1
|
Liao Y, Niu L, Ling J, Cui Y, Huang Z, Xu J, Jiang Y, Yu P, Liu X. Turning sour into sweet: Lactylation modification as a promising target in cardiovascular health. Metabolism 2025; 168:156234. [PMID: 40113080 DOI: 10.1016/j.metabol.2025.156234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 02/26/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
Lactylation, a recently identified posttranslational modification (PTM), has emerged as a critical regulatory mechanism in cardiovascular diseases (CVDs). This PTM involves the addition of lactyl groups to lysine residues on histones and nonhistone proteins, influencing gene expression and cellular metabolism. The discovery of lactylation has revealed new directions for understanding metabolic and immune processes, particularly in the context of CVDs. This review describes the intricate roles of specific lactylated proteins and enzymes, such as H3K18, HMGB1, MCT1/4, and LDH, in the regulation of cardiovascular pathology. This study also highlights the unique impact of lactylation on myocardial hypertrophy and distinguishes it from other PTMs, such as SUMOylation and acetylation, underscoring its potential as a therapeutic target. Emerging drugs targeting lactate transporters and critical enzymes involved in lactylation offer promising avenues for novel CVD therapies. This review calls for further research to elucidate the mechanisms linking lactylation to CVDs, emphasizing the need for comprehensive studies at the molecular, cellular, and organismal levels to pave the way for innovative preventive, diagnostic, and treatment strategies in cardiovascular medicine.
Collapse
Affiliation(s)
- Yajie Liao
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China; Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China; Queen Mary School, Nanchang University, Nanchang, Jiangxi, China
| | - Liyan Niu
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jitao Ling
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yuzhen Cui
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China; Queen Mary School, Nanchang University, Nanchang, Jiangxi, China
| | - Zixuan Huang
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China; Queen Mary School, Nanchang University, Nanchang, Jiangxi, China
| | - Jingdong Xu
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China; Queen Mary School, Nanchang University, Nanchang, Jiangxi, China
| | - Yuan Jiang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| | - Peng Yu
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China; Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| | - Xiao Liu
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China; Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Jin H, Liu Q, Li J, Zhao S, Tuo B. Multifaceted roles of lactate dehydrogenase in liver cancer (Review). Int J Oncol 2025; 66:50. [PMID: 40417916 PMCID: PMC12118952 DOI: 10.3892/ijo.2025.5756] [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: 02/21/2025] [Accepted: 05/05/2025] [Indexed: 05/27/2025] Open
Abstract
Hepatocellular carcinoma (HCC) has high morbidity and mortality rates, and metabolic reprogramming of HCC cells supports the proliferation and development of tumor cells. Lactate dehydrogenase (LDH), a key metabolic enzyme, can maintain the rapid proliferative demand of tumor cells by promoting glycolysis and lactate production in HCC cells. In addition, LDH regulates redox homeostasis and influences lipid synthesis and signaling pathways, further promoting tumor invasion and metastasis. In the tumor microenvironment, LDH affects the function of immune cells and stromal cells by regulating the lactate concentration in and promoting the immune escape and angiogenesis of tumor cells. Since elevated levels of LDH are closely associated with tumor load, invasiveness and poor prognosis, LDH also has promising applications in the early diagnosis, treatment and prognostic assessment of HCC. The present study reviewed the roles of LDH in the occurrence, development, diagnosis, prognosis and treatment of HCC and explored its value as an important biomarker and potential therapeutic target, with the aim of providing a comprehensive reference for HCC‑related research and clinical practice.
Collapse
Affiliation(s)
| | | | - Jin Li
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Siyu Zhao
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
3
|
Cheng P, Gan L, Wu J, Hao X, Li Q, Chen L. ALDH2 delays ventricular pressure overload-induced heart failure by promoting cardiomyocyte proliferation in mice. Exp Cell Res 2025; 448:114571. [PMID: 40273968 DOI: 10.1016/j.yexcr.2025.114571] [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: 02/20/2025] [Revised: 04/21/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
The adult mammalian heart is a terminally differentiated organ in which the majority of cardiomyocytes are in a state of cell cycle arrest, rendering them incapable of effectively proliferating to replace damaged cells. ALDH2, an enzyme known for alleviating oxidative stress, has been demonstrated to play a critical role in cardiac protection. However, whether ALDH2 regulates cardiomyocyte proliferation has not been conclusively established. We found that activation of ALDH2 activity significantly promotes cardiomyocyte proliferation and extends the proliferation window during early postnatal development in neonatal mice. Furthermore, administration of Alda-1 to activate ALDH2 in adult mice subjected to transverse aortic constriction markedly enhanced cardiomyocyte proliferation and delayed the onset of pressure overload-induced heart failure. In summary, our findings identify ALDH2 as a potential target for regulating cardiomyocyte proliferation and offer a novel therapeutic approach for treating heart failure.
Collapse
Affiliation(s)
- Peng Cheng
- Department of Physiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Lu Gan
- Department of Physiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Jieyun Wu
- Department of Physiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Xiaodan Hao
- Department of Physiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Qiyong Li
- Department of Cardiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Li Chen
- Department of Physiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China.
| |
Collapse
|
4
|
Wang J, Zhang Y, Cao C, Hua J, Xing L, Wu C. The anti-atherosclerosis effect and molecular mechanism of AMPKα1 by polarizing monocytes to an M2 phenotype via cell-intrinsic lysosomal lipolysis. Cardiovasc Pathol 2025; 78:107742. [PMID: 40354887 DOI: 10.1016/j.carpath.2025.107742] [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: 11/27/2024] [Revised: 05/09/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025] Open
Abstract
Regulating the differentiation of monocytes into M2 macrophages can promote the regression of Atherosclerosis (AS) plaque. However, the key molecules regulating the differentiation of monocytes to M2 are unknown. In this study, we reported that adenosine-activated protein kinase α1 (AMPKα1) plays an anti-AS role by polarizing monocytes to an M2 phenotype via promoting fatty acid oxidation (FAO). AMPKα1 enhances the decomposition of cholesterol esters by increasing lysosomal acid lipase expression to provide fatty acids for FAO. Furthermore, AMPKα1 can induce lysosomal biogenesis and enhance lipolysis by promoting the transcription factor EB (TFEB) expression and facilitating TFEB nuclear translocation. In conclusion, AMPKα1 enhances the decomposition of cholesterol esters by increasing lysosomal acid lipase expression to produce fatty acids, which may represent a mechanism to promote FAO and inflammatory monocytes differentiation towards M2 phenotype.
Collapse
Affiliation(s)
- Jing Wang
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan, 030006, Shanxi province, China.
| | - Yahui Zhang
- School of Life Science, Shanxi University, Taiyuan, 030006, China
| | - Caixing Cao
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan, 030006, Shanxi province, China
| | - Jiale Hua
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan, 030006, Shanxi province, China
| | - Li Xing
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan, 030006, Shanxi province, China
| | - Changxin Wu
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan, 030006, Shanxi province, China.
| |
Collapse
|
5
|
Wang T, Wang X, Ren W, Sun Z, Zhang Y, Wu N, Diao H. Cardiomyocyte proliferation: Advances and insights in macrophage-targeted therapy for myocardial injury. Genes Dis 2025; 12:101332. [PMID: 39935606 PMCID: PMC11810708 DOI: 10.1016/j.gendis.2024.101332] [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: 08/17/2023] [Revised: 02/18/2024] [Accepted: 03/20/2024] [Indexed: 02/13/2025] Open
Abstract
In the mammalian heart, cardiomyocytes undergo a transient window of proliferation that leads to regenerative impairment, limiting cardiomyocyte proliferation and myocardial repair capacity. Cardiac developmental patterns exacerbate the progression of heart disease characterized by myocardial cell loss, ultimately leading to cardiac dysfunction and heart failure. Myocardial infarction causes the death of partial cardiomyocytes, which triggers an immune response to remove debris and restore tissue integrity. Interestingly, when transient myocardial injury triggers irreversible loss of cardiomyocytes, the subsequent macrophages responsible for proliferation and regeneration have a unique immune phenotype that promotes the formation of pre-existing new cardiomyocytes. During mammalian regeneration, mononuclear-derived macrophages and self-renewing resident cardiac macrophages provide multiple cytokines and molecular signals that create a regenerative environment and cellular plasticity capacity in postnatal cardiomyocytes, a pivotal strategy for achieving myocardial repair. Consistent with other human tissues, cardiac macrophages originating from the embryonic endothelium produce a hierarchy of contributions to monocyte recruitment and fate specification. In this review, we discuss the novel functions of macrophages in triggering cardiac regeneration and repair after myocardial infarction and provide recent advances and prospective insights into the phenotypic transformation and heterogeneous features involving cardiac macrophages. In conclusion, macrophages contribute critically to regeneration, repair, and remodeling, and are challenging targets for cardiovascular therapeutic interventions.
Collapse
Affiliation(s)
- Tao Wang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Xueyao Wang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Weibin Ren
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Zeyu Sun
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Yanhui Zhang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Nanping Wu
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Hongyan Diao
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
6
|
Chen X, Zhang Z, Wang K. Lactate released by lung adenocarcinoma (LUAD) cells promotes M2 macrophage polarization via the GPR132/cAMP/PKA pathway. Genes Genomics 2025; 47:521-531. [PMID: 40053234 DOI: 10.1007/s13258-025-01622-1] [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: 11/03/2024] [Accepted: 01/31/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Increasing evidence suggests that lactate is an essential compound in the tumor microenvironment, and especially for macrophage cells. However, the mechanism by which lactate affects macrophages remains unclear. OBJECTIVE This study investigated whether and how lactate affects macrophage polarization in lung adenocarcinoma (LUAD). METHODS Clinical samples of LUAD and paracancerous tissue were obtained for evaluation of lactate dehydrogenase A (LDHA) expression. LUAD cell lines and THP-1 induced macrophages were used in this study. Quantitative real-time PCR (QPCR), western blotting, and immunohistochemical (IHC) staining were performed to detect gene expression. Flow cytometry and ELISA assays were used to detect the levels of M1 macrophage and M2 macrophage biomarkers. RESULTS LDHA was highly expressed in the LUAD tissues. Culture medium supernatants derived from LUAD cells (CM) promoted macrophage M2 polarization, and lactate levels were elevated in the CM. Inhibition of LDHA in LUAD cells decreased lactate levels and suppressed M2 macrophage polarization. Moreover, overexpression of GPR132 in macrophages promoted, while GPR132 knockdown in macrophages suppressed M2 macrophage polarization and cAMP (Cyclic Adenosine 3',5'-Monophosphate)/PKA (Protein Kinase) pathway activation induced by lactate. The effect of GPR132 overexpression was reversed by a PKA inhibitor (H-89). CONCLUSION Collectively, our results confirmed that lactate released by LUAD cells promoted M2 macrophage polarization via the GPR132/cAMP/PKA pathway.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Geriatrics, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Zhongzheng Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical University, 287 Chang Huai Road, Bengbu, 233000, Anhui, China
| | - Kangwu Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical University, 287 Chang Huai Road, Bengbu, 233000, Anhui, China.
| |
Collapse
|
7
|
Jia X, Dong Y, Lu J, Yang Z, Xu R, Zhang X, Jiao J, Zhang Z, Lin Y, Chu F, Wang P, Zhong T, Lei H. A self-assembly enzyme-like hydrogel with ROS scavenging and immunomodulatory capability for microenvironment-responsive wound healing acceleration. Int J Pharm 2025; 675:125529. [PMID: 40158760 DOI: 10.1016/j.ijpharm.2025.125529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/12/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
On-demand responsive hydrogels are a promising solution for effective wound management as they can adjust their properties in response to changes in the wound environment, allowing them to provide tailored support for the healing process. However, the conventional hydrogels may not fully meet the diverse demands of the intricate healing process. Herein, a novel glycyrrhizic acid (GA) based self-assembly hydrogel coordinated with copper and polyphenol (GCP hydrogel) was developed to exhibit triggered release behavior in response to the microenvironment. The GCP hydrogel coordinated with copper and protocatechuic acid (PA) and self-assembled with GA, also exhibits enzyme-like properties by mimicking the cascade process of superoxide dismutase (SOD) and catalase (CAT), effectively scavenging reactive oxygen species (ROS). Furthermore, the on-demand release of Cu2+ at different stages of the wound healing process can not only enhance the antibacterial ability of methicillin-resistant Staphylococcus aureus (MRSA) but also intelligently promote angiogenesis with outstanding biocompatibility. In addition, the GCP hydrogel effectively modulated the activity of macrophages in response to inflammatory stimuli, exhibiting remarkable anti-inflammatory abilities and promoting tissue regeneration. The multifunctional GCP hydrogel platform has the potential to create a dynamic microenvironment that is conducive to tissue regeneration, making it an ideal candidate for smart wound management.
Collapse
Affiliation(s)
- Xiaohui Jia
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Yuhe Dong
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Jihui Lu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhenyuan Yang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Ran Xu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiang Zhang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jingyi Jiao
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zixuan Zhang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yixuan Lin
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Fuhao Chu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Penglong Wang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Tian Zhong
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macao 999078, China.
| | - Haimin Lei
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
8
|
Li H, Ren Q, Shi M, Ma L, Fu P. Lactate metabolism and acute kidney injury. Chin Med J (Engl) 2025; 138:916-924. [PMID: 38802283 PMCID: PMC12037090 DOI: 10.1097/cm9.0000000000003142] [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: 01/22/2024] [Indexed: 05/29/2024] Open
Abstract
ABSTRACT Acute kidney injury (AKI) is a common clinically critical syndrome in hospitalized patients with high morbidity and mortality. At present, the mechanism of AKI has not been fully elucidated, and no therapeutic drugs exist. As known, glycolytic product lactate is a key metabolite in physiological and pathological processes. The kidney is an important gluconeogenic organ, where lactate is the primary substrate of renal gluconeogenesis in physiological conditions. During AKI, altered glycolysis and gluconeogenesis in kidneys significantly disturb the lactate metabolic balance, which exert impacts on the severity and prognosis of AKI. Additionally, lactate-derived posttranslational modification, namely lactylation, is novel to AKI as it could regulate gene transcription of metabolic enzymes involved in glycolysis or Warburg effect. Protein lactylation widely exists in human tissues and may severely affect non-histone functions. Moreover, the strategies of intervening lactate metabolic pathways are expected to bring a new dawn for the treatment of AKI. This review focused on renal lactate metabolism, especially in proximal renal tubules after AKI, and updated recent advances of lactylation modification, which may help to explore potential therapeutic targets against AKI.
Collapse
Affiliation(s)
- Hui Li
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Qian Ren
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Min Shi
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Liang Ma
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Ping Fu
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
9
|
Koopmans T, van Rooij E. Molecular gatekeepers of endogenous adult mammalian cardiomyocyte proliferation. Nat Rev Cardiol 2025:10.1038/s41569-025-01145-y. [PMID: 40195566 DOI: 10.1038/s41569-025-01145-y] [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] [Accepted: 03/10/2025] [Indexed: 04/09/2025]
Abstract
Irreversible cardiac fibrosis, cardiomyocyte death and chronic cardiac dysfunction after myocardial infarction pose a substantial global health-care challenge, with no curative treatments available. To regenerate the injured heart, cardiomyocytes must proliferate to replace lost myocardial tissue - a capability that adult mammals have largely forfeited to adapt to the demanding conditions of life. Using various preclinical models, our understanding of cardiomyocyte proliferation has progressed remarkably, leading to the successful reactivation of cell cycle induction in adult animals, with functional recovery after cardiac injury. Central to this success is the targeting of key pathways and structures that drive cardiomyocyte maturation after birth - nucleation and ploidy, sarcomere structure, developmental signalling, chromatin and epigenetic regulation, the microenvironment and metabolic maturation - forming a complex regulatory framework that allows efficient cellular contraction but restricts cardiomyocyte proliferation. In this Review, we explore the molecular pathways underlying these core mechanisms and how their manipulation can reactivate the cell cycle in cardiomyocytes, potentially contributing to cardiac repair.
Collapse
Affiliation(s)
- Tim Koopmans
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, Netherlands
| | - Eva van Rooij
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, Netherlands.
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands.
| |
Collapse
|
10
|
Cao SH, Ma RY, Cao T, Hu T, Yang S, Ren ZY, Niu JL, Zheng MQ, Han M, Dong LH. PKM2 crotonylation reprograms glycolysis in VSMCs, contributing to phenotypic switching. Oncogene 2025:10.1038/s41388-025-03353-9. [PMID: 40181154 DOI: 10.1038/s41388-025-03353-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 02/19/2025] [Accepted: 03/13/2025] [Indexed: 04/05/2025]
Abstract
Post-translational modifications (PTMs) of pyruvate kinase M2 (PKM2) play a vital role in regulating its activity and function. Recently, we found PKM2 can undergo crotonylation in vascular smooth muscle cell (VSMC) phenotypic switching. However, the role of PKM2 crotonylation remains unknown. Here, we verify a crucial role of PKM2 crotonylation in VSMC metabolic reprogramming. In PDGF-BB-induced synthetic VSMCs, PKM2 crotonylation was upregulated and promotes its nuclear translocation, thereby facilitating the expression of Glut1 and Ldha. Furthermore, crotonylation facilitated the dimeric formation of PKM2. Then we identified the highly conserved crotonylation site at K305 across different species. The crotonylation of PKM2 was compromised by PKM2 K305 mutation, resulting in the suppression of PKM2 dimeric configuration and nuclear relocation, and ultimately reducing glycolysis rate. Furthermore, PKM2 K305 crotonylation was necessary for VSMC phenotypic switching in vitro and intimal hyperplasia in vivo via infection of PKM2 recombinant adenovirus. In summary, PKM2 K305 crotonylation facilitates VSMC aerobic glycolysis by enhancing PKM2 dimeric form.
Collapse
Affiliation(s)
- Shan-Hu Cao
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
- Department of Cardiology, Hebei Key Laboratory of Heart and Metabolism, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China
- Hebei Medical University Clinical Medicine Postdoctoral Mobile Station, Shijiazhuang, China
| | - Ru-Yuan Ma
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Tong Cao
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Tao Hu
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Shu Yang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Zhi-Yan Ren
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Jiang-Ling Niu
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Ming-Qi Zheng
- Department of Cardiology, Hebei Key Laboratory of Heart and Metabolism, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China.
| | - Mei Han
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, Hebei, China.
| | - Li-Hua Dong
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, Hebei, China.
- Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
11
|
Wan T, Liang Y, Wei T, Chen Z, Li Y. Targeting Lactic Acid Modification in Ischemic Heart Diseases: Novel Therapeutics and Mechanism. J Cardiovasc Transl Res 2025; 18:257-267. [PMID: 39920549 DOI: 10.1007/s12265-025-10593-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/20/2025] [Indexed: 02/09/2025]
Abstract
Ischemic heart disease (IHD), especially acute myocardial infarction (AMI), has a high mortality rate and poses a great threat to human health. When myocardial infarction occurs, the structure and function of the myocardium are significantly damaged, and its metabolisms switch from oxidative phosphorylation to glycolysis, producing lactate. Lactylation, as a newly discovered post-translational modification (PMT) in recent years, is involved in the regulation of gene expression, and cell proliferation. Emerging studies have revealed that lactate and lactylation modifications participate in inflammation and cardiac repair, and play an important role in cardiovascular diseases, such as myocardial infarction, myocardial fibrosis, and heart failure. Therefore, in this review, we discuss how glucose metabolism, glycolytic end-product lactate, and lactylation potentially interact with pathological processes, including inflammation, cardiac fibrosis, and heart failure. And targeting glycolysis and lactylation modification could provide a promising future for cardiovascular diseases.
Collapse
Affiliation(s)
- Tangjiang Wan
- Department of Cardiology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215000, Jiangsu Province, China
| | - Yucheng Liang
- Department of Cardiology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215000, Jiangsu Province, China
| | - Tianwen Wei
- Department of Cardiovascular Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zijie Chen
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Ischemic Heart Diseases, Fudan University, Shanghai, China
| | - Yafei Li
- Department of Cardiology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215000, Jiangsu Province, China.
| |
Collapse
|
12
|
Zhao L, Deng H, Zhang J, Zamboni N, Yang H, Gao Y, Yang Z, Xu D, Zhong H, van Geest G, Bruggmann R, Zhou Q, Schmid RA, Marti TM, Dorn P, Peng RW. Lactate dehydrogenase B noncanonically promotes ferroptosis defense in KRAS-driven lung cancer. Cell Death Differ 2025; 32:632-645. [PMID: 39643712 PMCID: PMC11982314 DOI: 10.1038/s41418-024-01427-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 11/21/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024] Open
Abstract
Ferroptosis is an oxidative, non-apoptotic cell death frequently inactivated in cancer, but the underlying mechanisms in oncogene-specific tumors remain poorly understood. Here, we discover that lactate dehydrogenase (LDH) B, but not the closely related LDHA, subunits of active LDH with a known function in glycolysis, noncanonically promotes ferroptosis defense in KRAS-driven lung cancer. Using murine models and human-derived tumor cell lines, we show that LDHB silencing impairs glutathione (GSH) levels and sensitizes cancer cells to blockade of either GSH biosynthesis or utilization by unleashing KRAS-specific, ferroptosis-catalyzed metabolic synthetic lethality, culminating in increased glutamine metabolism, oxidative phosphorylation (OXPHOS) and mitochondrial reactive oxygen species (mitoROS). We further show that LDHB suppression upregulates STAT1, a negative regulator of SLC7A11, thereby reducing SLC7A11-dependent GSH metabolism. Our study uncovers a previously undefined mechanism of ferroptosis resistance involving LDH isoenzymes and provides a novel rationale for exploiting oncogene-specific ferroptosis susceptibility to treat KRAS-driven lung cancer.
Collapse
Affiliation(s)
- Liang Zhao
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Haibin Deng
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Second Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
| | - Jingyi Zhang
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Nicola Zamboni
- Department of Biology, Institute of Molecular Systems Biology, Swiss Federal Institute of Technology/ETH Zürich, Zurich, Switzerland
- PHRT Swiss Multi-Omics Center, smoc.ethz.ch, Zurich, Switzerland
| | - Haitang Yang
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yanyun Gao
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Zhang Yang
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou City, Fujian, China
| | - Duo Xu
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Haiqing Zhong
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Geert van Geest
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Rémy Bruggmann
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Qinghua Zhou
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Ralph A Schmid
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| | - Thomas M Marti
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| | - Patrick Dorn
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| | - Ren-Wang Peng
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| |
Collapse
|
13
|
Ghirotto B, Gonçalves LE, Ruder V, James C, Gerasimova E, Rizo T, Wend H, Farrell M, Gerez JA, Prymaczok NC, Kuijs M, Shulman M, Hartebrodt A, Prots I, Gessner A, Zunke F, Winkler J, Blumenthal DB, Theis FJ, Riek R, Günther C, Neurath M, Gupta P, Winner B. TNF-α disrupts the malate-aspartate shuttle, driving metabolic rewiring in iPSC-derived enteric neural lineages from Parkinson's Disease patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.25.644826. [PMID: 40196623 PMCID: PMC11974853 DOI: 10.1101/2025.03.25.644826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Gastrointestinal (GI) dysfunction emerges years before motor symptoms in Parkinson's disease (PD), implicating the enteric nervous system (ENS) in early disease progression. However, the mechanisms linking the PD hallmark protein, α-synuclein (α-syn), to ENS dysfunction - and whether these mechanisms are influenced by inflammation - remains elusive. Using iPSC-derived enteric neural lineages from patients with α-syn triplications, we reveal that TNF-α increases mitochondrial-α-syn interactions, disrupts the malate-aspartate shuttle, and forces a metabolic shift toward glutamine oxidation. These alterations drive mitochondrial dysfunction, characterizing metabolic impairment under cytokine stress. Interestingly, targeting glutamate metabolism with Chicago Sky Blue 6B restores mitochondrial function, reversing TNF-α-driven metabolic disruption. Our findings position the ENS as a central player in PD pathogenesis, establishing a direct link between cytokines, α-syn accumulation, metabolic stress and mitochondrial dysfunction. By uncovering a previously unrecognized metabolic vulnerability in the ENS, we highlight its potential as a therapeutic target for early PD intervention.
Collapse
Affiliation(s)
- Bruno Ghirotto
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
- International Max Planck Research School in Physics and Medicine, Erlangen, Germany
| | - Luís Eduardo Gonçalves
- Department of Medicine 1, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Vivien Ruder
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Christina James
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Elizaveta Gerasimova
- Dental Clinic 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Tania Rizo
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
- Present address: Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, USA
| | - Holger Wend
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Michaela Farrell
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Juan Atilio Gerez
- Institute of Molecular Physical Sciences, ETH Zürich, Zürich, Switzerland
| | | | - Merel Kuijs
- Institute of Computational Biology, Helmholtz Center, Munich, Germany
- TUM, School of Computation, Information and Technology, Technical University of Munich, Germany
- TUM School of Life Sciences, Technical University of Munich, Germany
| | - Maiia Shulman
- Institute of Computational Biology, Helmholtz Center, Munich, Germany
- TUM, School of Computation, Information and Technology, Technical University of Munich, Germany
- TUM School of Life Sciences, Technical University of Munich, Germany
| | - Anne Hartebrodt
- Biomedical Network Science Lab, Department Artificial Intelligence in Biomedical Engineering , Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Iryna Prots
- Dental Clinic 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Arne Gessner
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Friederike Zunke
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Jürgen Winkler
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - David B Blumenthal
- Biomedical Network Science Lab, Department Artificial Intelligence in Biomedical Engineering , Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Center, Munich, Germany
- TUM, School of Computation, Information and Technology, Technical University of Munich, Germany
- TUM School of Life Sciences, Technical University of Munich, Germany
| | - Roland Riek
- Institute of Molecular Physical Sciences, ETH Zürich, Zürich, Switzerland
| | - Claudia Günther
- Department of Medicine 1, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Markus Neurath
- Department of Medicine 1, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Pooja Gupta
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Beate Winner
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
- Center of Rare Diseases Erlangen, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
14
|
Che Z, Yan W, Zhao Q. Extracellular Vesicles in the Mesenchymal Stem Cell/Macrophage Axis: Potential Targets for Inflammatory Treatment. Int J Mol Sci 2025; 26:2827. [PMID: 40141469 PMCID: PMC11943156 DOI: 10.3390/ijms26062827] [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: 02/21/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 03/28/2025] Open
Abstract
Mesenchymal stem cells (MSCs) have been widely used for the treatment of autoimmune and inflammatory diseases due to their pluripotent differentiation potential and immunomodulatory function. Macrophage (Mφ) polarization also acts an essential and central role in regulating inflammation, basically the dynamic balance of pro-inflammatory M1-like (M1φ) and anti-inflammatory M2-like macrophages (M2φ), affecting the occurrence and progression of inflammatory diseases. Since a pivotal molecular crosstalk between MSCs and Mφ has been elucidated using in vitro and in vivo preclinical studies, we presume that the mesenchymal stem cell/macrophages axis (MSC/Mφ axis) acts an important role in pathophysiological mechanisms of inflammatory diseases and should be the potential therapeutic target. However, the crucial effects of EVs as intercellular communicators and therapeutic agents in the MSC/Mφ axis remains explorable. Therefore, this review elaborated on the mechanisms of EVs mediating the MSC/Mφ axis regulating inflammation in-depth, hoping to provide more references for related research in the future.
Collapse
Affiliation(s)
- Zhen Che
- Experimental Orthopedics and Trauma Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany;
| | - Wenbin Yan
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University (SYSU), Guangzhou 510120, China
| | - Qun Zhao
- Experimental Orthopedics and Trauma Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany;
| |
Collapse
|
15
|
Zhu Y, Song Y, Jiang W, Zhang J, Yin L, Lin X, Lu Y, Tao D, Ma Y. A novel tumor-associated macrophage risk signature predicts prognosis and immunotherapy response in lung adenocarcinoma. Am J Cancer Res 2025; 15:876-893. [PMID: 40226479 PMCID: PMC11982730 DOI: 10.62347/squf6988] [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: 01/07/2025] [Accepted: 02/28/2025] [Indexed: 04/15/2025] Open
Abstract
OBJECTIVE To systematically characterize tumor-associated macrophage (TAM) subsets in lung adenocarcinoma (LUAD) and establish a TAM-based prognostic risk signature for LUAD patients. METHODS Single-cell RNA sequencing (scRNA-seq) and bulk transcriptomic data were integrated to identify TAM subsets linked to LUAD prognosis. Prognostic genes were screened using univariate Cox regression, refined via Least Absolute Shrinkage and Selection Operator (LASSO) regression, and used to construct a 10-gene risk signature. The signature's performance was validated in independent cohorts through receiver operating characteristic curves, Kaplan-Meier survival analysis, and a nomogram. Its predictive ability for immune checkpoint inhibitor (ICI) therapy response was assessed in the IMvigor210 and GSE78220 datasets. RESULTS Six distinct TAM subpopulations were identified, with two subsets significantly correlated with poor prognosis. The 10-gene risk signature, derived from TAM-related genes, demonstrated strong prognostic performance in both training and validation cohorts. High-risk patients exhibited markedly worse overall survival compared to low-risk patients. Additionally, the signature effectively stratified patients based on their response to anti-PD-L1 therapy, with high-risk patients exhibiting reduced clinical benefit. A nomogram combining the risk signature with clinicopathological parameters further enhanced survival prediction accuracy, supporting its clinical applicability. CONCLUSION This study established a novel TAM-based prognostic risk signature with robust predictive power for both survival outcomes and immunotherapy response in LUAD. These findings enhance our understanding of TAMs' clinical significance and provide a foundation for personalized immunotherapy strategies in LUAD.
Collapse
Affiliation(s)
- Yingchuan Zhu
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University Chengdu 610041, Sichuan, China
| | - Yue Song
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University Chengdu 610041, Sichuan, China
| | - Wenhao Jiang
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University Chengdu 610041, Sichuan, China
| | - Jingfei Zhang
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University Chengdu 610041, Sichuan, China
| | - Lan Yin
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University Chengdu 610041, Sichuan, China
| | - Xinyu Lin
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University Chengdu 610041, Sichuan, China
| | - Yilu Lu
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University Chengdu 610041, Sichuan, China
| | - Dachang Tao
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University Chengdu 610041, Sichuan, China
| | - Yongxin Ma
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University Chengdu 610041, Sichuan, China
| |
Collapse
|
16
|
Sharma G, Vela RJ, Powell L, Deja S, Fu X, Burgess SC, Malloy CR, Jessen ME, Peltz M. Metabolic and transcriptomic insights into temperature controlled hypothermic preservation of human donor hearts. J Heart Lung Transplant 2025:S1053-2498(25)01836-4. [PMID: 40081628 DOI: 10.1016/j.healun.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Heart transplantation (HT) is the gold standard for end-stage heart disease. Donor heart preservation is an important factor that influences post-transplant success. Recently, temperature-controlled storage has demonstrated reduced primary graft dysfunction compared to standard cold storage though mechanisms are poorly understood. We hypothesized that alterations in gene expression and metabolomics offer insight into improved outcomes observed with temperature-controlled storage. METHODS We conducted a comprehensive study to investigate the metabolic and transcriptomic responses of donor hearts preserved for 6 hours using a temperature-controlled hypothermic preservation (TCHP) system compared to conventional static cold storage (SCS). Metabolic assessments were carried out using high-resolution 1H and 31P nuclear magnetic resonance (NMR), and liquid chromatography/mass spectrometry (LC-MS) analysis on tissues obtained from various cardiac regions. Lactate, alanine, adenosine triphosphate (ATP), adenosine diphosphate (ADP), adenosine monophosphate (AMP), nicotinamide adenine dinucleotide (NAD), reduced nicotinamide adenine dinucleotide (NADH), phosphocreatine, and inorganic phosphate were measured, and metabolite ratios were calculated. Transcriptomic profiling was conducted using high throughput RNA sequencing followed by bioinformatic analysis to explore gene expression changes associated with different preservation methods. RESULTS Metabolic analyses revealed largely similar profiles between hearts preserved with TCHP and SCS. Energy metabolite ratios were comparable between preservation methods. Transcriptomic analysis unveiled a high correlation between preservation methods but also showed differential gene expression in energy metabolism and inflammation/immune-related pathways. CONCLUSIONS Our study demonstrates that TCHP maintains similar high-energy phosphate reserves to SCS but leads to alterations in gene expression of several metabolic and immunomodulatory pathways. These findings may offer important insight into reduced primary graft dysfunction observed in TCHP- hearts.
Collapse
Affiliation(s)
- Gaurav Sharma
- Department of Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, Texas; Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ryan J Vela
- Department of Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - LaShondra Powell
- Department of Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Stanislaw Deja
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Xiaorong Fu
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Shawn C Burgess
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Craig R Malloy
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Michael E Jessen
- Department of Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Matthias Peltz
- Department of Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
17
|
Wang Y, Cui Y, Liu X, Liang S, Zhong L, Guo R. Cathepsin K inhibition promotes efficient differentiation of human embryonic stem cells to mature cardiomyocytes by mediating glucolipid metabolism and cellular energy homeostasis. Stem Cell Res Ther 2025; 16:118. [PMID: 40045408 PMCID: PMC11883997 DOI: 10.1186/s13287-025-04231-7] [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: 10/21/2024] [Accepted: 02/14/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND AND AIM Generation of functional cardiomyocytes from human pluripotent stem cells (hPSCs) offers promising applications for cardiac regenerative medicine. Proper control of pluripotency and differentiation is vital for generating high-quality cardiomyocytes and repairing damaged myocardium. Cathepsin K, a lysosomal cysteine protease, is a potential target for cardiovascular disease treatment; however, its role in cardiomyocyte differentiation and regeneration is unclear. This study aims to investigate the effects and mechanisms of cathepsin K inhibition on the differentiation of human embryonic stem cell-induced cardiomyocytes (hESC-CMs) and myocardial generation. METHODS We cultured H9-hESCs in CDM3 medium to induce myocardial differentiation, adding cathepsin K inhibitor II (1 μM) on days 2, 5 and 8, respectively. Cells were observed and collected 48 h after each treatment. The morphology and contractile clusters of H9-hESCs were tracked with microscopy and video recording. Pluripotency and cardiac markers were assessed at each stage of differentiation. We also examined glucose and lipid metabolism, mitochondrion-related markers, apoptosis and autophagy. RESULTS CDM3 medium effectively differentiated high-density H9-hESCs into mature, spontaneously contracting cardiomyocytes. Cathepsin K inhibition accelerates the differentiation of H9-hESCs into cardiac mesoderm and cardiac precursor cells (CPCs) by reducing apoptosis, decreasing glycolysis and fatty acid metabolism at the early and middle stages, and subsequently facilitate the development and differentiation of cardiomyocytes by enhancing glucolipid metabolism and oxidative phosphorylation at the late stage. Meanwhile, cathepsin K inhibition enhanced mitochondrial function and lysosome-related gene transcription during the differentiation process. CONCLUSION Our study highlights the potential of cathepsin K inhibition for renewable cardiomyocytes and suggests exploring metabolic pathways and signaling to improve cardiac regeneration and organoid development.
Collapse
Affiliation(s)
- Ying Wang
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China
| | - Yang Cui
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China
| | - Xiaoyu Liu
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China
| | - Shengxian Liang
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China
| | - Li Zhong
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Rui Guo
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China.
- The Key Laboratory of Zoological Systematics and Application, College of Life Sciences, Hebei University, Baoding, 071002, China.
| |
Collapse
|
18
|
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
|
19
|
Li W, Li Y, Xiao L, Xie Z, Peng J, Huang W, Li X, Meng Y. Micheliolide attenuates sepsis-induced acute lung injury by suppressing mitochondrial oxidative stress and PFKFB3-driven glycolysis. J Transl Med 2025; 23:181. [PMID: 39953547 PMCID: PMC11829335 DOI: 10.1186/s12967-024-05906-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 11/19/2024] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Sepsis is a potentially fatal condition with a significant risk of death. Acute lung injury (ALI) is a life-threatening complication of sepsis, and the inflammatory response plays a critical role in sepsis-induced ALI. The protective effects of micheliolide (MCL) against renal fibrosis and leukemia have been demonstrated, but the precise underlying mechanisms remain unclear. METHODS In vitro, lipopolysaccharides (LPS) and interferon-gamma (IFN-γ) were used to stimulate RAW264.7 cells and bone marrow-derived macrophages (BMDMs) to investigate the protective effect of MCL on sepsis-induced ALI. Cecal ligation and puncture (CLP) models were constructed in mice to induce ALI in vivo. The expression of inflammatory factors, macrophage polarization markers, and the glycolysis-related enzyme 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) were measured in vivo. Mitochondrial function, oxidative stress, and mitochondrial-related proteins were evaluated in vitro. RESULTS MCL inhibited CLP-induced ALI, as evidenced by improvements in proinflammatory factor levels, lung wet/dry ratios, and histopathological findings. In vitro, MCL treatment significantly suppressed LPS + IFN-γ-induced M1-type polarization of RAW264.7 cells and BMDMs, as well as the production of inflammatory factors and oxidative stress. Mechanistic experiments revealed that MCL suppresses PFKFB3-driven glycolysis to reduce inflammation and activates the mitochondrial unfolded protein response (UPRmt) to alleviate mitochondrial stress. However, the therapeutic effect of MCL was diminished when PFKFB3 was overexpressed in cells. CONCLUSION This study is the first to demonstrate that MCL attenuates sepsis-induced ALI by reducing M1-type macrophage polarization. Its therapeutic effect is closely related to the suppression of oxidative stress and PFKFB3-driven glycolysis.
Collapse
Affiliation(s)
- Wenhan Li
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China
| | - Yuhan Li
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Linjie Xiao
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China
| | - Zhanzhan Xie
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China
| | - Jun Peng
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China
| | - Wenhui Huang
- Department of Respiratory and Critical Care Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, China
| | - Xu Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China.
| | - Ying Meng
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China.
| |
Collapse
|
20
|
Wu J, Lu L, Dai B, Yu A. Unraveling the role of LDHA and VEGFA in oxidative stress: A pathway to therapeutic interventions in cerebral aneurysms. BIOMOLECULES & BIOMEDICINE 2025; 25:360-374. [PMID: 38829380 PMCID: PMC11734822 DOI: 10.17305/bb.2024.10510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 06/05/2024]
Abstract
Cerebral aneurysms (CA) are critical conditions often associated with oxidative stress in vascular endothelial cells (VECs). The enzyme lactate dehydrogenase A (LDHA) plays a crucial role in glycolysis and lactate metabolism, processes implicated in the pathogenesis of aneurysms. Understanding these molecular mechanisms can inform the development of novel therapeutic targets. This study investigated the role of lactate metabolism and lactate-related genes, particularly LDHA and vascular endothelial growth factor A (VEGFA) genes, in VECs during oxidative stress. Using the GSE26969 dataset, we identified differential expression of lactate-related genes and performed functional enrichment analysis, revealing significant associations with glycolysis and lactate metabolic pathways. To induce oxidative stress, VECs were treated with H2O2, and the expression of LDHA and VEGFA was analyzed using quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting (WB) assays. Under oxygen-glucose deprivation/reperfusion (OGD/R) conditions, the effects of LDHA overexpression and VEGFA knockdown on cell viability and apoptosis were evaluated. Immunoprecipitation combined with western blotting was used to detect the lactylation status of LDHA following OGD/R stimulation and treatment with lactic acid (LA) and 2-deoxyglucose (2-DG). Our results indicated that oxidative stress modulates LDHA expression, glucose uptake, and lactate production, suggesting a metabolic shift towards glycolysis. LDHA overexpression improved cell survival and reduced apoptosis, while VEGFA knockdown had the opposite effect. Additionally, 2-DG treatment reduced LDHA lactylation and apoptosis. Our findings demonstrated that LDHA plays a critical role in the oxidative stress response of VECs, highlighting the potential therapeutic value of targeting glycolysis in CA. This study contributes to the understanding of metabolic adaptations in vascular pathologies and suggests new avenues for therapeutic intervention in CA management.
Collapse
Affiliation(s)
- Jiaying Wu
- Department of Neurology, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lixia Lu
- Department of Neurology, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Beibei Dai
- Department of Ultrasound, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Aiyong Yu
- Department of Neurology, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Xie B, Li J, Lou Y, Chen Q, Yang Y, Zhang R, Liu Z, He L, Cheng Y. Reprogramming macrophage metabolism following myocardial infarction: A neglected piece of a therapeutic opportunity. Int Immunopharmacol 2024; 142:113019. [PMID: 39217876 DOI: 10.1016/j.intimp.2024.113019] [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: 05/11/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Given the global prevalence of myocardial infarction (MI) as the leading cause of mortality, there is an urgent need to devise novel strategies that target reducing infarct size, accelerating cardiac tissue repair, and preventing detrimental left ventricular (LV) remodeling. Macrophages, as a predominant type of innate immune cells, undergo metabolic reprogramming following MI, resulting in alterations in function and phenotype that significantly impact the progression of MI size and LV remodeling. This article aimed to delineate the characteristics of macrophage metabolites during reprogramming in MI and elucidate their targets and functions in cardioprotection. Furthermore, we summarize the currently proposed regulatory mechanisms of macrophage metabolic reprogramming and identify the regulators derived from endogenous products and natural small molecules. Finally, we discussed the challenges of macrophage metabolic reprogramming in the treatment of MI, with the goal of inspiring further fundamental and clinical research into reprogramming macrophage metabolism and validating its potential therapeutic targets for MI.
Collapse
Affiliation(s)
- Baoping Xie
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Provincial Key Laboratory of Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Jiahua Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China
| | - Yanmei Lou
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China
| | - Qi Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China
| | - Ying Yang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China
| | - Rong Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China
| | - Zhongqiu Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China.
| | - Liu He
- Department of Endocrinology, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong 510006, China.
| | - Yuanyuan Cheng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China.
| |
Collapse
|
22
|
O'Hern C, Caywood S, Aminova S, Kiselev A, Volmert B, Wang F, Sewavi ML, Cao W, Dionise M, Muniyandi P, Popa M, Basrai H, Skoric M, Boulos G, Huang A, Nuñez-Regueiro I, Chalfoun N, Park S, Ashammakhi N, Zhou C, Contag C, Aguirre A. Human heart assembloids with autologous tissue-resident macrophages recreate physiological immuno-cardiac interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.13.623051. [PMID: 39677610 PMCID: PMC11642760 DOI: 10.1101/2024.11.13.623051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Interactions between the developing heart and the embryonic immune system are essential for proper cardiac development and maintaining homeostasis, with disruptions linked to various diseases. While human pluripotent stem cell (hPSC)-derived organoids are valuable models for studying human organ function, they often lack critical tissue-resident immune cells. Here, we introduce an advanced human heart assembloid model, termed hHMA (human heart-macrophage assembloid), which fully integrates autologous cardiac tissue- resident macrophages (MPs) with pre-existing human heart organoids (hHOs). Through multi-omic analyses, we confirmed that these MPs are phenotypically similar to embryonic cardiac tissue-resident MPs and remain viable in the assembloids over time. The inclusion of MPs significantly impacts hHMA development, influencing cardiac cellular composition, boosting cellular communication, remodeling the extracellular matrix, promoting ventricular morphogenesis, and enhancing sarcomeric maturation. Our findings indicate that MPs contribute to homeostasis via efferocytosis, integrate into the cardiomyocyte electrical system, and support catabolic metabolism. To demonstrate the versatility of this model, we developed a platform to study cardiac arrhythmias by chronic exposure to pro-inflammatory factors linked to arrhythmogenesis in clinical settings, successfully replicating key features of inflammasome-mediated atrial fibrillation. Overall, this work introduces a robust platform for examining the role of immune cells in cardiac development, disease mechanisms, and drug discovery, bridging the gap between in vitro models and human physiology. These findings offer insights into cardiogenesis and inflammation-driven heart disease, positioning the hHMA system as an invaluable tool for future cardiovascular research and therapeutic development.
Collapse
|
23
|
Cao S, Wang S, Luo H, Guo J, Xuan L, Sun L. The effect of macrophage-cardiomyocyte interactions on cardiovascular diseases and development of potential drugs. Mol Biol Rep 2024; 51:1056. [PMID: 39417949 DOI: 10.1007/s11033-024-09944-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
The interaction between macrophages and cardiomyocytes plays an important role not only in maintaining cardiac homeostasis, but also in the development of many cardiovascular diseases (CVDs), such as myocardial infarction (MI) and heart failure (HF). In addition to supporting cardiomyocytes, macrophages and cardiomyocytes have a close and complex relationship. By studying their cross-talk, we can better understand novel mechanisms and target pathogenic mechanisms, and improve the treatment of CVDs. We review macrophage-cardiomyocyte communication through connexin 43 (Cx43)-containing gap junctions (GJs) directly, secreted protein factors indirectly, and discuss the implications of these interactions in cardiac homeostasis and the development of various CVDs, including MI, HF, arrhythmia, cardiac fibrosis and myocarditis. In this section, we review various drugs that work by modulating cytokines or other proteins to reduce inflammation in CVDs. The clinical findings from targeting inflammation in CVDs are also discussed. Additionally, we examine the challenges and opportunities for improving our understanding of macrophage-cardiomyocyte coupling as it relates to pathophysiological disease processes, extending our research scope, and helping identify new molecular targets and improve the effectiveness of existing therapies.
Collapse
Affiliation(s)
- Shoupeng Cao
- Department of Pharmacology, College of Pharmacy, Harbin Medical University (Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Shengjie Wang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University (Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Huishan Luo
- Department of Pharmacology, College of Pharmacy, Harbin Medical University (Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Jianjun Guo
- Department of Pharmacology, College of Pharmacy, Harbin Medical University (Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Lina Xuan
- Department of Pharmacology, College of Pharmacy, Harbin Medical University (Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang, China.
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medicial University, Harbin, 157 Baojian Road, Nangang District, 150081, heilongjiang, China.
| | - Lihua Sun
- Department of Pharmacology, College of Pharmacy, Harbin Medical University (Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang, China.
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medicial University, Harbin, 157 Baojian Road, Nangang District, 150081, heilongjiang, China.
| |
Collapse
|
24
|
He Y, Zhang Y, Zhu S, Liu YF, Liu S, Xu YJ. Monomethyl Branched-Chain Fatty Acids Suppress M1 Macrophage Polarization via FABP4/PPAR-γ Signaling Pathway. Mol Nutr Food Res 2024; 68:e2400310. [PMID: 39318069 DOI: 10.1002/mnfr.202400310] [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: 04/28/2024] [Revised: 08/27/2024] [Indexed: 09/26/2024]
Abstract
SCOPE Monomethyl-branched chain fatty acids (mmBCFAs) are found in a variety of food sources and are of great interest due to their potent antiinflammatory properties. However, most of the current researches have concentrated on the relationship between mmBCFAs and intestinal inflammation, and there is a large gap in the biological mechanisms involved behind their antiinflammatory effects. METHODS AND RESULTS The present study examines the role of mmBCFAs in modulating macrophage polarization. The results demonstrate that iso-C16:0 significantly inhibits macrophages M1 proinflammatory polarization through regulating FABP4/PPAR-γ pathway. Proteomics and molecular biology experiments verify that metabolic reprogramming is involved in the inhibition of M1 macrophage, referring to the upregulation of fatty acid oxidation, TCA cycle, and oxidative phosphorylation, as well as downregulation of glycolytic flux. CONCLUSION In summary, this study offers a novel perspective on the antiinflammatory effects mediated by mmBCFAs.
Collapse
Affiliation(s)
- Yuan He
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, National Engineering Reacher Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, No. 1800, Lihu Road, Wuxi, Jiangsu, 214122, People's Republic of China
| | - Yu Zhang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, National Engineering Reacher Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, No. 1800, Lihu Road, Wuxi, Jiangsu, 214122, People's Republic of China
| | - Shuang Zhu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, National Engineering Reacher Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, No. 1800, Lihu Road, Wuxi, Jiangsu, 214122, People's Republic of China
| | - Yuan-Fa Liu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, National Engineering Reacher Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, No. 1800, Lihu Road, Wuxi, Jiangsu, 214122, People's Republic of China
| | - Sha Liu
- Wuxi Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu, 214071, China
| | - Yong-Jiang Xu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, National Engineering Reacher Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, No. 1800, Lihu Road, Wuxi, Jiangsu, 214122, People's Republic of China
| |
Collapse
|
25
|
Chen YX, Zhao AR, Wei TW, Wang H, Wang LS. Progress of Mitochondrial Function Regulation in Cardiac Regeneration. J Cardiovasc Transl Res 2024; 17:1097-1105. [PMID: 38647881 DOI: 10.1007/s12265-024-10514-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024]
Abstract
Heart failure and myocardial infarction, global health concerns, stem from limited cardiac regeneration post-injury. Myocardial infarction, typically caused by coronary artery blockage, leads to cardiac muscle cell damage, progressing to heart failure. Addressing the adult heart's minimal self-repair capability is crucial, highlighting cardiac regeneration research's importance. Studies reveal a metabolic shift from anaerobic glycolysis to oxidative phosphorylation in neonates as a key factor in impaired cardiac regeneration, with mitochondria being central. The heart's high energy demands rely on a robust mitochondrial network, essential for cellular energy, cardiac health, and regenerative capacity. Mitochondria's influence extends to redox balance regulation, signaling molecule interactions, and apoptosis. Changes in mitochondrial morphology and quantity also impact cardiac cell regeneration. This article reviews mitochondria's multifaceted role in cardiac regeneration, particularly in myocardial infarction and heart failure models. Understanding mitochondrial function in cardiac regeneration aims to enhance myocardial infarction and heart failure treatment methods and insights.
Collapse
Affiliation(s)
- Yi-Xi Chen
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - An-Ran Zhao
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Tian-Wen Wei
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Hao Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Lian-Sheng Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
26
|
Huang H, Chen H, Yao Y, Lou X. Branched-chain amino acids supplementation induces insulin resistance and pro-inflammatory macrophage polarization via INFGR1/JAK1/STAT1 signal pathway. Mol Med 2024; 30:149. [PMID: 39267003 PMCID: PMC11391606 DOI: 10.1186/s10020-024-00894-9] [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: 04/28/2024] [Accepted: 08/05/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Obesity is a global epidemic, and the low-grade chronic inflammation of adipose tissue in obese individuals can lead to insulin resistance and type 2 diabetes. Adipose tissue macrophages (ATMs) are the main source of pro-inflammatory cytokines in adipose tissue, making them an important target for therapy. While branched-chain amino acids (BCAA) have been strongly linked to obesity and type 2 diabetes in humans, the relationship between BCAA catabolism and adipose tissue inflammation is unclear. This study aims to investigate whether disrupted BCAA catabolism influences the function of adipose tissue macrophages and the secretion of pro-inflammatory cytokines in adipose tissue, and to determine the underlying mechanism. This research will help us better understand the role of BCAA catabolism in adipose tissue inflammation, obesity, and type 2 diabetes. METHODS In vivo, we examined whether the BCAA catabolism in ATMs was altered in high-fat diet-induced obesity mice, and if BCAA supplementation would influence obesity, glucose tolerance, insulin sensitivity, adipose tissue inflammation and ATMs polarization in mice. In vitro, we isolated ATMs from standard chow and high BCAA-fed group mice, using RNA-sequencing to investigate the potential molecular pathway regulated by BCAA accumulation. Finally, we performed targeted gene silence experiment and used immunoblotting assays to verify our findings. RESULTS We found that BCAA catabolic enzymes in ATMs were influenced by high-fat diet induced obesity mice, which caused the accumulation of both BCAA and its downstream BCKA. BCAA supplementation will cause obesity and insulin resistance compared to standard chow (STC) group. And high BCAA diet will induce pro-inflammatory cytokines including Interlukin-1beta (IL-1β), Tumor Necrosis Factor alpha (TNF-α) and monocyte chemoattractant protein-1 (MCP-1) secretion in adipose tissue as well as promoting ATMs M1 polarization (pro-inflammatory phenotype). Transcriptomic analysis revealed that a high BCAA diet would activate IFNGR1/JAK1/STAT1 pathway, and IFNGR1 specific silence can abolish the effect of BCAA supplementation-induced inflammation and ATMs M1 polarization. CONCLUSIONS The obesity mice model reveals the catabolism of BCAA was disrupted which will cause the accumulation of BCAA, and high-level BCAA will promote ATMs M1 polarization and increase the pro-inflammatory cytokines in adipose tissue which will cause the insulin resistance in further. Therefore, reducing the circulating level of BCAA can be a therapeutic strategy in obesity and insulin resistance patients.
Collapse
Affiliation(s)
- Huaying Huang
- Department of Endocrinology and Metabolism, JinHua Municipal Central Hospital, No. 365, Renmin East Road, Wucheng District, Jinhua, Zhejiang, China
| | - Heye Chen
- Department of Endocrinology and Metabolism, JinHua Municipal Central Hospital, No. 365, Renmin East Road, Wucheng District, Jinhua, Zhejiang, China
| | - Yu Yao
- Department of Neurology, JinHua Municipal Central Hospital, Jinhua, 321000, Zhejiang, China
| | - Xueyong Lou
- Department of Endocrinology and Metabolism, JinHua Municipal Central Hospital, No. 365, Renmin East Road, Wucheng District, Jinhua, Zhejiang, China.
| |
Collapse
|
27
|
Liu X, Wang B. Histone lactylation regulates autophagy of hyperplastic scar fibroblasts by inhibiting the transcriptional activity of phosphatase and tensin homologue. Wound Repair Regen 2024; 32:725-734. [PMID: 38764180 DOI: 10.1111/wrr.13188] [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: 01/03/2024] [Revised: 04/18/2024] [Accepted: 05/01/2024] [Indexed: 05/21/2024]
Abstract
Hyperplastic scar (HS) is an overreaction of tissue to skin injury caused by local fibroblast proliferation and excessive collagen production. Histone posttranslational modification patterns are important epigenetic processes that control various biological activities. This study was designed to investigate the effects of histone lactylation on HS and the underlying mechanism. Western blot was used to analyse the lactylation level in HS patients and fibroblasts (HSFs). In vitro experiments, western blot, cell counting kit-8, and immunofluorescence staining were performed to detect the collagen level, cell viability, and autophagy, respectively. The relationship between snai2 (SLUG) and phosphatase and tensin homologue (PTEN) was assessed by RNA immunoprecipitation and dual-luciferase reporter assays. The results showed that the histone lactylation level was upregulated in HS tissues and HSFs. HSFs showed increased collagen production and cell viability, and decreased autophagy. Silencing of lactate dehydrogenase A (LDHA) promoted the transcription of PTEN by inhibiting SLUG, thus promoting autophagy. Knockdown of LDHA inhibited collagen deposition and cell viability, and increased autophagy in HSFs, and the results were reversed after PTEN inhibition. In summary, histone lactylation inhibited the transcription activity of PTEN by promoting SLUG, thereby suppressing autophagy and promoting collagen deposition and cell viability of HSFs, which might provide effective therapeutic strategies in HS.
Collapse
Affiliation(s)
- Xiaosong Liu
- Department of Surgery, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Biao Wang
- Department of Surgery, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
28
|
Zhu W, Guo S, Sun J, Zhao Y, Liu C. Lactate and lactylation in cardiovascular diseases: current progress and future perspectives. Metabolism 2024; 158:155957. [PMID: 38908508 DOI: 10.1016/j.metabol.2024.155957] [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: 12/06/2023] [Revised: 06/10/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024]
Abstract
Cardiovascular diseases (CVDs) are often linked to structural and functional impairments, such as heart defects and circulatory dysfunction, leading to compromised peripheral perfusion and heightened morbidity risks. Metabolic remodeling, particularly in the context of cardiac fibrosis and inflammation, is increasingly recognized as a pivotal factor in the pathogenesis of CVDs. Metabolic syndromes further predispose individuals to these conditions, underscoring the need to elucidate the metabolic underpinnings of CVDs. Lactate, a byproduct of glycolysis, is now recognized as a key molecule that connects cellular metabolism with the regulation of cellular activity. The transport of lactate between different cells is essential for metabolic homeostasis and signal transduction. Disruptions to lactate dynamics are implicated in various CVDs. Furthermore, lactylation, a novel post-translational modification, has been identified in cardiac cells, where it influences protein function and gene expression, thereby playing a significant role in CVD pathogenesis. In this review, we summarized recent advancements in understanding the role of lactate and lactylation in CVDs, offering fresh insights that could guide future research directions and therapeutic interventions. The potential of lactate metabolism and lactylation as innovative therapeutic targets for CVD is a promising avenue for exploration.
Collapse
Affiliation(s)
- Wengen Zhu
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China; Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou 510080, PR China.
| | - Siyu Guo
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China; Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou 510080, PR China
| | - Junyi Sun
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China
| | - Yudan Zhao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430023, PR China.
| | - Chen Liu
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China; Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou 510080, PR China.
| |
Collapse
|
29
|
Jin J, Xia X, Ruan C, Luo Z, Yang Y, Wang D, Qin Y, Li D, Zhang Y, Hu Y, Lei P. GAPDH-Silence Microsphere via Reprogramming Macrophage Metabolism and eradicating Bacteria for Diabetic infection bone regeneration. J Nanobiotechnology 2024; 22:517. [PMID: 39210435 PMCID: PMC11361104 DOI: 10.1186/s12951-024-02787-9] [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: 06/19/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024] Open
Abstract
Macrophage metabolism dysregulation, which is exacerbated by persistent stimulation in infectious and inflammatory diseases, such as diabetic infectious bone defects (DIBD), eventually leads to the failure of bone repair. Here, we have developed an injectable, macrophage-modulated GAPDH-Silence drug delivery system. This microsphere comprises chondroitin sulfate methacrylate (CM) and methacrylated gelatin (GM), while the dimethyl fumarate (DMF)-loaded liposome (D-lip) is encapsulated within the microsphere (CM@GM), named D-lip/CM@GM. Triggered by the over-expressed collagenase in DIBD, the microspheres degrade and release the encapsulated D-lip. D-lip could modulate metabolism by inhibiting GAPDH, which suppresses the over-activation of glycolysis, thus preventing the inflammatory response of macrophages in vitro. While beneficial for macrophages, D-lip/CM@GM is harmful to bacteria. GAPDH, while crucial for glycolysis of staphylococcal species (S. aureus), can be effectively countered by D-lip/CM@GM. We are utilizing existing drugs in innovative ways to target central metabolism for effective eradication of bacteria. In the DIBD model, our results confirmed that the D-lip/CM@GM enhanced bacteria clearance and reprogrammed dysregulated metabolism, thereby significantly improving bone regeneration. In conclusion, this GAPDH-Silence microsphere system may provide a viable strategy to promote diabetic infection bone regeneration.
Collapse
Affiliation(s)
- Jiale Jin
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xiaowei Xia
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
| | - Chengxin Ruan
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zhiyuan Luo
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yiqi Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Dongyu Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yifang Qin
- Department of Endocrinology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310052, China
| | - Dongdong Li
- Department of Orthopedic Surgery, Ningxia Medical University, Yinchuan, 200233, China
| | - Yong Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China.
| | - Yihe Hu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Pengfei Lei
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
30
|
Moreira DC, Hermes-Lima M. Dynamics of Redox Metabolism during Complete Metamorphosis of Insects: Insights from the Sunflower Caterpillar Chlosyne lacinia (Lepidoptera). Antioxidants (Basel) 2024; 13:959. [PMID: 39199204 PMCID: PMC11351957 DOI: 10.3390/antiox13080959] [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: 07/02/2024] [Revised: 08/01/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024] Open
Abstract
Complete insect metamorphosis requires substantial metabolic and physiological adjustments. Although oxidative stress has been implicated in metamorphosis, details on redox metabolism during larva-to-pupa and pupa-to-adult remain scarce. This study explores redox metabolism during metamorphosis of a lepidopteran (Chlosyne lacinia), focusing on core metabolism, antioxidant systems and oxidative stress. The larva-to-pupa transition was characterized by increased lactate dehydrogenase and glutathione peroxidase (GPX) activities, coupled with depletion of reduced glutathione (GSH), high disulfide-to-total-glutathione ratio (GSSG/tGSH), and increased lipid peroxidation. As metamorphosis progressed, metabolic enzyme activities, citrate synthase and glucose 6-phosphate dehydrogenase increased, indicating heightened oxidative metabolism associated with adult development. Concurrently, GSH and GPX levels returned to larval levels and GSSG/tGSH reached its most reduced state right before adult emergence. Adult emergence was marked by a further increase in oxidative metabolism, accompanied by redox imbalance and enhanced antioxidant mechanisms. These findings highlight a fluctuation in redox balance throughout metamorphosis, with periods of oxidative eustress followed by compensatory antioxidant responses. This study is the first to identify concurrent changes in metabolism, antioxidants, redox balance and oxidative stress throughout metamorphosis. Our findings extend knowledge on redox metabolism adjustments and highlight redox adaptations and oxidative stress as natural components of complete insect metamorphosis.
Collapse
Affiliation(s)
- Daniel C. Moreira
- Research Center in Morphology and Applied Immunology, Faculty of Medicine, University of Brasilia, Brasilia 70910-900, Brazil
- Cell Biology Department, Biological Sciences Institute, University of Brasilia, Brasilia 70910-900, Brazil;
| | - Marcelo Hermes-Lima
- Cell Biology Department, Biological Sciences Institute, University of Brasilia, Brasilia 70910-900, Brazil;
| |
Collapse
|
31
|
Wu D, Wang S, Wang F, Zhang Q, Zhang Z, Li X. Lactate dehydrogenase A (LDHA)-mediated lactate generation promotes pulmonary vascular remodeling in pulmonary hypertension. J Transl Med 2024; 22:738. [PMID: 39103838 PMCID: PMC11302077 DOI: 10.1186/s12967-024-05543-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/26/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND High levels of lactate are positively associated with prognosis and mortality in pulmonary hypertension (PH). Lactate dehydrogenase A (LDHA) is a key enzyme for the production of lactate. This study is undertaken to investigate the role and molecular mechanisms of lactate and LDHA in PH. METHODS Lactate levels were measured by a lactate assay kit. LDHA expression and localization were detected by western blot and Immunofluorescence. Proliferation and migration were determined by CCK8, western blot, EdU assay and scratch-wound assay. The right heart catheterization and right heart ultrasound were measured to evaluate cardiopulmonary function. RESULTS In vitro, we found that lactate promoted proliferation and migration of pulmonary artery smooth muscle cells (PASMCs) in an LDHA-dependent manner. In vivo, we found that LDHA knockdown reduced lactate overaccumulation in the lungs of mice exposed to hypoxia. Furthermore, LDHA knockdown ameliorated hypoxia-induced vascular remodeling and right ventricular dysfunction. In addition, the activation of Akt signaling by hypoxia was suppressed by LDHA knockdown both in vivo and in vitro. The overexpression of Akt reversed the inhibitory effect of LDHA knockdown on proliferation in PASMCs under hypoxia. Finally, LDHA inhibitor attenuated vascular remodeling and right ventricular dysfunction in Sugen/hypoxia mouse PH model, Monocrotaline (MCT)-induced rat PH model and chronic hypoxia-induced mouse PH model. CONCLUSIONS Thus, LDHA-mediated lactate production promotes pulmonary vascular remodeling in PH by activating Akt signaling pathway, suggesting the potential role of LDHA in regulating the metabolic reprogramming and vascular remodeling in PH.
Collapse
Affiliation(s)
- Daiqian Wu
- Department of Cardiology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu Cardiovascular Disease Research Institute, Chengdu, 610014, PR China
| | - Shuo Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, 400042, PR China
| | - Fengxian Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, 400042, PR China
| | - Qing Zhang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, PR China
| | - Zhen Zhang
- Department of Cardiology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu Cardiovascular Disease Research Institute, Chengdu, 610014, PR China.
| | - Xingbing Li
- Department of Cardiology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, PR China.
| |
Collapse
|
32
|
Yu T, Wei Q, Tang Y, Cai L, Chen B, Yang M. A novel long non-coding RNA linc-93.2 participates in bisphenol induced oxidative stress and macrophage polarization in red common carp (Cyprinus carpio). FISH & SHELLFISH IMMUNOLOGY 2024; 151:109716. [PMID: 38909636 DOI: 10.1016/j.fsi.2024.109716] [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: 05/07/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024]
Abstract
Previous studies show that bisphenol A (BPA) and its analogs induce oxidative stress and promote inflammatory response. However, the key molecules in regulating this process remain unclear. Here, we report significant inductive effects of BPA and bisphenol AF (BPAF) on a newly found long non-coding RNA linc-93.2 accompanied by oxidative stress and activation of pro-inflammatory pathways in treated fish and fish primary macrophages. Silencing linc-93.2 in fish primary macrophages in vitro or fish in vivo significantly promotes the expression of anti-oxidative stress-related genes and anti-inflammatory cytokines. This inhibition of pro-inflammatory cytokine expression, showing cell status disruption towards to M2 polarization. Followed by exposure to BPA or BPAF, silencing linc-93.2 in vitro or in vivo significantly attenuates the increased production of reactive oxygen species and malondialdehyde level aroused by bisphenol treatment, possibly owing to the enhancement of total antioxidant capacity observed in cells and tissue after linc-93.2 knockdown. RNA-sequencing further revealed regulation of nuclear factor-kappa b (NF-κB) in linc-93.2's downstream network, combining with our previous observation on the upstream regulation of linc-93.2 via NF-κB, which together suggest a critical role of linc-93.2 in promoting NF-κB positive feedback loop that may be an important molecular event initiating the immunotoxicity of bisphenols.
Collapse
Affiliation(s)
- Ting Yu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China; Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, 361005, China
| | - Qing Wei
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Yiran Tang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Ling Cai
- Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, 361005, China.
| | - Bei Chen
- Fisheries Research Institute of Fujian, Key Laboratory of Cultivation and High-Value Utilization of Marine Organisms in Fujian Province, Xiamen, 361013, China
| | - Ming Yang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
33
|
Yan L, Wang J, Cai X, Liou Y, Shen H, Hao J, Huang C, Luo G, He W. Macrophage plasticity: signaling pathways, tissue repair, and regeneration. MedComm (Beijing) 2024; 5:e658. [PMID: 39092292 PMCID: PMC11292402 DOI: 10.1002/mco2.658] [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: 03/03/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 08/04/2024] Open
Abstract
Macrophages are versatile immune cells with remarkable plasticity, enabling them to adapt to diverse tissue microenvironments and perform various functions. Traditionally categorized into classically activated (M1) and alternatively activated (M2) phenotypes, recent advances have revealed a spectrum of macrophage activation states that extend beyond this dichotomy. The complex interplay of signaling pathways, transcriptional regulators, and epigenetic modifications orchestrates macrophage polarization, allowing them to respond to various stimuli dynamically. Here, we provide a comprehensive overview of the signaling cascades governing macrophage plasticity, focusing on the roles of Toll-like receptors, signal transducer and activator of transcription proteins, nuclear receptors, and microRNAs. We also discuss the emerging concepts of macrophage metabolic reprogramming and trained immunity, contributing to their functional adaptability. Macrophage plasticity plays a pivotal role in tissue repair and regeneration, with macrophages coordinating inflammation, angiogenesis, and matrix remodeling to restore tissue homeostasis. By harnessing the potential of macrophage plasticity, novel therapeutic strategies targeting macrophage polarization could be developed for various diseases, including chronic wounds, fibrotic disorders, and inflammatory conditions. Ultimately, a deeper understanding of the molecular mechanisms underpinning macrophage plasticity will pave the way for innovative regenerative medicine and tissue engineering approaches.
Collapse
Affiliation(s)
- Lingfeng Yan
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Jue Wang
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Xin Cai
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Yih‐Cherng Liou
- Department of Biological SciencesFaculty of ScienceNational University of SingaporeSingaporeSingapore
- National University of Singapore (NUS) Graduate School for Integrative Sciences and EngineeringNational University of SingaporeSingaporeSingapore
| | - Han‐Ming Shen
- Faculty of Health SciencesUniversity of MacauMacauChina
| | - Jianlei Hao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and TreatmentZhuhai Institute of Translational MedicineZhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University)Jinan UniversityZhuhaiGuangdongChina
- The Biomedical Translational Research InstituteFaculty of Medical ScienceJinan UniversityGuangzhouGuangdongChina
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospitaland West China School of Basic Medical Sciences and Forensic MedicineSichuan University, and Collaborative Innovation Center for BiotherapyChengduChina
| | - Gaoxing Luo
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Weifeng He
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| |
Collapse
|
34
|
Cao X, Luo B, Mu Y, Wang C, Lu R, Yao Y, Chen S. The regulatory effect of TiO 2 nanotubes loaded with graphene oxide on macrophage polarization in an inflammatory environment. BMC Oral Health 2024; 24:824. [PMID: 39033148 PMCID: PMC11265100 DOI: 10.1186/s12903-024-04608-9] [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: 01/31/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Excessive inflammation is a major cause of implant failure. The surface morphology, hydrophilicity, and loading of biomaterials are major properties modulating anti-inflammatory macrophage activation. This paper investigates the regulatory effects of modifying the surface of Titanium dioxide nanotubes (TNTs) with graphene oxide (GO) on the polarization of mouse monocyte macrophages (RAW264.7). METHODS TNT was produced by the anodic oxidation of titanium. GO was subsequently electrodeposited on the TNT to obtain a TNT-GO composite. The samples were characterised through scanning electron microscopy (SEM), Raman spectroscopy, and X-ray diffraction. RAW264.7 cells were separately seeded onto the surface of three groups of samples: pure Ti, TNT, and TNT-GO. Under the condition of lipopolysaccharide stimulation, the influence of the sample surfaces on the gene expression profiles was investigated through RNA sequence analysis. In addition, cell spreading was observed through SEM, cell adhesion and proliferation were analysed using the CCK8 assay, and the expression of inflammation-related factors was investigated by ELISA and cellular immunofluorescence staining. The production of reactive oxygen species (ROS) in the RAW264.7 cells on the surface of the three groups was detected via immunofluorescence staining. RESULTS The CCK8 results indicated that the adhesion and proliferation of the RAW264.7 cells were reduced on the TNT and TNT-GO surfaces. ELISA results revealed significant differences in the pro-inflammatory factors tumour necrosis factor-α and interleukin-6 secretion among the three groups at 24 h (p < 0.05). The secretion of pro-inflammatory factors significantly reduced and the expression of anti-inflammatory factor IL-10 increased on the TNT and TNT-GO surfaces. The RNA sequencing, ELISA, and cell immunofluorescence staining test results suggested that the inflammatory response of M1 polarization was reduced and the M2 polarization of macrophages was induced on the TNT-GO surface, which may be attributed to the reduction in ROS production. CONCLUSIONS Under lipopolysaccharide stimulation, the inflammatory response of the RAW264.7 cells was reduced and the M2 polarization of macrophages was promoted on the TNT-GO surface, which may be caused by the reduced ROS production. Consequently, the designed TNT-GO material is promising for implants owing to its excellent inflammation regulation ability.
Collapse
Affiliation(s)
- Xu Cao
- Laboratory of Biomaterials and Biomechanics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Bin Luo
- Laboratory of Biomaterials and Biomechanics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Yanting Mu
- Laboratory of Biomaterials and Biomechanics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Caiyun Wang
- Laboratory of Biomaterials and Biomechanics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Ran Lu
- Laboratory of Biomaterials and Biomechanics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Yao Yao
- Laboratory of Biomaterials and Biomechanics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Su Chen
- Laboratory of Biomaterials and Biomechanics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
35
|
Cao J, Hong K, Lv C, Jiang W, Chen Q, Wang R, Wang Y. Reduction-sensitive polymeric carrier for the targeted delivery of a quinazoline derivative for enhanced generation of reactive oxygen species against cancer. Biomater Sci 2024; 12:2626-2638. [PMID: 38526801 DOI: 10.1039/d3bm02136j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Hepatocellular carcinoma (HCC) is one of the deadliest malignant tumors and the development of effective therapeutics against HCC is urgently needed. A novel quinazoline derivative 04NB-03 (Qd04) has been proved to be highly effective against HCC without obvious toxic side-effects. However, the poor water solubility and low bioavailability in vivo severely limit its clinical application. In addition, Qd04 kills tumor cells by inducing an accumulation of endogenous reactive oxygen species (ROS), which is highly impeded by the overexpression of glutathione (GSH) in tumor cells. Herein, we designed a disulfide cross-linked polyamino acid micelle to deliver Qd04 for HCC therapy. The disulfide linkage not only endowed a tumor-targeted delivery of Qd04 by responding to tumor cell GSH but also depleted GSH to achieve increased levels of ROS generation, which improved the therapeutic efficiency of Qd04. Both in vitro and in vivo results demonstrated that the synthesized nanodrug exerted good anti-hepatoma effects, which provided a potential application for HCC therapy in clinics.
Collapse
Affiliation(s)
- Jianrong Cao
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China.
| | - Keze Hong
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China.
| | - Chengqi Lv
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China.
| | - Weiting Jiang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China.
| | - Qi Chen
- Department of Gynecology and Obstetrics, the First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Rongze Wang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China.
| | - Yong Wang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
36
|
Chen X, Wu H, Liu Y, Liu L, Houser SR, Wang WE. Metabolic Reprogramming: A Byproduct or a Driver of Cardiomyocyte Proliferation? Circulation 2024; 149:1598-1610. [PMID: 38739695 DOI: 10.1161/circulationaha.123.065880] [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] [Indexed: 05/16/2024]
Abstract
Defining mechanisms of cardiomyocyte proliferation should guide the understanding of endogenous cardiac regeneration and could lead to novel treatments for diseases such as myocardial infarction. In the neonatal heart, energy metabolic reprogramming (phenotypic alteration of glucose, fatty acid, and amino acid metabolism) parallels cell cycle arrest of cardiomyocytes. The metabolic reprogramming occurring shortly after birth is associated with alterations in blood oxygen levels, metabolic substrate availability, hemodynamic stress, and hormone release. In the adult heart, myocardial infarction causes metabolic reprogramming but these changes cannot stimulate sufficient cardiomyocyte proliferation to replace those lost by the ischemic injury. Some putative pro-proliferative interventions can induce the metabolic reprogramming. Recent data show that altering the metabolic enzymes PKM2 [pyruvate kinase 2], LDHA [lactate dehydrogenase A], PDK4 [pyruvate dehydrogenase kinase 4], SDH [succinate dehydrogenase], CPT1b [carnitine palmitoyl transferase 1b], or HMGCS2 [3-hydroxy-3-methylglutaryl-CoA synthase 2] is sufficient to partially reverse metabolic reprogramming and promotes adult cardiomyocyte proliferation. How metabolic reprogramming regulates cardiomyocyte proliferation is not clearly defined. The possible mechanisms involve biosynthetic pathways from the glycolysis shunts and the epigenetic regulation induced by metabolic intermediates. Metabolic manipulation could represent a new approach to stimulate cardiac regeneration; however, the efficacy of these manipulations requires optimization, and novel molecular targets need to be defined. In this review, we summarize the features, triggers, and molecular regulatory networks responsible for metabolic reprogramming and discuss the current understanding of metabolic reprogramming as a critical determinant of cardiomyocyte proliferation.
Collapse
Affiliation(s)
- Xiaokang Chen
- Department of Geriatrics (X.C., H.W., Y.L., L.L., W.E.W.), Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Cardiovascular Center (X.C., H.W., Y.L., L.L., W.E.W.), Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hao Wu
- Department of Geriatrics (X.C., H.W., Y.L., L.L., W.E.W.), Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Cardiovascular Center (X.C., H.W., Y.L., L.L., W.E.W.), Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Ya Liu
- Department of Geriatrics (X.C., H.W., Y.L., L.L., W.E.W.), Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Cardiovascular Center (X.C., H.W., Y.L., L.L., W.E.W.), Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lingyan Liu
- Department of Geriatrics (X.C., H.W., Y.L., L.L., W.E.W.), Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Cardiovascular Center (X.C., H.W., Y.L., L.L., W.E.W.), Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Steven R Houser
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (S.R.H.)
| | - Wei Eric Wang
- Department of Geriatrics (X.C., H.W., Y.L., L.L., W.E.W.), Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Cardiovascular Center (X.C., H.W., Y.L., L.L., W.E.W.), Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
37
|
Wang M, Li C, Liu Y, Jin Y, Yu Y, Tan X, Zhang C. The effect of macrophages and their exosomes in ischemic heart disease. Front Immunol 2024; 15:1402468. [PMID: 38799471 PMCID: PMC11116575 DOI: 10.3389/fimmu.2024.1402468] [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: 03/17/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
Ischemic heart disease (IHD) is a leading cause of disability and death worldwide, with immune regulation playing a crucial role in its pathogenesis. Various immune cells are involved, and as one of the key immune cells residing in the heart, macrophages play an indispensable role in the inflammatory and reparative processes during cardiac ischemia. Exosomes, extracellular vesicles containing lipids, nucleic acids, proteins, and other bioactive molecules, have emerged as important mediators in the regulatory functions of macrophages and hold promise as a novel therapeutic target for IHD. This review summarizes the regulatory mechanisms of different subsets of macrophages and their secreted exosomes during cardiac ischemia over the past five years. It also discusses the current status of clinical research utilizing macrophages and their exosomes, as well as strategies to enhance their therapeutic efficacy through biotechnology. The aim is to provide valuable insights for the treatment of IHD.
Collapse
Affiliation(s)
- Minrui Wang
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yuchang Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yuanyuan Jin
- The Key Laboratory of Medical Electrophysiology of the Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
| | - Yang Yu
- The Key Laboratory of Medical Electrophysiology of the Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaoqiu Tan
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
- The Key Laboratory of Medical Electrophysiology of the Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chunxiang Zhang
- The Key Laboratory of Medical Electrophysiology of the Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
38
|
Zhang Q, Luo Y, Qian B, Cao X, Xu C, Guo K, Wan R, Jiang Y, Wang T, Mei Z, Liu J, Lv C. A systematic pan-cancer analysis identifies LDHA as a novel predictor for immunological, prognostic, and immunotherapy resistance. Aging (Albany NY) 2024; 16:8000-8018. [PMID: 38709280 PMCID: PMC11132014 DOI: 10.18632/aging.205800] [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: 08/31/2023] [Accepted: 03/18/2024] [Indexed: 05/07/2024]
Abstract
Lactate dehydrogenase A (LDHA), a critical enzyme involved in glycolysis, is broadly involved multiple biological functions in human cancers. It is reported that LDHA can impact tumor immune surveillance and induce the transformation of tumor-associated macrophages, highlighting its unnoticed function of LDHA in immune system. However, in human cancers, the role of LDHA in prognosis and immunotherapy hasn't been investigated. In this study, we analyzed the expression pattern and prognostic value of LDHA in pan-cancer and explored its association between tumor microenvironment (TME), immune infiltration subtype, stemness scores, tumor mutation burden (TMB), and immunotherapy resistance. We found that LDHA expression is tumor heterogeneous and that its high expression is associated with poor prognosis in multiple human cancers. In addition, LDHA expression was positively correlated with the presence of mononuclear/macrophage cells, and also promoted the infiltration of a range of immune cells. Genomic alteration of LDHA was common in different types of cancer, while with prognostic value in pan-cancers. Pan-cancer analysis revealed that the significant correlations existed between LDHA expression and tumor microenvironment (including stromal cells and immune cells) as well as stemness scores (DNAss and RNAss) across cancer types. Drug sensitivity analysis also revealed that LDHA was able to predict response to chemotherapy and immunotherapy. Furthermore, it was confirmed that knockdown of LDHA reduced proliferation and migration ability of lung cancer cells. Taken together, LDHA could serve as a prognostic biomarker and a potential immunotherapy marker.
Collapse
Affiliation(s)
- Qiqi Zhang
- The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, P.R. China
- Southwest Medical University, Luzhou, P.R. China
| | - Yuanning Luo
- The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, P.R. China
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, P.R. China
| | - Bingshuo Qian
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, P.R. China
- School of Pharmacy, Henan University, Kaifeng, P.R. China
| | - Xiuhua Cao
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, P.R. China
| | - Caijun Xu
- Southwest Medical University, Luzhou, P.R. China
| | - Kan Guo
- The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, P.R. China
- Southwest Medical University, Luzhou, P.R. China
| | - Runlan Wan
- The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, P.R. China
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, P.R. China
| | - Yaling Jiang
- Southwest Medical University, Luzhou, P.R. China
| | - Tiecheng Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, P.R. China
| | - Zhiqiang Mei
- The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, P.R. China
- Southwest Medical University, Luzhou, P.R. China
| | - Jinbiao Liu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P.R. China
| | - Chaoxiang Lv
- The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, P.R. China
- Southwest Medical University, Luzhou, P.R. China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, P.R. China
| |
Collapse
|
39
|
Shan J, Jin X, Zhang C, Huang M, Xing J, Li Q, Cui Y, Niu Q, Chen XL, Wang X. Metal natural product complex Ru-procyanidins with quadruple enzymatic activity combat infections from drug-resistant bacteria. Acta Pharm Sin B 2024; 14:2298-2316. [PMID: 38799629 PMCID: PMC11121202 DOI: 10.1016/j.apsb.2023.12.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/20/2023] [Accepted: 12/23/2023] [Indexed: 05/29/2024] Open
Abstract
Bacterial infection hampers wound repair by impeding the healing process. Concurrently, inflammation at the wound site triggers the production of reactive oxygen species (ROS), causing oxidative stress and damage to proteins and cells. This can lead to chronic wounds, posing severe risks. Therefore, eliminating bacterial infection and reducing ROS levels are crucial for effective wound healing. Nanozymes, possessing enzyme-like catalytic activity, can convert endogenous substances into highly toxic substances, such as ROS, to combat bacteria and biofilms without inducing drug resistance. However, the current nanozyme model with single enzyme activity falls short of meeting the complex requirements of antimicrobial therapy. Thus, developing nanozymes with multiple enzymatic activities is essential. Herein, we engineered a novel metalloenzyme called Ru-procyanidin nanoparticles (Ru-PC NPs) with diverse enzymatic activities to aid wound healing and combat bacterial infections. Under acidic conditions, due to their glutathione (GSH) depletion and peroxidase (POD)-like activity, Ru-PC NPs combined with H2O2 exhibit excellent antibacterial effects. However, in a neutral environment, the Ru-PC NPs, with catalase (CAT) activity, decompose H2O2 to O2, alleviating hypoxia and ensuring a sufficient oxygen supply. Furthermore, Ru-PC NPs possess exceptional antioxidant capacity through their superior superoxide dismutase (SOD) enzyme activity, effectively scavenging excess ROS and reactive nitrogen species (RNS) in a neutral environment. This maintains the balance of the antioxidant system and prevents inflammation. Ru-PC NPs also promote the polarization of macrophages from M1 to M2, facilitating wound healing. More importantly, Ru-PC NPs show good biosafety with negligible toxicity. In vivo wound infection models have confirmed the efficacy of Ru-PC NPs in inhibiting bacterial infection and promoting wound healing. The focus of this work highlights the quadruple enzymatic activity of Ru-PC NPs and its potential to reduce inflammation and promote bacteria-infected wound healing.
Collapse
Affiliation(s)
- Jie Shan
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
| | - Xu Jin
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
| | - Cong Zhang
- Division of Gastroenterology, the First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Muchen Huang
- The Second Clinical Medical College, Anhui Medical University, Hefei 230022, China
| | - Jianghao Xing
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Qingrong Li
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
| | - Yuyu Cui
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Qiang Niu
- The Second Clinical Medical College, Anhui Medical University, Hefei 230022, China
| | - Xu Lin Chen
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Xianwen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
- College and Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| |
Collapse
|
40
|
Carey CM, Hollins HL, Schmid AV, Gagnon JA. Distinct features of the regenerating heart uncovered through comparative single-cell profiling. Biol Open 2024; 13:bio060156. [PMID: 38526188 PMCID: PMC11007736 DOI: 10.1242/bio.060156] [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: 09/18/2023] [Accepted: 03/14/2024] [Indexed: 03/26/2024] Open
Abstract
Adult humans respond to heart injury by forming a permanent scar, yet other vertebrates are capable of robust and complete cardiac regeneration. Despite progress towards characterizing the mechanisms of cardiac regeneration in fish and amphibians, the large evolutionary gulf between mammals and regenerating vertebrates complicates deciphering which cellular and molecular features truly enable regeneration. To better define these features, we compared cardiac injury responses in zebrafish and medaka, two fish species that share similar heart anatomy and common teleost ancestry but differ in regenerative capability. We used single-cell transcriptional profiling to create a time-resolved comparative cell atlas of injury responses in all major cardiac cell types across both species. With this approach, we identified several key features that distinguish cardiac injury response in the non-regenerating medaka heart. By comparing immune responses to injury, we found altered cell recruitment and a distinct pro-inflammatory gene program in medaka leukocytes, and an absence of the injury-induced interferon response seen in zebrafish. In addition, we found a lack of pro-regenerative signals, including nrg1 and retinoic acid, from medaka endothelial and epicardial cells. Finally, we identified alterations in the myocardial structure in medaka, where they lack primordial layer cardiomyocytes and fail to employ a cardioprotective gene program shared by regenerating vertebrates. Our findings reveal notable variation in injury response across nearly all major cardiac cell types in zebrafish and medaka, demonstrating how evolutionary divergence influences the hidden cellular features underpinning regenerative potential in these seemingly similar vertebrates.
Collapse
Affiliation(s)
- Clayton M. Carey
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - Hailey L. Hollins
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - Alexis V. Schmid
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - James A. Gagnon
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
- Henry Eyring Center for Cell & Genome Science, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
41
|
Bi YW, Li LS, Ru N, Zhang B, Lei X. Nicotinamide adenine dinucleotide phosphate oxidase in pancreatic diseases: Mechanisms and future perspectives. World J Gastroenterol 2024; 30:429-439. [PMID: 38414585 PMCID: PMC10895600 DOI: 10.3748/wjg.v30.i5.429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/17/2023] [Accepted: 01/12/2024] [Indexed: 01/31/2024] Open
Abstract
Pancreatitis and pancreatic cancer (PC) stand as the most worrisome ailments affecting the pancreas. Researchers have dedicated efforts to unraveling the mechanisms underlying these diseases, yet their true nature continues to elude their grasp. Within this realm, oxidative stress is often believed to play a causal and contributory role in the development of pancreatitis and PC. Excessive accumulation of reactive oxygen species (ROS) can cause oxidative stress, and the key enzyme responsible for inducing ROS production in cells is nicotinamide adenine dinucleotide phosphate hydrogen oxides (NOX). NOX contribute to pancreatic fibrosis and inflammation by generating ROS that injure acinar cells, activate pancreatic stellate cells, and mediate macrophage polarization. Excessive ROS production occurs during malignant transformation and pancreatic carcinogenesis, creating an oxidative microenvironment that can cause abnormal apoptosis, epithelial to mesenchymal transition and genomic instability. Therefore, understanding the role of NOX in pancreatic diseases contributes to a more in-depth exploration of the exact pathogenesis of these diseases. In this review, we aim to summarize the potential roles of NOX and its mechanism in pancreatic disorders, aiming to provide novel insights into understanding the mechanisms underlying these diseases.
Collapse
Affiliation(s)
- Ya-Wei Bi
- Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Long-Song Li
- Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Nan Ru
- Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Bo Zhang
- Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Xiao Lei
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
42
|
Lv Z, Sun L, Xie X, Yao X, Tian S, Wang C, Wang F, Liu J. TMEM225 Is Essential for Sperm Maturation and Male Fertility by Modifying Protein Distribution of Sperm in Mice. Mol Cell Proteomics 2024; 23:100720. [PMID: 38246484 PMCID: PMC10875271 DOI: 10.1016/j.mcpro.2024.100720] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 01/07/2024] [Accepted: 01/13/2024] [Indexed: 01/23/2024] Open
Abstract
Nonobstructive azoospermia is the leading cause of male infertility. Abnormal levels of transmembrane protein 225 (TMEM225), a testis-specific protein, have been found in patients with nonobstructive azoospermia, suggesting that TMEM225 plays an essential role in male fertility. Here, we generated a Tmem225 KO mouse model to explore the function and mechanism of TMEM225 in male reproduction. Male Tmem225 KO mice were infertile. Surprisingly, Tmem225 deletion did not affect spermatogenesis, but TMEM225-null sperm exhibited abnormalities during epididymal maturation, resulting in reduced sperm motility and an abnormal hairpin-loop configuration. Furthermore, proteomics analyses of cauda sperm revealed that signaling pathways related to mitochondrial function, the glycolytic pathway, and sperm flagellar morphology were abnormal in Tmem225 KO sperm, and spermatozoa lacking TMEM225 exhibited high reactive oxygen species levels, reduced motility, and flagellar folding, leading to typical asthenospermia. These findings suggest that testicular TMEM225 may control the sperm maturation process by regulating the expression of proteins related to mitochondrial function, glycolysis, and sperm flagellar morphology in epididymal spermatozoa.
Collapse
Affiliation(s)
- Zheng Lv
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Longjie Sun
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xiaomei Xie
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xiaohong Yao
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Shuang Tian
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Chaofan Wang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Fengchao Wang
- Transgenic Animal Center, National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China.
| | - Jiali Liu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China.
| |
Collapse
|
43
|
Yang X, Li L, Zeng C, Wang WE. The characteristics of proliferative cardiomyocytes in mammals. J Mol Cell Cardiol 2023; 185:50-64. [PMID: 37918322 DOI: 10.1016/j.yjmcc.2023.10.010] [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: 12/31/2022] [Revised: 10/03/2023] [Accepted: 10/16/2023] [Indexed: 11/04/2023]
Abstract
Better understanding of the mechanisms regulating the proliferation of pre-existing cardiomyocyte (CM) should lead to better options for regenerating injured myocardium. The absence of a perfect research model to definitively identify newly formed mammalian CMs is lacking. However, methodologies are being developed to identify and enrich proliferative CMs. These methods take advantages of the different proliferative states of CMs during postnatal development, before and after injury in the neonatal heart. New approaches use CMs labeled in lineage tracing animals or single cell technique-based CM clusters. This review aims to provide a timely update on the characteristics of the proliferative CMs, including their structural, functional, genetic, epigenetic and metabolic characteristics versus non-proliferative CMs. A better understanding of the characteristics of proliferative CMs should lead to the mechanisms for inducing endogenous CMs to self-renew, which is a promising therapeutic strategy to treat cardiac diseases that cause CM death in humans.
Collapse
Affiliation(s)
- Xinyue Yang
- Department of Geriatrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Liangpeng Li
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Wei Eric Wang
- Department of Geriatrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
44
|
Chen S, Zou Y, Song C, Cao K, Cai K, Wu Y, Zhang Z, Geng D, Sun W, Ouyang N, Zhang N, Li Z, Sun G, Zhang Y, Sun Y, Zhang Y. The role of glycolytic metabolic pathways in cardiovascular disease and potential therapeutic approaches. Basic Res Cardiol 2023; 118:48. [PMID: 37938421 PMCID: PMC10632287 DOI: 10.1007/s00395-023-01018-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/09/2023]
Abstract
Cardiovascular disease (CVD) is a major threat to human health, accounting for 46% of non-communicable disease deaths. Glycolysis is a conserved and rigorous biological process that breaks down glucose into pyruvate, and its primary function is to provide the body with the energy and intermediate products needed for life activities. The non-glycolytic actions of enzymes associated with the glycolytic pathway have long been found to be associated with the development of CVD, typically exemplified by metabolic remodeling in heart failure, which is a condition in which the heart exhibits a rapid adaptive response to hypoxic and hypoxic conditions, occurring early in the course of heart failure. It is mainly characterized by a decrease in oxidative phosphorylation and a rise in the glycolytic pathway, and the rise in glycolysis is considered a hallmark of metabolic remodeling. In addition to this, the glycolytic metabolic pathway is the main source of energy for cardiomyocytes during ischemia-reperfusion. Not only that, the auxiliary pathways of glycolysis, such as the polyol pathway, hexosamine pathway, and pentose phosphate pathway, are also closely related to CVD. Therefore, targeting glycolysis is very attractive for therapeutic intervention in CVD. However, the relationship between glycolytic pathway and CVD is very complex, and some preclinical studies have confirmed that targeting glycolysis does have a certain degree of efficacy, but its specific role in the development of CVD has yet to be explored. This article aims to summarize the current knowledge regarding the glycolytic pathway and its key enzymes (including hexokinase (HK), phosphoglucose isomerase (PGI), phosphofructokinase-1 (PFK1), aldolase (Aldolase), phosphoglycerate metatase (PGAM), enolase (ENO) pyruvate kinase (PKM) lactate dehydrogenase (LDH)) for their role in cardiovascular diseases (e.g., heart failure, myocardial infarction, atherosclerosis) and possible emerging therapeutic targets.
Collapse
Affiliation(s)
- Shuxian Chen
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Yuanming Zou
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Chunyu Song
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Kexin Cao
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Kexin Cai
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Yanjiao Wu
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Zhaobo Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Danxi Geng
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Wei Sun
- Department of Thyroid Surgery, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Nanxiang Ouyang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Naijin Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
- Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
- Key Laboratory of Reproductive and Genetic Medicine, China Medical University, National Health Commission, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| | - Zhao Li
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Guozhe Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Yixiao Zhang
- Department of Urology Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
- Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| | - Ying Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
- Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| |
Collapse
|
45
|
Zuo W, Sun R, Ji Z, Ma G. Macrophage-driven cardiac inflammation and healing: insights from homeostasis and myocardial infarction. Cell Mol Biol Lett 2023; 28:81. [PMID: 37858035 PMCID: PMC10585879 DOI: 10.1186/s11658-023-00491-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/19/2023] [Indexed: 10/21/2023] Open
Abstract
Early and prompt reperfusion therapy has markedly improved the survival rates among patients enduring myocardial infarction (MI). Nonetheless, the resulting adverse remodeling and the subsequent onset of heart failure remain formidable clinical management challenges and represent a primary cause of disability in MI patients worldwide. Macrophages play a crucial role in immune system regulation and wield a profound influence over the inflammatory repair process following MI, thereby dictating the degree of myocardial injury and the subsequent pathological remodeling. Despite numerous previous biological studies that established the classical polarization model for macrophages, classifying them as either M1 pro-inflammatory or M2 pro-reparative macrophages, this simplistic categorization falls short of meeting the precision medicine standards, hindering the translational advancement of clinical research. Recently, advances in single-cell sequencing technology have facilitated a more profound exploration of macrophage heterogeneity and plasticity, opening avenues for the development of targeted interventions to address macrophage-related factors in the aftermath of MI. In this review, we provide a summary of macrophage origins, tissue distribution, classification, and surface markers. Furthermore, we delve into the multifaceted roles of macrophages in maintaining cardiac homeostasis and regulating inflammation during the post-MI period.
Collapse
Affiliation(s)
- Wenjie Zuo
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, No. 87, Dingjiaqiao, Nanjing, 210009, China
| | - Renhua Sun
- Department of Cardiology, Yancheng No. 1 People's Hospital, No. 66 South Renmin Road, Yancheng, 224000, China
| | - Zhenjun Ji
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, No. 87, Dingjiaqiao, Nanjing, 210009, China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, No. 87, Dingjiaqiao, Nanjing, 210009, China.
| |
Collapse
|
46
|
Li S, Liu L, Luo G, Yuan Y, Hu D, Xiao F. The crosstalk between M1 macrophage polarization and energy metabolism disorder contributes to polystyrene nanoplastics-triggered testicular inflammation. Food Chem Toxicol 2023; 180:114002. [PMID: 37634612 DOI: 10.1016/j.fct.2023.114002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/06/2023] [Accepted: 08/22/2023] [Indexed: 08/29/2023]
Abstract
Ubiquitous microplastics have become a threat to animal and human health, due to their potential toxicity, persistent nature and consequent bioaccumulation. Supporting evidence elucidates that polystyrene nanoplastics (PS-NPs) can destroy blood-testis barrier integrity, thus causing testicular hypoplasia and impairment of spermatogenesis. Nevertheless, how PS-NPs modulate macrophage polarization-energy metabolism crosstalk has not been fully investigated in testicular tissue. Here, we observed that polystyrene PS-NPs exposure contributes to severe vacuolization in the seminiferous tubules, accompanied by apoptosis of testicular tissue and infiltration of M1 macrophages. Meanwhile, we found that PS-NPs could trigger the M1 polarization phenotype, which activated ROS-macrophage migration inhibitory factor (MIF)/NF-κB signaling that in turn induced apoptosis of GC2 cells in the GC2-macrophage cell coculture model. Simultaneously, we confirmed that PS-NPs exposure increased 3-phospho-D-glycerate, phosphoenolpyruvate and lactate concentrations, accompanied by decreased pyruvate and adenosine triphosphate (ATP) production, likely due to downregulated pyruvate kinase M2 (PKM2) dimer expression. In conclusion, the mechanism of PS-NPs-induced testicular inflammation can be mediated by promoting the infiltration of M1 macrophages, thereby resulting in an ROS burst and subsequent induction of energy metabolism disorders. The current study will provide new insights into PS-NPs-induced male reproductive toxicity and highlight the context-specific roles of testicular macrophages.
Collapse
Affiliation(s)
- Siwen Li
- Xiangya School of Public Health, Central South University, Changsha, 410078, PR China
| | - Lemei Liu
- Xiangya School of Public Health, Central South University, Changsha, 410078, PR China
| | - Gang Luo
- Xiangya School of Public Health, Central South University, Changsha, 410078, PR China
| | - Yu Yuan
- Xiangya School of Public Health, Central South University, Changsha, 410078, PR China
| | - Die Hu
- Xiangya School of Public Health, Central South University, Changsha, 410078, PR China
| | - Fang Xiao
- Xiangya School of Public Health, Central South University, Changsha, 410078, PR China.
| |
Collapse
|
47
|
Abstract
Interest in cardioimmunology has reached new heights as the experimental cardiology field works to tap the unrealized potential of immunotherapy for clinical care. Within this space is the cardiac macrophage, a key modulator of cardiac function in health and disease. After a myocardial infarction, myeloid macrophages both protect and harm the heart. To varying degrees, such outcomes are a function of myeloid ontogeny and heterogeneity, as well as functional cellular plasticity. Diversity is further shaped by the extracellular milieu, which fluctuates considerably after coronary occlusion. Ischemic limitation of nutrients constrains the metabolic potential of immune cells, and accumulating evidence supports a paradigm whereby macrophage metabolism is coupled to divergent inflammatory consequences, although experimental evidence for this in the heart is just emerging. Herein we examine the heterogeneous cardiac macrophage response following ischemic injury, with a focus on integrating putative contributions of immunometabolism and implications for therapeutically relevant cardiac injury versus cardiac repair.
Collapse
|
48
|
Wang J, Wu W, Xia J, Chen L, Liu D, Wang G, Wang L, Zheng Q. Dynamic changes in macrophage subtypes during lung cancer progression and metastasis at single-cell resolution. J Thorac Dis 2023; 15:4456-4471. [PMID: 37691661 PMCID: PMC10482613 DOI: 10.21037/jtd-23-1012] [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: 06/27/2023] [Accepted: 08/18/2023] [Indexed: 09/12/2023]
Abstract
Background Lung cancer remains a major global health challenge. Macrophages (Macs) are one important component of tumor microenvironments (TMEs); however, their prognostic relevance to lung cancer is currently unknown due to the complexity of their phenotypes. Methods In the present study, reanalysis and atlas reconstruction of downloaded single-cell RNA sequencing (scRNAseq) data were used to systematically compare the component and transcriptional changes in Mac subtypes across different stages of lung cancer. Results We found that with the progression of lung cancer, the proportion of alveolar macrophages (aMacs) gradually decreased, while the proportions of Macs and monocytes (Monos) gradually increased, suggesting a chemotaxis process followed by a Mono-Mac differentiation process. Meanwhile, through ligand-receptor (LR) screening, we identified 9 Mac-specific interactions that were enriched during the progression and metastasis of lung cancer, which could potential promote M2 polarization or the infiltration of M2 Macs. Moreover, we found that the expression of SPP1 in Macs increased with lung cancer progression, and identified 9 genes that were correlated with the expression of SPP1 in Macs, which might also contribute to the immunosuppression process in lung cancer. Conclusions Our results revealed detailed changes in Macs at different stages of lung cancer progression and metastasis and provided potential therapeutic targets that could be used in future lung cancer treatments.
Collapse
Affiliation(s)
- Jian Wang
- Department of Thoracic Surgery, The Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen, China
| | - Weiqing Wu
- Department of Health Management, Shenzhen People’s Hospital, Shenzhen, China
| | - Jinquan Xia
- Department of Respiratory and Critical Care Medicine, Shenzhen People’s Hospital, Shenzhen, China
| | - Lipeng Chen
- Department of Respiratory and Critical Care Medicine, Shenzhen People’s Hospital, Shenzhen, China
| | - Dongcheng Liu
- Department of Respiratory and Critical Care Medicine, Shenzhen People’s Hospital, Shenzhen, China
| | - Guangsuo Wang
- Department of Thoracic Surgery, The Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen, China
| | - Lingwei Wang
- Department of Respiratory and Critical Care Medicine, Shenzhen People’s Hospital, Shenzhen, China
| | - Qijun Zheng
- Department of Cardiac Surgery, The Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen, China
| |
Collapse
|
49
|
Carey CM, Hollins HL, Schmid AV, Gagnon JA. Distinct features of the regenerating heart uncovered through comparative single-cell profiling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.04.547574. [PMID: 37461520 PMCID: PMC10349989 DOI: 10.1101/2023.07.04.547574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Adult humans respond to heart injury by forming a permanent scar, yet other vertebrates are capable of robust and complete cardiac regeneration. Despite progress towards characterizing the mechanisms of cardiac regeneration in fish and amphibians, the large evolutionary gulf between mammals and regenerating vertebrates complicates deciphering which cellular and molecular features truly enable regeneration. To better define these features, we compared cardiac injury responses in zebrafish and medaka, two fish species that share similar heart anatomy and common teleost ancestry but differ in regenerative capability. We used single-cell transcriptional profiling to create a time-resolved comparative cell atlas of injury responses in all major cardiac cell types across both species. With this approach, we identified several key features that distinguish cardiac injury response in the non-regenerating medaka heart. By comparing immune responses to injury, we found altered cell recruitment and a distinct pro-inflammatory gene program in medaka leukocytes, and an absence of the injury-induced interferon response seen in zebrafish. In addition, we found a lack of pro-regenerative signals, including nrg1 and retinoic acid, from medaka endothelial and epicardial cells. Finally, we identified alterations in the myocardial structure in medaka, where they lack embryonic-like primordial layer cardiomyocytes, and fail to employ a cardioprotective gene program shared by regenerating vertebrates. Our findings reveal notable variation in injury response across nearly all major cardiac cell types in zebrafish and medaka, demonstrating how evolutionary divergence influences the hidden cellular features underpinning regenerative potential in these seemingly similar vertebrates.
Collapse
Affiliation(s)
- Clayton M. Carey
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - Hailey L. Hollins
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - Alexis V. Schmid
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - James A. Gagnon
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
- Henry Eyring Center for Cell & Genome Science, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
50
|
DeBerge M, Chaudhary R, Schroth S, Thorp EB. Immunometabolism at the Heart of Cardiovascular Disease. JACC Basic Transl Sci 2023; 8:884-904. [PMID: 37547069 PMCID: PMC10401297 DOI: 10.1016/j.jacbts.2022.12.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/21/2022] [Accepted: 12/27/2022] [Indexed: 08/08/2023]
Abstract
Immune cell function among the myocardium, now more than ever, is appreciated to regulate cardiac function and pathophysiology. This is the case for both innate immunity, which includes neutrophils, monocytes, dendritic cells, and macrophages, as well as adaptive immunity, which includes T cells and B cells. This function is fueled by cell-intrinsic shifts in metabolism, such as glycolysis and oxidative phosphorylation, as well as metabolite availability, which originates from the surrounding extracellular milieu and varies during ischemia and metabolic syndrome. Immune cell crosstalk with cardiac parenchymal cells, such as cardiomyocytes and fibroblasts, is also regulated by complex cellular metabolic circuits. Although our understanding of immunometabolism has advanced rapidly over the past decade, in part through valuable insights made in cultured cells, there remains much to learn about contributions of in vivo immunometabolism and directly within the myocardium. Insight into such fundamental cell and molecular mechanisms holds potential to inform interventions that shift the balance of immunometabolism from maladaptive to cardioprotective and potentially even regenerative. Herein, we review our current working understanding of immunometabolism, specifically in the settings of sterile ischemic cardiac injury or cardiometabolic disease, both of which contribute to the onset of heart failure. We also discuss current gaps in knowledge in this context and therapeutic implications.
Collapse
Affiliation(s)
| | | | - Samantha Schroth
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Edward B. Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|