1
|
Bess SN, Igoe MJ, Muldoon TJ. The Physiological and Therapeutic Role of CD47 in Macrophage Function and Cancer. Immunol Invest 2025; 54:112-146. [PMID: 39415597 PMCID: PMC11774679 DOI: 10.1080/08820139.2024.2415409] [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: 10/19/2024]
Abstract
BACKGROUND Immunotherapy is an emerging strategy in cancer therapeutics aimed at modulating the immune system to inhibit pro-tumor pathways and increase a tumor's sensitivity to chemotherapy. Several clinically approved immunotherapy treatments, such as monoclonal antibody treatments, have been successful in solid tumors such as breast, colorectal, and pancreatic. However, an outstanding challenge of these strategies is tumor cell resistance. One target of interest for immune cell modulation is targeting macrophages that enter the tumor microenvironment. More specifically, an immune checkpoint of interest is CD47. CD47 is a transmembrane protein that inhibits phagocytic activity by acting as a "don't eat me" signal. In both mice and humans, healthy cells can express CD47, while solid malignancies like colorectal and breast cancer express it most strongly. METHODS Analysis of literature data on the physiological and functional roles of tissue-resident macrophages, along with the structure and mechanisms of action of the CD47 pathway was explored. We also explored how CD47 can influence different aspects of the tumor microenvironment (i.e. cellular metabolism and hypoxia) in addition to current clinical strategies and challenges associated with targeting CD47. RESULTS Overall, it was discovered that CD47 is overexpressed in a variety of cancer types in addition to normal tissue, making it a promising treatment regimen to enhance the capability of macrophages to phagocytose tumor cells. However, treatment efficacy is varied in pre-clinical and clinical models due to various challenges such as off-target effects. CONCLUSION This review emphasizes the diverse functionality of macrophages in normal and cancerous tissue, while also emphasizing the importance of macrophage targeting and their clinical significance.
Collapse
Affiliation(s)
- Shelby N. Bess
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701
| | - Matthew J. Igoe
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701
| | - Timothy J. Muldoon
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701
| |
Collapse
|
2
|
Ghorbanzadeh S, Khojini JY, Abouali R, Alimardan S, Zahedi M, Tahershamsi Z, Tajbakhsh A, Gheibihayat SM. Clearing the Path: Exploring Apoptotic Cell Clearance in Inflammatory and Autoimmune Disorders for Therapeutic Advancements. Mol Biotechnol 2024:10.1007/s12033-024-01222-6. [PMID: 38935260 DOI: 10.1007/s12033-024-01222-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 01/22/2024] [Indexed: 06/28/2024]
Abstract
Inflammatory and autoimmune disorders, characterized by dysregulated immune responses leading to tissue damage and chronic inflammation, present significant health challenges. This review uniquely focuses on efferocytosis-the phagocyte-mediated clearance of apoptotic cells-and its pivotal role in these disorders. We delve into the intricate mechanisms of efferocytosis' four stages and their implications in disease pathogenesis, distinguishing our study from previous literature. Our findings highlight impaired efferocytosis in conditions like atherosclerosis and asthma, proposing its targeting as a novel therapeutic strategy. We discuss the therapeutic potential of efferocytosis in modulating immune responses and resolving inflammation, offering a new perspective in treating inflammatory disorders.
Collapse
Affiliation(s)
- Shadi Ghorbanzadeh
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Science, Bandar Abbas, Iran
| | - Javad Yaghmoorian Khojini
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, P.O. Box: 8915173143, Yazd, IR, Iran
| | - Reza Abouali
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
| | - Sajad Alimardan
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Zahedi
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Tahershamsi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, P.O. Box: 8915173143, Yazd, IR, Iran.
- Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
3
|
Moon HG, Kim SJ, Kim KH, Kim YM, Rehman J, Lee H, Wu YC, Lee SSY, Christman JW, Ackerman SJ, Kim M, You S, Park GY. CX 3CR 1+ Macrophage Facilitates the Resolution of Allergic Lung Inflammation via Interacting CCL26. Am J Respir Crit Care Med 2023; 207:1451-1463. [PMID: 36790376 PMCID: PMC10263139 DOI: 10.1164/rccm.202209-1670oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 02/15/2023] [Indexed: 02/16/2023] Open
Abstract
Rationale: The resolution of inflammation is an active process coordinated by mediators and immune cells to restore tissue homeostasis. However, the mechanisms for resolving eosinophilic allergic lung inflammation triggered by inhaled allergens have not been fully elucidated. Objectives: Our objectives were to investigate the cellular mechanism of tissue-resident macrophages involved in the resolution process of eosinophilic lung inflammation. Methods: For the study, we used the institutional review board-approved protocol for human subsegmental bronchoprovocation with allergen, mouse models for allergic lung inflammation, and novel transgenic mice, including a conditional CCL26 knockout. The samples were analyzed using mass cytometry, single-cell RNA sequencing, and biophysical and immunological analyses. Measurements and Main Results: We compared alveolar macrophage (AM) subsets in the BAL before and after allergen provocation. In response to provocation with inhaled allergens, the subsets of AMs are dynamically changed in humans and mice. In the steady state, the AM subset expressing CX3CR1 is a relatively small fraction in bronchoalveolar space and lung tissue but drastically increases after allergen challenges. This subset presents unique patterns of gene expression compared with classical AMs, expressing high C1q family genes. CX3CR1+ macrophages are activated by airway epithelial cell-derived CCL26 via a receptor-ligand interaction. The binding of CCL26 to the CX3CR1+ receptor induces CX3CR1+ macrophages to secrete C1q, subsequently facilitating the clearance of eosinophils. Furthermore, the depletion of CX3CR1 macrophages or CCL26 in airway epithelial cells delays the resolution of allergic lung inflammation displaying prolonged tissue eosinophilia. Conclusions: These findings indicate that the CCL26-CX3CR1 pathway is pivotal in resolving eosinophilic allergic lung inflammation.
Collapse
Affiliation(s)
- Hyung-Geun Moon
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine
| | - Seung-jae Kim
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine
| | - Ki-Hyun Kim
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine
| | | | | | - Hyun Lee
- Department of Medicinal Chemistry & Pharmacognosy, Center for Biomolecular Sciences
| | | | | | - John W. Christman
- Section of Pulmonary, Critical Care, and Sleep Medicine, Davis Heart and Lung Research Center, The Ohio State University, Columbus, Ohio
| | - Steven J. Ackerman
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois
| | - Minhyung Kim
- Departments of Surgery, Cedars-Sinai Medical Center, Los Angeles, California; and
| | - Sungyoung You
- Departments of Surgery, Cedars-Sinai Medical Center, Los Angeles, California; and
| | - Gye Young Park
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| |
Collapse
|
4
|
Cell death in skin function, inflammation, and disease. Biochem J 2022; 479:1621-1651. [PMID: 35929827 PMCID: PMC9444075 DOI: 10.1042/bcj20210606] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 11/17/2022]
Abstract
Cell death is an essential process that plays a vital role in restoring and maintaining skin homeostasis. It supports recovery from acute injury and infection and regulates barrier function and immunity. Cell death can also provoke inflammatory responses. Loss of cell membrane integrity with lytic forms of cell death can incite inflammation due to the uncontrolled release of cell contents. Excessive or poorly regulated cell death is increasingly recognised as contributing to cutaneous inflammation. Therefore, drugs that inhibit cell death could be used therapeutically to treat certain inflammatory skin diseases. Programmes to develop such inhibitors are already underway. In this review, we outline the mechanisms of skin-associated cell death programmes; apoptosis, necroptosis, pyroptosis, NETosis, and the epidermal terminal differentiation programme, cornification. We discuss the evidence for their role in skin inflammation and disease and discuss therapeutic opportunities for targeting the cell death machinery.
Collapse
|
5
|
Mahmoudi A, Firouzjaei AA, Darijani F, Navashenaq JG, Taghizadeh E, Darroudi M, Gheibihayat SM. Effect of diabetes on efferocytosis process. Mol Biol Rep 2022; 49:10849-10863. [PMID: 35902446 DOI: 10.1007/s11033-022-07725-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/04/2022] [Accepted: 06/16/2022] [Indexed: 11/28/2022]
Abstract
Diabetes is a complex of genetic, metabolic, and autoimmune disorders that are characterized by hyperglycemia. Elevated apoptotic cell count following defective clearance of dead cells that can cause chronic inflammation is a hallmark of the diabetic wound. Effective dead cell clearance is a prerequisite for rapid inflammation resolution and successful recovery. Efferocytosis is a multistep process in which phagocytes engulf the dead cells. Cell body elimination is of great significance in disease and homeostasis. Recent research has clarified that diabetic wounds have an enhanced load of the apoptotic cell, which is partly attributed to the dysfunction of macrophages in apoptotic clearance at the site of the diabetic wounds. In the current work, we highlight the pathways implicated in efferocytosis, from the diagnosis of apoptotic cells to the phagocytic swallowing and the homeostatic resolution, and explain the possible pathophysiological episodes occurring when the proceeding is abrogated. Also, we describe the last development in the management of inflammation in diabetes wound and future directions of surveillance.
Collapse
Affiliation(s)
- Ali Mahmoudi
- Department of medical biotechnology and nanotechnology, faculty of medicine, Mashhad University of Medical science, Mashhad, Iran
| | - Ali Ahmadizad Firouzjaei
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Darijani
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Eskandar Taghizadeh
- Department of Medical Genetic, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Majid Darroudi
- Nuclear Medicine Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, P.O. Box: 8915173143, Yazd, Iran.
| |
Collapse
|
6
|
Efferocytosis in lung mucosae: implications for health and disease. Immunol Lett 2022; 248:109-118. [PMID: 35843361 DOI: 10.1016/j.imlet.2022.07.005] [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: 03/11/2022] [Revised: 05/15/2022] [Accepted: 07/11/2022] [Indexed: 11/21/2022]
Abstract
Efferocytosis is imperative to maintain lung homeostasis and control inflammation. Populations of lung macrophages are the main efferocytes in this tissue, responsible for controlling immune responses and avoiding unrestrained inflammation and autoimmunity through the expression of a plethora of receptors that recognize multiple 'eat me' signals on apoptotic cells. Efferocytosis is essentially anti-inflammatory and tolerogenic. However, in some situations, apoptotic cells phagocytosis can elicit inflammatory and immunogenic immune responses. Here, we summarized the current knowledge of the mechanisms of efferocytosis, and how any abnormality in this process may have an important contribution to the lung pathophysiology of many chronic inflammatory lung diseases such as asthma, acute lung injury, chronic obstructive pulmonary disease, and cystic fibrosis. Further, we consider the consequences of the dual role of efferocytosis on the susceptibility or resistance to pulmonary microbial infections. Understanding how efferocytosis works in different contexts will be useful to the development of new and more effective strategies to control the diversity of lung diseases.
Collapse
|
7
|
Tajbakhsh A, Yousefi F, Abedi SM, Rezaee M, Savardashtaki A, Teng Y, Sahebkar A. The cross-talk between soluble "Find me" and "Keep out" signals as an initial step in regulating efferocytosis. J Cell Physiol 2022; 237:3113-3126. [PMID: 35578547 DOI: 10.1002/jcp.30770] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/16/2022] [Accepted: 04/21/2022] [Indexed: 12/20/2022]
Abstract
The rapid clearance of apoptotic cells (ACs), known as efferocytosis, prompts the inhibition of inflammatory responses and autoimmunity and maintains homeostatic cell turnover by controlling the release of intracellular contents. The fast clearance of ACs requires professional and nonprofessional phagocytic cells that can accurately and promptly recognize ACs and migrate towards them. Cells undergoing apoptosis alarm their presence by releasing special soluble chemotactic factors, such as lactoferrin, that act as "Find me," "Keep out," or "Stay away" signals to recruit phagocytic cells, such as macrophages or prevent granulocyte migration. Efferocytosis effectively serves to prevent damage-associated molecular pattern release and secondary necrosis and inhibit inflammation/autoimmunity at the very first step. Since less attention has been given to the cross-talk and balance of "Find me" and "Keep out" signals released from ACs in efferocytosis, we set out to investigate the current knowledge of the roles of "Find me" and "Keep out" signals in the efferocytosis process.
Collapse
Affiliation(s)
- Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Yousefi
- Department of Biological Sciences, Faculty of Genetics, Tarbiat Modares University, Tehran, Iran
| | - Seyedeh M Abedi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Rezaee
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Amir Savardashtaki
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yong Teng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Medicine, The University of Western Australia, Perth, Western Australia, Australia.,Depatment of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
8
|
Kang MS, Kim SH, Yang MJ, Kim HY, Kim IH, Kang JW, Choi HS, Jin SW, Park EJ. Polyhexamethylene guanidine phosphate-induced necrosis may be linked to pulmonary fibrosis. Toxicol Lett 2022; 362:1-16. [DOI: 10.1016/j.toxlet.2022.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/05/2022] [Accepted: 03/24/2022] [Indexed: 10/18/2022]
|
9
|
Onishchenko NA, Gonikova ZZ, Nikolskaya AO, Kirsanova LA, Sevastianov VI. Programmed cell death and liver diseases. RUSSIAN JOURNAL OF TRANSPLANTOLOGY AND ARTIFICIAL ORGANS 2022; 24:72-88. [DOI: 10.15825/1995-1191-2022-1-72-88] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Cell death represents the most critical pathologic entity in liver disease, which dictates pathologic consequences such as inflammation, fibrosis, and cell transformation. We analyzed the conclusions of studies on the involvement of different types of programmed cell death (PCD) in the pathogenesis of liver diseases. Three main forms of PCD (autophagy, apoptosis, necrosis) and five additional, still insufficiently studied PCD – necroptosis, ferroptosis, pyroptosis, partanatosis and entosis – observed in the liver in various acute and chronic diseases are considered. The involvement of several PCD at once in the development of any one pathology and one type of PCD in different pathologies was established. This indicates the existence of cross-regulation of metabolism in the liver cells with different levels of damage in the formation of the main dominant type of PCD. Available results indicate the possibility of attenuation (correction) of functional and morphological manifestations of PCD in the organ by controlled blocking of effector-mediated PCD pathways, as well as targeted induction of autophagy, anti-apoptotic and anti-necrotic mechanisms in liver cells.
Collapse
Affiliation(s)
- N. A. Onishchenko
- Shumakov National Medical Research Center of Transplantology and Artificial Organs
| | - Z. Z. Gonikova
- Shumakov National Medical Research Center of Transplantology and Artificial Organs
| | - A. O. Nikolskaya
- Shumakov National Medical Research Center of Transplantology and Artificial Organs
| | - L. A. Kirsanova
- Shumakov National Medical Research Center of Transplantology and Artificial Organs
| | - V. I. Sevastianov
- Shumakov National Medical Research Center of Transplantology and Artificial Organs
| |
Collapse
|
10
|
Zhao J, Zhang W, Wu T, Wang H, Mao J, Liu J, Zhou Z, Lin X, Yan H, Wang Q. Efferocytosis in the Central Nervous System. Front Cell Dev Biol 2021; 9:773344. [PMID: 34926460 PMCID: PMC8678611 DOI: 10.3389/fcell.2021.773344] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/25/2021] [Indexed: 11/23/2022] Open
Abstract
The effective clearance of apoptotic cells is essential for maintaining central nervous system (CNS) homeostasis and restoring homeostasis after injury. In most cases of physiological apoptotic cell death, efferocytosis prevents inflammation and other pathological conditions. When apoptotic cells are not effectively cleared, destruction of the integrity of the apoptotic cell membrane integrity, leakage of intracellular contents, and secondary necrosis may occur. Efferocytosis is the mechanism by which efferocytes quickly remove apoptotic cells from tissues before they undergo secondary necrosis. Cells with efferocytosis functions, mainly microglia, help to eliminate apoptotic cells from the CNS. Here, we discuss the impacts of efferocytosis on homeostasis, the mechanism of efferocytosis, the associations of efferocytosis failure and CNS diseases, and the current clinical applications of efferocytosis. We also identify efferocytosis as a novel potential target for exploring the causes and treatments of CNS diseases.
Collapse
Affiliation(s)
- Jiayi Zhao
- Department of Anesthesia, Zhejiang Hospital, Hangzhou, China
| | - Weiqi Zhang
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Tingting Wu
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Hongyi Wang
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jialiang Mao
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jian Liu
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ziheng Zhou
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xianfeng Lin
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huige Yan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingqing Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
11
|
Lebo DPV, McCall K. Murder on the Ovarian Express: A Tale of Non-Autonomous Cell Death in the Drosophila Ovary. Cells 2021; 10:cells10061454. [PMID: 34200604 PMCID: PMC8228772 DOI: 10.3390/cells10061454] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/05/2021] [Accepted: 06/07/2021] [Indexed: 12/30/2022] Open
Abstract
Throughout oogenesis, Drosophila egg chambers traverse the fine line between survival and death. After surviving the ten early and middle stages of oogenesis, egg chambers drastically change their size and structure to produce fully developed oocytes. The development of an oocyte comes at a cost, the price is the lives of the oocyte’s 15 siblings, the nurse cells. These nurse cells do not die of their own accord. Their death is dependent upon their neighbors—the stretch follicle cells. Stretch follicle cells are nonprofessional phagocytes that spend the final stages of oogenesis surrounding the nurse cells and subsequently forcing the nurse cells to give up everything for the sake of the oocyte. In this review, we provide an overview of cell death in the ovary, with a focus on recent findings concerning this phagocyte-dependent non-autonomous cell death.
Collapse
|
12
|
Having an Old Friend for Dinner: The Interplay between Apoptotic Cells and Efferocytes. Cells 2021; 10:cells10051265. [PMID: 34065321 PMCID: PMC8161178 DOI: 10.3390/cells10051265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 02/02/2023] Open
Abstract
Apoptosis, the programmed and intentional death of senescent, damaged, or otherwise superfluous cells, is the natural end-point for most cells within multicellular organisms. Apoptotic cells are not inherently damaging, but if left unattended, they can lyse through secondary necrosis. The resulting release of intracellular contents drives inflammation in the surrounding tissue and can lead to autoimmunity. These negative consequences of secondary necrosis are avoided by efferocytosis—the phagocytic clearance of apoptotic cells. Efferocytosis is a product of both apoptotic cells and efferocyte mechanisms, which cooperate to ensure the rapid and complete removal of apoptotic cells. Herein, we review the processes used by apoptotic cells to ensure their timely removal, and the receptors, signaling, and cellular processes used by efferocytes for efferocytosis, with a focus on the receptors and signaling driving this process.
Collapse
|
13
|
Sprooten J, De Wijngaert P, Vanmeerbeerk I, Martin S, Vangheluwe P, Schlenner S, Krysko DV, Parys JB, Bultynck G, Vandenabeele P, Garg AD. Necroptosis in Immuno-Oncology and Cancer Immunotherapy. Cells 2020; 9:E1823. [PMID: 32752206 PMCID: PMC7464343 DOI: 10.3390/cells9081823] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/23/2020] [Accepted: 07/29/2020] [Indexed: 12/12/2022] Open
Abstract
Immune-checkpoint blockers (ICBs) have revolutionized oncology and firmly established the subfield of immuno-oncology. Despite this renaissance, a subset of cancer patients remain unresponsive to ICBs due to widespread immuno-resistance. To "break" cancer cell-driven immuno-resistance, researchers have long floated the idea of therapeutically facilitating the immunogenicity of cancer cells by disrupting tumor-associated immuno-tolerance via conventional anticancer therapies. It is well appreciated that anticancer therapies causing immunogenic or inflammatory cell death are best positioned to productively activate anticancer immunity. A large proportion of studies have emphasized the importance of immunogenic apoptosis (i.e., immunogenic cell death or ICD); yet, it has also emerged that necroptosis, a programmed necrotic cell death pathway, can also be immunogenic. Emergence of a proficient immune profile for necroptosis has important implications for cancer because resistance to apoptosis is one of the major hallmarks of tumors. Putative immunogenic or inflammatory characteristics driven by necroptosis can be of great impact in immuno-oncology. However, as is typical for a highly complex and multi-factorial disease like cancer, a clear cause versus consensus relationship on the immunobiology of necroptosis in cancer cells has been tough to establish. In this review, we discuss the various aspects of necroptosis immunobiology with specific focus on immuno-oncology and cancer immunotherapy.
Collapse
Affiliation(s)
- Jenny Sprooten
- Department of Cellular and Molecular Medicine, Laboratory of Cell Stress & Immunity (CSI), KU Leuven, 3000 Leuven, Belgium
| | - Pieter De Wijngaert
- Department of Cellular and Molecular Medicine, Laboratory of Cell Stress & Immunity (CSI), KU Leuven, 3000 Leuven, Belgium
| | - Isaure Vanmeerbeerk
- Department of Cellular and Molecular Medicine, Laboratory of Cell Stress & Immunity (CSI), KU Leuven, 3000 Leuven, Belgium
| | - Shaun Martin
- Department of Cellular and Molecular Medicine, Laboratory of Cellular Transport Systems, KU Leuven, 3000 Leuven, Belgium
| | - Peter Vangheluwe
- Department of Cellular and Molecular Medicine, Laboratory of Cellular Transport Systems, KU Leuven, 3000 Leuven, Belgium
| | - Susan Schlenner
- Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Dmitri V Krysko
- Department of Human Structure and Repair, Cell Death Investigation and Therapy Laboratory, Ghent University, 9000 Ghent, Belgium
- Department of Pathophysiology, Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia
| | - Jan B Parys
- Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Laboratory of Molecular and Cellular Signaling, KU Leuven, 3000 Leuven, Belgium
| | - Geert Bultynck
- Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Laboratory of Molecular and Cellular Signaling, KU Leuven, 3000 Leuven, Belgium
| | - Peter Vandenabeele
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
- VIB Center for Inflammation Research, 9052 Ghent, Belgium
- Methusalem Program, Ghent University, 9000 Ghent, Belgium
| | - Abhishek D Garg
- Department of Cellular and Molecular Medicine, Laboratory of Cell Stress & Immunity (CSI), KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
14
|
Cell death in chronic inflammation: breaking the cycle to treat rheumatic disease. Nat Rev Rheumatol 2020; 16:496-513. [PMID: 32641743 DOI: 10.1038/s41584-020-0455-8] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2020] [Indexed: 02/08/2023]
Abstract
Cell death is a vital process that occurs in billions of cells in the human body every day. This process helps maintain tissue homeostasis, supports recovery from acute injury, deals with infection and regulates immunity. Cell death can also provoke inflammatory responses, and lytic forms of cell death can incite inflammation. Loss of cell membrane integrity leads to the uncontrolled release of damage-associated molecular patterns (DAMPs), which are normally sequestered inside cells. Such DAMPs increase local inflammation and promote the production of cytokines and chemokines that modulate the innate immune response. Cell death can be both a consequence and a cause of inflammation, which can be difficult to distinguish in chronic diseases. Despite this caveat, excessive or poorly regulated cell death is increasingly recognized as a contributor to chronic inflammation in rheumatic disease and other inflammatory conditions. Drugs that inhibit cell death could, therefore, be used therapeutically for the treatment of these diseases, and programmes to develop such inhibitors are already underway. In this Review, we outline pathways for the major cell death programmes (apoptosis, necroptosis, pyroptosis and NETosis) and their potential roles in chronic inflammation. We also discuss current and developing therapies that target the cell death machinery.
Collapse
|
15
|
Radiation-induced bystander and abscopal effects: important lessons from preclinical models. Br J Cancer 2020; 123:339-348. [PMID: 32581341 PMCID: PMC7403362 DOI: 10.1038/s41416-020-0942-3] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 03/10/2020] [Accepted: 05/28/2020] [Indexed: 12/11/2022] Open
Abstract
Radiotherapy is a pivotal component in the curative treatment of patients with localised cancer and isolated metastasis, as well as being used as a palliative strategy for patients with disseminated disease. The clinical efficacy of radiotherapy has traditionally been attributed to the local effects of ionising radiation, which induces cell death by directly and indirectly inducing DNA damage, but substantial work has uncovered an unexpected and dual relationship between tumour irradiation and the host immune system. In clinical practice, it is, therefore, tempting to tailor immunotherapies with radiotherapy in order to synergise innate and adaptive immunity against cancer cells, as well as to bypass immune tolerance and exhaustion, with the aim of facilitating tumour regression. However, our understanding of how radiation impacts on immune system activation is still in its early stages, and concerns and challenges regarding therapeutic applications still need to be overcome. With the increasing use of immunotherapy and its common combination with ionising radiation, this review briefly delineates current knowledge about the non-targeted effects of radiotherapy, and aims to provide insights, at the preclinical level, into the mechanisms that are involved with the potential to yield clinically relevant combinatorial approaches of radiotherapy and immunotherapy.
Collapse
|
16
|
Meng F, Kwon S, Wang J, Yeo Y. Immunoactive drug carriers in cancer therapy. BIOMATERIALS FOR CANCER THERAPEUTICS 2020:53-94. [DOI: 10.1016/b978-0-08-102983-1.00003-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
17
|
Huang W, Fan Y, Cheng X, Liang H, Pan H, Xiao T, Chen M, Guan J. A preliminary Study on the Effect of Head and Neck Chemoradiotherapy on Systematic Immunity. Dose Response 2019; 17:1559325819884186. [PMID: 31695581 PMCID: PMC6820191 DOI: 10.1177/1559325819884186] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 09/06/2019] [Accepted: 09/24/2019] [Indexed: 12/24/2022] Open
Abstract
Background This study was designed initially to explore the effect of chemoradiotherapy on patients diagnosed with head and neck cancer (HNC) with respect to the alteration of systematic immunity. Methods We did a retrospective study enrolling patients received concurrent chemoradiotherapy (CCRT), with or without induction chemotherapy (IC). Blood tests were performed before IC, before and after CCRT. Flow cytometric analysis and turbidimetric inhibition immunoassay were used for detection. Results A total number of 58 patients were included from April 1, 2018, to March 31, 2019. Levels of immunoglobulins (Ig), including IgA, IgG, and IgM, declined after 2 to 3 cycles of IC and CCRT, respectively. Serum level of total hemolytic complement (CH50) increased (P < .001) after IC, but kept stably post-CCRT. Natural killer (NK) cells decreased (P < .01) after IC and enhanced (P < .001) post-CCRT. The number of CD3+CD4+ T cells got increased (P < .01) after IC and decreased (P < .001) post-CCRT. Consistently, both IC and CCRT induced the increase in CD3+CD8+ T cells significantly (P < .001 vs P < .01). Conclusion Both radiotherapy (RT) and chemotherapy (CT) induced dual effect of immune response. Concurrent chemoradiotherapy created an active immune response based on the effect induced by IC, suggesting that RT exerted a potential function on mobilizing immune system.
Collapse
Affiliation(s)
- Weiqiang Huang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yao Fan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoya Cheng
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Huazhen Liang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hua Pan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ting Xiao
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Min Chen
- Department of Radiation Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Jian Guan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
18
|
Abstract
Tissue macrophages rapidly recognize and engulf apoptotic cells. These events require the display of so-called eat-me signals on the apoptotic cell surface, the most fundamental of which is phosphatidylserine (PtdSer). Externalization of this phospholipid is catalysed by scramblase enzymes, several of which are activated by caspase cleavage. PtdSer is detected both by macrophage receptors that bind to this phospholipid directly and by receptors that bind to a soluble bridging protein that is independently bound to PtdSer. Prominent among the latter receptors are the MER and AXL receptor tyrosine kinases. Eat-me signals also trigger macrophages to engulf virus-infected or metabolically traumatized, but still living, cells, and this 'murder by phagocytosis' may be a common phenomenon. Finally, the localized presentation of PtdSer and other eat-me signals on delimited cell surface domains may enable the phagocytic pruning of these 'locally dead' domains by macrophages, most notably by microglia of the central nervous system.
Collapse
Affiliation(s)
- Greg Lemke
- Molecular Neurobiology Laboratory, Immunobiology and Microbial Pathogenesis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
19
|
Liu L, Sun B. Neutrophil pyroptosis: new perspectives on sepsis. Cell Mol Life Sci 2019; 76:2031-2042. [PMID: 30877336 PMCID: PMC11105444 DOI: 10.1007/s00018-019-03060-1] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 01/01/2023]
Abstract
Pyroptosis is a caspase-1 or caspase-4/5/11-dependent programmed cell death associated with inflammation, which is initiated by inflammasomes or cytosolic LPS in innate immunity. Sepsis is a life-threatening organ dysfunction caused by an imbalance in the body's response to infection. It is a complex interaction between the pathogen and the host's immune system. Neutrophils play the role of a double-edged sword in sepsis, and a number of studies have previously shown that regulation of neutrophils is the most crucial part of sepsis treatment. Pyroptosis is one of the important forms for neutrophils to function, which is increasingly understood as a host active immune response. There is ample evidence that neutrophil pyroptosis may play an important role in sepsis. In recent years, a breakthrough in pyroptosis research has revealed the main mechanism of pyroptosis. However, the potential value of neutrophil pyroptosis in the treatment of sepsis did not draw enough attention. A literature review was performed on the main mechanism of pyroptosis in sepsis and the potential value of neutrophils pyroptosis in sepsis, which may be suitable targets for sepsis treatment in future.
Collapse
Affiliation(s)
- Lu Liu
- Department of Burns and Plastic Surgery, Affiliated Hospital, Jiangsu University, 438 Jiefang Rd., Zhenjiang, 212001, Jiangsu, China
| | - Bingwei Sun
- Department of Burns and Plastic Surgery, Affiliated Hospital, Jiangsu University, 438 Jiefang Rd., Zhenjiang, 212001, Jiangsu, China.
| |
Collapse
|
20
|
Voices from the dead: The complex vocabulary and intricate grammar of dead cells. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2019; 116:1-90. [PMID: 31036289 DOI: 10.1016/bs.apcsb.2019.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Of the roughly one million cells per second dying throughout the body, the vast majority dies by apoptosis, the predominant form of regulated cell death in higher organisms. Long regarded as mere waste, apoptotic cells are now recognized as playing a prominent and active role in homeostatic maintenance, especially resolution of inflammation, and in the sculpting of tissues during development. The activities associated with apoptotic cells are continually expanding, with more recent studies demonstrating their ability to modulate such vital functions as proliferation, survival, differentiation, metabolism, migration, and angiogenesis. In each case, the role of apoptotic cells is active, exerting their effects via new activities acquired during the apoptotic program. Moreover, the capacity to recognize and respond to apoptotic cells is not limited to professional phagocytes. Most, if not all, cells receive and integrate an array of signals from cells dying in their vicinity. These signals comprise a form of biochemical communication. As reviewed in this chapter, this communication is remarkably sophisticated; each of its three critical steps-encoding, transmission, and decoding of the apoptotic cell's "message"-is endowed with exquisite robustness. Together, the abundance and intricacy of the variables at each step comprise the vocabulary and grammar of the language by which dead cells achieve their post-mortem voice. The combinatorial complexity of the resulting communication network permits dying cells, through the signals they emit and the responses those signals elicit, to partake of an expanded role in homeostasis, acting as both sentinels of environmental change and agents of adaptation.
Collapse
|
21
|
Three cell deaths and a funeral: macrophage clearance of cells undergoing distinct modes of cell death. Cell Death Discov 2019; 5:65. [PMID: 30774993 PMCID: PMC6368547 DOI: 10.1038/s41420-019-0146-x] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/11/2019] [Accepted: 01/14/2019] [Indexed: 02/07/2023] Open
Abstract
Macrophage clearance of apoptotic cells has been extensively investigated, but less is known regarding the clearance of cells dying by other forms of programmed cell death, e.g., necroptosis or ferroptosis. Here, we established a model of three different cell deaths using the same cell line and the occurrence of distinct cell death modalities was verified by using the specific inhibitors, zVAD-fmk, necrostatin-1, and ferrostatin-1, respectively. Cell death was characterized by using transmission electron microscopy (TEM), the gold standard for the demarcation of different cell death modalities. Moreover, using annexin V as a probe, we could detect surface exposure of phosphatidylserine (PS) in all three types of cell death, and this was confirmed by using specific anti-PS antibodies. We then co-cultured the cells with human monocyte-derived macrophages and found that cells dying by all three death modalities were engulfed by macrophages. Macrophage clearance of apoptotic cells was more efficient when compared to necroptotic and ferroptotic cells with multiple internalized target cells per macrophage, as shown by TEM. We propose that clearance of dying cells also should be taken into account in the classification of different cell death modalities.
Collapse
|
22
|
Krombach J, Hennel R, Brix N, Orth M, Schoetz U, Ernst A, Schuster J, Zuchtriegel G, Reichel CA, Bierschenk S, Sperandio M, Vogl T, Unkel S, Belka C, Lauber K. Priming anti-tumor immunity by radiotherapy: Dying tumor cell-derived DAMPs trigger endothelial cell activation and recruitment of myeloid cells. Oncoimmunology 2018; 8:e1523097. [PMID: 30546963 PMCID: PMC6287777 DOI: 10.1080/2162402x.2018.1523097] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 09/04/2018] [Accepted: 09/08/2018] [Indexed: 12/30/2022] Open
Abstract
The major goal of radiotherapy is the induction of tumor cell death. Additionally, radiotherapy can function as in situ cancer vaccination by exposing tumor antigens and providing adjuvants for anti-tumor immune priming. In this regard, the mode of tumor cell death and the repertoire of released damage-associated molecular patterns (DAMPs) are crucial. However, optimal dosing and fractionation of radiotherapy remain controversial. Here, we examined the initial steps of anti-tumor immune priming by different radiation regimens (20 Gy, 4 × 2 Gy, 2 Gy, 0 Gy) with cell lines of triple-negative breast cancer in vitro and in vivo. Previously, we have shown that especially high single doses (20 Gy) induce a delayed type of primary necrosis with characteristics of mitotic catastrophe and plasma membrane disintegration. Now, we provide evidence that protein DAMPs released by these dying cells stimulate sequential recruitment of neutrophils and monocytes in vivo. Key players in this regard appear to be endothelial cells revealing a distinct state of activation upon exposure to supernatants of irradiated tumor cells as characterized by high surface expression of adhesion molecules and production of a discrete cytokine/chemokine pattern. Furthermore, irradiated tumor cell-derived protein DAMPs enforced differentiation and maturation of dendritic cells as hallmarked by upregulation of co-stimulatory molecules and improved T cell-priming. Consistently, a recurring pattern was observed: The strongest effects were detected with 20 Gy-irradiated cells. Obviously, the initial steps of radiotherapy-induced anti-tumor immune priming are preferentially triggered by high single doses – at least in models of triple-negative breast cancer.
Collapse
Affiliation(s)
- Julia Krombach
- Department of Radiation Oncology, University Hospital, Munich, Germany
| | - Roman Hennel
- Department of Radiation Oncology, University Hospital, Munich, Germany
| | - Nikko Brix
- Department of Radiation Oncology, University Hospital, Munich, Germany
| | - Michael Orth
- Department of Radiation Oncology, University Hospital, Munich, Germany.,German Cancer Consortium (DKTK), Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ulrike Schoetz
- Department of Radiation Oncology, University Hospital, Munich, Germany.,Department of Radiotherapy and Radiooncology, Philipps-University Marburg, University Hospital Gießen and Marburg, Marburg, Germany
| | - Anne Ernst
- Department of Radiation Oncology, University Hospital, Munich, Germany.,Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Jessica Schuster
- Department of Radiation Oncology, University Hospital, Munich, Germany
| | - Gabriele Zuchtriegel
- Department of Otorhinolaryngology, University Hospital, Munich, Germany.,Walter Brendel Centre of Experimental Medicine, University Hospital, Munich, Germany.,Translational research in haematology/oncology, Institute of Experimental Infectious Diseases and Cancer Research, Division of the University Children's Hospital of Zurich, Zurich, Switzerland
| | - Christoph A Reichel
- Department of Otorhinolaryngology, University Hospital, Munich, Germany.,Walter Brendel Centre of Experimental Medicine, University Hospital, Munich, Germany
| | - Susanne Bierschenk
- Walter Brendel Centre of Experimental Medicine, University Hospital, Munich, Germany
| | - Markus Sperandio
- Walter Brendel Centre of Experimental Medicine, University Hospital, Munich, Germany
| | - Thomas Vogl
- Institute of Immunology, University of Münster, Münster, Germany
| | - Steffen Unkel
- Department of Medical Statistics, University Medical Center, Goettingen, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, Munich, Germany.,German Cancer Consortium (DKTK), Munich, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, University Hospital, Munich, Germany.,German Cancer Consortium (DKTK), Munich, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| |
Collapse
|
23
|
Tajbakhsh A, Rezaee M, Kovanen PT, Sahebkar A. Efferocytosis in atherosclerotic lesions: Malfunctioning regulatory pathways and control mechanisms. Pharmacol Ther 2018; 188:12-25. [DOI: 10.1016/j.pharmthera.2018.02.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
24
|
Trenti A, Boscaro C, Tedesco S, Cignarella A, Trevisi L, Bolego C. Effects of digitoxin on cell migration in ovarian cancer inflammatory microenvironment. Biochem Pharmacol 2018; 154:414-423. [DOI: 10.1016/j.bcp.2018.06.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/07/2018] [Indexed: 12/14/2022]
|
25
|
Abdolmaleki F, Farahani N, Gheibi Hayat SM, Pirro M, Bianconi V, Barreto GE, Sahebkar A. The Role of Efferocytosis in Autoimmune Diseases. Front Immunol 2018; 9:1645. [PMID: 30083153 PMCID: PMC6064952 DOI: 10.3389/fimmu.2018.01645] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 07/04/2018] [Indexed: 12/28/2022] Open
Abstract
Apoptosis happens continuously for millions of cells along with the active removal of apoptotic debris in order to maintain tissue homeostasis. In this respect, efferocytosis, i.e., the process of dead cell clearance, is orchestrated through cell exposure of a set of "find me," "eat me," and "tolerate me" signals facilitating the engulfment of dying cells through phagocytosis by macrophages and dendritic cells. The clearance of dead cells via phagocytes is of utmost importance to maintain the immune system tolerance to self-antigens. Accordingly, this biological activity prevents the release of autoantigens by dead cells, thus potentially suppressing the undesirable autoreactivity of immune cells and the appearance of inflammatory autoimmune disorders as systemic lupus erythematous and rheumatoid arthritis. In the present study, the apoptosis pathways and their immune regulation were reviewed. Moreover, efferocytosis process and its impairment in association with some autoimmune diseases were discussed.
Collapse
Affiliation(s)
- Fereshte Abdolmaleki
- Cellular and Molecular Research Center, School of Paramedical Sciences, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Najmeh Farahani
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Matteo Pirro
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Vanessa Bianconi
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - George E. Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
26
|
He X, Xiong C, Liu A, Zhao W, Xia X, Peng S, Li C, Zhou M, Li Y, Shi X, Shan Z, Teng W. Phagocytosis Deficiency of Macrophages in NOD.H-2 h4 Mice Accelerates the Severity of Iodine-Induced Autoimmune Thyroiditis. Biol Trace Elem Res 2018; 184:196-205. [PMID: 29052174 DOI: 10.1007/s12011-017-1183-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 10/02/2017] [Indexed: 01/15/2023]
Abstract
Apoptosis occurs in many autoimmune diseases. Excess iodine induces thyrocyte apoptosis and increases the incidence and prevalence of autoimmune thyroiditis (AIT). However, the sequence of events between the appearance of thyrocyte apoptosis and the occurrence of thyroiditis remains uncharacterized. Furthermore, few studies have investigated the role of macrophage phagocytosis in the development of AIT. Therefore, we evaluated the relationship between apoptosis and inflammatory infiltration in NOD.H-2h4 mouse thyroids by comparing the sequence of events in tissue samples. We also investigated the role of macrophages by comparing macrophage phagocytosis function in BALB/c, C57BL/6, and NOD.H-2h4 mice treated with different levels of iodine. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assays and thyroid inflammatory scores revealed that apoptosis (2 weeks) occurred before inflammatory infiltration (4 weeks). Phosphatidylserine (PS) expression on the extracellular surface of the cell membrane and double-stranded DNA fragments associated with apoptosis appeared at 2 and 8 weeks, respectively. Additionally, although apoptosis was enhanced in the thyroids of mice supplemented with excess iodine (0.05 ± 0.12 vs 1.63 ± 0.82% for BALB/c, 0.09 ± 0.14 vs 1.51 ± 0.34% for C57BL/6, and 0.07 ± 1.11 vs 4.72 ± 0.62% for NOD.H-2h4 mice), only NOD.H-2h4 mouse thyroids presented with inflammation. Furthermore, macrophages from NOD.H-2h4 mice (44.46 ± 1.79%) exhibited decreased phagocytotic activity relative to that in BALB/c (54.21 ± 4.58%) and C57BL/6 (58.96 ± 4.04%) mice. There were no differences in phagocytosis function between NOD.H-2h4 mice supplemented with excess iodine or left untreated (24.50 ± 2.66 vs 21.71 ± 1.79%, p = 0.06). In conclusion, deficiencies in the apoptosis clearance of macrophages in NOD.H-2h4 mice may constitute an early pathogenic mechanism in AIT that is not influenced by iodine intake.
Collapse
Affiliation(s)
- Xue He
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Chuhui Xiong
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Aihua Liu
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Wei Zhao
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Xinghai Xia
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Shiqiao Peng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Chenyan Li
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Mi Zhou
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, 17033, USA
| | - Yushu Li
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Xiaoguang Shi
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China.
| | - Zhongyan Shan
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Weiping Teng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
27
|
McGrath JJC, Stampfli MR. The immune system as a victim and aggressor in chronic obstructive pulmonary disease. J Thorac Dis 2018; 10:S2011-S2017. [PMID: 30023106 DOI: 10.21037/jtd.2018.05.63] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Joshua J C McGrath
- Medical Sciences Graduate Program, Firestone Institute for Respiratory Health at St. Joseph's Health Care, McMaster University, Hamilton, ON, Canada
| | - Martin R Stampfli
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Firestone Institute for Respiratory Health at St. Joseph's Health Care, McMaster University, Hamilton, ON, Canada.,Department of Medicine, Firestone Institute for Respiratory Health at St. Joseph's Health Care, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
28
|
Brix N, Tiefenthaller A, Anders H, Belka C, Lauber K. Abscopal, immunological effects of radiotherapy: Narrowing the gap between clinical and preclinical experiences. Immunol Rev 2018; 280:249-279. [PMID: 29027221 DOI: 10.1111/imr.12573] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Radiotherapy-despite being a local therapy that meanwhile is characterized by an impressively high degree of spatial accuracy-can stimulate systemic phenomena which occasionally lead to regression and rejection of non-irradiated, distant tumor lesions. These abscopal effects of local irradiation have been observed in sporadic clinical case reports since the beginning of the 20th century, and extensive preclinical work has contributed to identify systemic anti-tumor immune responses as the underlying driving forces. Although abscopal tumor regression still remains a rare event in the radiotherapeutic routine, increasing numbers of cases are being reported, particularly since the clinical implementation of immune checkpoint inhibiting agents. Accordingly, interests to systematically exploit the therapeutic potential of radiotherapy-stimulated systemic responses are constantly growing. The present review briefly delineates the history of radiotherapy-induced abscopal effects and the activation of systemic anti-tumor immune responses by local irradiation. We discuss preclinical and clinical reports with specific focus on the corresponding controversies, and we propose issues that should be addressed in the future in order to narrow the gap between preclinical knowledge and clinical experiences.
Collapse
Affiliation(s)
- Nikko Brix
- Department of Radiation Oncology, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Anna Tiefenthaller
- Department of Radiation Oncology, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Heike Anders
- Department of Radiation Oncology, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany.,German Cancer Consortium Partner Site München, Munich, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| |
Collapse
|
29
|
Muñoz LE, Leppkes M, Fuchs TA, Hoffmann M, Herrmann M. Missing in action-The meaning of cell death in tissue damage and inflammation. Immunol Rev 2018; 280:26-40. [PMID: 29027227 DOI: 10.1111/imr.12569] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Billions of cells die every day in higher organisms as part of the normal process of tissue homeostasis. During special conditions like in development, acute infections, mechanical injuries, and immunity, cell death is a common denominator and it exerts profound effects in the outcome of these scenarios. To prevent the accumulation of aged, superfluous, infected, damaged and dead cells, professional phagocytes act in a rapid and efficient manner to clear the battle field and avoid spread of the destruction. Neutrophils are the most abundant effector immune cells that extravasate into tissues and can turn injured tissues into gory battle fields. In peace times, neutrophils tend to patrol tissues without provoking inflammatory reactions. We discuss in this review actual and forgotten knowledge about the meaning of cell death during homeostatic processes and drive the attention to the importance of the action of neutrophils during patrolling and for the maintenance or recovery of the homeostatic state once the organism gets attacked or injured, respectively. In this fashion, we disclose several disease conditions that arise as collateral damage of physiological responses to death.
Collapse
Affiliation(s)
- Luis E Muñoz
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
| | - Moritz Leppkes
- Department of Internal Medicine 1 - Gastroenterology, Pulmonology and Endocrinology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
| | - Tobias A Fuchs
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Hoffmann
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
| | - Martin Herrmann
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
30
|
Feng L, Vujicic S, Dietrich ME, Litbarg N, Setty S, Antoni A, Rauch J, Levine JS. Repeated exposure of epithelial cells to apoptotic cells induces the specific selection of an adaptive phenotype: Implications for tumorigenesis. J Biol Chem 2018; 293:10245-10263. [PMID: 29769319 DOI: 10.1074/jbc.ra117.001290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 05/08/2018] [Indexed: 11/06/2022] Open
Abstract
The consequences of apoptosis extend beyond the mere death of the cell. We have shown that receptor-mediated recognition of apoptotic target cells by viable kidney proximal tubular epithelial cells (PTECs) inhibits PTEC proliferation, growth, and survival. Here, we tested the hypothesis that continual exposure to apoptotic targets can induce a phenotypic change in responding PTECs, as in other instances of natural selection. In particular, we demonstrate that repeated exposure to apoptotic targets leads to emergence of a PTEC line (denoted BU.MPTSEL) resistant to apoptotic target-induced death. Resistance is exquisitely specific. Not only are BU.MPTSEL responders fully resistant to apoptotic target-induced death (∼85% survival versus <10% survival of nonselected cells) but do so while retaining sensitivity to all other target-induced responses, including inhibition of proliferation and growth. Moreover, the resistance of BU.MPTSEL responders is specific to target-induced apoptosis, as apoptosis in response to other suicidal stimuli occurs normally. Comparison of the signaling events induced by apoptotic target exposure in selected versus nonselected responders indicated that the acquired resistance of BU.MPTSEL cells lies in a regulatory step affecting the generation of the pro-apoptotic protein, truncated BH3 interacting-domain death agonist (tBID), most likely at the level of BID cleavage by caspase-8. This specific adaptation has especial relevance for cancer, in which the prominence and persistence of cell death entail magnification of the post-mortem effects of apoptotic cells. Just as cancer cells acquire specific resistance to chemotherapeutic agents, we propose that cancer cells may also adapt to their ongoing exposure to apoptotic targets.
Collapse
Affiliation(s)
- Lanfei Feng
- From the Section of Nephrology, Department of Medicine, and.,the Section of Nephrology, Department of Medicine, Jesse Brown Veterans Affairs Hospital, Chicago, Illinois 60612
| | - Snezana Vujicic
- From the Section of Nephrology, Department of Medicine, and.,the Section of Nephrology, Department of Medicine, Jesse Brown Veterans Affairs Hospital, Chicago, Illinois 60612
| | | | - Natalia Litbarg
- From the Section of Nephrology, Department of Medicine, and.,the Section of Nephrology, Department of Medicine, Jesse Brown Veterans Affairs Hospital, Chicago, Illinois 60612
| | - Suman Setty
- Department of Pathology, University of Illinois, Chicago, Illinois 60612
| | - Angelika Antoni
- the Department of Biology, Kutztown University of Pennsylvania, Kutztown, Pennsylvania 19530, and
| | - Joyce Rauch
- the Division of Rheumatology, Department of Medicine, Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Jerrold S Levine
- From the Section of Nephrology, Department of Medicine, and .,the Section of Nephrology, Department of Medicine, Jesse Brown Veterans Affairs Hospital, Chicago, Illinois 60612
| |
Collapse
|
31
|
Abstract
Within an organism, environmental stresses can trigger cell death, particularly apoptotic cell death. Apoptotic cells, themselves, are potent regulators of their cellular environment, involved primarily in effecting homeostatic control. Tumors, especially, exist in a dynamic balance of cell proliferation and cell death. This special feature of the tumorous microenvironment—namely, the prominence and persistence of cell death—necessarily entails a magnification of the extrinsic, postmortem effects of dead cells. In both normal and malignant tissues, apoptotic regulation is exerted through immune as well as non-immune mechanisms. Apoptotic cells suppress the repertoire of immune reactivities, both by attenuating innate (especially inflammatory) responses and by abrogating adaptive responses. In addition, apoptotic cells modulate multiple vital cell activities, including survival, proliferation (cell number), and growth (cell size). While the microenvironment of the tumor may contribute to apoptosis, the postmortem effects of apoptotic cells feature prominently in the reciprocal acclimatization between the tumor and its environment. In much the same way that pathogens evade the host’s defenses through exploitation of key aspects of innate and adaptive immunity, cancer cells subvert several normal homeostatic processes, in particular wound healing and organ regeneration, to transform and overtake their environment. In understanding this subversion, it is crucial to view a tumor not simply as a clone of malignant cells, but rather as a complex and highly organized structure in which there exists a multidirectional flow of information between the cancer cells themselves and the multiple other cell types and extracellular matrix components of which the tumor is comprised. Apoptotic cells, therefore, have the unfortunate consequence of facilitating tumorigenesis and tumor survival.
Collapse
Affiliation(s)
- David S Ucker
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL, United States
| | - Jerrold S Levine
- Department of Medicine, Division of Nephrology, University of Illinois College of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States
| |
Collapse
|
32
|
Meng MB, Wang HH, Cui YL, Wu ZQ, Shi YY, Zaorsky NG, Deng L, Yuan ZY, Lu Y, Wang P. Necroptosis in tumorigenesis, activation of anti-tumor immunity, and cancer therapy. Oncotarget 2018; 7:57391-57413. [PMID: 27429198 PMCID: PMC5302997 DOI: 10.18632/oncotarget.10548] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/20/2016] [Indexed: 02/05/2023] Open
Abstract
While the mechanisms underlying apoptosis and autophagy have been well characterized over recent decades, another regulated cell death event, necroptosis, remains poorly understood. Elucidating the signaling networks involved in the regulation of necroptosis may allow this form of regulated cell death to be exploited for diagnosis and treatment of cancer, and will contribute to the understanding of the complex tumor microenvironment. In this review, we have summarized the mechanisms and regulation of necroptosis, the converging and diverging features of necroptosis in tumorigenesis, activation of anti-tumor immunity, and cancer therapy, as well as attempts to exploit this newly gained knowledge to provide therapeutics for cancer.
Collapse
Affiliation(s)
- Mao-Bin Meng
- Department of Radiation Oncology, Tianjin's Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Huan-Huan Wang
- Department of Radiation Oncology, Tianjin's Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Yao-Li Cui
- Department of Lymphoma, Tianjin's Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Zhi-Qiang Wu
- Department of Radiation Oncology, Tianjin's Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Yang-Yang Shi
- Stanford University School of Medicine, Stanford, CA, United States of America
| | - Nicholas G Zaorsky
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, United States of America
| | - Lei Deng
- Department of Thoracic Cancer and Huaxi Student Society of Oncology Research, West China Hospital, West China School of Medicine, Sichuan University, Sichuan Province, China
| | - Zhi-Yong Yuan
- Department of Radiation Oncology, Tianjin's Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - You Lu
- Department of Thoracic Cancer and Huaxi Student Society of Oncology Research, West China Hospital, West China School of Medicine, Sichuan University, Sichuan Province, China
| | - Ping Wang
- Department of Radiation Oncology, Tianjin's Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
33
|
Dallenga T, Repnik U, Corleis B, Eich J, Reimer R, Griffiths GW, Schaible UE. M. tuberculosis-Induced Necrosis of Infected Neutrophils Promotes Bacterial Growth Following Phagocytosis by Macrophages. Cell Host Microbe 2017; 22:519-530.e3. [DOI: 10.1016/j.chom.2017.09.003] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 05/27/2017] [Accepted: 09/01/2017] [Indexed: 01/05/2023]
|
34
|
Nainu F, Shiratsuchi A, Nakanishi Y. Induction of Apoptosis and Subsequent Phagocytosis of Virus-Infected Cells As an Antiviral Mechanism. Front Immunol 2017; 8:1220. [PMID: 29033939 PMCID: PMC5624992 DOI: 10.3389/fimmu.2017.01220] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 09/14/2017] [Indexed: 01/14/2023] Open
Abstract
Viruses are infectious entities that hijack host replication machineries to produce their progeny, resulting, in most cases, in disease and, sometimes, in death in infected host organisms. Hosts are equipped with an array of defense mechanisms that span from innate to adaptive as well as from humoral to cellular immune responses. We previously demonstrated that mouse cells underwent apoptosis in response to influenza virus infection. These apoptotic, virus-infected cells were then targeted for engulfment by macrophages and neutrophils. We more recently reported similar findings in the fruit fly Drosophila melanogaster, which lacks adaptive immunity, after an infection with Drosophila C virus. In these experiments, the inhibition of phagocytosis led to severe influenza pathologies in mice and early death in Drosophila. Therefore, the induction of apoptosis and subsequent phagocytosis of virus-infected cells appear to be an antiviral innate immune mechanism that is conserved among multicellular organisms. We herein discuss the underlying mechanisms and significance of the apoptosis-dependent phagocytosis of virus-infected cells. Investigations on the molecular and cellular features responsible for this underrepresented virus–host interaction may provide a promising avenue for the discovery of novel substances that are targeted in medical treatments against virus-induced intractable diseases.
Collapse
Affiliation(s)
- Firzan Nainu
- Laboratory of Pharmacology and Toxicology, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia.,Laboratory of Host Defense and Responses, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Akiko Shiratsuchi
- Laboratory of Host Defense and Responses, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yoshinobu Nakanishi
- Laboratory of Host Defense and Responses, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
35
|
Shen C, Li J, Zhang Y, Li Y, Shen G, Zhu J, Tao J. Polyethylenimine-based micro/nanoparticles as vaccine adjuvants. Int J Nanomedicine 2017; 12:5443-5460. [PMID: 28814862 PMCID: PMC5546778 DOI: 10.2147/ijn.s137980] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Vaccines have shown great success in treating and preventing tumors and infections, while adjuvants are always demanded to ensure potent immune responses. Polyethylenimine (PEI), as one of the well-studied cationic polymers, has been used as a transfection reagent for decades. However, increasing evidence has shown that PEI-based particles are also capable of acting as adjuvants. In this paper, we briefly review the physicochemical properties and the broad applications of PEI in different fields, and elaborate on the intracellular processes of PEI-based vaccines. In addition, we sum up the proof of their in vivo and clinical applications. We also highlight some mechanisms proposed for the intrinsic immunoactivation function of PEI, followed by the challenges and future perspectives of the applications of PEI in the vaccines, as well as some strategies to elicit the desirable immune responses.
Collapse
Affiliation(s)
- Chen Shen
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Li
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Zhang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuce Li
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Guanxin Shen
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jintao Zhu
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Tao
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
36
|
Endoplasmic Reticulum Stress, NRF2 Signalling and Cardiovascular Diseases in a Nutshell. Curr Atheroscler Rep 2017; 19:33. [DOI: 10.1007/s11883-017-0669-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
37
|
Trofimenko AS. Elimination of Nucleoproteins in Systemic Lupus Erythematosus and Antinuclear Autoantibodies Production. Lupus 2017. [DOI: 10.5772/intechopen.68496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
38
|
Chen J, Chaurio RA, Maueröder C, Derer A, Rauh M, Kost A, Liu Y, Mo X, Hueber A, Bilyy R, Herrmann M, Zhao Y, Muñoz LE. Inosine Released from Dying or Dead Cells Stimulates Cell Proliferation via Adenosine Receptors. Front Immunol 2017; 8:504. [PMID: 28496447 PMCID: PMC5406388 DOI: 10.3389/fimmu.2017.00504] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 04/12/2017] [Indexed: 02/05/2023] Open
Abstract
Introduction Many antitumor therapies induce apoptotic cell death in order to cause tumor regression. Paradoxically, apoptotic cells are also known to promote wound healing, cell proliferation, and tumor cell repopulation in multicellular organisms. We aimed to characterize the nature of the regenerative signals concentrated in the micromilieu of dead and dying cells. Methods Cultures of viable melanoma B16F10 cells, mouse fibroblasts, and primary human fibroblast-like synoviocytes (FLS) in the presence of dead and dying cells, their supernatants (SNs), or purified agonists and antagonists were used to evaluate the stimulation of proliferation. Viable cell quantification was performed by either flow cytometry of harvested cells or by crystal violet staining of adherent cells. High-performance liquid chromatography and liquid chromatography coupled with mass spectrometry of cell SNs were deployed to identify the nature of growth-promoting factors. Coimplantation of living cells in the presence of SNs collected from dead and dying cells and specific agonists was used to evaluate tumor growth in vivo. Results The stimulation of proliferation of few surviving cells by bystander dead cells was confirmed for melanoma cells, mouse fibroblasts, and primary FLS. We found that small soluble molecules present in the protein-free fraction of SNs of dead and dying cells were responsible for the promotion of proliferation. The nucleoside inosine released by dead and dying cells acting via adenosine receptors was identified as putative inducer of proliferation of surviving tumor cells after irradiation and heat treatment. Conclusion Inosine released by dead and dying cells mediates tumor cell proliferation via purinergic receptors. Therapeutic strategies surmounting this pathway may help to reduce the rate of recurrence after radio- and chemotherapy.
Collapse
Affiliation(s)
- Jin Chen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China.,Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ricardo A Chaurio
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christian Maueröder
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
| | - Anja Derer
- Department of Radiation Oncology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
| | - Manfred Rauh
- Kinder- und Jugendklinik, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Andriy Kost
- Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Xianming Mo
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Axel Hueber
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
| | - Rostyslav Bilyy
- Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Martin Herrmann
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
| | - Yi Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Luis E Muñoz
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
39
|
Elliott MR, Koster KM, Murphy PS. Efferocytosis Signaling in the Regulation of Macrophage Inflammatory Responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 198:1387-1394. [PMID: 28167649 PMCID: PMC5301545 DOI: 10.4049/jimmunol.1601520] [Citation(s) in RCA: 284] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 09/23/2016] [Indexed: 02/07/2023]
Abstract
Since the pioneering work of Elie Metchnikoff and the discovery of cellular immunity, the phagocytic clearance of cellular debris has been considered an integral component of resolving inflammation and restoring function of damaged and infected tissues. We now know that the phagocytic clearance of dying cells (efferocytosis), particularly by macrophages and other immune phagocytes, has profound consequences on innate and adaptive immune responses in inflamed tissues. These immunomodulatory effects result from an array of molecular signaling events between macrophages, dying cells, and other tissue-resident cells. In recent years, many of these molecular pathways have been identified and studied in the context of tissue inflammation, helping us better understand the relationship between efferocytosis and inflammation. We review specific types of efferocytosis-related signals that can impact macrophage immune responses and discuss their relevance to inflammation-related diseases.
Collapse
Affiliation(s)
- Michael R Elliott
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Kyle M Koster
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Patrick S Murphy
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| |
Collapse
|
40
|
Vujicic S, Feng L, Antoni A, Rauch J, Levine JS. Identification of Intracellular Signaling Events Induced in Viable Cells by Interaction with Neighboring Cells Undergoing Apoptotic Cell Death. J Vis Exp 2016. [PMID: 28060335 DOI: 10.3791/54980] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Cells dying by apoptosis, also referred to as regulated cell death, acquire multiple new activities that enable them to influence the function of adjacent live cells. Vital activities, such as survival, proliferation, growth, and differentiation, are among the many cellular functions modulated by apoptotic cells. The ability to recognize and respond to apoptotic cells appears to be a universal feature of all cells, regardless of lineage or organ of origination. However, the diversity and complexity of the response to apoptotic cells mandates that great care be taken in dissecting the signaling events and pathways responsible for any particular outcome. In particular, one must distinguish among the multiple mechanisms by which apoptotic cells can influence intracellular signaling pathways within viable responder cells, including: receptor-mediated recognition of the apoptotic cell, release of soluble mediators by the apoptotic cell, and/or engagement of the phagocytic machinery. Here, we provide a protocol for identifying intracellular signaling events that are induced in viable responder cells following their exposure to apoptotic cells. A major advantage of the protocol lies in the attention it pays to dissection of the mechanism by which apoptotic cells modulate signaling events within responding cells. While the protocol is specific for a conditionally immortalized mouse kidney proximal tubular cell line (BU.MPT cells), it is easily adapted to cell lines that are non-epithelial in origin and/or derived from organs other than the kidney. The use of dead cells as a stimulus introduces several unique factors that can hinder the detection of intracellular signaling events. These problems, as well as strategies to minimize or circumvent them, are discussed within the protocol. Application of this protocol should aid our expanding knowledge of the broad influence that dead or dying cells exert on their live neighbors, both in health and in disease.
Collapse
Affiliation(s)
- Snezana Vujicic
- Section of Nephrology, Department of Medicine, University of Illinois at Chicago; Section of Nephrology, Department of Medicine, Jesse Brown Veterans Affairs Medical Center
| | - Lanfei Feng
- Section of Nephrology, Department of Medicine, University of Illinois at Chicago; Section of Nephrology, Department of Medicine, Jesse Brown Veterans Affairs Medical Center
| | | | - Joyce Rauch
- Division of Rheumatology, Department of Medicine, Research Institute of the McGill University Health Centre
| | - Jerrold S Levine
- Section of Nephrology, Department of Medicine, University of Illinois at Chicago; Section of Nephrology, Department of Medicine, Jesse Brown Veterans Affairs Medical Center; Department of Microbiology & Immunology, University of Illinois at Chicago;
| |
Collapse
|
41
|
Gonzalez L, Trigatti BL. Macrophage Apoptosis and Necrotic Core Development in Atherosclerosis: A Rapidly Advancing Field with Clinical Relevance to Imaging and Therapy. Can J Cardiol 2016; 33:303-312. [PMID: 28232016 DOI: 10.1016/j.cjca.2016.12.010] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/14/2016] [Accepted: 12/14/2016] [Indexed: 01/11/2023] Open
Abstract
Cardiovascular diseases represent 1 of the main causes of death worldwide, and atherosclerosis is 1 of the major contributors leading to ischemic heart disease. Macrophages actively participate in all stages of atherosclerosis development, from plaque initiation to the transition to vulnerable plaques. Macrophage apoptosis, in particular, has been recognized as a critical step in the formation of the necrotic core, a key characteristic of unstable lesions. In this review, we discuss the role of macrophage apoptosis and clearance of apoptotic cells by efferocytosis in the development of atherosclerosis, with particular emphasis on their contribution to the development of the necrotic core and the clinical implications of this process for plaque stabilization. We consider the molecular triggers of macrophage apoptosis during atherogenesis, the role of endoplasmic reticulum (ER) stress, the roles of key cellular mediators of apoptosis and efferocytosis, and mechanisms of defective efferocytosis in the progression of atherosclerotic plaques. Finally, we discuss the important clinical implications of rapidly evolving macrophage science, such as novel approaches to imaging vulnerable atherosclerotic plaques with macrophage-sensitive positron emission tomography and magnetic resonance imaging, the role of macrophages in mediating beneficial pleiotropic actions of lipid-lowering therapies, and novel therapeutic modalities targeting ER stress, autophagy, and deficient efferocytosis. Advances in understanding the critical role of macrophages in the progression and destabilization of atherosclerosis have the potential to greatly improve the prevention and management of atherosclerotic diseases over the next decade.
Collapse
Affiliation(s)
- Leticia Gonzalez
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Bernardo Louis Trigatti
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
42
|
Jorgensen I, Zhang Y, Krantz BA, Miao EA. Pyroptosis triggers pore-induced intracellular traps (PITs) that capture bacteria and lead to their clearance by efferocytosis. J Exp Med 2016; 213:2113-28. [PMID: 27573815 PMCID: PMC5030797 DOI: 10.1084/jem.20151613] [Citation(s) in RCA: 290] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 07/29/2016] [Indexed: 12/21/2022] Open
Abstract
When inflammasomes trigger pyroptosis, the plasma membrane tears release-soluble cytosolic contents but retains organelles and intracellular bacteria. Neutrophils then efferocytose the pyroptotic cell debris and kill the entrapped bacteria. Inflammasomes activate caspase-1 in response to cytosolic contamination or perturbation. This inflammatory caspase triggers the opening of the GSDMD pore in the plasma membrane, resulting in lytic cell death called pyroptosis. We had previously assumed that pyroptosis releases intracellular bacteria to the extracellular space. Here, we find that viable bacteria instead remain trapped within the cellular debris of pyroptotic macrophages. This trapping appears to be an inevitable consequence of how osmotic lysis ruptures the plasma membrane, and may also apply to necroptosis and some forms of nonprogrammed necrosis. Although membrane tears release soluble cytosolic contents, they are small enough to retain organelles and bacteria. We call this structure the pore-induced intracellular trap (PIT), which is conceptually parallel to the neutrophil extracellular trap (NET). The PIT coordinates innate immune responses via complement and scavenger receptors to drive recruitment of and efferocytosis by neutrophils. Ultimately, this secondary phagocyte kills the bacteria. Hence, caspase-1–driven pore-induced cell death triggers a multifaceted defense against intracellular bacteria facilitated by trapping the pathogen within the cellular debris. Bona fide intracellular bacterial pathogens, such as Salmonella, must prevent or delay pyroptosis to avoid being trapped in the PIT and subsequently killed by neutrophils.
Collapse
Affiliation(s)
- Ine Jorgensen
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Yue Zhang
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Bryan A Krantz
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, MD 21201
| | - Edward A Miao
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
43
|
Brauer R, Ge L, Schlesinger SY, Birkland TP, Huang Y, Parimon T, Lee V, McKinney BL, McGuire JK, Parks WC, Chen P. Syndecan-1 Attenuates Lung Injury during Influenza Infection by Potentiating c-Met Signaling to Suppress Epithelial Apoptosis. Am J Respir Crit Care Med 2016; 194:333-44. [PMID: 26959387 PMCID: PMC4970595 DOI: 10.1164/rccm.201509-1878oc] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 03/09/2016] [Indexed: 12/16/2022] Open
Abstract
RATIONALE Syndecan-1 is a cell surface heparan sulfate proteoglycan primarily expressed in the lung epithelium. Because the influenza virus is tropic to the airway epithelium, we investigated the role of syndecan-1 in influenza infection. OBJECTIVES To determine the mechanism by which syndecan-1 regulates the lung mucosal response to influenza infection. METHODS Wild-type (WT) and Sdc1(-/-) mice were infected with a H1N1 virus (PR8) as an experimental model of influenza infection. Human and murine airway epithelial cell cultures were also infected with PR8 to study the mechanism by which syndecan-1 regulates the inflammatory response. MEASUREMENT AND MAIN RESULTS We found worsened outcomes and lung injury in Sdc1(-/-) mice compared with WT mice after influenza infection. Our data demonstrated that syndecan-1 suppresses bronchial epithelial apoptosis during influenza infection to limit widespread lung inflammation. Furthermore, we determined that syndecan-1 attenuated apoptosis by crosstalking with c-Met to potentiate its cytoprotective signals in airway epithelial cells during influenza infection. CONCLUSIONS Our work shows that cell-associated syndecan-1 has an important role in regulating lung injury. Our findings demonstrate a novel mechanism in which cell membrane-associated syndecan-1 regulates the innate immune response to influenza infection by facilitating cytoprotective signals through c-Met signaling to limit bronchial epithelial apoptosis, thereby attenuating lung injury and inflammation.
Collapse
Affiliation(s)
- Rena Brauer
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center; Los Angeles, California; and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Lingyin Ge
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center; Los Angeles, California; and
| | | | - Timothy P. Birkland
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center; Los Angeles, California; and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Ying Huang
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center; Los Angeles, California; and
| | - Tanyalak Parimon
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center; Los Angeles, California; and
| | - Vivian Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | | | - John K. McGuire
- Department of Pediatrics, University of Washington, Seattle, Washington
| | - William C. Parks
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center; Los Angeles, California; and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Peter Chen
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center; Los Angeles, California; and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| |
Collapse
|
44
|
Zaenker P, Gray E, Ziman M. Autoantibody Production in Cancer—The Humoral Immune Response toward Autologous Antigens in Cancer Patients. Autoimmun Rev 2016; 15:477-83. [DOI: 10.1016/j.autrev.2016.01.017] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 01/23/2016] [Indexed: 12/21/2022]
|
45
|
Velásquez SY, Killian D, Schulte J, Sticht C, Thiel M, Lindner HA. Short Term Hypoxia Synergizes with Interleukin 15 Priming in Driving Glycolytic Gene Transcription and Supports Human Natural Killer Cell Activities. J Biol Chem 2016; 291:12960-77. [PMID: 27129235 DOI: 10.1074/jbc.m116.721753] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Indexed: 01/08/2023] Open
Abstract
Natural killer (NK) cells induce apoptosis in infected and transformed cells and are important producers of immunoregulatory cytokines. Therefore, they operate under low oxygen conditions (hypoxia) in inflammatory and tumor environments. In vitro studies of NK cells are, however, commonly performed in ambient air (normoxia). We used global gene expression profiling to evaluate changes in transcriptional pathways in primary human NK cells following short term culture under hypoxia compared with normoxia and in response to interleukin 15 (IL-15) priming using a 2 × 2 factorial design. The largest contrasts observed were priming dependences for associations between hypoxia and the hypoxia-inducible factor (Hif) 1 signaling and glycolysis pathways. RT-PCR confirmed positive synergistic hypoxia/IL-15 interactions for genes of key regulatory and metabolic enzymes. IL-15 primes NK cells for effector functions, which were recently demonstrated to depend on glycolytic switching. We did not, however, observe important increases in glycolytic flux through hypoxia and priming alone. Chemical Hif-1α inhibition suggested equal importance of this transcription factor for glycolysis and energy production under normoxia and hypoxia. Hypoxia promoted secretion of CC chemokines Ccl3/4/5 and macrophage migration inhibitory factor. Unexpectedly, hypoxia also stimulated migration of NK cells through the extracellular matrix and shifted amounts of susceptible leukemia target cells toward late apoptosis in a cell killing assay. We conclude that short term hypoxia supports these activities by positively interacting with NK cell priming at the level of glycolytic gene transcription. Hypoxic conditioning of NK cells may thus benefit their use in cell-based immunotherapy of cancer.
Collapse
Affiliation(s)
- Sonia Y Velásquez
- From the Department of Anesthesiology and Surgical Intensive Care Medicine, and
| | - Doreen Killian
- From the Department of Anesthesiology and Surgical Intensive Care Medicine, and
| | - Jutta Schulte
- From the Department of Anesthesiology and Surgical Intensive Care Medicine, and
| | - Carsten Sticht
- Medical Research Center, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Manfred Thiel
- From the Department of Anesthesiology and Surgical Intensive Care Medicine, and
| | - Holger A Lindner
- From the Department of Anesthesiology and Surgical Intensive Care Medicine, and
| |
Collapse
|
46
|
Singh V, Roth S, Veltkamp R, Liesz A. HMGB1 as a Key Mediator of Immune Mechanisms in Ischemic Stroke. Antioxid Redox Signal 2016; 24:635-51. [PMID: 26493086 DOI: 10.1089/ars.2015.6397] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
SIGNIFICANCE Stroke is the leading cause of morbidity and mortality worldwide. Inflammatory cascades have a major impact on outcome and regeneration after ischemic stroke. High-mobility group box 1 (HMGB1) has come into the focus of experimental and clinical stroke research because it is released from necrotic brain tissue and its differential redox forms attract and activate immune cells after ischemic brain injury. HMGB1 is a potent inducer of inflammatory cascades, and thereby, secondary deterioration of neurological outcome. RECENT ADVANCES The role of HMGB1 in sterile inflammation is well established. Emerging evidence suggests that HMGB1 modulates neuroinflammation after experimental brain ischemia and that it may be a useful prognostic biomarker for stroke patients. CRITICAL ISSUES HMGB1 is instantly released from necrotic cells in the ischemic core and activates an early inflammatory response. In addition, brain-released HMGB1 can be redox modified in the circulation and activate peripheral immune cells. HMGB1 concentrations correlate with disease severity and outcome after brain injury. This is the first review depicting the crucial role of HMGB1 in the initiation and perpetuation of secondary immune alterations after experimental and clinical stroke. FUTURE DIRECTIONS HMGB1-dependent signaling pathways are on the verge and have the potential to become a central topic in experimental stroke research. Current and upcoming projects in this field will be paving the way for future translational approaches targeting the center of poststroke inflammation to improve stroke recovery and long-term outcome.
Collapse
Affiliation(s)
- Vikramjeet Singh
- 1 Institute for Stroke and Dementia Research , Klinikum der Universität München, Munich, Germany .,2 Munich Cluster for Systems Neurology (SyNergy) , Munich, Germany
| | - Stefan Roth
- 1 Institute for Stroke and Dementia Research , Klinikum der Universität München, Munich, Germany .,2 Munich Cluster for Systems Neurology (SyNergy) , Munich, Germany
| | - Roland Veltkamp
- 3 Division of Brain Sciences, Imperial College London , London, United Kingdom
| | - Arthur Liesz
- 1 Institute for Stroke and Dementia Research , Klinikum der Universität München, Munich, Germany .,2 Munich Cluster for Systems Neurology (SyNergy) , Munich, Germany
| |
Collapse
|
47
|
The clearance of dying cells: table for two. Cell Death Differ 2016; 23:915-26. [PMID: 26990661 PMCID: PMC4987729 DOI: 10.1038/cdd.2015.172] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/15/2015] [Accepted: 12/16/2015] [Indexed: 12/19/2022] Open
Abstract
Phagocytic cells of the immune system must constantly survey for, recognize, and efficiently clear the billions of cellular corpses that arise as a result of development, stress, infection, or normal homeostasis. This process, termed efferocytosis, is critical for the prevention of autoimmune and inflammatory disorders, and persistence of dead cells in tissue is characteristic of many human autoimmune diseases, notably systemic lupus erythematosus. The most notable characteristic of the efferocytosis of apoptotic cells is its ‘immunologically silent' response. Although the mechanisms by which phagocytes facilitate engulfment of dead cells has been a well-studied area, the pathways that coordinate to process the ingested corpse and direct the subsequent immune response is an area of growing interest. The recently described pathway of LC3 (microtubule-associated protein 1A/1B-light chain 3)-associated phagocytosis (LAP) has shed some light on this issue. LAP is triggered when an extracellular particle, such as a dead cell, engages an extracellular receptor during phagocytosis, induces the translocation of autophagy machinery, and ultimately LC3 to the cargo-containing phagosome, termed the LAPosome. In this review, we will examine efferocytosis and the impact of LAP on efferocytosis, allowing us to reimagine the impact of the autophagy machinery on innate host defense mechanisms.
Collapse
|
48
|
Grabiec AM, Hussell T. The role of airway macrophages in apoptotic cell clearance following acute and chronic lung inflammation. Semin Immunopathol 2016; 38:409-23. [PMID: 26957481 PMCID: PMC4896990 DOI: 10.1007/s00281-016-0555-3] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 01/29/2016] [Indexed: 12/19/2022]
Abstract
Acute and chronic inflammatory responses in the lung are associated with the accumulation of large quantities of immune and structural cells undergoing apoptosis, which need to be engulfed by phagocytes in a process called ‘efferocytosis’. Apoptotic cell recognition and removal from the lung is mediated predominantly by airway macrophages, though immature dendritic cells and non-professional phagocytes, such as epithelial cells and mesenchymal cells, can also display this function. Efficient clearance of apoptotic cells from the airways is essential for successful resolution of inflammation and the return to lung homeostasis. Disruption of this process leads to secondary necrosis of accumulating apoptotic cells, release of necrotic cell debris and subsequent uncontrolled inflammatory activation of the innate immune system by the released ‘damage associated molecular patterns’ (DAMPS). To control the duration of the immune response and prevent autoimmune reactions, anti-inflammatory signalling cascades are initiated in the phagocyte upon apoptotic cell uptake, mediated by a range of receptors that recognise specific phospholipids or proteins externalised on, or secreted by, the apoptotic cell. However, prolonged activation of apoptotic cell recognition receptors, such as the family of receptor tyrosine kinases Tyro3, Axl and MerTK (TAM), may delay or prevent inflammatory responses to subsequent infections. In this review, we will discuss recent advances in our understanding of the mechanism controlling apoptotic cell recognition and removal from the lung in homeostasis and during inflammation, the contribution of defective efferocytosis to chronic inflammatory lung diseases, such as chronic obstructive pulmonary disease, asthma and cystic fibrosis, and implications of the signals triggered by apoptotic cells in the susceptibility to pulmonary microbial infections.
Collapse
Affiliation(s)
- Aleksander M Grabiec
- Manchester Collaborative Centre for Inflammation Research, Core Technology Facility, The University of Manchester, 46 Grafton Street, M13 9NT, Manchester, UK
| | - Tracy Hussell
- Manchester Collaborative Centre for Inflammation Research, Core Technology Facility, The University of Manchester, 46 Grafton Street, M13 9NT, Manchester, UK.
| |
Collapse
|
49
|
Do not let death do us part: 'find-me' signals in communication between dying cells and the phagocytes. Cell Death Differ 2016; 23:979-89. [PMID: 26891690 DOI: 10.1038/cdd.2016.13] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 01/04/2016] [Accepted: 01/07/2016] [Indexed: 12/12/2022] Open
Abstract
The turnover and clearance of cells is an essential process that is part of many physiological and pathological processes. Improper or deficient clearance of apoptotic cells can lead to excessive inflammation and autoimmune disease. The steps involved in cell clearance include: migration of the phagocyte toward the proximity of the dying cells, specific recognition and internalization of the dying cell, and degradation of the corpse. The ability of phagocytes to recognize and react to dying cells to perform efficient and immunologically silent engulfment has been well-characterized in vitro and in vivo. However, how apoptotic cells themselves initiate the corpse removal and also influence the cells within the neighboring environment during clearance was less understood. Recent exciting observations suggest that apoptotic cells can attract phagocytes through the regulated release of 'find-me' signals. More recent studies also suggest that these find-me signals can have additional roles outside of phagocyte attraction to help orchestrate engulfment. This review will discuss our current understanding of the different find-me signals released by apoptotic cells, how they may be relevant in vivo, and their additional roles in facilitating engulfment.
Collapse
|
50
|
Mahajan A, Herrmann M, Muñoz LE. Clearance Deficiency and Cell Death Pathways: A Model for the Pathogenesis of SLE. Front Immunol 2016; 7:35. [PMID: 26904025 PMCID: PMC4745266 DOI: 10.3389/fimmu.2016.00035] [Citation(s) in RCA: 200] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 01/24/2016] [Indexed: 12/21/2022] Open
Abstract
Alterations of cell death pathways, including apoptosis and the neutrophil specific kind of death called NETosis, can represent a potential source of autoantigens. Defects in the clearance of apoptotic cells may be responsible for the initiation of systemic autoimmunity in several chronic inflammatory diseases, including systemic lupus erythematosus (SLE). Autoantigens are released mainly from secondary necrotic cells because of a defective clearance of apoptotic cells or an inefficient degradation of DNA-containing neutrophil extracellular traps (NETs). These modified autoantigens are presented by follicular dendritic cells to autoreactive B cells in germinal centers of secondary lymphoid organs. This results in the loss of self-tolerance and production of autoantibodies, a unifying feature of SLE. Immune complexes (IC) are formed from autoantibodies bound to uncleared cellular debris in blood or tissues. Clearance of IC by blood phagocytes, macrophages, and dendritic cells leads to proinflammatory cytokine secretion. In particular, plasmacytoid dendritic cells produce high amounts of interferon-α upon IC uptake, thereby contributing to the interferon signature of patients with SLE. The clearance of antinuclear IC via Fc-gamma receptors is considered a central event in amplifying inflammatory immune responses in SLE. Along with this, the accumulation of cell remnants represents an initiating event of the etiology, while the subsequent generation of autoantibodies against nuclear antigens (including NETs) results in the perpetuation of inflammation and tissue damage in patients with SLE. Here, we discuss the implications of defective clearance of apoptotic cells and NETs in the development of clinical manifestations in SLE.
Collapse
Affiliation(s)
- Aparna Mahajan
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3, Rheumatology and Immunology , Erlangen , Germany
| | - Martin Herrmann
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3, Rheumatology and Immunology , Erlangen , Germany
| | - Luis E Muñoz
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3, Rheumatology and Immunology , Erlangen , Germany
| |
Collapse
|