1
|
He Z, Li F, Yan J, Liu M, Chen Y, Guo C. The dual role of autophagy during porcine reproductive and respiratory syndrome virus infection: A review. Int J Biol Macromol 2024; 282:136978. [PMID: 39471930 DOI: 10.1016/j.ijbiomac.2024.136978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/12/2024] [Accepted: 10/25/2024] [Indexed: 11/01/2024]
Abstract
Autophagy is a highly conserved catabolic process that transports cellular components to lysosomes for degradation and reuse. It impacts various cellular functions, including innate and adaptive immunity. It can exhibit a dual role in viral infections, either promoting or inhibiting viral replication depending on the virus and the stage of the infection cycle. Porcine reproductive and respiratory syndrome virus (PRRSV) is a significant pathogen impacting the sustainable development of the global pork industry. Recent research has shown that PRRSV has evolved specific mechanisms to facilitate or impede autophagosome maturation, thereby evading innate and adaptive immune responses. These primary mechanisms involve viral proteins that target multiple regulators of autophagosome formation, including autophagy receptors, tethering proteins, autophagy-related (ATG) genes, as well as the functional proteins of autophagosomes and late endosomes/lysosomes. Additionally, these mechanisms are related to the post-translational modification of key components, viral antigens for presentation to T lymphocytes, interferon production, and the biogenesis and function of lysosomes. This review discusses the specific mechanisms by which PRRSV targets autophagy in host defence and virus survival, summarizes the role of viral proteins in subverting the autophagic process, and examines how the host utilizes the antiviral functions of autophagy to prevent PRRSV infection.
Collapse
Affiliation(s)
- Zhan He
- Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, PR China
| | - Fangfang Li
- Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, PR China
| | - Jiecong Yan
- Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, PR China
| | - Min Liu
- Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, PR China
| | - Yongjie Chen
- Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, PR China
| | - Chunhe Guo
- Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
2
|
Judith D, Versapuech M, Bejjani F, Palaric M, Verlhac P, Kuster A, Lepont L, Gallois-Montbrun S, Janvier K, Berlioz-Torrent C. ATG5 selectively engages virus-tethered BST2/tetherin in an LC3C-associated pathway. Proc Natl Acad Sci U S A 2023; 120:e2217451120. [PMID: 37155854 PMCID: PMC10193943 DOI: 10.1073/pnas.2217451120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 03/15/2023] [Indexed: 05/10/2023] Open
Abstract
Bone marrow stromal antigen 2 (BST2)/tetherin is a restriction factor that reduces HIV-1 dissemination by tethering virus at the cell surface. BST2 also acts as a sensor of HIV-1 budding, establishing a cellular antiviral state. The HIV-1 Vpu protein antagonizes BST2 antiviral functions via multiple mechanisms, including the subversion of an LC3C-associated pathway, a key cell intrinsic antimicrobial mechanism. Here, we describe the first step of this viral-induced LC3C-associated process. This process is initiated at the plasma membrane through the recognition and internalization of virus-tethered BST2 by ATG5, an autophagy protein. ATG5 and BST2 assemble as a complex, independently of the viral protein Vpu and ahead of the recruitment of the ATG protein LC3C. The conjugation of ATG5 with ATG12 is dispensable for this interaction. ATG5 recognizes cysteine-linked homodimerized BST2 and specifically engages phosphorylated BST2 tethering viruses at the plasma membrane, in an LC3C-associated pathway. We also found that this LC3C-associated pathway is used by Vpu to attenuate the inflammatory responses mediated by virion retention. Overall, we highlight that by targeting BST2 tethering viruses, ATG5 acts as a signaling scaffold to trigger an LC3C-associated pathway induced by HIV-1 infection.
Collapse
Affiliation(s)
- Delphine Judith
- Université Paris Cité, Institut Cochin, INSERM, CNRS, F-75014Paris, France
| | - Margaux Versapuech
- Université Paris Cité, Institut Cochin, INSERM, CNRS, F-75014Paris, France
| | - Fabienne Bejjani
- Université Paris Cité, Institut Cochin, INSERM, CNRS, F-75014Paris, France
| | - Marjory Palaric
- Université Paris Cité, Institut Cochin, INSERM, CNRS, F-75014Paris, France
| | - Pauline Verlhac
- Université Paris Cité, Institut Cochin, INSERM, CNRS, F-75014Paris, France
| | - Aurelia Kuster
- Université Paris Cité, Institut Cochin, INSERM, CNRS, F-75014Paris, France
| | - Leslie Lepont
- Université Paris Cité, Institut Cochin, INSERM, CNRS, F-75014Paris, France
| | | | - Katy Janvier
- Université Paris Cité, Institut Cochin, INSERM, CNRS, F-75014Paris, France
| | | |
Collapse
|
3
|
The role of lysosomes in metabolic and autoimmune diseases. Nat Rev Nephrol 2023; 19:366-383. [PMID: 36894628 DOI: 10.1038/s41581-023-00692-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2023] [Indexed: 03/11/2023]
Abstract
Lysosomes are catabolic organelles that contribute to the degradation of intracellular constituents through autophagy and of extracellular components through endocytosis, phagocytosis and macropinocytosis. They also have roles in secretory mechanisms, the generation of extracellular vesicles and certain cell death pathways. These functions make lysosomes central organelles in cell homeostasis, metabolic regulation and responses to environment changes including nutrient stresses, endoplasmic reticulum stress and defects in proteostasis. Lysosomes also have important roles in inflammation, antigen presentation and the maintenance of long-lived immune cells. Their functions are tightly regulated by transcriptional modulation via TFEB and TFE3, as well as by major signalling pathways that lead to activation of mTORC1 and mTORC2, lysosome motility and fusion with other compartments. Lysosome dysfunction and alterations in autophagy processes have been identified in a wide variety of diseases, including autoimmune, metabolic and kidney diseases. Deregulation of autophagy can contribute to inflammation, and lysosomal defects in immune cells and/or kidney cells have been reported in inflammatory and autoimmune pathologies with kidney involvement. Defects in lysosomal activity have also been identified in several pathologies with disturbances in proteostasis, including autoimmune and metabolic diseases such as Parkinson disease, diabetes mellitus and lysosomal storage diseases. Targeting lysosomes is therefore a potential therapeutic strategy to regulate inflammation and metabolism in a variety of pathologies.
Collapse
|
4
|
Peña-Martinez C, Rickman AD, Heckmann BL. Beyond autophagy: LC3-associated phagocytosis and endocytosis. SCIENCE ADVANCES 2022; 8:eabn1702. [PMID: 36288309 PMCID: PMC9604515 DOI: 10.1126/sciadv.abn1702] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 07/26/2022] [Indexed: 05/08/2023]
Abstract
Noncanonical functions of the autophagy machinery in pathways including LC3-associated phagocytosis and LC3-associated endocytosis have garnered increasing interest in both normal physiology and pathobiology. New discoveries over the past decade of noncanonical uses of the autophagy machinery in these distinct molecular mechanisms have led to robust investigation into the roles of single-membrane LC3 lipidation. Noncanonical autophagy pathways have now been implicated in the regulation of multiple processes ranging from debris clearance, cellular signaling, and immune regulation and inflammation. Accumulating evidence is demonstrating roles in a variety of disease states including host-pathogen responses, autoimmunity, cancer, and neurological and neurodegenerative pathologies. Here, we broadly summarize the differences in the mechanistic regulation between autophagy and LAP and LANDO and highlight some of the key roles of LAP and LANDO in innate immune function, inflammation, and disease pathology.
Collapse
Affiliation(s)
- Carolina Peña-Martinez
- Department of Molecular Medicine, USF Morsani College of Medicine, Tampa, FL, USA
- Byrd Alzheimer’s Center, USF Health Neuroscience Institute, Tampa, FL, USA
| | - Alexis D. Rickman
- Department of Molecular Medicine, USF Morsani College of Medicine, Tampa, FL, USA
- Byrd Alzheimer’s Center, USF Health Neuroscience Institute, Tampa, FL, USA
| | - Bradlee L. Heckmann
- Department of Molecular Medicine, USF Morsani College of Medicine, Tampa, FL, USA
- Byrd Alzheimer’s Center, USF Health Neuroscience Institute, Tampa, FL, USA
| |
Collapse
|
5
|
Wu B, Wang Q, Li B, Jiang M. LAMTOR1 degrades MHC-II via the endocytic in hepatocellular carcinoma. Carcinogenesis 2022; 43:1059-1070. [PMID: 36070764 PMCID: PMC9890926 DOI: 10.1093/carcin/bgac075] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 08/11/2022] [Accepted: 09/06/2022] [Indexed: 02/04/2023] Open
Abstract
Tumor cell surface antigen recognition is a major hallmark of cancer therapy, and loss of major histocompatibility complex class I (MHC-I) is the most common mechanism that impairs tumor cell surface antigen processing and expression. In addition to this, MHC-II regulates antigen presentation in CD4+ T cell immune responses involved in tumor killing by CD8+ T cells, whereas the regulation of endocytosis regulating MHC-II antigen presentation has not been reported. Therefore, the regulation of the endocytosis pathway on the expression of MHC-II surface level and antitumor T cell response remains to be explored. In this experiment, we found that LAMTOR1 regulates the endocytic pathway through the GTPase domain of DNM2 and triggers the formation of autophagosomes. We performed flow cytometry and western blotting analyses, which revealed that the expression of MHC-II molecules on the surface of cells is influenced by LAMTOR1 through the endocytic pathway. We showed that the expression of MHC-II molecules, which recognize CD4+ T cells on the surface of cells, was regulated by LAMTOR1 through an endocytic pathway. By coculture experiments, we showed that CD8+/CD4+ T cells exhibit substantially higher levels of tumor cell apoptosis than those observed when hepatocellular carcinoma (HCC) cells were cocultured with CD8+ T cells alone. This study revealed that LAMTOR1 decreases the expression levels of MHC-II on cell surfaces in order to reduce antigen expression, leading to a decrease in antitumor T cell responses.
Collapse
Affiliation(s)
- Bo Wu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China
| | - Qian Wang
- Department of Radiology, The Fifth Hospital of Xiamen, Xiamen 361101, China
| | - Bowen Li
- Department of Oncological and Endoscopic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Meixi Jiang
- To whom correspondence should be addressed. Tel: +86 13782206769; Fax: +86 02462255001;
| |
Collapse
|
6
|
Nakamura M, Nakamura J, Mochizuki C, Kuroda C, Kato S, Haruta T, Kakefuda M, Sato S, Tamanoi F, Sugino N. Analysis of cell-nanoparticle interactions and imaging of in vitro labeled cells showing barcorded endosomes using fluorescent thiol-organosilica nanoparticles surface-functionalized with polyethyleneimine. NANOSCALE ADVANCES 2022; 4:2682-2703. [PMID: 36132282 PMCID: PMC9417756 DOI: 10.1039/d1na00839k] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 04/18/2022] [Indexed: 06/15/2023]
Abstract
Biomedical imaging using cell labeling is an important technique to visualize cell dynamics in the body. To label cells, thiol-organosilica nanoparticles (thiol-OS) containing fluorescein (thiol-OS/Flu) and rhodamine B (thiol-OS/Rho) were surface-functionalized with polyethyleneimine (PEI) (OS/Flu-PEI and OS/Rho-PEI) with 4 molecular weights (MWs). We hypothesized PEI structures such as brush, bent brush, bent lie-down, and coiled types on the surface depending on MWs based on dynamic light scattering and thermal gravimetric analyses. The labeling efficacy of OS/Flu-PEIs was dependent on the PEI MW and the cell type. A dual-particle administration study using thiol-OS and OS-PEIs revealed differential endosomal sorting of the particles depending on the surface of the NPs. The endosomes in the labeled cells using OS/Flu-PEI and thiol-OS/Rho revealed various patterns of fluorescence termed barcoded endosomes. The cells labeled with OS-PEI in vitro were administrated to mice intraperitoneally after in situ labeling of peritoneal cells using thiol-OS/Rho. The in vitro labeled cells were detected and identified in cell aggregates in vivo seamlessly. The labeled cells with barcoded endosomes were also identified in cell aggregates. Biomedical imaging of in vitro OS-PEI-labeled cells combined with in situ labeled cells showed high potential for observation of cell dynamics.
Collapse
Affiliation(s)
- Michihiro Nakamura
- Department of Organ Anatomy and Nanomedicine, Graduate School of Medicine, Yamaguchi University 1-1-1 Minami-Kogushi Ube Yamaguchi 755-8505 Japan
| | - Junna Nakamura
- Department of Organ Anatomy and Nanomedicine, Graduate School of Medicine, Yamaguchi University 1-1-1 Minami-Kogushi Ube Yamaguchi 755-8505 Japan
| | - Chihiro Mochizuki
- Department of Organ Anatomy and Nanomedicine, Graduate School of Medicine, Yamaguchi University 1-1-1 Minami-Kogushi Ube Yamaguchi 755-8505 Japan
| | - Chika Kuroda
- Department of Organ Anatomy and Nanomedicine, Graduate School of Medicine, Yamaguchi University 1-1-1 Minami-Kogushi Ube Yamaguchi 755-8505 Japan
| | - Shigeki Kato
- Department of Organ Anatomy and Nanomedicine, Graduate School of Medicine, Yamaguchi University 1-1-1 Minami-Kogushi Ube Yamaguchi 755-8505 Japan
| | | | - Mayu Kakefuda
- EM Application Group, EM Business Unit, JEOL Ltd. Japan
| | - Shun Sato
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Yamaguchi University 1-1-1 Minami-Kogushi Ube Yamaguchi 755-8505 Japan
| | - Fuyuhiko Tamanoi
- Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles CA 90095 USA
- Institute for Integrated Cell-Material Sciences, Institute for Advanced Study, Kyoto University Yoshida-Honmachi, Sakyo-ku Kyoto 606-8501 Japan
| | - Norihiro Sugino
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Yamaguchi University 1-1-1 Minami-Kogushi Ube Yamaguchi 755-8505 Japan
| |
Collapse
|
7
|
Khan A, Zhang K, Singh VK, Mishra A, Kachroo P, Bing T, Won JH, Mani A, Papanna R, Mann LK, Ledezma-Campos E, Aguillon-Duran G, Canaday DH, David SA, Restrepo BI, Viet NN, Phan H, Graviss EA, Musser JM, Kaushal D, Gauduin MC, Jagannath C. Human M1 macrophages express unique innate immune response genes after mycobacterial infection to defend against tuberculosis. Commun Biol 2022; 5:480. [PMID: 35590096 PMCID: PMC9119986 DOI: 10.1038/s42003-022-03387-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/21/2022] [Indexed: 12/23/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) is responsible for approximately 1.5 million deaths each year. Though 10% of patients develop tuberculosis (TB) after infection, 90% of these infections are latent. Further, mice are nearly uniformly susceptible to Mtb but their M1-polarized macrophages (M1-MΦs) can inhibit Mtb in vitro, suggesting that M1-MΦs may be able to regulate anti-TB immunity. We sought to determine whether human MΦ heterogeneity contributes to TB immunity. Here we show that IFN-γ-programmed M1-MΦs degrade Mtb through increased expression of innate immunity regulatory genes (Inregs). In contrast, IL-4-programmed M2-polarized MΦs (M2-MΦs) are permissive for Mtb proliferation and exhibit reduced Inregs expression. M1-MΦs and M2-MΦs express pro- and anti-inflammatory cytokine-chemokines, respectively, and M1-MΦs show nitric oxide and autophagy-dependent degradation of Mtb, leading to increased antigen presentation to T cells through an ATG-RAB7-cathepsin pathway. Despite Mtb infection, M1-MΦs show increased histone acetylation at the ATG5 promoter and pro-autophagy phenotypes, while increased histone deacetylases lead to decreased autophagy in M2-MΦs. Finally, Mtb-infected neonatal macaques express human Inregs in their lymph nodes and macrophages, suggesting that M1 and M2 phenotypes can mediate immunity to TB in both humans and macaques. We conclude that human MФ subsets show unique patterns of gene expression that enable differential control of TB after infection. These genes could serve as targets for diagnosis and immunotherapy of TB.
Collapse
Affiliation(s)
- Arshad Khan
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, USA
| | - Kangling Zhang
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Vipul K Singh
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, USA
| | - Abhishek Mishra
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, USA
| | - Priyanka Kachroo
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, USA
| | - Tian Bing
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jong Hak Won
- Department of Obstetrics, Gynecology and Reproductive Sciences, UTHSC, Houston, TX, USA
| | - Arunmani Mani
- Department of Obstetrics, Gynecology and Reproductive Sciences, UTHSC, Houston, TX, USA
| | - Ramesha Papanna
- Department of Obstetrics, Gynecology and Reproductive Sciences, UTHSC, Houston, TX, USA
| | - Lovepreet K Mann
- Department of Obstetrics, Gynecology and Reproductive Sciences, UTHSC, Houston, TX, USA
| | | | | | - David H Canaday
- Division of Infectious Disease, Case Western Reserve University Cleveland VA, Cleveland, OH, USA
| | - Sunil A David
- Virovax, LLC, Adjuvant Division, Lawrence, Kansas, USA
| | - Blanca I Restrepo
- UT School of Public Health, Brownsville, and STDOI, UT Rio Grande Valley, Brownsville, TX, USA
| | | | - Ha Phan
- Center for Promotion of Advancement of Society, Ha Noi, Vietnam
| | - Edward A Graviss
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, USA
| | - James M Musser
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, USA
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Marie Claire Gauduin
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, USA.
| |
Collapse
|
8
|
Aliahmad P, Miyake-Stoner SJ, Geall AJ, Wang NS. Next generation self-replicating RNA vectors for vaccines and immunotherapies. Cancer Gene Ther 2022:10.1038/s41417-022-00435-8. [PMID: 35194198 PMCID: PMC8861484 DOI: 10.1038/s41417-022-00435-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/18/2022] [Accepted: 01/28/2022] [Indexed: 01/06/2023]
Abstract
RNA technology has recently come to the forefront of innovative medicines and is being explored for a wide range of therapies, including prophylactic and therapeutic vaccines, biotherapeutic protein expression and gene therapy. In addition to conventional mRNA platforms now approved for prophylactic SARS-CoV2 vaccines, synthetic self-replicating RNA vaccines are currently being evaluated in the clinic for infectious disease and oncology. The prototypical srRNA vectors in clinical development are derived from alphaviruses, specifically Venezuelan Equine Encephalitis Virus (VEEV). While non-VEEV alphaviral strains have been explored as single cycle viral particles, their use as synthetic vectors largely remains under-utilized in clinical applications. Here we describe the potential commonalities and differences in synthetic alphaviral srRNA vectors in host cell interactions, immunogenicity, cellular delivery, and cargo expression. Thus, unlike the current thinking that VEEV-based srRNA is a one-size-fits-all platform, we argue that a new drug development approach leveraging panels of customizable, synthetic srRNA vectors will be required for clinical success.
Collapse
|
9
|
Dong F, Song X, Xing J, Tang X, Sheng X, Chi H, Zhan W. Immunological characteristics of dendritic cells marker CD83 in flounder (Paralichthys olivaceus). FISH AND SHELLFISH IMMUNOLOGY REPORTS 2021; 2:100030. [DOI: 10.1016/j.fsirep.2021.100030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/08/2021] [Accepted: 10/08/2021] [Indexed: 12/18/2022] Open
|
10
|
Daussy CF, Pied N, Wodrich H. Understanding Post Entry Sorting of Adenovirus Capsids; A Chance to Change Vaccine Vector Properties. Viruses 2021; 13:1221. [PMID: 34202573 PMCID: PMC8310329 DOI: 10.3390/v13071221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/25/2022] Open
Abstract
Adenovirus vector-based genetic vaccines have emerged as a powerful strategy against the SARS-CoV-2 health crisis. This success is not unexpected because adenoviruses combine many desirable features of a genetic vaccine. They are highly immunogenic and have a low and well characterized pathogenic profile paired with technological approachability. Ongoing efforts to improve adenovirus-vaccine vectors include the use of rare serotypes and non-human adenoviruses. In this review, we focus on the viral capsid and how the choice of genotypes influences the uptake and subsequent subcellular sorting. We describe how understanding capsid properties, such as stability during the entry process, can change the fate of the entering particles and how this translates into differences in immunity outcomes. We discuss in detail how mutating the membrane lytic capsid protein VI affects species C viruses' post-entry sorting and briefly discuss if such approaches could have a wider implication in vaccine and/or vector development.
Collapse
Affiliation(s)
| | | | - Harald Wodrich
- Microbiologie Fondamentale et Pathogénicité, MFP CNRS UMR 5234, University of Bordeaux, 146 rue Leo Saignat, CEDEX, 33076 Bordeaux, France; (C.F.D.); (N.P.)
| |
Collapse
|
11
|
Bustos SO, Antunes F, Rangel MC, Chammas R. Emerging Autophagy Functions Shape the Tumor Microenvironment and Play a Role in Cancer Progression - Implications for Cancer Therapy. Front Oncol 2020; 10:606436. [PMID: 33324568 PMCID: PMC7724038 DOI: 10.3389/fonc.2020.606436] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 10/22/2020] [Indexed: 12/15/2022] Open
Abstract
The tumor microenvironment (TME) is a complex environment where cancer cells reside and interact with different types of cells, secreted factors, and the extracellular matrix. Additionally, TME is shaped by several processes, such as autophagy. Autophagy has emerged as a conserved intracellular degradation pathway for clearance of damaged organelles or aberrant proteins. With its central role, autophagy maintains the cellular homeostasis and orchestrates stress responses, playing opposite roles in tumorigenesis. During tumor development, autophagy also mediates autophagy-independent functions associated with several hallmarks of cancer, and therefore exerting several effects on tumor suppression and/or tumor promotion mechanisms. Beyond the concept of degradation, new different forms of autophagy have been described as modulators of cancer progression, such as secretory autophagy enabling intercellular communication in the TME by cargo release. In this context, the synthesis of senescence-associated secretory proteins by autophagy lead to a senescent phenotype. Besides disturbing tumor treatment responses, autophagy also participates in innate and adaptive immune signaling. Furthermore, recent studies have indicated intricate crosstalk between autophagy and the epithelial-mesenchymal transition (EMT), by which cancer cells obtain an invasive phenotype and metastatic potential. Thus, autophagy in the cancer context is far broader and complex than just a cell energy sensing mechanism. In this scenario, we will discuss the key roles of autophagy in the TME and surrounding cells, contributing to cancer development and progression/EMT. Finally, the potential intervention in autophagy processes as a strategy for cancer therapy will be addressed.
Collapse
Affiliation(s)
- Silvina Odete Bustos
- Instituto do Cancer do Estado de São Paulo, Faculdade de Medicina de São Paulo, Brazil
| | - Fernanda Antunes
- Instituto do Cancer do Estado de São Paulo, Faculdade de Medicina de São Paulo, Brazil
| | - Maria Cristina Rangel
- Instituto do Cancer do Estado de São Paulo, Faculdade de Medicina de São Paulo, Brazil
| | - Roger Chammas
- Instituto do Cancer do Estado de São Paulo, Faculdade de Medicina de São Paulo, Brazil
| |
Collapse
|
12
|
Muñoz-Sánchez S, van der Vaart M, Meijer AH. Autophagy and Lc3-Associated Phagocytosis in Zebrafish Models of Bacterial Infections. Cells 2020; 9:cells9112372. [PMID: 33138004 PMCID: PMC7694021 DOI: 10.3390/cells9112372] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/24/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023] Open
Abstract
Modeling human infectious diseases using the early life stages of zebrafish provides unprecedented opportunities for visualizing and studying the interaction between pathogens and phagocytic cells of the innate immune system. Intracellular pathogens use phagocytes or other host cells, like gut epithelial cells, as a replication niche. The intracellular growth of these pathogens can be counteracted by host defense mechanisms that rely on the autophagy machinery. In recent years, zebrafish embryo infection models have provided in vivo evidence for the significance of the autophagic defenses and these models are now being used to explore autophagy as a therapeutic target. In line with studies in mammalian models, research in zebrafish has shown that selective autophagy mediated by ubiquitin receptors, such as p62, is important for host resistance against several bacterial pathogens, including Shigella flexneri, Mycobacterium marinum, and Staphylococcus aureus. Furthermore, an autophagy related process, Lc3-associated phagocytosis (LAP), proved host beneficial in the case of Salmonella Typhimurium infection but host detrimental in the case of S. aureus infection, where LAP delivers the pathogen to a replication niche. These studies provide valuable information for developing novel therapeutic strategies aimed at directing the autophagy machinery towards bacterial degradation.
Collapse
|
13
|
Cicco S, Cicco G, Racanelli V, Vacca A. Neutrophil Extracellular Traps (NETs) and Damage-Associated Molecular Patterns (DAMPs): Two Potential Targets for COVID-19 Treatment. Mediators Inflamm 2020; 2020:7527953. [PMID: 32724296 PMCID: PMC7366221 DOI: 10.1155/2020/7527953] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/11/2020] [Accepted: 06/30/2020] [Indexed: 12/17/2022] Open
Abstract
COVID-19 is a pandemic disease caused by the new coronavirus SARS-CoV-2 that mostly affects the respiratory system. The consequent inflammation is not able to clear viruses. The persistent excessive inflammatory response can build up a clinical picture that is very difficult to manage and potentially fatal. Modulating the immune response plays a key role in fighting the disease. One of the main defence systems is the activation of neutrophils that release neutrophil extracellular traps (NETs) under the stimulus of autophagy. Various molecules can induce NETosis and autophagy; some potent activators are damage-associated molecular patterns (DAMPs) and, in particular, the high-mobility group box 1 (HMGB1). This molecule is released by damaged lung cells and can induce a robust innate immunity response. The increase in HMGB1 and NETosis could lead to sustained inflammation due to SARS-CoV-2 infection. Therefore, blocking these molecules might be useful in COVID-19 treatment and should be further studied in the context of targeted therapy.
Collapse
Affiliation(s)
- Sebastiano Cicco
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro Medical School, Piazza G. Cesare 11, I-70124 Bari, Italy
| | - Gerolamo Cicco
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro Medical School, Piazza G. Cesare 11, I-70124 Bari, Italy
| | - Vito Racanelli
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro Medical School, Piazza G. Cesare 11, I-70124 Bari, Italy
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro Medical School, Piazza G. Cesare 11, I-70124 Bari, Italy
| |
Collapse
|
14
|
Liang X, Liu L, Wang Y, Guo H, Fan H, Zhang C, Hou L, Liu Z. Autophagy-driven NETosis is a double-edged sword - Review. Biomed Pharmacother 2020; 126:110065. [PMID: 32200255 DOI: 10.1016/j.biopha.2020.110065] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 02/25/2020] [Accepted: 02/28/2020] [Indexed: 02/06/2023] Open
Abstract
Autophagy is a cellular mechanism responsible for delivering protein aggregates or damaged organelles to lysosomes for degradation. It is also simultaneously a precise regulatory process, which is crucial for dealing with hunger, oxidative stress, and pathogen defense. Neutrophil Extracellular Traps (NETs), which form a part of a newly described bactericidal process, are reticular structures composed of a DNA backbone and multiple functional proteins, formed via a process known as NETosis. NETs exert their anti-infection activity by capturing pathogenic microorganisms, inhibiting their spread and inactivating virulence factors. However, NETs may also activate an immune response in non-infectious diseases, leading to tissue damage. Although the mechanism underlying this phenomenon is unclear, a large number of studies have suggested that autophagy may be involved. Autophagy-mediated NETs not only induce inflammation and tissue damage, but can also lead to cell senescence, malignant transformation, and cell death. Autophagy-dependent NETs also play a beneficial role in the hostwith respect to pathogen clearance and immune defense. Through careful review of the literature, we have found that the distinct roles of autophagy in NETosis may be dependent on the extent of autophagy and the specific manner in which it was induced. This article summarizes numerous recent studies, and reviews the role of autophagy-driven NETosis in various diseases, in the hope that this will lead to the development of more effective treatments.
Collapse
Affiliation(s)
- Xiaofei Liang
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, 161005, China; Department of Laboratory Medicine, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, 161005, China
| | - Li Liu
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, 161005, China; Department of Laboratory Medicine, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, 161005, China.
| | - Yan Wang
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, 161005, China; Department of Laboratory Medicine, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, 161005, China
| | - Haipeng Guo
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, 161005, China; Department of Laboratory Medicine, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, 161005, China
| | - Hua Fan
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, 161005, China; Department of Laboratory Medicine, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, 161005, China
| | - Chao Zhang
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, 161005, China; Department of Laboratory Medicine, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, 161005, China
| | - Lili Hou
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, 161005, China; Department of Laboratory Medicine, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, 161005, China
| | - Zhibo Liu
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, 161005, China; Department of Laboratory Medicine, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, 161005, China
| |
Collapse
|
15
|
Kang R, Zeh H, Lotze M, Tang D. The Multifaceted Effects of Autophagy on the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1225:99-114. [PMID: 32030650 DOI: 10.1007/978-3-030-35727-6_7] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The tumor microenvironment is composed of cancer cells, noncancer cells (e.g., immune cells, stromal cells, endothelial cells, and adipocytes), and various mediators (e.g., cytokines, chemokines, growth factors, and humoral factors) that work together to support cancer growth, progression, and resistance to therapies. Autophagy is an evolutionarily conserved degradation mechanism by which various cytosolic cargos (e.g., damaged organelles, unused molecules, or invaded pathogens) are engulfed by double-membrane autophagosomes, and then delivered into the lysosome for degradation and recycling. The level of autophagy is a crucial threshold to either promote cell survival or induce cell death in response to environmental stresses. Autophagy plays a context-dependent role in tumorigenesis and anticancer therapy via shaping the inflammatory, hypoxic, immunosuppressive, and metabolic tumor microenvironment. In particular, impaired autophagy flux is associated with chronic inflammation, immunosuppression, stromal formation, cancer stemness, angiogenesis, metastasis, and metabolic reprogramming in the tumor microenvironment. Understanding the molecular machinery of autophagy and its communication with hallmarks of cancer could lead to potential new anticancer strategies or drugs.
Collapse
Affiliation(s)
- Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Herbert Zeh
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Michael Lotze
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
16
|
Ishimwe N, Zhang W, Qian J, Zhang Y, Wen L. Autophagy regulation as a promising approach for improving cancer immunotherapy. Cancer Lett 2020; 475:34-42. [PMID: 32014460 DOI: 10.1016/j.canlet.2020.01.034] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/24/2020] [Accepted: 01/29/2020] [Indexed: 02/07/2023]
Abstract
Autophagy plays a critical role in intracellular metabolism and maintaining cellular homeostasis. Certain tumor cells present a higher basal autophagy rate and autophagy inhibition can lead to impaired metabolic dysfunction in autophagy-dependent tumor cells. Autophagy status in immune cells dictates their fate and response to antigen; however, autophagy in immune cells may be beneficial or detrimental depending on the developmental stage of the cell and more specifically its degree of differentiation. Autophagy-deficient hosts present variations in many metabolites, proteins and enzymes that may have tumor-promoting or -inhibiting effects. The centrality of autophagy in the metabolism of some cancers and immune cells poses as a critical target whose mechanisms must be further unraveled to optimize patient response and prevent tumor recurrence.
Collapse
Affiliation(s)
- Nestor Ishimwe
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, 230027, PR China; Guangzhou First People's Hospital, School of Medicine and Institutes for Life Sciences, South China University of Technology, Guangzhou, Guangdong, 510006, PR China.
| | - Wenbin Zhang
- Guangzhou First People's Hospital, School of Medicine and Institutes for Life Sciences, South China University of Technology, Guangzhou, Guangdong, 510006, PR China
| | - Jieying Qian
- Guangzhou First People's Hospital, School of Medicine and Institutes for Life Sciences, South China University of Technology, Guangzhou, Guangdong, 510006, PR China
| | - Yunjiao Zhang
- Guangzhou First People's Hospital, School of Medicine and Institutes for Life Sciences, South China University of Technology, Guangzhou, Guangdong, 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, Guangdong, 510006, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, And Innovation Center for Tissue Restoration and Reconstruction, Guangzhou, Guangdong, 510006, PR China.
| | - Longping Wen
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, 230027, PR China; Guangzhou First People's Hospital, School of Medicine and Institutes for Life Sciences, South China University of Technology, Guangzhou, Guangdong, 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, Guangdong, 510006, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, And Innovation Center for Tissue Restoration and Reconstruction, Guangzhou, Guangdong, 510006, PR China.
| |
Collapse
|
17
|
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
|