1
|
Endo-Umeda K, Makishima M. Exploring the Roles of Liver X Receptors in Lipid Metabolism and Immunity in Atherosclerosis. Biomolecules 2025; 15:579. [PMID: 40305368 PMCID: PMC12024750 DOI: 10.3390/biom15040579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 05/02/2025] Open
Abstract
Hypercholesterolemia causes atherosclerosis by inducing immune cell migration and chronic inflammation in arterial walls. Recent single-cell analyses reveal the presence of lipid-enriched foamy macrophages, as well as other macrophage subtypes, neutrophils, T cells, and B cells, in atherosclerotic plaques in both animal models and humans. These cells interact with each other and other cells, including non-immune cells such as endothelial cells and smooth muscle cells. They thereby regulate metabolic, inflammatory, phagocytic, and cell death processes, thus affecting the progression and stability of atherosclerotic plaques. The nuclear receptors liver X receptor (LXR)α and LXRβ are transcription factors that are activated by oxysterols and regulate lipid metabolism and immune responses. LXRs regulate cholesterol homeostasis by controlling cholesterol's transport, absorption, synthesis, and breakdown in the liver and intestine. LXRs are also highly expressed in tissue-resident and monocyte-derived macrophages and other immune cells, including both myeloid cells and lymphocytes, and they regulate both innate and adaptive immune responses. Interestingly, LXRs have immunosuppressive and immunoregulatory functions that are cell-type-dependent. In animal models of atherosclerosis, LXRs have been shown to be involved in both progression and regression phases. The pharmacological activation of LXR enhances cholesterol efflux from macrophages and promotes atherosclerosis progression. Deleting LXR in immune cells, especially myeloid cells, accelerates atherosclerosis by increasing monocyte migration, macrophage proliferation and activation, and neutrophil extracellular traps (NETs); furthermore, the deletion of hematopoietic LXRs impairs the regression of atherosclerotic plaques. Therefore, LXRs in immune cells may be a potent therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Kaori Endo-Umeda
- Division of Biochemistry, Department of Biomedical Sciences, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan;
| | | |
Collapse
|
2
|
Fang S, Wu S, Chen P. Targeting Caveolin-1 for enhanced rotator cuff repair: findings from single-cell RNA sequencing. Cell Death Discov 2025; 11:88. [PMID: 40044676 PMCID: PMC11882801 DOI: 10.1038/s41420-025-02359-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 02/05/2025] [Accepted: 02/13/2025] [Indexed: 03/09/2025] Open
Abstract
Rotator cuff injury (RCI), a prevalent cause of shoulder pain and disability, often leads to significant functional impairments due to adipocyte infiltration into the damaged tissue. Caveolin-1 (Cav-1), a critical membrane protein, plays a significant role in adipocyte differentiation and lipid metabolism. This study utilized single-cell RNA sequencing (scRNA-seq) to investigate the heterogeneity of cell subpopulations in RCI tissues and assess the regulatory effects of Cav-1. The findings revealed that Cav-1 expression negatively correlates with adipogenic activity, and its modulation through exercise or targeted therapies can significantly reduce adipocyte infiltration and enhance tissue repair. Further, Cav-1 knockout and overexpression models demonstrated the protein's impact on key genes involved in adipocyte differentiation and lipid metabolism, such as Scd1, fatty acid synthase (FASN), and peroxisome proliferator-activated receptor gamma (Pparg). Animal studies corroborated these results, showing that exercise intervention increased Cav-1 expression, decreased adipocyte infiltration, and promoted structural repair. These insights suggest that targeting Cav-1 could offer a novel therapeutic strategy for improving RCI outcomes.
Collapse
Affiliation(s)
- Shanhong Fang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China
- Department of Sports Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, PR China
- Fujian Orthopaedics Research Institute, Fuzhou, PR China
- Fujian Orthopedic Bone and Joint Disease and Sports Rehabilitation Clinical Medical Research Center, Fuzhou, PR China
| | - Songye Wu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China
- Department of Sports Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, PR China
- Fujian Orthopaedics Research Institute, Fuzhou, PR China
- Fujian Orthopedic Bone and Joint Disease and Sports Rehabilitation Clinical Medical Research Center, Fuzhou, PR China
| | - Peng Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China.
- Department of Sports Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, PR China.
- Fujian Orthopaedics Research Institute, Fuzhou, PR China.
- Fujian Orthopedic Bone and Joint Disease and Sports Rehabilitation Clinical Medical Research Center, Fuzhou, PR China.
| |
Collapse
|
3
|
Ma X, Huang T, Chen X, Li Q, Liao M, Fu L, Huang J, Yuan K, Wang Z, Zeng Y. Molecular mechanisms in liver repair and regeneration: from physiology to therapeutics. Signal Transduct Target Ther 2025; 10:63. [PMID: 39920130 PMCID: PMC11806117 DOI: 10.1038/s41392-024-02104-8] [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: 11/08/2023] [Revised: 09/02/2024] [Accepted: 12/12/2024] [Indexed: 02/09/2025] Open
Abstract
Liver repair and regeneration are crucial physiological responses to hepatic injury and are orchestrated through intricate cellular and molecular networks. This review systematically delineates advancements in the field, emphasizing the essential roles played by diverse liver cell types. Their coordinated actions, supported by complex crosstalk within the liver microenvironment, are pivotal to enhancing regenerative outcomes. Recent molecular investigations have elucidated key signaling pathways involved in liver injury and regeneration. Viewed through the lens of metabolic reprogramming, these pathways highlight how shifts in glucose, lipid, and amino acid metabolism support the cellular functions essential for liver repair and regeneration. An analysis of regenerative variability across pathological states reveals how disease conditions influence these dynamics, guiding the development of novel therapeutic strategies and advanced techniques to enhance liver repair and regeneration. Bridging laboratory findings with practical applications, recent clinical trials highlight the potential of optimizing liver regeneration strategies. These trials offer valuable insights into the effectiveness of novel therapies and underscore significant progress in translational research. In conclusion, this review intricately links molecular insights to therapeutic frontiers, systematically charting the trajectory from fundamental physiological mechanisms to innovative clinical applications in liver repair and regeneration.
Collapse
Affiliation(s)
- Xiao Ma
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Tengda Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiangzheng Chen
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qian Li
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Mingheng Liao
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Fu
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jiwei Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Kefei Yuan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhen Wang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Yong Zeng
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
4
|
Teng Y, Xu H, He X, Zhuang Q, Lu H, Xu R, Xue D. LXRα agonist differentially regulates BAFF expression and biological effects in RAW264.7 cells depending on growth status: LXRα activation and BAFF signaling in RAW264.7 cells. Biochem Biophys Res Commun 2025; 742:151067. [PMID: 39632295 DOI: 10.1016/j.bbrc.2024.151067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
B- cell-activating factor (BAFF), which is essential for the survival and development of B cells, is mainly produced by myeloid cells such as macrophages. Abnormal macrophage infiltration and high BAFF expression in kidney allografts are associated with the occurrence and development of antibody-mediated rejection (ABMR). Nuclear hormone receptor Liver X receptors (LXRs), is a nonnegligible participant in regulating cholesterol metabolism and inflammatory responses. Nowadays the effects of LXRα activation on macrophages have been widely studied, however the effects of LXRα activation on BAFF expression and cell function due to the change of BAFF signaling have not yet been fully investigated. In the present study, LXRα activation alone was found to downregulate BAFF expression in quiescent RAW 264.7 cells, whereas LXRα agonist significantly upregulated BAFF expression in cells pretreated with lipopolysaccharide (LPS) for 6 h. The increased BAFF signaling promoted M1 polarization and enhanced cell viability, migration, and phagocytic ability. LXRα can directly bind to the BAFF promoter region and decrease BAFF expression in RAW264.7 cells. LXRα activation enhanced mitochondrial metabolism, which promoted BAFF expression in the LPS-activated cells. Our results indicate that subtle changes in the microenvironment would affect the biological function of macrophages, in which a variety of BAFF signaling pathways may also be involved, providing a new perspective on exploring the mechanism of allograft rejection and uncovering the potential reason for the unstable efficacy of anti-BAFF preparations in kidney transplant recipients.
Collapse
Affiliation(s)
- Yisa Teng
- Urology Department, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Haiyan Xu
- Urology Department, Third Affiliated Hospital of Soochow University, Changzhou, China.
| | - Xiaozhou He
- Urology Department, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Qianfeng Zhuang
- Urology Department, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Hao Lu
- Urology Department, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Renfang Xu
- Urology Department, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Dong Xue
- Urology Department, Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
5
|
Back P, Yu M, Modaresahmadi S, Hajimirzaei S, Zhang Q, Islam MR, Schwendeman AA, La-Beck NM. Immune Implications of Cholesterol-Containing Lipid Nanoparticles. ACS NANO 2024; 18:28480-28501. [PMID: 39388645 PMCID: PMC11505898 DOI: 10.1021/acsnano.4c06369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/12/2024]
Abstract
The majority of clinically approved nanoparticle-mediated therapeutics are lipid nanoparticles (LNPs), and most of these LNPs are liposomes containing cholesterol. LNP formulations significantly alter the drug pharmacokinetics (PK) due to the propensity of nanoparticles for uptake by macrophages. In addition to readily engulfing LNPs, the high expression of cholesterol hydroxylases and reactive oxygen species (ROS) in macrophages suggests that they will readily produce oxysterols from LNP-associated cholesterol. Oxysterols are a heterogeneous group of cholesterol oxidation products that have potent immune modulatory effects. Oxysterols are implicated in the pathogenesis of atherosclerosis and certain malignancies; they have also been found in commercial liposome preparations. Yet, the in vivo metabolic fate of LNP-associated cholesterol remains unclear. We review herein the mechanisms of cellular uptake, trafficking, metabolism, and immune modulation of endogenous nanometer-sized cholesterol particles (i.e., lipoproteins) that are also relevant for cholesterol-containing nanoparticles. We believe that it would be imperative to better understand the in vivo metabolic fate of LNP-associated cholesterol and the immune implications for LNP-therapeutics. We highlight critical knowledge gaps that we believe need to be addressed in order to develop safer and more efficacious lipid nanoparticle delivery systems.
Collapse
Affiliation(s)
- Patricia
Ines Back
- Department
of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of
Pharmacy, Texas Tech University Health Sciences
Center, Abilene, Texas 79601, United States
| | - Minzhi Yu
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, North Campus Research Complex, 2800 Plymouth Road, Ann Arbor, Michigan 48109, United States
| | - Shadan Modaresahmadi
- Department
of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of
Pharmacy, Texas Tech University Health Sciences
Center, Abilene, Texas 79601, United States
| | - Sahelosadat Hajimirzaei
- Department
of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of
Pharmacy, Texas Tech University Health Sciences
Center, Abilene, Texas 79601, United States
| | - Qisheng Zhang
- Division
of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Md Rakibul Islam
- Department
of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of
Pharmacy, Texas Tech University Health Sciences
Center, Abilene, Texas 79601, United States
| | - Anna A. Schwendeman
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, North Campus Research Complex, 2800 Plymouth Road, Ann Arbor, Michigan 48109, United States
- Biointerfaces
Institute, University of Michigan, North
Campus Research Complex, 2800 Plymouth Road, Ann Arbor, Michigan 48109, United States
| | - Ninh M. La-Beck
- Department
of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of
Pharmacy, Texas Tech University Health Sciences
Center, Abilene, Texas 79601, United States
- Department
of Pharmacy Practice, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, Texas 79601, United States
| |
Collapse
|
6
|
Wang HN, Wang PH, Jiang MR, Zhang JQ, Ma SY, Hu YF, Wang YZ. The processed Euphorbia lathyris L. alleviates the inflammatory injury via regulating LXRα/ABCA1 expression and TLR4 positioning to lipid rafts. Fitoterapia 2024; 177:106111. [PMID: 38971330 DOI: 10.1016/j.fitote.2024.106111] [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: 04/08/2024] [Revised: 06/10/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024]
Abstract
Euphorbia lathyris L. (EL) is a traditional poisonous herbal medicine used to treat dropsy, ascites, amenorrhea, anuria and constipation. Processing to reduce toxicity of EL is essential for its safe and effective application. However, there is little known regarding the molecular mechanism of reducing toxicity after EL processing. This research aimed to screen the differential markers for EL and PEL, explore the differential mechanisms of inflammatory injury induced by EL and processed EL (PEL) to expound the mechanism of alleviating toxicity after EL processing. The results showed that 15 potential biomarkers, mainly belonging to diterpenoids, were screened to distinguish EL from PEL. EL promoted the expressions of TLR4, NLRP3, NF-κB p65, IL-1β and TNF-α, increased lipid rafts abundance and promoted TLR4 positioning to lipid rafts. Meanwhile, EL decreased LXRα and ABCA1 expression, and reduced cholesterol efflux. In contrast to EL, the effects of PEL on these indicators were markedly weakened. In addition, Euphorbia factors L1, L2, and L3 affected LXRα, ABCA1, TLR4, NLRP3, NF-κB p65, TNF-α and IL-1β expression, influenced cholesterol efflux and lipid rafts abundance, and interfered with the colocalization of TLR4 and lipid rafts. The inflammatory injury caused by processed EL was significantly weaker than that caused by crude EL, and reduction of Euphorbia factors L1, L2, and L3 as well as attenuation of inflammatory injury participated in processing-based detoxification of EL. Our results provide valuable insights into the attenuated mechanism of EL processing and will guide future research on the processing mechanism of toxic traditional Chinese medicine.
Collapse
Affiliation(s)
- Hui-Nan Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Pei-Hua Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Ming-Rui Jiang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Jing-Qiu Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Si-Yuan Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Yu-Feng Hu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Ying-Zi Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, PR China.
| |
Collapse
|
7
|
Liu S, Huang J, Luo J, Bian Q, Weng Y, Li L, Chen J. Therapeutic biomaterials with liver X receptor agonists based on the horizon of material biology to regulate atherosclerotic plaque regression in situ for devices surface engineering. Regen Biomater 2024; 11:rbae089. [PMID: 39165884 PMCID: PMC11335375 DOI: 10.1093/rb/rbae089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 08/22/2024] Open
Abstract
Percutaneous coronary interventional is the main treatment for coronary atherosclerosis. At present, most studies focus on blood components and smooth muscle cells to achieve anticoagulation or anti-proliferation effects, while the mediated effects of materials on macrophages are also the focus of attention. Macrophage foam cells loaded with elevated cholesterol is a prominent feature of atherosclerotic plaque. Activation of liver X receptor (LXR) to regulate cholesterol efflux and efferocytosis and reduce the number of macrophage foam cells in plaque is feasible for the regression of atherosclerosis. However, cholesterol efflux promotion remains confined to targeted therapies. Herein, LXR agonists (GW3965) were introduced on the surface of the material and delivered in situ to atherogenic macrophages to improve drug utilization for anti-atherogenic therapy and plaque regression. LXR agonists act as plaque inhibition mediated by multichannel regulation macrophages, including lipid metabolism (ABCA1, ABCG1 and low-density lipoprotein receptor), macrophage migration (CCR7) and efferocytosis (MerTK). Material loaded with LXR agonists significantly reduced plaque burden in atherosclerotic model rats, most importantly, it did not cause hepatotoxicity and adverse reactions such as restenosis and thrombosis after material implantation. Both in vivo and in vitro evaluations confirmed its anti-atherosclerotic capability and safety. Overall, multi-functional LXR agonist-loaded materials with pathological microenvironment regulation effect are expected to be promising candidates for anti-atherosclerosis and have potential applications in cardiovascular devices surface engineering.
Collapse
Affiliation(s)
- Sainan Liu
- Key Laboratory of Advanced Technology for Materials of Chinese Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Jinquan Huang
- Key Laboratory of Advanced Technology for Materials of Chinese Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Jiayan Luo
- Key Laboratory of Advanced Technology for Materials of Chinese Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Qihao Bian
- Key Laboratory of Advanced Technology for Materials of Chinese Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yajun Weng
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Li Li
- School of Health Management, West China University, Chengdu 610039, China
| | - Junying Chen
- Key Laboratory of Advanced Technology for Materials of Chinese Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| |
Collapse
|
8
|
Qin B, Hu C, Zhang Y, Chen Y, Lei Y. ABCA1 Deletion Does Not Affect Aqueous Humor Outflow Function in Mice. J Ophthalmol 2024; 2024:7195550. [PMID: 39049847 PMCID: PMC11268963 DOI: 10.1155/2024/7195550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 11/08/2023] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Background ATP binding cassette transporter A1 (ABCA1) is a candidate gene within a POAG susceptibility locus by GWAS analysis, and it is involved in IOP modulation via the Cav1/eNOS/NO signaling pathway. We aim to examine the phenotype of ABCA1 deletion in the ABCA1 gene knockout (Abca1-/-) mice. Methods The anterior segments of Abca1-/- eyes were imaged by slit-lamp microscopy and anterior segment OCT. IOPs were measured by rebound tonometry. By perfusing enucleated eyes at various pressures, the aqueous humor outflow facility was determined. The mRNA expressions of ABCA1, Cav1, and eNOS were measured by RT-qPCR. The protein expressions were analyzed by western blot and immunofluorescence staining. Results There was no significant difference in the anterior segment morphology of Abca1-/- mice. IOP and aqueous humor outflow facility did not change in Abca1-/- mice compared with wild-type mice. mRNA and protein expressions of ABCA1 were significantly lower in the outflow tissue of Abca1-/- eyes. The expressions of Cav1 and eNOS were both significantly upregulated in the outflow tissue of Abca1-/- eyes. Conclusion ABCA1 deletion does not affect IOP and aqueous humor outflow function but the Cav1/eNOS/NO pathway is changed in Abca1-/- mice. The function of ABCA1 in aqueous humor outflow still requires further research.
Collapse
Affiliation(s)
- Bo Qin
- Department of Ophthalmology and Visual ScienceEye and ENT HospitalShanghai Medical CollegeFudan University, Shanghai, China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesFudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and RestorationFudan University, Shanghai, China
| | - Chunchun Hu
- Department of Ophthalmology and Visual ScienceEye and ENT HospitalShanghai Medical CollegeFudan University, Shanghai, China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesFudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and RestorationFudan University, Shanghai, China
| | - Youjia Zhang
- Department of Ophthalmology and Visual ScienceEye and ENT HospitalShanghai Medical CollegeFudan University, Shanghai, China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesFudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and RestorationFudan University, Shanghai, China
| | - Yuhong Chen
- Department of Ophthalmology and Visual ScienceEye and ENT HospitalShanghai Medical CollegeFudan University, Shanghai, China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesFudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and RestorationFudan University, Shanghai, China
| | - Yuan Lei
- Department of Ophthalmology and Visual ScienceEye and ENT HospitalShanghai Medical CollegeFudan University, Shanghai, China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesFudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and RestorationFudan University, Shanghai, China
| |
Collapse
|
9
|
Hernández-Hernández I, De La Rosa JV, Martín-Rodríguez P, Díaz-Sarmiento M, Recio C, Guerra B, Fernández-Pérez L, León TE, Torres R, Font-Díaz J, Roig A, de Mora F, Boscá L, Díaz M, Valledor AF, Castrillo A, Tabraue C. Endogenous LXR signaling controls pulmonary surfactant homeostasis and prevents lung inflammation. Cell Mol Life Sci 2024; 81:287. [PMID: 38970705 PMCID: PMC11335212 DOI: 10.1007/s00018-024-05310-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/04/2024] [Accepted: 06/04/2024] [Indexed: 07/08/2024]
Abstract
Lung type 2 pneumocytes (T2Ps) and alveolar macrophages (AMs) play crucial roles in the synthesis, recycling and catabolism of surfactant material, a lipid/protein fluid essential for respiratory function. The liver X receptors (LXR), LXRα and LXRβ, are transcription factors important for lipid metabolism and inflammation. While LXR activation exerts anti-inflammatory actions in lung injury caused by lipopolysaccharide (LPS) and other inflammatory stimuli, the full extent of the endogenous LXR transcriptional activity in pulmonary homeostasis is incompletely understood. Here, using mice lacking LXRα and LXRβ as experimental models, we describe how the loss of LXRs causes pulmonary lipidosis, pulmonary congestion, fibrosis and chronic inflammation due to defective de novo synthesis and recycling of surfactant material by T2Ps and defective phagocytosis and degradation of excess surfactant by AMs. LXR-deficient T2Ps display aberrant lamellar bodies and decreased expression of genes encoding for surfactant proteins and enzymes involved in cholesterol, fatty acids, and phospholipid metabolism. Moreover, LXR-deficient lungs accumulate foamy AMs with aberrant expression of cholesterol and phospholipid metabolism genes. Using a house dust mite aeroallergen-induced mouse model of asthma, we show that LXR-deficient mice exhibit a more pronounced airway reactivity to a methacholine challenge and greater pulmonary infiltration, indicating an altered physiology of LXR-deficient lungs. Moreover, pretreatment with LXR agonists ameliorated the airway reactivity in WT mice sensitized to house dust mite extracts, confirming that LXR plays an important role in lung physiology and suggesting that agonist pharmacology could be used to treat inflammatory lung diseases.
Collapse
Affiliation(s)
- Irene Hernández-Hernández
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Juan V De La Rosa
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Departamento de Bioquímica y Biología Molecular, Fisiología, Genética e Inmunología, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Farmacología Molecular y Traslacional, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Patricia Martín-Rodríguez
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Departamento de Morfología, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Farmacología Molecular y Traslacional, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Mercedes Díaz-Sarmiento
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Carlota Recio
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Farmacología Molecular y Traslacional, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Borja Guerra
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Farmacología Molecular y Traslacional, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Leandro Fernández-Pérez
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Farmacología Molecular y Traslacional, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Theresa E León
- Department of Cell Biology, Physiology and Immunology, School of Biology, University of Barcelona, Barcelona, Spain
| | - Rosa Torres
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Joan Font-Díaz
- Department of Cell Biology, Physiology and Immunology, School of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
| | - Angela Roig
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Fernando de Mora
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Lisardo Boscá
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos 3-5, P-11, Madrid, 28029, Spain
| | - Mario Díaz
- Laboratory of Membrane Physiology and Biophysics, School of Physics, Faculty of Sciences, University of La Laguna, San Cristóbal de La Laguna, Tenerife, Spain
| | - Annabel F Valledor
- Department of Cell Biology, Physiology and Immunology, School of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
| | - Antonio Castrillo
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), CSIC-UAM, Madrid, Spain.
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Farmacología Molecular y Traslacional, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.
| | - Carlos Tabraue
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.
- Departamento de Morfología, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Farmacología Molecular y Traslacional, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.
| |
Collapse
|
10
|
Vladimir de la Rosa J, Tabraue C, Huang Z, Orizaola MC, Martin‐Rodríguez P, Steffensen KR, Zapata JM, Boscá L, Tontonoz P, Alemany S, Treuter E, Castrillo A. Reprogramming of the LXRα Transcriptome Sustains Macrophage Secondary Inflammatory Responses. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307201. [PMID: 38549193 PMCID: PMC11132038 DOI: 10.1002/advs.202307201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 03/01/2024] [Indexed: 05/29/2024]
Abstract
Macrophages regulate essential aspects of innate immunity against pathogens. In response to microbial components, macrophages activate primary and secondary inflammatory gene programs crucial for host defense. The liver X receptors (LXRα, LXRβ) are ligand-dependent nuclear receptors that direct gene expression important for cholesterol metabolism and inflammation, but little is known about the individual roles of LXRα and LXRβ in antimicrobial responses. Here, the results demonstrate that induction of LXRα transcription by prolonged exposure to lipopolysaccharide (LPS) supports inflammatory gene expression in macrophages. LXRα transcription is induced by NF-κB and type-I interferon downstream of TLR4 activation. Moreover, LPS triggers a reprogramming of the LXRα cistrome that promotes cytokine and chemokine gene expression through direct LXRα binding to DNA consensus sequences within cis-regulatory regions including enhancers. LXRα-deficient macrophages present fewer binding of p65 NF-κB and reduced histone H3K27 acetylation at enhancers of secondary inflammatory response genes. Mice lacking LXRα in the hematopoietic compartment show impaired responses to bacterial endotoxin in peritonitis models, exhibiting reduced neutrophil infiltration and decreased expansion and inflammatory activation of recruited F4/80lo-MHC-IIhi peritoneal macrophages. Together, these results uncover a previously unrecognized function for LXRα-dependent transcriptional cis-activation of secondary inflammatory gene expression in macrophages and the host response to microbial ligands.
Collapse
Affiliation(s)
- Juan Vladimir de la Rosa
- Unidad de Biomedicina (Unidad Asociada al CSIC)Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran CanariaLas Palmas35016Spain
| | - Carlos Tabraue
- Unidad de Biomedicina (Unidad Asociada al CSIC)Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran CanariaLas Palmas35016Spain
- Departamento de MorfologíaUniversidad de Las Palmas de Gran CanariaLas Palmas35016Spain
| | - Zhiqiang Huang
- Department of Biosciences and NutritionKarolinska Institutet, NEOHuddinge14183Sweden
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular MedicineMedical SchoolNanjing UniversityNanjing210093P. R. China
| | - Marta C. Orizaola
- Department of Metabolic and Immune Diseases. Instituto de Investigaciones Biomédicas Sols‐MorrealeCentro Mixto Consejo Superior de Investigaciones Científicas CSIC‐Universidad Autónoma de MadridMadrid28029Spain
| | - Patricia Martin‐Rodríguez
- Unidad de Biomedicina (Unidad Asociada al CSIC)Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran CanariaLas Palmas35016Spain
| | - Knut R. Steffensen
- Division of Clinical Chemistry, Department of Laboratory MedicineKarolinska InstituteHuddinge14186Sweden
| | - Juan Manuel Zapata
- Department of Metabolic and Immune Diseases. Instituto de Investigaciones Biomédicas Sols‐MorrealeCentro Mixto Consejo Superior de Investigaciones Científicas CSIC‐Universidad Autónoma de MadridMadrid28029Spain
| | - Lisardo Boscá
- Department of Metabolic and Immune Diseases. Instituto de Investigaciones Biomédicas Sols‐MorrealeCentro Mixto Consejo Superior de Investigaciones Científicas CSIC‐Universidad Autónoma de MadridMadrid28029Spain
- Centro de Investigación Biomedica en Red sobre Enfermedades Cardiovasculares (CIBERCV)Madrid28029Spain
| | - Peter Tontonoz
- Department of Pathology and Laboratory MedicineUniversity of California Los AngelesUCLACalifornia90095USA
| | - Susana Alemany
- Department of Metabolic and Immune Diseases. Instituto de Investigaciones Biomédicas Sols‐MorrealeCentro Mixto Consejo Superior de Investigaciones Científicas CSIC‐Universidad Autónoma de MadridMadrid28029Spain
| | - Eckardt Treuter
- Department of Biosciences and NutritionKarolinska Institutet, NEOHuddinge14183Sweden
| | - Antonio Castrillo
- Unidad de Biomedicina (Unidad Asociada al CSIC)Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran CanariaLas Palmas35016Spain
- Department of Metabolic and Immune Diseases. Instituto de Investigaciones Biomédicas Sols‐MorrealeCentro Mixto Consejo Superior de Investigaciones Científicas CSIC‐Universidad Autónoma de MadridMadrid28029Spain
| |
Collapse
|
11
|
Wang TT, Yang CY, Peng L, Li L, Chen NT, Feng X, Xie J, Wu TC, Xu T, Chen YZ. QiShenYiQi pill inhibits atherosclerosis by promoting TTC39B-LXR mediated reverse cholesterol transport in liver. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155192. [PMID: 37951148 DOI: 10.1016/j.phymed.2023.155192] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/02/2023] [Accepted: 11/04/2023] [Indexed: 11/13/2023]
Abstract
BACKGROUND Tetranucleotide repeat domain protein 39B (TTC39B) was found to combine with ubiquitin ligase E3, and promote the ubiquitination modification of liver X receptor (LXR), which led to the inhibition of reverse cholesterol transport and development of atherosclerosis. QiShenYiQi pill (QSYQ) is a modern Chinese patent drug for treating ischemic cardiovascular diseases, the underlying mechanism is found to promote the expression of LXR-α/ ATP-binding cassette transporter G5 (ABCG5) in the liver of atherosclerotic mice. PURPOSE The aim of this study is to investigate the effect of QSYQ on TTC39B-LXR mediated reverse cholesterol transport in atherosclerotic mice. STUDY DESIGN AND METHODS Male apolipoprotein E gene knockout mice (7 weeks old) were fed with high-fat diet and treated with low dose of QSYQ (QSYQ-l, 0.3 g/kg·d), high dose of QSYQ (QSYQ-H, 1.2 g/kg·d) and LXR-α agonist (LXR-A, GW3965 10 mg/kg·d) for 8 weeks. C57BL/6 J mice were fed with normal diet and used as negative control. Oil red O staining, HE staining, ELISA, RNA sequencing, western blot, immunohistochemistry, RT-PCR, cell culture and RNA interference were performed to analyze the effect of QSYQ on atherosclerosis. RESULTS HE staining showed that QSYQ reduced the atherosclerotic lesion significantly when compared to the control group. ELISA measurement showed that QSYQ decreased serum VLDL and increased serum ApoA1. Oil Red O staining showed that QSYQ reduced the lipid content of liver and protect liver function. Comparative transcriptome RNA-sequence of liver showed that DEGs after QSYQ treatment enriched in high-density lipoprotein particle, ubiquitin ligase complex, bile secretion, etc. Immunohistochemical staining and western blot proved that QSYQ increased the protein expression of hepatic SR-B1, LXR-α, LXR-β, CYP7A1 and ABCG5. Targeted inhibiting Ttc39b gene in vitro further established that QSYQ inhibited the gene expression of Ttc39b, increased the protein expression of SR-B1, LXR-α/β, CYP7A1 and ABCG5 in rat hepatocyte. CONCLUSION Our results demonstrated the new anti-atherosclerotic mechanism of QSYQ by targeting TTC39B-LXR mediated reverse cholesterol transport in liver. QSYQ not only promoted reverse cholesterol transport, but also improved fatty liver and protected liver function.
Collapse
Affiliation(s)
- Tao-Tao Wang
- Graduate School, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550025, China
| | - Cheng-Yong Yang
- Graduate School, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550025, China
| | - Li Peng
- Department of Cardiovascular Internal Medicine, Second Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China.
| | - Li Li
- Graduate School, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550025, China
| | - Nan-Ting Chen
- Graduate School, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550025, China
| | - Xue Feng
- Graduate School, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550025, China
| | - Jing Xie
- Graduate School, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550025, China
| | - Ting-Chun Wu
- Department of Cardiovascular Internal Medicine, Second Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Tao Xu
- Department of Cardiovascular Internal Medicine, Second Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Yun-Zhi Chen
- Basic Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550025, China
| |
Collapse
|
12
|
Sun D, Chang Q, Lu F. Immunomodulation in diabetic wounds healing: The intersection of macrophage reprogramming and immunotherapeutic hydrogels. J Tissue Eng 2024; 15:20417314241265202. [PMID: 39071896 PMCID: PMC11283672 DOI: 10.1177/20417314241265202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/12/2024] [Indexed: 07/30/2024] Open
Abstract
Diabetic wound healing presents a significant clinical challenge due to the interplay of systemic metabolic disturbances and local inflammation, which hinder the healing process. Macrophages undergo a phenotypic shift from M1 to M2 during wound healing, a transition pivotal for effective tissue repair. However, in diabetic wounds, the microenvironment disrupts this phenotypic polarization, perpetuating inflammation, and impeding healing. Reprograming macrophages to restore their M2 phenotype offers a potential avenue for modulating the wound immune microenvironment and promoting healing. This review elucidates the mechanisms underlying impaired macrophage polarization toward the M2 phenotype in diabetic wounds and discusses novel strategies, including epigenetic and metabolic interventions, to promote macrophage conversion to M2. Hydrogels, with their hydrated 3D cross-linked structure, closely resemble the physiological extracellular matrix and offer advantageous properties such as biocompatibility, tunability, and versatility. These characteristics make hydrogels promising candidates for developing immunomodulatory materials aimed at addressing diabetic wounds. Understanding the role of hydrogels in immunotherapy, particularly in the context of macrophage reprograming, is essential for the development of advanced wound care solutions. This review also highlights recent advancements in immunotherapeutic hydrogels as a step toward precise and effective treatments for diabetic wounds.
Collapse
Affiliation(s)
- Dan Sun
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiang Chang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Feng Lu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
13
|
Vassiliou E, Farias-Pereira R. Impact of Lipid Metabolism on Macrophage Polarization: Implications for Inflammation and Tumor Immunity. Int J Mol Sci 2023; 24:12032. [PMID: 37569407 PMCID: PMC10418847 DOI: 10.3390/ijms241512032] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Macrophage polarization is influenced by lipids, which also exert significant control over macrophage functions. Lipids and their metabolites are players in intricate signaling pathways that modulate macrophages' responses to pathogens, phagocytosis, ferroptosis, and inflammation. This review focuses on lipid metabolism and macrophage functions and addresses potential molecular targets for the treatment of macrophage-related diseases. While lipogenesis is crucial for lipid accumulation and phagocytosis in M1 macrophages, M2 macrophages likely rely on fatty acid β-oxidation to utilize fatty acids as their primary energy source. Cholesterol metabolism, regulated by factors such as SREBPs, PPARs, and LXRs, is associated with the cholesterol efflux capacity and the formation of foam cells (M2-like macrophages). Foam cells, which are targets for atherosclerosis, are associated with an increase in inflammatory cytokines. Lipolysis and fatty acid uptake markers, such as CD36, also contribute to the production of cytokines. Enhancing the immune system through the inhibition of lipid-metabolism-related factors can potentially serve as a targeted approach against tumor cells. Cyclooxygenase inhibitors, which block the conversion of arachidonic acid into various inflammatory mediators, influence macrophage polarization and have generated attention in cancer research.
Collapse
Affiliation(s)
- Evros Vassiliou
- Department of Biological Sciences, Kean University, Union, NJ 07083, USA;
| | - Renalison Farias-Pereira
- Department of Biological Sciences, Kean University, Union, NJ 07083, USA;
- Department of Plant Biology, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| |
Collapse
|
14
|
Na Y, Ke L, Jie Z, Jinping W, Tao M, Jie Z, Liu Y, Yueqin Z. Amelioration of Cholesterol Rich diet-induced Impaired Cognition in AD Transgenic Mice by an LXR Agonist TO901317 Is Associated with the Activation of the LXR-β-RXR-α-ABCA1 Transmembrane Transport System and Improving the Composition of Lipid Raft. Exp Aging Res 2023; 49:214-225. [PMID: 35792710 DOI: 10.1080/0361073x.2022.2095605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 06/26/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND It has been reported that LXR agonist can inhibit Aβ generation and alleviate Aβ-induced various adverse reactions in vivo and in vitro experiments, but the mechanisms have not been clarified. The study aimed to observe the effect of LXR agonist TO901317 on the cognitive function of AD transgenic mice fed with cholesterol-rich diet (CRD), and to explore the possible mechanism. Methods: 32 male 6-month-old double transgenic AD mice were enrolled and randomly divided into 4 groups: control (normal diet) group, CRD treatment group, TO901317 treatment group and GSK2033 treatment group. After 3 month, Morris water maze was for the changes of spatial exploration and memory ability; ELISA was for detecting the production of Aβ42 in the brain; the concentration of total cholesterol (TC), low density lipoprotein (LDL) and high density lipoprotein (HDL) in serum were detected by cholesterol enzyme colorimetry; Finally, the expression of LXR-β, RXR-α, ABCA1, caveolin-1, BACE1 and APP at protein level in the brains was measured by Western blotting. RESULTS Compared with the control group, the learning, memory ability and spatial exploration ability of the mice were more significantly serious in the CRD group (P<0.05); The contents of TC and LDL in the serum and the production of Aβ42 in the brains were significantly increased (P<0.05), but HDL was remarkably decreased (P<0.05); The protein levels of LXR-β, RXR-α and ABCA1 were also significantly decreased (P<0.05); The expression of caveolin-1, APP and BACE1 were evidently increased (P<0.05). However, after treatment with TO901317, the impaired learning and memory and spatial exploration ability of the mice were significantly improved (P<0.05); The contents of TC and LDL in serum and the production of Aβ42 in the brains were significantly decreased (P<0.05), but HLD was increased (P<0.05); The protein levels of LXR-β, RXR-α, ABCA1were all significantly increased (P<0.05), while, the expression of caveolin-1, APP and BACE1 were all significantly decreased (P<0.05). All the changes were reversed by GSK2033 (P<0.05). CONCLUSIONS TO901317 attenuated the more serious impairment of spatial exploration, learning and memory in transgenic AD mice induced by CRD, and the mechanism may be that TO901317 could activate the LXR-β/RXR-α/ABCA1 transmembrane transport system, promote the cholesterol efflux, and decreased caveolin-1, APP and BACE1, further reduce Aβ42 in the brains.
Collapse
Affiliation(s)
- Yang Na
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Lin Ke
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Zhang Jie
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Wang Jinping
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Meng Tao
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Zhu Jie
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Yang Liu
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Zhou Yueqin
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| |
Collapse
|
15
|
Sotodosos-Alonso L, Pulgarín-Alfaro M, Del Pozo MA. Caveolae Mechanotransduction at the Interface between Cytoskeleton and Extracellular Matrix. Cells 2023; 12:cells12060942. [PMID: 36980283 PMCID: PMC10047380 DOI: 10.3390/cells12060942] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/30/2023] Open
Abstract
The plasma membrane (PM) is subjected to multiple mechanical forces, and it must adapt and respond to them. PM invaginations named caveolae, with a specific protein and lipid composition, play a crucial role in this mechanosensing and mechanotransduction process. They respond to PM tension changes by flattening, contributing to the buffering of high-range increases in mechanical tension, while novel structures termed dolines, sharing Caveolin1 as the main component, gradually respond to low and medium forces. Caveolae are associated with different types of cytoskeletal filaments, which regulate membrane tension and also initiate multiple mechanotransduction pathways. Caveolar components sense the mechanical properties of the substrate and orchestrate responses that modify the extracellular matrix (ECM) according to these stimuli. They perform this function through both physical remodeling of ECM, where the actin cytoskeleton is a central player, and via the chemical alteration of the ECM composition by exosome deposition. Here, we review mechanotransduction regulation mediated by caveolae and caveolar components, focusing on how mechanical cues are transmitted through the cellular cytoskeleton and how caveolae respond and remodel the ECM.
Collapse
Affiliation(s)
- Laura Sotodosos-Alonso
- Mechanoadaptation and Caveolae Biology Laboratory, Novel Mechanisms of Atherosclerosis Program, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
| | - Marta Pulgarín-Alfaro
- Mechanoadaptation and Caveolae Biology Laboratory, Novel Mechanisms of Atherosclerosis Program, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
| | - Miguel A Del Pozo
- Mechanoadaptation and Caveolae Biology Laboratory, Novel Mechanisms of Atherosclerosis Program, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
| |
Collapse
|
16
|
Zhan N, Wang B, Martens N, Liu Y, Zhao S, Voortman G, van Rooij J, Leijten F, Vanmierlo T, Kuipers F, Jonker JW, Bloks VW, Lütjohann D, Palumbo M, Zimetti F, Adorni MP, Liu H, Mulder MT. Identification of Side Chain Oxidized Sterols as Novel Liver X Receptor Agonists with Therapeutic Potential in the Treatment of Cardiovascular and Neurodegenerative Diseases. Int J Mol Sci 2023; 24:ijms24021290. [PMID: 36674804 PMCID: PMC9863018 DOI: 10.3390/ijms24021290] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/27/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023] Open
Abstract
The nuclear receptors-liver X receptors (LXR α and β) are potential therapeutic targets in cardiovascular and neurodegenerative diseases because of their key role in the regulation of lipid homeostasis and inflammatory processes. Specific oxy(phyto)sterols differentially modulate the transcriptional activity of LXRs providing opportunities to develop compounds with improved therapeutic characteristics. We isolated oxyphytosterols from Sargassum fusiforme and synthesized sidechain oxidized sterol derivatives. Five 24-oxidized sterols demonstrated a high potency for LXRα/β activation in luciferase reporter assays and induction of LXR-target genes APOE, ABCA1 and ABCG1 involved in cellular cholesterol turnover in cultured cells: methyl 3β-hydroxychol-5-en-24-oate (S1), methyl (3β)-3-aldehydeoxychol-5-en-24-oate (S2), 24-ketocholesterol (S6), (3β,22E)-3-hydroxycholesta-5,22-dien-24-one (N10) and fucosterol-24,28 epoxide (N12). These compounds induced SREBF1 but not SREBP1c-mediated lipogenic genes such as SCD1, ACACA and FASN in HepG2 cells or astrocytoma cells. Moreover, S2 and S6 enhanced cholesterol efflux from HepG2 cells. All five oxysterols induced production of the endogenous LXR agonists 24(S)-hydroxycholesterol by upregulating the CYP46A1, encoding the enzyme converting cholesterol into 24(S)-hydroxycholesterol; S1 and S6 may also act via the upregulation of desmosterol production. Thus, we identified five novel LXR-activating 24-oxidized sterols with a potential for therapeutic applications in neurodegenerative and cardiovascular diseases.
Collapse
Affiliation(s)
- Na Zhan
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Department of Internal Medicine, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Boyang Wang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Nikita Martens
- Department of Internal Medicine, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
| | - Yankai Liu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Shangge Zhao
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Gardi Voortman
- Department of Internal Medicine, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Jeroen van Rooij
- Department of Internal Medicine, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Frank Leijten
- Department of Internal Medicine, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Tim Vanmierlo
- Department of Neuroscience, Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
- School for Mental Health and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Folkert Kuipers
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Johan W. Jonker
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Vincent W. Bloks
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53105 Bonn, Germany
| | - Marcella Palumbo
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| | - Maria Pia Adorni
- Unit of Neurosciences, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy
| | - Hongbing Liu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Correspondence: (H.L.); (M.T.M.)
| | - Monique T. Mulder
- Department of Internal Medicine, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
- Correspondence: (H.L.); (M.T.M.)
| |
Collapse
|
17
|
Wißfeld J, Werner A, Yan X, ten Bosch N, Cui G. Metabolic regulation of immune responses to cancer. Cancer Biol Med 2022; 19:j.issn.2095-3941.2022.0381. [PMID: 36269001 PMCID: PMC9724228 DOI: 10.20892/j.issn.2095-3941.2022.0381] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The tumor microenvironment is an ecosystem composed of multiple types of cells, such as tumor cells, immune cells, and cancer-associated fibroblasts. Cancer cells grow faster than non-cancerous cells and consume larger amounts of nutrients. The rapid growth characteristic of cancer cells fundamentally alters nutrient availability in the tumor microenvironment and results in reprogramming of immune cell metabolic pathways. Accumulating evidence suggests that cellular metabolism of nutrients, such as lipids and amino acids, beyond being essential to meet the bioenergetic and biosynthetic demands of immune cells, also regulates a broad spectrum of cellular signal transduction, and influences immune cell survival, differentiation, and anti-tumor effector function. The cancer immunometabolism research field is rapidly evolving, and exciting new discoveries are reported in high-profile journals nearly weekly. Therefore, all new findings in this field cannot be summarized within this short review. Instead, this review is intended to provide a brief introduction to this rapidly developing research field, with a focus on the metabolism of two classes of important nutrients-lipids and amino acids-in immune cells. We highlight recent research on the roles of lipids and amino acids in regulating the metabolic fitness and immunological functions of T cells, macrophages, and natural killer cells in the tumor microenvironment. Furthermore, we discuss the possibility of "editing" metabolic pathways in immune cells to act synergistically with currently available immunotherapies in enhancing anti-tumor immune responses.
Collapse
Affiliation(s)
- Jannis Wißfeld
- Helmholtz Institute for Translational Oncology (HI-TRON), Mainz 55131, Germany,T Cell Metabolism Group (D192), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Anke Werner
- Helmholtz Institute for Translational Oncology (HI-TRON), Mainz 55131, Germany,T Cell Metabolism Group (D192), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Xin Yan
- Helmholtz Institute for Translational Oncology (HI-TRON), Mainz 55131, Germany,T Cell Metabolism Group (D192), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany,Faculty of Biosciences, Heidelberg University, Heidelberg 69120, Germany
| | - Nora ten Bosch
- Helmholtz Institute for Translational Oncology (HI-TRON), Mainz 55131, Germany,T Cell Metabolism Group (D192), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Guoliang Cui
- Helmholtz Institute for Translational Oncology (HI-TRON), Mainz 55131, Germany,T Cell Metabolism Group (D192), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany,Faculty of Biosciences, Heidelberg University, Heidelberg 69120, Germany,Correspondence to: Guoliang Cui, E-mail:
| |
Collapse
|
18
|
Aranda JF, Pérez-García A, Torrecilla-Parra M, Fernández-de Frutos M, Martín-Martín Y, Mateos-Gómez PA, Pardo-Marqués V, Busto R, Ramírez CM. Role of miR-199a-5p in the post-transcriptional regulation of ABCA1 in response to hypoxia in peritoneal macrophages. Front Cardiovasc Med 2022; 9:994080. [PMID: 36407436 PMCID: PMC9669644 DOI: 10.3389/fcvm.2022.994080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/19/2022] [Indexed: 01/03/2025] Open
Abstract
Hypoxia is a crucial factor contributing to maintenance of atherosclerotic lesions. The ability of ABCA1 to stimulate the efflux of cholesterol from cells in the periphery, particularly foam cells in atherosclerotic plaques, is an important anti-atherosclerotic mechanism. The posttranscriptional regulation by miRNAs represents a key regulatory mechanism of a number of signaling pathways involved in atherosclerosis. Previously, miR-199a-5p has been shown to be implicated in the endocytic and retrograde intracellular transport. Although the regulation of miR-199a-5p and ABCA1 by hypoxia has been already reported independently, the role of miR-199a-5p in macrophages and its possible role in atherogenic processes such us regulation of lipid homeostasis through ABCA1 has not been yet investigated. Here, we demonstrate that both ABCA1 and miR-199a-5p show an inverse regulation by hypoxia and Ac-LDL in primary macrophages. Moreover, we demonstrated that miR-199a-5p regulates ABCA1 mRNA and protein levels by directly binding to its 3'UTR. As a result, manipulation of cellular miR-199a-5p levels alters ABCA1 expression and cholesterol efflux in primary mouse macrophages. Taken together, these results indicate that the correlation between ABCA1-miR-199a-5p could be exploited to control macrophage cholesterol efflux during the onset of atherosclerosis, where cholesterol alterations and hypoxia play a pathogenic role.
Collapse
Affiliation(s)
- Juan Francisco Aranda
- Department of Basic Medical Sciences, CEU San Pablo University, CEU Universities, Madrid, Spain
| | - Ana Pérez-García
- IMDEA Research Institute of Food and Health Sciences, Madrid, Spain
| | | | | | | | - Pedro A. Mateos-Gómez
- Department of Systems Biology, School of Medicine and Health Sciences, University of Alcalá, Madrid, Spain
| | | | - Rebeca Busto
- Department of Clinical Biochemistry, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | | |
Collapse
|
19
|
Stifel U, Caratti G, Tuckermann J. Novel insights into the regulation of cellular catabolic metabolism in macrophages through nuclear receptors. FEBS Lett 2022; 596:2617-2629. [PMID: 35997656 DOI: 10.1002/1873-3468.14474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 06/14/2022] [Accepted: 07/04/2022] [Indexed: 11/07/2022]
Abstract
Regulation of cellular catabolic metabolism in immune cells has recently become a major concept for resolution of inflammation. Nuclear receptors (NRs), including peroxisome proliferator activator receptors (PPARs), 1,25-dihydroxyvitamin D(3) receptor (VDR), liver X receptors (LXRs), glucocorticoid receptors (GRs), estrogen-related receptor α (ERRα) and Nur77, have been identified as major modulators of inflammation, affecting innate immune cells, such as macrophages. Evidence emerges on how NRs regulate cellular metabolism in macrophages during inflammatory processes and contribute to the resolution of inflammation. This could have new implications for our understanding of how NRs shape immune responses and inform anti-inflammatory drug design. This review will highlight the recent developments about NRs and their role in cellular metabolism in macrophages.
Collapse
Affiliation(s)
- Ulrich Stifel
- Institute of Comparative Molecular Endocrinology (CME), Ulm University, Ulm, Germany
| | - Giorgio Caratti
- Institute of Comparative Molecular Endocrinology (CME), Ulm University, Ulm, Germany.,NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology (CME), Ulm University, Ulm, Germany
| |
Collapse
|
20
|
Ong KL, Cochran BBiotech BJ, Manandhar B, Thomas S, Rye KA. HDL maturation and remodelling. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159119. [PMID: 35121104 DOI: 10.1016/j.bbalip.2022.159119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/16/2022] [Accepted: 01/20/2022] [Indexed: 11/29/2022]
Abstract
Cholesterol in the circulation is mostly transported in an esterified form as a component of lipoproteins. The majority of these cholesteryl esters are produced in nascent, discoidal high density lipoproteins (HDLs) by the enzyme, lecithin:cholesterol acyltransferase (LCAT). Discoidal HDLs are discrete populations of particles that consist of a phospholipid bilayer, the hydrophobic acyl chains of which are shielded from the aqueous environment by apolipoproteins that also confer water solubility on the particles. The progressive LCAT-mediated accumulation of cholesteryl esters in discoidal HDLs generates the spherical HDLs that predominate in normal human plasma. Spherical HDLs contain a core of water insoluble, neutral lipids (cholesteryl esters and triglycerides) that is surrounded by a surface monolayer of phospholipids with which apolipoproteins associate. Although spherical HDLs all have the same basic structure, they are extremely diverse in size, composition, and function. This review is concerned with how the biogenesis of discoidal and spherical HDLs is regulated and the mechanistic basis of their size and compositional heterogeneity. Current understanding of the impact of this heterogeneity on the therapeutic potential of HDLs of varying size and composition is also addressed in the context of several disease states.
Collapse
Affiliation(s)
- Kwok-Leung Ong
- School of Medical Sciences, Faculty of Medicine, University of New South Wales Sydney, New South Wales, Australia
| | - Blake J Cochran BBiotech
- School of Medical Sciences, Faculty of Medicine, University of New South Wales Sydney, New South Wales, Australia
| | - Bikash Manandhar
- School of Medical Sciences, Faculty of Medicine, University of New South Wales Sydney, New South Wales, Australia
| | - Shane Thomas
- School of Medical Sciences, Faculty of Medicine, University of New South Wales Sydney, New South Wales, Australia
| | - Kerry-Anne Rye
- School of Medical Sciences, Faculty of Medicine, University of New South Wales Sydney, New South Wales, Australia.
| |
Collapse
|