1
|
Ghorbanzadeh S, Khojini JY, Abouali R, Alimardan S, Zahedi M, Tahershamsi Z, Tajbakhsh A, Gheibihayat SM. Clearing the Path: Exploring Apoptotic Cell Clearance in Inflammatory and Autoimmune Disorders for Therapeutic Advancements. Mol Biotechnol 2025; 67:2223-2238. [PMID: 38935260 DOI: 10.1007/s12033-024-01222-6] [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: 12/13/2023] [Accepted: 01/22/2024] [Indexed: 06/28/2024]
Abstract
Inflammatory and autoimmune disorders, characterized by dysregulated immune responses leading to tissue damage and chronic inflammation, present significant health challenges. This review uniquely focuses on efferocytosis-the phagocyte-mediated clearance of apoptotic cells-and its pivotal role in these disorders. We delve into the intricate mechanisms of efferocytosis' four stages and their implications in disease pathogenesis, distinguishing our study from previous literature. Our findings highlight impaired efferocytosis in conditions like atherosclerosis and asthma, proposing its targeting as a novel therapeutic strategy. We discuss the therapeutic potential of efferocytosis in modulating immune responses and resolving inflammation, offering a new perspective in treating inflammatory disorders.
Collapse
Affiliation(s)
- Shadi Ghorbanzadeh
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Science, Bandar Abbas, Iran
| | - Javad Yaghmoorian Khojini
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, P.O. Box: 8915173143, Yazd, IR, Iran
| | - Reza Abouali
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
| | - Sajad Alimardan
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Zahedi
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Tahershamsi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, P.O. Box: 8915173143, Yazd, IR, Iran.
- Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
2
|
Yuan J, Liao YS, Zhang TC, Tang YQ, Yu P, Liu YN, Cai DJ, Yu SG, Zhao L. Integrating Bulk RNA and Single-Cell Sequencing Data Unveils Efferocytosis Patterns and ceRNA Network in Ischemic Stroke. Transl Stroke Res 2025; 16:733-746. [PMID: 38678526 DOI: 10.1007/s12975-024-01255-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/17/2024] [Accepted: 04/18/2024] [Indexed: 05/01/2024]
Abstract
Excessive inflammatory response following ischemic stroke (IS) injury is a key factor affecting the functional recovery of patients. The efferocytic clearance of apoptotic cells within ischemic brain tissue is a critical mechanism for mitigating inflammation, presenting a promising avenue for the treatment of ischemic stroke. However, the cellular and molecular mechanisms underlying efferocytosis in the brain after IS and its impact on brain injury and recovery are poorly understood. This study explored the roles of inflammation and efferocytosis in IS with bioinformatics. Three Gene Expression Omnibus Series (GSE) (GSE137482-3 m, GSE137482-18 m, and GSE30655) were obtained from NCBI (National Center for Biotechnology Information) and GEO (Gene Expression Omnibus). Differentially expressed genes (DEGs) were processed for GSEA (Gene Set Enrichment Analysis), GO (Gene Ontology Functional Enrichment Analysis), and KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway analyses. Efferocytosis-related genes were identified from the existing literature, following which the relationship between Differentially Expressed Genes (DEGs) and efferocytosis-related genes was examined. The single-cell dataset GSE174574 was employed to investigate the distinct expression profiles of efferocytosis-related genes. The identified hub genes were verified using the dataset of human brain and peripheral blood sample datasets GSE56267 and GSE122709. The dataset GSE215212 was used to predict competing endogenous RNA (ceRNA) network, and GSE231431 was applied to verify the expression of differential miRNAs. At last, the middle cerebral artery (MCAO) model was established to validate the efferocytosis process and the expression of hub genes. DEGs in two datasets were significantly enriched in pathways involved in inflammatory response and immunoregulation. Based on the least absolute shrinkage and selection operator (LASSO) analyses, we identified hub efferocytosis-related genes (Abca1, C1qc, Ptx3, Irf5, and Pros1) and key transcription factors (Stat5). The scRNA-seq analysis showed that these hub genes were mainly expressed in microglia and macrophages which are the main cells with efferocytosis function in the brain. We then identified miR-125b-5p as a therapeutic target of IS based on the ceRNA network. Finally, we validated the phagocytosis and clearance of dead cells by efferocytosis and the expression of hub gene Abca1 in MCAO mice models.
Collapse
Affiliation(s)
- Jing Yuan
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan Province, China
| | - Yu-Sha Liao
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan Province, China
| | - Tie-Chun Zhang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan Province, China
| | - Yu-Qi Tang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan Province, China
| | - Pei Yu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan Province, China
| | - Ya-Ning Liu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan Province, China
| | - Ding-Jun Cai
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan Province, China
| | - Shu-Guang Yu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan Province, China
| | - Ling Zhao
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan Province, China.
| |
Collapse
|
3
|
Ju K, Liu X, Wang Q, Liu X, Li D, Tan B. Integration of Machine Learning Algorithms and Single-Cell Sequencing Analysis Reveals the Efferocytosis-Related Molecular Subtype and Prognostic Scoring Index in Colon Adenocarcinoma. J Gastroenterol Hepatol 2025. [PMID: 40296254 DOI: 10.1111/jgh.16985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 04/08/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Colon adenocarcinoma (COAD) is a leading cause of cancer-related mortality, with limited therapies for advanced stages. Efferocytosis, the clearance of apoptotic cells, modulates tumor immunity and progression. We investigated efferocytosis-related genes (ERRGs) in COAD through multiomics integration. METHODS We analyzed multiomics data from public databases to identify differentially expressed ERRGs and their molecular subtypes. An ERRG score index was developed using integrated machine learning algorithms to evaluate its predictive capacity. Single-cell sequencing and in vitro functional assays were performed to validate key findings. RESULTS Among 162 ERRGs, 22 were dysregulated in COAD. Three molecular subtypes exhibited distinct prognoses, immune profiles, and therapy responses. The ERRG score system accurately predicted clinical outcomes, with low scores correlating with improved survival and sensitivity to certain drugs. Single-cell analysis highlighted TIMP1 as a key regulator, confirmed by its knockdown suppressing tumor proliferation and migration in vitro. CONCLUSION ERRGs demonstrate prognostic and therapeutic relevance in COAD, providing insights into molecular subtyping and immunotherapy prediction. TIMP1 emerges as a potential therapeutic target, warranting further clinical validation.
Collapse
Affiliation(s)
- Kun Ju
- Department of Emergency, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaolei Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qian Wang
- Medical Records Management Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xichun Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dalue Li
- Department of Emergency, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bin Tan
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
4
|
Liu YQ, Li ZZ, Han YL, Wang QB. The role of efferocytosis in inflammatory bowel disease. Front Immunol 2025; 16:1524058. [PMID: 40040696 PMCID: PMC11876057 DOI: 10.3389/fimmu.2025.1524058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/16/2025] [Indexed: 03/06/2025] Open
Abstract
Efferocytosis is the process by which various phagocytes clear apoptotic cells. In recent years, an increasing body of evidence has emphasized the importance of efferocytosis in maintaining internal homeostasis. Intestinal macrophages play a crucial role in modulating intestinal inflammation and promoting tissue repair. Inflammatory bowel disease (IBD) is a chronic, progressive, and relapsing condition, primarily marked by the presence of ulcers in the digestive tract. The exact mechanisms underlying IBD are not yet fully understood, and current treatment approaches mainly aim at repairing the damaged intestinal mucosa and reducing inflammatory responses to ease symptoms.This article provides new perspectives on IBD treatment and clinical management by examining the expression of macrophage efferocytosis-related molecules, the effects of efferocytosis on IBD development, the various roles of macrophage efferocytosis in IBD, and treatment strategies for IBD that focus on efferocytosis.
Collapse
Affiliation(s)
- Yi-Qian Liu
- Institute of Acupuncture and Moxibustion, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Zhan-Zhan Li
- Academy of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yong-Li Han
- Acupuncture Department, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Qing-Bo Wang
- Acupuncture Department, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
5
|
Bavarsad SB, Shahryarhesami S, Karami N, Naseri N, Tajbakhsh A, Gheibihayat SM. Efferocytosis and infertility: Implications for diagnosis and therapy. J Reprod Immunol 2025; 167:104413. [PMID: 39631138 DOI: 10.1016/j.jri.2024.104413] [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: 09/30/2024] [Revised: 11/15/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
Recent research has shed light on the intricate connection between efferocytosis and infertility, revealing its dysregulation as a contributing factor in various reproductive diseases. Despite the multifaceted nature of infertility etiology, the impact of insufficient clearance of apoptotic cells on fertility has emerged as a focal point. Notably, the removal of apoptotic cells through phagocytosis in the female reproductive system has been a subject of extensive investigation in the field of infertility. Additionally, special functions performed by immune system cell types, such as macrophages and Sertoli cells, in the male reproductive system underscore their significance in spermatogenesis and the efferocytosis of apoptotic germ cells. Dysregulation of efferocytosis emerges as a critical factor contributing to reproductive challenges, such as low pregnancy rates, miscarriages, and implantation failures. Moreover, defective efferocytosis can lead to compromised implantation, recurrent miscarriages, and unsuccessful assisted reproductive procedures. This review article aims to provide a comprehensive overview of efferocytosis in the context of infertility. Molecular mechanisms underlying efferocytosis, its relevance in both female and male infertility, and its implications in various reproductive diseases are elucidated. The elucidation of the intricate relationship between efferocytosis and infertility not only facilitates diagnosis but also paves the way for targeted therapeutic interventions.
Collapse
Affiliation(s)
| | - Soroosh Shahryarhesami
- Functional Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, Heidelberg 69120, Germany.
| | - Noorodin Karami
- Genetics Department, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Nasim Naseri
- Department of Animal Sciences and Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
6
|
Laddha K, Sobhia ME. Optimizing antibody stability and efficacy in CD47- SIRPα inhibition via computational approaches. Mol Divers 2025:10.1007/s11030-024-11037-x. [PMID: 39832086 DOI: 10.1007/s11030-024-11037-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 10/28/2024] [Indexed: 01/22/2025]
Abstract
CD47, a cell surface protein, serves as a "don't eat me" signal that prevents immune cells from engulfing healthy cells upon its interaction with SIRPα. Cancer cells exploit this mechanism by overexpressing CD47 to evade immune destruction. Blocking the interaction between CD47 and its receptor, SIRPα, is a promising therapeutic strategy. Targeting the interactions between these surface proteins with small molecules is quite challenging, and on the other hand, antibodies offer potential. However, the interactions between antigen (CD47) and antibody (B6H12.2) play a crucial role in this scenario, and increasing the affinity by mutating the interacting residues might impact the inclination and effectiveness of the antibody towards antigen. Thus, this study focuses on designing antibodies with increased affinity and stability towards the antigen compared to the wild-type. Residual scanning calculations were performed to mutate the interacting as well as the hydrophobic residues of the antibody and affinity was assessed. Computational approaches, including antigen-antibody docking studies and molecular dynamics simulations, were employed to evaluate the affinity, stability and therapeutic potential of these modified antibodies.
Collapse
Affiliation(s)
- Kapil Laddha
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, 160062, India
| | - M Elizabeth Sobhia
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, 160062, India.
| |
Collapse
|
7
|
Vafadar A, Tajbakhsh A, Hosseinpour-Soleimani F, Savardshtaki A, Hashempur MH. Phytochemical-mediated efferocytosis and autophagy in inflammation control. Cell Death Discov 2024; 10:493. [PMID: 39695119 DOI: 10.1038/s41420-024-02254-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/06/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
Efferocytosis, the clearance of apoptotic cells, is a critical process that maintains tissue homeostasis and immune regulation. Defective efferocytosis is linked to the development of chronic inflammatory conditions, including atherosclerosis, neurological disorders, and autoimmune diseases. Moreover, the interplay between autophagy and efferocytosis is crucial for inflammation control, as autophagy enhances the ability of phagocytic cells. Efficient efferocytosis, in turn, regulates autophagic pathways, fostering a balanced cellular environment. Dysregulation of this balance can contribute to the pathogenesis of various disorders. Phytochemicals, bioactive compounds found in plants, have emerged as promising therapeutic agents owing to their diverse pharmacological properties, including antioxidant, anti-inflammatory, and immunomodulatory effects. This review aims to highlight the pivotal role of phytochemicals in enhancing efferocytosis and autophagy and explore their potential in the prevention and treatment of related disorders. This study examines how phytochemicals influence key aspects of efferocytosis, including phagocytic cell activation, macrophage polarization, and autophagy induction. The therapeutic potential of phytochemicals in atherosclerosis and neurological diseases is highlighted, emphasizing their ability to enhance efferocytosis and autophagy and reduce inflammation. This review also discusses innovative approaches, such as nanoformulations and combination therapies to improve the targeting and bioavailability of phytochemicals. Ultimately, this study inspires further research and clinical applications in phytochemical-mediated efferocytosis enhancement for managing chronic inflammatory and autoimmune conditions.
Collapse
Affiliation(s)
- Asma Vafadar
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Hosseinpour-Soleimani
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Applied Cell Sciences and Tissue Engineering, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardshtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hashem Hashempur
- Research Center for Traditional Medicine and History of Medicine, Department of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
8
|
Gao L, Manaenko A, Zeng F, Li J, Liu L, Xie R, Zhang X, Zhang JH, Mei Q, Tang J, Hu Q. Efferocytosis: A new therapeutic target for stroke. Chin Med J (Engl) 2024; 137:2843-2850. [PMID: 39528491 DOI: 10.1097/cm9.0000000000003363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Indexed: 11/16/2024] Open
Abstract
ABSTRACT Efferocytosis refers to the process that phagocytes recognize and remove the apoptotic cells, which is essential for maintaining tissue homeostasis both in physiological and pathological conditions. Numerous studies have demonstrated that efferocytosis can prevent secondary necrosis and proinflammatory factor release, leading to the resolution of inflammation and tissue immunological tolerance in numerous diseases such as stroke. Stroke is a leading cause of death and morbidity for adults worldwide. Persistent inflammation triggered by the dead cells or cell debris is a major contributor to post-stroke brain damage. Effective efferocytosis might be an efficient strategy to minimize inflammation and restore brain homeostasis for neuronal regeneration and function recovery. In this review, we will discuss the phagocytes in the brain, the molecular mechanisms underlying efferocytosis, the role of efferocytosis in inflammation resolution, and the potential therapeutic applications targeting efferocytosis in stroke.
Collapse
Affiliation(s)
- Li Gao
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Anatol Manaenko
- Clinical Neuroanatomy, Department of Neurology, School of Medicine, Ulm University, Ulm 89081, Germany
| | - Feng Zeng
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Jingchen Li
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Lele Liu
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Ruichuan Xie
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Xiaohua Zhang
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA
| | - Qiyong Mei
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA
| | - Qin Hu
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai 200127, China
| |
Collapse
|
9
|
Mo K, Wang Y, Lu C, Li Z. Insight into the role of macrophages in periodontitis restoration and development. Virulence 2024; 15:2427234. [PMID: 39535076 PMCID: PMC11572313 DOI: 10.1080/21505594.2024.2427234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 08/31/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Periodontitis is one of the chronic diseases that have the greatest impact on human health, and it is associated with several other chronic diseases. Tissue damage associated with periodontitis is often connected with immune response. Immune cells are a crucial component of the human immune system and are directly involved in periodontitis during the inflammatory phase of the disease. Macrophages, as a key component of the immune system, are responsible for defence, antigen presentation and phagocytosis in healthy tissue. They are also closely linked to the development and resolution of periodontitis, through mechanisms such as macrophage polarization, pattern recognition receptors recognition, efferocytosis, and Specialized Pro-resolving Mediators (SPMs) production. Additionally, apoptosis and autophagy are also known to play a role in the recovery of periodontitis. This review aims to investigate the aforementioned mechanisms in more detail and identify novel therapeutic approaches for periodontitis.
Collapse
Affiliation(s)
- Keyin Mo
- School of Stomatology, Jinan University, Guangzhou, China
| | - Yijue Wang
- West China School of Stomatology, Sichuan University, Chengdu, China
| | - Chunting Lu
- Science and Education Office, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Zejian Li
- Hospital of Stomatology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| |
Collapse
|
10
|
Xie Y, Chen H, Zhang X, Zhang J, Zhang K, Wang X, Min S, Wang X, Lian C. Integration of the bulk transcriptome and single-cell transcriptome reveals efferocytosis features in lung adenocarcinoma prognosis and immunotherapy by combining deep learning. Cancer Cell Int 2024; 24:388. [PMID: 39580462 PMCID: PMC11585238 DOI: 10.1186/s12935-024-03571-3] [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: 04/17/2024] [Accepted: 11/10/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Efferocytosis (ER) refers to the process of phagocytic clearance of programmed dead cells, and studies have shown that it is closely related to tumor immune escape. METHODS This study was based on a comprehensive analysis of TCGA, GEO and CTRP databases. ER-related genes were collected from previous literature, univariate Cox regression was performed and consistent clustering was performed to categorize lung adenocarcinoma (LUAD) patients into two subgroups. Lasso regression and multivariate Cox regression analyses were used to construct ER-related prognostic features, and multiple immune infiltration algorithms were used to assess the correlation between the extracellular burial-related risk score (ERGRS) and tumor microenvironment (TME). And the key gene HAVCR1 was identified by deep learning, etc. Finally, pan-cancer analysis of the key genes was performed and in vitro experiments were conducted to verify the promotional effect of HAVCR1 on LUAD progression. RESULTS A total of 33 ER-related genes associated with the prognosis of LUAD were identified, and the prognostic signature of ERGRS was successfully constructed to predict the overall survival (OS) and treatment response of LUAD patients. The high-risk group was highly enriched in some oncogenic pathways, while the low-ERGRS group was highly enriched in some immune-related pathways. In addition, the high ERGRS group had higher TMB, TNB and TIDE scores and lower immune scores. The low-risk group had better immunotherapeutic response and less likelihood of immune escape. Drug sensitivity analysis revealed that BRD-K92856060, monensin and hexaminolevulinate may be potential therapeutic agents for the high-risk group. And ERGRS was validated in several cohorts. In addition, HAVCR1 is one of the key genes, and knockdown of HAVCR1 in vitro significantly reduced the proliferation, migration and invasion ability of lung adenocarcinoma cells. CONCLUSION Our study developed a novel prognostic signature of efferocytosis-related genes. This prognostic signature accurately predicted survival prognosis as well as treatment outcome in LUAD patients and explored the role of HAVCR1 in lung adenocarcinoma progression.
Collapse
Affiliation(s)
- Yiluo Xie
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233030, China
- Department of Clinical Medicine, Bengbu Medical University, Bengbu, 233030, China
| | - Huili Chen
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, 233030, China
| | - Xueying Zhang
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233030, China
| | - Jing Zhang
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu, 233030, China
| | - Kai Zhang
- Department of Clinical Medicine, Bengbu Medical University, Bengbu, 233030, China
| | - Xinyu Wang
- Department of Clinical Medicine, Bengbu Medical University, Bengbu, 233030, China
| | - Shengping Min
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233030, China
| | - Xiaojing Wang
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233030, China.
| | - Chaoqun Lian
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, 233030, China.
| |
Collapse
|
11
|
Li W, Huang Y, Liu J, Zhou Y, Sun H, Fan Y, Liu F. Defective macrophage efferocytosis in advanced atherosclerotic plaque and mitochondrial therapy. Life Sci 2024; 359:123204. [PMID: 39491771 DOI: 10.1016/j.lfs.2024.123204] [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: 08/05/2024] [Revised: 10/02/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease primarily affecting large and medium-sized arterial vessels, characterized by lipoprotein disorders, intimal thickening, smooth muscle cell proliferation, and the formation of vulnerable plaques. Macrophages (MΦs) play a vital role in the inflammatory response throughout all stages of atherosclerotic development and are considered significant therapeutic targets. In early lesions, macrophage efferocytosis rapidly eliminates harmful cells. However, impaired efferocytosis in advanced plaques perpetuates the inflammatory microenvironment of AS. Defective efferocytosis has emerged as a key factor in atherosclerotic pathogenesis and the progression to severe cardiovascular disease. Herein, this review probes into investigate the potential mechanisms at the cellular, molecular, and organelle levels underlying defective macrophage efferocytosis in advanced lesion plaques. In the inflammatory microenvironments of AS with interactions among diverse inflammatory immune cells, impaired macrophage efferocytosis is strongly linked to multiple factors, such as a lower absolute number of phagocytes, the aberrant expression of crucial molecules, and impaired mitochondrial energy provision in phagocytes. Thus, focusing on molecular targets to enhance macrophage efferocytosis or targeting mitochondrial therapy to restore macrophage metabolism homeostasis has emerged as a potential strategy to mitigate the progression of advanced atherosclerotic plaque, providing various treatment options.
Collapse
Affiliation(s)
- Wanling Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Yaqing Huang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Jun Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Yue Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Hongyu Sun
- The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Yonghong Fan
- The General Hospital of Western Theater Command, Chengdu 610083, China.
| | - Feila Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China.
| |
Collapse
|
12
|
Xing J, Wang K, Xu YC, Pei ZJ, Yu QX, Liu XY, Dong YL, Li SF, Chen Y, Zhao YJ, Yao F, Ding J, Hu W, Zhou RP. Efferocytosis: Unveiling its potential in autoimmune disease and treatment strategies. Autoimmun Rev 2024; 23:103578. [PMID: 39004157 DOI: 10.1016/j.autrev.2024.103578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 07/06/2024] [Accepted: 07/07/2024] [Indexed: 07/16/2024]
Abstract
Efferocytosis is a crucial process whereby phagocytes engulf and eliminate apoptotic cells (ACs). This intricate process can be categorized into four steps: (1) ACs release "find me" signals to attract phagocytes, (2) phagocytosis is directed by "eat me" signals emitted by ACs, (3) phagocytes engulf and internalize ACs, and (4) degradation of ACs occurs. Maintaining immune homeostasis heavily relies on the efficient clearance of ACs, which eliminates self-antigens and facilitates the generation of anti-inflammatory and immunosuppressive signals that maintain immune tolerance. However, any disruptions occurring at any of the efferocytosis steps during apoptosis can lead to a diminished efficacy in removing apoptotic cells. Factors contributing to this inefficiency encompass dysregulation in the release and recognition of "find me" or "eat me" signals, defects in phagocyte surface receptors, bridging molecules, and other signaling pathways. The inadequate clearance of ACs can result in their rupture and subsequent release of self-antigens, thereby promoting immune responses and precipitating the onset of autoimmune diseases such as systemic lupus erythematosus, rheumatoid arthritis, type 1 diabetes, and multiple sclerosis. A comprehensive understanding of the efferocytosis process and its implications can provide valuable insights for developing novel therapeutic strategies that target this process to prevent or treat autoimmune diseases.
Collapse
Affiliation(s)
- Jing Xing
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; School of pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ke Wang
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yu-Cai Xu
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; School of pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ze-Jun Pei
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; School of pharmacy, Anhui Medical University, Hefei 230032, China
| | - Qiu-Xia Yu
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; School of pharmacy, Anhui Medical University, Hefei 230032, China
| | - Xing-Yu Liu
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; School of pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ya-Lu Dong
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; School of pharmacy, Anhui Medical University, Hefei 230032, China
| | - Shu-Fang Li
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yong Chen
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Ying-Jie Zhao
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Feng Yao
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Jie Ding
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Wei Hu
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China.
| | - Ren-Peng Zhou
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
13
|
Chen P, Li Z, Li N. Establishment of a novel efferocytosis potential index predicts prognosis and immunotherapy response in cancers. Heliyon 2024; 10:e30337. [PMID: 38707349 PMCID: PMC11068824 DOI: 10.1016/j.heliyon.2024.e30337] [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: 10/09/2023] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/07/2024] Open
Abstract
The biological function and prognostic value of efferocytosis in cancer remains unclear. In this study, we systematically analysed the expression profiles and genetic variations of 50 efferocytosis-related regulator genes in 33 cancer types. Using data from The Cancer Genome Atlas, we established an efferocytosis potential index (EPI) model to represent the efferocytosis level in each cancer type. The relationship between the EPI and prognosis, immune-related molecules, specific pathways, and drug sensitivity was determined. We found that efferocytosis regulator genes were abnormally expressed in cancer tissue, perhaps owing to copy number variations, gene alterations, and DNA methylation. For the most part, the EPI was higher in tumour vs. normal tissues. In most of the 33 cancer types, it positively correlated with cell death- and immune-related pathway enrichment, the tumour microenvironment, immune infiltration, and drug sensitivity. For specific cancers, a high EPI may be a prognostic risk factor and, in patients treated receiving immune checkpoint therapy, a predictor of poor prognosis. Our study reveals the biological functions of efferocytosis-related regulator genes in distinct cancers and highlights the potential of efferocytosis intervention in cancer therapy.
Collapse
Affiliation(s)
- Peng Chen
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Zhanzhan Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Na Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| |
Collapse
|
14
|
Zaid A, Ariel A. Harnessing anti-inflammatory pathways and macrophage nano delivery to treat inflammatory and fibrotic disorders. Adv Drug Deliv Rev 2024; 207:115204. [PMID: 38342241 DOI: 10.1016/j.addr.2024.115204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 12/08/2023] [Accepted: 02/05/2024] [Indexed: 02/13/2024]
Abstract
Targeting specific organs and cell types using nanotechnology and sophisticated delivery methods has been at the forefront of applicative biomedical sciences lately. Macrophages are an appealing target for immunomodulation by nanodelivery as they are heavily involved in various aspects of many diseases and are highly plastic in their nature. Their continuum of functional "polarization" states has been a research focus for many years yielding a profound understanding of various aspects of these cells. The ability of monocyte-derived macrophages to metamorphose from pro-inflammatory to reparative and consequently to pro-resolving effectors has raised significant interest in its therapeutic potential. Here, we briefly survey macrophages' ontogeny and various polarization phenotypes, highlighting their function in the inflammation-resolution shift. We review their inducing mediators, signaling pathways, and biological programs with emphasis on the nucleic acid sensing-IFN-I axis. We also portray the polarization spectrum of macrophages and the characteristics of their transition between different subtypes. Finally, we highlighted different current drug delivery methods for targeting macrophages with emphasis on nanotargeting that might lead to breakthroughs in the treatment of wound healing, bone regeneration, autoimmune, and fibrotic diseases.
Collapse
Affiliation(s)
- Ahmad Zaid
- Department of Biology and Human Biology, University of Haifa, Haifa, 3498838 Israel
| | - Amiram Ariel
- Department of Biology and Human Biology, University of Haifa, Haifa, 3498838 Israel.
| |
Collapse
|
15
|
Soldano S, Smith V, Montagna P, Gotelli E, Campitiello R, Pizzorni C, Paolino S, Sulli A, Cere A, Cutolo M. Nintedanib downregulates the profibrotic M2 phenotype in cultured monocyte-derived macrophages obtained from systemic sclerosis patients affected by interstitial lung disease. Arthritis Res Ther 2024; 26:74. [PMID: 38509595 PMCID: PMC10953168 DOI: 10.1186/s13075-024-03308-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/10/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Systemic sclerosis (SSc) is an autoimmune connective tissue disease characterized by vasculopathy and progressive fibrosis of skin and several internal organs, including lungs. Macrophages are the main cells involved in the immune-inflammatory damage of skin and lungs, and alternatively activated (M2) macrophages seem to have a profibrotic role through the release of profibrotic cytokines (IL10) and growth factors (TGFβ1). Nintedanib is a tyrosine kinase inhibitor targeting several fibrotic mediators and it is approved for the treatment of SSc-related interstitial lung disease (ILD). The study aimed to evaluate the effect of nintedanib in downregulating the profibrotic M2 phenotype in cultured monocyte-derived macrophages (MDMs) obtained from SSc-ILD patients. METHODS Fourteen SSc patients, fulfilling the 2013 ACR/EULAR criteria for SSc, 10 SSc patients affected by ILD (SSc-ILD pts), 4 SSc patients non affected by ILD (SSc pts no-ILD), and 5 voluntary healthy subjects (HSs), were recruited at the Division of Clinical Rheumatology-University of Genova, after obtaining Ethical Committee approval and patients' informed consent. Monocytes were isolated from peripheral blood, differentiated into MDMs, and then maintained in growth medium without any treatment (untreated cells), or treated with nintedanib (0.1 and 1µM) for 3, 16, and 24 h. Gene expression of macrophage scavenger receptors (CD204, CD163), mannose receptor-1 (CD206), Mer tyrosine kinase (MerTK), identifying M2 macrophages, together with TGFβ1 and IL10, were evaluated by quantitative real-time polymerase chain reaction. Protein synthesis was investigated by Western blotting and the level of active TGFβ1 was evaluated by ELISA. Statistical analysis was carried out using non-parametric Wilcoxon test. RESULTS Cultured untreated SSc-ILD MDMs showed a significant increased protein synthesis of CD206 (p < 0.05), CD204, and MerTK (p < 0.01), together with a significant upregulation of the gene expression of MerTK and TGFβ1 (p < 0.05; p < 0.01) compared to HS-MDMs. Moreover, the protein synthesis of CD206 and MerTK and the gene expression of TGFβ1 were significantly higher in cultured untreated MDMs from SSc-ILD pts compared to MDMs without ILD (p < 0.05; p < 0.01). In cultured SSc-ILD MDMs, nintedanib 0.1 and 1µM significantly downregulated the gene expression and protein synthesis of CD204, CD206, CD163 (p < 0.05), and MerTK (p < 0.01) compared to untreated cells after 24 h of treatment. Limited to MerTK and IL10, both nintedanib concentrations significantly downregulated their gene expression already after 16 h of treatment (p < 0.05). In cultured SSc-ILD MDMs, nintedanib 0.1 and 1µM significantly reduced the release of active TGFβ1 after 24 h of treatment (p < 0.05 vs. untreated cells). CONCLUSIONS In cultured MDMs from SSc-ILD pts, nintedanib seems to downregulate the profibrotic M2 phenotype through the significant reduction of gene expression and protein synthesis of M2 cell surface markers, together with the significant reduction of TGFβ1 release, and notably MerTK, a tyrosine kinase receptor involved in lung fibrosis.
Collapse
Affiliation(s)
- Stefano Soldano
- Laboratory of Experimental Rheumatology, Division of Clinical Rheumatology, Department of Internal Medicine, University of Genova, Genoa, Italy
| | - Vanessa Smith
- Department of Internal Medicine, Ghent University, Ghent, Belgium
- Department of Rheumatology, Ghent University Hospital, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB Inflammation Research Centre, Ghent, Belgium
| | - Paola Montagna
- Laboratory of Experimental Rheumatology, Division of Clinical Rheumatology, Department of Internal Medicine, University of Genova, Genoa, Italy
| | - Emanuele Gotelli
- Laboratory of Experimental Rheumatology, Division of Clinical Rheumatology, Department of Internal Medicine, University of Genova, Genoa, Italy
| | - Rosanna Campitiello
- Laboratory of Experimental Rheumatology, Division of Clinical Rheumatology, Department of Internal Medicine, University of Genova, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Carmen Pizzorni
- Laboratory of Experimental Rheumatology, Division of Clinical Rheumatology, Department of Internal Medicine, University of Genova, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Sabrina Paolino
- Laboratory of Experimental Rheumatology, Division of Clinical Rheumatology, Department of Internal Medicine, University of Genova, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Alberto Sulli
- Laboratory of Experimental Rheumatology, Division of Clinical Rheumatology, Department of Internal Medicine, University of Genova, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Andrea Cere
- Laboratory of Experimental Rheumatology, Division of Clinical Rheumatology, Department of Internal Medicine, University of Genova, Genoa, Italy
| | - Maurizio Cutolo
- Laboratory of Experimental Rheumatology, Division of Clinical Rheumatology, Department of Internal Medicine, University of Genova, Genoa, Italy.
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| |
Collapse
|
16
|
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
|
17
|
Jamalvandi M, Khayyatzadeh SS, Hayati MJ, Gheibihayat SM. The role of fat-soluble vitamins in efferocytosis. Cell Biochem Funct 2024; 42:e3972. [PMID: 38500392 DOI: 10.1002/cbf.3972] [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: 12/19/2023] [Revised: 02/08/2024] [Accepted: 03/02/2024] [Indexed: 03/20/2024]
Abstract
Cell death and the efficient removal of dead cells are two basic mechanisms that maintain homeostasis in multicellular organisms. efferocytosis, which includes four steps recruitment, recognition, binding and signaling, and engulfment. Effectively and quickly removes apoptotic cells from the body. Any alteration in efferocytosis can lead to several diseases, including autoimmune and inflammatory conditions, atherosclerosis, and cancer. A wide range of dietary components affects apoptosis and, subsequently, efferocytosis. Some vitamins, including fat-soluble vitamins, affect different stages of efferocytosis. Among other things, by affecting macrophages, they are effective in the apoptotic cleansing of cells. Also, polyphenols indirectly intervene in efferocytosis through their effect on apoptosis. Considering that there are limited articles on the effect of nutrition on efferocytosis, in this article we will examine the effect of some dietary components on efferocytosis.
Collapse
Affiliation(s)
- Mona Jamalvandi
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Sayyed Saeid Khayyatzadeh
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammad Javad Hayati
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
- Yazd Cardiovascular Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
18
|
Zhang Z, Zhou Y, Lv Q, Gao K, Li Z, Miao Q, Shen L. Gegen Qinlian Decoction Modulates Atherosclerosis and Lipid Metabolism Through Cellular Interplay and Signaling Pathways. Comb Chem High Throughput Screen 2024; 27:2609-2621. [PMID: 38486386 DOI: 10.2174/0113862073285562240305113728] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 09/27/2024]
Abstract
OBJECTIVE The objective of this study is to investigate Gegen Qinlian decoction (GQD) effects on lipid metabolism and explore its mechanism for preventing and treating atherosclerosis. METHODS An atherosclerotic rat model was established, and after an 8-week high-fat diet, atherosclerosis and non-alcoholic fatty liver disease were assessed. Subsequently, GQD was administered at low and high doses. Histopathological aortic wall changes, hepatic lipid deposition, and blood lipid changes were evaluated. ELISA indicated the influence of TNF-α and IL-13, and Western blotting revealed MerTK, ABCA1, and LXR-α expression. A foam macrophage model was established, and Cell activity was detected by the MTT method. ELISA indicated the influence of PPAR-γ. The expression of ABCA1, ABCA7, ABCG1, GAS6, MerTK, SCARB1, LXR- α and LXR-β mRNA were detected by qPCR. and Western blotting revealed MerTK and LXR-α expression. The impact of drug-containing serum of GQD on efferocytosis-related factors was studied. RESULTS GQD improved atherosclerosis and non-alcoholic fatty liver disease and reduced serum low-density lipoprotein levels in the high-dose group. The high- and low-dose groups showed upregulated ABCA1, MerTK, and LXR-α expression in blood vessels and the liver, respectively. GQD decreased serum TNF-α and increased IL-13 levels. PPAR-γ expression was elevated in the high-, and low-dose groups. In the high-and low-dose groups, ABCA7, GAS6, SCARB1, and LXR-α, ABCA1 and MerTK, and ABCG1 gene expression were upregulated, respectively. Both low- and high-dose serum-containing drugs promoted LXR-β gene expression, and LXR-α protein expression was improved in the high-dose group. CONCLUSION GQD improves rat atherosclerosis and hepatic lipid metabolism by regulating PPAR-γ, LXR-α, LXR-β, ABCA1, ABCA7, and ABCG1 expression and augmenting cellular intercalation through the GAS6/TAM pathway.
Collapse
Affiliation(s)
- Zhiwei Zhang
- Department of Digestive Endocrinology, Beijing Fengtai Hospital of Integrated Traditional and Western Medicine, Beijing, China
| | - Yu Zhou
- Department of Nephrology Endocrinology, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, Beijing, China
| | - Qin Lv
- Department of Digestive Endocrinology, Beijing Fengtai Hospital of Integrated Traditional and Western Medicine, Beijing, China
| | - Kun Gao
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhiguo Li
- Department of Digestive Endocrinology, Beijing Fengtai Hospital of Integrated Traditional and Western Medicine, Beijing, China
| | - Qing Miao
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Shen
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
19
|
Panahi S, Yazdi Z, Sanchouli M, Sajadi F, Motavasselian F, Maghareh Abed H, Beygi Z, Gheibihayat SM. The Role of Extracellular Vesicles in Efferocytosis. Mini Rev Med Chem 2024; 24:634-641. [PMID: 37859308 DOI: 10.2174/0113895575247690230926113455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 06/19/2023] [Accepted: 08/10/2023] [Indexed: 10/21/2023]
Abstract
Efferocytosis is the physiological process of phagocytic clearance of apoptotic cells by both professional phagocytic cells, such as macrophages, and non-professional phagocytic cells, such as epithelial cells. This process is crucial for maintaining tissue homeostasis in normal physiology. Any defects in efferocytosis can lead to pathological consequences and result in inflammatory diseases. Extracellular vesicles (EVs), including exosomes, microvesicles (MVs), and apoptotic vesicles (ApoVs), play a crucial role in proper efferocytosis. These EVs can significantly impact efferocytosis by affecting the polarization of macrophages and impacting calreticulin (CRT), TAM receptors, and MFG-E8. With further knowledge of these effects, new treatment strategies can be proposed for many inflammatory diseases caused by efferocytosis disorders. This review article aims to investigate the role of EVs during efferocytosis and its potential clinical applications in inflammatory diseases.
Collapse
Affiliation(s)
- Susan Panahi
- Department of Microbiology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Zohreh Yazdi
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mahboubeh Sanchouli
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Fatemehsadat Sajadi
- Oral and Dental Diseases Research Center, Department of Pediatric Dentistry, School of Dentistry, Kerman University of Medical Sciences, Kerman, Iran
| | - Fateme Motavasselian
- Department of Persian Medicine, School of Persian Medicine, Shahid Sadoughi University of Medical Sciences, Ardakan, Iran
- The Research Center of Persian Medicine, Shahid Sadoughi University of Medical Sciences, Ardakan, Iran
| | - Hedyeh Maghareh Abed
- Department of Biochemistry, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | - Zahra Beygi
- Department of Biostatistics and Epidemiology, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of midwife and nursing, Maybod Branch, Islamic Azad University, Maybod, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
20
|
Vafadar A, Vosough P, Jahromi HK, Tajbakhsh A, Savardshtaki A, Butler AE, Sahebkar A. The role of efferocytosis and transplant rejection: Strategies in promoting transplantation tolerance using apoptotic cell therapy and/or synthetic particles. Cell Biochem Funct 2023; 41:959-977. [PMID: 37787641 DOI: 10.1002/cbf.3852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/26/2023] [Accepted: 08/24/2023] [Indexed: 10/04/2023]
Abstract
Recently, efforts have been made to recognize the precise reason(s) for transplant failure and the process of rejection utilizing the molecular signature. Most transplant recipients do not appreciate the unknown length of survival of allogeneic grafts with the existing standard of care. Two noteworthy immunological pathways occur during allogeneic transplant rejection. A nonspecific innate immune response predominates in the early stages of the immune reaction, and allogeneic antigens initiate a donor-specific adaptive reaction. Though the adaptive response is the major cause of allograft rejection, earlier pro-inflammatory responses that are part of the innate immune response are also regarded as significant in graft loss. The onset of the innate and adaptive immune response causes chronic and acute transplant rejection. Currently employed immunosuppressive medications have shown little or no influence on chronic rejection and, as a result, on overall long-term transplant survival. Furthermore, long-term pharmaceutical immunosuppression is associated with side effects, toxicity, and an increased risk of developing diseases, both infectious and metabolic. As a result, there is a need for the development of innovative donor-specific immunosuppressive medications to regulate the allorecognition pathways that induce graft loss and to reduce the side effects of immunosuppression. Efferocytosis is an immunomodulatory mechanism with fast and efficient clearance of apoptotic cells (ACs). As such, AC therapy strategies have been suggested to limit transplant-related sequelae. Efferocytosis-based medicines/treatments can also decrease the use of immunosuppressive drugs and have no detrimental side effects. Thus, this review aims to investigate the impact of efferocytosis on transplant rejection/tolerance and identify approaches using AC clearance to increase transplant viability.
Collapse
Affiliation(s)
- Asma Vafadar
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parisa Vosough
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Kargar Jahromi
- Research Center for Non-Communicable Disease, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Amir Tajbakhsh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardshtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland - Bahrain, Adliya, Bahrain
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
21
|
Mohammad-Rafiei F, Moadab F, Mahmoudi A, Navashenaq JG, Gheibihayat SM. Efferocytosis: a double-edged sword in microbial immunity. Arch Microbiol 2023; 205:370. [PMID: 37925389 DOI: 10.1007/s00203-023-03704-8] [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: 09/03/2023] [Revised: 09/30/2023] [Accepted: 10/10/2023] [Indexed: 11/06/2023]
Abstract
Efferocytosis is characterized as the rapid and efficient process by which dying or dead cells are removed. This type of clearance is initiated via "find-me" signals, and then, carries on by "eat-me" and "don't-eat-me" ones. Efferocytosis has a critical role to play in tissue homeostasis and innate immunity. However, some evidence suggests it as a double-edged sword in microbial immunity. In other words, some pathogens have degraded efferocytosis by employing efferocytic mechanisms to bypass innate immune detection and promote infection, despite the function of this process for the control and clearance of pathogens. In this review, the efferocytosis mechanisms from the recognition of dying cells to phagocytic engulfment are initially presented, and then, its diverse roles in inflammation and immunity are highlighted. In this case, much focus is also laid on some bacterial, viral, and parasitic infections caused by Mycobacterium tuberculosis (M. tb), Mycobacterium marinum (M. marinum), Listeria monocytogenes (L. monocytogenes), Chlamydia pneumoniae (CP), Klebsiella pneumoniae (KP), Influenza A virus (IAV), human immunodeficiency virus (HIV), and Leishmania, respectively.
Collapse
Affiliation(s)
- Fatemeh Mohammad-Rafiei
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Moadab
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, USA
| | - Ali Mahmoudi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | | | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
22
|
Lu N, Zhu JF, Lv HF, Zhang HP, Wang PL, Yang JJ, Wang XW. Modulation of oxidized low-density lipoprotein-affected macrophage efferocytosis by mitochondrial calcium uniporter in a murine model. Immunol Lett 2023; 263:14-24. [PMID: 37689315 DOI: 10.1016/j.imlet.2023.09.003] [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: 04/06/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
OBJECTIVE Efferocytosis dysfunction contributes to the progression and rupture of atherosclerotic plaques. Efferocytosis is crucially modulated by intracytoplasmic Ca2+, and mitochondrial calcium uniporter (MCU) complex proteins serve as key channels for regulating Ca2+ concentration. Therefore, it was speculated that MCU may affect the development of atherosclerosis (AS) by regulating efferocytosis. In the present study, we aimed to investigate whether MCU could affect foam cell formation by regulating efferocytosis. METHODS We stimulated primary macrophages (Møs) using oxidized low-density lipoprotein (ox-LDL) to mimic the atherosclerotic microenvironment and treated them with Ru360, an MCU-specific inhibitor, and UNC1062, an inhibitor of efferocytosis. Additionally, we conducted double staining to determine the Mø efferocytosis rate. We measured the expression of MCU complexes and efferocytosis-associated proteins using western blotting (WB) and real-time quantitative polymerase chain reaction (RT-qPCR), respectively. In addition, we separately detected the Ca2+ level in the cytoplasm and mitochondria (MT) using Fluo-4 AM and Rhod-2 methods. We separately determined the reactive oxygen species (ROS) level in cytoplasm and MT using dichlorodihydrofluorescein diacetate (DCFH-DA) fluorescent probing method and Mito-SOXTM superoxide indicator staining. Additionally, we conducted the enzyme-linked immunosorbent assay (ELISA) to detect the production of interleukin-6 (IL-6), interleukin-18 (IL-18), interleukin-1β (IL-1β), and tumor necrosis factor-alpha (TNF-α). Oil Red O staining was performed to measure cytoplasmic lipid levels. RESULTS Ru360 attenuated ox-LDL-induced efferocytosis dysfunction, and attenuated the upregulation of MCU and MCUR1 induced by ox-LDL, and meanwhile attenuated the downregulation of MCUb induced by ox-LDL. Ru360 attenuated the decrease of intracytoplasmic Ca2+ concentration induced by ox- LDL, Ru360 also attenuated the ROS production induced by ox- LDL, attenuated the release of IL-6, IL-18, IL-1β, and TNF-α induced by ox- LDL, and attenuated the increase of intracytoplasmic lipid content induced by ox-LDL. UNC1062 attenuated the effects of Ru360 in reducing inflammatory cytokines and intracytoplasmic lipid content. CONCLUSIONS In this study, we found that MCU inhibition modulated intracytoplasmic Ca2+ concentration, improved impaired Mø efferocytosis, and reduced ROS generation. Macrophage efferocytosis removed apoptotic cells and prevented the release of inflammatory factor and foam cell formation, and this can be a potential new therapeutic target for alleviating atherosclerosis.
Collapse
Affiliation(s)
- Na Lu
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.
| | - Jun-Fan Zhu
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - He-Fan Lv
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Hai-Peng Zhang
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Peng-le Wang
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jing-Jing Yang
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Xian-Wei Wang
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
23
|
Zhao S, Wang Q, Liu Y, Zhang P, Ji W, Xie J, Cheng C. Interaction, immune infiltration characteristics and prognostic modeling of efferocytosis-related subtypes in glioblastoma. BMC Med Genomics 2023; 16:248. [PMID: 37853449 PMCID: PMC10583324 DOI: 10.1186/s12920-023-01688-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 10/04/2023] [Indexed: 10/20/2023] Open
Abstract
BACKGROUND Efferocytosis is a biological process in which phagocytes remove apoptotic cells and vesicles from tissues. This process is initiated by the release of inflammatory mediators from apoptotic cells and plays a crucial role in resolving inflammation. The signals associated with efferocytosis have been found to regulate the inflammatory response and the tumor microenvironment (TME), which promotes the immune escape of tumor cells. However, the role of efferocytosis in glioblastoma multiforme (GBM) is not well understood and requires further investigation. METHODS In this study, we conducted a comprehensive analysis of 22 efferocytosis-related genes (ERGs) by searching for studies related to efferocytosis. Using bulk RNA-Seq and single-cell sequencing data, we analyzed the expression and mutational characteristics of these ERGs. By using an unsupervised clustering algorithm, we obtained ERG clusters from 549 GBM patients and evaluated the immune infiltration characteristics of each cluster. We then identified differential genes (DEGs) in the two ERG clusters and classified GBM patients into different gene clusters using univariate cox analysis and unsupervised clustering algorithms. Finally, we utilized the Boruta algorithm to screen for prognostic genes and reduce dimensionality, and the PCA algorithm was applied to create a novel efferocytosis-related scoring system. RESULTS Differential expression of ERGs in glioma cell lines and normal cells was analyzed by rt-PCR. Cell function experiments, on the other hand, validated TIMD4 as a tumor risk factor in GBM. We found that different ERG clusters and gene clusters have distinct prognostic and immune infiltration profiles. The ERG signature we developed provides insight into the tumor microenvironment of GBM. Patients with lower ERG scores have a better survival rate and a higher likelihood of benefiting from immunotherapy. CONCLUSIONS Our novel efferocytosis-related signature has the potential to be used in clinical practice for risk stratification of GBM patients and for selecting individuals who are likely to respond to immunotherapy. This can help clinicians design appropriate targeted therapies before initiating clinical treatment.
Collapse
Affiliation(s)
- Songyun Zhao
- Department of Neurosurgery, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Qi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yuankun Liu
- Department of Neurosurgery, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Pengpeng Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Ji
- Department of Neurosurgery, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Jiaheng Xie
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, China.
| | - Chao Cheng
- Department of Neurosurgery, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China.
| |
Collapse
|
24
|
Zheng W, Zhou Z, Guo X, Zuo X, Zhang J, An Y, Zheng H, Yue Y, Wang G, Wang F. Efferocytosis and Respiratory Disease. Int J Mol Sci 2023; 24:14871. [PMID: 37834319 PMCID: PMC10573909 DOI: 10.3390/ijms241914871] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
Cells are the smallest units that make up living organisms, which constantly undergo the processes of proliferation, differentiation, senescence and death. Dead cells need to be removed in time to maintain the homeostasis of the organism and keep it healthy. This process is called efferocytosis. If the process fails, this may cause different types of diseases. More and more evidence suggests that a faulty efferocytosis process is closely related to the pathological processes of respiratory diseases. In this review, we will first introduce the process and the related mechanisms of efferocytosis of the macrophage. Secondly, we will propose some methods that can regulate the function of efferocytosis at different stages of the process. Next, we will discuss the role of efferocytosis in different lung diseases and the related treatment approaches. Finally, we will summarize the drugs that have been applied in clinical practice that can act upon efferocytosis, in order to provide new ideas for the treatment of lung diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Guoqiang Wang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (W.Z.); (Z.Z.); (X.G.); (X.Z.); (J.Z.); (Y.A.); (H.Z.); (Y.Y.)
| | - Fang Wang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (W.Z.); (Z.Z.); (X.G.); (X.Z.); (J.Z.); (Y.A.); (H.Z.); (Y.Y.)
| |
Collapse
|
25
|
Rochette L, Dogon G, Rigal E, Zeller M, Cottin Y, Vergely C. Interplay between efferocytosis and atherosclerosis. Arch Cardiovasc Dis 2023; 116:474-484. [PMID: 37659915 DOI: 10.1016/j.acvd.2023.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/26/2023] [Accepted: 07/31/2023] [Indexed: 09/04/2023]
Abstract
In an adult human, billions of cells die and turn over daily. During this process, many apoptotic cells are produced and subsequently cleared by phagocytes - a process termed efferocytosis, which plays a critical role in tissue homeostasis. Efferocytosis is an important mechanism in the control of inflammatory processes. Efficient efferocytosis inhibits accumulation of apoptotic cells/debris and maintains homeostasis before the onset of necrosis (secondary necrosis), which promotes inflammation or injury. During efferocytosis, mitochondrial fission and the oxidative stress process are linked through reactive oxygen species production and oxidative stress control. Autophagy plays an important role in inhibiting inflammation and apoptosis, and in promoting efferocytosis by activated inflammatory cells, particularly neutrophils and macrophages. Autophagy in neutrophils is activated by phagocytosis of pathogens or activation of pattern recognition receptors. Autophagy is essential for major neutrophil functions, including degranulation, reactive oxygen species production, oxidative stress and release of neutrophil extracellular cytokines. Failed efferocytosis is a key mechanism driving the development and progression of chronic inflammatory diseases, including atherosclerosis, cardiometabolic pathology, neurodegenerative disease and cancer. Impairment of efferocytosis in apoptotic macrophages is a determinant of atherosclerosis severity and the vulnerability of plaques to rupture. Recent results suggest that inhibition of efferocytosis in the protection of the myocardium results in reduced infiltration of reparatory macrophages into the tissue, in association with oxidative stress reduction. Activated macrophages play a central role in the development and resolution of inflammation. The resolution of inflammation through efferocytosis is an endogenous process that protects host tissues from prolonged or excessive inflammation. Accordingly, therapeutic strategies that ameliorate efferocytosis control would be predicted to dampen inflammation and improve resolution. Thus, therapies targeting efferocytosis will provide a new means of treating and preventing cardiovascular and metabolic diseases involving the chronic inflammatory state.
Collapse
Affiliation(s)
- Luc Rochette
- Équipe d'accueil (EA 7460) : physiopathologie et épidémiologie cérébro-cardiovasculaires (PEC2), faculté des sciences de santé, université de Bourgogne-Franche-Comté, 7, boulevard Jeanne-d'Arc, 21000 Dijon, France.
| | - Geoffrey Dogon
- Équipe d'accueil (EA 7460) : physiopathologie et épidémiologie cérébro-cardiovasculaires (PEC2), faculté des sciences de santé, université de Bourgogne-Franche-Comté, 7, boulevard Jeanne-d'Arc, 21000 Dijon, France
| | - Eve Rigal
- Équipe d'accueil (EA 7460) : physiopathologie et épidémiologie cérébro-cardiovasculaires (PEC2), faculté des sciences de santé, université de Bourgogne-Franche-Comté, 7, boulevard Jeanne-d'Arc, 21000 Dijon, France
| | - Marianne Zeller
- Équipe d'accueil (EA 7460) : physiopathologie et épidémiologie cérébro-cardiovasculaires (PEC2), faculté des sciences de santé, université de Bourgogne-Franche-Comté, 7, boulevard Jeanne-d'Arc, 21000 Dijon, France
| | - Yves Cottin
- Service de cardiologie, CHU de Dijon, 21000 Dijon, France
| | - Catherine Vergely
- Équipe d'accueil (EA 7460) : physiopathologie et épidémiologie cérébro-cardiovasculaires (PEC2), faculté des sciences de santé, université de Bourgogne-Franche-Comté, 7, boulevard Jeanne-d'Arc, 21000 Dijon, France
| |
Collapse
|
26
|
Shook PL, Singh M, Singh K. Macrophages in the Inflammatory Phase following Myocardial Infarction: Role of Exogenous Ubiquitin. BIOLOGY 2023; 12:1258. [PMID: 37759657 PMCID: PMC10526096 DOI: 10.3390/biology12091258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/19/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023]
Abstract
Cardiovascular disease (CVD) is one of the leading causes of death worldwide. One of the most common implications of CVD is myocardial infarction (MI). Following MI, the repair of the infarcted heart occurs through three distinct, yet overlapping phases of inflammation, proliferation, and maturation. Macrophages are essential to the resolution of the inflammatory phase due to their role in phagocytosis and efferocytosis. However, excessive and long-term macrophage accumulation at the area of injury and dysregulated function can induce adverse cardiac remodeling post-MI. Ubiquitin (UB) is a highly evolutionarily conserved small protein and is a normal constituent of plasma. Levels of UB are increased in the plasma during a variety of pathological conditions, including ischemic heart disease. Treatment of mice with UB associates with decreased inflammatory response and improved heart function following ischemia/reperfusion injury. This review summarizes the role of macrophages in the infarct healing process of the heart post-MI, and discusses the role of exogenous UB in myocardial remodeling post-MI and in the modulation of macrophage phenotype and function.
Collapse
Affiliation(s)
- Paige L. Shook
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (P.L.S.); (M.S.)
| | - Mahipal Singh
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (P.L.S.); (M.S.)
| | - Krishna Singh
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (P.L.S.); (M.S.)
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- James H. Quillen Veterans Affairs Medical Center, Mountain Home, TN 37684, USA
| |
Collapse
|
27
|
Zheng H, Cheng X, Jin L, Shan S, Yang J, Zhou J. Recent advances in strategies to target the behavior of macrophages in wound healing. Biomed Pharmacother 2023; 165:115199. [PMID: 37517288 DOI: 10.1016/j.biopha.2023.115199] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/05/2023] [Accepted: 07/18/2023] [Indexed: 08/01/2023] Open
Abstract
Chronic wounds and scar formation are widespread due to limited suitable remedies. The macrophage is a crucial regulator in wound healing, controlling the onset and termination of inflammation and regulating other processes related to wound healing. The current breakthroughs in developing new medications and drug delivery methods have enabled the accurate targeting of macrophages in oncology and rheumatic disease therapies through clinical trials. These successes have cleared the way to utilize drugs targeting macrophages in various disorders. This review thus summarizes macrophage involvement in normal and pathologic wound healing. It further details the targets available for macrophage intervention and therapeutic strategies for targeting the behavior of macrophages in tissue repair and regeneration.
Collapse
Affiliation(s)
- Hongkun Zheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Xinwei Cheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Lu Jin
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Shengzhou Shan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Jun Yang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| | - Jia Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| |
Collapse
|
28
|
Lin YC, Tsai WH, Chang SC, Hsu HC. Apoptotic Cell-Derived CD14(+) Microparticles Promote the Phagocytic Activity of Neutrophilic Precursor Cells in the Phagocytosis of Apoptotic Cells. Cells 2023; 12:1983. [PMID: 37566062 PMCID: PMC10417108 DOI: 10.3390/cells12151983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/12/2023] Open
Abstract
Membranous CD14 is crucial in the phagocytic activity of neutrophils. However, the role of CD14(+) microparticles (MPs) derived from apoptotic neutrophils (apo-MP) during the phagocytic process is not clear. All trans-retinoic acid (ATRA) induces acute promyelocytic leukemic NB4 cells along granulocytic differentiation. In this study, we investigated the role of CD14(+)apo-MP in the cell-cell interaction during the phagocytic process of apoptotic cells by viable ATRA-NB4 cells. We firstly demonstrate that CD14 expression and phagocytic activity of NB4 cells were upregulated simultaneously after ATRA treatment in a time-dependent manner, and both were significantly enhanced via concurrent lipopolysaccharide treatment. The phagocytic activity of ATRA-NB4 cells and lipopolysaccharide-treated ATRA-NB4 cells were both significantly attenuated by pre-treating cells with an antibody specific to either CD14 or TLR4. Further flow cytometric analysis demonstrates that apoptotic ATRA-NB4 cells release CD14(+)apo-MP in an idarubicin dosage-dependent manner. Both CD14 expression and the phagocytic activity of viable ATRA-NB4 cells were significantly enhanced after incubation with apo-MP harvested from apoptotic ATRA-NB4 cells, and the apo-MP-enhanced phagocytic activity was significantly attenuated by pre-treating apo-MP with an anti-CD14 antibody before incubation with viable cells. We conclude that CD14(+)apo-MP derived from apoptotic ATRA-NB4 cells promotes the phagocytic activity of viable ATRA-NB4 cells in engulfing apoptotic cells.
Collapse
Affiliation(s)
- Yu-Chieh Lin
- Department of Physiology, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei 112, Taiwan; (Y.-C.L.)
- Sleep Medicine Center, Division of Chest Medicine, Taichung Tzu Chi Hospital, Taichung 427, Taiwan
| | - Wen-Hui Tsai
- Department of Respiratory Therapy, Taipei Medical University, Taipei 106, Taiwan;
| | - Shao-Chi Chang
- Department of Physiology, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei 112, Taiwan; (Y.-C.L.)
| | - Hui-Chi Hsu
- Department of Medicine, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei 112, Taiwan
- Division of General Medicine, Department of Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Division of Hematology & Oncology, Department of Medicine, Cheng-Hsin General Hospital, Taipei 112, Taiwan
| |
Collapse
|
29
|
Serna MF, Mosquera Escudero M, García-Perdomo HA. Lipoxins and their relationship with inflammation-associated diseases. A systematic review. Obes Res Clin Pract 2023; 17:298-307. [PMID: 37316341 DOI: 10.1016/j.orcp.2023.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 05/23/2023] [Accepted: 06/06/2023] [Indexed: 06/16/2023]
Abstract
AIM To determine the relationship of lipoxin levels with inflammation and disease development in adults and children. METHODS We conducted a systematic review. The search strategy included Medline, Ovid, EMBASE, LILACS, The Cochrane Central Register of Controlled Trials, and Open Gray. We included Clinical trials, cohort studies, case-control studies, and cross-sectional studies. Animal experiments were excluded. RESULTS We included fourteen studies in this review, nine consistently showing decreased lipoxin levels and anti-inflammatory markers or increased pro-inflammatory markers in cardiovascular disease, metabolic syndrome, Alzheimer's disease, periodontitis, or autism. Five studies showed increased lipoxin levels and pro-inflammatory markers in pre-eclampsia, asthma, and coronary disease. On the other hand, one showed increased lipoxin levels and decreased pro-inflammatory marker levels. CONCLUSIONS Decreases in lipoxins are associated with developing pathologies such as cardiovascular and neurological diseases, indicating that lipoxins protect against these pathologies. However, in other pathologies, such as asthma, pre-eclampsia, and periodontitis, which are associated with chronic inflammation despite increased levels of LXA4, the increase in inflammation suggests a possible failure of this regulatory pathway. Therefore, further studies are necessary to evaluate the role of LXA4 in the pathogenesis of inflammatory diseases.
Collapse
Affiliation(s)
- Maria Fernanda Serna
- Nutrition Group. School of Basic Sciences. Universidad del Valle, Cali, Colombia
| | | | - Herney Andrés García-Perdomo
- Nutrition Group. School of Basic Sciences. Universidad del Valle, Cali, Colombia; Division of Urology/Urooncology. Department of Surgery. School of Medicine. Universidad del Valle, Cali, Colombia.
| |
Collapse
|
30
|
She Y, Xu X, Yu Q, Yang X, He J, Tang XX. Elevated expression of macrophage MERTK exhibits profibrotic effects and results in defective regulation of efferocytosis function in pulmonary fibrosis. Respir Res 2023; 24:118. [PMID: 37120511 PMCID: PMC10148433 DOI: 10.1186/s12931-023-02424-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 03/08/2023] [Indexed: 05/01/2023] Open
Abstract
Increased apoptosis of alveolar epithelial cells is a prominent feature of pulmonary fibrosis. Macrophage efferocytosis, phagocytosis of apoptotic cells by macrophages, is crucial for maintaining tissue homeostasis. Expression of Mer tyrosine kinase (MERTK, an important recognition receptor in efferocytosis) in macrophages is thought to be associated with fibrosis. However, how macrophage MERTK affects pulmonary fibrosis and whether it depends on efferocytosis are not yet clear. Here, we found elevated MERTK expression in lung macrophages from IPF patients and mice with bleomycin-induced pulmonary fibrosis. In vitro experiments showed that macrophages overexpressing MERTK exhibit profibrotic effects and that macrophage efferocytosis abrogates the profibrotic effect of MERTK by downregulating MERTK, forming a negative regulatory loop. In pulmonary fibrosis, this negative regulation is defective, and MERTK mainly exhibits profibrotic effects. Our study reveals a previously unsuspected profibrotic effect of elevated macrophage MERTK in pulmonary fibrosis and defective regulation of efferocytosis function as a result of that elevation, suggesting that targeting MERTK in macrophages may help to attenuate pulmonary fibrosis.
Collapse
Affiliation(s)
- Yixin She
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xin Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qingyang Yu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiangsheng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianxing He
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Xiao Xiao Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou Laboratory, Bio-Island, Guangzhou, China.
| |
Collapse
|
31
|
Zhuang WR, Wang Y, Nie W, Lei Y, Liang C, He J, Zuo L, Huang LL, Xie HY. Bacterial outer membrane vesicle based versatile nanosystem boosts the efferocytosis blockade triggered tumor-specific immunity. Nat Commun 2023; 14:1675. [PMID: 36966130 PMCID: PMC10039929 DOI: 10.1038/s41467-023-37369-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 03/14/2023] [Indexed: 03/27/2023] Open
Abstract
Efferocytosis inhibition is emerging as an attractive strategy for antitumor immune therapy because of the subsequent leak of abundant immunogenic contents. However, the practical efficacy is seriously impeded by the immunosuppressive tumor microenvironments. Here, we construct a versatile nanosystem that can not only inhibit the efferocytosis but also boost the following antitumor immunity. MerTK inhibitor UNC2025 is loaded into the bacterial outer membrane vesicles (OMVs), which are then modified with maleimide (mU@OMVs). The prepared mU@OMVs effectively inhibits the efferocytosis by promoting the uptake while preventing the MerTK phosphorylation of tumor associated macrophages, and then captures the released antigens through forming universal thioether bonds. The obtained in situ vaccine effectively transfers to lymph nodes by virtue of the intrinsic features of OMVs, and then provokes intense immune responses that can efficiently prevent the growth, metastasis and recurrence of tumors in mice, providing a generalizable strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Wan-Ru Zhuang
- School of Life Science, Beijing Institute of Technology, 100081, Beijing, P.R. China
| | - Yunfeng Wang
- School of Life Science, Beijing Institute of Technology, 100081, Beijing, P.R. China
| | - Weidong Nie
- School of Life Science, Beijing Institute of Technology, 100081, Beijing, P.R. China
| | - Yao Lei
- School of Life Science, Beijing Institute of Technology, 100081, Beijing, P.R. China
| | - Chao Liang
- School of Life Science, Beijing Institute of Technology, 100081, Beijing, P.R. China
| | - Jiaqi He
- School of Life Science, Beijing Institute of Technology, 100081, Beijing, P.R. China
| | - Liping Zuo
- School of Life Science, Beijing Institute of Technology, 100081, Beijing, P.R. China
| | - Li-Li Huang
- School of Medical Technology, Beijing Institute of Technology, 100081, Beijing, P.R. China
| | - Hai-Yan Xie
- School of Life Science, Beijing Institute of Technology, 100081, Beijing, P.R. China.
| |
Collapse
|
32
|
Dilara Fatma Akin, Özkan D. Molecular profiling of TAM tyrosine kinase receptors and ligands in endometrial carcinoma: An in silico-study. Taiwan J Obstet Gynecol 2023; 62:311-324. [PMID: 36965901 DOI: 10.1016/j.tjog.2022.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2022] [Indexed: 03/27/2023] Open
Abstract
OBJECTIVES TAM Receptors (TYRO3, AXL, and MerTK) and their ligands on tumor-associated macrophages are promising therapeutic targets for most solid cancers. However, in endometrial cancer, the most common invasive gynecologic malignancy, the TAM receptor-mediated activation pathway, its molecular mechanisms, and its pathophysiology are unknown. The goal of this research; to uncover the comprehensive genetic profile of TAM receptors and ligands in endometrial cancer. MATERIAL AND METHODS Mutation and expression profiles of the Uterine Corpus Endometrial Carcinoma (UCEC) cohort (n = 509) were obtained using bioinformatics tools providing data from The Cancer Genome Atlas (TCGA). PolyPhen-2 and SNAP tools were used to predict the oncogenic/pathogenic properties of the identified mutations for UCEC. STRING network analysis was performed to better understand the functional relationships of the mutant proteins in cellular processes. Furthermore to the mutation profile, gene expression and survival profiles were also determined. Finally, the correlation between target genes and macrophage infiltration was investigated using the tool TIMER. RESULTS A total of 229 mutations were detected in 6 genes, and 81 missense mutations are pathogenic. In the UCEC cohort, the expression level of MerTK, AXL, GAS6, and PROS1 was statistically significantly lower in the patient group, while the expression level of CD47 was higher in the patient group than in the healthy group (p < 0.01). Protein-protein interaction analysis identified target genes, SRC protein responsible for important cellular mechanisms such as cell proliferation, adhesion and migration, ITGB3, ITGAV and THSB1 proteins involved in endothelial mesenchymal transition and tumor metabolism reprogramming, and FOLR1 involved in DNA replication and damage repair. CONCLUSION We believe that TAM receptors and their ligands may be attractive molecular targets for the treatment of endometrial carcinoma because they act as pleiotropic inhibitors of immune cells, effectively regulate phagocytic clearance of apoptotic cells, and make the tumor microenvironment a more suitable niche for the tumour.
Collapse
Affiliation(s)
- Dilara Fatma Akin
- Nigde Ömer Halisdemir University, Faculty of Medicine, Medical Biology, Nigde, Turkey.
| | - Didem Özkan
- Istanbul Okan University, Vocational School of Health Service, Istanbul, Turkey
| |
Collapse
|
33
|
Baraniecki Ł, Tokarz-Deptuła B, Syrenicz A, Deptuła W. Macrophage efferocytosis in atherosclerosis. Scand J Immunol 2022; 97:e13251. [PMID: 36583598 DOI: 10.1111/sji.13251] [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: 08/23/2022] [Revised: 11/17/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022]
Abstract
This paper presents the role of macrophage efferocytosis, the process of elimination of apoptotic bodies-elements formed during vascular atherosclerosis. The mechanisms of macrophage efferocytosis are presented, introducing the specific signals of this process, that is, 'find me', 'eat me' and 'don't eat me'. The role of the process of efferocytosis in the formation of vascular atherosclerosis is also presented, including the factors and mechanisms that determine it, as well as the factors that determine the maintenance of homeostasis in the vessels, including the formation of vascular atherosclerosis.
Collapse
Affiliation(s)
| | | | - Anhelli Syrenicz
- Department of Endocrinology, Metabolic Diseases and Internal Diseases, Pomeranian Medical University, Szczecin, Poland
| | - Wiesław Deptuła
- Faculty of Biological and Veterinary Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Toruń, Toruń, Poland
| |
Collapse
|
34
|
Shi H, Duan J, Wang J, Li H, Wu Z, Wang S, Wu X, Lu M. 1,25(OH) 2D 3 Promotes Macrophage Efferocytosis Partly by Upregulating ASAP2 Transcription via the VDR-Bound Enhancer Region and ASAP2 May Affect Antiviral Immunity. Nutrients 2022; 14:4935. [PMID: 36432619 PMCID: PMC9699620 DOI: 10.3390/nu14224935] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/11/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
The active form of vitamin D3, i.e., 1,25(OH)2D3, exerts an anti-inflammatory effect on the immune system, especially macrophage-mediated innate immunity. In a previous study, we identified 1,25(OH)2D3-responsive and vitamin D receptor (VDR)-bound super-enhancer regions in THP-1 cells. Herein, we examined the transcriptional regulation of ArfGAP with SH3 Domain, Ankyrin Repeat and PH Domain 2 (ASAP2) (encoding a GTPase-activating protein) by 1,25(OH)2D3 through the top-ranked VDR-bound super-enhancer region in the first intron of ASAP2 and potential functions of ASAP2 in macrophages. First, we validated the upregulation of ASAP2 by 1,25(OH)2D3 in both THP-1 cells and macrophages. Subsequently, we identified three regulatory regions (i.e., the core, 1,25(OH)2D3-responsive, and inhibitory regions) in the VDR bound-enhancer of ASAP2. ASAP2 promoted RAC1-activity and macrophage efferocytosis in vitro. Next, we assessed the functions of ASAP2 by mass spectrometry and RNA sequencing analyses. ASAP2 upregulated the expressions of antiviral-associated genes and interacted with SAM and HD domain-containing deoxynucleoside triphosphate triphosphohydrolase 1 (SAMHD1). In vivo, vitamin D reduced the number of apoptotic cells in experimental autoimmune encephalomyelitis (EAE) and promoted macrophage efferocytosis in peritonitis without changing the mRNA level of ASAP2. Thus, we could better understand the regulatory mechanism underlying ASAP2 transcription and the function of ASAP2, which may serve as a potential treatment target against inflammatory diseases and virus infections.
Collapse
Affiliation(s)
- Hui Shi
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Jiangling Duan
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Jiayu Wang
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Haohao Li
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Zhiheng Wu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Shuaideng Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Xueyan Wu
- Department of Human Anatomy, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Ming Lu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
35
|
Tajbakhsh A, Gheibihayat SM, Askari H, Savardashtaki A, Pirro M, Johnston TP, Sahebkar A. Statin-regulated phagocytosis and efferocytosis in physiological and pathological conditions. Pharmacol Ther 2022; 238:108282. [DOI: 10.1016/j.pharmthera.2022.108282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/10/2022] [Accepted: 09/13/2022] [Indexed: 10/14/2022]
|
36
|
Ji H, Zhang C, Xu F, Mao Q, Xia R, Chen M, Wang W, Lv S, Li W, Shi X. Inhaled Pro-Efferocytic Nanozymes Promote Resolution of Acute Lung Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201696. [PMID: 35859230 PMCID: PMC9475552 DOI: 10.1002/advs.202201696] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/06/2022] [Indexed: 06/01/2023]
Abstract
Acute lung injury (ALI) is a significant contributor to the morbidity and mortality of sepsis. Characterized by uncontrolled inflammation and excessive inflammatory cells infiltration in lung, ALI has been exacerbated by impaired efferocytosis (clearance of apoptotic cells by macrophages). Through specific receptor recognition and activation of downstream signaling, efferocytic macrophages promote resolution of inflammation by efficiently engulfing dying cells, avoiding the consequent release of cellular inflammatory contents. Here, inspired by the intrinsic recovery mechanism of efferocytosis, an apoptotic cell membrane (ACM) coated antioxidant nanozyme (AOzyme) is engineered, thus obtaining an inhalable pro-efferocytic nanozyme (AOzyme@ACM). Notably, AOzyme@ACM can efficiently increase apoptotic cell removal by combing enhanced macrophages recognition of "eat me" signals through apoptotic body mimicking and scavenge of intracellular excessive reactive oxygen species (ROS), a significant barrier for efferocytosis. AOzyme@ACM can significantly inhibit inflammatory response, promote pro-resolving (M2) phenotype transition of macrophage, and alleviate ALI in endotoxemia mice compared with AOzyme group. By addressing the critical factor in the pathogenesis of sepsis-related ALI through restoring efferocytosis activity, the ACM-based antioxidant nanozyme in this study is envisioned to provide a promising strategy to treat this complex and challenging disease.
Collapse
Affiliation(s)
- Haiying Ji
- Department of Anesthesiology and Intensive Care UnitXinhua HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200092China
| | - Chengmi Zhang
- Department of Anesthesiology and Intensive Care UnitXinhua HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200092China
| | - Fengying Xu
- Department of AnesthesiologyNo. 971 Hospital of People's Liberation Army NavyQingdao266000China
| | - Qianyun Mao
- Department of EndocrinologyXinhua HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200092China
| | - Ran Xia
- Department of Anesthesiology and Intensive Care UnitXinhua HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200092China
| | - Muqiao Chen
- Department of Anesthesiology and Intensive Care UnitXinhua HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200092China
| | - Wei Wang
- Department of Anesthesiology and Intensive Care UnitXinhua HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200092China
| | - Shunan Lv
- Department of Anesthesiology and Intensive Care UnitXinhua HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200092China
| | - Weiwei Li
- Department of Anesthesiology and Intensive Care UnitXinhua HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200092China
| | - Xueyin Shi
- Department of Anesthesiology and Intensive Care UnitXinhua HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200092China
| |
Collapse
|
37
|
Methodologies and tools to shed light on erythrophagocytosis. Biochimie 2022; 202:166-179. [PMID: 35952949 DOI: 10.1016/j.biochi.2022.07.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 11/20/2022]
Abstract
Red blood cells (RBC) are the most abundant circulating cell of the human body. RBC are constantly exposed to multiple stresses in the circulation, leading to molecular and structural impairments and death. The physiological process of RBC senescence or ageing is referred to as eryptosis. At the end of their lifespan, aged RBC are recognized and removed from the blood by professional phagocytes via a phenomenon called erythrophagocytosis (EP); the phagocytosis of RBC. Some genetic and acquired diseases can influence eryptosis, thereby affecting RBC lifespan and leading to hemolytic anemia. In some diseases, such as diabetes and atherosclerosis, eryptosis and EP can participate in disease progression with both professional and non-professional phagocytes. Therefore, investigating the process of EP in vivo and in vitro, as well as in different cell types, will not only contribute to the understanding of the physiological steps of EP, but also to the deciphering of the specific mechanisms involving RBC and EP that underlie certain pathologies. In this review, the process of EP is introduced and the different methods for studying EP are discussed together with examples of the experimental procedures and materials required.
Collapse
|
38
|
Cellular Carcinogenesis: Role of Polarized Macrophages in Cancer Initiation. Cancers (Basel) 2022; 14:cancers14112811. [PMID: 35681791 PMCID: PMC9179569 DOI: 10.3390/cancers14112811] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/26/2022] [Accepted: 06/02/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Inflammation is a hallmark of many cancers. Macrophages are key participants in innate immunity and important drivers of inflammation. When chronically polarized beyond normal homeostatic responses to infection, injury, or aging, macrophages can express several pro-carcinogenic phenotypes. In this review, evidence supporting polarized macrophages as endogenous sources of carcinogenesis is discussed. In addition, the depletion or modulation of macrophages by small molecule inhibitors and probiotics are reviewed as emerging strategies in cancer prevention. Abstract Inflammation is an essential hallmark of cancer. Macrophages are key innate immune effector cells in chronic inflammation, parainflammation, and inflammaging. Parainflammation is a form of subclinical inflammation associated with a persistent DNA damage response. Inflammaging represents low-grade inflammation due to the dysregulation of innate and adaptive immune responses that occur with aging. Whether induced by infection, injury, or aging, immune dysregulation and chronic macrophage polarization contributes to cancer initiation through the production of proinflammatory chemokines/cytokines and genotoxins and by modulating immune surveillance. This review presents pre-clinical and clinical evidence for polarized macrophages as endogenous cellular carcinogens in the context of chronic inflammation, parainflammation, and inflammaging. Emerging strategies for cancer prevention, including small molecule inhibitors and probiotic approaches, that target macrophage function and phenotype are also discussed.
Collapse
|
39
|
Trybus E, Król T, Trybus W. The Multidirectional Effect of Azelastine Hydrochloride on Cervical Cancer Cells. Int J Mol Sci 2022; 23:5890. [PMID: 35682572 PMCID: PMC9180047 DOI: 10.3390/ijms23115890] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/22/2022] [Accepted: 05/23/2022] [Indexed: 02/01/2023] Open
Abstract
A major cause of cancer cell resistance to chemotherapeutics is the blocking of apoptosis and induction of autophagy in the context of cell adaptation and survival. Therefore, new compounds are being sought, also among drugs that are commonly used in other therapies. Due to the involvement of histamine in the regulation of processes occurring during the development of many types of cancer, antihistamines are now receiving special attention. Our study concerned the identification of new mechanisms of action of azelastine hydrochloride, used in antiallergic treatment. The study was performed on HeLa cells treated with different concentrations of azelastine (15-90 µM). Cell cycle, level of autophagy (LC3 protein activity) and apoptosis (annexin V assay), activity of caspase 3/7, anti-apoptotic protein of Bcl-2 family, ROS concentration, measurement of mitochondrial membrane potential (Δψm), and level of phosphorylated H2A.X in response to DSB were evaluated by cytometric method. Cellular changes were also demonstrated at the level of transmission electron microscopy and optical and fluorescence microscopy. Lysosomal enzyme activities-cathepsin D and L and cell viability (MTT assay) were assessed spectrophotometrically. Results: Azelastine in concentrations of 15-25 µM induced degradation processes, vacuolization, increase in cathepsin D and L activity, and LC3 protein activation. By increasing ROS, it also caused DNA damage and blocked cells in the S phase of the cell cycle. At the concentrations of 45-90 µM, azelastine clearly promoted apoptosis by activation of caspase 3/7 and inactivation of Bcl-2 protein. Fragmentation of cell nucleus was confirmed by DAPI staining. Changes were also found in the endoplasmic reticulum and mitochondria, whose damage was confirmed by staining with rhodamine 123 and in the MTT test. Azelastine decreased the mitotic index and induced mitotic catastrophe. Studies demonstrated the multidirectional effects of azelastine on HeLa cells, including anti-proliferative, cytotoxic, autophagic, and apoptotic properties, which were the predominant mechanism of death. The revealed novel properties of azelastine may be practically used in anti-cancer therapy in the future.
Collapse
Affiliation(s)
- Ewa Trybus
- Department of Medical Biology, The Jan Kochanowski University, Uniwersytecka 7, 25-406 Kielce, Poland;
| | - Teodora Król
- Department of Medical Biology, The Jan Kochanowski University, Uniwersytecka 7, 25-406 Kielce, Poland;
| | | |
Collapse
|
40
|
Tajbakhsh A, Yousefi F, Abedi SM, Rezaee M, Savardashtaki A, Teng Y, Sahebkar A. The cross-talk between soluble "Find me" and "Keep out" signals as an initial step in regulating efferocytosis. J Cell Physiol 2022; 237:3113-3126. [PMID: 35578547 DOI: 10.1002/jcp.30770] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/16/2022] [Accepted: 04/21/2022] [Indexed: 12/20/2022]
Abstract
The rapid clearance of apoptotic cells (ACs), known as efferocytosis, prompts the inhibition of inflammatory responses and autoimmunity and maintains homeostatic cell turnover by controlling the release of intracellular contents. The fast clearance of ACs requires professional and nonprofessional phagocytic cells that can accurately and promptly recognize ACs and migrate towards them. Cells undergoing apoptosis alarm their presence by releasing special soluble chemotactic factors, such as lactoferrin, that act as "Find me," "Keep out," or "Stay away" signals to recruit phagocytic cells, such as macrophages or prevent granulocyte migration. Efferocytosis effectively serves to prevent damage-associated molecular pattern release and secondary necrosis and inhibit inflammation/autoimmunity at the very first step. Since less attention has been given to the cross-talk and balance of "Find me" and "Keep out" signals released from ACs in efferocytosis, we set out to investigate the current knowledge of the roles of "Find me" and "Keep out" signals in the efferocytosis process.
Collapse
Affiliation(s)
- Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Yousefi
- Department of Biological Sciences, Faculty of Genetics, Tarbiat Modares University, Tehran, Iran
| | - Seyedeh M Abedi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Rezaee
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Amir Savardashtaki
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yong Teng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Medicine, The University of Western Australia, Perth, Western Australia, Australia.,Depatment of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
41
|
Geng R, Lin Y, Ji M, Chang Q, Li Z, Xu L, Zhang W, Lu J. MFG-E8 promotes tendon-bone healing by regualting macrophage efferocytosis and M2 polarization after anterior cruciate ligament reconstruction. J Orthop Translat 2022; 34:11-21. [PMID: 35615640 PMCID: PMC9109120 DOI: 10.1016/j.jot.2022.04.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/18/2022] [Accepted: 04/15/2022] [Indexed: 11/24/2022] Open
|
42
|
Su W, Liang L, Zhou L, Cao Y, Zhou X, Liu S, Wang Q, Zhang H. Macrophage Paired Immunoglobulin-Like Receptor B Deficiency Promotes Peripheral Atherosclerosis in Apolipoprotein E–Deficient Mice. Front Cell Dev Biol 2022; 9:783954. [PMID: 35321392 PMCID: PMC8936951 DOI: 10.3389/fcell.2021.783954] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/13/2021] [Indexed: 01/08/2023] Open
Abstract
Background: Peripheral atherosclerotic disease (PAD) is the narrowing or blockage of arteries that supply blood to the lower limbs. Given its complex nature, bioinformatics can help identify crucial genes involved in the progression of peripheral atherosclerosis. Materials and Methods: Raw human gene expression data for 462 PAD arterial plaque and 23 normal arterial samples were obtained from the GEO database. The data was analyzed using an integrated, multi-layer approach involving differentially-expressed gene analysis, KEGG pathway analysis, GO term enrichment analysis, weighted gene correlation network analysis, and protein-protein interaction analysis. The monocyte/macrophage-expressed leukocyte immunoglobulin-like receptor B2 (LILRB2) was strongly associated with the human PAD phenotype. To explore the role of the murine LILRB2 homologue PirB in vivo, we created a myeloid-specific PirB-knockout Apoe−/− murine model of PAD (PirBMΦKO) to analyze femoral atherosclerotic burden, plaque features of vulnerability, and monocyte recruitment to femoral atherosclerotic lesions. The phenotypes of PirBMΦKO macrophages under various stimuli were also investigated in vitro. Results:PirBMΦKO mice displayed increased femoral atherogenesis, a more vulnerable plaque phenotype, and enhanced monocyte recruitment into lesions. PirBMΦKO macrophages showed enhanced pro-inflammatory responses and a shift toward M1 over M2 polarization under interferon-γ and oxidized LDL exposure. PirBMΦKO macrophages also displayed enhanced efferocytosis and reduced lipid efflux under lipid exposure. Conclusion: Macrophage PirB reduces peripheral atherosclerotic burden, stabilizes peripheral plaque composition, and suppresses macrophage accumulation in peripheral lesions. Macrophage PirB inhibits pro-inflammatory activation, inhibits efferocytosis, and promotes lipid efflux, characteristics critical to suppressing peripheral atherogenesis.
Collapse
Affiliation(s)
- Wenhua Su
- Department of Cardiology, First People’s Hospital of Yunnan Province, Kunming, China
- Faculty of Life Science and Biotechnology, Kunming University of Science and Technology, Kunming, China
| | - Liwen Liang
- Department of Cardiology, First People’s Hospital of Yunnan Province, Kunming, China
| | - Liang Zhou
- Department of Cardiology, First People’s Hospital of Yunnan Province, Kunming, China
| | - Yu Cao
- Department of Cardiology, First People’s Hospital of Yunnan Province, Kunming, China
- Department of Cardiovascular Surgery, First People’s Hospital of Yunnan Province, Kunming, China
| | - Xiuli Zhou
- Department of Cardiology, First People’s Hospital of Yunnan Province, Kunming, China
| | - Shiqi Liu
- Department of Cardiology, First People’s Hospital of Yunnan Province, Kunming, China
| | - Qian Wang
- Department of Cardiology, First People’s Hospital of Yunnan Province, Kunming, China
| | - Hong Zhang
- Department of Cardiology, First People’s Hospital of Yunnan Province, Kunming, China
- *Correspondence: Hong Zhang,
| |
Collapse
|
43
|
Pan X, Yuan S, Xun X, Fan Z, Xue X, Zhang C, Wang J, Deng J. Long-Term Recruitment of Endogenous M2 Macrophages by Platelet Lysate-Rich Plasma Macroporous Hydrogel Scaffold for Articular Cartilage Defect Repair. Adv Healthc Mater 2022; 11:e2101661. [PMID: 34969180 DOI: 10.1002/adhm.202101661] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 12/21/2021] [Indexed: 01/08/2023]
Abstract
After cartilage damage, a large number of monocytes/macrophages infiltrate into adjacent synovium and the resident macrophages in synovial tissue transform to activated macrophages (M1), which secrete pro-inflammatory cytokines to induce sustained inflammation and chondrocyte apoptotic. However, current clinical therapies for cartilage repair can rarely achieve long-term anti-inflammatory regulation and satisfactory outcomes. Herein, a platelet lysate-rich plasma macroporous hydrogel (PLPMH) scaffold with around 100 µm pore size and 1.25 MPa Young's modulus is developed to sustainedly recruit and polarize endogenous anti-inflammatory macrophages (M2) for improving cartilage defect repair. PLPMH scaffold can steadily release sphingosine1-phosphate and proteins via gradual degradation, thus inducing M2 macrophages migration or resting (M0) macrophages migration and then polarization to M2 phenotype, and improving the secretion of anti-inflammatory cytokines. Furthermore, PLPMH scaffold exhibits negligible inflammatory responses in vivo and promotes endogenous M2 macrophage infiltration in large numbers and long-time duration to provide a local anti-inflammatory microenvironment, which even lasts for 42 d. In a rabbit model of cartilage defect, PLPMH scaffold increases the ratio of M2 macrophages and improves cartilage tissue regeneration. These studies support that PLPMH scaffold may have a great potential in articular cartilage tissue engineering by providing an anti-inflammatory and pro-regenerative microenvironment.
Collapse
Affiliation(s)
- Xiaoyun Pan
- Department of Orthopaedics The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou Zhejiang 325000 China
- Key Laboratory of Orthopaedics of Zhejiang Province Wenzhou Medical University Wenzhou Zhejiang 325000 China
| | - Shanshan Yuan
- Wenzhou Institute University of Chinese Academy Sciences Wenzhou Zhejiang 325000 China
| | - Xiaojie Xun
- Wenzhou Institute University of Chinese Academy Sciences Wenzhou Zhejiang 325000 China
| | | | - Xinghe Xue
- Department of Orthopaedics The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou Zhejiang 325000 China
- Key Laboratory of Orthopaedics of Zhejiang Province Wenzhou Medical University Wenzhou Zhejiang 325000 China
| | - Changhuan Zhang
- Wenzhou Institute University of Chinese Academy Sciences Wenzhou Zhejiang 325000 China
| | - Jilong Wang
- Wenzhou Institute University of Chinese Academy Sciences Wenzhou Zhejiang 325000 China
| | - Junjie Deng
- Department of Orthopaedics The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou Zhejiang 325000 China
- Key Laboratory of Orthopaedics of Zhejiang Province Wenzhou Medical University Wenzhou Zhejiang 325000 China
- Wenzhou Institute University of Chinese Academy Sciences Wenzhou Zhejiang 325000 China
| |
Collapse
|
44
|
Zhang Y, Wang Y, Ding J, Liu P. Efferocytosis in multisystem diseases (Review). Mol Med Rep 2022; 25:13. [PMID: 34779503 PMCID: PMC8600411 DOI: 10.3892/mmr.2021.12529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/15/2021] [Indexed: 01/22/2023] Open
Abstract
Efferocytosis, the phagocytosis of apoptotic cells performed by both specialized phagocytes (such as macrophages) and non‑specialized phagocytes (such as epithelial cells), is involved in tissue repair and homeostasis. Effective efferocytosis prevents secondary necrosis, terminates inflammatory responses, promotes self‑tolerance and activates pro‑resolving pathways to maintain homeostasis. When efferocytosis is impaired, apoptotic cells that could not be cleared in time aggregate, resulting in the necrosis of apoptotic cells and release of pro‑inflammatory factors. In addition, defective efferocytosis inhibits the intracellular cholesterol reverse transportation pathways, which may lead to atherosclerosis, lung damage, non‑alcoholic fatty liver disease and neurodegenerative diseases. The uncleared apoptotic cells can also release autoantigens, which can cause autoimmune diseases. Cancer cells escape from phagocytosis via efferocytosis. Therefore, new treatment strategies for diseases related to defective efferocytosis are proposed. This review illustrated the mechanisms of efferocytosis in multisystem diseases and organismal homeostasis and the pathophysiological consequences of defective efferocytosis. Several drugs and treatments available to enhance efferocytosis are also mentioned in the review, serving as new evidence for clinical application.
Collapse
Affiliation(s)
- Yifan Zhang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P.R. China
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Yiru Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P.R. China
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Jie Ding
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P.R. China
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Ping Liu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P.R. China
| |
Collapse
|
45
|
Apoptotic cell-derived metabolites in efferocytosis-mediated resolution of inflammation. Cytokine Growth Factor Rev 2021; 62:42-53. [PMID: 34742632 DOI: 10.1016/j.cytogfr.2021.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 10/13/2021] [Indexed: 12/21/2022]
Abstract
The resolution of inflammation, as part of standard host defense mechanism, is the process to guarantee timely termination of inflammatory responses and eventual restoration of tissue homeostasis . It is mainly achieved via efferocytosis, during which pro-resolving macrophages clear apoptotic neutrophils at the inflammatory site. Unfortunately, impaired resolution can be the leading cause of chronic inflammatory disorders and some autoimmune diseases. Existing studies have provided relatively comprehensive understandings about the recognition and uptake of apoptotic neutrophils by macrophages during early phases of efferocytosis. However, lack of information concerns macrophage metabolism of apoptotic cell-derived metabolites after being released from phagolysosomes or the relationship between such metabolism and efferocytosis. Notwithstanding, three recent studies have revealed macrophage metabolism of cholesterol, fatty acids and arginine, as well as their respective functions in the context of inflammation-resolution. This review provides an overview of the resolution of inflammation, efferocytosis and the key players involved, followed by a focus on the metabolism of apoptotic cell-derived metabolites within efferocytes. Hypotheses of more potential apoptotic cell-derived metabolites and their possible roles in the resolution are also formulated. Understanding the effect of these metabolites further advances the concept that apoptotic cells act as active players to regulate resolution, and also suggests novel therapeutic strategies for diseases driven by defective resolution and even cancer that may be treated through enhanced efferocytosis.
Collapse
|
46
|
Physiological Roles of Apoptotic Cell Clearance: Beyond Immune Functions. Life (Basel) 2021; 11:life11111141. [PMID: 34833017 PMCID: PMC8621940 DOI: 10.3390/life11111141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
The clearance of apoptotic cells is known to be a critical step in maintaining tissue and organism homeostasis. This process is rapidly/promptly mediated by recruited or resident phagocytes. Phagocytes that engulf apoptotic cells have been closely linked to the release of anti-inflammatory cytokines to eliminate inflammatory responses. Defective clearance of apoptotic cells can cause severe inflammation and autoimmune responses due to secondary necrosis of apoptotic cells. Recently accumulated evidence indicates that apoptotic cells and their clearance have important physiological roles in addition to immune-related functions. Herein, we review the current understanding of the mechanisms and fundamental roles of apoptotic cell clearance and the beneficial roles of apoptotic cells in physiological processes such as differentiation and development.
Collapse
|
47
|
Wang Y, Carion TW, Ebrahim AS, Sosne G, Berger EA. Adjunctive Thymosin Beta-4 Treatment Influences MΦ Effector Cell Function to Improve Disease Outcome in Pseudomonas aeruginosa-Induced Keratitis. Int J Mol Sci 2021; 22:ijms222011016. [PMID: 34681676 PMCID: PMC8537948 DOI: 10.3390/ijms222011016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 11/16/2022] Open
Abstract
Our previous work has shown that topical thymosin beta 4 (Tβ4) as an adjunct to ciprofloxacin treatment reduces inflammatory mediators and inflammatory cell infiltrates (neutrophils/PMN and macrophages/MΦ) while enhancing bacterial killing and wound healing pathway activation in an experimental model of P. aeruginosa-induced keratitis. This study aimed to mechanistically examine how Tβ4 influences MΦ function in particular, leading to reduced inflammation and enhanced host defense following P. aeruginosa-induced infection of the cornea. Flow cytometry was conducted to profile the phenotype of infiltrating MΦ after infection, while generation of reactive nitrogen species and markers of efferocytosis were detected to assess functional activity. In vitro studies were performed utilizing RAW 264.7 cells to verify and extend the in vivo findings. Tβ4 treatment decreases MΦ infiltration and regulates the activation state in response to infected corneas. MΦ functional data demonstrated that the adjunctive Tβ4 treatment group significantly downregulated reactive nitrogen species (RNS) production and efferocytotic activity. In addition, the in vitro studies showed that both Tβ4 alone and adjunctive Tβ4 treatment influenced MΦ cellular function following LPS stimulation. Collectively, these data provide further evidence that adjunctive Tβ4 + ciprofloxacin treatment offers a more efficacious option for treating bacterial keratitis. Not only does the adjunctive therapy address both the infectious pathogen and corneal wound healing response, but it also influences MΦ infiltration, activation, and function, as revealed by the current study.
Collapse
|
48
|
Mertens C, Marques O, Horvat NK, Simonetti M, Muckenthaler MU, Jung M. The Macrophage Iron Signature in Health and Disease. Int J Mol Sci 2021; 22:ijms22168457. [PMID: 34445160 PMCID: PMC8395084 DOI: 10.3390/ijms22168457] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022] Open
Abstract
Throughout life, macrophages are located in every tissue of the body, where their main roles are to phagocytose cellular debris and recycle aging red blood cells. In the tissue niche, they promote homeostasis through trophic, regulatory, and repair functions by responding to internal and external stimuli. This in turn polarizes macrophages into a broad spectrum of functional activation states, also reflected in their iron-regulated gene profile. The fast adaptation to the environment in which they are located helps to maintain tissue homeostasis under physiological conditions.
Collapse
Affiliation(s)
- Christina Mertens
- Department of Pediatric Hematology, Oncology and Immunology, University of Heidelberg, INF 350, 69120 Heidelberg, Germany; (O.M.); (N.K.H.); (M.U.M.)
- Correspondence: (C.M.); (M.J.); Tel.: +(49)-622-156-4582 (C.M.); +(49)-696-301-6931 (M.J.)
| | - Oriana Marques
- Department of Pediatric Hematology, Oncology and Immunology, University of Heidelberg, INF 350, 69120 Heidelberg, Germany; (O.M.); (N.K.H.); (M.U.M.)
- Molecular Medicine Partnership Unit, 69120 Heidelberg, Germany
| | - Natalie K. Horvat
- Department of Pediatric Hematology, Oncology and Immunology, University of Heidelberg, INF 350, 69120 Heidelberg, Germany; (O.M.); (N.K.H.); (M.U.M.)
- Molecular Medicine Partnership Unit, 69120 Heidelberg, Germany
- European Molecular Biology Laboratory (EMBL), Collaboration for Joint PhD Degree between EMBL and the Faculty of Biosciences, University of Heidelberg, 69117 Heidelberg, Germany
| | - Manuela Simonetti
- Institute of Pharmacology, Medical Faculty Heidelberg, Heidelberg University, INF 366, 69120 Heidelberg, Germany;
| | - Martina U. Muckenthaler
- Department of Pediatric Hematology, Oncology and Immunology, University of Heidelberg, INF 350, 69120 Heidelberg, Germany; (O.M.); (N.K.H.); (M.U.M.)
- Molecular Medicine Partnership Unit, 69120 Heidelberg, Germany
| | - Michaela Jung
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany
- Correspondence: (C.M.); (M.J.); Tel.: +(49)-622-156-4582 (C.M.); +(49)-696-301-6931 (M.J.)
| |
Collapse
|
49
|
Senatus L, MacLean M, Arivazhagan L, Egaña-Gorroño L, López-Díez R, Manigrasso MB, Ruiz HH, Vasquez C, Wilson R, Shekhtman A, Gugger PF, Ramasamy R, Schmidt AM. Inflammation Meets Metabolism: Roles for the Receptor for Advanced Glycation End Products Axis in Cardiovascular Disease. IMMUNOMETABOLISM 2021; 3:e210024. [PMID: 34178389 PMCID: PMC8232874 DOI: 10.20900/immunometab20210024] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Fundamental modulation of energy metabolism in immune cells is increasingly being recognized for the ability to impart important changes in cellular properties. In homeostasis, cells of the innate immune system, such as monocytes, macrophages and dendritic cells (DCs), are enabled to respond rapidly to various forms of acute cellular and environmental stress, such as pathogens. In chronic stress milieus, these cells may undergo a re-programming, thereby triggering processes that may instigate tissue damage and failure of resolution. In settings of metabolic dysfunction, moieties such as excess sugars (glucose, fructose and sucrose) accumulate in the tissues and may form advanced glycation end products (AGEs), which are signaling ligands for the receptor for advanced glycation end products (RAGE). In addition, cellular accumulation of cholesterol species such as that occurring upon macrophage engulfment of dead/dying cells, presents these cells with a major challenge to metabolize/efflux excess cholesterol. RAGE contributes to reduced expression and activities of molecules mediating cholesterol efflux. This Review chronicles examples of the roles that sugars and cholesterol, via RAGE, play in immune cells in instigation of maladaptive cellular signaling and the mediation of chronic cellular stress. At this time, emerging roles for the ligand-RAGE axis in metabolism-mediated modulation of inflammatory signaling in immune cells are being unearthed and add to the growing body of factors underlying pathological immunometabolism.
Collapse
Affiliation(s)
- Laura Senatus
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Michael MacLean
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Lakshmi Arivazhagan
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Lander Egaña-Gorroño
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Raquel López-Díez
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Michaele B. Manigrasso
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Henry H. Ruiz
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Carolina Vasquez
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Robin Wilson
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | | | - Paul F. Gugger
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ravichandran Ramasamy
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
50
|
Chen W, Li L, Wang J, Zhang R, Zhang T, Wu Y, Wang S, Xing D. The ABCA1-efferocytosis axis: A new strategy to protect against atherosclerosis. Clin Chim Acta 2021; 518:1-8. [PMID: 33741356 DOI: 10.1016/j.cca.2021.02.025] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/24/2021] [Accepted: 02/26/2021] [Indexed: 12/13/2022]
Abstract
Atherosclerosis, a disease process characterized by lipid accumulation and inflammation, is the main cause of coronary heart disease (CHD) and myocardial infarction (MI). Efferocytosis involves the clearance of apoptotic cells by phagocytes. Successful engulfment triggers the release of anti-inflammatory cytokines to suppress atherosclerosis. ABCA1 is a key mediator of cholesterol efflux to apoA-I for the generation of HDL-C in reverse cholesterol transport (RCT). Intriguingly, ABCA1 promotes not only cholesterol efflux but also efferocytosis. ABCA1 promotes efferocytosis by regulating the release of "find-me" ligands, including LPC, and the exposure, release, and expression of "eat-me" ligands, including PtdSer, ANXA1, ANXA5, MEGF10, and GULP1. ABCA1 has a pathway similar to TG2, which is an "eat-me" ligand. ABCA1 has the highest known homology to ABCA7, which controls efferocytosis as the engulfment and processing ligand. In addition, ABCA1 can form several regulatory feedback axes with ANXA1, MEGF10, GULP1, TNFα, and IL-6. Therefore, ABCA1 is the central factor that links cholesterol efflux and apoptotic cell clearance. Several drugs have been studied or approved for apoptotic cell clearance, such as CD47 antibody and PD1-/PD-L1 antibody. In this article, we review the role and mechanism of action of ABCA1 in efferocytosis and focus on new insights into the ABCA1-efferocytosis axis and its potential as a novel therapeutic target in atherosclerosis.
Collapse
Affiliation(s)
- Wujun Chen
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Lu Li
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Jie Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Renshuai Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Tingting Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Yudong Wu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China.
| | - Shuai Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China; School of Medical Imaging, Radiotherapy Department of Affiliated Hospital, Weifang Medical University, Weifang, Shandong 261053, China.
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|