1
|
Qi X, Guo H, Xia X, Liu Y, Qiu S, Lin T, He W, Jin L, Cheng J, Hao L, Liu W, Hu H. Paeoniflorin alleviated STZ-induced diabetic retinopathy via regulation of the PDI/ADAM17/MerTK pathway. Int Immunopharmacol 2025; 155:114571. [PMID: 40209310 DOI: 10.1016/j.intimp.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: 12/16/2024] [Revised: 02/11/2025] [Accepted: 03/26/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Diabetic retinopathy (DR) is a severe microvascular complication of diabetes and a leading cause of vision impairment in diabetic patients. The accumulation of apoptotic cells and inflammation are key pathological mechanisms in DR. The Mer tyrosine kinase (MerTK) receptor plays a critical role in maintaining retinal homeostasis. Proteolytic cleavage of MerTK by disintegrin and metalloproteinase-17 (ADAM17) disrupts MerTK-dependent clearance of apoptotic cells and diminishes its anti-inflammatory effects. Therefore, reducing the cleavage activity ADAM17's and promoting MerTK-dependent anti-inflammatory effects may represent potent strategy to alleviate DR. METHODS The DR mouse model was established using streptozotocin (STZ), and a high-glucose (HG)-induced in vitro model was developed using human retinal pigment epithelial (ARPE-19) cells. Relevant signaling molecules were analyzed through western blotting and immunohistochemistry. RESULTS Hyperglycemia promoted the accumulation of apoptotic cells and disrupted retinal microvascular growth. In both vivo and vitro model, MerTK expression was significantly reduced, while ADAM17 phosphorylation levels were markedly increased. In STZ-treated mice, protein disulfide isomerase (PDI) secretion initially rose but subsequently declined, whereas PDI secretion decreased under HG conditions. We then utilized paeoniflorin to increase the expression of this endogenous inhibitor of ADAM17. Results showed that paeoniflorin upregulated PDI production, suppressed ADAM17 expression, and enhanced MerTK phosphorylation in the eye tissues of STZ-induced mice. Additionally, paeoniflorin elevated the expression of suppressor of cytokine signaling 3 (SOCS3) and decreased the level of matrix metalloproteinase 9 (MMP9) both in vivo and in vitro. CONCLUSION Paeoniflorin may alleviate diabetic retinopathy by suppressing inflammation through modulation of the PDI/ADAM17/MerTK signaling pathway.
Collapse
Affiliation(s)
- Xiuting Qi
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Haiyue Guo
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Xinyue Xia
- The First Clinical College, Nanjing Medical University, Nanjing 211166, China
| | - Yanmei Liu
- The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, Jiangsu 224005, China
| | - Shenghui Qiu
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Tongtong Lin
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligence Manufacture, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Wenqi He
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Lai Jin
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Jing Cheng
- Department of Gastroenterology, Lianyungang Municipal Oriental Hospital, Lianyungang, Jiangsu, China; Department of Gastroenterology,Shanghai General Hospial of Nanjing Medical University, Shanghai, China
| | - Lanxiang Hao
- The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, Jiangsu 224005, China.
| | - Wentao Liu
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211100, China.
| | - Haitao Hu
- The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, Jiangsu 224005, China.
| |
Collapse
|
2
|
Kanuri B, Maremanda KP, Chattopadhyay D, Essop MF, Lee MKS, Murphy AJ, Nagareddy PR. Redefining Macrophage Heterogeneity in Atherosclerosis: A Focus on Possible Therapeutic Implications. Compr Physiol 2025; 15:e70008. [PMID: 40108774 DOI: 10.1002/cph4.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/20/2025] [Accepted: 03/08/2025] [Indexed: 03/22/2025]
Abstract
Atherosclerosis is a lipid disorder where modified lipids (especially oxidized LDL) induce macrophage foam cell formation in the aorta. Its pathogenesis involves a continuum of persistent inflammation accompanied by dysregulated anti-inflammatory responses. Changes in the immune cell status due to differences in the lesional microenvironment are crucial in terms of plaque development, its progression, and plaque rupture. Ly6Chi monocytes generated through both medullary and extramedullary cascades act as one of the major sources of plaque macrophages and thereby foam cells. Both monocytes and monocyte-derived macrophages also participate in pathological events in atherosclerosis-associated multiple organ systems through inter-organ communications. For years, macrophage phenotypes M1 and M2 have been shown to perpetuate inflammatory and resolution responses; nevertheless, such a dualistic classification is too simplistic and contains severe drawbacks. As the lesion microenvironment is enriched with multiple mediators that possess the ability to activate macrophages to diverse phenotypes, it is obvious that such cells should demonstrate substantial heterogeneity. Considerable research in this regard has indicated the presence of additional macrophage phenotypes that are exclusive to atherosclerotic plaques, namely Mox, M4, Mhem, and M(Hb) type. Furthermore, although the concept of macrophage clusters has come to the fore in recent years with the evolution of high-dimensional techniques, classifications based on such 'OMICS' approaches require extensive functional validation as well as metabolic phenotyping. Bearing this in mind, the current review provides an overview of the status of different macrophage populations and their role during atherosclerosis and also outlines possible therapeutic implications.
Collapse
Affiliation(s)
- Babunageswararao Kanuri
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, USA
| | - Krishna P Maremanda
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, USA
| | - Dipanjan Chattopadhyay
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, USA
| | - M Faadiel Essop
- Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Man Kit Sam Lee
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Andrew J Murphy
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Prabhakara R Nagareddy
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, USA
| |
Collapse
|
3
|
Oggero S, Voisin MB, Picco F, Huerta MÁ, Cecconello C, Burgoyne T, Perretti M, Malcangio M. Activation of proresolving macrophages in dorsal root ganglia attenuates persistent arthritis pain. Proc Natl Acad Sci U S A 2025; 122:e2416343122. [PMID: 40063821 PMCID: PMC11929478 DOI: 10.1073/pnas.2416343122] [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: 08/15/2024] [Accepted: 01/31/2025] [Indexed: 03/25/2025] Open
Abstract
Pain independent of disease activity is frequently reported by rheumatoid arthritis patients and remains undertreated. Preclinical evidence suggests that imbalance of neuroimmune proresolving interactions within dorsal root ganglia (DRG) rather than at the site of inflammation plays mechanistic roles in persistent arthritis pain. Here, we inhibited production of proresolving lipid mediators by silencing 12/15-lipoxygenase expression in CX3CR1+ monocyte/macrophages conditional knockout (cKO) mice. In an arthritis model, hind paw mechanical hypersensitivity is exacerbated in male and female cKO mice in association with DRG infiltration of neutrophils, which migrate in response to leukotriene B4 released by macrophages through 5-lipoxygenase conversion of arachidonic acid provided by neuron-derived vesicles. Neutrophils apoptosis promotes primary macrophage efferocytosis which is defective in cKO macrophages. In wild-type (WT) and cKO mice, intrathecal injection of MerTK activating antibody, attenuates persistent hypersensitivity and polarizes DRG macrophages toward a proresolving phenotype with production of antinociceptive lipoxin A4. Thus, we delineate a neuron-macrophage-neutrophil bidirectional circuit that can be exploited to reduce persistent arthritis pain.
Collapse
Affiliation(s)
- Silvia Oggero
- Sensory, Pain and Regeneration Centre, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, Guys’ Campus, LondonSE1 1UL, United Kingdom
| | - Mathieu-Benoit Voisin
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, LondonEC1M 6BQ, United Kingdom
| | - Francesca Picco
- Sensory, Pain and Regeneration Centre, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, Guys’ Campus, LondonSE1 1UL, United Kingdom
| | - Miguel Á. Huerta
- Sensory, Pain and Regeneration Centre, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, Guys’ Campus, LondonSE1 1UL, United Kingdom
- Department of Pharmacology, University of Granada, Granada18016, Spain
| | - Chiara Cecconello
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, LondonEC1M 6BQ, United Kingdom
| | - Thomas Burgoyne
- Institute of Ophthalmology, Faculty of Brain Sciences, University College London, LondonEC1V 9EL, United Kingdom
- Pediatric Respiratory Medicine, Royal Brompton Hospital, Guy’s and St Thomas’ National Health System Foundation Trust, LondonSW3 6NP, United Kingdom
| | - Mauro Perretti
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, LondonEC1M 6BQ, United Kingdom
| | - Marzia Malcangio
- Sensory, Pain and Regeneration Centre, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, Guys’ Campus, LondonSE1 1UL, United Kingdom
| |
Collapse
|
4
|
de Jager C, Soliman E, Theus MH. Interrogating mediators of single-cell transcriptional changes in the acute damaged cerebral cortex: Insights into endothelial-astrocyte interactions. Mol Cell Neurosci 2025; 133:104003. [PMID: 40090391 DOI: 10.1016/j.mcn.2025.104003] [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: 11/26/2024] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/18/2025] Open
Abstract
Traumatic brain injury (TBI) induces complex cellular and molecular changes, challenging recovery and therapeutic development. Although molecular pathways have been implicated in TBI pathology, the cellular specificity of these mechanisms remains underexplored. Here, we investigate the role of endothelial cell (EC) EphA4, a receptor tyrosine kinase receptor involved in axonal guidance, in modulating cell-specific transcriptomic changes within the damaged cerebral cortex. Utilizing single-cell RNA sequencing (scRNA-seq) in an experimental TBI model, we mapped transcriptional changes across various cell types, with a focus on astrocytes and ECs. Our analysis reveals that EC-specific knockout (KO) of EphA4 triggers significant alterations in astrocyte gene expression and shifts predominate subclusters. We identified six distinct astrocyte clusters (C0-C5) in the damaged cortex including as C0-Mobp/Plp1+; C1-Slc1a3/Clu+; C2-Hbb-bs/Hba-a1/Ndrg2+; C3-GFAP/Lcn2+; C4-Gli3/Mertk+, and C5-Cox8a+. We validate a new Sox9+ cluster expressing Mertk and Gas, which mediates efferocytosis to facilitate apoptotic cell clearance and anti-inflammatory responses. Transcriptomic and CellChat analyses of EC-KO cells highlights upregulation of neuroprotective pathways, including increased amyloid precursor protein (APP) and Gas6. Key pathways predicted to be modulated in astrocytes from EC-KO mice include oxidative phosphorylation and FOXO signaling, mitochondrial dysfunction and ephrin B signaling. Concurrently, metabolic and signaling pathways in endothelial cells-such as ceramide and sphingosine phosphate metabolism and NGF-stimulated transcription-indicate an adaptive response to a metabolically demanding post-injury hypoxic environment. These findings elucidate potential interplay between astrocytic and endothelial responses as well as transcriptional networks underlying cortical tissue damage.
Collapse
Affiliation(s)
- Caroline de Jager
- Translational Biology Medicine and Health Graduate Program, Blacksburg, VA 24061, USA
| | - Eman Soliman
- Department of Biomedical Sciences and Pathobiology, Blacksburg, VA 24061, USA
| | - Michelle H Theus
- Department of Biomedical Sciences and Pathobiology, Blacksburg, VA 24061, USA; Center for Engineered Health, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
5
|
Lantz C, Becker A, DeBerge M, Filipp M, Glinton K, Ananthakrishnan A, Urbanczyk J, Cetlin M, Alzamroon A, Abdel-Latif A, Spite M, Ge ZD, Thorp EB. Early-age efferocytosis directs macrophage arachidonic acid metabolism for tissue regeneration. Immunity 2025; 58:344-361.e7. [PMID: 39938482 PMCID: PMC11839170 DOI: 10.1016/j.immuni.2024.11.018] [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: 08/16/2023] [Revised: 09/20/2024] [Accepted: 11/21/2024] [Indexed: 02/14/2025]
Abstract
In response to organ injury in adults, macrophages often promote scarring, yet during early life, they are required for tissue regeneration. To elucidate the mechanisms underlying age-associated regeneration, we compared the macrophage injury response in newborn versus adult hearts. Single-cell analysis revealed an accumulation of tissue-resident macrophages in neonates that were selectively polarized for apoptotic cell recognition and uptake (efferocytosis). Ablation of the apoptotic cell recognition receptor Mertk in newborns prevented cardiac regeneration. These findings could be attributed to reprogramming of macrophage gene expression that was required for biosynthesis of the eicosanoid thromboxane A2, which unexpectedly activated parenchymal cell proliferation. Markers of thromboxane A2 production were suppressed in adult macrophages after efferocytosis. Moreover, macrophage-neighboring neonatal cardiomyocytes expressed the thromboxane A2 receptor, whose activation induced a metabolic shift that supported cellular proliferation. Our data reveal a fundamental age-defined macrophage response in which lipid mitogens produced during efferocytosis support receptor-mediated tissue regeneration.
Collapse
Affiliation(s)
- Connor Lantz
- Department of Pathology, Feinberg School of Medicine, Chicago, IL 60611, USA; Comprehensive Transplant Center, Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611, USA.
| | - Amanda Becker
- Department of Pediatrics, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Matthew DeBerge
- Department of Pathology, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Mallory Filipp
- Department of Pathology, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Kristofor Glinton
- Department of Pathology, Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | - Jessica Urbanczyk
- Department of Pathology, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Madeline Cetlin
- Department of Pathology, Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | - Matthew Spite
- Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Zhi-Dong Ge
- Department of Pathology, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Edward B Thorp
- Department of Pathology, Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Pediatrics, Feinberg School of Medicine, Chicago, IL 60611, USA; Heart Center, Stanley Manne Children's Research Institute, Ann & Robert Lurie Children's Hospital, Chicago, IL 60611, USA; Comprehensive Transplant Center, Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
6
|
Liu Y, Hu W, Yang F, Zou S, Ren H, Zuo Y, Qu L. Chitinase-3-like Protein 1 Reduces the Stability of Atherosclerotic Plaque via Impairing Macrophagic Efferocytosis. J Cardiovasc Transl Res 2025; 18:3-16. [PMID: 39813006 DOI: 10.1007/s12265-024-10576-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: 03/10/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025]
Abstract
CHI3L1 is strongly associated with atherosclerosis, but its role in macrophages remains unknown. In this study, we observed a significant up-regulation of CHI3L1 in both carotid plaques and serum of symptomatic patients, and demonstrated that CHI3L1 impairs the efferocytosis of macrophages by down-regulating crucial efferocytic mediator MFGE8 through inhibiting ATF2, which binds directly to the enhancer of MFGE8. In human plaques, we observed a negative correlation between CHI3L1 expression and both ATF2 and MFGE8 levels, further proved their involvement in plaque destabilization. Using Ldlr-/- mice with tandem carotid stenosis surgery, we demonstrated that administration of CHI3L1 protein resulted in enlarged atherosclerotic necrotic cores and decreased MFGE8 and ATF2 levels. Conversely, treatment with a CHI3L1 blocking antibody exhibited the opposite trend.In conclusion, CHI3L1 destabilizes atherosclerotic plaque by impairing macrophagic efferocytosis through the down-regulation of ATF2-induced MFGE8 expression. Targeting CHI3L1 may offer a promising therapeutic strategy for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yandong Liu
- Department of Geriatrics, 905, Hospital of PLA NAVY, Shanghai, China
- Department of Vascular and Endovascular Surgery, Changzheng Hospital, Affiliated to the Naval Medical University, Shanghai, 200003, China
| | - Weilin Hu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital, Affiliated to the Naval Medical University, Shanghai, 200003, China
| | - Futang Yang
- Department of Vascular and Endovascular Surgery, Changzheng Hospital, Affiliated to the Naval Medical University, Shanghai, 200003, China
| | - Sili Zou
- Department of Vascular and Endovascular Surgery, Changzheng Hospital, Affiliated to the Naval Medical University, Shanghai, 200003, China
| | - Huiqiong Ren
- Department of Geriatrics, 905, Hospital of PLA NAVY, Shanghai, China.
| | - Yong Zuo
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lefeng Qu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital, Affiliated to the Naval Medical University, Shanghai, 200003, China.
| |
Collapse
|
7
|
Wang J, Li M, Zhao B, Chang R, Wu W, Zhang H, Usman M, Loor JJ, Xu C. A Disintegrin and Metalloproteinase 17 Disrupts Bovine Macrophage MER Proto-Oncogene Tyrosine Kinase Integrity to Impede Apoptotic Cell Clearance and Promote Inflammation in Clinical Mastitis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:549-561. [PMID: 39731564 DOI: 10.1021/acs.jafc.4c09164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2024]
Abstract
In clinical mastitis of dairy cows, the abnormal accumulation of apoptotic cells (ACs) and subsequent secondary necrosis and inflammation pose significant concerns, with macrophage-mediated efferocytosis, crucial for ACs clearance, remaining unexplored in this context. In nonruminants, MER proto-oncogene tyrosine kinase (MERTK) receptors are essential for efferocytosis and A Disintegrin and Metalloproteinase 17 (ADAM17) is thought to play a role in regulating MERTK integrity. This study aimed to delineate the in situ role of efferocytosis in clinical mastitis, with a particular focus on the interaction between MERTK and ADAM17 in bovine macrophages. In mastitic mammary tissue, a significant accumulation of ACs was observed, along with active macrophage efferocytosis. Western blotting analysis revealed elevated expressions of MERTK and ADAM17, and immunofluorescence confirmed that MERTK is predominantly localized within CD163+ macrophages. Additionally, elevated levels of soluble MERTK (sol-MER) in serum indicated impaired integrity and functionality of MERTK. In vitro experiments with the bovine macrophage cell line Bo-mac cells selectively phagocytosed apoptotic MAC-T cells, a process associated with increased MERTK phosphorylation and an anti-inflammatory phenotype. The activation of ADAM17 by Phorbol 12-myristate 13-acetate (PMA) induced sol-Mer release and impaired efferocytosis, with these effects reversed by the ADAM17 inhibitor TAPI-1. Bo-mac efferocytosis was influenced by the presence and activation of MERTK. Silencing MERTK interrupted efferocytosis, a phenomenon also observed with the inhibition of MERTK phosphorylation by UNC2025, which concurrently suppressed anti-inflammatory cytokine production. These findings suggest that targeting the MERTK-ADAM17 axis could enhance inflammatory resolution, providing a novel therapeutic strategy for dairy cattle health management.
Collapse
Affiliation(s)
- Jingyi Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road, Beijing 100193, China
| | - Ming Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road, Beijing 100193, China
| | - Bichen Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road, Beijing 100193, China
| | - Renxu Chang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road, Beijing 100193, China
| | - Wenda Wu
- School of Food and Biological Engineering, Engineering Research Center of Bio-Process, Ministry of Education, Hefei University of Technology, Hefei 230009, China
| | - Huijing Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road, Beijing 100193, China
| | - Muhammad Usman
- Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Juan J Loor
- Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Chuang Xu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road, Beijing 100193, China
| |
Collapse
|
8
|
Bu S, Ling JY, Wu X, Zhang L, Shi X, Huang L, Zhao Z, Yang Y, Xiang Z, Liu YU, Liu Y, Zhang Y. Downregulation of MerTK in circulating T cells of patients with non-proliferative diabetic retinopathy. Front Endocrinol (Lausanne) 2025; 15:1509445. [PMID: 39845889 PMCID: PMC11750652 DOI: 10.3389/fendo.2024.1509445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/09/2024] [Indexed: 01/24/2025] Open
Abstract
Objective To explore the differential gene expression in peripheral blood immune cells of individuals with type 2 diabetes mellitus (DM), comparing those with and without non-proliferative diabetic retinopathy (NPDR). Methods From a pool of 126 potential participants, 60 were selected for detailed analysis. This group included 12 healthy donors (HDs), 22 individuals with DM, and 26 with NPDR. We analyzed peripheral blood mononuclear cells (PBMCs) using RNA sequencing and quantitative PCR (qPCR) to pinpoint differentially expressed genes (DEGs). Western blot and flow cytometry were also employed to evaluate the protein expression of specific genes. Results In patients with NPDR compared to those with DM alone, MerTK-a gene implicated in inherited retinal dystrophies due to its mutations-was notably downregulated in PBMCs. Through flow cytometry, we assessed the protein levels and cellular distribution of MerTK, finding a predominant expression in monocytes and myeloid-derived suppressor cells (MDSCs), with a marked reduction in CD4+ and CD8+ T cells, as well as in natural killer T (NKT) cells. Patients with DM demonstrated a significant deviation in the PBMCs composition, particularly in B cells, CD4+ T cells, and NK cells, when compared to HDs. Conclusions The study indicates that MerTK expression in T cells within PBMCs could act as a viable blood biomarker for NPDR risk in patients with DM. Furthermore, the regulation of T cells by MerTK might represent a critical pathway through which DM evolves into NPDR.
Collapse
Affiliation(s)
- Shimiao Bu
- Department of Ophthalmology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jiang-Yue Ling
- Department of Ophthalmology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiaojun Wu
- Department of Ophthalmology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Liting Zhang
- Department of Ophthalmology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiangyu Shi
- Department of Ophthalmology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Lang Huang
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Zheng Zhao
- Department of Neurology, The First Affiliated Hospital, School of Medicine, Jinan University, Guangzhou, China
| | - Ying Yang
- Department of Neurology, The First Affiliated Hospital, School of Medicine, Jinan University, Guangzhou, China
| | - Zongqin Xiang
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yong U. Liu
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Yufeng Liu
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yuehong Zhang
- Department of Ophthalmology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
9
|
Vanoni EM, Enderlin J, Rieu Q, Hamieh F, Réty S, Nandrot EF. Oxidative Stress and Energetic Failure: Common Features and Dissimilarities in 3 Different Mouse Models of Retinal Pigment Epithelium Phagocytosis Defects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1468:259-263. [PMID: 39930206 DOI: 10.1007/978-3-031-76550-6_43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Amidst the various crucial functions ensured by retinal pigment epithelial (RPE) cells is the circadian phagocytosis of oxidized photoreceptor outer segments (POS) extremities. We have been exploring three mouse models with defective RPE phagocytosis: β5-/- mice inactivated for the αvβ5 integrin synchronizing phagocytosis, MerTKCR knockin mice devoid of the MerTK internalization receptor cleavage site, and Pre-mRNA Processing Factors 31 knockout mice, PRPF splicing factor mutations constituting the second most important cause of autosomic dominant retinitis pigmentosa in patients. Failure in mitochondrial activity and energetic metabolism has been detected in all three models. Signs of cellular stress and increasing oxidative processes were observed in β5-/- and Prpf31+/- RPE cells, while MerTKCR mutants seem to be sensitive to light-derived stress associated with augmented retinal inflammation. Taken together, these results highlight some common pathological mechanisms in these mice, as well as particular features related to the specific function of each protein.
Collapse
Affiliation(s)
- Elora M Vanoni
- Sorbonne Université, CNRS, Inserm, Institut de la Vision, F-75012, Paris, France
| | - Julie Enderlin
- Sorbonne Université, CNRS, Inserm, Institut de la Vision, F-75012, Paris, France
| | - Quentin Rieu
- Sorbonne Université, CNRS, Inserm, Institut de la Vision, F-75012, Paris, France
| | - Florian Hamieh
- Sorbonne Université, CNRS, Inserm, Institut de la Vision, F-75012, Paris, France
| | - Salomé Réty
- Sorbonne Université, CNRS, Inserm, Institut de la Vision, F-75012, Paris, France
| | - Emeline F Nandrot
- Sorbonne Université, CNRS, Inserm, Institut de la Vision, F-75012, Paris, France.
| |
Collapse
|
10
|
Chen F, Li Y, Zhao L, Lin C, Zhou Y, Ye W, Wan W, Zou H, Xue Y. Anti-inflammatory effects of MerTK by inducing M2 macrophage polarization via PI3K/Akt/GSK-3β pathway in gout. Int Immunopharmacol 2024; 142:112942. [PMID: 39217874 DOI: 10.1016/j.intimp.2024.112942] [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: 04/09/2024] [Revised: 08/03/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
Mer tyrosine kinase (MerTK) has been found to regulate the secretion of inflammatory factors and exert immunosuppressive effects, but its role in gout remains unclear. In this study, we aimed to clarify the immnue effects of MerTK in gout. MerTK in synovium or serum of gout patients was determined by immunohistochemistry, enzyme-linked immunosorbent assay (ELISA), and real-time quantitative polymerase chain reaction (RT-qPCR). In monosodium urate (MSU)-induced gout mice, the effect of MerTK inhibitor (UNC2250) on inflammation and polarization was also assessed. After inhibition, knockdown or overexpression of MerTK, inflammatory response and polarization level in THP1-derived macrophages were evaluated by RT-qPCR and flow cytometry. Regulation of MerTK inhibitors on mitochondrial function and downstream pathway in THP1-derived macrophages were detected. MerTK in synovium and serum of gout patients were increased. MerTK inhibitor stimulated the inflammation and M1 polarization in MSU-induced gout mice. MerTK inhibition, knock-down, or overexpression affected inflammatory response, polarization and mitochondrial function in vitro in gout model. The PI3K/Akt/GSK-3β pathway was identified to reduce after MerTK inhibition and the relevant results were as expected, validated by knock-down or overexpressing MerTK. In conclusion, MerTK was detected to increase in both gout patients and model. MerTK influenced inflammatory response and polarization markers through PI3K/Akt/GSK-3β pathway. Interfering MerTK/PI3K/Akt/GSK-3β axis may provide a new therapeutic target for gout.
Collapse
Affiliation(s)
- Fangfang Chen
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Yixuan Li
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Li Zhao
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Cong Lin
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Yingzi Zhou
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Wenjing Ye
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Weiguo Wan
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Hejian Zou
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China.
| | - Yu Xue
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China.
| |
Collapse
|
11
|
Prouse T, Majumder S, Majumder R. Functions of TAM Receptors and Ligands Protein S and Gas6 in Atherosclerosis and Cardiovascular Disease. Int J Mol Sci 2024; 25:12736. [PMID: 39684449 DOI: 10.3390/ijms252312736] [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: 09/27/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Atherosclerosis and cardiovascular disease are associated with high morbidity and mortality in industrialized nations. The Tyro3, Axl, and Mer (TAM) family of receptor tyrosine kinases is involved in the amplification or resolution of atherosclerosis pathology and other cardiovascular pathology. The ligands of these receptors, Protein S (PS) and growth arrest specific protein 6 (Gas6), are essential for TAM receptor functions in the amplification and resolution of atherosclerosis. The Axl-Gas6 interaction has various effects on cardiovascular disease. Mer and PS dampen inflammation, thereby protecting against atherosclerosis progression. Tyro3, the least studied TAM receptor in cardiovascular disease, appears to protect against fibrosis in post-myocardial infarction injury. Ultimately, PS, Gas6, and TAM receptors present an exciting avenue of potential therapeutic targets against inflammation associated with atherosclerosis and cardiovascular disease.
Collapse
Affiliation(s)
- Teagan Prouse
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Samarpan Majumder
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Rinku Majumder
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
12
|
Zhang B, Zou Y, Yuan Z, Jiang K, Zhang Z, Chen S, Zhou X, Wu Q, Zhang X. Efferocytosis: the resolution of inflammation in cardiovascular and cerebrovascular disease. Front Immunol 2024; 15:1485222. [PMID: 39660125 PMCID: PMC11628373 DOI: 10.3389/fimmu.2024.1485222] [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: 08/23/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
Cardiovascular and cerebrovascular diseases have surpassed cancer as significant global health challenges, which mainly include atherosclerosis, myocardial infarction, hemorrhagic stroke and ischemia stroke. The inflammatory response immediately following these diseases profoundly impacts patient prognosis and recovery. Efficient resolution of inflammation is crucial not only for halting the inflammatory process but also for restoring tissue homeostasis. Efferocytosis, the phagocytic clearance of dying cells by phagocytes, especially microglia and macrophages, plays a critical role in this resolution process. Upon tissue injury, phagocytes are recruited to the site of damage where they engulf and clear dying cells through efferocytosis. Efferocytosis suppresses the secretion of pro-inflammatory cytokines, stimulates the production of anti-inflammatory cytokines, modulates the phenotype of microglia and macrophages, accelerates the resolution of inflammation, and promotes tissue repair. It involves three main stages: recognition, engulfment, and degradation of dying cells. Optimal removal of apoptotic cargo by phagocytes requires finely tuned machinery and associated modifications. Key molecules in efferocytosis, such as 'Find-me signals', 'Eat-me signals', and 'Don't eat-me signals', have been shown to enhance efferocytosis following cardiovascular and cerebrovascular diseases. Moreover, various additional molecules, pathways, and mitochondrial metabolic processes have been identified to enhance prognosis and outcomes via efferocytosis in diverse experimental models. Impaired efferocytosis can lead to inflammation-associated pathologies and prolonged recovery periods. Therefore, this review consolidates current understanding of efferocytosis mechanisms and its application in cardiovascular and cerebrovascular diseases, proposing future research directions.
Collapse
Affiliation(s)
- Bingtao Zhang
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yan Zou
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zixuan Yuan
- Department of Neurosurgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Kun Jiang
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhaoxiang Zhang
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shujuan Chen
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiaoming Zhou
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qi Wu
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xin Zhang
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
13
|
Köröskényi K, Sós L, Rostás M, Papp AB, Kókai E, Garabuczi É, Deák D, Beke L, Méhes G, Szondy Z. Loss of MER Tyrosine Kinase Attenuates Adipocyte Hypertrophy and Leads to Enhanced Thermogenesis in Mice Exposed to High-Fat Diet. Cells 2024; 13:1902. [PMID: 39594650 PMCID: PMC11593050 DOI: 10.3390/cells13221902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/30/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Obesity is characterized by low-grade inflammation that originates predominantly from the expanding visceral adipose tissue, in which adipocytes respond to lipid overload with hypertrophy, and consequently die by apoptosis. Recruited adipose tissue macrophages (ATMs) take up the excess lipids and remove the dead cells; however, long-term exposure to high concentrations of lipids alters their phenotype to M1-like ATMs that produce pro-inflammatory cytokines and resistin leading to insulin resistance and other obesity-related pathologies. Mer tyrosine kinase is expressed by macrophages and by being an efferocytosis receptor, and by suppressing inflammation, we hypothesized that it might play a protective role against obesity. To our surprise, however, the loss of Mer protected mice against high-fat diet (HFD)-induced obesity. We report in this paper that Mer is also expressed by adipocytes of both white and brown adipose tissues, and while its activity facilitates adipocyte lipid storage both in vitro and in vivo in mice exposed to HFD, it simultaneously attenuates thermogenesis in the brown adipose tissue contributing to its 'whitening'. Our data indicate that Mer is one of the adipocyte tyrosine kinase receptors, the activity of which contributes to the metabolic decision about the fate of excess lipids favoring their storage within the body.
Collapse
Affiliation(s)
- Krisztina Köröskényi
- Division of Dental Biochemistry, Department of Basic Medical Sciences, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary; (K.K.); (E.K.)
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - László Sós
- Doctoral School of Dental Sciences, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary; (L.S.); (M.R.); (A.B.P.)
| | - Melinda Rostás
- Doctoral School of Dental Sciences, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary; (L.S.); (M.R.); (A.B.P.)
| | - Albert Bálint Papp
- Doctoral School of Dental Sciences, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary; (L.S.); (M.R.); (A.B.P.)
| | - Endre Kókai
- Division of Dental Biochemistry, Department of Basic Medical Sciences, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary; (K.K.); (E.K.)
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Éva Garabuczi
- Department of Integrative Health Sciences, Institute of Health Sciences, Faculty of Health Sciences, University of Debrecen, 4032 Debrecen, Hungary;
| | - Dávid Deák
- Laboratory Animal Facility, Life Science Building, University of Debrecen, 4032 Debrecen, Hungary;
| | - Lívia Beke
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (L.B.); (G.M.)
| | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (L.B.); (G.M.)
| | - Zsuzsa Szondy
- Division of Dental Biochemistry, Department of Basic Medical Sciences, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary; (K.K.); (E.K.)
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
14
|
Liu W, Hardaway BD, Kim E, Pauli J, Wettich JL, Yalcinkaya M, Hsu CC, Xiao T, Reilly MP, Tabas I, Maegdefessel L, Schlepckow K, Haass C, Wang N, Tall AR. Inflammatory crosstalk impairs phagocytic receptors and aggravates atherosclerosis in clonal hematopoiesis in mice. J Clin Invest 2024; 135:e182939. [PMID: 39531316 PMCID: PMC11684819 DOI: 10.1172/jci182939] [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: 05/13/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Clonal hematopoiesis (CH) increases inflammasome-linked atherosclerosis, but the mechanisms by which CH mutant cells transmit inflammatory signals to nonmutant cells are largely unknown. To address this question, we transplanted 1.5% Jak2V617F (Jak2VF) bone marrow (BM) cells with 98.5% WT BM cells into hyperlipidemic Ldlr-/- mice. Low-allele-burden (LAB) mice showed accelerated atherosclerosis with increased features of plaque instability, decreased levels of the macrophage phagocytic receptors c-Mer tyrosine kinase (MERTK) and triggering receptor expressed on myeloid cells 2 (TREM2), and increased neutrophil extracellular traps (NETs). These changes were reversed when Jak2VF BM was transplanted with Il1r1-/- BM. LAB mice with noncleavable MERTK in WT BM showed improvements in necrotic core and fibrous cap formation and reduced NETs. An agonistic TREM2 antibody (4D9) markedly increased fibrous caps in both control and LAB mice, eliminating the difference between the groups. Mechanistically, 4D9 increased TREM2+PDGFB+ macrophages and PDGF receptor-α+ fibroblast-like cells in the cap region. TREM2 and PDGFB mRNA levels were positively correlated in human carotid plaques and coexpressed in macrophages. In summary, low frequencies of Jak2VF mutations promoted atherosclerosis via IL-1 signaling from Jak2VF to WT macrophages and neutrophils, promoting cleavage of phagocytic receptors and features of plaque instability. Therapeutic approaches that stabilize MERTK or TREM2 could promote plaque stabilization, especially in CH- and inflammasome-driven atherosclerosis.
Collapse
Affiliation(s)
- Wenli Liu
- Division of Molecular Medicine, Department of Medicine, and
| | | | - Eunyoung Kim
- Division of Cardiology, Department of Medicine, Columbia University, New York, New York, USA
| | - Jessica Pauli
- Institute of Molecular Vascular Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany
| | - Justus Leonard Wettich
- Institute of Molecular Vascular Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany
| | | | | | - Tong Xiao
- Division of Molecular Medicine, Department of Medicine, and
| | - Muredach P. Reilly
- Division of Cardiology, Department of Medicine, Columbia University, New York, New York, USA
| | - Ira Tabas
- Division of Molecular Medicine, Department of Medicine, and
| | - Lars Maegdefessel
- Institute of Molecular Vascular Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Kai Schlepckow
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Metabolic Biochemistry, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Nan Wang
- Division of Molecular Medicine, Department of Medicine, and
| | - Alan R. Tall
- Division of Molecular Medicine, Department of Medicine, and
| |
Collapse
|
15
|
Watanabe H, Rana M, Son M, Chiu PY, Fei-Bloom Y, Choi K, Diamond B, Sherry B. Single cell RNA-seq reveals cellular and transcriptional heterogeneity in the splenic CD11b +Ly6C high monocyte population expanded in sepsis-surviving mice. Mol Med 2024; 30:202. [PMID: 39506629 PMCID: PMC11539566 DOI: 10.1186/s10020-024-00970-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Sepsis survivors exhibit immune dysregulation that contributes to poor long-term outcomes. Phenotypic and functional alterations within the myeloid compartment are believed to be a contributing factor. Here we dissect the cellular and transcriptional heterogeneity of splenic CD11b+Ly6Chigh myeloid cells that are expanded in mice that survive the cecal ligation and puncture (CLP) murine model of polymicrobial sepsis to better understand the basis of immune dysregulation in sepsis survivors. METHODS Sham or CLP surgeries were performed on C57BL/6J and BALB/c mice. Four weeks later splenic CD11b+Ly6Chigh cells from both groups were isolated for phenotypic (flow cytometry) and functional (phagocytosis and glycolysis) characterization and RNA was obtained for single-cell RNA-seq (scRNA-seq) and subsequent analysis. RESULTS CD11b+Ly6Chigh cells from sham and CLP surviving mice exhibit phenotypic and functional differences that relate to immune function, some of which are observed in both C57BL/6J and BALB/c strains and others that are not. To dissect disease-specific and strain-specific distinctions within the myeloid compartment, scRNA-seq analysis was performed on CD11b+Ly6Chigh cells from C57BL/6J and BALB/c sham and CLP mice. Uniform Manifold Approximation and Projection from both strains identified 13 distinct clusters of sorted CD11b+Ly6Chigh cells demonstrating significant transcriptional heterogeneity and expressing gene signatures corresponding to classical-monocytes, non-classical monocytes, M1- or M2-like macrophages, dendritic-like cells, monocyte-derived dendritic-like cells, and proliferating monocytic myeloid-derived suppressor cells (M-MDSCs). Frequency plots showed that the percentages of proliferating M-MDSCs (clusters 8, 11 and 12) were increased in CLP mice compared to sham mice in both strains. Pathway and UCell score analysis in CLP mice revealed that cell cycle and glycolytic pathways were upregulated in proliferating M-MDSCs in both strains. Notably, granule protease genes were upregulated in M-MDSCs from CLP mice. ScRNA-seq analyses also showed that phagocytic pathways were upregulated in multiple clusters including the classical monocyte cluster, confirming the increased phagocytic capacity in CD11b+Ly6Chigh cells from CLP mice observed in ex vivo functional assays in C57BL/6J mice. CONCLUSION The splenic CD11b+Ly6Chigh myeloid populations expanded in survivors of CLP sepsis correspond to proliferating cells that have an increased metabolic demand and gene signatures consistent with M-MDSCs, a population known to have immunosuppressive capacity.
Collapse
Affiliation(s)
- Haruki Watanabe
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA
| | - Minakshi Rana
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery at Weill Cornell Medicine, New York, New York, 10021, USA
| | - Myoungsun Son
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA
| | - Pui Yan Chiu
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA
| | - Yurong Fei-Bloom
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA
| | - Kwangmin Choi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Betty Diamond
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA.
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA.
| | - Barbara Sherry
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA.
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA.
| |
Collapse
|
16
|
Abstract
Cardiovascular disease is the leading cause of death worldwide, and it commonly results from atherosclerotic plaque progression. One of the increasingly recognized drivers of atherosclerosis is dysfunctional efferocytosis, a homeostatic mechanism responsible for the clearance of dead cells and the resolution of inflammation. In atherosclerosis, the capacity of phagocytes to participate in efferocytosis is hampered, leading to the accumulation of apoptotic and necrotic tissue within the plaque, which results in enlargement of the necrotic core, increased luminal stenosis and plaque inflammation, and predisposition to plaque rupture or erosion. In this Review, we describe the different forms of programmed cell death that can occur in the atherosclerotic plaque and highlight the efferocytic machinery that is normally implicated in cardiovascular physiology. We then discuss the mechanisms by which efferocytosis fails in atherosclerosis and other cardiovascular and cardiometabolic diseases, including myocardial infarction and diabetes mellitus, and discuss therapeutic approaches that might reverse this pathological process.
Collapse
Affiliation(s)
- Shaunak S Adkar
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford, CA, USA
| | - Nicholas J Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford, CA, USA.
| |
Collapse
|
17
|
Fredman G, Serhan CN. Specialized pro-resolving mediators in vascular inflammation and atherosclerotic cardiovascular disease. Nat Rev Cardiol 2024; 21:808-823. [PMID: 38216693 DOI: 10.1038/s41569-023-00984-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/07/2023] [Indexed: 01/14/2024]
Abstract
Timely resolution of the acute inflammatory response (or inflammation resolution) is an active, highly coordinated process that is essential to optimal health. Inflammation resolution is regulated by specific endogenous signalling molecules that function as 'stop signals' to terminate the inflammatory response when it is no longer needed; to actively promote healing, regeneration and tissue repair; and to limit pain. Specialized pro-resolving mediators are a superfamily of signalling molecules that initiate anti-inflammatory and pro-resolving actions. Without an effective and timely resolution response, inflammation can become chronic, a pathological state that is associated with many widely occurring human diseases, including atherosclerotic cardiovascular disease. Uncovering the mechanisms of inflammation resolution failure in cardiovascular diseases and identifying useful biomarkers for non-resolving inflammation are unmet needs. In this Review, we discuss the accumulating evidence that supports the role of non-resolving inflammation in atherosclerosis and the use of specialized pro-resolving mediators as therapeutic tools for the treatment of atherosclerotic cardiovascular disease. We highlight open questions about therapeutic strategies and mechanisms of disease to provide a framework for future studies on the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Gabrielle Fredman
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anaesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
18
|
Xie XD, Dong SS, Liu RJ, Shi LL, Zhu T. Mechanism of Efferocytosis in Determining Ischaemic Stroke Resolution-Diving into Microglia/Macrophage Functions and Therapeutic Modality. Mol Neurobiol 2024; 61:7583-7602. [PMID: 38409642 DOI: 10.1007/s12035-024-04060-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/17/2024] [Indexed: 02/28/2024]
Abstract
After ischaemic cerebral vascular injury, efferocytosis-a process known as the efficient clearance of apoptotic cells (ACs) by various phagocytes in both physiological and pathological states-is crucial for maintaining central nervous system (CNS) homeostasis and regaining prognosis. The mechanisms of efferocytosis in ischaemic stroke and its influence on preventing inflammation progression from secondary injury were still not fully understood, despite the fact that the fundamental process of efferocytosis has been described in a series of phases, including AC recognition, phagocyte engulfment, and subsequent degradation. The genetic reprogramming of macrophages and brain-resident microglia after an ischaemic stroke has been equated by some researchers to that of the peripheral blood and brain. Based on previous studies, some molecules, such as signal transducer and activator of transcription 6 (STAT6), peroxisome proliferator-activated receptor γ (PPARG), CD300A, and sigma non-opioid intracellular receptor 1 (SIGMAR1), were discovered to be largely associated with aspects of apoptotic cell elimination and accompanying neuroinflammation, such as inflammatory cytokine release, phenotype transformation, and suppressing of antigen presentation. Exacerbated stroke outcomes are brought on by defective efferocytosis and improper modulation of pertinent signalling pathways in blood-borne macrophages and brain microglia, which also results in subsequent tissue inflammatory damage. This review focuses on recent researches which contain a number of recently discovered mechanisms, such as studies on the relationship between benign efferocytosis and the regulation of inflammation in ischaemic stroke, the roles of some risk factors in disease progression, and current immune approaches that aim to promote efferocytosis to treat some autoimmune diseases. Understanding these pathways provides insight into novel pathophysiological processes and fresh characteristics, which can be used to build cerebral ischaemia targeting techniques.
Collapse
Affiliation(s)
- Xiao-Di Xie
- Department of Pathophysiology, School of Basic Medicine, Institute of Neuroregeneration & Neurorehabilitation, Qingdao University, No. 308 Ningxia Road, Qingdao, China
| | - Shan-Shan Dong
- Department of Pathophysiology, School of Basic Medicine, Institute of Neuroregeneration & Neurorehabilitation, Qingdao University, No. 308 Ningxia Road, Qingdao, China
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ru-Juan Liu
- Department of Pathophysiology, School of Basic Medicine, Institute of Neuroregeneration & Neurorehabilitation, Qingdao University, No. 308 Ningxia Road, Qingdao, China
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Liu-Liu Shi
- Department of Pathophysiology, School of Basic Medicine, Institute of Neuroregeneration & Neurorehabilitation, Qingdao University, No. 308 Ningxia Road, Qingdao, China
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ting Zhu
- Department of Pathophysiology, School of Basic Medicine, Institute of Neuroregeneration & Neurorehabilitation, Qingdao University, No. 308 Ningxia Road, Qingdao, China.
| |
Collapse
|
19
|
Leroy V, Manual Kollareth DJ, Tu Z, Valisno JAC, Woolet-Stockton M, Saha B, Emtiazjoo AM, Rackauskas M, Moldawer LL, Efron PA, Cai G, Atkinson C, Upchurch GR, Sharma AK. MerTK-dependent efferocytosis by monocytic-MDSCs mediates resolution of ischemia/reperfusion injury after lung transplant. JCI Insight 2024; 9:e179876. [PMID: 39172530 PMCID: PMC11466183 DOI: 10.1172/jci.insight.179876] [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: 01/29/2024] [Accepted: 08/15/2024] [Indexed: 08/24/2024] Open
Abstract
Lung transplantation (LTx) outcomes are impeded by ischemia/reperfusion injury (IRI) and subsequent chronic lung allograft dysfunction (CLAD). We examined the undefined role of receptor Mer tyrosine kinase (MerTK) on monocytic myeloid-derived suppressor cells (M-MDSCs) in efferocytosis to facilitate resolution of lung IRI. Single-cell RNA sequencing of lung tissue and bronchoalveolar lavage (BAL) from patients after LTx were analyzed. Murine lung hilar ligation and allogeneic orthotopic LTx models of IRI were used with BALB/c (WT), Cebpb-/- (MDSC-deficient), Mertk-/-, or MerTK-cleavage-resistant mice. A significant downregulation in MerTK-related efferocytosis genes in M-MDSC populations of patients with CLAD was observed compared with healthy individuals. In the murine IRI model, a significant increase in M-MDSCs, MerTK expression, and efferocytosis and attenuation of lung dysfunction was observed in WT mice during injury resolution that was absent in Cebpb-/- and Mertk-/- mice. Adoptive transfer of M-MDSCs in Cebpb-/- mice significantly attenuated lung dysfunction and inflammation. Additionally, in a murine orthotopic LTx model, increases in M-MDSCs were associated with resolution of lung IRI in the transplant recipients. In vitro studies demonstrated the ability of M-MDSCs to efferocytose apoptotic neutrophils in a MerTK-dependent manner. Our results suggest that MerTK-dependent efferocytosis by M-MDSCs can substantially contribute to the resolution of post-LTx IRI.
Collapse
Affiliation(s)
- Victoria Leroy
- Department of Surgery
- Department of Pharmacology and Therapeutics
| | | | - Zhenxiao Tu
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | | | | | - Biplab Saha
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Amir M. Emtiazjoo
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | | | | | | | - Guoshuai Cai
- Department of Surgery
- Department of Biostatistics, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Carl Atkinson
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | | | - Ashish K. Sharma
- Department of Surgery
- Department of Pharmacology and Therapeutics
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
20
|
Liu J, Gong W, Liu P, Li Y, Jiang H, Wu C, Wu X, Zhao Y, Ren J. Osteopontin regulation of MerTK + macrophages promotes Crohn's disease intestinal fibrosis. iScience 2024; 27:110226. [PMID: 39021800 PMCID: PMC11253513 DOI: 10.1016/j.isci.2024.110226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 04/04/2024] [Accepted: 06/06/2024] [Indexed: 07/20/2024] Open
Abstract
The pathogenesis of intestinal fibrosis in Crohn's disease (CD) remains unclear. Mer receptor tyrosine kinase (MerTK) is an immunosuppressive protein specifically expressed in macrophages. Osteopontin (OPN), also known as secreted phosphoprotein 1, contributes to inflammation and wound repair. This study investigates the potential profibrotic pathway in MerTK+ macrophages in order to provide a possible therapeutic target for intestinal fibrosis. MerTK expression in the inflamed and stenotic bowels was evaluated. The MerTK/ERK/TGF-β1 pathway was overactivated in the fibrotic intestinal tissues of patients with CD. This pathway was induced by epithelial cell apoptosis, resulting in activated fibroblasts with increased TGF-β1 secretion. OPN upregulated TGF production by altering ERK1/2 phosphorylation, as evidenced by OPN or MerTK knockdown and OPN overexpression in vitro. MerTK inhibitor UNC2025 alleviated intestinal fibrosis in mouse colitis models, suggesting a potential therapeutic target for intestinal fibrosis in patients with CD.
Collapse
Affiliation(s)
- Juanhan Liu
- Department of General Surgery, Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wenbin Gong
- Department of General Surgery, School of Medicine, Southeast University, Nanjing, China
| | - Peizhao Liu
- Department of General Surgery, Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yangguang Li
- Department of General Surgery, Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Haiyang Jiang
- Department of General Surgery, BenQ Medical Center, Nanjing Medical School, Nanjing, China
| | - Cunxia Wu
- Department of General Surgery, BenQ Medical Center, Nanjing Medical School, Nanjing, China
| | - Xiuwen Wu
- Department of General Surgery, Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yun Zhao
- Department of General Surgery, BenQ Medical Center, Nanjing Medical School, Nanjing, China
| | - Jianan Ren
- Department of General Surgery, Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
21
|
Shu LX, Cao LL, Guo X, Wang ZB, Wang SZ. Mechanism of efferocytosis in atherosclerosis. J Mol Med (Berl) 2024; 102:831-840. [PMID: 38727748 DOI: 10.1007/s00109-024-02439-3] [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: 05/04/2023] [Revised: 02/26/2024] [Accepted: 03/13/2024] [Indexed: 06/29/2024]
Abstract
Atherosclerosis (AS) is a chronic inflammatory vascular disease that occurs in the intima of large and medium-sized arteries with the immune system's involvement. It is a common pathological basis for high morbidity and mortality of cardiovascular diseases. Abnormal proliferation of apoptotic cells and necrotic cells leads to AS plaque expansion, necrotic core formation, and rupture. In the early stage of AS, macrophages exert an efferocytosis effect to engulf and degrade apoptotic, dead, damaged, or senescent cells by efferocytosis, thus enabling the regulation of the organism. In the early stage of AS, macrophages rely on this effect to slow down the process of AS. However, in the advanced stage of AS, the efferocytosis of macrophages within the plaque is impaired, which leads to the inability of macrophages to promptly remove the apoptotic cells (ACs) from the organism promptly, causing exacerbation of AS. Moreover, upregulation of CD47 expression in AS plaques also protects ACs from phagocytosis by macrophages, resulting in a large amount of residual ACs in the plaque, further expanding the necrotic core. In this review, we discussed the molecular mechanisms involved in the process of efferocytosis and how efferocytosis is impaired and regulated during AS, hoping to provide new insights for treating AS.
Collapse
Affiliation(s)
- Li-Xia Shu
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Liu-Li Cao
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Xin Guo
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Zong-Bao Wang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Shu-Zhi Wang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China.
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China.
| |
Collapse
|
22
|
Han Y, Hu J, Pan J, Song X, Zhou Y, Zhang J, Yang Y, Shi X, Yang J, Sun M. LPS exposure alleviates multiple tissues damage by facilitating macrophage efferocytosis. Int Immunopharmacol 2024; 135:112283. [PMID: 38772299 DOI: 10.1016/j.intimp.2024.112283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/05/2024] [Accepted: 05/14/2024] [Indexed: 05/23/2024]
Abstract
Toll-like receptors (TLRs) play a crucial role in mediating immune responses by recognizing pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs), as well as facilitating apoptotic cell (ACs) clearance (efferocytosis), thus contributing significantly to maintaining homeostasis and promoting tissue resolution. In this study, we investigate the impact of TLR agonists on macrophage efferocytosis. Our findings demonstrate that pretreatment with the TLR agonist lipopolysaccharide (LPS) significantly enhances macrophage phagocytic ability, thereby promoting efferocytosis both in vitro and in vivo. Moreover, LPS pretreatment confers tissue protection against damage by augmenting macrophage efferocytic capacity in murine models. Further examination reveals that LPS modulates efferocytosis by upregulating the expression of Tim4.These results underscore the pivotal role of TLR agonists in regulating the efferocytosis process and suggest potential therapeutic avenues for addressing inflammatory diseases. Overall, our study highlights the intricate interplay between LPS pretreatment and efferocytosis in maintaining tissue homeostasis and resolving inflammation.
Collapse
Affiliation(s)
- Yuwen Han
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Jiangsu Key Lab of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Keling Road No. 88, Suzhou 215163, China
| | - Jiukun Hu
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Jiangsu Key Lab of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Keling Road No. 88, Suzhou 215163, China
| | - Jinlin Pan
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Jiangsu Key Lab of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Keling Road No. 88, Suzhou 215163, China
| | - Xueyan Song
- Jiangsu Key Lab of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Keling Road No. 88, Suzhou 215163, China
| | - Yuanshuai Zhou
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Jiangsu Key Lab of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Keling Road No. 88, Suzhou 215163, China
| | - Jun Zhang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Jiangsu Key Lab of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Keling Road No. 88, Suzhou 215163, China
| | - Yue Yang
- Jiangsu Key Lab of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Keling Road No. 88, Suzhou 215163, China; Department of Chemistry, College of Sciences, Shanghai University, Shanghai 200444, China
| | - Xiaohua Shi
- Department of Gastroenterology, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 1 Lijiang Road, Suzhou 215153, China
| | - Jiao Yang
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Lijiang Road No. 1, Suzhou 215153, China.
| | - Minxuan Sun
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Jiangsu Key Lab of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Keling Road No. 88, Suzhou 215163, China.
| |
Collapse
|
23
|
Hilgendorf I, Frantz S, Frangogiannis NG. Repair of the Infarcted Heart: Cellular Effectors, Molecular Mechanisms and Therapeutic Opportunities. Circ Res 2024; 134:1718-1751. [PMID: 38843294 PMCID: PMC11164543 DOI: 10.1161/circresaha.124.323658] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/08/2024] [Indexed: 06/12/2024]
Abstract
The adult mammalian heart has limited endogenous regenerative capacity and heals through the activation of inflammatory and fibrogenic cascades that ultimately result in the formation of a scar. After infarction, massive cardiomyocyte death releases a broad range of damage-associated molecular patterns that initiate both myocardial and systemic inflammatory responses. TLRs (toll-like receptors) and NLRs (NOD-like receptors) recognize damage-associated molecular patterns (DAMPs) and transduce downstream proinflammatory signals, leading to upregulation of cytokines (such as interleukin-1, TNF-α [tumor necrosis factor-α], and interleukin-6) and chemokines (such as CCL2 [CC chemokine ligand 2]) and recruitment of neutrophils, monocytes, and lymphocytes. Expansion and diversification of cardiac macrophages in the infarcted heart play a major role in the clearance of the infarct from dead cells and the subsequent stimulation of reparative pathways. Efferocytosis triggers the induction and release of anti-inflammatory mediators that restrain the inflammatory reaction and set the stage for the activation of reparative fibroblasts and vascular cells. Growth factor-mediated pathways, neurohumoral cascades, and matricellular proteins deposited in the provisional matrix stimulate fibroblast activation and proliferation and myofibroblast conversion. Deposition of a well-organized collagen-based extracellular matrix network protects the heart from catastrophic rupture and attenuates ventricular dilation. Scar maturation requires stimulation of endogenous signals that inhibit fibroblast activity and prevent excessive fibrosis. Moreover, in the mature scar, infarct neovessels acquire a mural cell coat that contributes to the stabilization of the microvascular network. Excessive, prolonged, or dysregulated inflammatory or fibrogenic cascades accentuate adverse remodeling and dysfunction. Moreover, inflammatory leukocytes and fibroblasts can contribute to arrhythmogenesis. Inflammatory and fibrogenic pathways may be promising therapeutic targets to attenuate heart failure progression and inhibit arrhythmia generation in patients surviving myocardial infarction.
Collapse
Affiliation(s)
- Ingo Hilgendorf
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine at the University of Freiburg, Freiburg, Germany
| | - Stefan Frantz
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx NY
| |
Collapse
|
24
|
Ma J, Eadie K, Schippers M, Fahal A, Laleu B, Verbon A, van de Sande WWJ. Novel Compound MMV1804559 from the Global Health Priority Box Exhibits In Vitro and In Vivo Activity against Madurella mycetomatis. Int J Mol Sci 2024; 25:6227. [PMID: 38892422 PMCID: PMC11172423 DOI: 10.3390/ijms25116227] [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: 04/18/2024] [Revised: 05/24/2024] [Accepted: 06/02/2024] [Indexed: 06/21/2024] Open
Abstract
OBJECTIVES Eumycetoma is a neglected tropical disease (NTD) characterized by subcutaneous lesions and the formation of grains. Attempts to treat eumycetoma involve a combination of antifungal treatment and surgery, although the outcome is frequently disappointing. Therefore, there is a need to identify novel antifungal drugs to treat eumycetoma. In this respect, Medicines for Malaria Venture (MMV) has assembled libraries of compounds for researchers to use in drug discovery research against NTD. Therefore, we screened two MMVOpen compound libraries to identify novel leads for eumycetoma. METHODS A total of 400 compounds from the COVID Box and the Global Health Priority Box were screened in vitro at 100 µM and 25 µM against the most common causative agents of eumycetoma, namely Madurella mycetomatis and Falciformispora senegalensis, and the resulting IC50 and MIC50 values were obtained. Compounds with an IC50 < 8 µM were identified for possible in vivo efficacy studies using an M. mycetomatis grain model in Galleria mellonella larvae. RESULTS Out of the 400 compounds, 22 were able to inhibit both M. mycetomatis and F. senegalensis growth at 100 µM and 25 µM, with compounds MMV1593278, MMV020335, and MMV1804559 being selected for in vivo testing. Of these three, only the pyrazolopyrimidine derivative MMV1804559 was able to prolong the survival of M. mycetomatis-infected G. mellonella larvae. Furthermore, the grains in MMV1804559-treated larvae were significantly smaller compared to the PBS-treated group. CONCLUSION MMV1804559 shows promising in vitro and in vivo activity against M. mycetomatis.
Collapse
Affiliation(s)
- Jingyi Ma
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015GD Rotterdam, The Netherlands; (J.M.); (K.E.); (M.S.); (A.V.)
| | - Kimberly Eadie
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015GD Rotterdam, The Netherlands; (J.M.); (K.E.); (M.S.); (A.V.)
| | - Marij Schippers
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015GD Rotterdam, The Netherlands; (J.M.); (K.E.); (M.S.); (A.V.)
| | - Ahmed Fahal
- Mycetoma Research Centre, Khartoum 1115, Sudan;
| | - Benoît Laleu
- MMV Medicines for Malaria Venture, 1215 Geneva, Switzerland;
| | - Annelies Verbon
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015GD Rotterdam, The Netherlands; (J.M.); (K.E.); (M.S.); (A.V.)
| | - Wendy W. J. van de Sande
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015GD Rotterdam, The Netherlands; (J.M.); (K.E.); (M.S.); (A.V.)
| |
Collapse
|
25
|
Tan H, Li W, Pang Z, Weng X, Gao J, Chen J, Wang Q, Li Q, Yang H, Dong Z, Wang Z, Zhu G, Tan Y, Fu Y, Han C, Cai S, Qian J, Huang Z, Song Y, Ge J. Genetically Engineered Macrophages Co-Loaded with CD47 Inhibitors Synergistically Reconstruct Efferocytosis and Improve Cardiac Remodeling Post Myocardial Ischemia Reperfusion Injury. Adv Healthc Mater 2024; 13:e2303267. [PMID: 38198534 PMCID: PMC11468776 DOI: 10.1002/adhm.202303267] [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: 09/26/2023] [Revised: 12/16/2023] [Indexed: 01/12/2024]
Abstract
Efferocytosis, mediated by the macrophage receptor MerTK (myeloid-epithelial-reproductive tyrosine kinase), is a significant contributor to cardiac repair after myocardial ischemia-reperfusion (MI/R) injury. However, the death of resident cardiac macrophages (main effector cells), inactivation of MerTK (main effector receptor), and overexpression of "do not eat me" signals (brake signals, such as CD47), collectively lead to the impediment of efferocytosis in the post-MI/R heart. To date, therapeutic strategies targeting individual above obstacles are relatively lacking, let alone their effectiveness being limited due to constraints from the other concurrent two. Herein, inspired by the application research of chimeric antigen receptor macrophages (CAR-Ms) in solid tumors, a genetically modified macrophage-based synergistic drug delivery strategy that effectively challenging the three major barriers in an integrated manner is developed. This strategy involves the overexpression of exogenous macrophages with CCR2 (C-C chemokine receptor type 2) and cleavage-resistant MerTK, as well as surface clicking with liposomal PEP-20 (a CD47 antagonist). In MI/R mice model, this synergistic strategy can effectively restore cardiac efferocytosis after intravenous injection, thereby alleviating the inflammatory response, ultimately preserving cardiac function. This therapy focuses on inhibiting the initiation and promoting active resolution of inflammation, providing new insights for immune-regulatory therapy.
Collapse
Affiliation(s)
- Haipeng Tan
- Department of CardiologyZhongshan Hospital, Fudan UniversityShanghai Institute of Cardiovascular DiseasesShanghai20032P. R. China
- National Clinical Research Center for Interventional Medicine and Shanghai Clinical Research Center for Interventional MedicineShanghai200032P. R. China
- Key Laboratory of Viral Heart DiseasesNational Health CommissionShanghai20032P. R. China
| | - Weiyan Li
- Department of CardiologyZhongshan Hospital, Fudan UniversityShanghai Institute of Cardiovascular DiseasesShanghai20032P. R. China
- National Clinical Research Center for Interventional Medicine and Shanghai Clinical Research Center for Interventional MedicineShanghai200032P. R. China
- Key Laboratory of Viral Heart DiseasesNational Health CommissionShanghai20032P. R. China
| | - Zhiqing Pang
- Key Laboratory of Smart Drug DeliverySchool of PharmacyFudan UniversityMinistry of Education826 Zhangheng Road, Pudong New AreaShanghai201210P. R. China
| | - Xueyi Weng
- Department of CardiologyZhongshan Hospital, Fudan UniversityShanghai Institute of Cardiovascular DiseasesShanghai20032P. R. China
- National Clinical Research Center for Interventional Medicine and Shanghai Clinical Research Center for Interventional MedicineShanghai200032P. R. China
- Key Laboratory of Viral Heart DiseasesNational Health CommissionShanghai20032P. R. China
| | - Jinfeng Gao
- Department of CardiologyZhongshan Hospital, Fudan UniversityShanghai Institute of Cardiovascular DiseasesShanghai20032P. R. China
- National Clinical Research Center for Interventional Medicine and Shanghai Clinical Research Center for Interventional MedicineShanghai200032P. R. China
- Key Laboratory of Viral Heart DiseasesNational Health CommissionShanghai20032P. R. China
| | - Jing Chen
- Department of CardiologyZhongshan Hospital, Fudan UniversityShanghai Institute of Cardiovascular DiseasesShanghai20032P. R. China
- National Clinical Research Center for Interventional Medicine and Shanghai Clinical Research Center for Interventional MedicineShanghai200032P. R. China
- Key Laboratory of Viral Heart DiseasesNational Health CommissionShanghai20032P. R. China
| | - Qiaozi Wang
- Department of CardiologyZhongshan Hospital, Fudan UniversityShanghai Institute of Cardiovascular DiseasesShanghai20032P. R. China
- National Clinical Research Center for Interventional Medicine and Shanghai Clinical Research Center for Interventional MedicineShanghai200032P. R. China
- Key Laboratory of Viral Heart DiseasesNational Health CommissionShanghai20032P. R. China
| | - Qiyu Li
- Department of CardiologyZhongshan Hospital, Fudan UniversityShanghai Institute of Cardiovascular DiseasesShanghai20032P. R. China
- National Clinical Research Center for Interventional Medicine and Shanghai Clinical Research Center for Interventional MedicineShanghai200032P. R. China
- Key Laboratory of Viral Heart DiseasesNational Health CommissionShanghai20032P. R. China
| | - Hongbo Yang
- Department of CardiologyZhongshan Hospital, Fudan UniversityShanghai Institute of Cardiovascular DiseasesShanghai20032P. R. China
- National Clinical Research Center for Interventional Medicine and Shanghai Clinical Research Center for Interventional MedicineShanghai200032P. R. China
- Key Laboratory of Viral Heart DiseasesNational Health CommissionShanghai20032P. R. China
| | - Zheng Dong
- Department of CardiologyZhongshan Hospital, Fudan UniversityShanghai Institute of Cardiovascular DiseasesShanghai20032P. R. China
- National Clinical Research Center for Interventional Medicine and Shanghai Clinical Research Center for Interventional MedicineShanghai200032P. R. China
- Key Laboratory of Viral Heart DiseasesNational Health CommissionShanghai20032P. R. China
| | - Zhengmin Wang
- Department of CardiologyZhongshan Hospital, Fudan UniversityShanghai Institute of Cardiovascular DiseasesShanghai20032P. R. China
- National Clinical Research Center for Interventional Medicine and Shanghai Clinical Research Center for Interventional MedicineShanghai200032P. R. China
- Key Laboratory of Viral Heart DiseasesNational Health CommissionShanghai20032P. R. China
| | - Guangrui Zhu
- Department of CardiologyZhongshan Hospital, Fudan UniversityShanghai Institute of Cardiovascular DiseasesShanghai20032P. R. China
- National Clinical Research Center for Interventional Medicine and Shanghai Clinical Research Center for Interventional MedicineShanghai200032P. R. China
- Key Laboratory of Viral Heart DiseasesNational Health CommissionShanghai20032P. R. China
| | - Yiwen Tan
- Department of CardiologyZhongshan Hospital, Fudan UniversityShanghai Institute of Cardiovascular DiseasesShanghai20032P. R. China
- National Clinical Research Center for Interventional Medicine and Shanghai Clinical Research Center for Interventional MedicineShanghai200032P. R. China
- Key Laboratory of Viral Heart DiseasesNational Health CommissionShanghai20032P. R. China
| | - Yuyuan Fu
- Department of CardiologyZhongshan Hospital, Fudan UniversityShanghai Institute of Cardiovascular DiseasesShanghai20032P. R. China
- National Clinical Research Center for Interventional Medicine and Shanghai Clinical Research Center for Interventional MedicineShanghai200032P. R. China
- Key Laboratory of Viral Heart DiseasesNational Health CommissionShanghai20032P. R. China
| | - Chengzhi Han
- Department of CardiologyZhongshan Hospital, Fudan UniversityShanghai Institute of Cardiovascular DiseasesShanghai20032P. R. China
- National Clinical Research Center for Interventional Medicine and Shanghai Clinical Research Center for Interventional MedicineShanghai200032P. R. China
- Key Laboratory of Viral Heart DiseasesNational Health CommissionShanghai20032P. R. China
| | - Shiteng Cai
- Department of CardiologyZhongshan Hospital, Fudan UniversityShanghai Institute of Cardiovascular DiseasesShanghai20032P. R. China
- National Clinical Research Center for Interventional Medicine and Shanghai Clinical Research Center for Interventional MedicineShanghai200032P. R. China
- Key Laboratory of Viral Heart DiseasesNational Health CommissionShanghai20032P. R. China
| | - Juying Qian
- Department of CardiologyZhongshan Hospital, Fudan UniversityShanghai Institute of Cardiovascular DiseasesShanghai20032P. R. China
- National Clinical Research Center for Interventional Medicine and Shanghai Clinical Research Center for Interventional MedicineShanghai200032P. R. China
- Key Laboratory of Viral Heart DiseasesNational Health CommissionShanghai20032P. R. China
| | - Zheyong Huang
- Department of CardiologyZhongshan Hospital, Fudan UniversityShanghai Institute of Cardiovascular DiseasesShanghai20032P. R. China
- National Clinical Research Center for Interventional Medicine and Shanghai Clinical Research Center for Interventional MedicineShanghai200032P. R. China
- Key Laboratory of Viral Heart DiseasesNational Health CommissionShanghai20032P. R. China
| | - Yanan Song
- Department of CardiologyZhongshan Hospital, Fudan UniversityShanghai Institute of Cardiovascular DiseasesShanghai20032P. R. China
- National Clinical Research Center for Interventional Medicine and Shanghai Clinical Research Center for Interventional MedicineShanghai200032P. R. China
- Key Laboratory of Viral Heart DiseasesNational Health CommissionShanghai20032P. R. China
| | - Junbo Ge
- Department of CardiologyZhongshan Hospital, Fudan UniversityShanghai Institute of Cardiovascular DiseasesShanghai20032P. R. China
- National Clinical Research Center for Interventional Medicine and Shanghai Clinical Research Center for Interventional MedicineShanghai200032P. R. China
- Key Laboratory of Viral Heart DiseasesNational Health CommissionShanghai20032P. R. China
| |
Collapse
|
26
|
Ngai D, Sukka SR, Tabas I. Crosstalk between efferocytic myeloid cells and T-cells and its relevance to atherosclerosis. Front Immunol 2024; 15:1403150. [PMID: 38873597 PMCID: PMC11169609 DOI: 10.3389/fimmu.2024.1403150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/17/2024] [Indexed: 06/15/2024] Open
Abstract
The interplay between myeloid cells and T-lymphocytes is critical to the regulation of host defense and inflammation resolution. Dysregulation of this interaction can contribute to the development of chronic inflammatory diseases. Important among these diseases is atherosclerosis, which refers to focal lesions in the arterial intima driven by elevated apolipoprotein B-containing lipoproteins, notably low-density lipoprotein (LDL), and characterized by the formation of a plaque composed of inflammatory immune cells, a collection of dead cells and lipids called the necrotic core, and a fibrous cap. As the disease progresses, the necrotic core expands, and the fibrous cap becomes thin, which increases the risk of plaque rupture or erosion. Plaque rupture leads to a rapid thrombotic response that can give rise to heart attack, stroke, or sudden death. With marked lowering of circulating LDL, however, plaques become more stable and cardiac risk is lowered-a process known as atherosclerosis regression. A critical aspect of both atherosclerosis progression and regression is the crosstalk between innate (myeloid cells) and adaptive (T-lymphocytes) immune cells. Myeloid cells are specialized at clearing apoptotic cells by a process called efferocytosis, which is necessary for inflammation resolution. In advanced disease, efferocytosis is impaired, leading to secondary necrosis of apoptotic cells, inflammation, and, most importantly, defective tissue resolution. In regression, efferocytosis is reawakened aiding in inflammation resolution and plaque stabilization. Here, we will explore how efferocytosing myeloid cells could affect T-cell function and vice versa through antigen presentation, secreted factors, and cell-cell contacts and how this cellular crosstalk may contribute to the progression or regression of atherosclerosis.
Collapse
Affiliation(s)
- David Ngai
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Santosh R. Sukka
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, United States
- Department of Physiology, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
27
|
Grazda R, Seyfried AN, Maddipati KR, Fredman G, MacNamara KC. Resolvin E1 improves efferocytosis and rescues severe aplastic anemia in mice. Cell Death Dis 2024; 15:324. [PMID: 38724533 PMCID: PMC11082201 DOI: 10.1038/s41419-024-06705-7] [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: 10/24/2023] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/12/2024]
Abstract
Severe aplastic anemia (SAA) is a rare, fatal disease characterized by severe cytopenias and loss of hematopoietic stem cells (HSCs). Immune-mediated destruction and inflammation are known drivers of SAA, however, the underlying mechanisms driving persistent inflammation are unknown. Current treatments for SAA rely on immunosuppressive therapies or HSC transplantation, however, these treatments are not always effective. Using an established mouse model of SAA, we observed a significant increase in apoptotic cells within the bone marrow (BM) and impaired efferocytosis in SAA mice, relative to radiation controls. Single-cell transcriptomic analysis revealed heterogeneity among BM monocytes and unique populations emerged during SAA characterized by increased inflammatory signatures and significantly increased expression of Sirpa and Cd47. CD47, a "don't eat me" signal, was increased on both live and apoptotic BM cells, concurrent with markedly increased expression of signal regulatory protein alpha (SIRPα) on monocytes. Functionally, SIRPα blockade improved cell clearance and reduced accumulation of CD47-positive apoptotic cells. Lipidomic analysis revealed a reduction in the precursors of specialized pro-resolving lipid mediators (SPMs) and increased prostaglandins in the BM during SAA, indicative of impaired inflammation resolution. Specifically, 18-HEPE, a precursor of E-series resolvins, was significantly reduced in SAA-induced mice relative to radiation controls. Treatment of SAA mice with Resolvin E1 (RvE1) improved efferocytic function, BM cellularity, platelet output, and survival. Our data suggest that impaired efferocytosis and inflammation resolution contributes to SAA progression and demonstrate that SPMs, such as RvE1, offer new and/or complementary treatments for SAA that do not rely on immune suppression.
Collapse
Affiliation(s)
- Rachel Grazda
- Department of Immunology and Microbiology, Albany Medical College, Albany, NY, USA
| | - Allison N Seyfried
- Department of Immunology and Microbiology, Albany Medical College, Albany, NY, USA
- Institute for Clinical Pharmacodynamics, Schenectady, NY, USA
| | - Krishna Rao Maddipati
- Department of Pathology, Lipidomics Core Facility, Wayne State University, Detroit, MI, USA
| | - Gabrielle Fredman
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | | |
Collapse
|
28
|
Sheng Y, Hu W, Chen S, Zhu X. Efferocytosis by macrophages in physiological and pathological conditions: regulatory pathways and molecular mechanisms. Front Immunol 2024; 15:1275203. [PMID: 38779685 PMCID: PMC11109379 DOI: 10.3389/fimmu.2024.1275203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 04/17/2024] [Indexed: 05/25/2024] Open
Abstract
Efferocytosis is defined as the highly effective phagocytic removal of apoptotic cells (ACs) by professional or non-professional phagocytes. Tissue-resident professional phagocytes ("efferocytes"), such as macrophages, have high phagocytic capacity and are crucial to resolve inflammation and aid in homeostasis. Recently, numerous exciting discoveries have revealed divergent (and even diametrically opposite) findings regarding metabolic immune reprogramming associated with efferocytosis by macrophages. In this review, we highlight the key metabolites involved in the three phases of efferocytosis and immune reprogramming of macrophages under physiological and pathological conditions. The next decade is expected to yield further breakthroughs in the regulatory pathways and molecular mechanisms connecting immunological outcomes to metabolic cues as well as avenues for "personalized" therapeutic intervention.
Collapse
Affiliation(s)
- Yan−Ran Sheng
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Wen−Ting Hu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Siman Chen
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Xiao−Yong Zhu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, China
| |
Collapse
|
29
|
Chuang ST, Stein JB, Nevins S, Kilic Bektas C, Choi HK, Ko WK, Jang H, Ha J, Lee KB. Enhancing CAR Macrophage Efferocytosis Via Surface Engineered Lipid Nanoparticles Targeting LXR Signaling. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308377. [PMID: 38353580 PMCID: PMC11081841 DOI: 10.1002/adma.202308377] [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: 08/17/2023] [Revised: 02/05/2024] [Indexed: 02/24/2024]
Abstract
The removal of dying cells, or efferocytosis, is an indispensable part of resolving inflammation. However, the inflammatory microenvironment of the atherosclerotic plaque frequently affects the biology of both apoptotic cells and resident phagocytes, rendering efferocytosis dysfunctional. To overcome this problem, a chimeric antigen receptor (CAR) macrophage that can target and engulf phagocytosis-resistant apoptotic cells expressing CD47 is developed. In both normal and inflammatory circumstances, CAR macrophages exhibit activity equivalent to antibody blockage. The surface of CAR macrophages is modified with reactive oxygen species (ROS)-responsive therapeutic nanoparticles targeting the liver X receptor pathway to improve their cell effector activities. The combination of CAR and nanoparticle engineering activated lipid efflux pumps enhances cell debris clearance and reduces inflammation. It is further suggested that the undifferentiated CAR-Ms can transmigrate within a mico-fabricated vessel system. It is also shown that our CAR macrophage can act as a chimeric switch receptor (CSR) to withstand the immunosuppressive inflammatory environment. The developed platform has the potential to contribute to the advancement of next-generation cardiovascular disease therapies and further studies include in vivo experiments.
Collapse
Affiliation(s)
- Skylar T Chuang
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Joshua B Stein
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Sarah Nevins
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Cemile Kilic Bektas
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Hye Kyu Choi
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Wan-Kyu Ko
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Hyunjun Jang
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Jihun Ha
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| |
Collapse
|
30
|
Xiong W, Han Z, Ding S, Wang H, Du Y, Cui W, Zhang M. In Situ Remodeling of Efferocytosis via Lesion-Localized Microspheres to Reverse Cartilage Senescence. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400345. [PMID: 38477444 PMCID: PMC11109622 DOI: 10.1002/advs.202400345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/19/2024] [Indexed: 03/14/2024]
Abstract
Efferocytosis, an intrinsic regulatory mechanism to eliminate apoptotic cells, will be suppressed due to the delayed apoptosis process in aging-related diseases, such as osteoarthritis (OA). In this study, cartilage lesion-localized hydrogel microspheres are developed to remodel the in situ efferocytosis to reverse cartilage senescence and recruit endogenous stem cells to accelerate cartilage repair. Specifically, aldehyde- and methacrylic anhydride (MA)-modified hyaluronic acid hydrogel microspheres (AHM), loaded with pro-apoptotic liposomes (liposomes encapsulating ABT263, A-Lipo) and PDGF-BB, namely A-Lipo/PAHM, are prepared by microfluidic and photo-cross-linking techniques. By a degraded porcine cartilage explant OA model, the in situ cartilage lesion location experiment illustrated that aldehyde-functionalized microspheres promote affinity for degraded cartilage. In vitro data showed that A-Lipo induced apoptosis of senescent chondrocytes (Sn-chondrocytes), which can then be phagocytosed by the efferocytosis of macrophages, and remodeling efferocytosis facilitated the protection of normal chondrocytes and maintained the chondrogenic differentiation capacity of MSCs. In vivo experiments confirmed that hydrogel microspheres localized to cartilage lesion reversed cartilage senescence and promoted cartilage repair in OA. It is believed this in situ efferocytosis remodeling strategy can be of great significance for tissue regeneration in aging-related diseases.
Collapse
Affiliation(s)
- Wei Xiong
- Department of Foot and Ankle SurgeryBeijing Tongren HospitalCapital Medical UniversityBeijing100730P. R. China
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Zeyu Han
- Department of Foot and Ankle SurgeryBeijing Tongren HospitalCapital Medical UniversityBeijing100730P. R. China
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Sheng‐Long Ding
- Department of Foot and Ankle SurgeryBeijing Tongren HospitalCapital Medical UniversityBeijing100730P. R. China
| | - Haoran Wang
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Yawei Du
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Wenguo Cui
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Ming‐Zhu Zhang
- Department of Foot and Ankle SurgeryBeijing Tongren HospitalCapital Medical UniversityBeijing100730P. R. China
| |
Collapse
|
31
|
Singh S, Bensalem J, Hein LK, Casey A, Mäkinen VP, Sargeant TJ. epHero - a tandem-fluorescent probe to track the fate of apoptotic cells during efferocytosis. Cell Death Discov 2024; 10:179. [PMID: 38632247 PMCID: PMC11024195 DOI: 10.1038/s41420-024-01952-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 04/19/2024] Open
Abstract
The efficient removal of apoptotic cells via efferocytosis is critical for maintaining optimal tissue function. This involves the binding and engulfment of apoptotic cells by phagocytes and the subsequent maturation of the phagosome, culminating in lysosomal fusion and cargo destruction. However, current approaches to measure efferocytosis rely on labelling apoptotic targets with fluorescent dyes, which do not sufficiently distinguish between changes to the engulfment and acidification of apoptotic material. To address this limitation, we have developed a genetically coded ratiometric probe epHero which when expressed in the cytoplasm of target cells, bypasses the need for additional labelling steps. We demonstrate that epHero is a pH-sensitive reporter for efferocytosis and can be used to simultaneously track changes to apoptotic cell uptake and acidification, both in vitro and in mice. As proof-of-principle, we modify extracellular nutrition to show how epHero can distinguish between changes to cargo engulfment and acidification. Thus, tracking efferocytosis with epHero is a simple, cost-effective improvement on conventional techniques.
Collapse
Affiliation(s)
- Sanjna Singh
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- University of Adelaide, Adelaide, SA, Australia
| | - Julien Bensalem
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Leanne K Hein
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Aaron Casey
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Ville-Petteri Mäkinen
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Timothy J Sargeant
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
- University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
32
|
Lahey KC, Varsanyi C, Wang Z, Aquib A, Gadiyar V, Rodrigues AA, Pulica R, Desind S, Davra V, Calianese DC, Liu D, Cho JH, Kotenko SV, De Lorenzo MS, Birge RB. Regulation of Mertk Surface Expression via ADAM17 and γ-Secretase Proteolytic Processing. Int J Mol Sci 2024; 25:4404. [PMID: 38673989 PMCID: PMC11050108 DOI: 10.3390/ijms25084404] [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: 02/02/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Mertk, a type I receptor tyrosine kinase and member of the TAM family of receptors, has important functions in promoting efferocytosis and resolving inflammation under physiological conditions. In recent years, Mertk has also been linked to pathophysiological roles in cancer, whereby, in several cancer types, including solid cancers and leukemia/lymphomas. Mertk contributes to oncogenic features of proliferation and cell survival as an oncogenic tyrosine kinase. In addition, Mertk expressed on macrophages, including tumor-associated macrophages, promotes immune evasion in cancer and is suggested to act akin to a myeloid checkpoint inhibitor that skews macrophages towards inhibitory phenotypes that suppress host T-cell anti-tumor immunity. In the present study, to better understand the post-translational regulation mechanisms controlling Mertk expression in monocytes/macrophages, we used a PMA-differentiated THP-1 cell model to interrogate the regulation of Mertk expression and developed a novel Mertk reporter cell line to study the intracellular trafficking of Mertk. We show that PMA treatment potently up-regulates Mertk as well as components of the ectodomain proteolytic processing platform ADAM17, whereas PMA differentially regulates the canonical Mertk ligands Gas6 and Pros1 (Gas6 is down-regulated and Pros1 is up-regulated). Under non-stimulated homeostatic conditions, Mertk in PMA-differentiated THP1 cells shows active constitutive proteolytic cleavage by the sequential activities of ADAM17 and the Presenilin/γ-secretase complex, indicating that Mertk is cleaved homeostatically by the combined sequential action of ADAM17 and γ-secretase, after which the cleaved intracellular fragment of Mertk is degraded in a proteasome-dependent mechanism. Using chimeric Flag-Mertk-EGFP-Myc reporter receptors, we confirm that inhibitors of γ-secretase and MG132, which inhibits the 26S proteasome, stabilize the intracellular fragment of Mertk without evidence of nuclear translocation. Finally, the treatment of cells with active γ-carboxylated Gas6, but not inactive Warfarin-treated non-γ-carboxylated Gas6, regulates a distinct proteolytic itinerary-involved receptor clearance and lysosomal proteolysis. Together, these results indicate that pleotropic and complex proteolytic activities regulate Mertk ectodomain cleavage as a homeostatic negative regulatory event to safeguard against the overactivation of Mertk.
Collapse
Affiliation(s)
- Kevin C. Lahey
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ 07103, USA; (K.C.L.); (C.V.); (Z.W.); (A.A.); (A.A.R.); (R.P.); (S.D.); (V.D.); (D.C.C.); (S.V.K.)
| | - Christopher Varsanyi
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ 07103, USA; (K.C.L.); (C.V.); (Z.W.); (A.A.); (A.A.R.); (R.P.); (S.D.); (V.D.); (D.C.C.); (S.V.K.)
| | - Ziren Wang
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ 07103, USA; (K.C.L.); (C.V.); (Z.W.); (A.A.); (A.A.R.); (R.P.); (S.D.); (V.D.); (D.C.C.); (S.V.K.)
| | - Ahmed Aquib
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ 07103, USA; (K.C.L.); (C.V.); (Z.W.); (A.A.); (A.A.R.); (R.P.); (S.D.); (V.D.); (D.C.C.); (S.V.K.)
| | - Varsha Gadiyar
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ 07103, USA; (K.C.L.); (C.V.); (Z.W.); (A.A.); (A.A.R.); (R.P.); (S.D.); (V.D.); (D.C.C.); (S.V.K.)
| | - Alcina A. Rodrigues
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ 07103, USA; (K.C.L.); (C.V.); (Z.W.); (A.A.); (A.A.R.); (R.P.); (S.D.); (V.D.); (D.C.C.); (S.V.K.)
| | - Rachael Pulica
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ 07103, USA; (K.C.L.); (C.V.); (Z.W.); (A.A.); (A.A.R.); (R.P.); (S.D.); (V.D.); (D.C.C.); (S.V.K.)
| | - Samuel Desind
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ 07103, USA; (K.C.L.); (C.V.); (Z.W.); (A.A.); (A.A.R.); (R.P.); (S.D.); (V.D.); (D.C.C.); (S.V.K.)
| | - Viralkumar Davra
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ 07103, USA; (K.C.L.); (C.V.); (Z.W.); (A.A.); (A.A.R.); (R.P.); (S.D.); (V.D.); (D.C.C.); (S.V.K.)
| | - David C. Calianese
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ 07103, USA; (K.C.L.); (C.V.); (Z.W.); (A.A.); (A.A.R.); (R.P.); (S.D.); (V.D.); (D.C.C.); (S.V.K.)
| | - Dongfang Liu
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07101, USA; (D.L.); (J.-H.C.)
| | - Jong-Hyun Cho
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07101, USA; (D.L.); (J.-H.C.)
| | - Sergei V. Kotenko
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ 07103, USA; (K.C.L.); (C.V.); (Z.W.); (A.A.); (A.A.R.); (R.P.); (S.D.); (V.D.); (D.C.C.); (S.V.K.)
| | - Mariana S. De Lorenzo
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, 185 South Orange Ave, Newark, NJ 07103, USA;
| | - Raymond B. Birge
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ 07103, USA; (K.C.L.); (C.V.); (Z.W.); (A.A.); (A.A.R.); (R.P.); (S.D.); (V.D.); (D.C.C.); (S.V.K.)
| |
Collapse
|
33
|
Li Q, Liu H, Yin G, Xie Q. Efferocytosis: Current status and future prospects in the treatment of autoimmune diseases. Heliyon 2024; 10:e28399. [PMID: 38596091 PMCID: PMC11002059 DOI: 10.1016/j.heliyon.2024.e28399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/11/2024] Open
Abstract
Billions of apoptotic cells are swiftly removed from the human body daily. This clearance process is regulated by efferocytosis, an active anti-inflammatory process during which phagocytes engulf and remove apoptotic cells. However, impaired clearance of apoptotic cells is associated with the development of various autoimmune diseases, such as rheumatoid arthritis, systemic lupus erythematosus, and inflammatory bowel disease. In this review, we conducted a comprehensive search of relevant studies published from January 1, 2000, to the present, focusing on efferocytosis, autoimmune disease pathogenesis, regulatory mechanisms governing efferocytosis, and potential treatments targeting this process. Our review highlights the key molecules involved in different stages of efferocytosis-namely, the "find me," "eat me," and "engulf and digest" phases-while elucidating their relevance to autoimmune disease pathology. Furthermore, we explore the therapeutic potential of modulating efferocytosis to restore immune homeostasis and mitigate autoimmune responses. By providing theoretical underpinnings for the targeting of efferocytosis in the treatment of autoimmune diseases, this review contributes to the advancement of therapeutic strategies in this field.
Collapse
Affiliation(s)
- Qianwei Li
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Huan Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Geng Yin
- Department of General Practice, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Qibing Xie
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
34
|
Enderlin J, Rieu Q, Réty S, Vanoni EM, Roux S, Dégardin J, César Q, Augustin S, Nous C, Cai B, Fontaine V, Sennlaub F, Nandrot EF. Retinal atrophy, inflammation, phagocytic and metabolic disruptions develop in the MerTK-cleavage-resistant mouse model. Front Neurosci 2024; 18:1256522. [PMID: 38680449 PMCID: PMC11047123 DOI: 10.3389/fnins.2024.1256522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 03/11/2024] [Indexed: 05/01/2024] Open
Abstract
In the eye, cells from the retinal pigment epithelium (RPE) facing the neurosensory retina exert several functions that are all crucial for long-term survival of photoreceptors (PRs) and vision. Among those, RPE cells phagocytose under a circadian rhythm photoreceptor outer segment (POS) tips that are constantly subjected to light rays and oxidative attacks. The MerTK tyrosine kinase receptor is a key element of this phagocytic machinery required for POS internalization. Recently, we showed that MerTK is subjected to the cleavage of its extracellular domain to finely control its function. In addition, monocytes in retinal blood vessels can migrate inside the inner retina and differentiate into macrophages expressing MerTK, but their role in this context has not been studied yet. We thus investigated the ocular phenotype of MerTK cleavage-resistant (MerTKCR) mice to understand the relevance of this characteristic on retinal homeostasis at the RPE and macrophage levels. MerTKCR retinae appear to develop and function normally, as observed in retinal sections, by electroretinogram recordings and optokinetic behavioral tests. Monitoring of MerTKCR and control mice between the ages of 3 and 18 months showed the development of large degenerative areas in the central retina as early as 4 months when followed monthly by optical coherence tomography (OCT) plus fundus photography (FP)/autofluorescence (AF) detection but not by OCT alone. The degenerative areas were associated with AF, which seems to be due to infiltrated macrophages, as observed by OCT and histology. MerTKCR RPE primary cultures phagocytosed less POS in vitro, while in vivo, the circadian rhythm of POS phagocytosis was deregulated. Mitochondrial function and energy production were reduced in freshly dissected RPE/choroid tissues at all ages, thus showing a metabolic impairment not present in macrophages. RPE anomalies were detected by electron microscopy, including phagosomes retained in the apical area and vacuoles. Altogether, this new mouse model displays a novel phenotype that could prove useful to understanding the interplay between RPE and PRs in inflammatory retinal degenerations and highlights new roles for MerTK in the regulation of the energetic metabolism and the maintenance of the immune privilege in the retina.
Collapse
Affiliation(s)
- Julie Enderlin
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Quentin Rieu
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Salomé Réty
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Elora M. Vanoni
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Solène Roux
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Julie Dégardin
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Quénol César
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Sébastien Augustin
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Caroline Nous
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Bishuang Cai
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Valérie Fontaine
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Florian Sennlaub
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Emeline F. Nandrot
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| |
Collapse
|
35
|
Wang X, Du W, Li Y, Yang HH, Zhang Y, Akbar R, Morgan H, Peng T, Chen J, Sadayappan S, Hu YC, Fan Y, Huang W, Fan GC. Macrophage-enriched Sectm1a promotes efficient efferocytosis to attenuate ischemia/reperfusion-induced cardiac injury. JCI Insight 2024; 9:e173832. [PMID: 38456501 PMCID: PMC10972593 DOI: 10.1172/jci.insight.173832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 01/23/2024] [Indexed: 03/09/2024] Open
Abstract
Efficient clearance and degradation of apoptotic cardiomyocytes by macrophages (collectively termed efferocytosis) is critical for inflammation resolution and restoration of cardiac function after myocardial ischemia/reperfusion (I/R). Here, we define secreted and transmembrane protein 1a (Sectm1a), a cardiac macrophage-enriched gene, as a modulator of macrophage efferocytosis in I/R-injured hearts. Upon myocardial I/R, Sectm1a-KO mice exhibited impaired macrophage efferocytosis, leading to massive accumulation of apoptotic cardiomyocytes, cardiac inflammation, fibrosis, and consequently, exaggerated cardiac dysfunction. By contrast, therapeutic administration of recombinant SECTM1A protein significantly enhanced macrophage efferocytosis and improved cardiac function. Mechanistically, SECTM1A could elicit autocrine effects on the activation of glucocorticoid-induced TNF receptor (GITR) at the surface of macrophages, leading to the upregulation of liver X receptor α (LXRα) and its downstream efferocytosis-related genes and lysosomal enzyme genes. Our study suggests that Sectm1a-mediated activation of the Gitr/LXRα axis could be a promising approach to enhance macrophage efferocytosis for the treatment of myocardial I/R injury.
Collapse
Affiliation(s)
| | - Wa Du
- Department of Cancer Biology, and
| | - Yutian Li
- Department of Pharmacology and Systems Physiology
| | - Hui-Hui Yang
- Department of Pharmacology and Systems Physiology
| | - Yu Zhang
- Department of Pharmacology and Systems Physiology
| | - Rubab Akbar
- Department of Pharmacology and Systems Physiology
| | - Hannah Morgan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Tianqing Peng
- Centre for Critical Illness Research, Lawson Health Research Institute, London, Ontario, Canada
| | - Jing Chen
- Division of Biomedical Informatics and
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Yueh-Chiang Hu
- Transgenic Animal and Genome Editing Facility, Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | | | - Wei Huang
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | |
Collapse
|
36
|
Mytych JS, Pan Z, Lopez-Davis C, Redinger N, Lawrence C, Ziegler J, Popescu NI, James JA, Farris AD. Peptidoglycan from Bacillus anthracis Inhibits Human Macrophage Efferocytosis in Part by Reducing Cell Surface Expression of MERTK and TIM-3. Immunohorizons 2024; 8:269-280. [PMID: 38517345 PMCID: PMC10985058 DOI: 10.4049/immunohorizons.2300109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/06/2024] [Indexed: 03/23/2024] Open
Abstract
Bacillus anthracis peptidoglycan (PGN) is a major component of the bacterial cell wall and a key pathogen-associated molecular pattern contributing to anthrax pathology, including organ dysfunction and coagulopathy. Increases in apoptotic leukocytes are a late-stage feature of anthrax and sepsis, suggesting there is a defect in apoptotic clearance. In this study, we tested the hypothesis that B. anthracis PGN inhibits the capacity of human monocyte-derived macrophages (MΦ) to efferocytose apoptotic cells. Exposure of CD163+CD206+ MΦ to PGN for 24 h impaired efferocytosis in a manner dependent on human serum opsonins but independent of complement component C3. PGN treatment reduced cell surface expression of the proefferocytic signaling receptors MERTK, TYRO3, AXL, integrin αVβ5, CD36, and TIM-3, whereas TIM-1, αVβ3, CD300b, CD300f, STABILIN-1, and STABILIN-2 were unaffected. ADAM17 is a major membrane-bound protease implicated in mediating efferocytotic receptor cleavage. We found multiple ADAM17-mediated substrates increased in PGN-treated supernatant, suggesting involvement of membrane-bound proteases. ADAM17 inhibitors TAPI-0 and Marimastat prevented TNF release, indicating effective protease inhibition, and modestly increased cell-surface levels of MerTK and TIM-3 but only partially restored efferocytic capacity by PGN-treated MΦ. We conclude that human serum factors are required for optimal recognition of PGN by human MΦ and that B. anthracis PGN inhibits efferocytosis in part by reducing cell surface expression of MERTK and TIM-3.
Collapse
Affiliation(s)
- Joshua S. Mytych
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Zijian Pan
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Charmaine Lopez-Davis
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Nancy Redinger
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Christina Lawrence
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Jadith Ziegler
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Narcis I. Popescu
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Judith A. James
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - A. Darise Farris
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
37
|
Mohammad-Rafiei F, Negahdari S, Tahershamsi Z, Gheibihayat SM. Interface between Resolvins and Efferocytosis in Health and Disease. Cell Biochem Biophys 2024; 82:53-65. [PMID: 37794303 DOI: 10.1007/s12013-023-01187-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 09/25/2023] [Indexed: 10/06/2023]
Abstract
Acute inflammation resolution acts as a vital process for active host response, tissue support, and homeostasis maintenance, during which resolvin D (RvD) and E (RvE) as mediators derived from omega-3 polyunsaturated fatty acids display specific and stereoselective anti-inflammations like restricting neutrophil infiltration and pro-resolving activities. On the other side of the coin, potent macrophage-mediated apoptotic cell clearance, namely efferocytosis, is essential for successful inflammation resolution. Further studies mentioned a linkage between efferocytosis and resolvins. For instance, resolvin D1 (RvD1), which is endogenously formed from docosahexaenoic acid within the inflammation resolution, thereby provoking efferocytosis. There is still limited information regarding the mechanism of action of RvD1-related efferocytosis enhancement at the molecular level. The current review article was conducted to explore recent data on how the efferocytosis process and resolvins relate to each other during the inflammation resolution in illness and health. Understanding different aspects of this connection sheds light on new curative approaches for medical conditions caused by defective efferocytosis and disrupted inflammation resolution.
Collapse
Affiliation(s)
- Fatemeh Mohammad-Rafiei
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Samira Negahdari
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Zahra Tahershamsi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Munich, Germany.
| |
Collapse
|
38
|
Raju S, Botts SR, Blaser MC, Abdul-Samad M, Prajapati K, Khosraviani N, Ho TWW, Breda LC, Ching C, Galant NJ, Fiddes L, Wu R, Clift CL, Pham T, Lee WL, Singh SA, Aikawa E, Fish JE, Howe KL. Directional Endothelial Communication by Polarized Extracellular Vesicle Release. Circ Res 2024; 134:269-289. [PMID: 38174557 PMCID: PMC10826926 DOI: 10.1161/circresaha.123.322993] [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: 04/28/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Extracellular vesicles (EVs) contain bioactive cargo including miRNAs and proteins that are released by cells during cell-cell communication. Endothelial cells (ECs) form the innermost lining of all blood vessels, interfacing with cells in the circulation and vascular wall. It is unknown whether ECs release EVs capable of governing recipient cells within these 2 separate compartments. Given their boundary location, we propose ECs use bidirectional release of distinct EV cargo in quiescent (healthy) and activated (atheroprone) states to communicate with cells within the circulation and blood vessel wall. METHODS EVs were isolated from primary human aortic ECs (plate and transwell grown; ±IL [interleukin]-1β activation), quantified, visualized, and analyzed by miRNA transcriptomics and proteomics. Apical and basolateral EC-EV release was determined by miRNA transfer, total internal reflection fluorescence and electron microscopy. Vascular reprogramming (RNA sequencing) and functional assays were performed on primary human monocytes or smooth muscle cells±EC-EVs. RESULTS Activated ECs increased EV release, with miRNA and protein cargo related to atherosclerosis. EV-treated monocytes and smooth muscle cells revealed activated EC-EV altered pathways that were proinflammatory and atherogenic. ECs released more EVs apically, which increased with activation. Apical and basolateral EV cargo contained distinct transcriptomes and proteomes that were altered by EC activation. Notably, activated basolateral EC-EVs displayed greater changes in the EV secretome, with pathways specific to atherosclerosis. In silico analysis determined compartment-specific cargo released by the apical and basolateral surfaces of ECs can reprogram monocytes and smooth muscle cells, respectively, with functional assays and in vivo imaging supporting this concept. CONCLUSIONS Demonstrating that ECs are capable of polarized EV cargo loading and directional EV secretion reveals a novel paradigm for endothelial communication, which may ultimately enhance the design of endothelial-based therapeutics for cardiovascular diseases such as atherosclerosis where ECs are persistently activated.
Collapse
Affiliation(s)
- Sneha Raju
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (S.R., S.R.B., M.A.-S., K.P., N.K., L.C.D.B., C.C., R.W., J.E.F., K.L.H.)
- Institute of Medical Science (S.R., S.R.B., C.C., J.E.F., K.L.H.), University of Toronto, Toronto, ON, Canada
- Faculty of Medicine (S.R., S.R.B., L.F., K.L.H.), University of Toronto, Toronto, ON, Canada
- Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada (S.R., K.L.H.)
| | - Steven R. Botts
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (S.R., S.R.B., M.A.-S., K.P., N.K., L.C.D.B., C.C., R.W., J.E.F., K.L.H.)
- Institute of Medical Science (S.R., S.R.B., C.C., J.E.F., K.L.H.), University of Toronto, Toronto, ON, Canada
- Faculty of Medicine (S.R., S.R.B., L.F., K.L.H.), University of Toronto, Toronto, ON, Canada
| | - Mark C. Blaser
- Cardiovascular Division, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences (M.C.B., C.L.C., T.P., S.A.S., E.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Majed Abdul-Samad
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (S.R., S.R.B., M.A.-S., K.P., N.K., L.C.D.B., C.C., R.W., J.E.F., K.L.H.)
- Department of Laboratory Medicine and Pathobiology (M.A.-S., N.K., R.W., J.E.F.), University of Toronto, Toronto, ON, Canada
| | - Kamalben Prajapati
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (S.R., S.R.B., M.A.-S., K.P., N.K., L.C.D.B., C.C., R.W., J.E.F., K.L.H.)
| | - Negar Khosraviani
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (S.R., S.R.B., M.A.-S., K.P., N.K., L.C.D.B., C.C., R.W., J.E.F., K.L.H.)
- Department of Laboratory Medicine and Pathobiology (M.A.-S., N.K., R.W., J.E.F.), University of Toronto, Toronto, ON, Canada
| | - Tse Wing Winnie Ho
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada (T.W.W.H., W.L.L.)
| | - Leandro C.D. Breda
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (S.R., S.R.B., M.A.-S., K.P., N.K., L.C.D.B., C.C., R.W., J.E.F., K.L.H.)
| | - Crizza Ching
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (S.R., S.R.B., M.A.-S., K.P., N.K., L.C.D.B., C.C., R.W., J.E.F., K.L.H.)
- Institute of Medical Science (S.R., S.R.B., C.C., J.E.F., K.L.H.), University of Toronto, Toronto, ON, Canada
| | | | - Lindsey Fiddes
- Faculty of Medicine (S.R., S.R.B., L.F., K.L.H.), University of Toronto, Toronto, ON, Canada
| | - Ruilin Wu
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (S.R., S.R.B., M.A.-S., K.P., N.K., L.C.D.B., C.C., R.W., J.E.F., K.L.H.)
- Department of Laboratory Medicine and Pathobiology (M.A.-S., N.K., R.W., J.E.F.), University of Toronto, Toronto, ON, Canada
| | - Cassandra L. Clift
- Cardiovascular Division, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences (M.C.B., C.L.C., T.P., S.A.S., E.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Tan Pham
- Cardiovascular Division, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences (M.C.B., C.L.C., T.P., S.A.S., E.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Warren L. Lee
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada (T.W.W.H., W.L.L.)
| | - Sasha A. Singh
- Cardiovascular Division, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences (M.C.B., C.L.C., T.P., S.A.S., E.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine (S.A.S., E.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Elena Aikawa
- Cardiovascular Division, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences (M.C.B., C.L.C., T.P., S.A.S., E.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine (S.A.S., E.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Jason E. Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (S.R., S.R.B., M.A.-S., K.P., N.K., L.C.D.B., C.C., R.W., J.E.F., K.L.H.)
- Institute of Medical Science (S.R., S.R.B., C.C., J.E.F., K.L.H.), University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology (M.A.-S., N.K., R.W., J.E.F.), University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, Toronto General Hospital, Toronto, ON, Canada (J.E.F., K.L.H.)
| | - Kathryn L. Howe
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (S.R., S.R.B., M.A.-S., K.P., N.K., L.C.D.B., C.C., R.W., J.E.F., K.L.H.)
- Institute of Medical Science (S.R., S.R.B., C.C., J.E.F., K.L.H.), University of Toronto, Toronto, ON, Canada
- Faculty of Medicine (S.R., S.R.B., L.F., K.L.H.), University of Toronto, Toronto, ON, Canada
- Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada (S.R., K.L.H.)
- Peter Munk Cardiac Centre, Toronto General Hospital, Toronto, ON, Canada (J.E.F., K.L.H.)
| |
Collapse
|
39
|
Tutusaus A, Morales A, García de Frutos P, Marí M. GAS6/TAM Axis as Therapeutic Target in Liver Diseases. Semin Liver Dis 2024; 44:99-114. [PMID: 38395061 PMCID: PMC11027478 DOI: 10.1055/a-2275-0408] [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: 02/25/2024]
Abstract
TAM (TYRO3, AXL, and MERTK) protein tyrosine kinase membrane receptors and their vitamin K-dependent ligands GAS6 and protein S (PROS) are well-known players in tumor biology and autoimmune diseases. In contrast, TAM regulation of fibrogenesis and the inflammation mechanisms underlying metabolic dysfunction-associated steatohepatitis (MASH), cirrhosis, and, ultimately, liver cancer has recently been revealed. GAS6 and PROS binding to phosphatidylserine exposed in outer membranes of apoptotic cells links TAMs, particularly MERTK, with hepatocellular damage. In addition, AXL and MERTK regulate the development of liver fibrosis and inflammation in chronic liver diseases. Acute hepatic injury is also mediated by the TAM system, as recent data regarding acetaminophen toxicity and acute-on-chronic liver failure have uncovered. Soluble TAM-related proteins, mainly released from activated macrophages and hepatic stellate cells after hepatic deterioration, are proposed as early serum markers for disease progression. In conclusion, the TAM system is becoming an interesting pharmacological target in liver pathology and a focus of future biomedical research in this field.
Collapse
Affiliation(s)
- Anna Tutusaus
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, Barcelona, Catalunya, Spain
- Barcelona Clinic Liver Cancer (BCLC) Group, Barcelona, Spain
| | - Albert Morales
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, Barcelona, Catalunya, Spain
- Barcelona Clinic Liver Cancer (BCLC) Group, Barcelona, Spain
| | - Pablo García de Frutos
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, Barcelona, Catalunya, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Cardiovasculares (CIBERCV), Barcelona, Comunidad de Madrid, Spain
| | - Montserrat Marí
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, Barcelona, Catalunya, Spain
- Barcelona Clinic Liver Cancer (BCLC) Group, Barcelona, Spain
| |
Collapse
|
40
|
Gonzalez AL, Dungan MM, Smart CD, Madhur MS, Doran AC. Inflammation Resolution in the Cardiovascular System: Arterial Hypertension, Atherosclerosis, and Ischemic Heart Disease. Antioxid Redox Signal 2024; 40:292-316. [PMID: 37125445 PMCID: PMC11071112 DOI: 10.1089/ars.2023.0284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 04/12/2023] [Indexed: 05/02/2023]
Abstract
Significance: Chronic inflammation has emerged as a major underlying cause of many prevalent conditions in the Western world, including cardiovascular diseases. Although targeting inflammation has emerged as a promising avenue by which to treat cardiovascular disease, it is also associated with increased risk of infection. Recent Advances: Though previously assumed to be passive, resolution has now been identified as an active process, mediated by unique immunoresolving mediators and mechanisms designed to terminate acute inflammation and promote tissue repair. Recent work has determined that failures of resolution contribute to chronic inflammation and the progression of human disease. Specifically, failure to produce pro-resolving mediators and the impaired clearance of dead cells from inflamed tissue have been identified as major mechanisms by which resolution fails in disease. Critical Issues: Drawing from a rapidly expanding body of experimental and clinical studies, we review here what is known about the role of inflammation resolution in arterial hypertension, atherosclerosis, myocardial infarction, and ischemic heart disease. For each, we discuss the involvement of specialized pro-resolving mediators and pro-reparative cell types, including T regulatory cells, myeloid-derived suppressor cells, and macrophages. Future Directions: Pro-resolving therapies offer the promise of limiting chronic inflammation without impairing host defense. Therefore, it is imperative to better understand the mechanisms underlying resolution to identify therapeutic targets. Antioxid. Redox Signal. 40, 292-316.
Collapse
Affiliation(s)
- Azuah L. Gonzalez
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Matthew M. Dungan
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - C. Duncan Smart
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Meena S. Madhur
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Amanda C. Doran
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
41
|
Leroy V, Manual Kollareth DJ, Tu Z, Valisno JAC, Woolet-Stockton M, Saha B, Emtiazjoo AM, Rackauskas M, Moldawer LL, Efron PA, Cai G, Atkinson C, Upchurch GR, Sharma AK. MerTK-dependent efferocytosis by monocytic-MDSCs mediates resolution of post-lung transplant injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.576261. [PMID: 38328174 PMCID: PMC10849528 DOI: 10.1101/2024.01.18.576261] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Rationale Patients with end stage lung diseases require lung transplantation (LTx) that can be impeded by ischemia-reperfusion injury (IRI) leading to subsequent chronic lung allograft dysfunction (CLAD) and inadequate outcomes. Objectives We examined the undefined role of MerTK (receptor Mer tyrosine kinase) on monocytic myeloid-derived suppressor cells (M-MDSCs) in efferocytosis (phagocytosis of apoptotic cells) to facilitate resolution of lung IRI. Methods Single-cell RNA sequencing of lung tissue and BAL from post-LTx patients was analyzed. Murine lung hilar ligation and allogeneic orthotopic LTx models of IRI were used with Balb/c (WT), cebpb -/- (MDSC-deficient), Mertk -/- or MerTK-CR (cleavage resistant) mice. Lung function, IRI (inflammatory cytokine and myeloperoxidase expression, immunohistology for neutrophil infiltration), and flow cytometry of lung tissue for efferocytosis of apoptotic neutrophils were assessed in mice. Measurements and Main Results A significant downregulation in MerTK-related efferocytosis genes in M-MDSC populations of CLAD patients compared to healthy subjects was observed. In the murine IRI model, significant increase in M-MDSCs, MerTK expression and efferocytosis was observed in WT mice during resolution phase that was absent in cebpb -/- Land Mertk -/- mice. Adoptive transfer of M-MDSCs in cebpb -/- mice significantly attenuated lung dysfunction, and inflammation leading to resolution of IRI. Additionally, in a preclinical murine orthotopic LTx model, increases in M-MDSCs were associated with resolution of lung IRI in the transplant recipients. In vitro studies demonstrated the ability of M-MDSCs to efferocytose apoptotic neutrophils in a MerTK-dependent manner. Conclusions Our results suggest that MerTK-dependent efferocytosis by M-MDSCs can significantly contribute to the resolution of post-LTx IRI.
Collapse
|
42
|
Fan L, Jin L, Tang T, Zheng Y, Chen Z, Lin H, Ding C, Wang T, Chen H, Guo Y, Xu C, Zhou H, Wu X, Fu X, Yan F, Mao Z, Chen G. Neutrophil-like pH-responsive pro-efferocytic nanoparticles improve neurological recovery by promoting erythrophagocytosis after intracerebral hemorrhage. Theranostics 2024; 14:283-303. [PMID: 38164152 PMCID: PMC10750197 DOI: 10.7150/thno.90370] [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: 09/21/2023] [Accepted: 11/06/2023] [Indexed: 01/03/2024] Open
Abstract
Rationale: Intracerebral hemorrhage (ICH) is a devastating cerebrovascular disease resulting from blood extravasating into the brain parenchyma. Escalation of erythrophagocytosis (a form of efferocytosis), avoiding the consequent release of the detrimental erythrocyte lysates, may be a promising target of ICH management. The ADAM17 inhibitor and liver X receptor (LXR) agonist could promote efficient efferocytosis and injury repair. Nevertheless, the poor bioavailability and restriction of the blood-brain barrier (BBB) hinder their application. Therefore, it is needed that biocompatible and smart nanoplatforms were designed and synthesized to realize effective therapy targeting erythrophagocytosis. Methods: We first assessed the synergistic effect of therapeutic GW280264X (an ADAM17 inhibitor) and desmosterol (an LXR agonist) on erythrophagocytosis in vitro. Then a pH-responsive neutrophil membrane-based nanoplatform (NPEOz) served as a carrier to accurately deliver therapeutic GW280264X and desmosterol to the damaged brain was prepared via co-extrusion. Afterwards, their pH-responsive performance was valued in vitro and targeting ability was assessed through fluorescence image in vivo. Finally, the pro-erythrophagocytic and anti-neuroinflammatory ability of the nanomedicine and related mechanisms were investigated. Results: After the synergistical effect of the above two drugs on erythrophagocytosis was confirmed, we successfully developed neutrophil-disguised pH-responsive nanoparticles to efficiently co-deliver them. The nanoparticles could responsively release therapeutic agents under acidic environments, and elicit favorable biocompatibility and ability of targeting injury sites. D&G@NPEOz nanoparticles enhanced erythrophagocytosis through inhibiting shedding of the efferocytotic receptors MERTK/AXL mediated by ADAM17 and accelerating ABCA-1/ABCG-1-mediated cholesterol efflux regulated by LXR respectively. In addition, the nano-formulation was able to modulate the inflammatory microenvironment by transforming efferocytes towards a therapeutic phenotype with reducing the release of proinflammatory cytokines while increasing the secretion of anti-inflammatory factors, and improve neurological function. Conclusions: This biomimetic nanomedicine is envisaged to offer an encouraging strategy to effectively promote hematoma and inflammation resolution, consequently alleviate ICH progression.
Collapse
Affiliation(s)
- Linfeng Fan
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Lulu Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Tianchi Tang
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Yonghe Zheng
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Zihang Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Haopu Lin
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Chao Ding
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Tingting Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Huaijun Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Yinghan Guo
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Chaoran Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Hang Zhou
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Xinyan Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Xiongjie Fu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Feng Yan
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Gao Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| |
Collapse
|
43
|
Xu S, Xu C, Xu J, Zhang K, Zhang H. Macrophage Heterogeneity and Its Impact on Myocardial Ischemia-Reperfusion Injury: An Integrative Review. J Inflamm Res 2023; 16:5971-5987. [PMID: 38088942 PMCID: PMC10712254 DOI: 10.2147/jir.s436560] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/30/2023] [Indexed: 10/21/2024] Open
Abstract
The coronary reperfusion following acute myocardial infarction can paradoxically trigger myocardial ischemia-reperfusion (IR) injury. This complex phenomenon involves the intricate interplay of different subsets of macrophages. These macrophages are crucial players in the post-infarction inflammatory response and subsequent myocardial anti-inflammatory repair. However, their diverse functions can lead to both beneficial and detrimental effects. On one hand, these macrophages play a crucial role in orchestrating the inflammatory response, aiding in the clearance of cellular debris and initiating tissue repair mechanisms. On the other hand, their excessive infiltration and activation can contribute to the perpetuation of the inflammatory cascade, leading to additional myocardial injury and adverse cardiac remodeling. Multiple mechanisms contribute to the IR injury mediated by macrophages, including oxidative stress, apoptosis, and autophagy. These processes further exacerbate the damage to the already vulnerable myocardial tissue. To address this delicate balance, therapeutic strategies aiming to target and modulate macrophage polarization and function are being explored. By fine-tuning the immune inflammatory response, such interventions hold promise in mitigating post-infarction myocardial injury and fostering a more favorable environment for myocardial healing and recovery. Through advancements in this area of research, potential anti-inflammatory interventions may pave the way for improved clinical outcomes and better management of patients after acute myocardial infarction.
Collapse
Affiliation(s)
- Shuwan Xu
- Cardiovascular Department, the Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, People’s Republic of China
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Cong Xu
- Cardiovascular Department, the Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, People’s Republic of China
| | - Jiahua Xu
- Cardiovascular Department, the Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, People’s Republic of China
| | - Kun Zhang
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Huanji Zhang
- Cardiovascular Department, the Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
44
|
Schilperoort M, Ngai D, Sukka SR, Avrampou K, Shi H, Tabas I. The role of efferocytosis-fueled macrophage metabolism in the resolution of inflammation. Immunol Rev 2023; 319:65-80. [PMID: 37158427 PMCID: PMC10615666 DOI: 10.1111/imr.13214] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/20/2023] [Indexed: 05/10/2023]
Abstract
The phagocytosis of dying cells by macrophages, termed efferocytosis, is a tightly regulated process that involves the sensing, binding, engulfment, and digestion of apoptotic cells. Efferocytosis not only prevents tissue necrosis and inflammation caused by secondary necrosis of dying cells, but it also promotes pro-resolving signaling in macrophages, which is essential for tissue resolution and repair following injury or inflammation. An important factor that contributes to this pro-resolving reprogramming is the cargo that is released from apoptotic cells after their engulfment and phagolysosomal digestion by macrophages. The apoptotic cell cargo contains amino acids, nucleotides, fatty acids, and cholesterol that function as metabolites and signaling molecules to bring about this re-programming. Here, we review efferocytosis-induced changes in macrophage metabolism that mediate the pro-resolving functions of macrophages. We also discuss various strategies, challenges, and future perspectives related to drugging efferocytosis-fueled macrophage metabolism as strategy to dampen inflammation and promote resolution in chronic inflammatory diseases.
Collapse
Affiliation(s)
- Maaike Schilperoort
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - David Ngai
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Santosh R Sukka
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kleopatra Avrampou
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Hongxue Shi
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Physiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
45
|
Fredman G, Khan S. Specialized pro-resolving mediators enhance the clearance of dead cells. Immunol Rev 2023; 319:151-157. [PMID: 37787174 DOI: 10.1111/imr.13278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
The failure to resolve inflammation underpins to several prevalent diseases, like atherosclerosis, and so identifying ways to boost resolution is unmet clinical needs. The resolution of inflammation is governed by several factors such as specialized pro-resolving mediators (SPMs) that counter-regulate pro-inflammatory pathways and promote tissue repair without compromising host defense. A major function of nearly all SPMs is to enhance the clearance of dead cells or efferocytosis. As such, phagocytes, such as macrophages, are essential cellular players in the resolution of inflammation because of their ability to rapidly and efficiently clear dead cells. This review highlights the role of SPMs in the clearance of apoptotic and necroptotic cells and offers insights into how targeting efferocytosis may provide new treatments for non-resolving diseases, like atherosclerosis.
Collapse
Affiliation(s)
- Gabrielle Fredman
- The Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Sayeed Khan
- The Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| |
Collapse
|
46
|
Mytych JS, Pan Z, Lopez-Davis C, Redinger N, Lawrence C, Ziegler J, Popescu NI, James JA, Farris AD. Peptidoglycan from Bacillus anthracis Inhibits Human Macrophage Efferocytosis in Part by Reducing Cell Surface Expression of MERTK and TIM-3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.30.535001. [PMID: 37066181 PMCID: PMC10103956 DOI: 10.1101/2023.03.30.535001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Bacillus anthracis peptidoglycan (PGN) is a major component of the bacterial cell wall and a key pathogen-associated molecular pattern (PAMP) contributing to anthrax pathology, including organ dysfunction and coagulopathy. Increases in apoptotic lymphocytes are a late-stage feature of anthrax and sepsis, suggesting there is a defect in apoptotic clearance. Here, we tested the hypothesis that B. anthracis PGN inhibits the capacity of human monocyte-derived macrophages (MΦ) to efferocytose apoptotic cells. Exposure of CD163+CD206+ MΦ to PGN for 24h impaired efferocytosis in a manner dependent on human serum opsonins but independent of complement component C3. PGN treatment reduced cell surface expression of the pro-efferocytic signaling receptors MERTK, TYRO3, AXL, integrin αVβ5, CD36 and TIM-3, whereas TIM-1, αVβ3, CD300b, CD300f, STABILIN-1 and STABILIN-2 were unaffected. ADAM17 is a major membrane-bound protease implicated in mediating efferocytotic receptor cleavage. We found multiple ADAM17-mediated substrates increased in PGN-treated supernatant suggesting involvement of membrane-bound proteases. ADAM17 inhibitors TAPI-0 and Marimastat prevented TNF release, indicating effective protease inhibition, and modestly increased cell-surface levels of MerTK and TIM-3 but only partially restored efferocytic capacity by PGN-treated MΦ. We conclude that human serum factors are required for optimal recognition of PGN by human MΦ and that B. anthracis PGN inhibits efferocytosis in part by reducing cell surface expression of MERTK and TIM-3.
Collapse
Affiliation(s)
- Joshua S Mytych
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Oklahoma City, OK 73104, USA
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, 1100 N Lindsay Avenue, Oklahoma City, OK 73104, USA
| | - Zijian Pan
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Oklahoma City, OK 73104, USA
| | - Charmaine Lopez-Davis
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Oklahoma City, OK 73104, USA
| | - Nancy Redinger
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Oklahoma City, OK 73104, USA
| | - Christina Lawrence
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Oklahoma City, OK 73104, USA
| | - Jadith Ziegler
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Oklahoma City, OK 73104, USA
| | - Narcis I. Popescu
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Oklahoma City, OK 73104, USA
| | - Judith A. James
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Oklahoma City, OK 73104, USA
| | - A. Darise Farris
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Oklahoma City, OK 73104, USA
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, 1100 N Lindsay Avenue, Oklahoma City, OK 73104, USA
| |
Collapse
|
47
|
Hu L, Lv Z, Gu Y, Zheng T, Kong Y, Mao W. A bibliometric analysis of efferocytosis in cardiovascular diseases from 2001 to 2022. Medicine (Baltimore) 2023; 102:e34366. [PMID: 37773819 PMCID: PMC10545234 DOI: 10.1097/md.0000000000034366] [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: 04/24/2023] [Accepted: 06/26/2023] [Indexed: 10/01/2023] Open
Abstract
INTRODUCTION In recent years, efferocytosis in cardiovascular diseases has become an intense area of research. However, only a few bibliometric analyses have been conducted in this area. In this review, we used CiteSpace 5.7. R2 and VOSviewer 1.6.17 software to perform text mining and knowledge map analysis. This study summarizes the latest progress, development paths, frontier research hotspots, and future research trends in this field. MATERIALS AND METHODS Studies on efferocytosis in cardiovascular diseases were downloaded from the Web of Science Core Collection. RESULTS In total, 327 studies published by 506 institutions across 42 countries and regions were identified. The number of studies on efferocytosis in cardiovascular diseases has increased over time. Arteriosclerosis Thrombosis and Vascular Biology published the highest number of articles and was the top co-cited journal. Tabas Ira. was the most prolific researcher and co-cited the most. The most productive countries were the United States and China. Columbia University, Harvard Medical School, and Brigham Women's Hospital were the 3 most productive institutions in the field of research. Keyword Co-occurrence, Clusters, and Burst analyses showed that inflammation, atherosclerosis, macrophages, and phagocytosis appeared with the highest frequency in these studies. CONCLUSION Multinational cooperation and multidisciplinary intersections are characteristic trends of development in the field, and the immune microenvironment, glycolysis, and lipid metabolism will be the focus of future research.
Collapse
Affiliation(s)
- Luoxia Hu
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Integrative Chinese and Western Medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, Hangzhou, China
| | - Zhengtian Lv
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Integrative Chinese and Western Medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, Hangzhou, China
| | - Yangyang Gu
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Integrative Chinese and Western Medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, Hangzhou, China
| | - Tiantian Zheng
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Integrative Chinese and Western Medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, Hangzhou, China
| | - Youjin Kong
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Integrative Chinese and Western Medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, Hangzhou, China
| | - Wei Mao
- Key Laboratory of Integrative Chinese and Western Medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, Hangzhou, China
- Department of Cardiology, Zhengjiang Hospital, Hangzhou, China
| |
Collapse
|
48
|
Shi Q, Lin M, Cheng X, Zhang Z, Deng S, Lang K, Yang Z, Sun X. KPNB1-mediated nuclear import in cancer. Eur J Pharmacol 2023; 955:175925. [PMID: 37473981 DOI: 10.1016/j.ejphar.2023.175925] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 07/03/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
Dysregulation of nucleocytoplasmic shuttling impairs cellular homeostasis and promotes cancer development. KPNB1 is a member of karyopherin β family, mediating the transportation of proteins from the cytoplasm to the nucleus. In a variety of cancers, the expression of KPNB1 is upregulated to facilitate tumor growth and progression. Both downregulation of KPNB1 level and inhibition of KPNB1 activity prevent the entry of cancer-related transcription factors into the nucleus, subsequently suppressing the proliferation and metastasis of cancer cells. Currently, five KPNB1 inhibitors have been reported and exhibited good efficacy against cancer. This paper provides an overview of the role and mechanism of KPNB1 in different cancers and KPNB1-targeted anticancer compounds which hold promise for the future.
Collapse
Affiliation(s)
- Qiwen Shi
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Mengxia Lin
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Xiang Cheng
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Ziyuan Zhang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Shufen Deng
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Ke Lang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Zhikun Yang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Xuanrong Sun
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China.
| |
Collapse
|
49
|
Grazda R, Seyfried AN, Maddipatti KR, Fredman G, MacNamara KC. Resolvin E1 improves efferocytosis and rescues severe aplastic anemia in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.15.528688. [PMID: 36909559 PMCID: PMC10002513 DOI: 10.1101/2023.02.15.528688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Current treatments for severe aplastic anemia (SAA) rely on hematopoietic stem cell (HSC) transplantation and immunosuppressive therapies, however these treatments are not always effective. While immune-mediated destruction and inflammation are known drivers of SAA, the underlying mechanisms that lead to persistent inflammation are unknown. Using an established mouse model of SAA, we observed a significant increase in apoptotic cells within the bone marrow (BM) and demonstrate impaired efferocytosis in SAA mice, as compared to radiation controls. Single-cell transcriptomic analysis revealed heterogeneity among BM monocytes and unique populations emerged during SAA characterized by increased inflammatory signatures and significantly increased expression of Sirpa and Cd47. CD47, a "don't eat me" signal, was increased on both live and apoptotic BM cells, concurrent with markedly increased expression of signal regulatory protein alpha (SIRPα) on monocytes. Functionally, SIRPα blockade improved cell clearance and reduced accumulation of CD47-positive apoptotic cells. Lipidomic analysis revealed a reduction in the precursors of specialized pro-resolving lipid mediators (SPMs) and increased prostaglandins in the BM during SAA, indicative of impaired inflammation resolution. Specifically, 18-HEPE, a precursor of E-series resolvins, was significantly reduced in SAA-induced mice relative to radiation controls. Treatment of SAA mice with Resolvin E1 (RvE1) improved efferocytic function, BM cellularity, platelet output, and survival. Our data suggest that impaired efferocytosis and inflammation resolution contributes to SAA progression and demonstrate that SPMs, such as RvE1, offer new and/or complementary treatments for SAA that do not rely on immune suppression.
Collapse
Affiliation(s)
- Rachel Grazda
- Department of Immunology and Microbiology, Albany Medical College, Albany, New York, USA
| | - Allison N. Seyfried
- Department of Immunology and Microbiology, Albany Medical College, Albany, New York, USA
- Current address: Institute for Clinical Pharmacodynamics, Schenectady, NY, USA
| | - Krishna Rao Maddipatti
- Department of Pathology, Lipidomics Core Facility, Wayne State University, Detroit, Michigan, USA
| | - Gabrielle Fredman
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Katherine C. MacNamara
- Department of Immunology and Microbiology, Albany Medical College, Albany, New York, USA
| |
Collapse
|
50
|
Gadiyar V, Patel G, Chen J, Vigil D, Ji N, Campbell V, Sharma K, Shi Y, Weiss MM, Birge RB, Davra V. Targeted degradation of MERTK and other TAM receptor paralogs by heterobifunctional targeted protein degraders. Front Immunol 2023; 14:1135373. [PMID: 37545504 PMCID: PMC10397400 DOI: 10.3389/fimmu.2023.1135373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 05/26/2023] [Indexed: 08/08/2023] Open
Abstract
TAM receptors (TYRO3, AXL, and MERTK) comprise a family of homologous receptor tyrosine kinases (RTK) that are expressed across a range of liquid and solid tumors where they contribute to both oncogenic signaling to promote tumor proliferation and survival, as well as expressed on myeloid and immune cells where they function to suppress host anti-tumor immunity. In recent years, several strategies have been employed to inhibit TAM kinases, most notably small molecule tyrosine kinase inhibitors and inhibitory neutralizing monoclonal antibodies (mAbs) that block receptor dimerization. Targeted protein degraders (TPD) use the ubiquitin proteasome pathway to redirect E3 ubiquitin ligase activity and target specific proteins for degradation. Here we employ first-in-class TPDs specific for MERTK/TAMs that consist of a cereblon E3 ligase binder linked to a tyrosine kinase inhibitor targeting MERTK and/or AXL and TYRO3. A series of MERTK TPDs were designed and investigated for their capacity to selectively degrade MERTK chimeric receptors, reduce surface expression on primary efferocytic bone marrow-derived macrophages, and impact on functional reduction in efferocytosis (clearance of apoptotic cells). We demonstrate proof-of-concept and establish that TPDs can be tailored to either selectivity degrades MERTK or concurrently degrade multiple TAMs and modulate receptor expression in vitro and in vivo. This work demonstrates the utility of proteome editing, enabled by tool degraders developed here towards dissecting the therapeutically relevant pathway biology in preclinical models, and the ability for TPDs to degrade transmembrane proteins. These data also provide proof of concept that TPDs may serve as a viable therapeutic strategy for targeting MERTK and other TAMs and that this technology could be expanded to other therapeutically relevant transmembrane proteins.
Collapse
Affiliation(s)
- Varsha Gadiyar
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, NJ, United States
| | - Gopi Patel
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, NJ, United States
| | - Jesse Chen
- Department of Research and Development, Kymera Therapeutics, Watertown, MA, United States
| | - Dominico Vigil
- Department of Research and Development, Kymera Therapeutics, Watertown, MA, United States
| | - Nan Ji
- Department of Research and Development, Kymera Therapeutics, Watertown, MA, United States
| | - Veronica Campbell
- Department of Research and Development, Kymera Therapeutics, Watertown, MA, United States
| | - Kirti Sharma
- Department of Research and Development, Kymera Therapeutics, Watertown, MA, United States
| | - Yatao Shi
- Department of Research and Development, Kymera Therapeutics, Watertown, MA, United States
| | - Matthew M. Weiss
- Department of Research and Development, Kymera Therapeutics, Watertown, MA, United States
| | - Raymond B. Birge
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, NJ, United States
| | - Viralkumar Davra
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, NJ, United States
| |
Collapse
|