research Center for Urinary System Diseases, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Lanzhou, 730030, Gansu, China.
| | - Shanhui Liu
- Institute of Urology, Gansu Province Clinical Research Center for Urinary System Diseases, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
5
|
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
|
6
|
Zhang J, Lin F, Xu Y, Sun J, Zhang L, Chen W. Lactylation and Ischemic Stroke: Research Progress and Potential Relationship. Mol Neurobiol 2025; 62:5359-5376. [PMID: 39541071 DOI: 10.1007/s12035-024-04624-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Ischemic stroke is caused by interrupted cerebral blood flow and is a leading cause of mortality and disability worldwide. Significant advancements have been achieved in comprehending the pathophysiology of stroke and the fundamental mechanisms responsible for ischemic damage. Lactylation, as a newly discovered post-translational modification, has been reported to participate in several physiological and pathological processes. However, research on lactylation and ischemic stroke is scarce. This review summarized the current function of protein lactylation in other diseases or normal physiological processes and explored their potential link with the pathophysiological process and the reparative mechanism of ischemic stroke. We proposed that neuroinflammation, regulation of metabolism, regulation of messenger RNA translation, angiogenesis, and neurogenesis might be the bridge linking lactylation and ischemic stroke. Our study provided a novel perspective for comprehending the role of protein lactylation in the pathophysiological processes underlying ischemic stroke. Lactylation might be a promising target in drug development of ischemic stroke.
Collapse
Affiliation(s)
- Jingyuan Zhang
- Department of Cerebrovascular Disease, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Feng Lin
- Department of Cerebrovascular Disease, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Yue Xu
- Department of Cerebrovascular Disease, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Jiaxin Sun
- Department of Cerebrovascular Disease, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Lei Zhang
- Department of Cerebrovascular Disease, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong Province, China.
| | - Wenli Chen
- Department of Pharmacy, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong Province, China.
| |
Collapse
|
7
|
Sun Y, Wang H, Cui Z, Yu T, Song Y, Gao H, Tang R, Wang X, Li B, Li W, Wang Z. Lactylation in cancer progression and drug resistance. Drug Resist Updat 2025; 81:101248. [PMID: 40287994 DOI: 10.1016/j.drup.2025.101248] [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: 03/20/2025] [Revised: 04/15/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
Lactate plays a crucial role as an energy substrate, metabolite, and signaling molecule in cancer. Lactate has long been considered a byproduct of glycolysis. Still, the lactate shuttle hypothesis has changed the lactate paradigm, revealing the implications of lactate in cellular metabolism and cellular communications that can transcend the compartment barrier and occur within and between different cells, tissues, and organs. Due to the Warburg effect, the tumor produces a large amount of lactate, thus creating a low-nutrition, hypoxic, and low-pH tumor microenvironment (TME). Consequently, immunosuppressive networks are built to acquire immune evasion potential and regulate tumor growth. Lactylation is a newly discovered post-translational modification of lysine residues with the capacity for transcriptional regulation via histone modification and modulation of non-histone protein functions, which links gene regulation to cellular metabolism by aberrant metabolism activity and epigenetic modification. There is growing evidence that lactylation plays a crucial role in cancer progression and drug resistance. Targeting lactylation enzymes or metabolic pathways has shown promising effects in suppressing cancer progression and drug resistance, highlighting the therapeutic potential of this modification. Therefore, in this review, we offer a systematic overview of lactate homeostasis in physiological and pathological processes as well as discuss the influence of lactylation in cancer progression and drug resistance and underlying molecular mechanisms, providing a theoretical basis for further research.
Collapse
Affiliation(s)
- Yuxiu Sun
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - He Wang
- Department of Breast Medicine 2, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Zhe Cui
- Laboratory Department, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Tingting Yu
- Department of Gynecology Surgery 4, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Yuanming Song
- Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Haolai Gao
- First Clinical College, Liaoning University of Traditional Chinese Medicine Affiliated Hospital, Liaoning Provincial Traditional Chinese Medicine Hospital, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110847, China
| | - Ruihong Tang
- Medical Equipment Department, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Xinlei Wang
- Department of Interventional Therapy, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Binru Li
- Department of Thoracic Medicine 2, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China.
| | - Wenxin Li
- Second Ward of Hepatobiliary and Pancreatic Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China.
| | - Zhe Wang
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China.
| |
Collapse
|
8
|
Hou Y, Liu D, Guo Z, Wei C, Cao F, Xu Y, Feng Q, Liu F. Lactate and Lactylation in AKI-to-CKD: Epigenetic Regulation and Therapeutic Opportunities. Cell Prolif 2025:e70034. [PMID: 40207870 DOI: 10.1111/cpr.70034] [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: 12/27/2024] [Revised: 02/20/2025] [Accepted: 03/21/2025] [Indexed: 04/11/2025] Open
Abstract
Lactate is not only a byproduct of glycolysis, but is also considered an energy source, gluconeogenic precursor, signalling molecule and protein modifier during the process of cellular metabolism. The discovery of lactylation reveals the multifaceted functions of lactate in cellular metabolism and opens new avenues for lactate-related research. Both lactate and lactylation have been implicated in regulating numerous biological processes, including tumour progression, ischemic-hypoxic injury, neurodevelopment and immune-related inflammation. The kidney plays a crucial role in regulating lactate metabolism, influencing lactate levels while also being regulated by lactate. Previous studies have demonstrated the importance of lactate in the pathogenesis of acute kidney injury (AKI) and chronic kidney disease (CKD). This review explores the role of lactate and lactylation in these diseases, comparing the function and metabolic mechanisms of lactate in normal and diseased kidneys from the perspective of lactylation. The key regulatory roles of lactylation in different organs, multiple systems, various pathological states and underlying mechanisms in AKI-to-CKD progression are summarised. Moreover, potential therapeutic targets and future research directions for lactate and lactylation across multiple kidney diseases are identified.
Collapse
Affiliation(s)
- Yi Hou
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, Henan Province, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Dongwei Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, Henan Province, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
- Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of China, Zhengzhou, Henan Province, China
| | - Zuishuang Guo
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, Henan Province, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
- Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of China, Zhengzhou, Henan Province, China
| | - Cien Wei
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, Henan Province, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Fengyu Cao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, Henan Province, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yue Xu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, Henan Province, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Qi Feng
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, Henan Province, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
- Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of China, Zhengzhou, Henan Province, China
| | - Fengxun Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, Henan Province, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
- Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of China, Zhengzhou, Henan Province, China
| |
Collapse
|
9
|
Zhang W, Shan G, Bi G, Hu Z, Yi Y, Zeng D, Lin Z, Zhan C. Lactylation and regulated cell death. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119927. [PMID: 40023198 DOI: 10.1016/j.bbamcr.2025.119927] [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: 11/27/2024] [Revised: 02/11/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
Lactylation, a newly identified post-translational modification, entails the attachment of lactate to lysine residues within proteins, profoundly modulating diverse cellular mechanisms underlying regulated cell death (RCD). This modification encompasses two primary categories: histone lactylation and non-histone lactylation. Histone lactylation assumes a pivotal regulatory function in the RCD process, primarily by modulating the transcriptional landscape of genes implicated in cell death. In contrast, non-histone lactylation exerts its influence by targeting transferases, transcription, cell cycle progression, death pathways, and metabolic processes that are intricately involved in RCD. This review provides a comprehensive overview of recent breakthroughs in understanding how lactylation regulates RCD, while also offering insights into potential avenues for future research, thereby deepening our comprehension of cellular fate determination.
Collapse
Affiliation(s)
- Wenlong Zhang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Guangyao Shan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Zhengyang Hu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Yanjun Yi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Dejun Zeng
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Zongwu Lin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China.
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China.
| |
Collapse
|
10
|
Du Q, Meng C, Zhang W, Huang L, Xue C. Establishing a Prognostic Model Correlates to Inflammatory Response Pathways for Prostate Cancer via Multiomic Analysis of Lactylation-Related Genes. Int J Genomics 2025; 2025:6681711. [PMID: 40161494 PMCID: PMC11952923 DOI: 10.1155/ijog/6681711] [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: 12/14/2024] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Prostate cancer (PCa) continues to pose substantial clinical challenges, with molecular heterogeneity significantly impacting therapeutic decision-making and disease trajectories. Emerging evidence implicates protein lactylation-a novel epigenetic regulatory mechanism-in oncogenic processes, though its prognostic relevance in PCa remains underexplored. Through integrative bioinformatics interrogation of lactylation-associated molecular signatures, we established prognostic correlations using multivariable feature selection methodologies. Initial screening via differential expression analysis (limma package) coupled with Cox proportional hazards modeling revealed 11 survival-favorable regulators and 16 hazard-associated elements significantly linked to biochemical recurrence. To enhance predictive precision, ensemble machine learning frameworks were implemented, culminating in a 10-gene lactylation signature demonstrating robust discriminative capacity (concordance index = 0.738) across both primary (TCGA-PRAD) and external validation cohorts (DKFZ). Multivariable regression confirmed the lactylation score's prognostic independence, exhibiting prominent associations with clinicopathological parameters including tumor staging and metastatic potential. The developed clinical-molecular nomogram achieved superior predictive accuracy (C - index > 0.7) through the synergistic integration of biological and clinical covariates. Tumor microenvironment deconvolution uncovered distinct immunological landscapes, with high-risk stratification correlating with enriched stromal infiltration and immunosuppressive phenotypes. Pathway enrichment analyses implicated chromatin remodeling processes and cytokine-mediated inflammatory cascades as potential mechanistic drivers of prognostic divergence. Therapeutic vulnerability profiling demonstrated differential response patterns: low-risk patients exhibited enhanced immune checkpoint inhibitor responsiveness, whereas high-risk subgroups showed selective chemosensitivity to docetaxel and mitoxantrone. Functional validation in PC-3 models revealed AK5 silencing induced proapoptotic effects, suppressed metastatic potential of migration and invasion, and modulated immune checkpoint regulation through CD276 coexpression. These multimodal findings position lactylation dynamics, particularly AK5-mediated pathways, as promising therapeutic targets and stratification biomarkers in PCa management.
Collapse
Affiliation(s)
- Qinglong Du
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - CuiYu Meng
- The Department of EICU, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong, China
| | - Wenchao Zhang
- The Department of Urology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong, China
| | - Li Huang
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou, Guangdong, China
| | - Chunlei Xue
- The Department of Urology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong, China
| |
Collapse
|
11
|
Yi D, Zhou K, Pan Y, Cai H, Huang P. The lactylation modification of proteins plays a critical role in tumor progression. Front Oncol 2025; 15:1530567. [PMID: 40190564 PMCID: PMC11970033 DOI: 10.3389/fonc.2025.1530567] [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: 01/01/2025] [Accepted: 03/04/2025] [Indexed: 04/09/2025] Open
Abstract
Lactylation modifications have been shown to be a novel type of protein post-translational modifications (PTMs), providing a new perspective for understanding the interaction between cellular metabolic reprogramming and epigenetic regulation. Studies have shown that lactylation plays an important role in the occurrence, development, angiogenesis, invasion and metastasis of tumors. It can not only regulate the phenotypic expression and functional polarization of immune cells, but also participate in the formation of tumor drug resistance through a variety of molecular mechanisms. In this review, we review the latest research progress of lactylation modification in tumors, focusing on its mechanism of action in angiogenesis, immune cell regulation in tumor microenvironment (TME), and tumor drug resistance, aiming to provide a theoretical basis and research ideas for the discovery of new therapeutic targets and methods. Through the in-depth analysis of lactylation modification, it is expected to open up a new research direction for tumor treatment and provide potential strategies for overcoming tumor drug resistance and improving clinical efficacy.
Collapse
Affiliation(s)
- Dehao Yi
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Ke Zhou
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yinlong Pan
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Huazhong Cai
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Pan Huang
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
12
|
Wang FX, Mu G, Yu ZH, Shi ZA, Li XX, Fan X, Chen Y, Zhou J. Lactylation: a promising therapeutic target in ischemia-reperfusion injury management. Cell Death Discov 2025; 11:100. [PMID: 40082399 PMCID: PMC11906755 DOI: 10.1038/s41420-025-02381-4] [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: 03/02/2024] [Revised: 12/25/2024] [Accepted: 02/28/2025] [Indexed: 03/16/2025] Open
Abstract
Ischemia-reperfusion injury (IRI) is a critical condition that poses a significant threat to patient safety. The production of lactate increases during the process of IRI, and lactate serves as a crucial indicator for assessing the severity of such injury. Lactylation, a newly discovered post-translational modification in 2019, is induced by lactic acid and predominantly occurs on lysine residues of histone or nonhistone proteins. Extensive studies have demonstrated the pivotal role of lactylation in the pathogenesis and progression of various diseases, including melanoma, myocardial infarction, hepatocellular carcinoma, Alzheimer's disease, and nonalcoholic fatty liver disease. Additionally, a marked correlation between lactylation and inflammation has been observed. This article provides a comprehensive review of the mechanism underlying lactylation in IRI to establish a theoretical foundation for better understanding the interplay between lactylation and IRI.
Collapse
Affiliation(s)
- Fei-Xiang Wang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, China
| | - Guo Mu
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Department of Anesthesiology, Zigong Fourth People's Hospital, Zigong, Sichuan, China
| | - Zi-Hang Yu
- Department of Anesthesiology, Fushun County People's Hospital, Zigong, Sichuan, China
| | - Zu-An Shi
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, China
| | - Xue-Xin Li
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, China
| | - Xin Fan
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, China
| | - Ye Chen
- Department of Traditional Chinese Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Jun Zhou
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China.
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
13
|
Gao XD, Ding JE, Xie JX, Xu HM. Epigenetic regulation of iron metabolism and ferroptosis in Parkinson's disease: Identifying novel epigenetic targets. Acta Pharmacol Sin 2025:10.1038/s41401-025-01499-6. [PMID: 40069488 DOI: 10.1038/s41401-025-01499-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/28/2025] [Indexed: 03/17/2025]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease, and emerging evidence has shown that iron deposition, ferroptosis and epigenetic modifications are implicated in the pathogenesis of PD. However, the interplay among these factors in PD has not been fully understood. In this review, we provide an overview of the current research progress on iron metabolism, ferroptosis and epigenetic alterations associated with PD. Furthermore, we present new frontiers concerning various epigenetic modifications related to iron metabolism and ferroptosis that might contribute to the pathology of PD. Notably, epigenetic modifications of iron metabolism and ferroptosis as both diagnostic and therapeutic targets in PD have been discussed. This opens new avenues for the regulation of iron homeostasis and ferroptosis in PD from epigenetic perspectives, and provides evidence for their potential implications in the diagnosis and treatment of PD.
Collapse
Affiliation(s)
- Xiao-Die Gao
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Brain Diseases and State Key Disciplines: Physiology, Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Jian-E Ding
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Brain Diseases and State Key Disciplines: Physiology, Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Jun-Xia Xie
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China.
| | - Hua-Min Xu
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Brain Diseases and State Key Disciplines: Physiology, Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
14
|
Fang J, Wu S, Zhao H, Zhou C, Xue L, Lei Z, Li H, Shan Z. New Types of Post-Translational Modification of Proteins in Cardiovascular Diseases. J Cardiovasc Transl Res 2025:10.1007/s12265-025-10600-7. [PMID: 40032789 DOI: 10.1007/s12265-025-10600-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/18/2025] [Indexed: 03/05/2025]
Abstract
Post-translational modifications (PTMs), which are covalent alterations of proteins after their synthesis, are critical for their proper function and the maintenance of cellular physiology. The significance of PTMs in the context of cardiovascular diseases (CVDs) has been increasingly recognized due to their potential to influence protein stability, activity, and localization, thereby affecting the progression of CVDs. The identification and understanding of PTMs in CVDs at the molecular level are vital for the discovery of new biomarkers and new targets for clinical interventions. This article provides a comprehensive overview of the role and mechanisms of new types of PTMs, such as acetylation, crotonylation, succinylation, S-nitrosylation, malonylation, S-palmitonylation, β-hydroxybutyrylation and lactylation, in CVDs, highlighting their importance in advancing diagnostic and therapeutic approaches for CVDs.
Collapse
Affiliation(s)
- Juntao Fang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People'S Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China
| | - Shaoyu Wu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People'S Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China
| | - Hengli Zhao
- Medical Research Institute, Guangdong Provincial People'S Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Chuanmeng Zhou
- Medical Research Institute, Guangdong Provincial People'S Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Ling Xue
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People'S Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China
| | - Zhiyong Lei
- Department of Experimental Cardiology, University Medical Center Utrecht, 3508 GA, Utrecht, Netherlands
- CDL Research, University Medical Center Utrecht, 3508 GA, Utrecht, Netherlands
- Circulatory Health Laboratory, UMC Utrecht, Regenerative Medicine Center Utrecht, University Utrecht, 3508 GA, Utrecht, Netherlands
| | - Hui Li
- Medical Research Institute, Guangdong Provincial People'S Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Zhixin Shan
- Medical Research Institute, Guangdong Provincial People'S Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
15
|
Luo Y, Zhang N, Ye J, Wang Z, Zhou X, Liu J, Cai J, Li C, Chen L. Unveiling lactylation modification: A new hope for cancer treatment. Biomed Pharmacother 2025; 184:117934. [PMID: 39986235 DOI: 10.1016/j.biopha.2025.117934] [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: 12/29/2024] [Revised: 02/13/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025] Open
Abstract
This review article delves into the multifaceted role of lactylation modification in antitumor therapy, revealing the complex interplay between lactylation modification and the tumor microenvironment (TME), metabolic reprogramming, gene expression, and immunotherapy. As an emerging epigenetic modification, lactylation has a significant impact on the metabolic pathways of tumor cells, immune evasion, gene expression regulation, and sensitivity to chemotherapy drugs. Studies have shown that lactylation modification significantly alters the development and therapeutic response of tumors by affecting metabolites in the TME, immune cell functions, and signaling pathways. In the field of immunotherapy, the regulatory role of lactylation modification provides a new perspective and potential targets for tumor treatment, including modulating the sensitivity of tumors to immunotherapy by affecting the expression of immune checkpoint molecules and the infiltration of immune cells. Moreover, research progress on lactylation modification in various types of tumors indicates that it may serve as a biomarker to predict patients' responses to chemotherapy and immunotherapy. Overall, research on lactylation modification provides a theoretical foundation for the development of new tumor treatment strategies and holds promise for improving patient prognosis and quality of life. Future research will further explore the application potential of lactylation modification in tumor therapy and how to improve treatment efficacy by targeting lactylation modification.
Collapse
Affiliation(s)
- Yuxiang Luo
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Ning Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Jiarong Ye
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Zuao Wang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Xinchi Zhou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Jipeng Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Jing Cai
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Chen Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Institute of Orthopedics of Jiangxi Province, Nanchang, Jiangxi 330006, China; Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, Jiangxi 330006, China; Institute of Minimally Invasive Orthopedics, Nanchang University, Jiangxi 330006, China.
| | - Leifeng Chen
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Precision Oncology Medicine Center,The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, People's Republic of China.
| |
Collapse
|
16
|
Zhang M, Kong X, Wu C, Li J, Yang H, Huang L. The role of lactate and lactylation in ischemic cardiomyopathy: Mechanisms and gene expression. Exp Mol Pathol 2025; 141:104957. [PMID: 40020527 DOI: 10.1016/j.yexmp.2025.104957] [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: 05/30/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 03/03/2025]
Abstract
Ischemic cardiomyopathy (ICM) is a significant global public health issue, with its pathophysiology encompassing atherosclerotic plaque formation, thrombosis, hypoperfusion, ischemic cell death, and left ventricular remodeling. Lactate is not only regarded as an energy metabolite but also acts as a signaling molecule that influences various physiological processes, regulating metabolism and muscle contraction. Lactylation, an emerging epigenetic modification, affects protein functionality and gene expression through the P300 enzyme. In ICM, lactate accumulation leads to pH imbalance and myocardial cell dysfunction, impacting cellular signaling. This paper will analyze the role of lactylation in ICM, focusing on coronary artery disease (ASCVD) and myocardial infarction (MI). It will also explore the differential expression and immunological characteristics of lactylation-related genes in normal and ICM tissues, providing potential targets for future research.
Collapse
Affiliation(s)
- Mei Zhang
- Department of Clinical Nursing, The Second Xiangya Hospital of Central South University, Changsha, China; Xiangya School of Nursing, Central South University, Changsha, China
| | - Xue Kong
- Department of Clinical Nursing, The Second Xiangya Hospital of Central South University, Changsha, China; Xiangya School of Nursing, Central South University, Changsha, China
| | - Chenlu Wu
- Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Jiuhong Li
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Hui Yang
- Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China.
| | - Lingzhi Huang
- Department of Clinical Nursing, The Second Xiangya Hospital of Central South University, Changsha, China; Xiangya School of Nursing, Central South University, Changsha, China.
| |
Collapse
|
17
|
Li J, Jiang L, Ma Q, Zhang Z, Zheng S, Qiu J, Pang Y, Wang J. Evodiamine inhibits programmed cell death ligand 1 expression via the PI3K/AKT signaling pathway to regulate antitumor immunity in melanoma. Sci Rep 2025; 15:6649. [PMID: 39994441 PMCID: PMC11850830 DOI: 10.1038/s41598-025-91137-2] [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: 11/01/2024] [Accepted: 02/18/2025] [Indexed: 02/26/2025] Open
Abstract
Malignant melanoma, a rare and aggressive skin cancer, arises from the transformation of cutaneous melanocytes and is associated with a poor prognosis. Evodiamine (EVO), a bioactive compound derived from traditional Chinese herbal medicine, has demonstrated significant inhibitory effects on various tumor cells. In this study, we aimed to investigate the potential of EVO in regulating melanoma immunity and elucidate its underlying mechanisms. Experimental results revealed that the IC50 value of EVO in B16-F10 cells for 24, 48, and 72 h were 11.73, 5.083, and 4.604 µM, respectively. EVO inhibited the proliferation, invasion, and metastasis of B16-F10 cells by more than 50%, while promoting apoptosis of higher concentration of EVO. EVO also significantly suppressed tumor growth by more than 80% and reduced spleen damage in tumor-bearing mice. Treatment with EVO led to a marked increase in T-cell subsets in the spleen, bone marrow, and tumors, with immunohistochemical (IHC) staining in particular showing about 50% higher. Furthermore, EVO inhibited the expression of programmed cell death ligand 1 (PD-L1) and the PI3K/AKT signaling pathway-related proteins in both B16-F10 cells and tumors. These findings suggest that EVO exerts antitumor effects by enhancing the tumor immune microenvironment and indicates its potential as a therapeutic agent for melanoma.
Collapse
Affiliation(s)
- Jiamin Li
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China
| | - Li Jiang
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China
| | - Qianlong Ma
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China
| | - Zhenglong Zhang
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China
| | - Shengping Zheng
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China
| | - Jing Qiu
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China
| | - Yunqing Pang
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China.
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China.
| | - Jing Wang
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China.
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China.
| |
Collapse
|
18
|
Yang H, Zhang X, Jia Z, Wang H, Wu J, Wei X, Huang Y, Yan W, Lin Y. Targeting ferroptosis in prostate cancer management: molecular mechanisms, multidisciplinary strategies and translational perspectives. J Transl Med 2025; 23:166. [PMID: 39920771 PMCID: PMC11806579 DOI: 10.1186/s12967-025-06180-4] [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: 12/06/2024] [Accepted: 01/25/2025] [Indexed: 02/09/2025] Open
Abstract
Prostate cancer (PCa) is a kind of malignant solid tumor commonly observed among males worldwide. The dilemma of increasing incidence with therapeutic resistance has become the leading issue in PCa clinical management. Ferroptosis is a new form of regulatory cell death caused by iron-dependent lipid peroxidation, which has a dual role in PCa evolution and treatment due to the multi-omics cascade of interactions among pathways and environmental stimuli. Hence deciphering the role of ferroptosis in carcinogenesis would provide novel insights and strategies for precision medicine and personalized healthcare against PCa. In this study, the mechanisms of ferroptosis during cancer development were summarized both at the molecular and tumor microenvironment level. Then literature-reported ferroptosis-related signatures in PCa, e.g., genes, non-coding RNAs, metabolites, natural products and drug components, were manually collected and functionally compared as drivers/inducers, suppressors/inhibitors, and biomarkers according to their regulatory patterns in PCa ferroptosis and pathogenesis. The state-of-the-art techniques for ferroptosis-related data integration, knowledge identification, and translational application to PCa theranostics were discussed from a combinative perspective of artificial intelligence-powered modelling and advanced material-oriented therapeutic scheme design. The prospects and challenges in ferroptosis-based PCa researches were finally highlighted to light up future wisdoms for the flourishing of current findings from bench to bedside.
Collapse
Affiliation(s)
- Hubo Yang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Xuefeng Zhang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Zongming Jia
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - He Wang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Jixiang Wu
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Xuedong Wei
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Yuhua Huang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China.
| | - Wenying Yan
- Suzhou Key Lab of Multi-modal Data Fusion and Intelligent Healthcare, Suzhou, 215104, China.
- School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China.
| | - Yuxin Lin
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China.
- Suzhou Key Lab of Multi-modal Data Fusion and Intelligent Healthcare, Suzhou, 215104, China.
| |
Collapse
|
19
|
Chen S, Zhang X, Mo H, Peng Y, An Z, Wu J, Wei X, Zhang S, Xiong Y, Jiang W, Peng X, Zhuo L, Lei Z, Wang Z, Hu Z. Structure-activity relationship study of novel evodiamine amino acid conjugates with potent anti-colorectal cancer efficacy. Eur J Med Chem 2025; 283:117132. [PMID: 39647421 DOI: 10.1016/j.ejmech.2024.117132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 11/29/2024] [Accepted: 12/01/2024] [Indexed: 12/10/2024]
Abstract
Evodiamine has been a promising lead structure with broad-spectrum antitumor activity. Druggability optimization is the most challenging part of evodiamine-based lead-to-candidate campaign. Amino acids as building blocks for conjugates are widely incorporated into approved drug and drug candidates, demonstrating highly attractive druggability. Herein, a series of evodiamine amino acid conjugates were designed and synthesized based on the evodiamine lead compound (±)-8b discovered in our previous work. The structure-activity relationship (SAR) studies culminated in the identification of a promising conjugate (-)-15h featuring a N-Boc-l-glutamine group and a chiral carbon atom (sinister), which exhibited nanomolar antiproliferative activity against LoVo and RKO colorectal cancer cells. Moreover, (-)-15h could inhibit topoisomerases I, arrest the cell cycle in the G2/M phase, and induce apoptosis. Importantly, (-)-15h (tumor growth inhibition rate was 82.53 % in 40 mpk) showed better efficacy and tolerability to that of parent compound (-)-8b (tumor growth inhibition rate was 51.22 % in 40 mpk) in LoVo xenograft model. Further, (-)-15h (tumor growth inhibition rate was 70.09 % in 40 mpk) showed comparable efficacy and better tolerability to that of topotecan (tumor growth inhibition rate was 70.67 % in 0.5 mpk) in HT-29 xenograft model. Collectively, this study further provided a strong scientific basis for amino acid-based structural modifications and a drug lead for anti-colorectal cancer applications.
Collapse
Affiliation(s)
- Shuting Chen
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xi Zhang
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Hanxuan Mo
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Ying Peng
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zhigang An
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Junbo Wu
- Department of Colorectal Surgery, Affiliated Hengyang Hospital of Hunan Normal University & Hengyang Central Hospital, Hengyang, Hunan, 421001, China
| | - Xiuzhen Wei
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Siyi Zhang
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yongxia Xiong
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Weifan Jiang
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xue Peng
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Linsheng Zhuo
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Zhengwen Lei
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Zhen Wang
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, 810008, China; National Health Commission Key Laboratory of Birth Defect Research and Prevention Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, 410008, China; MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, China.
| | - Zecheng Hu
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
20
|
Liao Z, Chen B, Yang T, Zhang W, Mei Z. Lactylation modification in cardio-cerebral diseases: A state-of-the-art review. Ageing Res Rev 2025; 104:102631. [PMID: 39647583 DOI: 10.1016/j.arr.2024.102631] [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: 09/15/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 12/10/2024]
Abstract
Cardio-cerebral diseases (CCDs), encompassing conditions such as coronary heart disease, myocardial infarction, stroke, Alzheimer's disease, et al., represent a significant threat to human health and well-being. These diseases are often characterized by metabolic abnormalities and remodeling in the process of pathology. Glycolysis and hypoxia-induced lactate accumulation play critical roles in cellular energy dynamics and metabolic imbalances in CCDs. Lactylation, a post-translational modification driven by excessive lactate accumulation, occurs in both histone and non-histone proteins. It has been implicated in regulating protein function across various pathological processes in CCDs, including inflammation, angiogenesis, lipid metabolism dysregulation, and fibrosis. Targeting key proteins involved in lactylation, as well as the enzymes regulating this modification, holds promise as a therapeutic strategy to modulate disease progression by addressing these pathological mechanisms. This review provides a holistic picture of the types of lactylation and the associated modifying enzymes, highlights the roles of lactylation in different pathological processes, and synthesizes the latest clinical evidence and preclinical studies in a comprehensive view. We aim to emphasize the potential of lactylation as an innovative therapeutic target for preventing and treating CCD-related conditions.
Collapse
Affiliation(s)
- Zi Liao
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Bei Chen
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Tong Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Wenli Zhang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China.
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China.
| |
Collapse
|
21
|
Niu K, Chen Z, Li M, Ma G, Deng Y, Zhang J, Wei D, Wang J, Zhao Y. NSUN2 lactylation drives cancer cell resistance to ferroptosis through enhancing GCLC-dependent glutathione synthesis. Redox Biol 2025; 79:103479. [PMID: 39742570 PMCID: PMC11750563 DOI: 10.1016/j.redox.2024.103479] [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: 10/12/2024] [Revised: 11/29/2024] [Accepted: 12/19/2024] [Indexed: 01/03/2025] Open
Abstract
Lactate-mediated lactylation on target proteins is recently identified as the novel posttranslational modification with profound biological functions. RNA 5-methylcytosine (m5C) modification possesses dynamic and reversible nature, suggesting that activity of its methyltransferase NSUN2 is actively regulated. However, how NSUN2 activity is response to acidic condition in tumor microenvironment and then regulates cancer cell survival remain to be clarified. Here, we demonstrate that NSUN2 activity is enhanced by lactate-mediated lactylation at lysine 508, which then targets glutamate-cysteine ligase catalytic subunit (GCLC) mRNA to facilitates GCLC m5C formation and mRNA stabilization. The activated GCLC induces higher level of intracellular GSH accompanied by decreased lipid peroxidation and resistant phenotype to ferroptosis induction by doxorubicin (Dox) in gastric cancer cells. Specifically, the effect of NSUN2 lactylation-GCLC-GSH pathway is nearly lost when NSUN2 K508R or GCLC C-A mutant (five cytosine sites) was introduced into the cancer cells. We further identify the catalytic subunit N-α-acetyltransferase 10 (NAA10) as the lactytransferase of NSUN2, and lactate treatment substantially enhances their association and consequent NSUN2 activation. Taken together, our findings convincingly elucidate the signaling axis of NAA10-NSUN2-GCLC that potently antagonizes the ferroptosis under acidic condition, and therefore, targeting NSUN2 lactylation might be an effective strategy in improving the prognosis of cancer patients.
Collapse
Affiliation(s)
- Kaifeng Niu
- China National Center for Bioinformation, Beijing, 100101, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Zixiang Chen
- China National Center for Bioinformation, Beijing, 100101, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Mengge Li
- China National Center for Bioinformation, Beijing, 100101, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guannan Ma
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, 310030, China
| | - Yuchun Deng
- China National Center for Bioinformation, Beijing, 100101, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ji Zhang
- China National Center for Bioinformation, Beijing, 100101, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Di Wei
- China National Center for Bioinformation, Beijing, 100101, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jiaqi Wang
- China National Center for Bioinformation, Beijing, 100101, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yongliang Zhao
- China National Center for Bioinformation, Beijing, 100101, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
22
|
Jing F, Zhang J, Zhang H, Li T. Unlocking the multifaceted molecular functions and diverse disease implications of lactylation. Biol Rev Camb Philos Soc 2025; 100:172-189. [PMID: 39279350 DOI: 10.1111/brv.13135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/18/2024]
Abstract
In recent years, a significant breakthrough has emerged in biology, the identification of lactylation, a novel post-translational process. This intriguing modification is not limited to a specific class of proteins but occurs across a diverse range, including histones, signalling molecules, enzymes, and substrates. It can exert a broad regulatory role in various diseases, ranging from developmental anomalies and neurodegenerative disorders to inflammation and cancer. Thus, it presents exciting opportunities for exploring innovative treatment approaches. As a result, there has been a recent surge of research interest, leading to a deeper understanding of the molecular mechanisms and regulatory functions underlying lactylation within physiological and pathological processes. Here, we review the detection and molecular mechanisms of lactylation, from biological functions to disease effects, providing a systematic overview of the mechanisms and functions of this post-translational modification.
Collapse
Affiliation(s)
- Fengyang Jing
- Department of Oral Pathology, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
- Research Unit of Precision Pathologic Diagnosis in Tumors of the Oral and Maxillofacial Regions, Chinese Academy of Medical Sciences (2019RU034), No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
| | - Jianyun Zhang
- Department of Oral Pathology, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
- Research Unit of Precision Pathologic Diagnosis in Tumors of the Oral and Maxillofacial Regions, Chinese Academy of Medical Sciences (2019RU034), No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
| | - Heyu Zhang
- Research Unit of Precision Pathologic Diagnosis in Tumors of the Oral and Maxillofacial Regions, Chinese Academy of Medical Sciences (2019RU034), No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
- Central Laboratory, Peking University School and Hospital of Stomatology, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
| | - Tiejun Li
- Department of Oral Pathology, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
- Research Unit of Precision Pathologic Diagnosis in Tumors of the Oral and Maxillofacial Regions, Chinese Academy of Medical Sciences (2019RU034), No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
| |
Collapse
|
23
|
Yu D, Zhong Q, Wang Y, Yin C, Bai M, Zhu J, Chen J, Li H, Hong W. Lactylation: The metabolic accomplice shaping cancer's response to radiotherapy and immunotherapy. Ageing Res Rev 2025; 104:102670. [PMID: 39864560 DOI: 10.1016/j.arr.2025.102670] [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: 11/19/2024] [Revised: 01/09/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
Protein lactylation, an emerging post-translational modification, is providing new insights into tumor biology and challenging our current understanding of cancer mechanisms. Our review illuminates the intricate roles of lactylation in carcinogenesis, tumor progression, and therapeutic responses, positioning it as a critical linchpin connecting metabolic reprogramming, epigenetic modulation, and treatment outcomes. We provide an in-depth analysis of lactylation's molecular mechanisms and its far-reaching impact on cell cycle regulation, immune evasion strategies, and therapeutic resistance within the complex tumor microenvironment. Notably, this review dissects the paradoxical nature of lactylation in cancer immunotherapy and radiotherapy. While heightened lactylation can foster immune suppression and radioresistance, strategically targeting lactylation cascades opens innovative avenues for amplifying the efficacy of current treatment paradigms. We critically evaluate lactylation's potential as a robust diagnostic and prognostic biomarker and explore frontier therapeutic approaches targeting lactylation. The synergistic integration of multi-omics data and artificial intelligence in lactylation research is catalyzing significant strides towards personalized cancer management. This review not only consolidates current knowledge but also charts a course for future investigations. Key research imperatives include deciphering tumor-specific lactylation signatures, optimizing synergistic strategies combining lactylation modulation with immune checkpoint inhibitors and radiotherapy, and comprehensively assessing the long-term physiological implications of lactylation intervention. As our understanding of lactylation's pivotal role in tumor biology continues to evolve, this burgeoning field promises to usher in transformative advancements in cancer diagnosis, treatment modalitie.
Collapse
Affiliation(s)
- Danqing Yu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Qingping Zhong
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yanlin Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Chang Yin
- Nursing Department, Shanghai Sixth People's Hospital, Shanghai 200233, China
| | - Minghua Bai
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Ji Zhu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Jinggang Chen
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Huaming Li
- Department of Gastroenterology, Hangzhou Third Peoples Hospital, Hangzhou 310000, China.
| | - Weifeng Hong
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
24
|
Hua M, Li T. Multiomic machine learning on lactylation for molecular typing and prognosis of lung adenocarcinoma. Sci Rep 2025; 15:3075. [PMID: 39856156 PMCID: PMC11760357 DOI: 10.1038/s41598-025-87419-4] [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: 08/15/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
To integrate machine learning and multiomic data on lactylation-related genes (LRGs) for molecular typing and prognosis prediction in lung adenocarcinoma (LUAD). LRG mRNA and long non-coding RNA transcriptomes, epigenetic methylation data, and somatic mutation data from The Cancer Genome Atlas LUAD cohort were analyzed to identify lactylation cancer subtypes (CSs) using 10 multiomics ensemble clustering techniques. The findings were then validated using the GSE31210 and GSE13213 LUAD cohorts. A prognosis model for LUAD was developed using the identified hub LRGs to divide patients into high- and low-risk groups. The effectiveness of this model was validated. We identified two lactylation CSs, which were validated in the GSE31210 and GSE13213 LUAD cohorts. Nine hub LRGs, namely HNRNPC, PPIA, BZW1, GAPDH, H2AFZ, RAN, KIF2C, RACGAP1, and WBP11, were used to construct the prognosis model. In the subsequent prognosis validation, the high-risk group included more patients with stage T3 + 4, N1 + 2 + 3, M1, and III + IV cancer; higher recurrence/metastasis rates; and lower 1, 3, and 5 year overall survival rates. In the oncogenic pathway analysis, most of the oncogenic mutations were detected in the high-risk group. The tumor microenvironment analysis illustrated that immune activity was notably elevated in low-risk patients, indicating they might more strongly respond to immunotherapy than high-risk patients. Further, oncoPredict analysis revealed that low-risk patients have increased sensitivity to chemotherapeutics. Overall, we developed a model that combines multiomic analysis and machine learning for LUAD prognosis. Our findings represent a valuable reference for further understanding the important function of lactylation modification pathways in LUAD progression.
Collapse
Affiliation(s)
- Mengmeng Hua
- Department of Oral and Maxillofacial Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Tao Li
- Department of Respiratory Diseases, Qilu Hospital of Shandong University, No. 107, Culture West Road, Jinan, Shandong, China.
| |
Collapse
|
25
|
Wang W, Wang H, Wang Q, Yu X, Ouyang L. Lactate-induced protein lactylation in cancer: functions, biomarkers and immunotherapy strategies. Front Immunol 2025; 15:1513047. [PMID: 39867891 PMCID: PMC11757118 DOI: 10.3389/fimmu.2024.1513047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/27/2024] [Indexed: 01/28/2025] Open
Abstract
Lactate, long viewed as a byproduct of glycolysis and metabolic waste. Initially identified within the context of yogurt fermentation, lactate's role extends beyond culinary applications to its significance in biochemical processes. Contemporary research reveals that lactate functions not merely as the terminal product of glycolysis but also as a nexus for initiating physiological and pathological responses within the body. Lysine lactylation (Kla), a novel post-translational modification (PTM) of proteins, has emerged as a pivotal mechanism by which lactate exerts its regulatory influence. This epigenetic modification has the potential to alter gene expression patterns, thereby impacting physiological and pathological processes. Increasing evidence indicates a correlation between lactylation and adverse prognosis in various malignancies. Consequently, this review article aims to encapsulate the proteins that interact with lactate, elucidate the role of lactylation in tumorigenesis and progression, and explore the potential therapeutic targets afforded by the modulation of lactylation. The objective of this review is to clarify the oncogenic significance of lactylation and to provide a strategic framework for future research directions in this burgeoning field.
Collapse
Affiliation(s)
- Wenjuan Wang
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
| | - Hong Wang
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
| | - Qi Wang
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Life Sciences, Nanjing Forestry University, Nanjing, China
| | - Xiaojing Yu
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
| | - Liangliang Ouyang
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
| |
Collapse
|
26
|
Bao C, Ma Q, Ying X, Wang F, Hou Y, Wang D, Zhu L, Huang J, He C. Histone lactylation in macrophage biology and disease: from plasticity regulation to therapeutic implications. EBioMedicine 2025; 111:105502. [PMID: 39662177 PMCID: PMC11697715 DOI: 10.1016/j.ebiom.2024.105502] [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: 07/30/2024] [Revised: 11/10/2024] [Accepted: 12/02/2024] [Indexed: 12/13/2024] Open
Abstract
Epigenetic modifications have been identified as critical molecular determinants influencing macrophage plasticity and heterogeneity. Among these, histone lactylation is a recently discovered epigenetic modification. Research examining the effects of histone lactylation on macrophage activation and polarization has grown substantially in recent years. Evidence increasingly suggests that lactate-mediated changes in histone lactylation levels within macrophages can modulate gene transcription, thereby contributing to the pathogenesis of various diseases. This review provides a comprehensive analysis of the role of histone lactylation in macrophage activation, exploring its discovery, effects, and association with macrophage diversity and phenotypic variability. Moreover, it highlights the impact of alterations in macrophage histone lactylation in diverse pathological contexts, such as inflammation, tumorigenesis, neurological disorders, and other complex conditions, and demonstrates the therapeutic potential of drugs targeting these epigenetic modifications. This mechanistic understanding provides insights into the underlying disease mechanisms and opens new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Chuncha Bao
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, PR China
| | - Qing Ma
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xihong Ying
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Fengsheng Wang
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, PR China
| | - Yue Hou
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, PR China
| | - Dun Wang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, PR China
| | - Linsen Zhu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, PR China
| | - Jiapeng Huang
- Clinical Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, PR China.
| | - Chengqi He
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, PR China.
| |
Collapse
|
27
|
Liu X, Shen Q, Cheng L, Dai K, Wu Q, Liu X, Yao P, Zeng L. Synergistic inhibitory effects of tetramethylpyrazine and evodiamine on endometriosis development. J Steroid Biochem Mol Biol 2025; 245:106630. [PMID: 39486648 DOI: 10.1016/j.jsbmb.2024.106630] [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: 06/20/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Endometriosis (EMS) belongs to a gynecological disorder with inflammation and the existence of endometrial-like tissues beyond the uterus, often leading to infertility and pelvic pain. Estrogen receptor β (ERβ) is significantly expressed in endometriosis (EMS) and recognized as a promising therapeutic target for EMS treatment by inhibiting ERβ activity. In this study, we investigated the potential mechanisms for tetramethylpyrazine (TMP)-mediated ERβ suppression, and the synergistic inhibitory effect of TMP and evodiamine (EVO) on ERβ expression and EMS development. We found that TMP suppresses ERβ expression by reducing the association of Oct3/4 with the ERβ promoter and decreasing Oct3/4 protein levels without affecting Oct3/4 transcript levels. A minimum dosage of 10 µM TMP is required to inhibit ERβ expression. Neither TMP (5 µM) nor EVO (2 µM) alone had any effect, but their combination synergistically inhibited ERβ expression and modulated related cellular processes, including redox balance, mitochondrial function, inflammation, and proliferation. Additionally, the combination of TMP (10 mg/kg body weight) and EVO (5 mg/kg) synergistically inhibited ERβ expression and EMS development in the mouse model. In conclusion, TMP suppresses ERβ expression by reducing the association of Oct3/4 with the ERβ promoter. Neither TMP nor EVO alone effectively suppresses ERβ in both laboratory and live organism models. However, their combination synergistically inhibits ERβ expression and EMS development, suggesting a potential therapeutic strategy for EMS using TMP and EVO.
Collapse
Affiliation(s)
- Xiaohan Liu
- Department of gynecology, Sun Yat-Sen University Affiliated, No.8 Hospital, Shenzhen 518033, PR China
| | - Qingjun Shen
- Department of gynecology, Sun Yat-Sen University Affiliated, No.8 Hospital, Shenzhen 518033, PR China
| | - Liqin Cheng
- Department of gynecology, Sun Yat-Sen University Affiliated, No.8 Hospital, Shenzhen 518033, PR China
| | - Kailing Dai
- Department of gynecology, Sun Yat-Sen University Affiliated, No.8 Hospital, Shenzhen 518033, PR China
| | - Qiaozhu Wu
- Department of gynecology, Sun Yat-Sen University Affiliated, No.8 Hospital, Shenzhen 518033, PR China
| | - Xiaole Liu
- Department of gynecology, Sun Yat-Sen University Affiliated, No.8 Hospital, Shenzhen 518033, PR China
| | - Paul Yao
- Department of gynecology, Sun Yat-Sen University Affiliated, No.8 Hospital, Shenzhen 518033, PR China.
| | - Liqin Zeng
- Department of gynecology, Sun Yat-Sen University Affiliated, No.8 Hospital, Shenzhen 518033, PR China.
| |
Collapse
|
28
|
Yang Y, Shi J, Yu J, Zhao X, Zhu K, Wang S, Zhang X, Zhang X, Wei G, Cao W. New Posttranslational Modification Lactylation Brings New Inspiration for the Treatment of Rheumatoid Arthritis. J Inflamm Res 2024; 17:11845-11860. [PMID: 39758940 PMCID: PMC11697653 DOI: 10.2147/jir.s497240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 12/21/2024] [Indexed: 01/07/2025] Open
Abstract
Lactic acid (LA) is an essential glycolytic metabolite and energy source in the body, which is present in high levels in the synovial fluid of patients with rheumatoid arthritis (RA) and is a reliable indicator for identifying inflammatory arthritis. LA not only acts as an inflammatory amplifier in RA, recent studies have found that novel posttranslational modification (PTM) lactylation mediated by LA may also play a key role in RA. Single-cell sequencing showed that the RA lactylation score of patients with RA was significantly increased, and core lactylation-promoting genes, including NDUFB3, NGLY1, and other genes, were found to be potential biomarkers of RA. More studies have shown that lactylation can regulate genes in various cells, such as fibroblast-like synoviocytes (FLSs) and macrophages, thus playing a special role in the development and occurrence of autoimmune diseases, neurological diseases, and cancer diseases. In this paper, we review the research on lactylation in RA-related cells and mechanisms and bring new insights into the pathogenesis, diagnosis, and treatment of RA.
Collapse
Affiliation(s)
- Yue Yang
- Rheumatology Department, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Jinjie Shi
- Graduate School, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Jiming Yu
- The Second Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Xin Zhao
- Rheumatology Department, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Ke Zhu
- Rheumatology Department, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Shen Wang
- Orthopedics Department, Nanjing Integrated Traditional Chinese and Western Medicine Hospital, Nanjing, People’s Republic of China
| | - Xinwen Zhang
- Rheumatology Department, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Xieyu Zhang
- Rheumatology Department, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Guangcheng Wei
- Rheumatology Department, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Wei Cao
- Rheumatology Department, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
29
|
Hao ZN, Tan XP, Zhang Q, Li J, Xia R, Ma Z. Lactate and Lactylation: Dual Regulators of T-Cell-Mediated Tumor Immunity and Immunotherapy. Biomolecules 2024; 14:1646. [PMID: 39766353 PMCID: PMC11674224 DOI: 10.3390/biom14121646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/14/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Lactate and its derivative, lactylation, play pivotal roles in modulating immune responses within the tumor microenvironment (TME), particularly in T-cell-mediated cancer immunotherapy. Elevated lactate levels, a hallmark of the Warburg effect, contribute to immune suppression through CD8+ T cell functionality and by promoting regulatory T cell (Treg) activity. Lactylation, a post-translational modification (PTM), alters histone and non-histone proteins, influencing gene expression and further reinforcing immune suppression. In the complex TME, lactate and its derivative, lactylation, are not only associated with immune suppression but can also, under certain conditions, exert immunostimulatory effects that enhance cytotoxic responses. This review describes the dual roles of lactate and lactylation in T-cell-mediated tumor immunity, analyzing how these factors contribute to immune evasion, therapeutic resistance, and immune activation. Furthermore, the article highlights emerging therapeutic strategies aimed at inhibiting lactate production or disrupting lactylation pathways to achieve a balanced regulation of these dual effects. These strategies offer new insights into overcoming tumor-induced immune suppression and hold the potential to improve the efficacy of cancer immunotherapies.
Collapse
Affiliation(s)
- Zhi-Nan Hao
- Department of Gastroenterology, First Affiliated Hospital of Yangtze University, Health Science Center, Yangtze University, Jingzhou 434023, China; (Z.-N.H.); (Q.Z.); (J.L.)
- Digestive Disease Research Institution of Yangtze University, Yangtze University, Jingzhou 434023, China;
| | - Xiao-Ping Tan
- Digestive Disease Research Institution of Yangtze University, Yangtze University, Jingzhou 434023, China;
- The Third Clinical Medical College of Yangtze University, Jingzhou Hospital of Traditional Chinese Medicine, Jingzhou 434023, China
| | - Qing Zhang
- Department of Gastroenterology, First Affiliated Hospital of Yangtze University, Health Science Center, Yangtze University, Jingzhou 434023, China; (Z.-N.H.); (Q.Z.); (J.L.)
- Digestive Disease Research Institution of Yangtze University, Yangtze University, Jingzhou 434023, China;
| | - Jie Li
- Department of Gastroenterology, First Affiliated Hospital of Yangtze University, Health Science Center, Yangtze University, Jingzhou 434023, China; (Z.-N.H.); (Q.Z.); (J.L.)
- Digestive Disease Research Institution of Yangtze University, Yangtze University, Jingzhou 434023, China;
| | - Ruohan Xia
- Department of Gastroenterology, First Affiliated Hospital of Yangtze University, Health Science Center, Yangtze University, Jingzhou 434023, China; (Z.-N.H.); (Q.Z.); (J.L.)
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Zhaowu Ma
- Department of Gastroenterology, First Affiliated Hospital of Yangtze University, Health Science Center, Yangtze University, Jingzhou 434023, China; (Z.-N.H.); (Q.Z.); (J.L.)
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| |
Collapse
|
30
|
Godiyal Y, Maheshwari D, Taniguchi H, Zinzuwadia SS, Morera-Díaz Y, Tewari D, Bishayee A. Role of PD-1/PD-L1 signaling axis in oncogenesis and its targeting by bioactive natural compounds for cancer immunotherapy. Mil Med Res 2024; 11:82. [PMID: 39690423 DOI: 10.1186/s40779-024-00586-9] [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] [Accepted: 11/29/2024] [Indexed: 12/19/2024] Open
Abstract
Cancer is a global health problem and one of the leading causes of mortality. Immune checkpoint inhibitors have revolutionized the field of oncology, emerging as a powerful treatment strategy. A key pathway that has garnered considerable attention is programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1). The interaction between PD-L1 expressed on tumor cells and PD-1 reduces the innate immune response and thus compromises the capability of the body's immune system. Furthermore, it controls the phenotype and functionality of innate and adaptive immune components. A range of monoclonal antibodies, including avelumab, atezolizumab, camrelizumab, dostarlimab, durvalumab, sinitilimab, toripalimab, and zimberelimab, have been developed for targeting the interaction between PD-1 and PD-L1. These agents can induce a broad spectrum of autoimmune-like complications that may affect any organ system. Recent studies have focused on the effect of various natural compounds that inhibit immune checkpoints. This could contribute to the existing arsenal of anticancer drugs. Several bioactive natural agents have been shown to affect the PD-1/PD-L1 signaling axis, promoting tumor cell apoptosis, influencing cell proliferation, and eventually leading to tumor cell death and inhibiting cancer progression. However, there is a substantial knowledge gap regarding the role of different natural compounds targeting PD-1 in the context of cancer. Hence, this review aims to provide a common connection between PD-1/PD-L1 blockade and the anticancer effects of distinct natural molecules. Moreover, the primary focus will be on the underlying mechanism of action as well as the clinical efficacy of bioactive molecules. Current challenges along with the scope of future research directions targeting PD-1/PD-L1 interactions through natural substances are also discussed.
Collapse
Affiliation(s)
- Yogesh Godiyal
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Drishti Maheshwari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Hiroaki Taniguchi
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, 05-552, Magdalenka, Poland
- African Genome Center, Mohammed VI Polytechnic University, Hay Moulay Rachid, 43150, Ben Guerir, Morocco
| | - Shweta S Zinzuwadia
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA
| | - Yanelys Morera-Díaz
- Clinical Investigation and Biomedical Research Directions, Center for Genetic Engineering and Biotechnology, 11600, Havana, Cuba
| | - Devesh Tewari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India.
| | - Anupam Bishayee
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA.
| |
Collapse
|
31
|
Fernández-Nogueira P, Linzoain-Agos P, Cueto-Remacha M, De la Guia-Lopez I, Recalde-Percaz L, Parcerisas A, Gascon P, Carbó N, Gutierrez-Uzquiza A, Fuster G, Bragado P. Role of semaphorins, neuropilins and plexins in cancer progression. Cancer Lett 2024; 606:217308. [PMID: 39490515 DOI: 10.1016/j.canlet.2024.217308] [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: 08/02/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Progress in understanding nervous system-cancer interconnections has emphasized the functional role of semaphorins (SEMAs) and their receptors, neuropilins (NRPs) and plexins (PLXNs), in cancer progression. SEMAs are a conserved and extensive family of broadly expressed soluble and membrane-associated proteins that were first described as regulators of axon guidance and neural and vascular development. However, recent advances have shown that they can have a dual role in cancer progression, acting either as tumor promoters or suppressors. SEMAs effects result from their interaction with specific co-receptors/receptors NRPs/PLXNs, that have also been described to play a role in cancer progression. They can influence both cancer cells and tumor microenvironment components modulating various aspects of tumorigenesis such as oncogenesis, tumor growth, invasion and metastatic spread or treatment resistance. In this review we focus on the role of these axon guidance signals and their receptors and co-receptors in various aspects of cancer. Furthermore, we also highlight their potential application as novel approaches for cancer treatment in the future.
Collapse
Affiliation(s)
- P Fernández-Nogueira
- Department of Biomedicine, School of Medicine, Universitat de Barcelona, 08028, Barcelona, Spain; Biosciences Department, Faculty of Sciences, Technology and Engineering, University of Vic. Central University of Catalonia (UVic-UCC), 08500, Vic, Catalonia, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Institute of Biomedicine of the Universitat de Barcelona (IBUB), 08028, Barcelona, Spain
| | - P Linzoain-Agos
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040, Madrid, Spain; Health Research Institute of the Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - M Cueto-Remacha
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040, Madrid, Spain; Health Research Institute of the Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - I De la Guia-Lopez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040, Madrid, Spain; Health Research Institute of the Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - L Recalde-Percaz
- Department of Biomedicine, School of Medicine, Universitat de Barcelona, 08028, Barcelona, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Institute of Biomedicine of the Universitat de Barcelona (IBUB), 08028, Barcelona, Spain
| | - A Parcerisas
- Biosciences Department, Faculty of Sciences, Technology and Engineering, University of Vic. Central University of Catalonia (UVic-UCC), 08500, Vic, Catalonia, Spain; Tissue Repair and Regeneration Laboratory (TR2Lab), Institute of Research and Innovation of Life Sciences and Health, Catalunya Central (IRIS-CC), 08500, Vic, Catalonia, Spain
| | - P Gascon
- Department of Biomedicine, School of Medicine, Universitat de Barcelona, 08028, Barcelona, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Institute of Biomedicine of the Universitat de Barcelona (IBUB), 08028, Barcelona, Spain
| | - N Carbó
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Institute of Biomedicine of the Universitat de Barcelona (IBUB), 08028, Barcelona, Spain
| | - A Gutierrez-Uzquiza
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040, Madrid, Spain; Health Research Institute of the Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - G Fuster
- Department of Biomedicine, School of Medicine, Universitat de Barcelona, 08028, Barcelona, Spain; Biosciences Department, Faculty of Sciences, Technology and Engineering, University of Vic. Central University of Catalonia (UVic-UCC), 08500, Vic, Catalonia, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Institute of Biomedicine of the Universitat de Barcelona (IBUB), 08028, Barcelona, Spain; Tissue Repair and Regeneration Laboratory (TR2Lab), Institute of Research and Innovation of Life Sciences and Health, Catalunya Central (IRIS-CC), 08500, Vic, Catalonia, Spain.
| | - P Bragado
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040, Madrid, Spain; Health Research Institute of the Hospital Clínico San Carlos, 28040, Madrid, Spain.
| |
Collapse
|
32
|
He Y, Xiang L, Yuan J, Yan H. Lactylation Modification as a Promoter of Bladder Cancer: Insights from Multi-Omics Analysis. Curr Issues Mol Biol 2024; 46:12866-12885. [PMID: 39590360 PMCID: PMC11593262 DOI: 10.3390/cimb46110766] [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: 10/03/2024] [Revised: 11/06/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024] Open
Abstract
Bladder cancer (BLAC) is a malignant tumor with high morbidity and mortality. The establishment of a prognostic model for BLAC is of great significance for clinical prognosis prediction and treatment guidance. Lactylation modification is a newly discovered post-transcriptional modification of proteins, which is closely related to the occurrence and development of tumors. Multiple omics data of BLAC were obtained from the GEO database and TCGA database. The Lasso algorithm was used to establish a prognostic model related to lactylation modification, and its predictive ability was tested with a validation cohort. Functional enrichment analysis, tumor microenvironment analysis, and treatment response evaluation were performed on the high- and low-risk groups. Single-cell and spatial transcriptome data were used to analyze the distribution characteristics of model genes and their changes during epithelial carcinogenesis. A prognostic model consisting of 12 genes was constructed. The survival rate of the high-risk group was significantly lower than that of the low-risk group. The multiple ROC curve showed that the prediction efficiency of the model was higher than that of the traditional clinical tumor grading. Functional enrichment analysis showed that glycolysis and hypoxia pathways were significantly upregulated in the high-risk group. The high-risk group was more sensitive to most first-line chemotherapy drugs, while the low-risk group had a better response to immunotherapy. Single-cell sequencing analysis revealed the dynamic changes of model genes during the transition of epithelial cells to squamous-differentiated cells. Spatial transcriptome analysis showed the spatial distribution characteristics of the model genes. The lactylation-related models have a satisfactory predictive ability and the potential to guide the clinical treatment of BLAC. This model has significant biological implications at the single-cell level as well as at the spatial level.
Collapse
Affiliation(s)
- Yipeng He
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (Y.H.); (L.X.); (J.Y.)
- The First Clinical College, Wuhan University, Wuhan 430060, China
| | - Lingyan Xiang
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (Y.H.); (L.X.); (J.Y.)
| | - Jingping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (Y.H.); (L.X.); (J.Y.)
| | - Honglin Yan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (Y.H.); (L.X.); (J.Y.)
| |
Collapse
|
33
|
Hu XT, Wu XF, Xu JY, Xu X. Lactate-mediated lactylation in human health and diseases: Progress and remaining challenges. J Adv Res 2024:S2090-1232(24)00529-0. [PMID: 39522689 DOI: 10.1016/j.jare.2024.11.010] [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: 07/31/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Lactate was once considered as metabolic waste for a long time. In 2019, Professor Zhao Yingming's team from the University of Chicago found that lactate could also be used as a substrate to induce histone lactylation and regulate gene expression. Since then, researchers have discovered that lactate-mediated lactylation play important regulatory roles in various physiological and pathological processes. AIM OF REVIEW In this review, we aim to discuss the roles and mechanisms of lactylation in human health and diseases, as well as the effects of lactylation on proteins and metabolic modulators targeting lactylation. KEY SCIENTIFIC CONCEPTS OF REVIEW In this work, we emphasize the crucial regulatory roles of lactylation in the development of numerous physiological and pathological processes. Of relevance, we discuss the current issues and challenges pertaining to lactylation. This review provides directions and a theoretical basis for future research and clinical translation of lactylation.
Collapse
Affiliation(s)
- Xue-Ting Hu
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Xiao-Feng Wu
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jin-Yi Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| | - Xiang Xu
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
34
|
Azevedo T, Ferreira T, Peña‐Corona SI, Cortes H, Silva‐Reis R, da Costa RMG, Faustino‐Rocha AI, Oliveira PA, Calina D, Cardoso SM, Büsselberg D, Leyva‐Gómez G, Sharifi‐Rad J, Cho WC. Natural products‐based antiangiogenic agents: New frontiers in cancer therapy. FOOD FRONTIERS 2024; 5:2423-2466. [DOI: 10.1002/fft2.466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
AbstractAngiogenesis, vital for tumor growth and metastasis, is a promising target in cancer therapy. Natural compounds offer potential as antiangiogenic agents with reduced toxicity. This review provides a comprehensive overview of natural product‐based antiangiogenic therapies, focusing on molecular mechanisms and therapeutic potential. A systematic search identified relevant articles from 2019 to 2023. Various natural compounds, including polyphenols, terpenes, alkaloids, cannabinoids, omega‐3 fatty acids, polysaccharides, proteins, and carotenoids, were investigated for their antiangiogenic properties. Challenges such as dose standardization, routes of administration, and potential side effects remain. Further studies, including in‐depth animal models and human epidemiological studies, must elucidate clinical efficacy and safety. Synergistic effects with current antiangiogenic therapies, such as bevacizumab and tyrosine kinase inhibitors, should be explored. Additionally, the potential hormone‐dependent effects of compounds like genistein highlight the need for safety evaluation. In conclusion, natural products hold promise as adjunctive therapies to conventional antineoplastic drugs in modulating angiogenesis in cancer. However, robust clinical trials are needed to validate preclinical findings and ensure safety and efficacy.
Collapse
Affiliation(s)
- Tiago Azevedo
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
| | - Tiago Ferreira
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
| | - Sheila I. Peña‐Corona
- Departamento de Farmacia, Facultad de Química Universidad Nacional Autónoma de México Ciudad de México Mexico
| | - Hernán Cortes
- Laboratorio de Medicina Genómica, Departamento de Genómica Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra Ciudad de México Mexico
| | - Rita Silva‐Reis
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
- LAQV‐REQUIMTE, Department of Chemistry University of Aveiro Aveiro Portugal
| | - Rui M. Gil da Costa
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI‐IPOP)/RISE@CI‐IPOP (Health Research Network) Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center (Porto. CCC) Porto Portugal
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering University of Porto Porto Portugal
- Associate Laboratory in Chemical Engineering (ALiCE), Faculty of Engineering University of Porto Porto Portugal
- Postgraduate Programme in Adult Health (PPGSAD), Department of Morphology Federal University of Maranhão (UFMA), UFMA University Hospital (HUUFMA) São Luís Brazil
| | - Ana I. Faustino‐Rocha
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
- Comprehensive Health Research Center, Department of Zootechnics, School of Sciences and Technology University of Évora Evora Portugal
| | - Paula A. Oliveira
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
| | - Daniela Calina
- Department of Clinical Pharmacy University of Medicine and Pharmacy of Craiova Craiova Romania
| | - Susana M. Cardoso
- LAQV‐REQUIMTE, Department of Chemistry University of Aveiro Aveiro Portugal
| | | | - Gerardo Leyva‐Gómez
- Departamento de Farmacia, Facultad de Química Universidad Nacional Autónoma de México Ciudad de México Mexico
| | - Javad Sharifi‐Rad
- Centro de Estudios Tecnológicos y Universitarios del Golfo Veracruz Mexico
- Department of Medicine, College of Medicine Korea University Seoul Republic of Korea
- Facultad de Medicina Universidad del Azuay Cuenca Ecuador
| | - William C. Cho
- Department of Clinical Oncology Queen Elizabeth Hospital Kowloon Hong Kong
| |
Collapse
|
35
|
He Y, Song T, Ning J, Wang Z, Yin Z, Jiang P, Yuan Q, Yu W, Cheng F. Lactylation in cancer: Mechanisms in tumour biology and therapeutic potentials. Clin Transl Med 2024; 14:e70070. [PMID: 39456119 PMCID: PMC11511673 DOI: 10.1002/ctm2.70070] [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: 08/06/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Lactylation, a recently identified form of protein post-translational modification (PTM), has emerged as a key player in cancer biology. The Warburg effect, a hallmark of tumour metabolism, underscores the significance of lactylation in cancer progression. By regulating gene transcription and protein function, lactylation facilitates metabolic reprogramming, enabling tumours to adapt to nutrient limitations and sustain rapid growth. Over the past decade, extensive research has revealed the intricate regulatory network underlying lactylation in tumours. Large-scale sequencing and machine learning have confirmed the widespread occurrence of lactylation sites across the tumour proteome. Targeting lactylation enzymes or metabolic pathways has demonstrated promising anti-tumour effects, highlighting the therapeutic potential of this modification. This review comprehensively explores the mechanisms of lactylation in cancer cells and the tumour microenvironment. We expound on the application of advanced omics technologies for target identification and data modelling within the lactylation field. Additionally, we summarise existing anti-lactylation drugs and discuss their clinical implications. By providing a comprehensive overview of recent advancements, this review aims to stimulate innovative research and accelerate the translation of lactylation-based therapies into clinical practice. KEY POINTS: Lactylation significantly influences tumour metabolism and gene regulation, contributing to cancer progression. Advanced sequencing and machine learning reveal widespread lactylation sites in tumours. Targeting lactylation enzymes shows promise in enhancing anti-tumour drug efficacy and overcoming chemotherapy resistance. This review outlines the clinical implications and future research directions of lactylation in oncology.
Collapse
Affiliation(s)
- Yipeng He
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Tianbao Song
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Jinzhuo Ning
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Zefeng Wang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Zhen Yin
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Pengcheng Jiang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Qin Yuan
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Weimin Yu
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Fan Cheng
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| |
Collapse
|
36
|
Li Q, Tong Y, Chen J, Xie T. Targeting programmed cell death via active ingredients from natural plants: a promising approach to cancer therapy. Front Pharmacol 2024; 15:1491802. [PMID: 39584140 PMCID: PMC11582395 DOI: 10.3389/fphar.2024.1491802] [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: 09/05/2024] [Accepted: 10/09/2024] [Indexed: 11/26/2024] Open
Abstract
Cancer is a serious public health problem in humans, and prevention and control strategies are still necessary. Therefore, the development of new therapeutic drugs is urgently needed. Targeting programmed cell death, particularly via the induction of cancer cell apoptosis, is one of the cancer treatment approaches employed. Recently, an increasing number of studies have shown that compounds from natural plants can target programmed cell death and kill cancer cells, laying the groundwork for use in future anticancer treatments. In this review, we focus on the latest research progress on the role and mechanism of natural plant active ingredients in different forms of programmed cell death, such as apoptosis, autophagy, necroptosis, ferroptosis, and pyroptosis, to provide a strong theoretical basis for the clinical development of antitumor drugs.
Collapse
Affiliation(s)
- Qian Li
- School of Pharmacy and Department of Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yan Tong
- School of Pharmacy and Department of Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jianxiang Chen
- School of Pharmacy and Department of Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Tian Xie
- School of Pharmacy and Department of Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
37
|
Ma W, Jiang X, Jia R, Li Y. Mechanisms of ferroptosis and targeted therapeutic approaches in urological malignancies. Cell Death Discov 2024; 10:432. [PMID: 39384767 PMCID: PMC11464522 DOI: 10.1038/s41420-024-02195-w] [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: 06/02/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/11/2024] Open
Abstract
The prevalence of urological malignancies remains a significant global health concern, particularly given the challenging prognosis for patients in advanced disease stages. Consequently, there is a pressing need to explore the molecular mechanisms that regulate the development of urological malignancies to discover novel breakthroughs in diagnosis and treatment. Ferroptosis, characterized by iron-ion-dependent lipid peroxidation, is a form of programmed cell death (PCD) distinct from apoptosis, autophagy, and necrosis. Notably, lipid, iron, and glutathione metabolism intricately regulate intracellular ferroptosis, playing essential roles in the progression of various neoplasms and drug resistance. In recent years, ferroptosis has been found to be closely related to urological malignancies. This paper provides an overview of the involvement of ferroptosis in the pathogenesis and progression of urological malignancies, elucidates the molecular mechanisms governing its regulation, and synthesizes recent breakthroughs in diagnosing and treating these malignancies. We aim to provide a new direction for the clinical treatment of urological malignancies.
Collapse
Affiliation(s)
- Wenjie Ma
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Xiaotian Jiang
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.
| | - Yang Li
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.
| |
Collapse
|
38
|
Zhang X, Liang C, Wu C, Wan S, Xu L, Wang S, Wang J, Huang X, Xu L. A rising star involved in tumour immunity: Lactylation. J Cell Mol Med 2024; 28:e70146. [PMID: 39417674 PMCID: PMC11483924 DOI: 10.1111/jcmm.70146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 09/28/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
In recent years, continuous exploration worldwide has revealed that some metabolites produced during cellular and tissue metabolism can act as signalling molecules to exert different effects on the human body. These metabolites may act as cofactors for proteases or as post-translational modifications linked to proteins. Lactate, a traditional metabolite, is found at high levels in the tumour microenvironment (TME). Many studies have shown that lactate influences tumorigenesis and development via different mechanisms, not only through the metabolic reprogramming of tumours but also through its significant impact on tumour immunity. Previously, tumour cells were reported to use glucose and glutamine to fuel lactate metabolism; however, lactate serves not only as an energy source for tumour cells but also as a precursor substance needed for the post-translational modification of proteins. Recent studies identified a novel form of epigenetic modification, lactate-mediated histone lysine lactylation (Kla) and demonstrated that histone lactylation directly stimulates chromatin after gene transcription; consequently, lactylation has become a popular research topic in recent years. This article focuses on the research progress and application prospects of lactylation in the context of tumour immunity.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Gastrointestinal SurgeryThe First Affiliated Yijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
- Anhui Province Key Laboratory of Non‐coding RNA Basic and Clinical Transformation (Wannan Medical College)WuhuChina
| | - Changming Liang
- Department of Gastrointestinal SurgeryThe First Affiliated Yijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
- Anhui Province Key Laboratory of Non‐coding RNA Basic and Clinical Transformation (Wannan Medical College)WuhuChina
| | - Chengwei Wu
- Department of Gastrointestinal SurgeryThe First Affiliated Yijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
- Anhui Province Key Laboratory of Non‐coding RNA Basic and Clinical Transformation (Wannan Medical College)WuhuChina
| | - Senlin Wan
- Department of Gastrointestinal SurgeryThe First Affiliated Yijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
- Anhui Province Key Laboratory of Non‐coding RNA Basic and Clinical Transformation (Wannan Medical College)WuhuChina
| | - Lishuai Xu
- Department of Gastrointestinal SurgeryThe First Affiliated Yijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
- Anhui Province Key Laboratory of Non‐coding RNA Basic and Clinical Transformation (Wannan Medical College)WuhuChina
| | - Song Wang
- Department of Gastrointestinal SurgeryThe First Affiliated Yijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
- Anhui Province Key Laboratory of Non‐coding RNA Basic and Clinical Transformation (Wannan Medical College)WuhuChina
| | - Jiawei Wang
- Department of Gastrointestinal SurgeryThe First Affiliated Yijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
- Anhui Province Key Laboratory of Non‐coding RNA Basic and Clinical Transformation (Wannan Medical College)WuhuChina
| | - Xiaoxu Huang
- Department of Gastrointestinal SurgeryThe First Affiliated Yijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
- Anhui Province Key Laboratory of Non‐coding RNA Basic and Clinical Transformation (Wannan Medical College)WuhuChina
| | - Li Xu
- Department of Gastrointestinal SurgeryThe First Affiliated Yijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
- Anhui Province Key Laboratory of Non‐coding RNA Basic and Clinical Transformation (Wannan Medical College)WuhuChina
| |
Collapse
|
39
|
Yu H, Zhu T, Ma D, Cheng X, Wang S, Yao Y. The role of nonhistone lactylation in disease. Heliyon 2024; 10:e36296. [PMID: 39315193 PMCID: PMC11417196 DOI: 10.1016/j.heliyon.2024.e36296] [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: 05/18/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/25/2024] Open
Abstract
In 2019, a novel post-translational modification termed lactylation was identified, which established a connection among lactate, transcriptional regulation and epigenetics. Lactate, which is traditionally viewed as a metabolic byproduct, is now recognized for its significant functional role, including modulating the tumor microenvironment, engaging in signaling and interfering in immune regulation. While research on lactylation (KLA) is advancing, the focus has primarily been on histone lactylation. This paper aims to explore the less-studied area of nonhistone lactylation, highlighting its involvement in certain diseases and physiological processes. Additionally, the clinical relevance and potential implications of nonhistone lactylation will be discussed.
Collapse
Affiliation(s)
- Hao Yu
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tingting Zhu
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, School of Medicine, Southeast University, China
| | - Dongwen Ma
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, School of Medicine, Southeast University, China
| | - Xiaohan Cheng
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College, Nanjing Medical University, Nanjing, China
| | - Shengjia Wang
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, School of Medicine, Southeast University, China
| | - Yongzhong Yao
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, School of Medicine, Southeast University, China
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College, Nanjing Medical University, Nanjing, China
| |
Collapse
|
40
|
Wang P, Kong G. Comprehensive Analysis of Angiogenesis and Ferroptosis Genes for Predicting the Survival Outcome and Immunotherapy Response of Hepatocellular Carcinoma. J Hepatocell Carcinoma 2024; 11:1845-1859. [PMID: 39364435 PMCID: PMC11448465 DOI: 10.2147/jhc.s483647] [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: 08/03/2024] [Accepted: 09/26/2024] [Indexed: 10/05/2024] Open
Abstract
Background Angiogenesis and ferroptosis are both linked to hepatocellular carcinoma (HCC) development, recurrence, and medication resistance. As a result, a thorough examination of the link between genes associated with angiogenesis and ferroptosis and immunotherapy efficacy is required to improve the dismal prognosis of HCC patients. Methods The molecular subtypes were found using a non-negative matrix factorization technique (NMF) based on the genes associated with angiogenesis and ferroptosis. Based on the differentially expressed genes (DEGs) screed between different molecular subtypes, an angiogenesis and ferroptosis-related prognostic stratification model was built using LASSO-COX regression, random forest technique, and extreme gradient boosting (XGBoost), which was further validated in the ICGC and GSE14520 databases. The impact of this model on tumor microenvironment (TME) and immunotherapy sensitivity was also investigated. The expression levels of candidate genes were detected and validated by Real-Time PCR and immunohistochemistry between liver cancer tissues and adjacent non-tumor liver tissues. Results Both angiogenesis and ferroptosis-related genes can significantly divide HCC patients into two subgroups with different survival outcomes, mutation profiles, and immune microenvironments. We screened six core genes (SLC10A1, PAEP, DPYSL4, MSC, NQO1, and CD24) for the construction of prognostic models by three machine learning methods after intersecting DEGs between angiogenesis and ferroptosis-related subgroups. In both the TCGA, ICGC, and GSE14520 datasets, the model exhibits high prediction efficiency based on the analysis of KM survival curves and ROC curves. Immunomodulatory genes analysis suggested that the model could be used to predict which patients are most likely to benefit from immunotherapy. Furthermore, the transcriptional expression levels of SLC10A1 in the validation experiment matched the outcomes derived from public datasets. Conclusions We identified a new angiogenesis and ferroptosis-related signature that might offer the molecular characteristic information needed for an efficient prognostic assessment and perhaps tailored treatment for HCC patients.
Collapse
Affiliation(s)
- Peng Wang
- Department of Nuclear Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan, 450003, People's Republic of China
| | - Guilian Kong
- Department of Nuclear Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan, 450003, People's Republic of China
| |
Collapse
|
41
|
Yao W, Hu X, Wang X. Crossing epigenetic frontiers: the intersection of novel histone modifications and diseases. Signal Transduct Target Ther 2024; 9:232. [PMID: 39278916 PMCID: PMC11403012 DOI: 10.1038/s41392-024-01918-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/11/2024] [Accepted: 06/30/2024] [Indexed: 09/18/2024] Open
Abstract
Histone post-translational modifications (HPTMs), as one of the core mechanisms of epigenetic regulation, are garnering increasing attention due to their close association with the onset and progression of diseases and their potential as targeted therapeutic agents. Advances in high-throughput molecular tools and the abundance of bioinformatics data have led to the discovery of novel HPTMs which similarly affect gene expression, metabolism, and chromatin structure. Furthermore, a growing body of research has demonstrated that novel histone modifications also play crucial roles in the development and progression of various diseases, including various cancers, cardiovascular diseases, infectious diseases, psychiatric disorders, and reproductive system diseases. This review defines nine novel histone modifications: lactylation, citrullination, crotonylation, succinylation, SUMOylation, propionylation, butyrylation, 2-hydroxyisobutyrylation, and 2-hydroxybutyrylation. It comprehensively introduces the modification processes of these nine novel HPTMs, their roles in transcription, replication, DNA repair and recombination, metabolism, and chromatin structure, as well as their involvement in promoting the occurrence and development of various diseases and their clinical applications as therapeutic targets and potential biomarkers. Moreover, this review provides a detailed overview of novel HPTM inhibitors targeting various targets and their emerging strategies in the treatment of multiple diseases while offering insights into their future development prospects and challenges. Additionally, we briefly introduce novel epigenetic research techniques and their applications in the field of novel HPTM research.
Collapse
Affiliation(s)
- Weiyi Yao
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Xinting Hu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
- Taishan Scholars Program of Shandong Province, Jinan, Shandong, 250021, China.
| |
Collapse
|
42
|
Yu L, Qiu Y, Tong X. Ferroptosis in Renal Cancer Therapy: A Narrative Review of Drug Candidates. Cancers (Basel) 2024; 16:3131. [PMID: 39335103 PMCID: PMC11430741 DOI: 10.3390/cancers16183131] [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: 07/20/2024] [Revised: 08/26/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Renal cancer is a common and serious malignant tumor of the urinary system. While surgery effectively treats early-stage renal cancer, advanced cases pose a significant challenge due to poor treatment outcomes and chemotherapy resistance. Therefore, there is an urgent need to develop alternative therapeutic strategies. Ferroptosis is a newly defined form of programmed cell death characterized by the accumulation of iron-dependent lipid peroxides, which plays a critical role in tumor progression and drug resistance. Recent studies have shown that ferroptosis is involved in the occurrence and development of renal cancer, and ferroptosis-related genes can induce cell apoptosis and can be used as potential biomarkers for early diagnosis of renal cancer and participate in drug resistance of renal cancer chemotherapy. With the continuous improvement of the mechanism of ferroptosis, drugs targeting ferroptosis for the treatment of renal cancer are emerging in an endless stream. Based on the theoretical basis of the occurrence of ferroptosis, this paper reviewed drug-induced ferroptosis in renal cancer cells from the aspects of herbal medicine, natural compounds, drug resistance mechanisms, and nanomaterials, and delves into the clinical application potential of ferroptosis-related drugs in the treatment of renal cancer.
Collapse
Affiliation(s)
- Lingyan Yu
- Zhejiang Chinese Medical University, Hangzhou 310053, China
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Yuyueyang Qiu
- Department of Biology, Grinnell College, Grinnell, IA 50112, USA
| | - Xiangmin Tong
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou 310014, China
- Department of Laboratory Medicine, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| |
Collapse
|
43
|
Fan B, Guo Q, Wang S. The application of alkaloids in ferroptosis: A review. Biomed Pharmacother 2024; 178:117232. [PMID: 39098181 DOI: 10.1016/j.biopha.2024.117232] [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: 05/25/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/06/2024] Open
Abstract
Alkaloids have remarkable biological and pharmacological properties and have recently garnered extensive attention. Various alkaloids, including commercially available drugs such as berberine, substantially affect ferroptosis. In addition to the three main pathways of ferroptosis, iron metabolism, phospholipid metabolism, and the glutathione peroxidase 4-regulated pathway, novel mechanisms of ferroptosis are continuously being identified. Alkaloids can modulate the progression of various diseases through ferroptosis and exhibit the ability to exert varied effects depending on dosage and tissue type underscores their versatility. Therefore, this review comprehensively summarizes primary targets and the latest advancements of alkaloids in ferroptosis, as well as the dual roles of alkaloids in inhibiting and promoting ferroptosis.
Collapse
Affiliation(s)
- Bocheng Fan
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province 110013, China
| | - Qihao Guo
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province 110013, China
| | - Shu Wang
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province 110013, China.
| |
Collapse
|
44
|
Hao Y, Gu C, Luo W, Shen J, Xie F, Zhao Y, Song X, Han Z, He J. The role of protein post-translational modifications in prostate cancer. PeerJ 2024; 12:e17768. [PMID: 39148683 PMCID: PMC11326433 DOI: 10.7717/peerj.17768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/27/2024] [Indexed: 08/17/2024] Open
Abstract
Involving addition of chemical groups or protein units to specific residues of the target protein, post-translational modifications (PTMs) alter the charge, hydrophobicity, and conformation of a protein, which in turn influences protein function, protein-protein interaction, and protein aggregation. These alterations, which include phosphorylation, glycosylation, ubiquitination, methylation, acetylation, lipidation, and lactylation, are significant biological events in the development of cancer, and play vital roles in numerous biological processes. The processes behind essential functions, the screening of clinical illness signs, and the identification of therapeutic targets all depend heavily on further research into the PTMs. This review outlines the influence of several PTM types on prostate cancer (PCa) diagnosis, therapy, and prognosis in an effort to shed fresh light on the molecular causes and progression of the disease.
Collapse
Affiliation(s)
- Yinghui Hao
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chenqiong Gu
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenfeng Luo
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jian Shen
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fangmei Xie
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ying Zhao
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoyu Song
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zeping Han
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jinhua He
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
- Rehabilitation Medicine Institute of Panyu District, Guangzhou, Guangdong, China
| |
Collapse
|
45
|
Chen HJ, Yu MM, Huang JC, Lan FY, Liao HH, Xu ZH, Yu YJ, Huang YC, Chen F. SLC4A4 is a novel driver of enzalutamide resistance in prostate cancer. Cancer Lett 2024; 597:217070. [PMID: 38880227 DOI: 10.1016/j.canlet.2024.217070] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 06/03/2024] [Accepted: 06/12/2024] [Indexed: 06/18/2024]
Abstract
The androgen receptor signaling inhibitor (ARSI) enzalutamide (Enz) has shown critical efficacy in the treatment of advanced prostate cancer (PCa). However, the development of drug resistance is a significant factor contributing to mortality in PCa patients. We aimed to explore the key mechanisms of Enz-resistance. Through analysis of GEO databases, we identified SLC4A4 as a novel driver in Enz resistance. Long-term Enz treatment leads to the up-regulation of SLC4A4, which in turn mediates P53 lactylation via the NF-κB/STAT3/SLC4A4 axis, ultimately leading to the development of Enz resistance and progression of PCa. SLC4A4 knockdown overcomes Enz resistance both in vitro and in vivo. Hence, our results suggest that targeting SLC4A4 could be a promising therapeutic strategy for Enz resistance. STATEMENT OF SIGNIFICANCE: SLC4A4 is a novel driver of enzalutamide resistance.
Collapse
Affiliation(s)
- Hao-Jie Chen
- Department of Urology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China; Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Ming-Ming Yu
- Department of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jia-Cheng Huang
- Department of Urology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Fu-Ying Lan
- Department of Urology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Hai-Hong Liao
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Zi-Han Xu
- Department of Urology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Yong-Jiang Yu
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China.
| | - Yi-Chen Huang
- Department of Urology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China.
| | - Fang Chen
- Department of Urology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China.
| |
Collapse
|
46
|
Wang J, Liang Y, Liang X, Peng H, Wang Y, Xu M, Liang X, Yao H, Liu X, Zeng L, Yao P, Xiang D. Evodiamine suppresses endometriosis development induced by early EBV exposure through inhibition of ERβ. Front Pharmacol 2024; 15:1426660. [PMID: 39148548 PMCID: PMC11324466 DOI: 10.3389/fphar.2024.1426660] [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: 05/01/2024] [Accepted: 07/17/2024] [Indexed: 08/17/2024] Open
Abstract
Introduction: Endometriosis (EMS) is characterized as a prevalent gynecological inflammatory disorder marked by the existence of endometrial tissues situated beyond the uterus. This condition leads to persistent pelvic pain and may contribute to infertility. In this investigation, we explored the potential mechanism underlying the development of endometriosis (EMS) triggered by transient exposure to either latent membrane protein 1 (LMP1) or Epstein-Barr virus (EBV) in a mouse model. Additionally, we examined the potential inhibitory effect of evodiamine (EDM) on EMS. Methods: Immortalized human endometrial stromal cells (HESC) or epithelial cells (HEEC) were transiently exposed to either EBV or LMP1. The presence of evodiamine (EDM) was assessed for its impact on estrogen receptor β (ERβ) expression, as well as on cell metabolism parameters such as redox balance, mitochondrial function, inflammation, and proliferation. Additionally, a mixture of LMP1-treated HESC and HEEC was administered intraperitoneally to generate an EMS mouse model. Different dosages of EDM were employed for treatment to evaluate its potential suppressive effect on EMS development. Results: Transient exposure to either EBV or LMP1 triggers persistent ERβ expression through epigenetic modifications, subsequently modulating related cell metabolism for EMS development. Furthermore, 4.0 µM of EDM can efficiently reverse this effect in in vitro cell culture studies. Additionally, 20 mg/kg body weight of EDM treatment can partly suppress EMS development in the in vivo EMS mouse model. Conclusion: Transient EBV/LMP1 exposure triggers permanent ERβ expression, favoring later EMS development, EDM inhibits EMS development through ERβ suppression. This presents a novel mechanism for the development of endometriosis (EMS) in adulthood stemming from early Epstein-Barr virus (EBV) exposure during childhood. Moreover, evodiamine (EDM) stands out as a prospective candidate for treating EMS.
Collapse
Affiliation(s)
- Junling Wang
- Department of Gynecology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuanqi Liang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoru Liang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huijuan Peng
- Department of Gynecology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongxia Wang
- Department of Gynecology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Mingtao Xu
- Department of Gynecology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuefang Liang
- Department of Gynecology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Helen Yao
- University of California at Riverside, Riverside, CA, United States
| | - Xiaohan Liu
- Department of Gynecology, Sun Yat-Sen University Affiliated No. 8 Hospital, Shenzhen, China
| | - Liqin Zeng
- Department of Gynecology, Sun Yat-Sen University Affiliated No. 8 Hospital, Shenzhen, China
| | - Paul Yao
- Department of Gynecology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dongfang Xiang
- Department of Gynecology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
47
|
Lin J, Ren J. Lactate-induced lactylation and cardiometabolic diseases: From epigenetic regulation to therapeutics. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167247. [PMID: 38762059 DOI: 10.1016/j.bbadis.2024.167247] [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: 01/10/2024] [Revised: 04/23/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024]
Abstract
Cardiometabolic diseases (CMDs) denote a cadre of chronic and devastating cardiovascular anomalies routed from metabolic derangements including obesity, type 2 diabetes mellitus, and hypertension. Recent studies have demonstrated the association between histone lactylation, a unique form of post-translational modification, and pathogenesis of CMDs, apparently through epigenetic mechanisms. Lactylation has been indicated to regulate key aspects of metabolism, inflammation, and cardiovascular function in the realm of CMDs in a cellular and tissue-specific manner. A better understanding of the molecular, cellular and physiological domains of lactylation in the etiology of CMDs is expected to offer new insights into etiopathogenesis, hazardous factor control and therapeutic development for these challenging ailments.
Collapse
Affiliation(s)
- Jie Lin
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, 200032, China; Shanhai Institude of Cardiovascular Diseases, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, 200032, China; Shanhai Institude of Cardiovascular Diseases, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| |
Collapse
|
48
|
Xu K, Zhang K, Wang Y, Gu Y. Comprehensive review of histone lactylation: Structure, function, and therapeutic targets. Biochem Pharmacol 2024; 225:116331. [PMID: 38821374 DOI: 10.1016/j.bcp.2024.116331] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Histone lysine lactylation (Kla) has emerged as a distinct epigenetic modification that differs markedly from established acylation modifications through the unique addition of a lactyl group to a lysine residue. Such modifications not only alter nucleosome structure but also significantly impact chromatin dynamics and gene expression, thus playing a crucial role in cellular metabolism, inflammatory responses, and embryonic development. The association of histone Kla with various metabolic processes, particularly glycolysis and glutamine metabolism, underscores its pivotal role in metabolic reprogramming, including in cancerous tissues, where it contributes to tumorigenesis, immune evasion, and angiogenesis. In addition, histone Kla is involved in the pathogenesis of various diseases, particularly several cancers and neurodegenerative diseases. The identification of histone Kla opens new avenues for therapeutic interventions targeting specific Kla sites. In this review, we summarize the differences between histone Kla modifications and other acylation modifications, discuss the mechanisms and roles of histone Kla in disease, and conclude by describing existing drugs and potential targets. This study provides new insights into the mechanisms linking histone Kla to diseases and into the discovery of new drugs and targets.
Collapse
Affiliation(s)
- Kaiwen Xu
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China
| | - Keyi Zhang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China
| | - Yanshuang Wang
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine and Laboratory Medicine, Hainan Medical University, Haikou 571199, China
| | - Yue Gu
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
49
|
Wu Z, Chai Z, Cai X, Wang J, Wang H, Yue B, Zhang M, Wang J, Wang H, Zhong J, Xin J. Protein Lactylation Profiles Provide Insights into Molecular Mechanisms Underlying Metabolism in Yak. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 38850252 DOI: 10.1021/acs.jafc.4c01800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2024]
Abstract
Protein lysine lactylation, a recently discovered post-translational modification (PTM), is prevalent across tissues and cells of diverse species, serving as a regulator of glycolytic flux and biological metabolism. The yak (Bos grunniens), a species that has inhabited the Qinghai-Tibetan Plateau for millennia, has evolved intricate adaptive mechanisms to cope with the region's unique geographical and climatic conditions, exhibiting remarkable energy utilization and metabolic efficiency. Nonetheless, the specific landscape of lysine lactylation in yaks remains poorly understood. Herein, we present the first comprehensive lactylome profile of the yak, effectively identifying 421, 308, and 650 lactylated proteins in the heart, muscles, and liver, respectively. These lactylated proteins are involved in glycolysis/gluconeogenesis, the tricarboxylic acid cycle, oxidative phosphorylation, and metabolic process encompassing carbohydrates, lipids, and proteins during both anaerobic and aerobic glucose bio-oxidation, implying their crucial role in material and energy metabolism, as well as in maintaining homeostasis in yaks.
Collapse
Affiliation(s)
- Zhijuan Wu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, Sichuan 610225, China
| | - Zhixin Chai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, Sichuan 610225, China
| | - Xin Cai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, Sichuan 610225, China
| | - Jiabo Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, Sichuan 610225, China
| | - Hui Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, Sichuan 610225, China
| | - Binglin Yue
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, Sichuan 610225, China
| | - Ming Zhang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, Sichuan 610225, China
| | - Jikun Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, Sichuan 610225, China
| | - Haibo Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, Sichuan 610225, China
| | - Jincheng Zhong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, Sichuan 610225, China
- Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan 610225, China
| | - Jinwei Xin
- State Key Laboratory of Hulless Barley and Yak Germplasm Resources and Genetic Improvement, Lhasa, Tibet 850000, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, Tibet 850009, China
| |
Collapse
|
50
|
Pan J, Zhang J, Lin J, Cai Y, Zhao Z. Constructing lactylation-related genes prognostic model to effectively predict the disease-free survival and treatment responsiveness in prostate cancer based on machine learning. Front Genet 2024; 15:1343140. [PMID: 38566813 PMCID: PMC10985269 DOI: 10.3389/fgene.2024.1343140] [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: 11/27/2023] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
Background: Prostate cancer (PCa) is one of the most common malignancies in men with a poor prognosis. It is therefore of great clinical importance to find reliable prognostic indicators for PCa. Many studies have revealed the pivotal role of protein lactylation in tumor development and progression. This research aims to analyze the effect of lactylation-related genes on PCa prognosis. Methods: By downloading mRNA-Seq data of TCGA PCa, we obtained the differential genes related to lactylation in PCa. Five machine learning algorithms were used to screen for lactylation-related key genes for PCa, then the five overlapping key genes were used to construct a survival prognostic model by lasso cox regression analysis. Furthermore, the relationships between the model and related pathways, tumor mutation and immune cell subpopulations, and drug sensitivity were explored. Moreover, two risk groups were established according to the risk score calculated by the five lactylation-related genes (LRGs). Subsequently, a nomogram scoring system was established to predict disease-free survival (DFS) of patients by combining clinicopathological features and lactylation-related risk scores. In addition, the mRNA expression levels of five genes were verified in PCa cell lines by qPCR. Results: We identified 5 key LRGs (ALDOA, DDX39A, H2AX, KIF2C, RACGAP1) and constructed the LRGs prognostic model. The AUC values for 1 -, 3 -, and 5-year DFS in the TCGA dataset were 0.762, 0.745, and 0.709, respectively. The risk score was found a better predictor of DFS than traditional clinicopathological features in PCa. A nomogram that combined the risk score with clinical variables accurately predicted the outcome of the patients. The PCa patients in the high-risk group have a higher proportion of regulatory T cells and M2 macrophage, a higher tumor mutation burden, and a worse prognosis than those in the low-risk group. The high-risk group had a lower IC50 for certain chemotherapeutic drugs, such as Docetaxel, and Paclitaxel than the low-risk group. Furthermore, five key LRGs were found to be highly expressed in castration-resistant PCa cells. Conclusion: The lactylation-related genes prognostic model can effectively predict the DFS and therapeutic responses in patients with PCa.
Collapse
Affiliation(s)
| | | | | | | | - Zhigang Zhao
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Provincial Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally Invasive Surgery Robot and Intelligent Equipment, Guangzhou Institute of Urology, Guangzhou, China
| |
Collapse
|