1
|
Wang C, Zhuo JJ, Li WQ, Zhou ML, Cheng KJ. Role of autophagy and mitophagy of group 2 innate lymphoid cells in allergic and local allergic rhinitis. World Allergy Organ J 2024; 17:100852. [PMID: 38298830 PMCID: PMC10827603 DOI: 10.1016/j.waojou.2023.100852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/14/2023] [Accepted: 12/07/2023] [Indexed: 02/02/2024] Open
Abstract
Background Roles of ILC2s in allergic rhinitis (AR) and local allergic rhinitis (LAR) are unclear. In this study, we are determined to find the levels of autophagy and mitophagy of ILC2s in allergic nasal inflammation. Methods ELISA was used to detect type 2 inflammatory cytokines. Hematoxylin and eosin (H&E) staining were used to compare the eosinophil (EOS) infiltration of nasal tissue specimens. Flow cytometry was used to detect the levels of ILC2s and Th2 cells. Immunohistochemistry (IHC) and Western blot (WB) were used to detect the levels of Beclin1, LC3, p62, PINK1, Parkin, FUNDC1, and BNIP3 in nasal mucosa. The levels of autophagy related proteins and mitophagy related proteins of the ILC2s were detected by WB. The number of autophagosomes of ILC2s was observed by transmission electron microscopy. The co-localization levels of GFP-LC3 and Mito tracker in ILC2s were observed by confocal microscopy using immunofluorescence. Results We found that the level of type 2 inflammation in AR and LAR mice was significantly increased. The levels of autophagy and mitophagy of AR and LAR mice in nasal mucosa and ILC2s were both increased. Conclusions ILC2s may be associated with the occurrence and development of nasal allergic inflammation. The abnormal increase of autophagy and mitophagy levels in the nose may be associated with the incidence of AR and LAR. Abnormal autophagy and mitophagy levels of ILC2s cells may be one of the causes of allergic nasal inflammation.
Collapse
Affiliation(s)
- Chen Wang
- Department of Otolaryngology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jin-Jing Zhuo
- Department of Otolaryngology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wen-Qian Li
- Department of Otolaryngology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Min-Li Zhou
- Department of Otolaryngology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ke-Jia Cheng
- Department of Otolaryngology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
2
|
Hosseini A, Germic N, Markov N, Stojkov D, Oberson K, Yousefi S, Simon HU. The regulatory role of eosinophils in adipose tissue depends on autophagy. Front Immunol 2024; 14:1331151. [PMID: 38235134 PMCID: PMC10792036 DOI: 10.3389/fimmu.2023.1331151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/06/2023] [Indexed: 01/19/2024] Open
Abstract
Introduction Obesity is a metabolic condition that elevates the risk of all-cause mortality. Brown and beige adipose tissues, known for their thermogenic properties, offer potential therapeutic targets for combating obesity. Recent reports highlight the role of immune cells, including eosinophils, in adipose tissue homeostasis, while the underlying mechanisms are poorly understood. Methods To study the role of autophagy in eosinophils in this process, we used a genetic mouse model lacking autophagy-associated protein 5 (Atg5), specifically within the eosinophil lineage (Atg5 eoΔ). Results The absence of Atg5 in eosinophils led to increased body weight, impaired glucose metabolism, and alterations in the cellular architecture of adipose tissue. Our findings indicate that Atg5 modulates the functional activity of eosinophils within adipose tissue rather than their abundance. Moreover, RNA-seq analysis revealed upregulation of arginase 2 (Arg2) in Atg5-knockout eosinophils. Increased Arg2 activity was shown to suppress adipocyte beiging. Furthermore, we observed enrichment of the purine pathway in the absence of Atg5 in eosinophils, leading to a pro-inflammatory shift in macrophages and a further reduction in beiging. Discussion The data shed light on the importance of autophagy in eosinophils and its impact on adipose tissue homeostasis by suppressing Arg2 expression and limiting inflammation in adipose tissue.
Collapse
Affiliation(s)
- Aref Hosseini
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Nina Germic
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Nikita Markov
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Darko Stojkov
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Kevin Oberson
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Shida Yousefi
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- Institute of Biochemistry, Brandenburg Medical School, Neuruppin, Germany
| |
Collapse
|
3
|
Ma Z, Wang J, Hu L, Wang S. Function of Innate Lymphoid Cells in Periodontal Tissue Homeostasis: A Narrative Review. Int J Mol Sci 2023; 24:ijms24076099. [PMID: 37047071 PMCID: PMC10093809 DOI: 10.3390/ijms24076099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/16/2023] [Accepted: 03/18/2023] [Indexed: 04/14/2023] Open
Abstract
Periodontitis is an irreversible inflammatory response that occurs in periodontal tissues. Given the size and diversity of natural flora in the oral mucosa, host immunity must strike a balance between pathogen identification and a complicated system of tolerance. The innate immune system, which includes innate lymphoid cells (ILCs), certainly plays a crucial role in regulating this homeostasis because pathogens are quickly recognized and responded to. ILCs are a recently discovered category of tissue-resident lymphocytes that lack adaptive antigen receptors. ILCs are found in both lymphoid and non-lymphoid organs and are particularly prevalent at mucosal barrier surfaces, where they control inflammatory response and homeostasis. Recent studies have shown that ILCs are important players in periodontitis; however, the mechanisms that govern the innate immune response in periodontitis still require further investigation. This review focuses on the intricate crosstalk between ILCs and the microenvironment in periodontal tissue homeostasis, with the purpose of regulating or improving immune responses in periodontitis prevention and therapy.
Collapse
Affiliation(s)
- Zhiyu Ma
- Beijing Laboratory of Oral Health, School of Basic Medicine, School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Jinsong Wang
- Beijing Laboratory of Oral Health, School of Basic Medicine, School of Stomatology, Capital Medical University, Beijing 100050, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Capital Medical University Beijing 100070, China
| | - Lei Hu
- Beijing Laboratory of Oral Health, School of Basic Medicine, School of Stomatology, Capital Medical University, Beijing 100050, China
- Department of Prosthodontics, School of Stomatology, Capital Medical University, Beijing 100050, China
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing 100070, China
| | - Songlin Wang
- Beijing Laboratory of Oral Health, School of Basic Medicine, School of Stomatology, Capital Medical University, Beijing 100050, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Capital Medical University Beijing 100070, China
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing 100070, China
- Laboratory for Oral and General Health Integration and Translation, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Research Unit of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, Beijing 100700, China
| |
Collapse
|
4
|
Makhijani P, Basso PJ, Chan YT, Chen N, Baechle J, Khan S, Furman D, Tsai S, Winer DA. Regulation of the immune system by the insulin receptor in health and disease. Front Endocrinol (Lausanne) 2023; 14:1128622. [PMID: 36992811 PMCID: PMC10040865 DOI: 10.3389/fendo.2023.1128622] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/08/2023] [Indexed: 03/14/2023] Open
Abstract
The signaling pathways downstream of the insulin receptor (InsR) are some of the most evolutionarily conserved pathways that regulate organism longevity and metabolism. InsR signaling is well characterized in metabolic tissues, such as liver, muscle, and fat, actively orchestrating cellular processes, including growth, survival, and nutrient metabolism. However, cells of the immune system also express the InsR and downstream signaling machinery, and there is increasing appreciation for the involvement of InsR signaling in shaping the immune response. Here, we summarize current understanding of InsR signaling pathways in different immune cell subsets and their impact on cellular metabolism, differentiation, and effector versus regulatory function. We also discuss mechanistic links between altered InsR signaling and immune dysfunction in various disease settings and conditions, with a focus on age related conditions, such as type 2 diabetes, cancer and infection vulnerability.
Collapse
Affiliation(s)
- Priya Makhijani
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Buck Institute for Research in Aging, Novato, CA, United States
| | - Paulo José Basso
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Yi Tao Chan
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Nan Chen
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Jordan Baechle
- Buck Institute for Research in Aging, Novato, CA, United States
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, United States
| | - Saad Khan
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
| | - David Furman
- Buck Institute for Research in Aging, Novato, CA, United States
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, United States
- Stanford 1, 000 Immunomes Project, Stanford School of Medicine, Stanford University, Stanford, CA, United States
- Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Pilar, Argentina
| | - Sue Tsai
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Daniel A. Winer
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Buck Institute for Research in Aging, Novato, CA, United States
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, United States
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
5
|
Hasan KMM, Haque MA. Autophagy and Its Lineage-Specific Roles in the Hematopoietic System. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:8257217. [PMID: 37180758 PMCID: PMC10171987 DOI: 10.1155/2023/8257217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 02/26/2023] [Accepted: 03/17/2023] [Indexed: 05/16/2023]
Abstract
Autophagy is a dynamic process that regulates the selective and nonselective degradation of cytoplasmic components, such as damaged organelles and protein aggregates inside lysosomes to maintain tissue homeostasis. Different types of autophagy including macroautophagy, microautophagy, and chaperon-mediated autophagy (CMA) have been implicated in a variety of pathological conditions, such as cancer, aging, neurodegeneration, and developmental disorders. Furthermore, the molecular mechanism and biological functions of autophagy have been extensively studied in vertebrate hematopoiesis and human blood malignancies. In recent years, the hematopoietic lineage-specific roles of different autophagy-related (ATG) genes have gained more attention. The evolution of gene-editing technology and the easy access nature of hematopoietic stem cells (HSCs), hematopoietic progenitors, and precursor cells have facilitated the autophagy research to better understand how ATG genes function in the hematopoietic system. Taking advantage of the gene-editing platform, this review has summarized the roles of different ATGs at the hematopoietic cell level, their dysregulation, and pathological consequences throughout hematopoiesis.
Collapse
Affiliation(s)
- Kazi Md Mahmudul Hasan
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia 7003, Bangladesh
- Department of Neurology, David Geffen School of Medicine, The University of California, 710 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Md Anwarul Haque
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia 7003, Bangladesh
| |
Collapse
|
6
|
Li Y, Law HKW. Deciphering the role of autophagy in the immunopathogenesis of inflammatory bowel disease. Front Pharmacol 2022; 13:1070184. [DOI: 10.3389/fphar.2022.1070184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a typical immune-mediated chronic inflammatory disorder. Following the industrialization and changes in lifestyle, the incidence of IBD in the world is rising, which makes health concerns and heavy burdens all over the world. However, the pathogenesis of IBD remains unclear, and the current understanding of the pathogenesis involves dysregulation of mucosal immunity, gut microbiome dysbiosis, and gut barrier defect based on genetic susceptibility and environmental triggers. In recent years, autophagy has emerged as a key mechanism in IBD development and progression because Genome-Wide Association Study revealed the complex interactions of autophagy in IBD, especially immunopathogenesis. Besides, autophagy markers are also suggested to be potential biomarkers and target treatment in IBD. This review summarizes the autophagy-related genes regulating immune response in IBD. Furthermore, we explore the evolving evidence that autophagy interacts with intestinal epithelial and immune cells to contribute to the inflammatory changes in IBD. Finally, we discuss how novel discovery could further advance our understanding of the role of autophagy and inform novel therapeutic strategies in IBD.
Collapse
|
7
|
Lian J, Liang Y, Zhang H, Lan M, Ye Z, Lin B, Qiu X, Zeng J. The role of polyamine metabolism in remodeling immune responses and blocking therapy within the tumor immune microenvironment. Front Immunol 2022; 13:912279. [PMID: 36119047 PMCID: PMC9479087 DOI: 10.3389/fimmu.2022.912279] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
The study of metabolism provides important information for understanding the biological basis of cancer cells and the defects of cancer treatment. Disorders of polyamine metabolism is a common metabolic change in cancer. With the deepening of understanding of polyamine metabolism, including molecular functions and changes in cancer, polyamine metabolism as a new anti-cancer strategy has become the focus of attention. There are many kinds of polyamine biosynthesis inhibitors and transport inhibitors, but not many drugs have been put into clinical application. Recent evidence shows that polyamine metabolism plays essential roles in remodeling the tumor immune microenvironment (TIME), particularly treatment of DFMO, an inhibitor of ODC, alters the immune cell population in the tumor microenvironment. Tumor immunosuppression is a major problem in cancer treatment. More and more studies have shown that the immunosuppressive effect of polyamines can help cancer cells to evade immune surveillance and promote tumor development and progression. Therefore, targeting polyamine metabolic pathways is expected to become a new avenue for immunotherapy for cancer.
Collapse
Affiliation(s)
- Jiachun Lian
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Yanfang Liang
- Department of Pathology, Dongguan Hospital Affiliated to Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Hailiang Zhang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Minsheng Lan
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Ziyu Ye
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Department of Pathology, Dongguan Hospital Affiliated to Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China
- Dongguan Metabolite Analysis Engineering Technology Center of Cells for Medical Use, Guangdong Xinghai Institute of Cell, Dongguan, China
| | - Bihua Lin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Key Laboratory of Medical Bioactive Molecular Research for Department of Education of Guangdong Province, Collaborative Innovation Center for Antitumor Active Substance Research and Development, Zhanjiang, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangdong Medical University, Zhanjiang, China
| | - Xianxiu Qiu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Key Laboratory of Medical Bioactive Molecular Research for Department of Education of Guangdong Province, Collaborative Innovation Center for Antitumor Active Substance Research and Development, Zhanjiang, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangdong Medical University, Zhanjiang, China
| | - Jincheng Zeng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Dongguan Metabolite Analysis Engineering Technology Center of Cells for Medical Use, Guangdong Xinghai Institute of Cell, Dongguan, China
- Key Laboratory of Medical Bioactive Molecular Research for Department of Education of Guangdong Province, Collaborative Innovation Center for Antitumor Active Substance Research and Development, Zhanjiang, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
8
|
Nrf2 Pathway and Autophagy Crosstalk: New Insights into Therapeutic Strategies for Ischemic Cerebral Vascular Diseases. Antioxidants (Basel) 2022; 11:antiox11091747. [PMID: 36139821 PMCID: PMC9495910 DOI: 10.3390/antiox11091747] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
Cerebrovascular disease is highly prevalent and has a complex etiology and variable pathophysiological activities. It thus poses a serious threat to human life and health. Currently, pathophysiological research on cerebrovascular diseases is gradually improving, and oxidative stress and autophagy have been identified as important pathophysiological activities that are gradually attracting increasing attention. Many studies have found some effects of oxidative stress and autophagy on cerebrovascular diseases, and studies on the crosstalk between the two in cerebrovascular diseases have made modest progress. However, further, more detailed studies are needed to determine the specific mechanisms. This review discusses nuclear factor erythroid 2-related factor 2 (Nrf2) molecules, which are closely associated with oxidative stress and autophagy, and the crosstalk between them, with the aim of providing clues for studying the two important pathophysiological changes and their crosstalk in cerebrovascular diseases as well as exploring new target treatments.
Collapse
|
9
|
Autophagy: Guardian of Skin Barrier. Biomedicines 2022; 10:biomedicines10081817. [PMID: 36009363 PMCID: PMC9405116 DOI: 10.3390/biomedicines10081817] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 11/23/2022] Open
Abstract
Autophagy is a major degradation pathway that removes harmful intracellular substances to maintain homeostasis. Various stressors, such as starvation and oxidative stress, upregulate autophagy, and the dysregulation of autophagy is associated with various human diseases, including cancer and skin diseases. The skin is the first defense barrier against external environmental hazards such as invading pathogens, ultraviolet rays, chemical toxins, and heat. Although the skin is exposed to various stressors that can activate autophagy, the roles of autophagy in the skin have not yet been fully elucidated. Accumulating evidence suggests that autophagy is closely associated with pathogenesis and the treatment of immune-related skin diseases. In this study, we review how autophagy interacts with skin cells, including keratinocytes and immune cells, enabling them to successfully perform their protective functions by eliminating pathogens and maintaining skin homeostasis. Furthermore, we discuss the implications of autophagy in immune-related skin diseases, such as alopecia areata, psoriasis, and atopic dermatitis, and suggest that a combination of autophagy modulators with conventional therapies may be a better strategy for the treatment of these diseases.
Collapse
|
10
|
Manzoor S, Muhammad JS, Maghazachi AA, Hamid Q. Autophagy: A Versatile Player in the Progression of Colorectal Cancer and Drug Resistance. Front Oncol 2022; 12:924290. [PMID: 35912261 PMCID: PMC9329589 DOI: 10.3389/fonc.2022.924290] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Colorectal cancer (CRC) is among the topmost malignancies for both genders. Despite the high incidence rate and advances in diagnostic tools, treatment in many cases is still ineffective. Most cancerous lesions in CRC begin as benign, followed by the development of invasive forms and metastases. The development of CRC has been linked to defects in autophagy, which plays both a pro-and anti-tumor role and is mainly context-dependent. Autophagy suppression could enhance apoptosis via p53 activation, or autophagy also promotes tumor progression by maintaining tumor growth and increasing resistance to chemotherapy. Autophagy promotes the invasion and metastasis of CRC cells via increased epithelial-mesenchymal transition (EMT). Moreover, dysbiosis of gut microbiota upregulated autophagy and metastasis markers. Autophagy responses may also modulate the tumor microenvironment (TME) via regulating the differentiation process of several innate immune cells. Treatments that promote tumor cell death by stimulating or inhibiting autophagy could be beneficial if used as an adjunct treatment, but the precise role of various autophagy-modulating drugs in CRC patients is needed to be explored. In this article, we present an overview of the autophagy process and its role in the pathogenesis and therapeutic resistance of CRC. Also, we focused on the current understanding of the role of the EMT and TME, including its relation to gut microbiota and immune cells, in autophagic manipulation of CRC. We believe that there is a potential link between autophagy, TME, EMT, and drug resistance, suggesting that further studies are needed to explore this aspect.
Collapse
Affiliation(s)
- Shaista Manzoor
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Jibran Sualeh Muhammad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Azzam A. Maghazachi
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Qutayba Hamid
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Center, Montreal, QC, Canada
- *Correspondence: Qutayba Hamid,
| |
Collapse
|
11
|
Chen L, Kong D, Xia S, Wang F, Li Z, Zhang F, Zheng S. Crosstalk Between Autophagy and Innate Immunity: A Pivotal Role in Hepatic Fibrosis. Front Pharmacol 2022; 13:891069. [PMID: 35656309 PMCID: PMC9152088 DOI: 10.3389/fphar.2022.891069] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 03/29/2022] [Indexed: 11/13/2022] Open
Abstract
Liver fibrosis is a repair process of chronic liver injuries induced by toxic substances, pathogens, and inflammation, which exhibits a feature such as deposition of the extracellular matrix. The initiation and progression of liver fibrosis heavily relies on excessive activation of hepatic stellate cells (HSCs). The activated HSCs express different kinds of chemokine receptors to further promote matrix remodulation. The long-term progression of liver fibrosis will contribute to dysfunction of the liver and ultimately cause hepatocellular carcinoma. The liver also has abundant innate immune cells, including DCs, NK cells, NKT cells, neutrophils, and Kupffer cells, which conduct complicated functions to activation and expansion of HSCs and liver fibrosis. Autophagy is one specific type of cell death, by which the aberrantly expressed protein and damaged organelles are transferred to lysosomes for further degradation, playing a crucial role in cellular homeostasis. Autophagy is also important to innate immune cells in various aspects. The previous studies have shown that dysfunction of autophagy in hepatic immune cells can result in the initiation and progression of inflammation in the liver, directly or indirectly causing activation of HSCs, which ultimately accelerate liver fibrosis. Given the crosstalk between innate immune cells, autophagy, and fibrosis progression is complicated, and the therapeutic options for liver fibrosis are quite limited, the exploration is essential. Herein, we review the previous studies about the influence of autophagy and innate immunity on liver fibrosis and the molecular mechanism to provide novel insight into the prevention and treatment of liver fibrosis.
Collapse
Affiliation(s)
- Li Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Desong Kong
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Siwei Xia
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Feixia Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhanghao Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
12
|
Klapan K, Simon D, Karaulov A, Gomzikova M, Rizvanov A, Yousefi S, Simon HU. Autophagy and Skin Diseases. Front Pharmacol 2022; 13:844756. [PMID: 35370701 PMCID: PMC8971629 DOI: 10.3389/fphar.2022.844756] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/22/2022] [Indexed: 12/15/2022] Open
Abstract
Autophagy is a highly conserved lysosomal degradation system that involves the creation of autophagosomes, which eventually fuse with lysosomes and breakdown misfolded proteins and damaged organelles with their enzymes. Autophagy is widely known for its function in cellular homeostasis under physiological and pathological settings. Defects in autophagy have been implicated in the pathophysiology of a variety of human diseases. The new line of evidence suggests that autophagy is inextricably linked to skin disorders. This review summarizes the principles behind autophagy and highlights current findings of autophagy's role in skin disorders and strategies for therapeutic modulation.
Collapse
Affiliation(s)
- Kim Klapan
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Dagmar Simon
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Alexander Karaulov
- Department of Clinical Immunology and Allergology, Sechenov University, Moscow, Russia
| | - Marina Gomzikova
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Albert Rizvanov
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Shida Yousefi
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland.,Department of Clinical Immunology and Allergology, Sechenov University, Moscow, Russia.,Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Institute of Biochemistry, Brandenburg Medical School, Neuruppin, Germany
| |
Collapse
|
13
|
Suzuki Y, Aono Y, Akiyama N, Horiike Y, Naoi H, Horiguchi R, Shibata K, Hozumi H, Karayama M, Furuhashi K, Enomoto N, Fujisawa T, Nakamura Y, Inui N, Suda T. Involvement of autophagy in exacerbation of eosinophilic airway inflammation in a murine model of obese asthma. Autophagy 2022; 18:2216-2228. [PMID: 35098856 PMCID: PMC9397451 DOI: 10.1080/15548627.2022.2025571] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Obesity is a common comorbidity in patients with asthma, and obese asthma patients present the most refractory phenotype among patients with severe asthma. Similar to the observations in non-obese asthma patients, clinical studies have revealed heterogeneity in obese asthma patients, including the occurrences of T helper (Th)2-high and Th2-low phenotypes. However, the mechanisms underlying obesity-related asthma are not completely understood. Though macroautophagy/autophagy is involved in asthma and obesity, its role in obesity-associated asthma is unknown. We hypothesized that autophagy is involved in the pathogenesis of obese asthma. For our investigations, we used high-fat diet-induced Atg5 (autophagy related 5)-deficient mice and epithelial cell-specific atg5−/− (Scgb1a1/CCSP-atg5−/−) obesity-induced mice. House dust mite (HDM)-sensitized atg5−/− obese mice exhibited marked eosinophilic inflammation and airway hyper-reactivity (AHR), compared to wild-type (WT) obese mice. Analyses of atg5−/− obese mice showed increased levels of Th2 cells but not ILC2s together with elevated expression of Th2 cytokines in the lung. In response to the HDM challenge, activated epithelial autophagy was observed in lean but not obese WT mice. Epithelium-specific deletion of Atg5 induced eosinophilic inflammation in Scgb1a1/CCSP-atg5−/− obese mice, and genetic analyses of epithelial cells from HDM-immunized atg5−/− obesity-induced mice showed an elevated expression of thymic stromal lymphopoietin (TSLP) and IL33. Notably, HDM-sensitized atg5−/− mice developed TSLP- and IL33-dependent eosinophilic inflammation and AHR. Our results suggest that autophagy contributes to the exacerbation of eosinophilic inflammation in obese asthma. Modulations of autophagy may be a therapeutic target in obesity-associated asthma. Abbreviations: AHR: airway hyper-reactivity; BAL: bronchoalveolar lavage; Cdyn: dynamic compliance; BM: bone marrow; HDM: house dust mite; HFD: high-fat diet; ILC2s: type 2 innate lymphocyte cells; ROS: reactive oxygen species; RL: lung resistance; TSLP: thymic stromal lymphopoietin; TCC: total cell count; WT: wild type.
Collapse
Affiliation(s)
- Yuzo Suzuki
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuya Aono
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Norimichi Akiyama
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yasuoki Horiike
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hyogo Naoi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Ryo Horiguchi
- Advanced Research Facilities and Services, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kiyoshi Shibata
- Advanced Research Facilities and Services, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hironao Hozumi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masato Karayama
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuki Furuhashi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Noriyuki Enomoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tomoyuki Fujisawa
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yutaro Nakamura
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Naoki Inui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
14
|
Zhang J, Rousseaux N, Walzer T. Eomes and T‐bet, a dynamic duo regulating NK cell differentiation. Bioessays 2022; 44:e2100281. [DOI: 10.1002/bies.202100281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 11/10/2022]
Affiliation(s)
- Jiang Zhang
- Department of Dermatology Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
| | - Noémi Rousseaux
- CIRI Centre International de Recherche en Infectiologie CNRS, UMR5308, ENS de Lyon Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1 Lyon France
| | - Thierry Walzer
- CIRI Centre International de Recherche en Infectiologie CNRS, UMR5308, ENS de Lyon Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1 Lyon France
| |
Collapse
|
15
|
Zhang J, Le Gras S, Pouxvielh K, Faure F, Fallone L, Kern N, Moreews M, Mathieu AL, Schneider R, Marliac Q, Jung M, Berton A, Hayek S, Vidalain PO, Marçais A, Dodard G, Dejean A, Brossay L, Ghavi-Helm Y, Walzer T. Sequential actions of EOMES and T-BET promote stepwise maturation of natural killer cells. Nat Commun 2021; 12:5446. [PMID: 34521844 PMCID: PMC8440589 DOI: 10.1038/s41467-021-25758-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 08/06/2021] [Indexed: 02/08/2023] Open
Abstract
EOMES and T-BET are related T-box transcription factors that control natural killer (NK) cell development. Here we demonstrate that EOMES and T-BET regulate largely distinct gene sets during this process. EOMES is dominantly expressed in immature NK cells and drives early lineage specification by inducing hallmark receptors and functions. By contrast, T-BET is dominant in mature NK cells, where it induces responsiveness to IL-12 and represses the cell cycle, likely through transcriptional repressors. Regardless, many genes with distinct functions are co-regulated by the two transcription factors. By generating two gene-modified mice facilitating chromatin immunoprecipitation of endogenous EOMES and T-BET, we show a strong overlap in their DNA binding targets, as well as extensive epigenetic changes during NK cell differentiation. Our data thus suggest that EOMES and T-BET may distinctly govern, via differential expression and co-factors recruitment, NK cell maturation by inserting partially overlapping epigenetic regulations.
Collapse
MESH Headings
- Animals
- Base Sequence
- Bone Marrow Cells/cytology
- Bone Marrow Cells/immunology
- CD11b Antigen/genetics
- CD11b Antigen/immunology
- Cell Cycle/drug effects
- Cell Cycle/genetics
- Cell Cycle/immunology
- Cell Differentiation
- Cell Lineage/drug effects
- Cell Lineage/genetics
- Cell Lineage/immunology
- Epigenesis, Genetic/immunology
- Interleukin-12/pharmacology
- Killer Cells, Natural/cytology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Promoter Regions, Genetic
- Protein Binding
- Spleen/cytology
- Spleen/immunology
- T-Box Domain Proteins/deficiency
- T-Box Domain Proteins/genetics
- T-Box Domain Proteins/immunology
- Transcription, Genetic
- Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics
- Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
Collapse
Affiliation(s)
- Jiang Zhang
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Stéphanie Le Gras
- IGBMC, CNRS UMR7104, Inserm U1258, Université de Strasbourg, Illkirch, France
- Plateforme GenomEast, infrastructure France Génomique, Illkirch, France
| | - Kevin Pouxvielh
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
| | - Fabrice Faure
- Institut NeuroMyoGène, INSERM U1217/CNRS UMR5310, Université de Lyon, Université Claude Bernard, Lyon 1, Lyon, France
| | - Lucie Fallone
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
| | - Nicolas Kern
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
| | - Marion Moreews
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
| | - Anne-Laure Mathieu
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
| | - Raphaël Schneider
- Institut de Génomique Fonctionnelle de Lyon, CNRS UMR 5242, Ecole Normale Supérieure de Lyon Université Claude Bernard Lyon 1, 46 allée d'Italie, F-69364, Lyon, France
| | - Quentin Marliac
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
| | - Mathieu Jung
- IGBMC, CNRS UMR7104, Inserm U1258, Université de Strasbourg, Illkirch, France
- Plateforme GenomEast, infrastructure France Génomique, Illkirch, France
| | - Aurore Berton
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
| | - Simon Hayek
- Equipe Chimie et Biologie, Modélisation et Immunologie pour la Thérapie (CBMIT), Université Paris Descartes, CNRS UMR 8601, 75006, Paris, France
| | - Pierre-Olivier Vidalain
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
- Equipe Chimie et Biologie, Modélisation et Immunologie pour la Thérapie (CBMIT), Université Paris Descartes, CNRS UMR 8601, 75006, Paris, France
| | - Antoine Marçais
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
| | - Garvin Dodard
- Department of Molecular Microbiology and Immunology, Division of Biology and Medicine, Brown University Alpert Medical School, Providence, RI, 02912, USA
| | - Anne Dejean
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), INSERM UMR1291 - CNRS UMR5051 - Université Toulouse III, Toulouse, France
| | - Laurent Brossay
- Department of Molecular Microbiology and Immunology, Division of Biology and Medicine, Brown University Alpert Medical School, Providence, RI, 02912, USA
| | - Yad Ghavi-Helm
- Institut de Génomique Fonctionnelle de Lyon, CNRS UMR 5242, Ecole Normale Supérieure de Lyon Université Claude Bernard Lyon 1, 46 allée d'Italie, F-69364, Lyon, France
| | - Thierry Walzer
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France.
| |
Collapse
|
16
|
Painter JD, Akbari O. Type 2 Innate Lymphoid Cells: Protectors in Type 2 Diabetes. Front Immunol 2021; 12:727008. [PMID: 34489979 PMCID: PMC8416625 DOI: 10.3389/fimmu.2021.727008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/29/2021] [Indexed: 12/13/2022] Open
Abstract
Type 2 innate lymphoid cells (ILC2) are the innate counterparts of Th2 cells and are critically involved in the maintenance of homeostasis in a variety of tissues. Instead of expressing specific antigen receptors, ILC2s respond to external stimuli such as alarmins released from damage. These cells help control the delicate balance of inflammation in adipose tissue, which is a determinant of metabolic outcome. ILC2s play a key role in the pathogenesis of type 2 diabetes mellitus (T2DM) through their protective effects on tissue homeostasis. A variety of crosstalk takes place between resident adipose cells and ILC2s, with each interaction playing a key role in controlling this balance. ILC2 effector function is associated with increased browning of adipose tissue and an anti-inflammatory immune profile. Trafficking and maintenance of ILC2 populations are critical for tissue homeostasis. The metabolic environment and energy source significantly affect the number and function of ILC2s in addition to affecting their interactions with resident cell types. How ILC2s react to changes in the metabolic environment is a clear determinant of the severity of disease. Treating sources of metabolic instability via critical immune cells provides a clear avenue for modulation of systemic homeostasis and new treatments of T2DM.
Collapse
Affiliation(s)
- Jacob D Painter
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
17
|
Germic N, Hosseini A, Yousefi S, Karaulov A, Simon HU. Regulation of eosinophil functions by autophagy. Semin Immunopathol 2021; 43:347-362. [PMID: 34019141 PMCID: PMC8241657 DOI: 10.1007/s00281-021-00860-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 04/22/2021] [Indexed: 12/13/2022]
Abstract
Eosinophils are granule-containing leukocytes which develop in the bone marrow. For many years, eosinophils have been recognized as cytotoxic effector cells, but recent studies suggest that they perform additional immunomodulatory and homeostatic functions. Autophagy is a conserved intracellular process which preserves cellular homeostasis. Autophagy defects have been linked to the pathogenesis of many human disorders. Evidence for abnormal regulation of autophagy, including decreased or increased expression of autophagy-related (ATG) proteins, has been reported in several eosinophilic inflammatory disorders, such as Crohn's disease, bronchial asthma, eosinophilic esophagitis, and chronic rhinosinusitis. Despite the increasing extent of research using preclinical models of immune cell-specific autophagy deficiency, the physiological relevance of autophagic pathway in eosinophils has remained unknown until recently. Owing to the increasing evidence that eosinophils play a role in keeping organismal homeostasis, the regulation of eosinophil functions is of considerable interest. Here, we discuss the most recent advances on the role of autophagy in eosinophils, placing particular emphasis on insights obtained in mouse models of infections and malignant diseases in which autophagy has genetically dismantled in the eosinophil lineage. These studies pointed to the possibility that autophagy-deficient eosinophils exaggerate inflammation. Therefore, the pharmacological modulation of the autophagic pathway in these cells could be used for therapeutic interventions.
Collapse
Affiliation(s)
- Nina Germic
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, 3010, Bern, Switzerland
| | - Aref Hosseini
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, 3010, Bern, Switzerland
| | - Shida Yousefi
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, 3010, Bern, Switzerland
| | - Alexander Karaulov
- Department of Clinical Immunology and Allergology, Sechenov University, 119991, Moscow, Russia
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, 3010, Bern, Switzerland. .,Department of Clinical Immunology and Allergology, Sechenov University, 119991, Moscow, Russia. .,Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420012, Kazan, Russia.
| |
Collapse
|
18
|
Xu C, Zang Y, Zhao Y, Cui W, Zhang H, Zhu Y, Xu M. Comprehensive Pan-Cancer Analysis Confirmed That ATG5 Promoted the Maintenance of Tumor Metabolism and the Occurrence of Tumor Immune Escape. Front Oncol 2021; 11:652211. [PMID: 33842365 PMCID: PMC8027486 DOI: 10.3389/fonc.2021.652211] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/25/2021] [Indexed: 01/05/2023] Open
Abstract
Background Autophagy related protein 5 (ATG5) is an important autophagosome formation related protein, and its involvement in the biological process of autophagy has been shown to correlate with tumor metabolic patterns and the formation of tumor heterogeneity. However, the role of ATG5 in tumor metabolism and tumor immunity remains unclear. Method In order to explore this problem, this study was designed to reveal the role of ATG5 in tumor metabolism and tumor immunity through pan-cancer analysis of multi-database. GTEx database, CCLE database, and TCGA database were used to describe the expression, prognosis, immune microenvironment, immune new antigen, immune checkpoint, TMB, and microsatellite instability of ATG5 in 33 types of tumors. A series of bioinformatics tools and methods were used for quantitative analysis and panoramic description, such as to Estimate, Scanneo and GSEA. Result The differential analysis results of multiple databases showed that ATG5 was ubiquitously highly expressed in pan-cancer, especially in solid tumors. Survival analysis revealed that ATG5 was universally associated with the prognosis of pan-cancer, and high ATG5 expression was significantly associated with poor patient prognosis in most cases. Further, the expression level of ATG5 was confirmed to be associated with tumor immune infiltration and tumor microenvironment, especially in BRCA, KIRC, and LIHC. In addition to this, ATG5 expression was confirmed to correlate with these clinically significant phenotypes, in conjunction with immune neoantigens and immune checkpoint gene expression profiles in pan-cancer. In addition to TMB and microsatellite instability in pan-cancer, we confirmed that ATG5 expression affects the expression of DNA repair genes and methyltransferases in pan-cancer, and found through gene set enrichment analysis that ATG5 is involved in the regulation of numerous signaling pathways involved in cancer metabolism and cancer immunity. Conclusions ATG5 participated in the formation of autophagosomal membrane important molecule LC3-II outside, and played an important role in tumor metabolism and tumor immunity. The comprehensive pan-cancer analysis not only revealed the potential of ATG5 in tumor-targeted therapy but also suggested ATG5 as a promising tumor predictive biomarker in most solid tumors.
Collapse
Affiliation(s)
- Chunxiao Xu
- Department of Cardiology, Sunshine Union Hospital, Weifang, China
| | - Yusheng Zang
- School of Medicine, Kunming University of Science and Technology, Kunming, China
| | - Yuxiang Zhao
- Institute of Bioengineering, Biotrans Technology Co., LTD, Shanghai, China.,United New Drug Research and Development Center, Biotrans Technology Co., LTD, Ningbo, China
| | - Weiqiang Cui
- Department of Anesthesiology, Sunshine Union Hospital, Weifang, China
| | - Hong Zhang
- Department of Internal Medicine, Weifang Maternal and Child Health Hospital, Weifang, China
| | - Yingcui Zhu
- Department of Radiology, Jinan Central Hospital, Jinan, China
| | - Man Xu
- School of Medicine, Shandong University, Jinan, China
| |
Collapse
|
19
|
Swadling L, Pallett LJ, Diniz MO, Baker JM, Amin OE, Stegmann KA, Burton AR, Schmidt NM, Jeffery-Smith A, Zakeri N, Suveizdyte K, Froghi F, Fusai G, Rosenberg WM, Davidson BR, Schurich A, Simon AK, Maini MK. Human Liver Memory CD8 + T Cells Use Autophagy for Tissue Residence. Cell Rep 2021; 30:687-698.e6. [PMID: 31968246 PMCID: PMC6988113 DOI: 10.1016/j.celrep.2019.12.050] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 11/04/2019] [Accepted: 12/13/2019] [Indexed: 12/11/2022] Open
Abstract
Tissue-resident memory T cells have critical roles in long-term pathogen and tumor immune surveillance in the liver. We investigate the role of autophagy in equipping human memory T cells to acquire tissue residence and maintain functionality in the immunosuppressive liver environment. By performing ex vivo staining of freshly isolated cells from human liver tissue, we find that an increased rate of basal autophagy is a hallmark of intrahepatic lymphocytes, particularly liver-resident CD8+ T cells. CD8+ T cells with increased autophagy are those best able to proliferate and mediate cytotoxicity and cytokine production. Conversely, blocking autophagy induction results in the accumulation of depolarized mitochondria, a feature of exhausted T cells. Primary hepatic stellate cells or the prototypic hepatic cytokine interleukin (IL)-15 induce autophagy in parallel with tissue-homing/retention markers. Inhibition of T cell autophagy abrogates tissue-residence programming. Thus, upregulation of autophagy adapts CD8+ T cells to combat mitochondrial depolarization, optimize functionality, and acquire tissue residence. An increased rate of basal autophagy is a hallmark of liver-resident CD8+ T cells Enhanced T cell autophagy can be imprinted by IL-15 or hepatic stellate cells Autophagy induction is required for tissue-residence programming in vitro Enhanced autophagy maintains TRM mitochondrial fitness in the liver
Collapse
Affiliation(s)
- Leo Swadling
- Division of Infection and Immunity, University College London, London, UK.
| | - Laura J Pallett
- Division of Infection and Immunity, University College London, London, UK
| | - Mariana O Diniz
- Division of Infection and Immunity, University College London, London, UK
| | - Josephine M Baker
- Division of Infection and Immunity, University College London, London, UK
| | - Oliver E Amin
- Division of Infection and Immunity, University College London, London, UK
| | - Kerstin A Stegmann
- Division of Infection and Immunity, University College London, London, UK
| | - Alice R Burton
- Division of Infection and Immunity, University College London, London, UK
| | - Nathalie M Schmidt
- Division of Infection and Immunity, University College London, London, UK
| | - Anna Jeffery-Smith
- Division of Infection and Immunity, University College London, London, UK; Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, QMUL, London, UK
| | - Nekisa Zakeri
- Division of Infection and Immunity, University College London, London, UK
| | | | - Farid Froghi
- Institute for Liver and Digestive Health, University College London, London, UK
| | - Giuseppe Fusai
- Institute for Liver and Digestive Health, University College London, London, UK
| | - William M Rosenberg
- Institute for Liver and Digestive Health, University College London, London, UK
| | - Brian R Davidson
- Institute for Liver and Digestive Health, University College London, London, UK
| | - Anna Schurich
- Division of Infection and Immunity, University College London, London, UK; Department of Infectious Diseases, Kings College London, London, UK
| | - A Katharina Simon
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| | - Mala K Maini
- Division of Infection and Immunity, University College London, London, UK.
| |
Collapse
|
20
|
ATG5 promotes eosinopoiesis but inhibits eosinophil effector functions. Blood 2021; 137:2958-2969. [PMID: 33598715 DOI: 10.1182/blood.2020010208] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/03/2021] [Indexed: 12/14/2022] Open
Abstract
Eosinophils are white blood cells that contribute to the regulation of immunity and are involved in the pathogenesis of numerous inflammatory diseases. In contrast to other cells of the immune system, no information is available regarding the role of autophagy in eosinophil differentiation and functions. To study the autophagic pathway in eosinophils, we generated conditional knockout mice in which Atg5 is deleted within the eosinophil lineage only (designated Atg5eoΔ mice). Eosinophilia was provoked by crossbreeding Atg5eoΔ mice with Il5 (IL-5) overexpressing transgenic mice (designated Atg5eoΔIl5tg mice). Deletion of Atg5 in eosinophils resulted in a dramatic reduction in the number of mature eosinophils in blood and an increase of immature eosinophils in the bone marrow. Atg5-knockout eosinophil precursors exhibited reduced proliferation under both in vitro and in vivo conditions but no increased cell death. Moreover, reduced differentiation of eosinophils in the absence of Atg5 was also observed in mouse and human models of chronic eosinophilic leukemia. Atg5-knockout blood eosinophils exhibited augmented levels of degranulation and bacterial killing in vitro. Moreover, in an experimental in vivo model, we observed that Atg5eoΔ mice achieve better clearance of the local and systemic bacterial infection with Citrobacter rodentium. Evidence for increased degranulation of ATG5low-expressing human eosinophils was also obtained in both tissues and blood. Taken together, mouse and human eosinophil hematopoiesis and effector functions are regulated by ATG5, which controls the amplitude of overall antibacterial eosinophil immune responses.
Collapse
|
21
|
Santos A, Ramos I. ATG3 Is Important for the Chorion Ultrastructure During Oogenesis in the Insect Vector Rhodnius prolixus. Front Physiol 2021; 12:638026. [PMID: 33613326 PMCID: PMC7888535 DOI: 10.3389/fphys.2021.638026] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/12/2021] [Indexed: 01/13/2023] Open
Abstract
In insects, the last stage of the oogenesis is the choriogenesis, a process where the multiple layers of the chorion are synthesized, secreted, and deposited in the surface of the oocytes by the follicle cells. The chorion is an extracellular matrix that serves as a highly specialized protective shield for the embryo, being crucial to impair water loss and to allow gas exchange throughout development. The E2-like enzyme ATG3 (autophagy related gene 3) is known for its canonical function in the autophagy pathway, in the conjugation of the ubiquitin-like ATG8/LC3 to the membranes of autophagosomes. Although the ATGs were originally described and annotated as genes related to autophagy, additional functions have been attributed to various of these genes. Here, we found that Rhodnius prolixus ATG3 is highly expressed in the ovaries of the adult vitellogenic females. Parental RNAi depletion of ATG3 resulted in a 15% decrease in the oviposition rates of depleted females and in the generation of unviable eggs. ATG3-depleted eggs are small and present one specific phenotype of altered chorion ultrastructure, observed by high resolution scanning electron microscopy. The amounts of the major chorion proteins Rp30, Rp45, Rp100, and Rp200 were decreased in the ATG3-depleted chorions, as well as the readings for dityrosine cross-linking and sulfur, detected by fluorescence emission under ultraviolet excitation and X-ray elemental detection and mapping. Altogether, we found that ATG3 is important for the proper chorion biogenesis and, therefore, crucial for this vector reproduction.
Collapse
Affiliation(s)
- Anna Santos
- Laboratório de Bioquímica de Insetos, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Paraná, Brazil
| | - Isabela Ramos
- Laboratório de Bioquímica de Insetos, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Paraná, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular - INCT-EM/CNPq, Rio de Janeiro, Brazil
| |
Collapse
|
22
|
Yamazaki T, Bravo-San Pedro JM, Galluzzi L, Kroemer G, Pietrocola F. Autophagy in the cancer-immunity dialogue. Adv Drug Deliv Rev 2021; 169:40-50. [PMID: 33301821 DOI: 10.1016/j.addr.2020.12.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/24/2020] [Accepted: 12/04/2020] [Indexed: 02/08/2023]
Abstract
Autophagy is quintessential for the maintenance of cellular homeostasis in all eukaryotic cells, explaining why both normal and malignant cells benefit from proficient autophagic responses. Moreover, autophagy is intimately involved in the immunological control of malignant transformation, tumor progression and response to therapy. However, the net effect of autophagy activation or inhibition on the natural growth or therapeutic response of tumors evolving in immunocompetent hosts exhibits a considerable degree of context dependency. Here, we discuss the complex cross-talk between autophagy and immuno-oncology as delineated by genetic and pharmacological approaches in mouse models of cancer.
Collapse
|
23
|
Jogalekar MP, Veerabathini A, Gangadaran P. Recent developments in autophagy-targeted therapies in cancer. Exp Biol Med (Maywood) 2020; 246:207-212. [PMID: 33167689 DOI: 10.1177/1535370220966545] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Autophagy plays a crucial role in cellular development and differentiation as well as in the maintenance of homeostasis in healthy cells. Autophagy is well documented in neurodegenerative disorders, aging, and infectious diseases. However, recognizing its significance in cancer has always been challenging due to its tumor-promoting and suppressive attributes. Various modulators targeting key components of autophagy machinery directly or indirectly have been developed over the years, and have shown promising results in preclinical models. Some of these compounds are even being tested in clinical trials for safety and efficacy. A detailed review of strategies used to target autophagy in cancer is presented including our opinion on developing better therapies and outstanding issues.
Collapse
Affiliation(s)
- Manasi P Jogalekar
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| |
Collapse
|
24
|
Zheng Z, Wang L, Cheng S, Wang Y, Zhao W. Autophagy and Leukemia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1207:601-613. [PMID: 32671778 DOI: 10.1007/978-981-15-4272-5_43] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Leukemia is a malignant clonal disease that originates from hematopoietic stem cells. As in-depth research examines the molecular biology and immunology of the hematopoietic system, leukemia treatment has evolved from a single cytotoxic drug to treatments that inducing differentiation and apoptosis. Meanwhile, autophagy has become a growing concern as a new form of cell death. The immune response, hematopoietic stem cell differentiation, and drug resistance of tumor cells are all potentially affected by autophagy. Regulating autophagy may become one of the promising directions in the field of targeted therapy.
Collapse
Affiliation(s)
- Zhong Zheng
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Wang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shu Cheng
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Wang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weili Zhao
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
25
|
Painter JD, Galle-Treger L, Akbari O. Role of Autophagy in Lung Inflammation. Front Immunol 2020; 11:1337. [PMID: 32733448 PMCID: PMC7358431 DOI: 10.3389/fimmu.2020.01337] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 05/26/2020] [Indexed: 12/11/2022] Open
Abstract
Autophagy is a cellular recycling system found in almost all types of eukaryotic organisms. The system is made up of a variety of proteins which function to deliver intracellular cargo to lysosomes for formation of autophagosomes in which the contents are degraded. The maintenance of cellular homeostasis is key in the survival and function of a variety of human cell populations. The interconnection between metabolism and autophagy is extensive, therefore it has a role in a variety of different cell functions. The disruption or dysfunction of autophagy in these cell types have been implicated in the development of a variety of inflammatory diseases including asthma. The role of autophagy in non-immune and immune cells both lead to the pathogenesis of lung inflammation. Autophagy in pulmonary non-immune cells leads to tissue remodeling which can develop into chronic asthma cases with long term effects. The role autophagy in the lymphoid and myeloid lineages in the pathology of asthma differ in their functions. Impaired autophagy in lymphoid populations have been shown, in general, to decrease inflammation in both asthma and inflammatory disease models. Many lymphoid cells rely on autophagy for effector function and maintained inflammation. In stark contrast, autophagy deficient antigen presenting cells have been shown to have an activated inflammasome. This is largely characterized by a TH17 response that is accompanied with a much worse prognosis including granulocyte mediated inflammation and steroid resistance. The cell specificity associated with changes in autophagic flux complicates its targeting for amelioration of asthmatic symptoms. Differing asthmatic phenotypes between TH2 and TH17 mediated disease may require different autophagic modulations. Therefore, treatments call for a more cell specific and personalized approach when looking at chronic asthma cases. Viral-induced lung inflammation, such as that caused by SARS-CoV-2, also may involve autophagic modulation leading to inflammation mediated by lung resident cells. In this review, we will be discussing the role of autophagy in non-immune cells, myeloid cells, and lymphoid cells for their implications into lung inflammation and asthma. Finally, we will discuss autophagy's role viral pathogenesis, immunometabolism, and asthma with insights into autophagic modulators for amelioration of lung inflammation.
Collapse
Affiliation(s)
- Jacob D Painter
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Lauriane Galle-Treger
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
26
|
Xu CN, Kong LH, Ding P, Liu Y, Fan ZG, Gao EH, Yang J, Yang LF. Melatonin ameliorates pressure overload-induced cardiac hypertrophy by attenuating Atg5-dependent autophagy and activating the Akt/mTOR pathway. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165848. [PMID: 32473999 DOI: 10.1016/j.bbadis.2020.165848] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 05/06/2020] [Accepted: 05/21/2020] [Indexed: 12/16/2022]
Abstract
Cardiac hypertrophy, including hypertension and valvular dysfunction, is a pathological feature of many cardiac diseases that ultimately leads to heart failure. Melatonin confers a protective role against pathological cardiac hypertrophy, but the underlying mechanisms remain elusive. In the present study, we hypothesized that melatonin protects against pressure overload-induced cardiac hypertrophy by attenuating Atg5-dependent autophagy and activating the Akt/mTOR pathway. Male C57BL/6 mice that received adenovirus carrying cardiac-specific Atg5 (under the cTNT promoter; Ad-cTNT-Atg5) underwent transverse aortic constriction (TAC) or sham operation and received an intraperitoneal injection of melatonin (10 mg/kg/d), vehicle or LY294002 (10 mg/kg/d) for 8 weeks. Melatonin treatment for 8 weeks markedly attenuated cardiac hypertrophy and restored impaired cardiac function, as indicated by a decreased HW/BW ratio, reduced cell cross-sectional area and fibrosis, downregulated the mRNA levels of ANP, BNP, and β-MHC and ameliorated adverse effects on the LVEF and LVFS. Melatonin treatment also inhibited apoptosis and alleviated autophagy dysfunction. Furthermore, melatonin inhibited Akt/mTOR pathway activation, while these effects were blocked by LY294002. In addition, the effect of melatonin regulation on TAC-induced autophagy dysfunction was inhibited by LY294002 or cardiac-specific Atg5 overexpression. As expected, Akt/mTOR pathway inhibition or cardiac-specific Atg5 overexpression restrained melatonin alleviation of pressure overload-induced cardiac hypertrophy. These results demonstrated that melatonin ameliorated pressure overload-induced cardiac hypertrophy by attenuating Atg5-dependent autophagy and activating the Akt/mTOR pathway.
Collapse
Affiliation(s)
- Chen-Nian Xu
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Ling-Heng Kong
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, China; School of Basic Medical Science, Xi'an Medical University, Xi'an 710021, China
| | - Peng Ding
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Yang Liu
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Zhen-Ge Fan
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Er-He Gao
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Jian Yang
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, China.
| | - Li-Fang Yang
- Department of Anesthesiology, Xi'an Children's Hospital, Xi'an 710003, China.
| |
Collapse
|
27
|
Abstract
The molecular machinery of macroautophagy consists of Atg proteins and supports cytoplasmic constituent degradation in lysosomes as its canonical function, phagosome maturation and exocytosis. These different biological processes contribute to cell intrinsic, innate and adaptive immunity. For the respective immune responses, Atg proteins mediate direct pathogen degradation, inflammation restriction, antigen presentation on MHC molecules and survival of memory lymphocyte populations. During adaptive immunity MHC class II presentation of antigens is supported and MHC class I presentation restricted by the macroautophagy machinery. Considering these various functions might allow us to predict the outcome of interventions that manipulate the machinery of Atg proteins as immunotherapies for the benefit of human health.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
28
|
Autophagy-Independent Functions of the Autophagy Machinery. Cell 2020; 177:1682-1699. [PMID: 31199916 PMCID: PMC7173070 DOI: 10.1016/j.cell.2019.05.026] [Citation(s) in RCA: 606] [Impact Index Per Article: 121.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 02/11/2019] [Accepted: 05/13/2019] [Indexed: 02/07/2023]
Abstract
Macroautophagy (herein referred to as autophagy) is an evolutionary ancient mechanism that culminates with the lysosomal degradation of superfluous or potentially dangerous cytosolic entities. Over the past 2 decades, the molecular mechanisms underlying several variants of autophagy have been characterized in detail. Accumulating evidence suggests that most, if not all, components of the molecular machinery for autophagy also mediate autophagy-independent functions. Here, we discuss emerging data on the non-autophagic functions of autophagy-relevant proteins.
Collapse
|
29
|
Xu ZD, Wang Y, Liang G, Liu ZQ, Ma WH, Chu CT, Wei HF. Propofol affects mouse embryonic fibroblast survival and proliferation in vitro via ATG5- and calcium-dependent regulation of autophagy. Acta Pharmacol Sin 2020; 41:303-310. [PMID: 31645660 PMCID: PMC7471456 DOI: 10.1038/s41401-019-0303-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/29/2019] [Indexed: 12/03/2022] Open
Abstract
Propofol is a commonly used intravenous anesthetic agent, which has been found to affect cell survival and proliferation especially in early life. Our previous studies show that propofol-induced neurodegeneration and neurogenesis are closely associated with cell autophagy. In the present study we explored the roles of autophagy-related gene 5 (ATG5) in propofol-induced autophagy in mouse embryonic fibroblasts (MEF) in vitro. We showed that ATG5 was functionally related to propofol-induced cell survival and damage: propofol significantly enhanced cell survival and proliferation at a clinically relevant dose (10 µM), but caused cell death at an extremely high concentration (200 µM) in ATG5−/− MEF, but not in WT cells. The dual effects found in ATG5−/− MEF could be blocked by intracellular Ca2+ channel antagonists. We also found that propofol evoked a moderate (promote cell growth) and extremely high (cause apoptosis) cytosolic Ca2+ elevation at the concentrations of 10 µM and 200 µM, respectively, only in ATG5−/− MEF. In addition, ATG5−/− MEF themselves released more Ca2+ in cytosolic space and endoplasmic reticulum compared with WT cells, suggesting that autophagy deficiency made intracellular calcium signaling more vulnerable to external stimuli (propofol). Altogether, our results reveal that ATG5 plays a crucial role in propofol regulation of cell survival and proliferation by affecting intracellular Ca2+ homeostasis.
Collapse
|
30
|
Living with Yourself: Innate Lymphoid Cell Immunometabolism. Cells 2020; 9:cells9020334. [PMID: 32024050 PMCID: PMC7072664 DOI: 10.3390/cells9020334] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 01/29/2020] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
Innate lymphoid cells (ILCs) are tissue-resident sentinels of the immune system that function to protect local tissue microenvironments against pathogens and maintain homeostasis. However, because ILCs are sensitively tuned to perturbations within tissues, they can also contribute to host pathology when critical activating signals become dysregulated. Recent work has demonstrated that the crosstalk between ILCs and their environment has a significant impact on host metabolism in health and disease. In this review, we summarize studies that support evidence for the ability of ILCs to influence tissue and systemic metabolism, as well as how ILCs can be regulated by environmental changes in systemic host metabolism. We also highlight studies demonstrating how ILC- intrinsic metabolism influences their activation, proliferation, and homeostasis. Finally, this review discusses the challenges and open questions in the rapidly expanding field of ILCs and immunometabolism.
Collapse
|
31
|
Koschade SE, Brandts CH. Selective Autophagy in Normal and Malignant Hematopoiesis. J Mol Biol 2020; 432:261-282. [DOI: 10.1016/j.jmb.2019.06.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/18/2019] [Accepted: 06/18/2019] [Indexed: 12/16/2022]
|
32
|
Abstract
Across all branches of the immune system, the process of autophagy is fundamentally important in cellular development, function and homeostasis. Strikingly, this evolutionarily ancient pathway for intracellular recycling has been adapted to enable a high degree of functional complexity and specialization. However, although the requirement for autophagy in normal immune cell function is clear, the mechanisms involved are much less so and encompass control of metabolism, selective degradation of substrates and organelles and participation in cell survival decisions. We review here the crucial functions of autophagy in controlling the differentiation and homeostasis of multiple immune cell types and discuss the potential mechanisms involved.
Collapse
|
33
|
Autophagy Regulation of Mammalian Immune Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1209:7-22. [PMID: 31728862 DOI: 10.1007/978-981-15-0606-2_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Autophagy is a fully competent cellular machinery able to carry out the clearance of macromolecules via fusion with the lysosome. Many studies conducted in recent years have revealed that autophagy not only plays a critical role in maintaining cell homeostasis, but can also promote bacterial elimination. Additionally, autophagy exists in most eukaryotic cells including immune cells, such as lymphocytes, neutrophils, eosinophils, mast cells, and natural killer cells. Presently, there are numerous studies focusing on the roles of autophagy in regulating immune response. Autophagy regulates the innate and adaptive immunity by modulating cell differentiation, survival, phagocytosis, antigen presentation, degranulation, and cytokine production. In this chapter, we will summarize how autophagy participates explicitly in the survival and function of the mammalian adaptive and innate immune cells.
Collapse
|
34
|
Galle-Treger L, Hurrell BP, Lewis G, Howard E, Jahani PS, Banie H, Razani B, Soroosh P, Akbari O. Autophagy is critical for group 2 innate lymphoid cell metabolic homeostasis and effector function. J Allergy Clin Immunol 2019; 145:502-517.e5. [PMID: 31738991 DOI: 10.1016/j.jaci.2019.10.035] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 10/07/2019] [Accepted: 10/11/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Allergic asthma is a chronic inflammatory disorder characterized by airway hyperreactivity (AHR) and driven by TH2 cytokine production. Group 2 innate lymphoid cells (ILC2s) secrete high amounts of TH2 cytokines and contribute to the development of AHR. Autophagy is a cellular degradation pathway that recycles cytoplasmic content. However, the role of autophagy in ILC2s remains to be fully elucidated. OBJECTIVE We characterized the effects of autophagy deficiency on ILC2 effector functions and metabolic balance. METHODS ILC2s from autophagy-deficient mice were isolated to evaluate proliferation, apoptosis, cytokine secretion, gene expression and cell metabolism. Also, autophagy-deficient ILC2s were adoptively transferred into Rag-/-GC-/- mice, which were then challenged with IL-33 and assessed for AHR and lung inflammation. RESULTS We demonstrate that autophagy is extensively used by activated ILC2s to maintain their homeostasis and effector functions. Deletion of the critical autophagy gene autophagy-related 5 (Atg5) resulted in decreased cytokine secretion and increased apoptosis. Moreover, lack of autophagy among ILC2s impaired their ability to use fatty acid oxidation and strikingly promoted glycolysis, as evidenced by our transcriptomic and metabolite analyses. This shift of fuel dependency led to impaired homeostasis and TH2 cytokine production, thus inhibiting the development of ILC2-mediated AHR. Notably, this metabolic reprogramming was also associated with an accumulation of dysfunctional mitochondria, producing excessive reactive oxygen species. CONCLUSION These findings provide new insights into the metabolic profile of ILC2s and suggest that modulation of fuel dependency by autophagy is a potentially new therapeutic approach to target ILC2-dependent inflammation.
Collapse
Affiliation(s)
- Lauriane Galle-Treger
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Benjamin P Hurrell
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Gavin Lewis
- Janssen Research and Development, San Diego, Calif
| | - Emily Howard
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Pedram Shafiei Jahani
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | | | - Babak Razani
- Departments of Medicine and Pathology & Immunology, Washington University School of Medicine and John Cochran VA Medical Center, St Louis, Mo
| | | | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif.
| |
Collapse
|
35
|
Vetters J, van Helden MJ, Wahlen S, Tavernier SJ, Martens A, Fayazpour F, Vergote K, Vanheerswynghels M, Deswarte K, Van Moorleghem J, De Prijck S, Takahashi N, Vandenabeele P, Boon L, van Loo G, Vivier E, Lambrecht BN, Janssens S. The ubiquitin-editing enzyme A20 controls NK cell homeostasis through regulation of mTOR activity and TNF. J Exp Med 2019; 216:2010-2023. [PMID: 31296735 PMCID: PMC6719426 DOI: 10.1084/jem.20182164] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 05/10/2019] [Accepted: 06/18/2019] [Indexed: 01/20/2023] Open
Abstract
The study of Vetters et al. identifies the ubiquitin-modifying enzyme A20 as a critical regulator of mTOR. Loss of A20 unleashes mTOR activity and induces NK cell death, underscoring the need for a tightly controlled mTOR pathway for proper NK cell homeostasis. The ubiquitin-editing enzyme A20 is a well-known regulator of immune cell function and homeostasis. In addition, A20 protects cells from death in an ill-defined manner. While most studies focus on its role in the TNF-receptor complex, we here identify a novel component in the A20-mediated decision between life and death. Loss of A20 in NK cells led to spontaneous NK cell death and severe NK cell lymphopenia. The few remaining NK cells showed an immature, hyperactivated phenotype, hallmarked by the basal release of cytokines and cytotoxic molecules. NK-A20−/− cells were hypersensitive to TNF-induced cell death and could be rescued, at least partially, by a combined deficiency with TNF. Unexpectedly, rapamycin, a well-established inhibitor of mTOR, also strongly protected NK-A20−/− cells from death, and further studies revealed that A20 restricts mTOR activation in NK cells. This study therefore maps A20 as a crucial regulator of mTOR signaling and underscores the need for a tightly balanced mTOR pathway in NK cell homeostasis.
Collapse
Affiliation(s)
- Jessica Vetters
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium.,Laboratory for Endoplasmic Reticulum Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium.,GROUP-ID Consortium, Ghent University and Ghent University Hospital, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Mary J van Helden
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Sigrid Wahlen
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Simon J Tavernier
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Arne Martens
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Cellular and Molecular (Patho)physiology, VIB Center for Inflammation Research, Ghent, Belgium
| | - Farzaneh Fayazpour
- Laboratory for Endoplasmic Reticulum Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium.,GROUP-ID Consortium, Ghent University and Ghent University Hospital, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Karl Vergote
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Manon Vanheerswynghels
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Kim Deswarte
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Justine Van Moorleghem
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Sofie De Prijck
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Nozomi Takahashi
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium
| | - Peter Vandenabeele
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium
| | | | - Geert van Loo
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Cellular and Molecular (Patho)physiology, VIB Center for Inflammation Research, Ghent, Belgium
| | - Eric Vivier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France.,Aix-Marseille University, Assistance Publique-Hôpitaux de Marseille, Centre d'Immunologie de Marseille-Luminy, Hôpital de la Timone, Marseille Immunopôle, Marseille, France
| | - Bart N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium .,GROUP-ID Consortium, Ghent University and Ghent University Hospital, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Sophie Janssens
- Laboratory for Endoplasmic Reticulum Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium .,GROUP-ID Consortium, Ghent University and Ghent University Hospital, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| |
Collapse
|
36
|
Autophagy in regulatory T cells: A double-edged sword in disease settings. Mol Immunol 2019; 109:43-50. [PMID: 30852245 DOI: 10.1016/j.molimm.2019.02.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/03/2019] [Accepted: 02/05/2019] [Indexed: 12/21/2022]
Abstract
Autophagy is an evolutionarily conserved catabolic process that directs cytoplasmic proteins, organelles and microbes to lysosomes for degradation. It not only represents an essential cell-intrinsic mechanism to protect against internal and external stresses but also shapes both innate and adaptive immunity. Regulatory T cells (Tregs) are a developmentally and functionally distinct T cell subpopulation engaged in sustaining immunological self-tolerance and homeostasis. There is compelling evidence that autophagy is actively regulated in Tregs and serves as a central signal-dependent controller for Tregs by restraining excessive apoptotic and metabolic activities. In this review, we discuss how autophagy modulates the stability and functionality of Tregs in different disease settings, and provide a perspective on how manipulation of autophagy enables better control of immune response by targeting the generation of Tregs and the maintenance of their stability.
Collapse
|
37
|
Germic N, Frangez Z, Yousefi S, Simon HU. Regulation of the innate immune system by autophagy: neutrophils, eosinophils, mast cells, NK cells. Cell Death Differ 2019; 26:703-714. [PMID: 30737478 PMCID: PMC6460399 DOI: 10.1038/s41418-019-0295-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 12/15/2022] Open
Abstract
Autophagy is an evolutionally conserved, highly regulated catabolic process that combines cellular functions required for the regulation of metabolic balance under conditions of stress with those needed for the degradation of damaged cell organelles via the lysosomal machinery. The importance of autophagy for cell homeostasis and survival has long been appreciated. Recent data suggest that autophagy is also involved in non-metabolic functions that impact the immune system. Here, we reflect in two review articles the recent literature pointing to an important role for autophagy in innate immune cells. In this article, we focus on neutrophils, eosinophils, mast cells, and natural killer cells. We mainly discuss the influence of autophagy on functional cellular responses and its importance for overall host defense. In the companion review, we present the role of autophagy in the functions performed by monocytes/macrophages and dendritic cells.
Collapse
Affiliation(s)
- Nina Germic
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Ziva Frangez
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Shida Yousefi
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland.
- Department of Clinical Immunology and Allergology, Sechenov University, Moscow, Russia.
| |
Collapse
|
38
|
Jung HE, Shim YR, Oh JE, Oh DS, Lee HK. The autophagy Protein Atg5 Plays a Crucial Role in the Maintenance and Reconstitution Ability of Hematopoietic Stem Cells. Immune Netw 2019; 19:e12. [PMID: 31089439 PMCID: PMC6494762 DOI: 10.4110/in.2019.19.e12] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 01/17/2019] [Accepted: 01/21/2019] [Indexed: 12/19/2022] Open
Abstract
Hematopoietic stem cells (HSCs) in bone marrow are pluripotent cells that can constitute the hematopoiesis system through self-renewal and differentiation into immune cells and red blood cells. To ensure a competent hematopoietic system for life, the maintenance of HSCs is tightly regulated. Although autophagy, a self-degradation pathway for cell homeostasis, is essential for hematopoiesis, the role of autophagy key protein Atg5 in HSCs has not been thoroughly investigated. In this study, we found that Atg5 deficiency in hematopoietic cells causes survival defects, resulting in severe lymphopenia and anemia in mice. In addition, the absolute numbers of HSCs and multiple-lineage progenitor cells were significantly decreased, and abnormal erythroid development resulted in reduced erythrocytes in blood of Vav_Atg5−/− mice. The proliferation of Lin−Sca-1+c-Kit+ HSCs was aberrant in bone marrow of Vav_Atg5−/− mice, and mature progenitors and terminally differentiated cells were also significantly altered. Furthermore, the reconstitution ability of HSCs in bone marrow chimeric mice was significantly decreased in the presence of Atg5 deficiency in HSCs. Mechanistically, impairment of autophagy-mediated clearance of damaged mitochondria was the underlying cause of the HSC functional defects. Taken together, these results define the crucial role of Atg5 in the maintenance and the reconstitution ability of HSCs.
Collapse
Affiliation(s)
- Hi Eun Jung
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Ye Ri Shim
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Ji Eun Oh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Dong Sun Oh
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Heung Kyu Lee
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea.,KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| |
Collapse
|
39
|
Therapeutic Modulation of Autophagy in Leukaemia and Lymphoma. Cells 2019; 8:cells8020103. [PMID: 30704144 PMCID: PMC6406467 DOI: 10.3390/cells8020103] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/26/2019] [Accepted: 01/28/2019] [Indexed: 02/07/2023] Open
Abstract
Haematopoiesis is a tightly orchestrated process where a pool of hematopoietic stem and progenitor cells (HSPCs) with high self-renewal potential can give rise to both lymphoid and myeloid lineages. The HSPCs pool is reduced with ageing resulting in few HSPC clones maintaining haematopoiesis thereby reducing blood cell diversity, a phenomenon called clonal haematopoiesis. Clonal expansion of HSPCs carrying specific genetic mutations leads to increased risk for haematological malignancies. Therefore, it comes as no surprise that hematopoietic tumours develop in higher frequency in elderly people. Unfortunately, elderly patients with leukaemia or lymphoma still have an unsatisfactory prognosis compared to younger ones highlighting the need to develop more efficient therapies for this group of patients. Growing evidence indicates that macroautophagy (hereafter referred to as autophagy) is essential for health and longevity. This review is focusing on the role of autophagy in normal haematopoiesis as well as in leukaemia and lymphoma development. Attenuated autophagy may support early hematopoietic neoplasia whereas activation of autophagy in later stages of tumour development and in response to a variety of therapies rather triggers a pro-tumoral response. Novel insights into the role of autophagy in haematopoiesis will be discussed in light of designing new autophagy modulating therapies in hematopoietic cancers.
Collapse
|
40
|
Iida T, Yokoyama Y, Wagatsuma K, Hirayama D, Nakase H. Impact of Autophagy of Innate Immune Cells on Inflammatory Bowel Disease. Cells 2018; 8:cells8010007. [PMID: 30583538 PMCID: PMC6356773 DOI: 10.3390/cells8010007] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 12/18/2018] [Accepted: 12/18/2018] [Indexed: 12/13/2022] Open
Abstract
Autophagy, an intracellular degradation mechanism, has many immunological functions and is a constitutive process necessary for maintaining cellular homeostasis and organ structure. One of the functions of autophagy is to control the innate immune response. Many studies conducted in recent years have revealed the contribution of autophagy to the innate immune response, and relationships between this process and various diseases have been reported. Inflammatory bowel disease is an intractable disorder with unknown etiology; however, immunological abnormalities in the intestines are known to be involved in the pathology of inflammatory bowel disease, as is dysfunction of autophagy. In Crohn's disease, many associations with autophagy-related genes, such as ATG16L1, IRGM, NOD2, and others, have been reported. Abnormalities in the ATG16L1 gene, in particular, have been reported to cause autophagic dysfunction, resulting in enhanced production of inflammatory cytokines by macrophages as well as abnormal function of Paneth cells, which are important in intestinal innate immunity. In this review, we provide an overview of the autophagy mechanism in innate immune cells in inflammatory bowel disease.
Collapse
Affiliation(s)
- Tomoya Iida
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo 060-8543, Japan.
| | - Yoshihiro Yokoyama
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo 060-8543, Japan.
| | - Kohei Wagatsuma
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo 060-8543, Japan.
| | - Daisuke Hirayama
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo 060-8543, Japan.
| | - Hiroshi Nakase
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo 060-8543, Japan.
| |
Collapse
|
41
|
Lee NR, Ban J, Lee NJ, Yi CM, Choi JY, Kim H, Lee JK, Seong J, Cho NH, Jung JU, Inn KS. Activation of RIG-I-Mediated Antiviral Signaling Triggers Autophagy Through the MAVS-TRAF6-Beclin-1 Signaling Axis. Front Immunol 2018; 9:2096. [PMID: 30258449 PMCID: PMC6143786 DOI: 10.3389/fimmu.2018.02096] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/24/2018] [Indexed: 12/26/2022] Open
Abstract
Autophagy has been implicated in innate immune responses against various intracellular pathogens. Recent studies have reported that autophagy can be triggered by pathogen recognizing sensors, including Toll-like receptors and cyclic guanosine monophosphate-adenosine monophosphate synthase, to participate in innate immunity. In the present study, we examined whether the RIG-I signaling pathway, which detects viral infections by recognizing viral RNA, triggers the autophagic process. The introduction of polyI:C into the cytoplasm, or Sendai virus infection, significantly induced autophagy in normal cells but not in RIG-I-deficient cells. PolyI:C transfection or Sendai virus infection induced autophagy in the cells lacking type-I interferon signaling. This demonstrated that the effect was not due to interferon signaling. RIG-I-mediated autophagy diminished by the deficiency of mitochondrial antiviral signaling protein (MAVS) or tumor necrosis factor receptor-associated factor (TRAF)6, showing that the RIG-I-MAVS-TRAF6 signaling axis was critical for RIG-I-mediated autophagy. We also found that Beclin-1 was translocated to the mitochondria, and it interacted with TRAF6 upon RIG-I activation. Furthermore, Beclin-1 underwent K63-polyubiquitination upon RIG-I activation, and the ubiquitination decreased in TRAF6-deficient cells. This suggests that the RIG-I-MAVS-TRAF6 axis induced K63-linked polyubiquitination of Beclin-1, which has been implicated in triggering autophagy. As deficient autophagy increases the type-I interferon response, the induction of autophagy by the RIG-I pathway might also contribute to preventing an excessive interferon response as a negative-feedback mechanism.
Collapse
Affiliation(s)
- Na-Rae Lee
- Department of Fundamental Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Junsu Ban
- Department of Fundamental Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Noh-Jin Lee
- Department of Fundamental Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Chae-Min Yi
- Department of Fundamental Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Ji-Yoon Choi
- Department of Fundamental Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Hyunbin Kim
- KHU-KIST Department of Converging Science and Technology, Graduate School, Kyung Hee University, Seoul, South Korea
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul, South Korea
| | - Jong Kil Lee
- Department of Fundamental Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Jihye Seong
- KHU-KIST Department of Converging Science and Technology, Graduate School, Kyung Hee University, Seoul, South Korea
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul, South Korea
| | - Nam-Hyuk Cho
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea
| | - Jae U. Jung
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Kyung-Soo Inn
- Department of Fundamental Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, South Korea
- KHU-KIST Department of Converging Science and Technology, Graduate School, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
42
|
Arbogast F, Gros F. Lymphocyte Autophagy in Homeostasis, Activation, and Inflammatory Diseases. Front Immunol 2018; 9:1801. [PMID: 30127786 PMCID: PMC6087746 DOI: 10.3389/fimmu.2018.01801] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 07/23/2018] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a catabolic mechanism, allowing the degradation of cytoplasmic content via lysosomal activity. Several forms of autophagy are described in mammals. Macroautophagy leads to integration of cytoplasmic portions into vesicles named autophagosomes that ultimately fuse with lysosomes. Chaperone-mediated autophagy is in contrast the direct translocation of protein in lysosomes. Macroautophagy is central to lymphocyte homeostasis. Although its role is controversial in lymphocyte development and in naive cell survival, it seems particularly involved in the maintenance of certain lymphocyte subtypes. Its importance in memory B and T cells biology has recently emerged. Moreover, some effector cells like plasma cells rely on autophagy for survival. Autophagy is central to glucose and lipid metabolism, and to the maintenance of organelles like mitochondria and endoplasmic reticulum. In addition macroautophagy, or individual components of its machinery, are also actors in antigen presentation by B cells, a crucial step to receive help from T cells, this crosstalk favoring their final differentiation into memory or plasma cells. Autophagy is deregulated in several autoimmune or autoinflammatory diseases like systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, and Crohn’s disease. Some treatments used in these pathologies impact autophagic activity, even if the causal link between autophagy regulation and the efficiency of the treatments has not yet been clearly established. In this review, we will first discuss the mechanisms linking autophagy to lymphocyte subtype survival and the signaling pathways involved. Finally, potential impacts of autophagy modulation in lymphocytes on the course of these diseases will be approached.
Collapse
Affiliation(s)
- Florent Arbogast
- CNRS UPR3572, Immunology, Immunopathology and Therapeutic Chemistry/Laboratory of Excellence MEDALIS, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France.,University of Strasbourg, Strasbourg, France
| | - Frédéric Gros
- CNRS UPR3572, Immunology, Immunopathology and Therapeutic Chemistry/Laboratory of Excellence MEDALIS, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France.,University of Strasbourg, Strasbourg, France
| |
Collapse
|
43
|
Mikami Y, Takada Y, Hagihara Y, Kanai T. Innate lymphoid cells in organ fibrosis. Cytokine Growth Factor Rev 2018; 42:27-36. [PMID: 30104153 DOI: 10.1016/j.cytogfr.2018.07.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 07/24/2018] [Indexed: 02/06/2023]
Abstract
Innate lymphoid cells (ILCs) are a recently identified family of lymphoid effector cells. ILCs are mainly clustered into 3 groups based on their unique cytokine profiles and transcription factors typically attributed to the subsets of T helper cells. ILCs have a critical role in the mucosal immune response through promptly responding to pathogens and producing large amount of effector cytokines of type 1, 2, or 3 responses. In addition to the role of early immune responses against infections, ILCs, particularly group 2 ILCs (ILC2), have recently gained attention for modulating remodeling and fibrosis especially in the mucosal tissues. Herein, we overview the current knowledge in this area, highlighting roles of ILCs on fibrosis in the mucosal tissues, especially focusing on the gut and lung. We also discuss some new directions for future research by extrapolating from knowledge derived from studies on Th cells.
Collapse
Affiliation(s)
- Yohei Mikami
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan.
| | - Yoshiaki Takada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Yuya Hagihara
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, 100-0004, Japan
| |
Collapse
|
44
|
Rapp M, Wiedemann GM, Sun JC. Memory responses of innate lymphocytes and parallels with T cells. Semin Immunopathol 2018; 40:343-355. [PMID: 29808388 PMCID: PMC6054893 DOI: 10.1007/s00281-018-0686-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/04/2018] [Indexed: 12/23/2022]
Abstract
Natural killer (NK) cells are classified as innate immune cells, given their ability to rapidly respond and kill transformed or virally infected cells without prior sensitization. Recently, accumulating evidence suggests that NK cells also exhibit many characteristics similar to cells of the adaptive immune system. Analogous to T cells, NK cells acquire self-tolerance during development, express antigen-specific receptors, undergo clonal-like expansion, and can become long-lived, self-renewing memory cells with potent effector function providing potent protection against reappearing pathogens. In this review, we discuss the requirements for memory NK cell generation and highlight the similarities with the formation of memory T cells.
Collapse
Affiliation(s)
- Moritz Rapp
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
- Immunology Program, Memorial Sloan Kettering Cancer Center, 408 East 69th Street, ZRC-1462, New York, NY, 10065, USA
| | - Gabriela M Wiedemann
- Immunology Program, Memorial Sloan Kettering Cancer Center, 408 East 69th Street, ZRC-1462, New York, NY, 10065, USA
| | - Joseph C Sun
- Immunology Program, Memorial Sloan Kettering Cancer Center, 408 East 69th Street, ZRC-1462, New York, NY, 10065, USA.
- Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY, 10065, USA.
| |
Collapse
|
45
|
Louis C, Burns C, Wicks I. TANK-Binding Kinase 1-Dependent Responses in Health and Autoimmunity. Front Immunol 2018; 9:434. [PMID: 29559975 PMCID: PMC5845716 DOI: 10.3389/fimmu.2018.00434] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 02/19/2018] [Indexed: 01/05/2023] Open
Abstract
The pathogenesis of autoimmune diseases, such as rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE) is driven by genetic predisposition and environmental triggers that lead to dysregulated immune responses. These include the generation of pathogenic autoantibodies and aberrant production of inflammatory cytokines. Current therapies for RA and other autoimmune diseases reduce inflammation by targeting inflammatory mediators, most of which are innate response cytokines, resulting in generalized immunosuppression. Overall, this strategy has been very successful, but not all patients respond, responses can diminish over time and numerous side effects can occur. Therapies that target the germinal center (GC) reaction and/or antibody-secreting plasma cells (PC) potentially provide a novel approach. TANK-binding kinase 1 (TBK1) is an IKK-related serine/threonine kinase best characterized for its involvement in innate antiviral responses through the induction of type I interferons. TBK1 is also gaining attention for its roles in humoral immune responses. In this review, we discuss the role of TBK1 in immunological pathways involved in the development and maintenance of antibody responses, with particular emphasis on its potential relevance in the pathogenesis of humoral autoimmunity. First, we review the role of TBK1 in the induction of type I IFNs. Second, we highlight how TBK1 mediates inducible T cell co-stimulator signaling to the GC T follicular B helper population. Third, we discuss emerging evidence on the contribution of TBK1 to autophagic pathways and the potential implications for immune cell function. Finally, we discuss the therapeutic potential of TBK1 inhibition in autoimmunity.
Collapse
Affiliation(s)
- Cynthia Louis
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Chris Burns
- Chemical Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Ian Wicks
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Rheumatology Unit, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
46
|
Abstract
Mammalian cells harness autophagy to eliminate physiological byproducts of metabolism and cope with microenvironmental perturbations. Moreover, autophagy connects cellular adaptation with extracellular circuitries that impinge on immunity and metabolism. As it links transformed and non-transformed components of the tumour microenvironment, such an autophagic network is important for cancer initiation, progression and response to therapy. Here, we discuss the mechanisms whereby the autophagic network interfaces with multiple aspects of malignant disease.
Collapse
|
47
|
Wilhelm C, Kharabi Masouleh S, Kazakov A. Metabolic Regulation of Innate Lymphoid Cell-Mediated Tissue Protection-Linking the Nutritional State to Barrier Immunity. Front Immunol 2017; 8:1742. [PMID: 29375541 PMCID: PMC5770634 DOI: 10.3389/fimmu.2017.01742] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/23/2017] [Indexed: 12/14/2022] Open
Abstract
Innate lymphoid cells (ILC) are a recently described group of tissue-resident immune cells that play essential roles in maintaining and protecting the tissue barrier against invading pathogens. Extensive research has revealed that ILC-mediated immune responses are controlled by dietary components and metabolites. An additional role of ILC as important direct regulators of host metabolism and glucose tolerance is emerging. This suggests that ILC may act as key dietary sensors integrating nutritional and metabolic stress to facilitate both maintenance of barrier sites and a coordinated immune response protecting these tissues. In this respect, investigations have begun to determine how different ILC responses are metabolically fueled and the impact of nutrient availability on the regulation of ILC function. Here, we discuss the current literature concerning dietary and metabolic control of ILC. In particular, we address whether the dietary and metabolic control of ILC and their simultaneous influence on host metabolism may function as a coordinated program of barrier defense.
Collapse
Affiliation(s)
- Christoph Wilhelm
- Unit for Immunopathology, Department of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Schekufe Kharabi Masouleh
- Unit for Immunopathology, Department of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Alexander Kazakov
- Unit for Immunopathology, Department of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany
| |
Collapse
|
48
|
O'Sullivan TE, Sun JC. Innate Lymphoid Cell Immunometabolism. J Mol Biol 2017; 429:3577-3586. [PMID: 28867535 PMCID: PMC5719889 DOI: 10.1016/j.jmb.2017.08.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 08/01/2017] [Accepted: 08/21/2017] [Indexed: 02/06/2023]
Abstract
Innate lymphoid cells (ILCs) are tissue-resident "first responders" of the immune system that function to protect epithelial barriers against pathogens and maintain tissue homeostasis. However, because ILCs are finely tuned to perturbations within tissue microenvironments, they can also contribute to host pathology when upstream activating signals are dysregulated. Recent work has demonstrated that the crosstalk between ILCs and their environment has a significant impact on host metabolism in health and disease. In this brief review, we summarize recent studies that demonstrate the ability of ILCs to influence tissue and systemic metabolism, as well as how ILC biology can be regulated by environmental changes in host metabolism. We also highlight studies showing how ILC-intrinsic metabolism influences their activation, proliferation, and homeostasis. Finally, this review discusses the challenges and open questions in the rapidly expanding field of immunometabolism.
Collapse
Affiliation(s)
- Timothy E O'Sullivan
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States
| | - Joseph C Sun
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, United States; Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY 10065, United States.
| |
Collapse
|
49
|
Abstract
The cellular degradative pathway of autophagy has a fundamental role in immunity. Here, we review the function of autophagy and autophagy proteins in inflammation. We discuss how the autophagy machinery controls the burden of infectious agents while simultaneously limiting inflammatory pathologies, which often involves processes that are distinct from conventional autophagy. Among the newly emerging processes we describe are LC3-associated phagocytosis and targeting by autophagy proteins, both of which require many of the same proteins that mediate conventional autophagy. We also discuss how autophagy contributes to differentiation of myeloid and lymphoid cell types, coordinates multicellular immunity, and facilitates memory responses. Together, these functions establish an intimate link between autophagy, mucosal immunity, and chronic inflammatory diseases. Finally, we offer our perspective on current challenges and barriers to translation.
Collapse
Affiliation(s)
- Yu Matsuzawa-Ishimoto
- Kimmel Center for Biology and Medicine at the Skirball Institute and.,Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA; ,
| | - Seungmin Hwang
- Department of Pathology, The University of Chicago, Chicago, Illinois 60637, USA;
| | - Ken Cadwell
- Kimmel Center for Biology and Medicine at the Skirball Institute and.,Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA; ,
| |
Collapse
|
50
|
Withers DR, Hepworth MR. Group 3 Innate Lymphoid Cells: Communications Hubs of the Intestinal Immune System. Front Immunol 2017; 8:1298. [PMID: 29085366 PMCID: PMC5649144 DOI: 10.3389/fimmu.2017.01298] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 09/27/2017] [Indexed: 12/12/2022] Open
Abstract
The maintenance of mammalian health requires the generation of appropriate immune responses against a broad range of environmental and microbial challenges, which are continually encountered at barrier tissue sites including the skin, lung, and gastrointestinal tract. Dysregulated barrier immune responses result in inflammation, both locally and systemically in peripheral organs. Group 3 innate lymphoid cells (ILC3) are constitutively present at barrier sites and appear to be highly specialized in their ability to sense a range of environmental and host-derived signals. Under homeostatic conditions, ILC3 respond to local cues to maintain tissue homeostasis and restrict inflammatory responses. In contrast, perturbations in the tissue microenvironment resulting from disease, infection, or tissue damage can drive dysregulated pro-inflammatory ILC3 responses and contribute to immunopathology. The tone of the ILC3 response is dictated by a balance of “exogenous” signals, such as dietary metabolites and commensal microbes, and “endogenous” host-derived signals from stromal cells, immune cells, and the nervous system. ILC3 must therefore have the capacity to simultaneously integrate a wide array of complex and dynamic inputs in order to regulate barrier function and tissue health. In this review, we discuss the concept of ILC3 as a “communications hub” in the intestinal tract and associated lymphoid tissues and address the variety of signals, derived from multiple biological systems, which are interpreted by ILC3 to modulate the release of downstream effector molecules and regulate cell–cell crosstalk. Successful integration of environmental cues by ILC3 and downstream propagation to the broader immune system is required to maintain a tolerogenic and anti-inflammatory tone and reinforce barrier function, whereas dysregulation of ILC3 responses can contribute to the onset or progression of clinically relevant chronic inflammatory diseases.
Collapse
Affiliation(s)
- David R Withers
- College of Medical and Dental Sciences, Institute of Immunology and Immunotherapy (III), University of Birmingham, Birmingham, United Kingdom
| | - Matthew R Hepworth
- Manchester Collaborative Centre for Inflammation Research (MCCIR), Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|