1
|
Yvan-Charvet L, Barouillet T, Borowczyk C. Haematometabolism rewiring in atherosclerotic cardiovascular disease. Nat Rev Cardiol 2025; 22:414-430. [PMID: 39743562 DOI: 10.1038/s41569-024-01108-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/14/2024] [Indexed: 01/04/2025]
Abstract
Atherosclerotic cardiovascular diseases are the most frequent cause of death worldwide. The clinical complications of atherosclerosis are closely linked to the haematopoietic and immune systems, which maintain homeostatic functions and vital processes in the body. The nodes linking metabolism and inflammation are receiving increasing attention because they are inextricably linked to inflammatory manifestations of non-communicable diseases, including atherosclerosis. Although metabolism and inflammation are essential to survival and involve all tissues, we still know little about how these processes influence each other. In an effort to understand these mechanisms, in this Review we explore whether and how potent cardiovascular risk factors and metabolic modifiers of atherosclerosis influence the molecular and cellular machinery of 'haematometabolism' (metabolic-dependent haematopoietic stem cell skewing) and 'efferotabolism' (metabolic-dependent efferocyte reprogramming). These changes might ultimately propagate a quantitative and qualitative drift of the macrophage supply chain and affect the clinical manifestations of atherosclerosis. Refining our understanding of the different metabolic requirements of these processes could open the possibility of developing therapeutics targeting haematometabolism that, in conjunction with improved dietary habits, help rebalance and promote efficient haematopoiesis and efferocytosis and decrease the risk of atherosclerosis complications.
Collapse
Affiliation(s)
- Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Nice, France.
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France.
- Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France.
| | - Thibault Barouillet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Nice, France
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
- Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France
| | - Coraline Borowczyk
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Nice, France.
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France.
- Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France.
| |
Collapse
|
2
|
Jin J, Ma L, Li L, Zhou X, Zhu S, Shen K, Xu Q, Jiang B, Gu Y, Ding Q, Qian H, Lv T, Song Y. Geranylgeranyl diphosphate synthase deficiency impairs efferocytosis and resolution of acute lung injury. Respir Res 2025; 26:189. [PMID: 40380222 PMCID: PMC12084987 DOI: 10.1186/s12931-025-03241-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 04/16/2025] [Indexed: 05/19/2025] Open
Abstract
Acute respiratory distress syndrome (ARDS) are major causes of mortality of critically ill patients. Impaired macrophage-mediated clearance of apoptotic cells (efferocytosis) in ARDS contributes to prolonged inflammation, yet the underlying mechanisms remain unclear. In this study, we investigated the role of geranylgeranyl diphosphate synthase (GGPPS) in efferocytosis during lung injury resolution. We identified dynamic changes in GGPPS expression in lung macrophages and circulating monocytes throughout the progression and resolution phases of acute lung injury (ALI). Myeloid-specific GGPPS knockout mice exhibited prolonged lung inflammation, increased accumulation of apoptotic neutrophils, a higher number of recruited macrophages, and a reduced number of resident macrophages. Notably, recruited macrophages play a dominant role in efferocytosis compared to resident macrophages. GGPPS deficiency suppressed efferocytosis in both macrophage subsets in vivo and in vitro. Mechanistically, GGPPS knockout disrupted AXL signaling in recruited macrophages. Importantly, administration of geranylgeraniol (GGOH) rescued the delayed resolution of lung injury, restored efferocytosis, and increased the suppressed AXL expression in CKO mice. Collectively, this study identifies GGPPS as a key regulator of AXL-mediated efferocytosis in recruited macrophages, highlighting its potential as a therapeutic target to accelerate ARDS resolution.
Collapse
Affiliation(s)
- Jiajia Jin
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing Medical University, #305, East Zhongshan Road, Nanjing, 210002, China
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing, 210002, China
| | - Lihong Ma
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing Medical University, #305, East Zhongshan Road, Nanjing, 210002, China
- Department of Respiratory and Critical Care Medicine, Wuxi No. 2 People's Hospital, Wuxi, 214086, China
| | - Lulu Li
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing, 210002, China
| | - Xinyu Zhou
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing University, Nanjing, 210002, China
| | - Suhua Zhu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing, 210002, China
| | - Kaikai Shen
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing University, Nanjing, 210002, China
| | - Qiuli Xu
- Southeast University Medical College, Jinling Hospital, Nanjing, 210000, China
| | - Bei Jiang
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing University of Chinese Medicine, Nanjing, 210000, China
| | - Yanli Gu
- Department of Respiratory and Critical Care Medicine, The Affiliated Huaian No.1 People's Hospital, Nanjing Medical University, Huai'an, 223000, China
| | - Qianshan Ding
- Nanjing First Hospital, affiliated with Nanjing Medical University, Nanjing, 210006, China
| | - Hong Qian
- Department of Orthopaedic Surgery, Jinling Hospital, the First School of Clinical Medicine, Southern Medical University, Nanjing, 210002, China.
| | - Tangfeng Lv
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing Medical University, #305, East Zhongshan Road, Nanjing, 210002, China.
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing, 210002, China.
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing University, Nanjing, 210002, China.
| | - Yong Song
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing Medical University, #305, East Zhongshan Road, Nanjing, 210002, China.
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing, 210002, China.
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing University, Nanjing, 210002, China.
| |
Collapse
|
3
|
Soliman AM, Soliman M, Shah SSH, Baig HA, Gouda NS, Alenezi BT, Alenezy A, Hegazy AMS, Jan M, Eltom EH. Molecular dynamics of inflammation resolution: therapeutic implications. Front Cell Dev Biol 2025; 13:1600149. [PMID: 40406415 PMCID: PMC12095172 DOI: 10.3389/fcell.2025.1600149] [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: 03/25/2025] [Accepted: 04/23/2025] [Indexed: 05/26/2025] Open
Abstract
Inflammation is a critical part of innate immune response that is essential for exclusion of harmful stimuli and restoration of tissue homeostasis. Nonetheless, failure to resolve inflammation results in chronic inflammatory conditions, including autoimmune diseases. Conventionally, resolution of inflammation was deemed a passive process; however, evidence indicates that it entails active, highly regulated molecular and cellular events involving efferocytosis-driven macrophage reprogramming, post-transcriptional regulatory mechanisms and the production of specialized pro-resolving mediators (SPMs). These processes collectively restore tissue homeostasis and prevent chronic inflammation. Emerging therapeutic approaches targeting these pathways demonstrate promising results in preclinical studies and clinical trials, enhancing resolution and improving overall disease outcome. This resulted in a paradigm shift from conventional anti-inflammatory strategies to resolution-focused treatment. Yet, challenges remain due to the complexity of resolution mechanisms and tissue-specific differences. This review summarizes current advances in inflammation resolution, emphasizing emerging concepts of resolution pharmacology. By employing endogenous mechanisms facilitating resolution, novel therapeutic applications can effectively manage several chronic inflammatory disorders.
Collapse
Affiliation(s)
- Amro M. Soliman
- Department of Biological Sciences, Faculty of Science, Concordia University of Edmonton, Edmonton, AB, Canada
| | - Mohamed Soliman
- Department of Microbiology, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia
| | - Syed Sajid Hussain Shah
- Department of Pathology, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia
| | - Habeeb Ali Baig
- Department of Microbiology, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia
| | - Nawal Salama Gouda
- Department of Microbiology, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia
| | - Bandar Theyab Alenezi
- Department of Pharmacology, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia
| | - Awwad Alenezy
- Department of Family and Community Medicine, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia
| | - Ahmed M. S. Hegazy
- Department of Anatomy, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia
| | - Muhammad Jan
- Department of Pharmacology, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia
| | - Elhassan Hussein Eltom
- Department of Pharmacology, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia
| |
Collapse
|
4
|
Tang S, Lu C, Meng Z, Ye Z, Qin Y, Na N, Xian S, Huang F, Zeng Z. USP22 enhances atherosclerotic plaque stability and macrophage efferocytosis by stabilizing PPARγ. Commun Biol 2025; 8:678. [PMID: 40301680 PMCID: PMC12041205 DOI: 10.1038/s42003-025-08116-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 04/22/2025] [Indexed: 05/01/2025] Open
Abstract
Atherosclerosis is a chronic inflammatory disease that strongly threatens human health, and macrophages play a pivotal role in its pathogenesis. Ubiquitin-specific peptidase 22 (USP22) is involved in the regulation of macrophage inflammation. However, its role in the atherosclerotic microenvironment remains unclear. In this study, we found that USP22 overexpression in macrophages alleviated atherosclerosis progression in ApoE-/- mice. In vitro, USP22 silencing enhanced macrophage inflammation and foam cell formation, and macrophage efferocytosis was significantly impaired. Mechanistically, USP22 bound to peroxisome proliferator-activated receptor γ (PPARγ) and inhibited its ubiquitination, thereby stabilizing PPARγ and promoting efferocytosis. In addition, intraperitoneal injection of the USP22 inhibitor USP22i-S02 exacerbated atherosclerosis in ApoE-/- mice. In summary, these findings indicate that USP22 may be a potential therapeutic target for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Senhu Tang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, Guangxi, China
| | - Chuanghong Lu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, Guangxi, China
| | - Zhongyuan Meng
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, Guangxi, China
| | - Zihua Ye
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, Guangxi, China
| | - Yue Qin
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, Guangxi, China
| | - Na Na
- Department of Neuroscience, Scripps Research Institute, San Diego, CA, USA
| | - Shenglin Xian
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, Guangxi, China
| | - Feng Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, Guangxi, China.
| | - Zhiyu Zeng
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, Guangxi, China.
| |
Collapse
|
5
|
Zhou X, Zhu D, Wu D, Li G, Liang H, Zhang W, Wu Y, Xu H, Zhang Z, Tong B, Song Y, Wang K, Feng X, Lei J, Wang H, Zhang X, Ma L, Chen Y, Wei J, Ou Z, Peng S, Wu X, Tan L, Wang B, Yang C. Microneedle delivery of CAR-M-like engineered macrophages alleviates intervertebral disc degeneration through enhanced efferocytosis capacity. Cell Rep Med 2025; 6:102079. [PMID: 40199328 PMCID: PMC12047514 DOI: 10.1016/j.xcrm.2025.102079] [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: 03/19/2024] [Revised: 10/09/2024] [Accepted: 03/18/2025] [Indexed: 04/10/2025]
Abstract
Macrophages eliminate apoptotic cells produced daily in the body through efferocytosis. Restricted clearance can cause inflammation-related diseases. In intervertebral discs (IVDs), apoptotic nucleus pulposus cells (NPCs) are difficult to effectively remove, and their accumulation can cause changes in the inflammatory microenvironment, disrupt IVD homeostasis, and lead to IVD degeneration (IDD). Here, we present chimeric antigen receptor-M-like engineered macrophages (CAR-eMs) with enhanced efferocytosis capacity for IDD treatment. Macrophages undergo phenotypic transformation and a reduction in phagocytic ability after phagocyting apoptotic NPCs, but their efferocytosis capacity recovers with upregulated brain-specific angiogenesis inhibitor 1 (BAI1) expression. We develop a CAR-eM system with enhanced BAI1 expression and an IVD circular microneedle (MN) delivery system that utilizes arrays of MNs to deliver CAR-eMs into the deep IVD layers, thereby clearing apoptotic NPCs, ameliorating the inflammatory microenvironment, and repairing damaged IVDs. Our study explores the therapeutic potential of CAR-eM efferocytosis for IDD treatment.
Collapse
Affiliation(s)
- Xingyu Zhou
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Dingchao Zhu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Di Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Gaocai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Huaizhen Liang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Weifeng Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yali Wu
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hanpeng Xu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhengdong Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Bide Tong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yu Song
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Kun Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaobo Feng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jie Lei
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hongchuan Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaoguang Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Liang Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuhang Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Junyu Wei
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zixuan Ou
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shuchang Peng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xinghuo Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lei Tan
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Bingjin Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen 518057, China.
| |
Collapse
|
6
|
Duan Y, Wu Y, Tian J, Yin Y, Yuan Z, Zhu W, Zhou S, Li C, Feng S. Elucidation of the mechanism Underlying the promotion of ferroptosis and enhanced antitumor immunity by citrus polymethoxyflavones in CRC cells. Front Pharmacol 2025; 16:1571178. [PMID: 40290432 PMCID: PMC12021823 DOI: 10.3389/fphar.2025.1571178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/01/2025] [Indexed: 04/30/2025] Open
Abstract
Background Colon cancer is a prevalent condition with a high mortality rate on a global scale. Research has indicated that Citrus polymethoxyflavones (PMFs), a class of flavonoids found in Citrus, possess the potential to demonstrate anti-tumor efficacy. Ferroptosis a form of cell death that is dependent on iron accumulation and lipid peroxidation. Immunotherapy is one of the most commonly used anti-tumor modalities in a clinical setting. Consequently, studies on the pharmacodynamic mechanism of Citrus to determine whether it can modulate tumor immunity through ferroptosis provide new ideas for the clinical treatment of colon cancer. Purpose The objective of this study is to ascertain whether Citrus inhibits PD-L1 through ferroptosis and promotes tumor immunity among patients with colon cancer. Methods The inhibitory effect of PMFs on colon cancer was proved by in vitro experiment and in vivo model. In addition, the occurrence of ferroptosis was detected by measuring key ferroptosis indicators. Bioinformatics analysis was then performed to identify the crossover genes for Citrus polymethoxylflavonoids, colon cancer, and ferroptosis. Finally, key genes were identified by immunocorrelation analysis including WB, Q-PCR and flow cytometry. These experiments were designed to reveal the potential mechanisms of PMFs on ferroptosis and anti-tumor immunity. Results In vitro cell proliferation experiment and the growth of transplanted tumor mice showed that PMFs had inhibitory effect on colon cancer. In addition, the change of ferroptosis index showed that PMFs promoted the occurrence of ferroptosis, followed by Q-PCR and WB detection of NOX4 and TIMP1, the key genes screened by bioinformatics, found that PMFs inhibited PD-L1 by down-regulating TIMP1, thus affecting colon cancer. Flow cytometry showed that CD4+ T expression increased and CD8+ T cell expression decreased after treatment, suggesting that anti-tumor immunity was activated. Conclusion It is conceivable that the tumor immune microenvironment may be subject to regulation during the inhibition of colon cancer through ferroptosis in PMFs. The ferroptosis-related gene TIMP1 has been observed to regulate PD-L1, thereby promoting anti-tumor immunity in colon cancer. However, further investigation is required to ascertain the underlyingprecise mechanisms.
Collapse
Affiliation(s)
- Yingying Duan
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yu Wu
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiaqi Tian
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yuqin Yin
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Zhongwen Yuan
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Wenting Zhu
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Suyue Zhou
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chen Li
- Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Senling Feng
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
7
|
Zang H, Ji X, Yao W, Wan L, Zhang C, Zhu C, Liu T. Role of efferocytosis in chronic pain -- From molecular perspective. Neurobiol Dis 2025; 207:106857. [PMID: 40015655 DOI: 10.1016/j.nbd.2025.106857] [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: 12/27/2024] [Revised: 02/23/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025] Open
Abstract
The complex nature of pain pathophysiology complicates the establishment of objective diagnostic criteria and targeted treatments. The heterogeneous manifestations of pain stemming from various primary diseases contribute to the complexity and diversity of underlying mechanisms, leading to challenges in treatment efficacy and undesirable side effects. Recent evidence suggests the presence of apoptotic cells at injury sites, the distal dorsal root ganglia (DRG), spinal cord, and certain brain regions, indicating a potential link between the ineffective clearance of dead cells and debris and pain persistence. This review highlights recent research findings indicating that efferocytosis plays a significant yet often overlooked role in lesion expansion while also representing a potentially reversible impairment that could be targeted therapeutically to mitigate chronic pain progression. We examine recent advances into how efferocytosis, a process by which phagocytes clear apoptotic cells without triggering inflammation, influences pain initiation and intensity in both human diseases and animal models. This review summarizes that efferocytosis contributes to pain progression from the perspective of defective and inefficient efferocytosis and its subsequent secondary necrocytosis, cascade inflammatory response, and the shift of phenotypic plasticity and metabolism. Additionally, we investigate the roles of newly discovered genetic alterations or modifications in biological signaling pathways in pain development and chronicity, providing insights into innovative treatment strategies that modulate efferocytosis, which are promising candidates and potential avenues for further research in pain management and prevention.
Collapse
Affiliation(s)
- Hu Zang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiaoyu Ji
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Wenlong Yao
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Li Wan
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chuanhan Zhang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chang Zhu
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| | - Tongtong Liu
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| |
Collapse
|
8
|
Cao X, Li F, Xie X, Ling G, Tang X, He W, Tian J, Ge Y. Efferocytosis and inflammation: a bibliometric and systematic analysis. Front Med (Lausanne) 2025; 12:1498503. [PMID: 39995691 PMCID: PMC11847848 DOI: 10.3389/fmed.2025.1498503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/22/2025] [Indexed: 02/26/2025] Open
Abstract
Objective To visualize and analyze the trends and hotspots of efferocytosis and inflammation via bibliometric methods. Methods Relevant articles and reviews from 2006 to 2023 were retrieved from the Web of Science Core Collection. The data were processed with CiteSpace, and some graphs were generated with Microsoft Excel (version 2016), VOSviewer, Scimago Graphica, Bibliometrix and R Studio. Results A total of 1,003 papers were included, revealing a significant upward trend in efferocytosis and inflammation research. The United States (456, 45.46%), China (164, 16.35%) and the United Kingdom (99, 9.87%) were the three countries with the highest numbers of publications. Harvard University (84, 6.74%) contributes the most out of the top 5 institutions. Among the researchers in this field, Serhan CN was the author with the highest number of articles in the field (35, 3.49%), and deCathelineau AM first named "efferocytosis" in 2003. Keyword analysis identified "activation," "tam receptors," "docosahexaenoic acid" "systemic lupus erythematosus," "myocardial infarction" and "alveolar macrophages" as core topics, indicating a concentrated trend in the mechanism of physiological state and inflammatory diseases such as autoimmune, cardiovascular, and pulmonary diseases. The latest surge words "inflammation resolution" and "cancer" in the keyword heatmap indicate future research directions. Conclusion Research on the association between efferocytosis and inflammation has been a promising field. Key areas of focus include the crucial role of efferocytosis on tissue homeostasis and the pathogenesis of nontumorous inflammatory diseases. Future research will likely continue to explore these frontiers, with an emphasis on understanding efferocytosis in the context of chronic diseases and cancer, as well as developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Xin Cao
- Department of Rheumatology & Immunology, The Second Xiangya Hospital, Central South University, Changsha, China
- Clinical Medical Research Center for Systemic Autoimmune Diseases, Changsha, China
| | - Fen Li
- Department of Rheumatology & Immunology, The Second Xiangya Hospital, Central South University, Changsha, China
- Clinical Medical Research Center for Systemic Autoimmune Diseases, Changsha, China
| | - Xi Xie
- Department of Rheumatology & Immunology, The Second Xiangya Hospital, Central South University, Changsha, China
- Clinical Medical Research Center for Systemic Autoimmune Diseases, Changsha, China
| | - Guanghui Ling
- Department of Rheumatology & Immunology, The Second Xiangya Hospital, Central South University, Changsha, China
- Clinical Medical Research Center for Systemic Autoimmune Diseases, Changsha, China
| | - Xiaoyu Tang
- Department of Rheumatology & Immunology, The Second Xiangya Hospital, Central South University, Changsha, China
- Clinical Medical Research Center for Systemic Autoimmune Diseases, Changsha, China
| | - Wenfang He
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jing Tian
- Department of Rheumatology & Immunology, The Second Xiangya Hospital, Central South University, Changsha, China
- Clinical Medical Research Center for Systemic Autoimmune Diseases, Changsha, China
| | - Yan Ge
- Department of Rheumatology & Immunology, The Second Xiangya Hospital, Central South University, Changsha, China
- Clinical Medical Research Center for Systemic Autoimmune Diseases, Changsha, China
| |
Collapse
|
9
|
Hilu-Dadia R, Ghanem A, Vogelesang S, Ayoub M, Hakim-Mishnaevski K, Kurant E. Santa-maria is a glial phagocytic receptor that acts with SIMU to recognize and engulf apoptotic neurons. Cell Rep 2025; 44:115201. [PMID: 39799566 DOI: 10.1016/j.celrep.2024.115201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 12/08/2024] [Accepted: 12/20/2024] [Indexed: 01/15/2025] Open
Abstract
The elimination of superfluous neurons via apoptosis and subsequent glial phagocytosis is crucial for the development of the central nervous system (CNS). In Drosophila, two glial phagocytic receptors, six-microns-under (SIMU) and Draper, mediate the phagocytosis of apoptotic neurons during embryogenesis. However, in simu;draper double-mutant embryos, some apoptotic neurons are still engulfed by the glia, suggesting the involvement of additional receptors. Here, we discover the Drosophila CD36 homolog Santa-maria, a transmembrane receptor, which is specifically expressed in embryonic phagocytic glia and plays a major role in the recognition and engulfment steps of phagocytosis. Our data demonstrate that santa-maria genetically interacts with simu and draper, while the protein product binds apoptotic cells and physically interacts with the SIMU protein. Moreover, we reveal that triple knockout of genes for all three glial phagocytic receptors (i.e., simu, draper, and santa-maria) causes partial lethality, thus illuminating their role in development, particularly in the developing nervous system.
Collapse
Affiliation(s)
- Reut Hilu-Dadia
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa 34988, Israel
| | - Aseel Ghanem
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa 34988, Israel
| | - Shelly Vogelesang
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa 34988, Israel
| | - Malak Ayoub
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa 34988, Israel
| | - Ketty Hakim-Mishnaevski
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa 34988, Israel
| | - Estee Kurant
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa 34988, Israel.
| |
Collapse
|
10
|
Liang X, Tian S, Zhang H, Sun S, Zhang P, Li J, Li Y, Zhang Y, Liu Z. Efferocytosis: A new star of atherosclerotic plaques reversal. Int Immunopharmacol 2025; 146:113904. [PMID: 39724733 DOI: 10.1016/j.intimp.2024.113904] [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: 03/21/2024] [Revised: 12/16/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Efferocytosis is considered the key to eliminate apoptotic cells (ACs) under physiological and pathological conditions in vivo, mainly through different types of macrophages to achieve this process. Especially, tissue-resident macrophages (TRMs) are very significant for inflammation regression and maintenance of homeostasis in vivo. Abnormal efferocytosis will lead to the accumulation of ACs and the release of a variety of pro-inflammatory factors, which mediates the occurrence of many inflammatory diseases, including atherosclerosis (AS). AS is a chronic inflammatory vascular disease with the participation of the immune system. Defective efferocytosis will accelerate the progress of AS to a certain extent. Therefore, it is of great significance to understand the mechanism of efferocytosis and realize the prevention and treatment of AS through efferocytosis. In this review, we will briefly describe the specific process of efferocytosis, deeply discuss the possible molecular mechanism of impaired efferocytosis promoting the development of AS, and summarize the ways to prevent and treat AS through efferocytosis intervention therapy.
Collapse
Affiliation(s)
- Xiangyu Liang
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Baoding, China.
| | - Shuoqi Tian
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Baoding, China.
| | - Han Zhang
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Baoding, China.
| | - Shusen Sun
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Baoding, China.
| | - Peixiang Zhang
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Baoding, China.
| | - Jiameng Li
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Baoding, China.
| | - Yong Li
- Beijing Yongkang Nian Health Technology Co., Ltd., Beijing, China.
| | - Yanfen Zhang
- Technology Transfer Center, Hebei University, Baoding, China.
| | - Zhongcheng Liu
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Baoding, China.
| |
Collapse
|
11
|
Yang Y, Wu A, Deng AN, Liu H, Lan Q, Mazhar M, Xue JY, Chen MT, Luo G, Liu MN. Macrophages after myocardial infarction: Mechanisms for repairing and potential as therapeutic approaches. Int Immunopharmacol 2024; 143:113562. [PMID: 39536484 DOI: 10.1016/j.intimp.2024.113562] [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: 09/21/2024] [Revised: 10/20/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Macrophages - one of the crucial immune cells, are recruited to the cardiac tissue by chemokines, cytokines and upregulated endothelial adhesion molecules after myocardial infarction (MI). During the course of inflammation in the cardiac tissue, necrotic cells and matrix debris is phagocytosed by M1 macrophages. During the resolution phase of cardiac inflammation, M2 macrophages promote cardiac recovery. Suppression or over expression of both the M1 and M2 macrophage subtypes significantly affect the reparation of infarction. Stem cells therapy, cytokine regulation and immune cells therapy are considered as effective interventions to regulate the phenotypic transformation of cardiac macrophages after MI. Intervention with macrophages in the myocardium has shown unique advantages. In this review, the mechanisms and role of macrophages in the development of MI are elaborated in detail, the promising therapeutic methods for regulating macrophage phenotypes, their limitations and possible future research directions are discussed.
Collapse
Affiliation(s)
- You Yang
- Department of Pediatrics, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Ai Wu
- Department of Pediatrics, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - An-Ni Deng
- Department of Pediatrics, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Hao Liu
- Department of Pediatrics, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Qi Lan
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Maryam Mazhar
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Jin-Yi Xue
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Ming-Tai Chen
- Department of Cardiovascular Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China.
| | - Gang Luo
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China.
| | - Meng-Nan Liu
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
12
|
Liu WT, Li CQ, Fu AN, Yang HT, Xie YX, Yao H, Yi GH. Therapeutic implication of targeting mitochondrial drugs designed for efferocytosis dysfunction. J Drug Target 2024; 32:1169-1185. [PMID: 39099434 DOI: 10.1080/1061186x.2024.2386620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/17/2024] [Accepted: 07/25/2024] [Indexed: 08/06/2024]
Abstract
Efferocytosis refers to the process by which phagocytes remove apoptotic cells and related apoptotic products. It is essential for the growth and development of the body, the repair of damaged or inflamed tissues, and the balance of the immune system. Damaged efferocytosis will cause a variety of chronic inflammation and immune system diseases. Many studies show that efferocytosis is a process mediated by mitochondria. Mitochondrial metabolism, mitochondrial dynamics, and communication between mitochondria and other organelles can all affect phagocytes' clearance of apoptotic cells. Therefore, targeting mitochondria to modulate phagocyte efferocytosis is an anticipated strategy to prevent and treat chronic inflammatory diseases and autoimmune diseases. In this review, we introduced the mechanism of efferocytosis and the pivoted role of mitochondria in efferocytosis. In addition, we focused on the therapeutic implication of drugs targeting mitochondria in diseases related to efferocytosis dysfunction.
Collapse
Affiliation(s)
- Wan-Ting Liu
- Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hunan, Hengyang, China
| | - Chao-Quan Li
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hunan, Hengyang, China
| | - Ao-Ni Fu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hunan, Hengyang, China
| | - Hao-Tian Yang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hunan, Hengyang, China
| | - Yu-Xin Xie
- Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hunan, Hengyang, China
| | - Hui Yao
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hunan, Hengyang, China
| | - Guang-Hui Yi
- Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hunan, Hengyang, China
| |
Collapse
|
13
|
Ruan S, Jia R, Hu L, Liu Y, Tian Q, Jiang K, Xia X, Tao X, Liu WT, Pan Y, Hu F. Ozone promotes macrophage efferocytosis and alleviates neuropathic pain by activating the AMPK/Gas6-MerTK/SOCS3 signaling pathway. Front Immunol 2024; 15:1455771. [PMID: 39628480 PMCID: PMC11611719 DOI: 10.3389/fimmu.2024.1455771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/28/2024] [Indexed: 12/06/2024] Open
Abstract
Background Neuropathic pain (NPP) is a multifaceted pain syndrome that occurs as a consequence of physical injury or underlying diseases, with an incidence rate of 7%-10%, NPP poses a significant clinical challenge as current treatment options are ineffective. The accumulation of apoptotic cells and neuroinflammation play crucial roles in the pathological mechanisms of NPP. Here, we aim to investigate strategies for effectively clearing apoptotic cells and provide therapeutic interventions for NPP. Methods CCI mice were treated with different concentrations of ozone (15μg, 30μg, 45μg) to investigate the effects on the accumulation of apoptotic cells and neuroinflammation. In vitro, the phagocytic function of BMDM towards apoptotic neutrophils after ozone treatment was examined. Results We found ozone at a concentration of 30μg significantly alleviated mechanical hypersensitivity in CCI mice and ozone significantly upregulates the phagocytic activity of BMDM. Furthermore, we investigated the mechanisms and found ozone could activate AMPK, upregulate Gas6 (but not Protein S), activate MerTK (a key receptor involved in apoptosis), and enhance the phagocytic function of BMDM towards apoptotic neutrophils. It caused the promotion of SOCS3 expression and the suppression of inflammatory factors IL-1β, IL-6, and TNF-a. Interestingly, the effect of ozone in alleviating CCI-induced pain was abolished by the AMPK inhibitor CC and the MerTK receptor inhibitor UNC2541. Conclusion Ozone facilitated macrophage clearance of apoptotic cells, decreased neuroinflammation by activation of p-AMPK/Gas6/MerTK/SOCS3 signaling pathway, which may become an effective therapeutic approach for neuropathic pain after further clinical validation.
Collapse
Affiliation(s)
- Shirong Ruan
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rumeng Jia
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Liang Hu
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuge Liu
- Department of Anesthesiology, Ili and Jiangsu Joint Institute of Health, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yining, Xinjiang, China
| | - Qingyan Tian
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Kunmao Jiang
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xinyue Xia
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xueyou Tao
- Department of Anesthesiology, Yangzhou Maternal and Child Health Hospital Affiliated to Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Wen-Tao Liu
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yinbing Pan
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fan Hu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
14
|
Bai M, Wang T, Xing Z, Huang H, Wu X, Adeli M, Wang M, Han X, Ye L, Cheng C. Electron-donable heterojunctions with synergetic Ru-Cu pair sites for biocatalytic microenvironment modulations in inflammatory mandible defects. Nat Commun 2024; 15:9592. [PMID: 39505847 PMCID: PMC11541594 DOI: 10.1038/s41467-024-53824-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 10/22/2024] [Indexed: 11/08/2024] Open
Abstract
The clinical treatments of maxillofacial bone defects pose significant challenges due to complex microenvironments, including severe inflammation, high levels of reactive oxygen species (ROS), and potential bacterial infection. Herein, we propose the de novo design of an efficient, versatile, and precise electron-donable heterojunction with synergetic Ru-Cu pair sites (Ru-Cu/EDHJ) for superior biocatalytic regeneration of inflammatory mandible defects and pH-controlled antibacterial therapies. Our studies demonstrate that the unique structure of Ru-Cu/EDHJ enhances the electron density of Ru atoms and optimizes the binding strength of oxygen species, thus improving enzyme-like catalytic performance. Strikingly, this biocompatible Ru-Cu/EDHJ can efficiently switch between ROS scavenging in neutral media and ROS generation in acidic media, thus simultaneously exhibiting superior repair functions and bioadaptive antibacterial properties in treating mandible defects in male mice. We believe synthesizing such biocatalytic heterojunctions with exceptional enzyme-like capabilities will offer a promising pathway for engineering ROS biocatalytic materials to treat trauma, tumors, or infection-caused maxillofacial bone defects.
Collapse
Grants
- 52161145402, 52173133, 52373148 National Natural Science Foundation of China (National Science Foundation of China)
- 82470962, 82001020 National Natural Science Foundation of China (National Science Foundation of China)
- U21A20368 National Natural Science Foundation of China (National Science Foundation of China)
- sklpme2021-4-02 State Key Laboratory of Polymer Materials Engineering
- National Key R&D Program of China (2021YFB3800700),Sichuan Science and Technology Program (2023YFH0008),the 1·3·5 Project for Disciplines of Excellence, West China Hospital, Sichuan University (ZYJC21047).
- Sichuan Science and Technology Program (2024NSFSC0672, 2021YFG0238),China Postdoctoral Science Foundation (2019M663525), Research Funding from West China School/Hospital of Stomatology Sichuan University (RCDWJS2023-16), and Research and Develop Program, West China Hospital of Stomatology Sichuan University (RD-02-202206).
- National Key R&D Program of China (2023YFC3605600), Sichuan Science and Technology Program (2023YFS0019), Med-X Innovation Programme of Med-X Center for Materials, Sichuan University (MCMGD202301)
Collapse
Affiliation(s)
- Mingru Bai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ting Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Zhenyu Xing
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Haoju Huang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Xizheng Wu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Mohsen Adeli
- Institute of Chemistry and Biochemistry, Free University of Berlin, Berlin, Germany
| | - Mao Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Xianglong Han
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Chong Cheng
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China.
| |
Collapse
|
15
|
Abstract
Cardiovascular disease is the leading cause of death worldwide, and it commonly results from atherosclerotic plaque progression. One of the increasingly recognized drivers of atherosclerosis is dysfunctional efferocytosis, a homeostatic mechanism responsible for the clearance of dead cells and the resolution of inflammation. In atherosclerosis, the capacity of phagocytes to participate in efferocytosis is hampered, leading to the accumulation of apoptotic and necrotic tissue within the plaque, which results in enlargement of the necrotic core, increased luminal stenosis and plaque inflammation, and predisposition to plaque rupture or erosion. In this Review, we describe the different forms of programmed cell death that can occur in the atherosclerotic plaque and highlight the efferocytic machinery that is normally implicated in cardiovascular physiology. We then discuss the mechanisms by which efferocytosis fails in atherosclerosis and other cardiovascular and cardiometabolic diseases, including myocardial infarction and diabetes mellitus, and discuss therapeutic approaches that might reverse this pathological process.
Collapse
Affiliation(s)
- Shaunak S Adkar
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford, CA, USA
| | - Nicholas J Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford, CA, USA.
| |
Collapse
|
16
|
Cao C, Liu W, Guo X, Weng S, Chen Y, Luo Y, Wang S, Zhu B, Liu Y, Peng D. Identification and validation of efferocytosis-related biomarkers for the diagnosis of metabolic dysfunction-associated steatohepatitis based on bioinformatics analysis and machine learning. Front Immunol 2024; 15:1460431. [PMID: 39497821 PMCID: PMC11532026 DOI: 10.3389/fimmu.2024.1460431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 10/02/2024] [Indexed: 11/07/2024] Open
Abstract
Background Metabolic dysfunction-associated steatohepatitis (MASH) is a highly prevalent liver disease globally, with a significant risk of progressing to cirrhosis and even liver cancer. Efferocytosis, a process implicated in a broad spectrum of chronic inflammatory disorders, has been reported to be associated with the pathogenesis of MASH; however, its precise role remains obscure. Thus, we aimed to identify and validate efferocytosis linked signatures for detection of MASH. Methods We retrieved gene expression patterns of MASH from the GEO database and then focused on assessing the differential expression of efferocytosis-related genes (EFRGs) between MASH and control groups. This analysis was followed by a series of in-depth investigations, including protein-protein interaction (PPI), correlation analysis, and functional enrichment analysis, to uncover the molecular interactions and pathways at play. To screen for biomarkers for diagnosis, we applied machine learning algorithm to identify hub genes and constructed a clinical predictive model. Additionally, we conducted immune infiltration and single-cell transcriptome analyses in both MASH and control samples, providing insights into the immune cell landscape and cellular heterogeneity in these conditions. Results This research pinpointed 39 genes exhibiting a robust correlation with efferocytosis in MASH. Among these, five potential diagnostic biomarkers-TREM2, TIMD4, STAB1, C1QC, and DYNLT1-were screened using two distinct machine learning models. Subsequent external validation and animal experimentation validated the upregulation of TREM2 and downregulation of TIMD4 in MASH samples. Notably, both TREM2 and TIMD4 demonstrated area under the curve (AUC) values exceeding 0.9, underscoring their significant potential in facilitating the diagnosis of MASH. Conclusion Our study comprehensively elucidated the relationship between MASH and efferocytosis, constructing a favorable diagnostic model. Furthermore, we identified potential therapeutic targets for MASH treatment and offered novel insights into unraveling the underlying mechanisms of this disease.
Collapse
Affiliation(s)
- Chenghui Cao
- Department of Cardiology, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Wenwu Liu
- Department of Cardiology, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Guo
- Department of Cardiology, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuwei Weng
- Department of Cardiology, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yang Chen
- Department of Cardiology, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yonghong Luo
- Department of Cardiology, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuai Wang
- Department of Cardiology, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Botao Zhu
- Department of Cardiology, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yuxuan Liu
- Department of Cardiology, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Daoquan Peng
- Department of Cardiology, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
17
|
Chu YD, Chen MC, Yeh CT, Lai MW. Hijacking host extracellular vesicle machinery by hepatotropic viruses: current understandings and future prospects. J Biomed Sci 2024; 31:97. [PMID: 39369194 PMCID: PMC11453063 DOI: 10.1186/s12929-024-01063-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 06/25/2024] [Indexed: 10/07/2024] Open
Abstract
Recent advances in studies exploring the roles of extracellular vesicles (EVs) in viral transmission and replication have illuminated hepatotropic viruses, such as hepatitis A (HAV), hepatitis B (HBV), hepatitis C (HCV), hepatitis D (HDV), and hepatitis E (HEV). While previous investigations have uncovered these viruses' ability to exploit cellular EV pathways for replication and transmission, most have focused on the impacts of exosomal pathways. With an improved understanding of EVs, four main subtypes, including exosomes, microvesicles, large oncosomes, and apoptotic bodies, have been categorized based on size and biogenic pathways. However, there remains a noticeable gap in comprehensive reviews summarizing recent findings and outlining future perspectives for EV studies related to hepatotropic viruses. This review aims to consolidate insights into EV pathways utilized by hepatotropic viruses, offering guidance for the future research direction in this field. By comprehending the diverse range of hepatotropic virus-associated EVs and their role in cellular communication during productive viral infections, this review may offer valuable insights for targeting therapeutics and devising strategies to combat virulent hepatotropic virus infections and the associated incidence of liver cancer.
Collapse
Affiliation(s)
- Yu-De Chu
- Liver Research Center, Chang Gung Memorial Hospital, 5F., No. 15, Wenhua 1st Rd., Guishan Dist., Taoyuan City, 333, Taiwan
| | - Mi-Chi Chen
- Liver Research Center, Chang Gung Memorial Hospital, 5F., No. 15, Wenhua 1st Rd., Guishan Dist., Taoyuan City, 333, Taiwan
- Department of Pediatric, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial Hospital, 5F., No. 15, Wenhua 1st Rd., Guishan Dist., Taoyuan City, 333, Taiwan.
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| | - Ming-Wei Lai
- Liver Research Center, Chang Gung Memorial Hospital, 5F., No. 15, Wenhua 1st Rd., Guishan Dist., Taoyuan City, 333, Taiwan.
- Department of Pediatric, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| |
Collapse
|
18
|
Xie XD, Dong SS, Liu RJ, Shi LL, Zhu T. Mechanism of Efferocytosis in Determining Ischaemic Stroke Resolution-Diving into Microglia/Macrophage Functions and Therapeutic Modality. Mol Neurobiol 2024; 61:7583-7602. [PMID: 38409642 DOI: 10.1007/s12035-024-04060-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/17/2024] [Indexed: 02/28/2024]
Abstract
After ischaemic cerebral vascular injury, efferocytosis-a process known as the efficient clearance of apoptotic cells (ACs) by various phagocytes in both physiological and pathological states-is crucial for maintaining central nervous system (CNS) homeostasis and regaining prognosis. The mechanisms of efferocytosis in ischaemic stroke and its influence on preventing inflammation progression from secondary injury were still not fully understood, despite the fact that the fundamental process of efferocytosis has been described in a series of phases, including AC recognition, phagocyte engulfment, and subsequent degradation. The genetic reprogramming of macrophages and brain-resident microglia after an ischaemic stroke has been equated by some researchers to that of the peripheral blood and brain. Based on previous studies, some molecules, such as signal transducer and activator of transcription 6 (STAT6), peroxisome proliferator-activated receptor γ (PPARG), CD300A, and sigma non-opioid intracellular receptor 1 (SIGMAR1), were discovered to be largely associated with aspects of apoptotic cell elimination and accompanying neuroinflammation, such as inflammatory cytokine release, phenotype transformation, and suppressing of antigen presentation. Exacerbated stroke outcomes are brought on by defective efferocytosis and improper modulation of pertinent signalling pathways in blood-borne macrophages and brain microglia, which also results in subsequent tissue inflammatory damage. This review focuses on recent researches which contain a number of recently discovered mechanisms, such as studies on the relationship between benign efferocytosis and the regulation of inflammation in ischaemic stroke, the roles of some risk factors in disease progression, and current immune approaches that aim to promote efferocytosis to treat some autoimmune diseases. Understanding these pathways provides insight into novel pathophysiological processes and fresh characteristics, which can be used to build cerebral ischaemia targeting techniques.
Collapse
Affiliation(s)
- Xiao-Di Xie
- Department of Pathophysiology, School of Basic Medicine, Institute of Neuroregeneration & Neurorehabilitation, Qingdao University, No. 308 Ningxia Road, Qingdao, China
| | - Shan-Shan Dong
- Department of Pathophysiology, School of Basic Medicine, Institute of Neuroregeneration & Neurorehabilitation, Qingdao University, No. 308 Ningxia Road, Qingdao, China
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ru-Juan Liu
- Department of Pathophysiology, School of Basic Medicine, Institute of Neuroregeneration & Neurorehabilitation, Qingdao University, No. 308 Ningxia Road, Qingdao, China
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Liu-Liu Shi
- Department of Pathophysiology, School of Basic Medicine, Institute of Neuroregeneration & Neurorehabilitation, Qingdao University, No. 308 Ningxia Road, Qingdao, China
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ting Zhu
- Department of Pathophysiology, School of Basic Medicine, Institute of Neuroregeneration & Neurorehabilitation, Qingdao University, No. 308 Ningxia Road, Qingdao, China.
| |
Collapse
|
19
|
Lan W, Yang L, Tan X. Crosstalk between ferroptosis and macrophages: potential value for targeted treatment in diseases. Mol Cell Biochem 2024; 479:2523-2543. [PMID: 37880443 DOI: 10.1007/s11010-023-04871-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 10/05/2023] [Indexed: 10/27/2023]
Abstract
Ferroptosis is a newly identified form of programmed cell death that is connected to iron-dependent lipid peroxidization. It involves a variety of physiological processes involving iron metabolism, lipid metabolism, oxidative stress, and biosynthesis of nicotinamide adenine dinucleotide phosphate, glutathione, and coenzyme Q10. So far, it has been discovered to contribute to the pathological process of many diseases, such as myocardial infarction, acute kidney injury, atherosclerosis, and so on. Macrophages are innate immune system cells that regulate metabolism, phagocytize pathogens and dead cells, mediate inflammatory reactions, promote tissue repair, etc. Emerging evidence shows strong associations between macrophages and ferroptosis, which can provide us with a deeper comprehension of the pathological process of diseases and new targets for the treatments. In this review, we summarized the crosstalk between macrophages and ferroptosis and anatomized the application of this association in disease treatments, both non-neoplastic and neoplastic diseases. In addition, we have also addressed problems that remain to be investigated, in the hope of inspiring novel therapeutic strategies for diseases.
Collapse
Affiliation(s)
- Wanxin Lan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Lei Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Xuelian Tan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
20
|
Balena T, Staley K. Neuronal Death: Now You See It, Now You Don't. Neuroscientist 2024:10738584241282632. [PMID: 39316584 DOI: 10.1177/10738584241282632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Fatally injured neurons may necrose and rupture immediately, or they may initiate a programmed cell death pathway and then wait for microglial phagocytosis. Biochemical and histopathologic assays of neuronal death assess the numbers of neurons awaiting phagocytosis at a particular time point after injury. This number varies with the fraction of neurons that have necrosed vs initiated programmed cell death, the time elapsed since injury, the rate of phagocytosis, and the assay's ability to detect neurons at different stages of programmed cell death. Many of these variables can be altered by putatively neurotoxic and neuroprotective interventions independent of the effects on neuronal death. This complicates analyses of neurotoxicity and neuroprotection and has likely contributed to difficulties with clinical translation of neuroprotective strategies after brain injury. Time-resolved assays of neuronal health, such as ongoing expression of transgenic fluorescent proteins, are a useful means of avoiding these problems.
Collapse
Affiliation(s)
- Trevor Balena
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA
| | - Kevin Staley
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
21
|
Matthee A, Aghababaie Z, Nisbet LA, Dowrick JM, Windsor JA, Sands GB, Angeli-Gordon TR. Pulsed-field ablation: an alternative ablative method for gastric electrophysiological intervention. Am J Physiol Gastrointest Liver Physiol 2024; 327:G456-G465. [PMID: 39010831 DOI: 10.1152/ajpgi.00124.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/02/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024]
Abstract
Pulsed-field ablation (PFA) is an emerging ablative technology that has been used successfully to eliminate cardiac arrhythmias. As a nonthermal technique, it has significant benefits over traditional radiofrequency ablation with improved target tissue specificity and reduced risk of adverse events during cardiac applications. We investigated whether PFA is safe for use in the stomach and whether it could modulate gastric slow waves. Female weaner pigs were fasted overnight before anesthesia was induced using tiletamine hydrochloride (50 mg·mL-1) and zolazepam hydrochloride (50 mg·mL-1) and maintained with propofol (Diprivan 2%, 0.2-0.4 mg·kg-1·min-1). Pulsed-field ablation was performed on their gastric serosa in vivo. Adjacent point lesions (n = 2-4) were used to create a linear injury using bipolar pulsed-field ablation consisting of 40 pulses (10 Hz frequency, 0.1 ms pulse width, 1,000 V amplitude). High-resolution electrical mapping defined baseline and postablation gastric slow-wave patterns. A validated five-point scale was used to evaluate tissue damage in hematoxylin and eosin-stained images. Results indicated that PFA successfully induced complete conduction blocks in all cases, with lesions through the entire thickness of the gastric muscle layers. Consistent postablation slow-wave patterns emerged immediately following ablation and persisted over the study period. Pulsed-field ablation induces rapid conduction blocks as a tool to modulate slow-wave patterns, indicating it may be suitable as an alternative to radiofrequency ablation.NEW & NOTEWORTHY Results show that pulsed-field ablation can serve as a gastric slow-wave intervention by preventing slow-wave propagation across the lesion site. Stable conduction blocks were established immediately following energy delivery, faster than previous examples of radiofrequency gastric ablation. Pulsed-field ablation may be an alternative for gastric slow-wave intervention, and further functional and posthealing studies are now warranted.
Collapse
Affiliation(s)
- Ashton Matthee
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Zahra Aghababaie
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Linley A Nisbet
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Jarrah M Dowrick
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - John A Windsor
- Department of Surgery, University of Auckland, Auckland, New Zealand
- Surgical and Translational Research Centre, University of Auckland, Auckland, New Zealand
| | - Gregory B Sands
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Timothy R Angeli-Gordon
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
- Department of Surgery, University of Auckland, Auckland, New Zealand
- Te Manawahoukura Rangahau Centre, Te Wānanga o Aotearoa, Te Awamutu, Aotearoa New Zealand
| |
Collapse
|
22
|
Saavedra PHV, Trzeciak AJ, Lipshutz A, Daman AW, O'Neal AJ, Liu ZL, Wang Z, Romero-Pichardo JE, Rojas WS, Zago G, van den Brink MRM, Josefowicz SZ, Lucas CD, Anderson CJ, Rudensky AY, Perry JSA. Broad-spectrum antibiotics disrupt homeostatic efferocytosis. Nat Metab 2024; 6:1682-1694. [PMID: 39122784 PMCID: PMC7616532 DOI: 10.1038/s42255-024-01107-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/11/2024] [Indexed: 08/12/2024]
Abstract
The clearance of apoptotic cells, termed efferocytosis, is essential for tissue homeostasis and prevention of autoimmunity1. Although past studies have elucidated local molecular signals that regulate homeostatic efferocytosis in a tissue2,3, whether signals arising distally also regulate homeostatic efferocytosis remains elusive. Here, we show that large peritoneal macrophage (LPM) display impairs efferocytosis in broad-spectrum antibiotics (ABX)-treated, vancomycin-treated and germ-free mice in vivo, all of which have a depleted gut microbiota. Mechanistically, the microbiota-derived short-chain fatty acid butyrate directly boosts efferocytosis efficiency and capacity in mouse and human macrophages, and rescues ABX-induced LPM efferocytosis defects in vivo. Bulk messenger RNA sequencing of butyrate-treated macrophages in vitro and single-cell messenger RNA sequencing of LPMs isolated from ABX-treated and butyrate-rescued mice reveals regulation of efferocytosis-supportive transcriptional programmes. Specifically, we find that the efferocytosis receptor T cell immunoglobulin and mucin domain containing 4 (TIM-4, Timd4) is downregulated in LPMs of ABX-treated mice but rescued by oral butyrate. We show that TIM-4 is required for the butyrate-induced enhancement of LPM efferocytosis capacity and that LPM efferocytosis is impaired beyond withdrawal of ABX. ABX-treated mice exhibit significantly worse disease in a mouse model of lupus. Our results demonstrate that homeostatic efferocytosis relies on distal metabolic signals and suggest that defective homeostatic efferocytosis may explain the link between ABX use and inflammatory disease4-7.
Collapse
Affiliation(s)
- Pedro H V Saavedra
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Biology, Northeastern University, Boston, MA, USA.
| | - Alissa J Trzeciak
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Allie Lipshutz
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew W Daman
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, Cornell University, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Anya J O'Neal
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zong-Lin Liu
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zhaoquan Wang
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Jesús E Romero-Pichardo
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Waleska Saitz Rojas
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Giulia Zago
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marcel R M van den Brink
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Steven Z Josefowicz
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, Cornell University, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Christopher D Lucas
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh BioQuarter, UK
- Institute for Regeneration and Repair, Edinburgh BioQuarter, UK
| | | | - Alexander Y Rudensky
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, Cornell University, New York, NY, USA
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Howard Hughes Medical Institute and Ludwig Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Justin S A Perry
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, Cornell University, New York, NY, USA.
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
23
|
Cooper KN, Potempa J, Bagaitkar J. Dying for a cause: The pathogenic manipulation of cell death and efferocytic pathways. Mol Oral Microbiol 2024; 39:165-179. [PMID: 37786286 PMCID: PMC10985052 DOI: 10.1111/omi.12436] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/21/2023] [Accepted: 09/21/2023] [Indexed: 10/04/2023]
Abstract
Cell death is a natural consequence of infection. However, although the induction of cell death was solely thought to benefit the pathogen, compelling data now show that the activation of cell death pathways serves as a nuanced antimicrobial strategy that couples pathogen elimination with the generation of inflammatory cytokines and the priming of innate and adaptive cellular immunity. Following cell death, the phagocytic uptake of the infected dead cell by antigen-presenting cells and the subsequent lysosomal fusion of the apoptotic body containing the pathogen serve as an important antimicrobial mechanism that furthers the development of downstream adaptive immune responses. Despite the complexity of regulated cell death pathways, pathogens are highly adept at evading them. Here, we provide an overview of the remarkable diversity of cell death and efferocytic pathways and discuss illustrative examples of virulence strategies employed by pathogens, including oral pathogens, to counter their activation and persist within the host.
Collapse
Affiliation(s)
- Kelley N Cooper
- Department of Immunology and Microbiology, University of Louisville, Louisville, KY
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY
| | - Jan Potempa
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Juhi Bagaitkar
- Center for Microbial Pathogenesis, Nationwide Children’s Hospital, Columbus, OH
- Department of Pediatrics, The Ohio State College of Medicine, Columbus, OH
| |
Collapse
|
24
|
Liu X, Guo L, Du J, Luo Z, Xu J, Bhawal UK, Li X, Liu Y. Macrophage-derived apoptotic bodies impair the osteogenic ability of osteoblasts in periodontitis. Oral Dis 2024; 30:3296-3307. [PMID: 37994174 DOI: 10.1111/odi.14808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 08/30/2023] [Accepted: 10/30/2023] [Indexed: 11/24/2023]
Abstract
OBJECTIVES Periodontitis is induced by the imbalance between osteoblast and osteoclast activity, which leads to periodontal tissue destruction. Macrophages play a vital role in periodontitis. However, the hypoxic periodontal environment will also induce macrophage apoptosis within a short time. Apoptotic bodies (ABs) are the major products generated from apoptotic cells, but whether macrophage-derived ABs play a regulatory role as their mother cells in periodontitis remains unknown. In the present study, we aimed to investigate the effects of ABs on osteoblasts. METHOD ABs derived from hypoxia-induced macrophages were co-cultured with osteoblasts and the impact of ABs on osteoblast differentiation in vitro was assessed. In vivo, periodontitis model was established and macrophages-derived ABs were injected into the gingival sulcus. The effects of ABs on periodontal bone resorption were determined. RESULTS The results showed that ABs significantly inhibit osteoblast differentiation and promoted alveolar bone resorption in periodontitis. MicroRNA (miRNAs) array analysis was performed and revealed that miR-483-5p is the key miRNA in ABs. Dual luciferase reporter assays were performed and confirmed that miR-483-5p targeted Col1A1 mRNA and attenuated its expression. CONCLUSION Macrophage-derived ABs inhibit osteoblast differentiation via the transfer of miR-483-5p, which downregulates Col1A1 expression and finally suppresses osteogenic activity.
Collapse
Affiliation(s)
- Xu Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Lijia Guo
- Department of Orthodontics School of Stomatology, Capital Medical University, Beijing, China
| | - Juan Du
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Zhenhua Luo
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Junji Xu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Ujjal Kumar Bhawal
- Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Matsudo, Chiba, Japan
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Xiaoyan Li
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
25
|
Ryan AT, Kim M, Lim K. Immune Cell Migration to Cancer. Cells 2024; 13:844. [PMID: 38786066 PMCID: PMC11120175 DOI: 10.3390/cells13100844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 04/27/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Immune cell migration is required for the development of an effective and robust immune response. This elegant process is regulated by both cellular and environmental factors, with variables such as immune cell state, anatomical location, and disease state that govern differences in migration patterns. In all cases, a major factor is the expression of cell surface receptors and their cognate ligands. Rapid adaptation to environmental conditions partly depends on intrinsic cellular immune factors that affect a cell's ability to adjust to new environment. In this review, we discuss both myeloid and lymphoid cells and outline key determinants that govern immune cell migration, including molecules required for immune cell adhesion, modes of migration, chemotaxis, and specific chemokine signaling. Furthermore, we summarize tumor-specific elements that contribute to immune cell trafficking to cancer, while also exploring microenvironment factors that can alter these cellular dynamics within the tumor in both a pro and antitumor fashion. Specifically, we highlight the importance of the secretome in these later aspects. This review considers a myriad of factors that impact immune cell trajectory in cancer. We aim to highlight the immunotherapeutic targets that can be harnessed to achieve controlled immune trafficking to and within tumors.
Collapse
Affiliation(s)
- Allison T. Ryan
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (A.T.R.); (M.K.)
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (A.T.R.); (M.K.)
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | - Kihong Lim
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (A.T.R.); (M.K.)
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| |
Collapse
|
26
|
Lim JJY, Murata Y, Yuri S, Kitamuro K, Kawai T, Isotani A. Generating an organ-deficient animal model using a multi-targeted CRISPR-Cas9 system. Sci Rep 2024; 14:10636. [PMID: 38724644 PMCID: PMC11082136 DOI: 10.1038/s41598-024-61167-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 05/02/2024] [Indexed: 05/12/2024] Open
Abstract
Gene-knockout animal models with organ-deficient phenotypes used for blastocyst complementation are generally not viable. Animals need to be maintained as heterozygous mutants, and homozygous mutant embryos yield only one-fourth of all embryos. In this study, we generated organ-deficient embryos using the CRISPR-Cas9-sgRNAms system that induces cell death with a single-guide RNA (sgRNAms) targeting multiple sites in the genome. The Cas9-sgRNAms system interrupted cell proliferation and induced cell ablation in vitro. The mouse model had Cas9 driven by the Foxn1 promoter with a ubiquitous expression cassette of sgRNAms at the Rosa26 locus (Foxn1Cas9; Rosa26_ms). It showed an athymic phenotype similar to that of nude mice but was not hairless. Eventually, a rat cell-derived thymus in an interspecies chimera was generated by blastocyst complementation of Foxn1Cas9; Rosa26_ms mouse embryos with rat embryonic stem cells. Theoretically, a half of the total embryos has the Cas9-sgRNAms system because Rosa26_ms could be maintained as homozygous.
Collapse
Affiliation(s)
- Jonathan Jun-Yong Lim
- Laboratory of Organ Developmental Engineering, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, 630-0912, Japan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | - Yamato Murata
- Laboratory of Organ Developmental Engineering, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, 630-0912, Japan
| | - Shunsuke Yuri
- Laboratory of Organ Developmental Engineering, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, 630-0912, Japan
| | - Kohei Kitamuro
- Laboratory of Organ Developmental Engineering, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, 630-0912, Japan
| | - Taro Kawai
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, 630-0912, Japan
- Life Science Collaboration Center (LiSCo), Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, 630-0192, Japan
| | - Ayako Isotani
- Laboratory of Organ Developmental Engineering, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, 630-0912, Japan.
- Life Science Collaboration Center (LiSCo), Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, 630-0192, Japan.
| |
Collapse
|
27
|
Chavula T, To S, Smith J, Pedroza M, Nimri J, Devaraj S, Wenderfer S, Vogel TP, Agarwal SK. CADHERIN-11 regulation of myeloid phagocytes and autoimmune inflammation in murine lupus. J Autoimmun 2024; 145:103197. [PMID: 38447248 DOI: 10.1016/j.jaut.2024.103197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/15/2024] [Accepted: 02/23/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND AND OBJECTIVE Understanding the regulation of efferocytosis by myeloid phagocytes is important in identifying novel targets in systemic lupus erythematosus (SLE). Cadherin-11 (CDH11), a cell adhesion molecule, is implicated in inflammatory arthritis and fibrosis and recently been shown to regulate macrophage phagocytosis. The extent and mechanism of this regulation is unknown. Our objective was to examine the extent to which CDH11 regulates myeloid phagocytes and contributes to autoimmunity and tissue inflammation. METHODS We analyzed efferocytosis in macrophages and dendritic cells (DCs) from WT and Cdh11-/- mice and investigated the mechanisms in vitro. We investigated the role of CDH11 in disease development in vivo using the pristane induced lupus model. To translate the clinical relevance of CDH11 in human disease, we measured serum CDH11 levels in two independent pediatric SLE (pSLE) cohorts and healthy controls. RESULTS Using bone marrow derived macrophages (BMDMs) and DCs (BMDCs), we found impaired efferocytosis in phagocytes from Cdh11-/- mice, mediated by downregulated efferocytosis receptor expression and RhoGTPase activation. Specifically, loss of CDH11 downregulated Mertk expression and Rac1 activation in BMDMs, and integrin αVβ3 expression and Cdc42 activation in BMDCs, highlighting distinct pathways. In vivo, Cdh11-/- mice displayed defective efferocytosis and increased accumulation of apoptotic debris in pristane-induced lupus. Further, Cdh11-/- mice had enhanced systemic inflammation and autoimmune inflammation with increased anti-dsDNA autoantibodies, splenomegaly, type I interferons, and inflammatory cytokines. Paradoxically, at the tissue level, Cdh11-/- mice were protected against glomerulonephritis, indicating a dual role in murine lupus. Finally, SLE patients had increased serum CDH11 compared to controls. CONCLUSION This study highlights a novel role of CDH11 in regulating myeloid cells and efferocytosis and its potential as a contributor to development in autoimmunity murine lupus. Despite the increase in autoimmunity, Cdh11-/- mice developed decreased tissue inflammation and damage.
Collapse
Affiliation(s)
- Thandiwe Chavula
- Section of Immunology, Allergy and Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Sarah To
- Section of Immunology, Allergy and Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jennifer Smith
- Section of Immunology, Allergy and Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Mesias Pedroza
- Section of Immunology, Allergy and Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jena Nimri
- Section of Immunology, Allergy and Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Sridevi Devaraj
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA; Pathology Department, Texas Children's Hospital, Houston, TX, USA
| | - Scott Wenderfer
- Department of Pediatric Nephrology, B.C. Children's Hospital, Vancouver, BC, Canada
| | - Tiphanie P Vogel
- Section of Immunology, Allergy and Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Department of Pediatrics-Rheumatology, Baylor College of Medicine, Houston, TX, USA; Division of Rheumatology, Texas Children's Hospital, Houston, TX, USA
| | - Sandeep K Agarwal
- Section of Immunology, Allergy and Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
28
|
Pang X, Liu X. Immune Dysregulation in Chronic Obstructive Pulmonary Disease. Immunol Invest 2024; 53:652-694. [PMID: 38573590 DOI: 10.1080/08820139.2024.2334296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a disease whose incidence increase with age and is characterised by chronic inflammation and significant immune dysregulation. Inhalation of toxic substances cause oxidative stress in the lung tissue as well as airway inflammation, under the recruitment of chemokines, immune cells gathered and are activated to play a defensive role. However, persistent inflammation damages the immune system and leads to immune dysregulation, which is mainly manifested in the reduction of the body's immune response to antigens, and immune cells function are impaired, further destroy the respiratory defensive system, leading to recurrent lower respiratory infections and progressive exacerbation of the disease, thus immune dysregulation play an important role in the pathogenesis of COPD. This review summarizes the changes of innate and adaptive immune-related cells during the pathogenesis of COPD, aiming to control COPD airway inflammation and improve lung tissue remodelling by regulating immune dysregulation, for further reducing the risk of COPD progression and opening new avenues of therapeutic intervention in COPD.
Collapse
Affiliation(s)
- Xichen Pang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gerontal Respiratory Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaoju Liu
- Department of Gerontal Respiratory Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
29
|
Yao X, Zhang L, Sun S, Fu A, Ge Y. Progress of research on the relationship between efferocytosis and tumor. Front Oncol 2024; 14:1361327. [PMID: 38655133 PMCID: PMC11035832 DOI: 10.3389/fonc.2024.1361327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
Tumors are genetic changes that develop in an organism as a result of many internal and external causes. They affect the biological behavior of cells, cause them to grow independently, and give rise to new, perpetually proliferating organisms. Recent research has supported the critical function of tumor-associated macrophages in the development, progression, and metastasis of tumors through efferocytosis. Yet, there is still much to learn about the mechanisms behind their contribution to tumor pathological processes. As a result, it's critical to actively investigate how cytosolic processes contribute to the growth of tumors and to create novel therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | | | - Yanlei Ge
- North China University of Science and Technology Affiliated Hospital, Tangshan, China
| |
Collapse
|
30
|
Krejčová G, Danielová A, Sehadová H, Dyčka F, Kubásek J, Moos M, Bajgar A. Macrophages play a nutritive role in post-metamorphic maturation in Drosophila. Development 2024; 151:dev202492. [PMID: 38456486 DOI: 10.1242/dev.202492] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/27/2024] [Indexed: 03/09/2024]
Abstract
In the body of multicellular organisms, macrophages play an indispensable role in maintaining tissue homeostasis by removing old, apoptotic and damaged cells. In addition, macrophages allow significant remodeling of body plans during embryonic morphogenesis, regeneration and metamorphosis. Although the huge amount of organic matter that must be removed during these processes represents a potential source of nutrients, their further use by the organism has not yet been addressed. Here, we document that, during metamorphosis, Drosophila larval adipose tissue is infiltrated by macrophages, which remove dying adipocytes by efferocytosis and engulf leaking RNA-protein granules and lipids. Consequently, the infiltrating macrophages transiently adopt the adipocyte-like metabolic profile to convert remnants of dying adipocytes to lipoproteins and storage peptides that nutritionally support post-metamorphic development. This process is fundamental for the full maturation of ovaries and the achievement of early fecundity of individuals. Whether macrophages play an analogous role in other situations of apoptotic cell removal remains to be elucidated.
Collapse
Affiliation(s)
- Gabriela Krejčová
- Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia, Ceske Budejovice, 37005, Czech Republic
| | - Adéla Danielová
- Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia, Ceske Budejovice, 37005, Czech Republic
| | - Hana Sehadová
- Institute of Entomology , Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, 37005, Czech Republic
| | - Filip Dyčka
- Department of Chemistry, Faculty of Science, University of South Bohemia, Ceske Budejovice, 37005, Czech Republic
| | - Jiří Kubásek
- Department of Experimental Plant Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice, 37005, Czech Republic
| | - Martin Moos
- Institute of Entomology , Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, 37005, Czech Republic
| | - Adam Bajgar
- Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia, Ceske Budejovice, 37005, Czech Republic
- Institute of Entomology , Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, 37005, Czech Republic
| |
Collapse
|
31
|
Deng W, Shang H, Tong Y, Liu X, Huang Q, He Y, Wu J, Ba X, Chen Z, Chen Y, Tang K. The application of nanoparticles-based ferroptosis, pyroptosis and autophagy in cancer immunotherapy. J Nanobiotechnology 2024; 22:97. [PMID: 38454419 PMCID: PMC10921615 DOI: 10.1186/s12951-024-02297-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 01/02/2024] [Indexed: 03/09/2024] Open
Abstract
Immune checkpoint blockers (ICBs) have been applied for cancer therapy and achieved great success in the field of cancer immunotherapy. Nevertheless, the broad application of ICBs is limited by the low response rate. To address this issue, increasing studies have found that the induction of immunogenic cell death (ICD) in tumor cells is becoming an emerging therapeutic strategy in cancer treatment, not only straightly killing tumor cells but also enhancing dying cells immunogenicity and activating antitumor immunity. ICD is a generic term representing different cell death modes containing ferroptosis, pyroptosis, autophagy and apoptosis. Traditional chemotherapeutic agents usually inhibit tumor growth based on the apoptotic ICD, but most tumor cells are resistant to the apoptosis. Thus, the induction of non-apoptotic ICD is considered to be a more efficient approach for cancer therapy. In addition, due to the ineffective localization of ICD inducers, various types of nanomaterials have been being developed to achieve targeted delivery of therapeutic agents and improved immunotherapeutic efficiency. In this review, we briefly outline molecular mechanisms of ferroptosis, pyroptosis and autophagy, as well as their reciprocal interactions with antitumor immunity, and then summarize the current progress of ICD-induced nanoparticles based on different strategies and illustrate their applications in the cancer therapy.
Collapse
Affiliation(s)
- Wen Deng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Haojie Shang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yonghua Tong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qiu Huang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jian Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuan Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
32
|
Cogno N, Bauer R, Durante M. Mechanistic model of radiotherapy-induced lung fibrosis using coupled 3D agent-based and Monte Carlo simulations. COMMUNICATIONS MEDICINE 2024; 4:16. [PMID: 38336802 PMCID: PMC10858213 DOI: 10.1038/s43856-024-00442-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Mechanistic modelling of normal tissue toxicities is unfolding as an alternative to the phenomenological normal tissue complication probability models. The latter, currently used in the clinics, rely exclusively on limited patient data and neglect spatial dose distribution information. Among the various approaches, agent-based models are appealing as they provide the means to include patient-specific parameters and simulate long-term effects in complex systems. However, Monte Carlo tools remain the state-of-the-art for modelling radiation transport and provide measurements of the delivered dose with unmatched precision. METHODS In this work, we develop and characterize a coupled 3D agent-based - Monte Carlo model that mechanistically simulates the onset of the radiation-induced lung fibrosis in an alveolar segment. To the best of our knowledge, this is the first such model. RESULTS Our model replicates extracellular matrix patterns, radiation-induced lung fibrosis severity indexes and functional subunits survivals that show qualitative agreement with experimental studies and are consistent with our past results. Moreover, in accordance with experimental results, higher functional subunits survival and lower radiation-induced lung fibrosis severity indexes are achieved when a 5-fractions treatment is simulated. Finally, the model shows increased sensitivity to more uniform protons dose distributions with respect to more heterogeneous ones from photon irradiation. CONCLUSIONS This study lays thus the groundwork for further investigating the effects of different radiotherapeutic treatments on the onset of radiation-induced lung fibrosis via mechanistic modelling.
Collapse
Affiliation(s)
- Nicolò Cogno
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, 64291, Darmstadt, Germany
- Institute for Condensed Matter Physics, Technische Universität Darmstadt, 64289, Darmstadt, Germany
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Roman Bauer
- Department of Computer Science, University of Surrey, Guildford, GU2 7XH, UK
| | - Marco Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, 64291, Darmstadt, Germany.
- Institute for Condensed Matter Physics, Technische Universität Darmstadt, 64289, Darmstadt, Germany.
- Department of Physics "Ettore Pancini", University Federico II, Naples, Italy.
| |
Collapse
|
33
|
Mihaila AC, Ciortan L, Tucureanu MM, Simionescu M, Butoi E. Anti-Inflammatory Neutrophils Reprogram Macrophages toward a Pro-Healing Phenotype with Increased Efferocytosis Capacity. Cells 2024; 13:208. [PMID: 38334600 PMCID: PMC10854927 DOI: 10.3390/cells13030208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/11/2024] [Accepted: 01/20/2024] [Indexed: 02/10/2024] Open
Abstract
Following myocardial infarction (MI), blood neutrophils quickly and extensively infiltrate the heart, where they are temporally polarized into pro-inflammatory (N1) and anti-inflammatory (N2) subpopulations. Neutrophil transmigration is rapidly followed by the accrual of macrophages (MACs), which are believed to undergo local phenotypic transformations from pro-inflammatory to pro-healing MACs that mediate inflammation resolution. We hypothesized that N2 neutrophils can reprogram MACs toward a healing phenotype with increased efferocytosis capacity. To examine this, human neutrophils isolated from healthy subjects were polarized in N1 and N2 neutrophils, and their secretome was added to human MACs derived from THP monocytes. The impact of neutrophil factors on macrophages was investigated using qPCR, ELISA, Western blot, immunofluorescence, or an efferocytosis assay. The results show that the MACs exposed to N2 neutrophil secretome exhibited (i) increased expression of the anti-inflammatory molecules CD206, TGF-β, and IL-10 and the nuclear factors associated with reparatory macrophages (PPARγ, Nur77, and KLF4); (ii) enhanced expression of efferocytosis receptors (MerTK, CD36, CX3CR1, and integrins αv/β5) and of the bridge molecules Mfage8 and Gas6; and (iii) enhanced efferocytosis. In conclusion, factors released by N2 neutrophils induce a pro-healing phenotype of MACs by upregulating anti-inflammatory molecules and efferocytosis receptors and ensuing the efferocytosis capacity. The data suggest that molecular therapy to foster N2 polarization, which boosts macrophages' pro-healing phenotype, could be a promising strategy to speed up inflammation resolution and tissue repair.
Collapse
Affiliation(s)
| | | | | | | | - Elena Butoi
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (A.C.M.); (L.C.); (M.S.)
| |
Collapse
|
34
|
Liu J, Dong J, Pei X. Apoptotic Extracellular Vesicles Derived from Human Umbilical Vein Endothelial Cells Promote Skin Repair by Enhancing Angiogenesis: From Death to Regeneration. Int J Nanomedicine 2024; 19:415-428. [PMID: 38250193 PMCID: PMC10799620 DOI: 10.2147/ijn.s441453] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/08/2024] [Indexed: 01/23/2024] Open
Abstract
Purpose The promotion of angiogenesis is an effective strategy for skin wound repair. While the transplantation of endothelial cells has shown promise in vascularization, the underlying mechanism remains unclear. Recent studies have suggested that transplanted cells undergo apoptosis in a short period and release apoptotic extracellular vesicles (ApoEVs) that may have therapeutic potential. Methods In this study, we isolated ApoEVs from human umbilical vein endothelial cells (HUVECs) and characterized their properties. In vitro, we assessed the effects of ApoEVs on the proliferation, migration, and differentiation of endothelial cells and fibroblasts. In vivo, we investigated the therapeutic role of ApoEVs-AT in full-thickness skin wounds, evaluating wound closure rate, re-epithelialization, granulation tissue formation, vascularization, scar area, and collagen 3(Col3)/collagen 1(Col 1) ratio. Results ApoEVs derived from HUVECs displayed typical characteristics. In vitro, ApoEVs significantly enhanced the proliferation, migration, tube formation, and expression of angiogenic-related genes in endothelial cells and slightly promoted the proliferation and migration of fibroblasts. In vivo, ApoEVs improved the wound closure rate, re-epithelialization, the formation of granulation tissue, and vascularization. Besides, ApoEVs reduced scar formation, accompanied by an increase in the Col 3/ Col 1 ratio. Conclusion Given their abundant source and effectiveness, this study provided a novel approach for angiogenesis in tissue regeneration and deepened the understanding of from death to regeneration.
Collapse
Affiliation(s)
- Jinzhao Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Jia Dong
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Xibo Pei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| |
Collapse
|
35
|
Wang Y, Li Y, Yang Y, Swift M, Zhang Z, Wu S, Sun Y, Yang K. In situ vaccination caused by diverse irradiation-driven cell death programs. Theranostics 2024; 14:1147-1167. [PMID: 38323315 PMCID: PMC10845208 DOI: 10.7150/thno.86004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 11/20/2023] [Indexed: 02/08/2024] Open
Abstract
Interest surrounding the effect of irradiation on immune activation has exponentially grown within the last decade. This includes work regarding mechanisms of the abscopal effect and the success achieved by combination of radiotherapy and immunotherapy. It is hypothesized that irradiation triggers the immune system to eliminate tumors by inducing tumor cells immunogenic cell death (ICD) in tumor cells. Activation of the ICD pathways can be exploited as an in situ vaccine. In this review, we provide fundamental knowledge of various forms of ICD caused by irradiation, describe the relationship between various cell death pathways and the immune activation effect driven by irradiation, and focus on the therapeutic value of exploiting these cell death programs in the context of irradiation. Furthermore, we summarize the immunomodulatory effect of different cell death programs on combinative radiotherapy and immunotherapy. In brief, differences in cell death programs significantly impact the irradiation-induced immune activation effect. Evaluating the transition between them will provide clues to develop new strategies for radiotherapy and its combination with immunotherapy.
Collapse
Affiliation(s)
- Yijun Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan 430048, China
| | - Yan Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan 430048, China
| | - Yuxin Yang
- University of Southern California, Department of Biochemistry and Molecular Medicine
| | - Michelle Swift
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Zhenyu Zhang
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, California 90095-1772, USA
| | - Shuhui Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yajie Sun
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan 430048, China
| | - Kunyu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan 430048, China
| |
Collapse
|
36
|
Ma F, Ghimire L, Ren Q, Fan Y, Chen T, Balasubramanian A, Hsu A, Liu F, Yu H, Xie X, Xu R, Luo HR. Gasdermin E dictates inflammatory responses by controlling the mode of neutrophil death. Nat Commun 2024; 15:386. [PMID: 38195694 PMCID: PMC10776763 DOI: 10.1038/s41467-023-44669-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 12/21/2023] [Indexed: 01/11/2024] Open
Abstract
Both lytic and apoptotic cell death remove senescent and damaged cells in living organisms. However, they elicit contrasting pro- and anti-inflammatory responses, respectively. The precise cellular mechanism that governs the choice between these two modes of death remains incompletely understood. Here we identify Gasdermin E (GSDME) as a master switch for neutrophil lytic pyroptotic death. The tightly regulated GSDME cleavage and activation in aging neutrophils are mediated by proteinase-3 and caspase-3, leading to pyroptosis. GSDME deficiency does not alter neutrophil overall survival rate; instead, it specifically precludes pyroptosis and skews neutrophil death towards apoptosis, thereby attenuating inflammatory responses due to augmented efferocytosis of apoptotic neutrophils by macrophages. In a clinically relevant acid-aspiration-induced lung injury model, neutrophil-specific deletion of GSDME reduces pulmonary inflammation, facilitates inflammation resolution, and alleviates lung injury. Thus, by controlling the mode of neutrophil death, GSDME dictates host inflammatory outcomes, providing a potential therapeutic target for infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Fengxia Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key Laboratory for Prevention and Control of Hematological Disease Treatment Related Infection, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences, Tianjin, China.
| | - Laxman Ghimire
- Department of Pathology, Dana-Farber/Harvard Cancer Center, PhD Program in Immunology, Harvard Medical School; Department of Laboratory Medicine, Boston Children's Hospital, Enders Research Building, Room 811, Boston, MA, 02115, USA
| | - Qian Ren
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key Laboratory for Prevention and Control of Hematological Disease Treatment Related Infection, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences, Tianjin, China
| | - Yuping Fan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key Laboratory for Prevention and Control of Hematological Disease Treatment Related Infection, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences, Tianjin, China
| | - Tong Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key Laboratory for Prevention and Control of Hematological Disease Treatment Related Infection, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences, Tianjin, China
| | - Arumugam Balasubramanian
- Department of Pathology, Dana-Farber/Harvard Cancer Center, PhD Program in Immunology, Harvard Medical School; Department of Laboratory Medicine, Boston Children's Hospital, Enders Research Building, Room 811, Boston, MA, 02115, USA
| | - Alan Hsu
- Department of Pathology, Dana-Farber/Harvard Cancer Center, PhD Program in Immunology, Harvard Medical School; Department of Laboratory Medicine, Boston Children's Hospital, Enders Research Building, Room 811, Boston, MA, 02115, USA
| | - Fei Liu
- Department of Pathology, Dana-Farber/Harvard Cancer Center, PhD Program in Immunology, Harvard Medical School; Department of Laboratory Medicine, Boston Children's Hospital, Enders Research Building, Room 811, Boston, MA, 02115, USA
| | - Hongbo Yu
- VA Boston Healthcare System, Department of Pathology and Laboratory Medicine, 1400 VFW Parkway, West Roxbury, MA, 02132, USA
| | - Xuemei Xie
- Department of Pathology, Dana-Farber/Harvard Cancer Center, PhD Program in Immunology, Harvard Medical School; Department of Laboratory Medicine, Boston Children's Hospital, Enders Research Building, Room 811, Boston, MA, 02115, USA
| | - Rong Xu
- Department of Pathology, Dana-Farber/Harvard Cancer Center, PhD Program in Immunology, Harvard Medical School; Department of Laboratory Medicine, Boston Children's Hospital, Enders Research Building, Room 811, Boston, MA, 02115, USA
| | - Hongbo R Luo
- Department of Pathology, Dana-Farber/Harvard Cancer Center, PhD Program in Immunology, Harvard Medical School; Department of Laboratory Medicine, Boston Children's Hospital, Enders Research Building, Room 811, Boston, MA, 02115, USA.
| |
Collapse
|
37
|
Abele TJ, Billman ZP, Li L, Harvest CK, Bryan AK, Magalski GR, Lopez JP, Larson HN, Yin XM, Miao EA. Apoptotic signaling clears engineered Salmonella in an organ-specific manner. eLife 2023; 12:RP89210. [PMID: 38055781 PMCID: PMC10699806 DOI: 10.7554/elife.89210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023] Open
Abstract
Pyroptosis and apoptosis are two forms of regulated cell death that can defend against intracellular infection. When a cell fails to complete pyroptosis, backup pathways will initiate apoptosis. Here, we investigated the utility of apoptosis compared to pyroptosis in defense against an intracellular bacterial infection. We previously engineered Salmonella enterica serovar Typhimurium to persistently express flagellin, and thereby activate NLRC4 during systemic infection in mice. The resulting pyroptosis clears this flagellin-engineered strain. We now show that infection of caspase-1 or gasdermin D deficient macrophages by this flagellin-engineered S. Typhimurium induces apoptosis in vitro. Additionally, we engineered S. Typhimurium to translocate the pro-apoptotic BH3 domain of BID, which also triggers apoptosis in macrophages in vitro. During mouse infection, the apoptotic pathway successfully cleared these engineered S. Typhimurium from the intestinal niche but failed to clear the bacteria from the myeloid niche in the spleen or lymph nodes. In contrast, the pyroptotic pathway was beneficial in defense of both niches. To clear an infection, cells may have specific tasks that they must complete before they die; different modes of cell death could initiate these 'bucket lists' in either convergent or divergent ways.
Collapse
Affiliation(s)
- Taylor J Abele
- Department of Integrative Immunobiology, Duke University School of MedicineDurhamUnited States
- Department of Molecular Genetics and Microbiology, Duke University School of MedicineDurhamUnited States
- Department of Cell Biology, Duke University School of MedicineDurhamUnited States
| | - Zachary P Billman
- Department of Integrative Immunobiology, Duke University School of MedicineDurhamUnited States
- Department of Molecular Genetics and Microbiology, Duke University School of MedicineDurhamUnited States
- Department of Cell Biology, Duke University School of MedicineDurhamUnited States
- Department of Microbiology and Immunology, University of North Carolina at Chapel HillChapel HillUnited States
| | - Lupeng Li
- Department of Integrative Immunobiology, Duke University School of MedicineDurhamUnited States
- Department of Molecular Genetics and Microbiology, Duke University School of MedicineDurhamUnited States
- Department of Cell Biology, Duke University School of MedicineDurhamUnited States
| | - Carissa K Harvest
- Department of Integrative Immunobiology, Duke University School of MedicineDurhamUnited States
- Department of Molecular Genetics and Microbiology, Duke University School of MedicineDurhamUnited States
- Department of Cell Biology, Duke University School of MedicineDurhamUnited States
- Department of Microbiology and Immunology, University of North Carolina at Chapel HillChapel HillUnited States
| | - Alexia K Bryan
- Department of Integrative Immunobiology, Duke University School of MedicineDurhamUnited States
- Department of Biomedical Engineering, Duke University Pratt School of EngineeringDurhamUnited States
| | - Gabrielle R Magalski
- Department of Microbiology and Immunology, University of North Carolina at Chapel HillChapel HillUnited States
| | - Joseph P Lopez
- Department of Microbiology and Immunology, University of North Carolina at Chapel HillChapel HillUnited States
| | - Heather N Larson
- Department of Integrative Immunobiology, Duke University School of MedicineDurhamUnited States
- Department of Molecular Genetics and Microbiology, Duke University School of MedicineDurhamUnited States
- Department of Cell Biology, Duke University School of MedicineDurhamUnited States
| | - Xiao-Ming Yin
- Department of Pathology and Laboratory Medicine, Tulane University School of MedicineNew OrleansUnited States
| | - Edward A Miao
- Department of Integrative Immunobiology, Duke University School of MedicineDurhamUnited States
- Department of Molecular Genetics and Microbiology, Duke University School of MedicineDurhamUnited States
- Department of Cell Biology, Duke University School of MedicineDurhamUnited States
| |
Collapse
|
38
|
Yang M, Harrison BR, Promislow DEL. Cellular age explains variation in age-related cell-to-cell transcriptome variability. Genome Res 2023; 33:1906-1916. [PMID: 37973195 PMCID: PMC10760448 DOI: 10.1101/gr.278144.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/12/2023] [Indexed: 11/19/2023]
Abstract
Organs and tissues age at different rates within a single individual. Such asynchrony in aging has been widely observed at multiple levels, from functional hallmarks, such as anatomical structures and physiological processes, to molecular endophenotypes, such as the transcriptome and metabolome. However, we lack a conceptual framework to understand why some components age faster than others. Just as demographic models explain why aging evolves, here we test the hypothesis that demographic differences among cell types, determined by cell-specific differences in turnover rate, can explain why the transcriptome shows signs of aging in some cell types but not others. Through analysis of mouse single-cell transcriptome data across diverse tissues and ages, we find that cellular age explains a large proportion of the variation in the age-related increase in transcriptome variance. We further show that long-lived cells are characterized by relatively high expression of genes associated with proteostasis and that the transcriptome of long-lived cells shows greater evolutionary constraint than short-lived cells. In contrast, in short-lived cell types, the transcriptome is enriched for genes associated with DNA repair. Based on these observations, we develop a novel heuristic model that explains how and why aging rates differ among cell types.
Collapse
Affiliation(s)
- Ming Yang
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington 98195, USA
| | - Benjamin R Harrison
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington 98195, USA
| | - Daniel E L Promislow
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington 98195, USA;
- Department of Biology, University of Washington, Seattle, Washington 98195, USA
| |
Collapse
|
39
|
Li X, Jiang Y, Liu X, Fu J, Du J, Luo Z, Xu J, Bhawal UK, Liu Y, Guo L. Mesenchymal stem cell-derived apoptotic bodies alleviate alveolar bone destruction by regulating osteoclast differentiation and function. Int J Oral Sci 2023; 15:51. [PMID: 38040672 PMCID: PMC10692139 DOI: 10.1038/s41368-023-00255-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/21/2023] [Accepted: 10/22/2023] [Indexed: 12/03/2023] Open
Abstract
Periodontitis is caused by overactive osteoclast activity that results in the loss of periodontal supporting tissue and mesenchymal stem cells (MSCs) are essential for periodontal regeneration. However, the hypoxic periodontal microenvironment during periodontitis induces the apoptosis of MSCs. Apoptotic bodies (ABs) are the major product of apoptotic cells and have been attracting increased attention as potential mediators for periodontitis treatment, thus we investigated the effects of ABs derived from MSCs on periodontitis. MSCs were derived from bone marrows of mice and were cultured under hypoxic conditions for 72 h, after which ABs were isolated from the culture supernatant using a multi-filtration system. The results demonstrate that ABs derived from MSCs inhibited osteoclast differentiation and alveolar bone resorption. miRNA array analysis showed that miR-223-3p is highly enriched in those ABs and is critical for their therapeutic effects. Targetscan and luciferase activity results confirmed that Itgb1 is targeted by miR-223-3p, which interferes with the function of osteoclasts. Additionally, DC-STAMP is a key regulator that mediates membrane infusion. ABs and pre-osteoclasts expressed high levels of DC-STAMP on their membranes, which mediates the engulfment of ABs by pre-osteoclasts. ABs with knock-down of DC-STAMP failed to be engulfed by pre-osteoclasts. Collectively, MSC-derived ABs are targeted to be engulfed by pre-osteoclasts via DC-STAMP, which rescued alveolar bone loss by transferring miR-223-3p to osteoclasts, which in turn led to the attenuation of their differentiation and bone resorption. These results suggest that MSC-derived ABs are promising therapeutic agents for the treatment of periodontitis.
Collapse
Grants
- National Key R&D Program of China (Grant NO. 2022YFC2504200), the National Nature Science Foundation of China (81991504 and 81974149), the Beijing Municipal Administration of Hospitals Clinical Medicine Development of Special Funding Support (ZYLX202121), Innovation Research Team Project of Beijing Stomatological Hospital, Capital Medical University (CXTD202202), the Beijing Municipal Administration of Hospitals’ Ascent Plan (DFL20181501)
- National Nature Science Foundation of China (82201052), Beijing Municipal Administration of Hospitals’ Youth Programme (QML20231505), the Beijing Stomatological Hospital, Capital Medical University Young Scientist Program (NO. YSP202103)
- Beijing Municipal Administration of Hospitals’ Youth Programme (QML20181501), Innovation Foundation of Beijing Stomatological Hospital, Capital Medical University (21-09-18)
Collapse
Affiliation(s)
- Xiaoyan Li
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Yiyang Jiang
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Xu Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Jingfei Fu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Juan Du
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Zhenhua Luo
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Junji Xu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Ujjal Kumar Bhawal
- Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Chiba, Japan.
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India.
| | - Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China.
| | - Lijia Guo
- Department of Orthodontics School of Stomatology, Capital Medical University, Beijing, China.
| |
Collapse
|
40
|
Yang J, Zhang X, Wang G, Ma S, Yu Y, Liao C, Wang Z, Liang C, Li M, Tian W, Liao L. ApoSEVs-Mediated Modulation of Versatile Target Cells Promotes Diabetic Wound Healing: Unveiling a Promising Strategy. Int J Nanomedicine 2023; 18:6955-6977. [PMID: 38026535 PMCID: PMC10676647 DOI: 10.2147/ijn.s436350] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/09/2023] [Indexed: 12/01/2023] Open
Abstract
Background Diabetic chronic wounds present a formidable challenge in clinical management, lacking effective treatment options. Mesenchymal stem cell (MSC) transplantation has emerged as a promising therapy for tissue repair and regeneration. However, transplanted MSCs often undergo rapid apoptosis, giving rise to heterogeneous extracellular vesicles (EVs), including apoptotic bodies (apoBDs) and apoptotic small extracellular vesicles (apoSEVs). The potential stimulatory role of these EVs in diabetic wound healing remains unknown. Methods In this study, we investigated the effects of apoSEVs derived from adipose-derived mesenchymal/stromal cells (ADSCs) on the recovery of diabetic wounds by modulating the function of versatile target cells. First, we characterized the apoSEVs and apoBDs derived from apoptotic ADSCs. Subsequently, we evaluated the effects of apoSEVs and apoBDs on macrophages, endothelial cells, and fibroblasts, three essential cell types in wound healing, under high-glucose conditions. Furthermore, we developed a gelatin methacryloyl (GelMA) hydrogel for the sustained release of apoSEVs and investigated its therapeutic effects on wound healing in type 2 diabetic mice in vivo. Results apoSEVs facilitated the polarization of M1 phenotype macrophages to M2 phenotype, promoted proliferation, migration, and tube formation of endothelial cells, and enhanced fibroblast proliferation and migration. However, apoBDs failed to improve the function of endothelial cells and fibroblasts. In vivo, the apoSEVs-loaded GelMA effectively promoted wound healing by facilitating collagen fiber deposition, angiogenesis, and immune regulation. Conclusion Our study elucidates the beneficial effects of apoSEVs on wound recovery in diabetes and introduces a novel strategy for diabetic wound treatment based on apoSEVs.
Collapse
Affiliation(s)
- Jian Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Xuanhao Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Guanyu Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Shixing Ma
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Yejia Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Chengcheng Liao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Zhuo Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Cheng Liang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Maojiao Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Li Liao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People’s Republic of China
| |
Collapse
|
41
|
Duszyc K, von Pein JB, Ramnath D, Currin-Ross D, Verma S, Lim F, Sweet MJ, Schroder K, Yap AS. Apical extrusion prevents apoptosis from activating an acute inflammatory program in epithelia. Dev Cell 2023; 58:2235-2248.e6. [PMID: 37647898 DOI: 10.1016/j.devcel.2023.08.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/20/2023] [Accepted: 08/07/2023] [Indexed: 09/01/2023]
Abstract
Apoptosis is traditionally considered to be an immunologically silent form of cell death. Multiple mechanisms exist to ensure that apoptosis does not stimulate the immune system to cause inflammation or autoimmunity. Against this expectation, we now report that epithelia are programmed to provoke, rather than suppress, inflammation in response to apoptosis. We found that an acute inflammatory response led by neutrophils occurs in zebrafish and cell culture when apoptotic epithelial cells cannot be expelled from the monolayer by apical extrusion. This reflects an intrinsic circuit where ATP released from apoptotic cells stimulates epithelial cells in the immediate vicinity to produce interleukin-8 (IL-8). Apical extrusion therefore prevents inappropriate epithelial inflammation by physically eliminating apoptotic cells before they can activate this pro-inflammatory circuit. This carries the implication that epithelia may be predisposed to inflammation, elicited by sporadic or induced apoptosis, if apical extrusion is compromised.
Collapse
Affiliation(s)
- Kinga Duszyc
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia.
| | - Jessica B von Pein
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Divya Ramnath
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Denni Currin-Ross
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Suzie Verma
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Fayth Lim
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Matthew J Sweet
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Kate Schroder
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Alpha S Yap
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia.
| |
Collapse
|
42
|
Qiu H, Shao Z, Wen X, Liu Z, Chen Z, Qu D, Ding X, Zhang L. Efferocytosis: An accomplice of cancer immune escape. Biomed Pharmacother 2023; 167:115540. [PMID: 37741255 DOI: 10.1016/j.biopha.2023.115540] [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: 08/06/2023] [Revised: 09/13/2023] [Accepted: 09/17/2023] [Indexed: 09/25/2023] Open
Abstract
The clearance of apoptotic cells by efferocytes such as macrophages and dendritic cells is termed as "efferocytosis", it plays critical roles in maintaining tissue homeostasis in multicellular organisms. Currently, available studies indicate that efferocytosis-related molecules and pathways are tightly associated with cancer development, metastasis and treatment resistance, efferocytosis also induces an immunosuppressive tumor microenvironment and assists cancer cells escape from immune surveillance. In this study, we reviewed the underlying mechanisms of efferocytosis in mediating the occurrence of cancer immune escape, and then emphatically summarized the strategies of using efferocytosis as therapeutic target to enhance the anti-tumor efficacies of immune checkpoint inhibitors, hoping to provide powerful evidences for more effective therapeutic regimens of malignant tumors.
Collapse
Affiliation(s)
- Hui Qiu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhiying Shao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xin Wen
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhengyang Liu
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ziqin Chen
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Debao Qu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xin Ding
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Longzhen Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
43
|
Long H, Lichtnekert J, Andrassy J, Schraml BU, Romagnani P, Anders HJ. Macrophages and fibrosis: how resident and infiltrating mononuclear phagocytes account for organ injury, regeneration or atrophy. Front Immunol 2023; 14:1194988. [PMID: 37868987 PMCID: PMC10587486 DOI: 10.3389/fimmu.2023.1194988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023] Open
Abstract
Mononuclear phagocytes (MP), i.e., monocytes, macrophages, and dendritic cells (DCs), are essential for immune homeostasis via their capacities to clear pathogens, pathogen components, and non-infectious particles. However, tissue injury-related changes in local microenvironments activate resident and infiltrating MP towards pro-inflammatory phenotypes that contribute to inflammation by secreting additional inflammatory mediators. Efficient control of injurious factors leads to a switch of MP phenotype, which changes the microenvironment towards the resolution of inflammation. In the same way, MP endorses adaptive structural responses leading to either compensatory hypertrophy of surviving cells, tissue regeneration from local tissue progenitor cells, or tissue fibrosis and atrophy. Under certain circumstances, MP contribute to the reversal of tissue fibrosis by clearance of the extracellular matrix. Here we give an update on the tissue microenvironment-related factors that, upon tissue injury, instruct resident and infiltrating MP how to support host defense and recover tissue function and integrity. We propose that MP are not intrinsically active drivers of organ injury and dysfunction but dynamic amplifiers (and biomarkers) of specific tissue microenvironments that vary across spatial and temporal contexts. Therefore, MP receptors are frequently redundant and suboptimal targets for specific therapeutic interventions compared to molecular targets upstream in adaptive humoral or cellular stress response pathways that influence tissue milieus at a contextual level.
Collapse
Affiliation(s)
- Hao Long
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University (LMU), Munich, Germany
- Department of Urology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Julia Lichtnekert
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Joachim Andrassy
- Department of General, Visceral and Transplant Surgery, University Hospital of Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Barbara U. Schraml
- Institute for Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig-Maximilians-University (LMU), Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Paola Romagnani
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio”, University of Firenze, Nephrology and Dialysis Unit, Meyer Children’s Hospital, Firenze, Italy
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University (LMU), Munich, Germany
| |
Collapse
|
44
|
Ravichandran KS. Phagocytic clearance of dying cells and its implications. Immunol Rev 2023; 319:4-6. [PMID: 37858307 DOI: 10.1111/imr.13285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023]
Affiliation(s)
- Kodi S Ravichandran
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Biomedical Molecular Biology, Ghent University, and the Inflammation Research Center, VIB, Ghent, Belgium
| |
Collapse
|
45
|
Schilperoort M, Ngai D, Sukka SR, Avrampou K, Shi H, Tabas I. The role of efferocytosis-fueled macrophage metabolism in the resolution of inflammation. Immunol Rev 2023; 319:65-80. [PMID: 37158427 PMCID: PMC10615666 DOI: 10.1111/imr.13214] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/20/2023] [Indexed: 05/10/2023]
Abstract
The phagocytosis of dying cells by macrophages, termed efferocytosis, is a tightly regulated process that involves the sensing, binding, engulfment, and digestion of apoptotic cells. Efferocytosis not only prevents tissue necrosis and inflammation caused by secondary necrosis of dying cells, but it also promotes pro-resolving signaling in macrophages, which is essential for tissue resolution and repair following injury or inflammation. An important factor that contributes to this pro-resolving reprogramming is the cargo that is released from apoptotic cells after their engulfment and phagolysosomal digestion by macrophages. The apoptotic cell cargo contains amino acids, nucleotides, fatty acids, and cholesterol that function as metabolites and signaling molecules to bring about this re-programming. Here, we review efferocytosis-induced changes in macrophage metabolism that mediate the pro-resolving functions of macrophages. We also discuss various strategies, challenges, and future perspectives related to drugging efferocytosis-fueled macrophage metabolism as strategy to dampen inflammation and promote resolution in chronic inflammatory diseases.
Collapse
Affiliation(s)
- Maaike Schilperoort
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - David Ngai
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Santosh R Sukka
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kleopatra Avrampou
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Hongxue Shi
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Physiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
46
|
Gregory CD. Hijacking homeostasis: Regulation of the tumor microenvironment by apoptosis. Immunol Rev 2023; 319:100-127. [PMID: 37553811 PMCID: PMC10952466 DOI: 10.1111/imr.13259] [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: 04/25/2023] [Accepted: 07/18/2023] [Indexed: 08/10/2023]
Abstract
Cancers are genetically driven, rogue tissues which generate dysfunctional, obdurate organs by hijacking normal, homeostatic programs. Apoptosis is an evolutionarily conserved regulated cell death program and a profoundly important homeostatic mechanism that is common (alongside tumor cell proliferation) in actively growing cancers, as well as in tumors responding to cytotoxic anti-cancer therapies. Although well known for its cell-autonomous tumor-suppressive qualities, apoptosis harbors pro-oncogenic properties which are deployed through non-cell-autonomous mechanisms and which generally remain poorly defined. Here, the roles of apoptosis in tumor biology are reviewed, with particular focus on the secreted and fragmentation products of apoptotic tumor cells and their effects on tumor-associated macrophages, key supportive cells in the aberrant homeostasis of the tumor microenvironment. Historical aspects of cell loss in tumor growth kinetics are considered and the impact (and potential impact) on tumor growth of apoptotic-cell clearance (efferocytosis) as well as released soluble and extracellular vesicle-associated factors are discussed from the perspectives of inflammation, tissue repair, and regeneration programs. An "apoptosis-centric" view is proposed in which dying tumor cells provide an important platform for intricate intercellular communication networks in growing cancers. The perspective has implications for future research and for improving cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Christopher D. Gregory
- Centre for Inflammation ResearchInstitute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarterEdinburghUK
| |
Collapse
|
47
|
Cao L, Ma L, Zhao J, Wang X, Fang X, Li W, Qi Y, Tang Y, Liu J, Peng S, Yang L, Zhou L, Li L, Hu X, Ji Y, Hou Y, Zhao Y, Zhang X, Zhao YY, Zhao Y, Wei Y, Malik AB, Saiyin H, Xu J. An unexpected role of neutrophils in clearing apoptotic hepatocytes in vivo. eLife 2023; 12:RP86591. [PMID: 37728612 PMCID: PMC10511239 DOI: 10.7554/elife.86591] [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] [Indexed: 09/21/2023] Open
Abstract
Billions of apoptotic cells are removed daily in a human adult by professional phagocytes (e.g. macrophages) and neighboring nonprofessional phagocytes (e.g. stromal cells). Despite being a type of professional phagocyte, neutrophils are thought to be excluded from apoptotic sites to avoid tissue inflammation. Here, we report a fundamental and unexpected role of neutrophils as the predominant phagocyte responsible for the clearance of apoptotic hepatic cells in the steady state. In contrast to the engulfment of dead cells by macrophages, neutrophils burrowed directly into apoptotic hepatocytes, a process we term perforocytosis, and ingested the effete cells from the inside. The depletion of neutrophils caused defective removal of apoptotic bodies, induced tissue injury in the mouse liver, and led to the generation of autoantibodies. Human autoimmune liver disease showed similar defects in the neutrophil-mediated clearance of apoptotic hepatic cells. Hence, neutrophils possess a specialized immunologically silent mechanism for the clearance of apoptotic hepatocytes through perforocytosis, and defects in this key housekeeping function of neutrophils contribute to the genesis of autoimmune liver disease.
Collapse
Affiliation(s)
- Luyang Cao
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduChina
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL)GuangzhouChina
| | - Lixiang Ma
- Department of Anatomy, Histology & Embryology, Shanghai Medical CollegeShanghaiChina
| | - Juan Zhao
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduChina
| | - Xiangyu Wang
- Department of Anatomy, Histology & Embryology, Shanghai Medical CollegeShanghaiChina
| | - Xinzou Fang
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduChina
| | - Wei Li
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL)GuangzhouChina
| | - Yawen Qi
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL)GuangzhouChina
| | - Yingkui Tang
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduChina
| | - Jieya Liu
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduChina
| | - Shengxian Peng
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduChina
| | - Li Yang
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduChina
| | - Liangxue Zhou
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduChina
| | - Li Li
- Department of Anatomy, Histology & Embryology, Shanghai Medical CollegeShanghaiChina
| | - Xiaobo Hu
- Clinical Laboratory, Longhua Hospital, Shanghai University of Traditional MedicineShanghaiChina
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital Fudan UniversityShanghaiChina
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital Fudan UniversityShanghaiChina
| | - Yi Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan UniversityChengduChina
| | - Xianming Zhang
- Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, and Department of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of MedicineChicagoUnited States
| | - You-yang Zhao
- Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, and Department of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of MedicineChicagoUnited States
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of SciencesBeijingChina
| | - Yuquan Wei
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduChina
| | - Asrar B Malik
- Department of Pharmacology, University of Illinois, College of MedicineChicagoUnited States
| | - Hexige Saiyin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan UniversityShanghaiChina
| | - Jingsong Xu
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
48
|
Hide I, Shiraki H, Masuda A, Maeda T, Kumagai M, Kunishige N, Yanase Y, Harada K, Tanaka S, Sakai N. P2Y 2 receptor mediates dying cell removal via inflammatory activated microglia. J Pharmacol Sci 2023; 153:55-67. [PMID: 37524455 DOI: 10.1016/j.jphs.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/04/2023] [Accepted: 06/23/2023] [Indexed: 08/02/2023] Open
Abstract
Microglial removal of dying cells plays a beneficial role in maintaining homeostasis in the CNS, whereas under some pathological conditions, inflammatory microglia can cause excessive clearance, leading to neuronal death. However, the mechanisms underlying dying cell removal by inflammatory microglia remain poorly understood. In this study, we performed live imaging to examine the purinergic regulation of dying cell removal by inflammatory activated microglia. Lipopolysaccharide (LPS) stimulation induces rapid death of primary rat microglia, and the surviving microglia actively remove dying cells. The nonselective P2 receptor antagonist, suramin, inhibited dying cell removal to the same degree as that of the selective P2Y2 antagonist, AR-C118925. This inhibition was more potent in LPS-stimulated microglia than in non-stimulated ones. LPS stimulation elicited distribution of the P2Y2 receptor on the leading edge of the plasma membrane and then induced drastic upregulation of P2Y2 receptor mRNA expression in microglia. LPS stimulation caused upregulation of the dying cell-sensing inflammatory Axl phagocytic receptor, which was suppressed by blocking the P2Y2 receptor and its downstream signaling effector, proline-rich tyrosine kinase (Pyk2). Together, these results indicate that inflammatory stimuli may activate the P2Y2 receptor, thereby mediating dying cell removal, at least partially, through upregulating phagocytic Axl in microglia.
Collapse
Affiliation(s)
- Izumi Hide
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Hiroko Shiraki
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Akihiro Masuda
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Takuya Maeda
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Mayuka Kumagai
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Nao Kunishige
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Yuhki Yanase
- Department of Pharmacotherapy, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Kana Harada
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Shigeru Tanaka
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Norio Sakai
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan.
| |
Collapse
|
49
|
Abele TJ, Billman ZP, Li L, Harvest CK, Bryan AK, Magalski GR, Lopez JP, Larson HN, Yin XM, Miao EA. Apoptotic signaling clears engineered Salmonella in an organ-specific manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.06.539681. [PMID: 37205464 PMCID: PMC10187329 DOI: 10.1101/2023.05.06.539681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Pyroptosis and apoptosis are two forms of regulated cell death that can defend against intracellular infection. Although pyroptosis and apoptosis have distinct signaling pathways, when a cell fails to complete pyroptosis, backup pathways will initiate apoptosis. Here, we investigated the utility of apoptosis compared to pyroptosis in defense against an intracellular bacterial infection. We previously engineered Salmonella enterica serovar Typhimurium to persistently express flagellin, and thereby activate NLRC4 during systemic infection in mice. The resulting pyroptosis clears this flagellin-engineered strain. We now show that infection of caspase-1 or gasdermin D deficient macrophages by this flagellin-engineered S. Typhimurium induces apoptosis in vitro. Additionally, we also now engineer S. Typhimurium to translocate the pro-apoptotic BH3 domain of BID, which also triggers apoptosis in macrophages in vitro. In both engineered strains, apoptosis occurred somewhat slower than pyroptosis. During mouse infection, the apoptotic pathway successfully cleared these engineered S. Typhimurium from the intestinal niche, but failed to clear the bacteria in the myeloid niche in the spleen or lymph nodes. In contrast, the pyroptotic pathway was beneficial in defense of both niches. In order to clear an infection, distinct cell types may have specific tasks that they must complete before they die. In some cells, either apoptotic or pyroptotic signaling may initiate the same tasks, whereas in other cell types these modes of cell death may lead to different tasks that may not be identical in defense against infection. We recently suggested that such diverse tasks can be considered as different cellular 'bucket lists' to be accomplished before a cell dies.
Collapse
|
50
|
Schrodt MV, Behan-Bush RM, Liszewski JN, Humpal-Pash ME, Boland LK, Scroggins SM, Santillan DA, Ankrum JA. Efferocytosis of viable versus heat-inactivated MSC induces human monocytes to distinct immunosuppressive phenotypes. Stem Cell Res Ther 2023; 14:206. [PMID: 37592321 PMCID: PMC10433682 DOI: 10.1186/s13287-023-03443-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Immunomodulation by mesenchymal stromal cells (MSCs) can occur through trophic factor mechanisms, however, intravenously infused MSCs are rapidly cleared from the body yet a potent immunotherapeutic response is still observed. Recent work suggests that monocytes contribute to the clearance of MSCs via efferocytosis, the body's natural mechanism for clearing dead and dying cells in a non-inflammatory manner. This begs the questions of how variations in MSC quality affect monocyte phenotype and if viable MSCs are even needed to elicit an immunosuppressive response. METHODS Herein, we sought to dissect MSC's trophic mechanism from their efferocytic mechanisms and determine if the viability of MSCs prior to efferocytosis influences the resultant phenotype of monocytes. We cultured viable or heat-inactivated human umbilical cord MSCs with human peripheral blood mononuclear cells for 24 h and observed changes in monocyte surface marker expression and secretion profile. To isolate the effect of efferocytosis from MSC trophic factors, we used cell separation techniques to remove non-efferocytosed MSCs before challenging monocytes to suppress T-cells or respond to inflammatory stimuli. For all experiments, viable and heat-inactivated efferocytic-licensing of monocytes were compared to non-efferocytic-licensing control. RESULTS We found that monocytes efferocytose viable and heat-inactivated MSCs equally, but only viable MSC-licensed monocytes suppress activated T-cells and suppression occurred even after depletion of residual MSCs. This provides direct evidence that monocytes that efferocytose viable MSCs are immunosuppressive. Further characterization of monocytes after efferocytosis showed that uptake of viable-but not heat inactivated-MSC resulted in monocytes secreting IL-10 and producing kynurenine. When monocytes were challenged with LPS, IL-2, and IFN-γ to simulate sepsis, monocytes that had efferocytosed viable MSC had higher levels of IDO while monocytes that efferocytosed heat inactivated-MSCs produced the lowest levels of TNF-α. CONCLUSION Collectively, these studies show that the quality of MSCs efferocytosed by monocytes polarize monocytes toward distinctive immunosuppressive phenotypes and highlights the need to tailor MSC therapies for specific indications.
Collapse
Affiliation(s)
- Michael V Schrodt
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA
| | - Riley M Behan-Bush
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA
| | - Jesse N Liszewski
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA
| | - Madeleine E Humpal-Pash
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA
| | - Lauren K Boland
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA
| | - Sabrina M Scroggins
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Center for Immunology and Immune Based Diseases, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Biomedical Sciences, Center for Immunology, Center for Clinical and Translational Science, University of Minnesota School of Medicine, Duluth, MN, USA
| | - Donna A Santillan
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Center for Immunology and Immune Based Diseases, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Biomedical Sciences, Center for Immunology, Center for Clinical and Translational Science, University of Minnesota School of Medicine, Duluth, MN, USA
| | - James A Ankrum
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA.
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA.
- , 103 S. Capitol St., 5621 SC, Iowa City, IA, 52242, USA.
| |
Collapse
|