1
|
Liu P, Ren D, Li G, Xu X, Presotto L, Liu W, Zhao N, Li D, Chen M, Wang J, Liu X, Zhao C, Lu L, Liu Q. Ectoparasites enhance survival by suppressing host exploration and limiting dispersal. Nat Commun 2025; 16:4318. [PMID: 40346081 PMCID: PMC12064801 DOI: 10.1038/s41467-025-59601-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 04/29/2025] [Indexed: 05/11/2025] Open
Abstract
Parasites enhance their fitness by manipulating host dispersal. However, the strategies used by ectoparasites to influence host movement and the underlying mechanisms remain poorly understood. Here, we show that ectoparasites alter metabolic activity in specific brain regions of mice, with evidence pointing to a potential role for microglial activation in the prefrontal cortex. This activation appears to contribute to synaptic changes and altered neuronal differentiation, particularly in GABAergic neurons. Consequently, exploratory behavior decreases-an effect likely mediated through the skin-brain axis. In both indoor and field experiments with striped hamsters, ectoparasites reduce host exploration and modify their dispersal patterns. This behavioral shift ultimately restricts the host's distribution, enabling parasites to avoid environmental pressures. Our findings reveal that ectoparasites limit host dispersal to improve their own fitness, offering key insights for parasite control strategies that promote health and preserve ecological stability within the One Health framework.
Collapse
Affiliation(s)
- Pengbo Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Dongsheng Ren
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Guichang Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiaoming Xu
- State Key Laboratory of Animal Biodiversity Conservation and Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Luca Presotto
- Department of physics G. Occhialini, University of Milano-Bicocca, Milano, MI, Italy
- Milan Centre for Neuroscience, University of Milano-Bicocca, Milan, Italy
| | - Wei Liu
- State Key Laboratory of Animal Biodiversity Conservation and Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Ning Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Dongmei Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Min Chen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jun Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiaobo Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Chunchun Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Liang Lu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
| | - Qiyong Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
| |
Collapse
|
2
|
Min R, Bai Y, Wang NR, Liu X. Gasdermins in pyroptosis, inflammation, and cancer. Trends Mol Med 2025:S1471-4914(25)00090-5. [PMID: 40307076 DOI: 10.1016/j.molmed.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025]
Abstract
Pyroptosis is a type of programmed inflammatory cell death characterized by balloon-like swelling, membrane rupture, and the release of inflammatory cytokines and danger signals. Pyroptosis is directly triggered by activated gasdermins (GSDMs) which bind to membrane phospholipids, oligomerize, and form pores in cell membranes. GSDM activation is mediated by various effector proteases via cleavage of the linker region or post-translational modification to release the active N-terminal fragment in response to a variety of pathogenic or intrinsic danger signals. GSDM-mediated pyroptosis is involved in the pathogenesis of an array of infectious and inflammatory diseases and cancers. This review discusses recent advances related to the physiological and pathological functions of GSDM-mediated pyroptosis, as well as therapeutic strategies targeting pyroptosis.
Collapse
Affiliation(s)
- Rui Min
- National Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Bai
- National Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ning-Rui Wang
- School of Laboratory Medicine, Nanchang Medical College, Nanchang, Jiangxi 330052, China
| | - Xing Liu
- National Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China; Shanghai Academy of Natural Sciences (SANS), Shanghai 200031, China.
| |
Collapse
|
3
|
Bai Y, Pan Y, Liu X. Mechanistic insights into gasdermin-mediated pyroptosis. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00837-0. [PMID: 40128620 DOI: 10.1038/s41580-025-00837-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2025] [Indexed: 03/26/2025]
Abstract
Pyroptosis, a novel mode of inflammatory cell death, is executed by membrane pore-forming gasdermin (GSDM) family members in response to extracellular or intracellular injury cues and is characterized by a ballooning cell morphology, plasma membrane rupture and the release of inflammatory mediators such as interleukin-1β (IL-1β), IL-18 and high mobility group protein B1 (HMGB1). It is a key effector mechanism for host immune defence and surveillance against invading pathogens and aberrant cancerous cells, and contributes to the onset and pathogenesis of inflammatory and autoimmune diseases. Manipulating the pore-forming activity of GSDMs and pyroptosis could lead to novel therapeutic strategies. In this Review, we discuss the current knowledge regarding how GSDM-mediated pyroptosis is initiated, executed and regulated, its roles in physiological and pathological processes, and the crosstalk between different modes of programmed cell death. We also highlight the development of drugs that target pyroptotic pathways for disease treatment.
Collapse
Affiliation(s)
- Yang Bai
- Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Youdong Pan
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xing Liu
- Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China.
- Shanghai Academy of Natural Sciences (SANS), Shanghai, China.
| |
Collapse
|
4
|
Matt SM, Nolan R, Manikandan S, Agarwal Y, Channer B, Oteju O, Daniali M, Canagarajah JA, LuPone T, Mompho K, Runner K, Nickoloff-Bybel E, Li B, Niu M, Schlachetzki JCM, Fox HS, Gaskill PJ. Dopamine-driven increase in IL-1β in myeloid cells is mediated by differential dopamine receptor expression and exacerbated by HIV. J Neuroinflammation 2025; 22:91. [PMID: 40122818 PMCID: PMC11931822 DOI: 10.1186/s12974-025-03403-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 03/01/2025] [Indexed: 03/25/2025] Open
Abstract
The catecholamine neurotransmitter dopamine is classically known for regulation of central nervous system (CNS) functions such as reward, movement, and cognition. Increasing evidence also indicates that dopamine regulates critical functions in peripheral organs and is an important immunoregulatory factor. We have previously shown that dopamine increases NF-κB activity, inflammasome activation, and the production of inflammatory cytokines such as IL-1β in human macrophages. As myeloid lineage cells are central to the initiation and resolution of acute inflammatory responses, dopamine-mediated dysregulation of these functions could both impair the innate immune response and exacerbate chronic inflammation. However, the exact pathways by which dopamine drives myeloid inflammation are not well defined, and studies in both rodent and human systems indicate that dopamine can impact the production of inflammatory mediators through both D1-like dopamine receptors (DRD1, DRD5) and D2-like dopamine receptors (DRD2, DRD3, and DRD4). Therefore, we hypothesized that dopamine-mediated production of IL-1β in myeloid cells is regulated by the ratio of different dopamine receptors that are activated. Our data in primary human monocyte-derived macrophages (hMDM) indicate that DRD1 expression is necessary for dopamine-mediated increases in IL-1β, and that changes in the expression of DRD2 and other dopamine receptors can alter the magnitude of the dopamine-mediated increase in IL-1β. Mature hMDM have a high D1-like to D2-like receptor ratio, which is different relative to monocytes and peripheral blood mononuclear cells (PBMCs). We further confirm in human microglia cell lines that a high ratio of D1-like to D2-like receptors promotes dopamine-induced increases in IL-1β gene and protein expression using pharmacological inhibition or overexpression of dopamine receptors. RNA-sequencing of dopamine-treated microglia shows that genes encoding functions in IL-1β signaling pathways, microglia activation, and neurotransmission increased with dopamine treatment. Finally, using HIV as an example of a chronic inflammatory disease that is substantively worsened by comorbid substance use disorders (SUDs) that impact dopaminergic signaling, we show increased effects of dopamine on inflammasome activation and IL-1β in the presence of HIV in both human macrophages and microglia. These data suggest that use of addictive substances and dopamine-modulating therapeutics could dysregulate the innate inflammatory response and exacerbate chronic neuroimmunological conditions like HIV. Thus, a detailed understanding of dopamine-mediated changes in inflammation, in particular pathways regulating IL-1β, will be critical to effectively tailor medication regimens.
Collapse
Affiliation(s)
- Stephanie M Matt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Rachel Nolan
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
- Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Samyuktha Manikandan
- Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Yash Agarwal
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
- Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Breana Channer
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
- Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Oluwatofunmi Oteju
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Marzieh Daniali
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Joanna A Canagarajah
- Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Teresa LuPone
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Krisna Mompho
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Kaitlyn Runner
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Emily Nickoloff-Bybel
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Benjamin Li
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Meng Niu
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Howard S Fox
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Peter J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA.
| |
Collapse
|
5
|
Li Y, Guo B. GSDMD-mediated pyroptosis: molecular mechanisms, diseases and therapeutic targets. MOLECULAR BIOMEDICINE 2025; 6:11. [PMID: 39994107 PMCID: PMC11850691 DOI: 10.1186/s43556-025-00249-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 02/26/2025] Open
Abstract
Pyroptosis is a regulated form of inflammatory cell death in which Gasdermin D (GSDMD) plays a central role as the key effector molecule. GSDMD-mediated pyroptosis is characterized by complex biological features and considerable heterogeneity in its expression, mechanisms, and functional outcomes across various tissues, cell types, and pathological microenvironments. This heterogeneity is particularly pronounced in inflammation-related diseases and tumors. In the context of inflammatory diseases, GSDMD expression is typically upregulated, and its activation in macrophages, neutrophils, T cells, epithelial cells, and mitochondria triggers both pyroptotic and non-pyroptotic pathways, leading to the release of pro-inflammatory cytokines and exacerbation of tissue damage. However, under certain conditions, GSDMD-mediated pyroptosis may also serve a protective immune function. The expression of GSDMD in tumors is regulated in a more complex manner, where it can either promote immune evasion or, in some instances, induce tumor cell death. As our understanding of GSDMD's role continues to progress, there have been advancements in the development of inhibitors targeting GSDMD-mediated pyroptosis; however, these therapeutic interventions remain in the preclinical phase. This review systematically examines the cellular and molecular complexities of GSDMD-mediated pyroptosis, with a particular emphasis on its roles in inflammation-related diseases and cancer. Furthermore, it underscores the substantial therapeutic potential of GSDMD as a target for precision medicine, highlighting its promising clinical applications.
Collapse
Affiliation(s)
- Yujuan Li
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, China.
| | - Bin Guo
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| |
Collapse
|
6
|
Huang Y, Zhou Y, He Z, Yang J, Gu J, Cui B, Li S, Deng H, Zhao W, Yang X, Sun F, He C, Pan W. Cellular Senescence Contributes to Colonic Barrier Integrity Impairment Induced by Toxoplasma gondii Infection. Inflammation 2025:10.1007/s10753-024-02213-0. [PMID: 39827329 DOI: 10.1007/s10753-024-02213-0] [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: 09/16/2024] [Revised: 11/30/2024] [Accepted: 12/06/2024] [Indexed: 01/22/2025]
Abstract
Toxoplasma gondii (T. gondii) induces gut barrier integrity impairment, which is crucial to the establishment of long-term infection in hosts. Cellular senescence is an imperative event that drives disease progression. Several studies have indicated that T. gondii induces oxidative stress and cell cycle blockade in the tissues of hosts, suggesting cellular senescence induced by the parasite. Here, we explored whether cell senescence is involved in T. gondii-mediated colonic barrier integrity damage in mice. C57BL/6J mice were infected with 10 cysts of T. gondii. Senolytic therapy (dasatinib and quercetin, DQ, a combination therapy for reducing senescent cells) was given by oral gavage 4 weeks post-infection. Alcian blue staining, immunofluorescence, western blot, quantitative PCR (qPCR), and enzyme-linked immunosorbent assay (ELISA) were employed to evaluate the thickness of the colonic mucus layer, the expression profiles of genes and proteins related to tight junction function and cellular senescence in the colonic tissues, and the levels of serum lipopolysaccharides (LPS), respectively. T. gondii-infected mice exhibited deteriorated secreted mucus, shortened length, decreased expression of zonula occludens-1 (ZO-1) and occludin in the colon, accompanied by elevated levels of serum LPS. Moreover, the infection upregulated cell senescence-related markers (p16INK4A, p21CIP1) while inhibiting Lamin B1 expression. In addition, the expression levels of senescence-associated secretory phenotypes (SASPs), including IL-1β, TNF-α, IL-6, MMP9 and CXCL10, were upregulated post-infection. Notably, reducing cell senescence with DQ administration, significantly ameliorated the colonic pathological alterations induced by T. gondii infection. This study uncovers for the first time that cellular senescence contributes to the colonic barrier integrity damage induced by chronic T. gondii infection. Importantly, we provide evidence that senolytic therapy exerts a therapeutic effect on the intestinal pathological lesions.
Collapse
Affiliation(s)
- Yingting Huang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yumeng Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhicheng He
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiayi Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jianqi Gu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Bingqian Cui
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Siyu Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Heng Deng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wendi Zhao
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fenfen Sun
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Cheng He
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
7
|
Yang Z, Chen J, Zhang C, Peng H. Pathological mechanisms of glial cell activation and neurodegenerative and neuropsychiatric disorders caused by Toxoplasma gondii infection. Front Microbiol 2024; 15:1512233. [PMID: 39723133 PMCID: PMC11668811 DOI: 10.3389/fmicb.2024.1512233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Toxoplasma gondii is an intracellular opportunistic parasite that exists in a latent form within the human central nervous system (CNS), even in immune-competent hosts. During acute infection, T. gondii traverses the blood-brain barrier (BBB). In the subsequent chronic infection phase, the infiltration of immune cells into the brain, driven by T. gondii infection and the formation of parasitic cysts, leads to persistent activation and proliferation of astrocytes and microglia. This process results in neuronal damages that are fatal in some cases. Through inducing systemic immune responses, T. gondii infection can dramatically alter the behavior of rodents and increase the risk of various neuropsychiatric disorders in humans. In this review, we explore some recent research progress on the major events involved in BBB disruption, glial cell activation and neuronal damage following T. gondii infection in hosts. It further discusses potential pathological mechanisms and the feasible treatment approaches for the neurodegenerative and neuropsychiatric disorders caused by T. gondii infection to extend our understanding for pathogenesis and preventive control of toxoplasmosis in humans.
Collapse
Affiliation(s)
| | | | | | - Hongjuan Peng
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Diseases Research, School of Public Health, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
8
|
Rouvray S, Drummond RA. The role of lipids in regulating macrophage antifungal immunity. mBio 2024; 15:e0305723. [PMID: 39207168 PMCID: PMC11481918 DOI: 10.1128/mbio.03057-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Macrophages are critical components of the antifungal immune response. Disturbance in the number or function of these innate immune cells can significantly increase susceptibility to invasive fungal infections. Pathogenic fungi cause billions of infections every year and have an unmet clinical need, with many infections associated with unacceptably high mortality rates that primarily affect vulnerable patients with underlying immune defects. Lipid metabolism has been increasingly appreciated to significantly influence macrophage function, particularly of macrophages residing in lipid-rich organs, such as the brain, or macrophages specialized at clearing dead cells including alveolar macrophages in the lungs. In this review, we provide an overview of macrophage lipid metabolism, and discuss how lipid recycling and dysregulation affect key macrophage functions relevant for antifungal immunity including phagocytosis, functional polarization, and inflammasome activation. We focus on the fungal pathogen Cryptococcus neoformans, as this is the most common cause of death from fungal infection in humans and because several lines of evidence have already linked lipid metabolism in the regulation of C. neoformans and macrophage interactions.
Collapse
Affiliation(s)
- Sophie Rouvray
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Rebecca A. Drummond
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
9
|
Babcock IW, Sibley LA, Labuzan SA, Cowan MN, Sethi I, Alemu S, Kelly AG, Kovacs MA, Lukens JR, Harris TH. Caspase-1 in Cx3cr1-expressing cells drives an IL-18-dependent T cell response that promotes parasite control during acute Toxoplasma gondii infection. PLoS Pathog 2024; 20:e1012006. [PMID: 39446964 PMCID: PMC11537422 DOI: 10.1371/journal.ppat.1012006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 11/05/2024] [Accepted: 10/13/2024] [Indexed: 10/26/2024] Open
Abstract
Inflammasome activation is a robust innate immune mechanism that promotes inflammatory responses through the release of alarmins and leaderless cytokines, including IL-1α, IL-1β, and IL-18. Various stimuli, including infectious agents and cellular stress, cause inflammasomes to assemble and activate caspase-1. Then, caspase-1 cleaves targets that lead to pore formation and leaderless cytokine activation and release. Toxoplasma gondii has been shown to promote inflammasome formation, but the cell types utilizing caspase-1 and the downstream effects on immunological outcomes during acute in vivo infection have not been explored. Here, using knockout mice, we examine the role of caspase-1 responses during acute T. gondii infection globally and in Cx3cr1-positive populations. We provide in vivo evidence that caspase-1 expression is critical for, IL-18 release, optimal interferon-γ (IFN-γ) production, monocyte and neutrophil recruitment to the site of infection, and parasite control. Specifically, we find that caspase-1 expression in Cx3cr1-positive cells drives IL-18 release, which potentiates CD4+ T cell IFN-γ production and parasite control. Notably, our Cx3cr1-Casp1 knockouts exhibited a selective T cell defect, mirroring the phenotype observed in Il18 knockouts. In further support of this finding, treatment of Cx3cr1-Casp1 knockout mice with recombinant IL-18 restored CD4+ T cell IFN-γ responses and parasite control. Additionally, we show that neutrophil recruitment is dependent on IL-1 receptor accessory protein (IL-1RAP) signaling but is dispensable for parasite control. Overall, these experiments highlight the multifaceted role of caspase-1 in multiple cell populations contributing to specific pathways that collectively contribute to caspase-1 dependent immunity to T. gondii.
Collapse
Affiliation(s)
- Isaac W. Babcock
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Lydia A. Sibley
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Sydney A. Labuzan
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Maureen N. Cowan
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Ish Sethi
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Seblework Alemu
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Abigail G. Kelly
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Michael A. Kovacs
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - John R. Lukens
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Tajie H. Harris
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
10
|
Wang Q, Cao Y, Ye S, Ding M, Ge W, Liang Y, Chen J. Trem2/Syk/PI3K axis contributes to the host protection against Toxoplasma gondii-induced adverse pregnancy outcomes via modulating decidual macrophages. PLoS Pathog 2024; 20:e1012543. [PMID: 39250507 PMCID: PMC11412541 DOI: 10.1371/journal.ppat.1012543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 09/19/2024] [Accepted: 08/28/2024] [Indexed: 09/11/2024] Open
Abstract
Decidual macrophages residing at the maternal-fetal interface have been recognized as pivotal factors for maintaining normal pregnancy; however, they are also key target cells of Toxoplasma gondii (T. gondii) in the pathology of T. gondii-induced adverse pregnancy. Trem2, as a functional receptor on macrophage surface, recognizes and binds various kinds of pathogens. The role and underlying mechanism of Trem2 in T. gondii infection remain elusive. In the present study, we found that T. gondii infection downregulated Trem2 expression and that Trem2-/- mice exhibited more severe adverse pregnancy outcomes than wildtype mice. We also demonstrated that T. gondii infection resulted in increased decidual macrophages, which were significantly reduced in the Trem2-/- pregnant mouse model as compared to wildtype control animals. We further described the inhibited proliferation, migration, and invasion functions of trophoblast cell by T. gondii antigens through macrophages as an "intermediate bridge", while this inhibition can be rescued by Trem2 agonist HSP60. Concurrently, Trem2 deficiency in bone marrow-derived macrophages (BMDMs) heightened the inhibitory effect of TgAg on the migration and invasion of trophoblast cells, accompanied by higher pro-inflammatory factors (IL-1β, IL-6 and TNF-α) but a lower chemokine (CXCL1) in T. gondii antigens-treated BMDMs. Furthermore, compelling evidence from animal models and in vitro cell experiments suggests that T. gondii inhibits the Trem2-Syk-PI3K signaling pathway, leading to impaired function of decidual macrophages. Therefore, our findings highlight Trem2 signaling as an essential pathway by which decidual macrophages respond to T. gondii infection, suggesting Trem2 as a crucial sensor of decidual macrophages and potential therapeutic target in the pathology of T. gondii-induced adverse pregnancy.
Collapse
Affiliation(s)
- Qing Wang
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, People’s Republic of China
| | - Yining Cao
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, People’s Republic of China
| | - Songyi Ye
- Department of Pediatric Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, People’s Republic of China
| | - Maoyuan Ding
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, People’s Republic of China
| | - Wenliang Ge
- Department of Pediatric Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, People’s Republic of China
| | - Yuejin Liang
- Department of Microbiology & Immunology, The University of Texas Medical Branch Galveston, Texas, United States of America
| | - Jinling Chen
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, People’s Republic of China
| |
Collapse
|
11
|
Wang H, Lan Y, Luo L, Xiao Y, Meng X, Zeng Y, Wu J. The Scutellaria-Coptis herb couple and its active small-molecule ingredient wogonoside alleviate cytokine storm by regulating the CD39/NLRP3/GSDMD signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 329:118155. [PMID: 38593962 DOI: 10.1016/j.jep.2024.118155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 03/28/2024] [Accepted: 04/04/2024] [Indexed: 04/11/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE A drug pair is a fundamental aspect of traditional Chinese medicine prescriptions. Scutellaria baicalensis Georgi and Coptis chinensis Franch, commonly used as an herb couple (SBCC), are representative heat-clearing and dampness-drying drugs. They possess functions such as clearing heat, drying dampness, purging fire, and detoxifying. These herbs are used in both traditional and modern medicine for treating inflammation. AIM OF THE STUDY This study investigated the effects of SBCC on cytokine storm syndrome (CSS) and explored its potential regulatory mechanism. MATERIALS AND METHODS We assessed the impact of SBCC in a sepsis-induced acute lung injury mouse model by administering an intraperitoneal injection of LPS (15 mg/kg). The cytokine levels in the serum and lungs, the wet-to-dry ratio of the lungs, and lung histopathological changes were evaluated. The macrophages in the lung tissue were examined through transmission electron microscopy. Western blot was used to measure the levels of the CD39/NLRP3/GSDMD pathway-related proteins. Immunofluorescence imaging was used to assess the activation of pro-caspase-1 and ASC and their interaction. AMP-Glo™ assay was used to screen for active ingredients in SBCC targeting CD39. One of the ingredients was selected, and its effect on cell viability was assessed. We induced inflammation in macrophages using LPS + ATP and detected the levels of proinflammatory factors. The images of cell membrane large pores were captured using scanning electron microscopy, the interaction between NLRP3 and ASC was detected using immunofluorescence imaging, and the levels of CD39/NLRP3/GSDMD pathway-related proteins were assessed using Western blot. RESULTS SBCC administration effectively mitigated LPS-induced cytokine storm, pulmonary edema and lung injury. Furthermore, it repressed the programmed death of lung tissue macrophages by inhibiting the NLRP3/GSDMD pyroptosis pathway and regulating the CD39 purinergic pathway. Based on the results of the AMP-Glo™ assay, we selected wogonoside for further valuation. Wogonoside alleviated LPS + ATP-induced inflammatory damage by regulating the inhibiting the NLRP3/GSDMD pyroptosis pathway and regulating the CD39 purinergic pathway. However, its effect on NLRP3 is not mediated though CD39. CONCLUSION SBCC and its active small-molecule ingredient, wogonoside, improved CSS by regulating the NLRP3/GSDMD pyroptosis pathway and its upstream CD39 purinergic pathway. It is essential to note that the regulatory effect of wogonoside on NLRP3 is not mediated by CD39.
Collapse
Affiliation(s)
- Huan Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Yuejia Lan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Liuling Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Yang Xiao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China; Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Yong Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China.
| | - Jiasi Wu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China.
| |
Collapse
|
12
|
Zhang C, Ma J, Zhang X, Zhou D, Cao Z, Qiao L, Chen G, Yang L, Ding BS. Processing of angiocrine alarmin IL-1α in endothelial cells promotes lung and liver fibrosis. Int Immunopharmacol 2024; 134:112176. [PMID: 38723369 DOI: 10.1016/j.intimp.2024.112176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/21/2024] [Accepted: 04/27/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND Fibrosis results from excessive scar formation after tissue injury. Injured cells release alarmins such as interleukin 1 (IL-1) α and β as primary mediators initiating tissue repair. However, how alarmins from different cell types differentially regulate fibrosis remains to be explored. METHODS Here, we used tissue specific knockout strategy to illustrate a unique contribution of endothelial cell-derived IL-1α to lung and liver fibrosis. The two fibrotic animal model triggered by bleomycin and CCl4 were used to study the effects of endothelial paracrine/angiocrine IL-1α in fibrotic progression. Human umbilical vein endothelial cells (HUVEC) were performed to explore the production of angiocrine IL-1α at both transcriptional and post-transcriptional levels in vitro. RESULTS We found that endothelial paracrine/angiocrine IL-1α primarily promotes lung and liver fibrosis during the early phase of organ repair. By contrast, myeloid cell-specific ablation of IL-1α in mice resulted in little influence on fibrosis, suggesting the specific pro-fibrotic role of IL-1α from endothelial cell but not macrophage. In vitro study revealed a coordinated regulation of IL-1α production in human primary endothelial cells at both transcriptional and post-transcriptional levels. Specifically, the transcription of IL-1α is regulated by RIPK1, and after caspase-8 (CASP8) cleaves the precursor form of IL-1α, its secretion is triggered by ion channel Pannexin 1 upon CASP8 cleavage. CONCLUSIONS Endothelial cell-produced IL-1α plays a unique role in promoting organ fibrosis. Furthermore, the release of this angiocrine alarmin relies on a unique molecular mechanism involving RIPK1, CASP8, and ion channel Pannexin 1.
Collapse
Affiliation(s)
- Chunxue Zhang
- Key Laboratory of Birth Defects of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, College of Life Sciences, Sichuan University, Chengdu 610041, China
| | - Jie Ma
- Key Laboratory of Birth Defects of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, College of Life Sciences, Sichuan University, Chengdu 610041, China
| | - Xu Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin 150081, China
| | - Dengcheng Zhou
- Key Laboratory of Birth Defects of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, College of Life Sciences, Sichuan University, Chengdu 610041, China
| | - Zhongwei Cao
- Key Laboratory of Birth Defects of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, College of Life Sciences, Sichuan University, Chengdu 610041, China
| | - Lina Qiao
- Key Laboratory of Birth Defects of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, College of Life Sciences, Sichuan University, Chengdu 610041, China.
| | - Guo Chen
- Department of Anesthesiology, The Research Units of West China(2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, China.
| | - Liming Yang
- Department of Pathophysiology, Harbin Medical University, Harbin 150081, China.
| | - Bi-Sen Ding
- Key Laboratory of Birth Defects of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, College of Life Sciences, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
13
|
Porte R, Belloy M, Audibert A, Bassot E, Aïda A, Alis M, Miranda-Capet R, Jourdes A, van Gisbergen KPJM, Masson F, Blanchard N. Protective function and differentiation cues of brain-resident CD8+ T cells during surveillance of latent Toxoplasma gondii infection. Proc Natl Acad Sci U S A 2024; 121:e2403054121. [PMID: 38838017 PMCID: PMC11181119 DOI: 10.1073/pnas.2403054121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/01/2024] [Indexed: 06/07/2024] Open
Abstract
Chronic Toxoplasma gondii infection induces brain-resident CD8+ T cells (bTr), but the protective functions and differentiation cues of these cells remain undefined. Here, we used a mouse model of latent infection by T. gondii leading to effective CD8+ T cell-mediated parasite control. Thanks to antibody depletion approaches, we found that peripheral circulating CD8+ T cells are dispensable for brain parasite control during chronic stage, indicating that CD8+ bTr are able to prevent brain parasite reactivation. We observed that the retention markers CD69, CD49a, and CD103 are sequentially acquired by brain parasite-specific CD8+ T cells throughout infection and that a majority of CD69/CD49a/CD103 triple-positive (TP) CD8+ T cells also express Hobit, a transcription factor associated with tissue residency. This TP subset develops in a CD4+ T cell-dependent manner and is associated with effective parasite control during chronic stage. Conditional invalidation of Transporter associated with Antigen Processing (TAP)-mediated major histocompatibility complex (MHC) class I presentation showed that presentation of parasite antigens by glutamatergic neurons and microglia regulates the differentiation of CD8+ bTr into TP cells. Single-cell transcriptomic analyses revealed that resistance to encephalitis is associated with the expansion of stem-like subsets of CD8+ bTr. In summary, parasite-specific brain-resident CD8+ T cells are a functionally heterogeneous compartment which autonomously ensure parasite control during T. gondii latent infection and which differentiation is shaped by neuronal and microglial MHC I presentation. A more detailed understanding of local T cell-mediated immune surveillance of this common parasite is needed for harnessing brain-resident CD8+ T cells in order to enhance control of chronic brain infections.
Collapse
Affiliation(s)
- Rémi Porte
- Toulouse Institute for Infectious and Inflammatory Diseases, Infinity, Inserm, CNRS, University of Toulouse, Toulouse31300, France
| | - Marcy Belloy
- Toulouse Institute for Infectious and Inflammatory Diseases, Infinity, Inserm, CNRS, University of Toulouse, Toulouse31300, France
| | - Alexis Audibert
- Toulouse Institute for Infectious and Inflammatory Diseases, Infinity, Inserm, CNRS, University of Toulouse, Toulouse31300, France
| | - Emilie Bassot
- Toulouse Institute for Infectious and Inflammatory Diseases, Infinity, Inserm, CNRS, University of Toulouse, Toulouse31300, France
| | - Amel Aïda
- Toulouse Institute for Infectious and Inflammatory Diseases, Infinity, Inserm, CNRS, University of Toulouse, Toulouse31300, France
| | - Marine Alis
- Toulouse Institute for Infectious and Inflammatory Diseases, Infinity, Inserm, CNRS, University of Toulouse, Toulouse31300, France
| | - Romain Miranda-Capet
- Toulouse Institute for Infectious and Inflammatory Diseases, Infinity, Inserm, CNRS, University of Toulouse, Toulouse31300, France
| | - Aurélie Jourdes
- Toulouse Institute for Infectious and Inflammatory Diseases, Infinity, Inserm, CNRS, University of Toulouse, Toulouse31300, France
| | | | - Frédérick Masson
- Toulouse Institute for Infectious and Inflammatory Diseases, Infinity, Inserm, CNRS, University of Toulouse, Toulouse31300, France
| | - Nicolas Blanchard
- Toulouse Institute for Infectious and Inflammatory Diseases, Infinity, Inserm, CNRS, University of Toulouse, Toulouse31300, France
| |
Collapse
|
14
|
Matt SM, Nolan R, Manikandan S, Agarwal Y, Channer B, Oteju O, Daniali M, Canagarajah JA, LuPone T, Mompho K, Runner K, Nickoloff-Bybel E, Li B, Niu M, Schlachetzki JCM, Fox HS, Gaskill PJ. Dopamine-driven Increase in IL-1β in Myeloid Cells is Mediated by Differential Dopamine Receptor Expression and Exacerbated by HIV. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.09.598137. [PMID: 38915663 PMCID: PMC11195146 DOI: 10.1101/2024.06.09.598137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
The catecholamine neurotransmitter dopamine is classically known for regulation of central nervous system (CNS) functions such as reward, movement, and cognition. Increasing evidence also indicates that dopamine regulates critical functions in peripheral organs and is an important immunoregulatory factor. We have previously shown that dopamine increases NF-κB activity, inflammasome activation, and the production of inflammatory cytokines such as IL-1β in human macrophages. As myeloid lineage cells are central to the initiation and resolution of acute inflammatory responses, dopamine-mediated dysregulation of these functions could both impair the innate immune response and exacerbate chronic inflammation. However, the exact pathways by which dopamine drives myeloid inflammation are not well defined, and studies in both rodent and human systems indicate that dopamine can impact the production of inflammatory mediators through both D1-like dopamine receptors (DRD1, DRD5) and D2-like dopamine receptors (DRD2, DRD3, and DRD4). Therefore, we hypothesized that dopamine-mediated production of IL-1β in myeloid cells is regulated by the ratio of different dopamine receptors that are activated. Our data in primary human monocyte-derived macrophages (hMDM) indicate that DRD1 expression is necessary for dopamine-mediated increases in IL-1β, and that changes in the expression of DRD2 and other dopamine receptors can alter the magnitude of the dopamine-mediated increase in IL-1β. Mature hMDM have a high D1-like to D2-like receptor ratio, which is different relative to monocytes and peripheral blood mononuclear cells (PBMCs). We further confirm in human microglia cell lines that a high ratio of D1-like to D2-like receptors promotes dopamine-induced increases in IL-1β gene and protein expression using pharmacological inhibition or overexpression of dopamine receptors. RNA-sequencing of dopamine-treated microglia shows that genes encoding functions in IL-1β signaling pathways, microglia activation, and neurotransmission increased with dopamine treatment. Finally, using HIV as an example of a chronic inflammatory disease that is substantively worsened by comorbid substance use disorders (SUDs) that impact dopaminergic signaling, we show increased effects of dopamine on inflammasome activation and IL-1β in the presence of HIV in both human macrophages and microglia. These data suggest that use of addictive substances and dopamine-modulating therapeutics could dysregulate the innate inflammatory response and exacerbate chronic neuroimmunological conditions like HIV. Thus, a detailed understanding of dopamine-mediated changes in inflammation, in particular pathways regulating IL-1β, will be critical to effectively tailor medication regimens.
Collapse
|
15
|
Liu J, Wang X, Wang X, Wang J, Ma Y, Cao Y, Zhang W. Chicken gasdermins mediate pyroptosis after the cleavage by caspases. Int J Biol Macromol 2024; 270:132476. [PMID: 38777016 DOI: 10.1016/j.ijbiomac.2024.132476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/22/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024]
Abstract
Gasdermin (GSDM) proteins are executioners of pyroptosis in many species. Gasdermin proteins can be cleaved at their linker region between the amino domain (NT) and carboxyl domain (CT) by enzymes. The released GSDM-NTs bind cell membrane and form pores, thereby leading to the release of cellular components and lytic cell death. GSDM-mediated pyroptosis is considered to play important role in immune responses. However, little is known about the GSDM proteins and GSDM-mediated pyroptosis in birds. In the current study, genes encoding chicken gasdermin A (chGSDMA) and chGSDME were cloned. The cleavage of chGSDMA and chGSDME by chicken caspase-1 (chCASP1), chCASP3 and chCASP7 and the cleavage sites were determined. The chGSDMA-NT obtained form chCASP1-mediated cleavage and chGSDME-NT obtained from chCASP3/chCASP7-mediated cleavage could bind and damage cell membrane and lead to cell death of HEK293 cells. chGSDMA-NT also strongly localized to and formed puncta in nucleus. Besides, both chGSDMA-NT and chGSDME-NT showed growth inhibition and bactericidal activity to bacteria. In chickens challenged with Pasteurella multocida and Salmonella typhimurium, the expression of chGSDMA and chGSDME was upregulated and the activation of chCASP3 and the cleavage of chGSDME were observed. The work provides essential information for expanding our knowledge on pyroptosis in birds.
Collapse
Affiliation(s)
- Jing Liu
- College of Veterinary Medicine, Northeast Agricultural University, Changjiang Road No.600, Harbin 150030, China; Northeastern Science Inspection Station, China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Harbin 150030, China
| | - Xin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Changjiang Road No.600, Harbin 150030, China; Northeastern Science Inspection Station, China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Harbin 150030, China
| | - Xinyu Wang
- College of Veterinary Medicine, Northeast Agricultural University, Changjiang Road No.600, Harbin 150030, China; Northeastern Science Inspection Station, China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Harbin 150030, China
| | - Junwei Wang
- College of Veterinary Medicine, Northeast Agricultural University, Changjiang Road No.600, Harbin 150030, China; Northeastern Science Inspection Station, China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Harbin 150030, China
| | - Yingying Ma
- College of Veterinary Medicine, Northeast Agricultural University, Changjiang Road No.600, Harbin 150030, China; Northeastern Science Inspection Station, China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Harbin 150030, China
| | - Yongsheng Cao
- College of Veterinary Medicine, Northeast Agricultural University, Changjiang Road No.600, Harbin 150030, China; Northeastern Science Inspection Station, China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Harbin 150030, China.
| | - Wenlong Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Changjiang Road No.600, Harbin 150030, China; Northeastern Science Inspection Station, China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Harbin 150030, China.
| |
Collapse
|
16
|
Matini M, Amini R, Foroughi-Parvar F. Glia Maturation Factor Beta: A Novel Neuro-Impairment Prediction Factor in Toxoplasmosis. IRANIAN JOURNAL OF PUBLIC HEALTH 2024; 53:1200-1208. [PMID: 38912132 PMCID: PMC11188646 DOI: 10.18502/ijph.v53i5.15602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/11/2023] [Indexed: 06/25/2024]
Abstract
Background Toxoplasma gondii, a neurotropic protozoan, infects up one to third of the world population. The parasite can invade a wide variety of nucleated cells but preferably glial cells. Glia maturation factor β (GMFβ), a 17KD protein expressed at high levels in the central nervous system is predominantly related to neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Multiple sclerosis. We aimed to determine the expression level of GMFβ and its relation to other pro-inflammatory factors (IL33, SDF1, and CCL2) on T. gondii infected human neuroblastoma cell line. Methods The human neuroblastoma (SK_NMC C535) cell line was infected by 5×106 (1:1 ratio). The supernatant was collected after cell lysis and centrifugation. Total RNA was extracted using the Yekta Tajhiz RNA extraction kit. cDNA was synthesized based on RevertAid First Strand cDNA Synthesis Kit manufacturer's protocol (Parstous, cDNA synthesis kit, Iran). The specificity of each primer pair (GMFβ, IL33, SDF1, and CCL2) was provided by NCBI BLAST. Gene expression level was measured using Real-Time PCR. All experiments were conducted at the Hamadan University of Medical Sciences, western Iran in 2022. Results The GMFβ increased significantly up to 1.35-fold (P=0.007). The increase in GMFβ expression in neuroblastoma cells was consistent with the increase in pro-inflammatory factors (CCL2 (0.47), IL33 (0.152) and, SDF1 (1.33)). Conclusion GMFβ upregulation can be a novel indicator of the destruction of nerve cells.
Collapse
Affiliation(s)
- Mohammad Matini
- Department of Medical Parasitology and Mycology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Razieh Amini
- Department of Molecular Medicine and Human Genetics, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Faeze Foroughi-Parvar
- Department of Medical Parasitology and Mycology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
17
|
Pollock NM, Fernandes JP, Woodfield J, Moussa E, Hlavay B, Branton WG, Wuest M, Mohammadzadeh N, Schmitt L, Plemel JR, Julien O, Wuest F, Power C. Gasdermin D activation in oligodendrocytes and microglia drives inflammatory demyelination in progressive multiple sclerosis. Brain Behav Immun 2024; 115:374-393. [PMID: 37914099 DOI: 10.1016/j.bbi.2023.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 10/20/2023] [Accepted: 10/22/2023] [Indexed: 11/03/2023] Open
Abstract
Neuroinflammation coupled with demyelination and neuro-axonal damage in the central nervous system (CNS) contribute to disease advancement in progressive multiple sclerosis (P-MS). Inflammasome activation accompanied by proteolytic cleavage of gasdermin D (GSDMD) results in cellular hyperactivation and lytic death. Using multiple experimental platforms, we investigated the actions of GSDMD within the CNS and its contributions to P-MS. Brain tissues from persons with P-MS showed significantly increased expression of GSDMD, NINJ1, IL-1β, and -18 within chronic active demyelinating lesions compared to MS normal appearing white matter and nonMS (control) white matter. Conditioned media (CM) from stimulated GSDMD+/+ human macrophages caused significantly greater cytotoxicity of oligodendroglial and neuronal cells, compared to CM from GSDMD-/- macrophages. Oligodendrocytes and CNS macrophages displayed increased Gsdmd immunoreactivity in the central corpus callosum (CCC) of cuprizone (CPZ)-exposed Gsdmd+/+ mice, associated with greater demyelination and reduced oligodendrocyte precursor cell proliferation, compared to CPZ-exposed Gsdmd-/- animals. CPZ-exposed Gsdmd+/+ mice exhibited significantly increased G-ratios and reduced axonal densities in the CCC compared to CPZ-exposed Gsdmd-/- mice. Proteomic analyses revealed increased brain complement C1q proteins and hexokinases in CPZ-exposed Gsdmd-/- animals. [18F]FDG PET imaging showed increased glucose metabolism in the hippocampus and whole brain with intact neurobehavioral performance in Gsdmd-/- animals after CPZ exposure. GSDMD activation in CNS macrophages and oligodendrocytes contributes to inflammatory demyelination and neuroaxonal injury, offering mechanistic and potential therapeutic insights into P-MS pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Laura Schmitt
- Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton AB, Canada.
| | | | | | | | - Christopher Power
- Department of Medicine (Neurology), Canada; Department of Medical Microbiology & Immunology, Canada.
| |
Collapse
|
18
|
Orchanian SB, Still K, Harris TH, Lodoen MB. Deficiency in astrocyte CCL2 production reduces neuroimmune control of Toxoplasma gondii infection. PLoS Pathog 2024; 20:e1011710. [PMID: 38206985 PMCID: PMC10807779 DOI: 10.1371/journal.ppat.1011710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 01/24/2024] [Accepted: 09/25/2023] [Indexed: 01/13/2024] Open
Abstract
Toxoplasma gondii is an obligate intracellular parasite that infects one-third of the world's human population and establishes infection in the brain. Cerebral immune cell infiltration is critical for controlling the parasite, but little is known about the molecular cues guiding immune cells to the brain during infection. Activated astrocytes produce CCL2, a chemokine that mediates inflammatory monocyte recruitment to tissues by binding to the CCR2 receptor. We detected elevated CCL2 production in the brains of C57BL/6J mice by 15 days after T. gondii infection. Utilizing confocal microscopy and intracellular flow cytometry, we identified microglia and brain-infiltrating myeloid cells as the main producers of CCL2 during acute infection, and CCL2 was specifically produced in regions of parasite infection in the brain. In contrast, astrocytes became the dominant CCL2 producer during chronic T. gondii infection. To determine the role of astrocyte-derived CCL2 in mobilizing immune cells to the brain and controlling T. gondii infection, we generated GFAP-Cre x CCL2fl/fl mice, in which astrocytes are deficient in CCL2 production. We observed significantly decreased immune cell recruitment and increased parasite burden in the brain during chronic, but not acute, infection of mice deficient in astrocyte CCL2 production, without an effect on peripheral immune responses. To investigate potential mechanisms explaining the reduced control of T. gondii infection, we analyzed key antimicrobial and immune players in host defense against T. gondii and detected a reduction in iNOS+ myeloid cells, and T. gondii-specific CD4+ T cells in the knockout mice. These data uncover a critical role for astrocyte-derived CCL2 in immune cell recruitment and parasite control in the brain during chronic, but not acute, T. gondii infection.
Collapse
Affiliation(s)
- Stephanie B. Orchanian
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, United States of America
- Institute for Immunology, University of California, Irvine, Irvine, California, United States of America
| | - Katherine Still
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Tajie H. Harris
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Melissa B. Lodoen
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, United States of America
- Institute for Immunology, University of California, Irvine, Irvine, California, United States of America
| |
Collapse
|
19
|
Mohamed SH, Fu MS, Hain S, Alselami A, Vanhoffelen E, Li Y, Bojang E, Lukande R, Ballou ER, May RC, Ding C, Velde GV, Drummond RA. Microglia are not protective against cryptococcal meningitis. Nat Commun 2023; 14:7202. [PMID: 37938547 PMCID: PMC10632471 DOI: 10.1038/s41467-023-43061-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/30/2023] [Indexed: 11/09/2023] Open
Abstract
Microglia provide protection against a range of brain infections including bacteria, viruses and parasites, but how these glial cells respond to fungal brain infections is poorly understood. We investigated the role of microglia in the context of cryptococcal meningitis, the most common cause of fungal meningitis in humans. Using a series of transgenic- and chemical-based microglia depletion methods we found that, contrary to their protective role during other infections, loss of microglia did not affect control of Cryptococcus neoformans brain infection which was replicated with several fungal strains. At early time points post-infection, we found that microglia depletion lowered fungal brain burdens, which was related to intracellular residence of C. neoformans within microglia. Further examination of extracellular and intracellular fungal populations revealed that C. neoformans residing in microglia were protected from copper starvation, whereas extracellular yeast upregulated copper transporter CTR4. However, the degree of copper starvation did not equate to fungal survival or abundance of metals within different intracellular niches. Taken together, these data show how tissue-resident myeloid cells may influence fungal phenotype in the brain but do not provide protection against this infection, and instead may act as an early infection reservoir.
Collapse
Affiliation(s)
- Sally H Mohamed
- Institute of Immunology & Immunotherapy, University of Birmingham, Birmingham, UK
| | - Man Shun Fu
- Institute of Immunology & Immunotherapy, University of Birmingham, Birmingham, UK
| | - Sofia Hain
- Institute of Immunology & Immunotherapy, University of Birmingham, Birmingham, UK
| | - Alanoud Alselami
- Institute of Immunology & Immunotherapy, University of Birmingham, Birmingham, UK
| | - Eliane Vanhoffelen
- Department of Imaging and Pathology, Biomedical MRI/MoSAIC, KU Leuven, Leuven, Belgium
| | - Yanjian Li
- College of Life and Health Sciences, Northeastern University, Shenyang, 110015, Liaoning, China
| | - Ebrima Bojang
- Institute of Immunology & Immunotherapy, University of Birmingham, Birmingham, UK
| | - Robert Lukande
- Department of Pathology, College of Health Sciences, Makerere University, Kampala, Uganda
| | | | - Robin C May
- Institute of Microbiology & Infection and School of Biosciences, University of Birmingham, Birmingham, UK
| | - Chen Ding
- College of Life and Health Sciences, Northeastern University, Shenyang, 110015, Liaoning, China
| | - Greetje Vande Velde
- Department of Imaging and Pathology, Biomedical MRI/MoSAIC, KU Leuven, Leuven, Belgium
| | - Rebecca A Drummond
- Institute of Immunology & Immunotherapy, University of Birmingham, Birmingham, UK.
- Institute of Microbiology & Infection and School of Biosciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
20
|
Xiao Z, Li Y, Xiong L, Liao J, Gao Y, Luo Y, Wang Y, Chen T, Yu D, Wang T, Zhang C, Chen Z. Recent Advances in Anti-Atherosclerosis and Potential Therapeutic Targets for Nanomaterial-Derived Drug Formulations. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302918. [PMID: 37698552 PMCID: PMC10582432 DOI: 10.1002/advs.202302918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/12/2023] [Indexed: 09/13/2023]
Abstract
Atherosclerosis, the leading cause of death worldwide, is responsible for ≈17.6 million deaths globally each year. Most therapeutic drugs for atherosclerosis have low delivery efficiencies and significant side effects, and this has hampered the development of effective treatment strategies. Diversified nanomaterials can improve drug properties and are considered to be key for the development of improved treatment strategies for atherosclerosis. The pathological mechanisms underlying atherosclerosis is summarized, rationally designed nanoparticle-mediated therapeutic strategies, and potential future therapeutic targets for nanodelivery. The content of this study reveals the potential and challenges of nanoparticle use for the treatment of atherosclerosis and highlights new effective design ideas.
Collapse
Affiliation(s)
- Zhicheng Xiao
- Shanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Yi Li
- Shanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Liyan Xiong
- Shanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Jun Liao
- Shanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Yijun Gao
- Shanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Yunchun Luo
- Shanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Yun Wang
- Shanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Ting Chen
- Shanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Dahai Yu
- Weihai Medical Area970 Hospital of Joint Logistic Support Force of PLAWeihai264200China
| | - Tingfang Wang
- Shanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Chuan Zhang
- Shanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Zhe‐Sheng Chen
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityNew York11439USA
| |
Collapse
|
21
|
Broz P. Unconventional protein secretion by gasdermin pores. Semin Immunol 2023; 69:101811. [PMID: 37473560 DOI: 10.1016/j.smim.2023.101811] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/14/2023] [Accepted: 07/14/2023] [Indexed: 07/22/2023]
Abstract
Unconventional protein secretion (UPS) allows the release of specific leaderless proteins independently of the classical endoplasmic reticulum (ER)-Golgi secretory pathway. While it remains one of the least understood mechanisms in cell biology, UPS plays an essential role in immunity as it controls the release of the IL-1 family of cytokines, which coordinate host defense and inflammatory responses. The unconventional secretion of IL-1β and IL-18, the two most prominent members of the IL-1 family, is initiated by inflammasome complexes - cytosolic signaling platforms that are assembled in response to infectious or noxious stimuli. Inflammasomes activate inflammatory caspases that proteolytically mature IL-1β/- 18, but also induce pyroptosis, a lytic form of cell death. Pyroptosis is caused by gasdermin-D (GSDMD), a member of the gasdermin protein family, which is activated by caspase cleavage and forms large β-barrel plasma membrane pores. This pore-forming activity is shared with other family members that are activated during infection or upon treatment with chemotherapy drugs. While the induction of cell death was assumed to be the main function of gasdermin pores, accumulating evidence suggests that they have also non-lytic functions, such as in the release of cytokines and alarmins, or in regulating ion fluxes. This has raised the possibility that gasdermin pores are one of the main mediators of UPS. Here, I summarize and discuss new insights into gasdermin activation and pore formation, how gasdermin pores achieve selective cargo release, and how gasdermin pore formation and ninjurin-1-driven plasma membrane rupture are executed and regulated.
Collapse
Affiliation(s)
- Petr Broz
- Department of Immunobiology, University of Lausanne, Switzerland.
| |
Collapse
|
22
|
Chou ML, Babamale AO, Walker TL, Cognasse F, Blum D, Burnouf T. Blood-brain crosstalk: the roles of neutrophils, platelets, and neutrophil extracellular traps in neuropathologies. Trends Neurosci 2023; 46:764-779. [PMID: 37500363 DOI: 10.1016/j.tins.2023.06.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/17/2023] [Accepted: 06/29/2023] [Indexed: 07/29/2023]
Abstract
Systemic inflammation, neurovascular dysfunction, and coagulopathy often occur concurrently in neuropathologies. Neutrophils and platelets have crucial synergistic roles in thromboinflammation and are increasingly suspected as effector cells contributing to the pathogenesis of neuroinflammatory diseases. In this review, we summarize the roles of platelet-neutrophil interactions in triggering complex pathophysiological events affecting the brain that may lead to the disruption of brain barriers, infiltration of toxic factors into the parenchyma, and amplification of neuroinflammation through the formation of neutrophil extracellular traps (NETs). We highlight the clinical significance of thromboinflammation in neurological disorders and examine the contributions of damage-associated molecular patterns (DAMPs) derived from platelets and neutrophils. These DAMPs originate from both infectious and non-infectious risk factors and contribute to the activation of inflammasomes during brain disorders. Finally, we identify knowledge gaps in the molecular mechanisms underlying neurodegenerative disease pathogenesis and emphasize the potential of interventions targeting platelets and neutrophils to treat neuroinflammatory diseases.
Collapse
Affiliation(s)
- Ming-Li Chou
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City 23561, Taiwan; INSERM UMRS 938, Centre de Recherche Saint-Antoine, Immune System and Neuroinflammation Laboratory, Hôpital Saint-Antoine, Paris 75012, France
| | - Abdulkareem Olarewaju Babamale
- Taiwan International Graduate Program in Molecular Medicine, Academia Sinica, Taipei 11266, Taiwan; Department of Zoology, Faculty of Life Sciences, University of Ilorin, Ilorin 240003, Nigeria
| | - Tara L Walker
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Fabrice Cognasse
- Etablissement Français du Sang Auvergne-Rhône-Alpes, 42023 Saint-Étienne, France; University Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 Sainbiose, 42023 Saint-Etienne, France
| | - David Blum
- University of Lille, INSERM, CHU Lille, UMR-S1172 LilNCog, Lille Neuroscience and Cognition, F-59000 Lille, France; Alzheimer & Tauopathies, LabEx DISTALZ, LiCEND, Lille F-59000, France; NeuroTMULille International Laboratory, University of Lille, F-59000 Lille, France
| | - Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City 23561, Taiwan; International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City 23561, Taiwan; NeuroTMULille International Laboratory, Taipei Medical University, Taipei 10031, Taiwan; Neuroscience Research Center, Taipei Medical University, Taipei 11031, Taiwan; Brain and Consciousness Research Centre, Taipei Medical University Shuang Ho Hospital, New Taipei City 23561, Taiwan.
| |
Collapse
|
23
|
Brito RMDM, da Silva MCM, Vieira-Santos F, de Almeida Lopes C, Souza JLN, Bastilho AL, de Barros Fernandes H, de Miranda AS, de Oliveira ACP, de Almeida Vitor RW, de Andrade-Neto VF, Bueno LL, Fujiwara RT, Magalhães LMD. Chronic infection by atypical Toxoplasma gondii strain induces disturbance in microglia population and altered behaviour in mice. Brain Behav Immun Health 2023; 30:100652. [PMID: 37396335 PMCID: PMC10308216 DOI: 10.1016/j.bbih.2023.100652] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 06/03/2023] [Accepted: 06/04/2023] [Indexed: 07/04/2023] Open
Abstract
Toxoplasma gondii chronic infection is characterized by the establishment of tissue cysts in the brain and increased levels of IFN-γ, which can lead to brain circuitry interference and consequently abnormal behaviour in mice. In this sense, the study presented here sought to investigate the impact of chronic infection by two T. gondii strains in the brain of infection-resistant mice, as a model for studying the involvement of chronic neuroinflammation with the development of behavioural alterations. For that, male BALB/c mice were divided into three groups: non-infected (Ni), infected with T. gondii ME49 clonal strain (ME49), and infected with TgCkBrRN2 atypical strain (CK2). Mice were monitored for 60 days to establish the chronic infection and then submitted to behavioural assessment. The enzyme-linked immunosorbent assay was used for measurement of specific IgG in the blood and levels of inflammatory cytokines and neurotrophic factors in the brain, and the cell's immunophenotype was determined by multiparametric flow cytometry. Mice infected with ME49 clonal strain displayed hyperlocomotor activity and memory deficit, although no signs of depressive- and/or anxiety-like behaviour were detected; on the other hand, chronic infection with CK2 atypical strain induced anxiety- and depressive-like behaviour. During chronic infection by CK2 atypical strain, mice displayed a higher number of T. gondii brain tissue cysts and inflammatory infiltrate, composed mainly of CD3+ T lymphocytes and Ly6Chi inflammatory monocytes, compared to mice infected with the ME49 clonal strain. Infected mice presented a marked decrease of microglia population compared to non-infected group. Chronic infection with CK2 strain produced elevated levels of IFN-γ and TNF-ɑ in the brain, decreased NGF levels in the prefrontal cortex and striatum, and altered levels of fractalkine (CX3CL1) in the prefrontal cortex and hippocampus. The persistent inflammation and the disturbance in the cerebral homeostasis may contribute to altered behaviour in mice, as the levels of IFN-γ were shown to be correlated with the behavioural parameters assessed here. Considering the high incidence and life-long persistence of T. gondii infection, this approach can be considered a suitable model for studying the impact of chronic infections in the brain and how it impacts in behavioural responses.
Collapse
Affiliation(s)
- Ramayana Morais de Medeiros Brito
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Laboratory of Malaria and Toxoplasmosis Biology, Department of Microbiology and Parasitology, Biosciences Centre, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Maria Carolina Machado da Silva
- Neuropharmacology Laboratory, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Flaviane Vieira-Santos
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Camila de Almeida Lopes
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jorge Lucas Nascimento Souza
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alexandre Lazoski Bastilho
- Laboratory of Malaria and Toxoplasmosis Biology, Department of Microbiology and Parasitology, Biosciences Centre, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Heliana de Barros Fernandes
- Laboratory of Neurobiology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Aline Silva de Miranda
- Laboratory of Neurobiology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Antônio Carlos Pinheiro de Oliveira
- Neuropharmacology Laboratory, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ricardo Wagner de Almeida Vitor
- Laboratory of Toxoplasmosis, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Valter Ferreira de Andrade-Neto
- Laboratory of Malaria and Toxoplasmosis Biology, Department of Microbiology and Parasitology, Biosciences Centre, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Lilian Lacerda Bueno
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ricardo Toshio Fujiwara
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luísa Mourão Dias Magalhães
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
24
|
Wei Y, You Y, Zhang J, Ban J, Min H, Li C, Chen J. Crystalline silica-induced macrophage pyroptosis interacting with mitophagy contributes to pulmonary fibrosis via modulating mitochondria homeostasis. JOURNAL OF HAZARDOUS MATERIALS 2023; 454:131562. [PMID: 37148789 DOI: 10.1016/j.jhazmat.2023.131562] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 04/05/2023] [Accepted: 05/01/2023] [Indexed: 05/08/2023]
Abstract
Environmental exposure to crystalline silica (CS) can lead to silicosis. Alveolar macrophages (AMs) play a crucial role in the pathogenesis of silicosis. Previously, we demonstrated that enhancing AMs mitophagy exerted protective effects on silicosis with a restrained inflammatory response. However, the exact molecular mechanisms are elusive. Pyroptosis and mitophagy are two different biological processes that determine cell fate. Exploring whether there were interactions or balances between these two processes in AMs would provide new insight into treating silicosis. Here we reported that crystalline silica induced pyroptosis in silicotic lungs and AMs with apparent mitochondria injury. Notably, we identified a reciprocal inhibitory effect between mitophagy and pyroptosis cascades in AMs. By enhancing or diminishing mitophagy, we demonstrated that PINK1-mediated mitophagy helped clear damaged mitochondria to negatively regulate CS-induced pyroptosis. While constraining pyroptosis cascades by NLRP3, Caspase1, and GSDMD inhibitors, respectively, displayed enhanced PINK1-dependent mitophagy with lessened CS-injured mitochondria. These observed effects were echoed in the mice with enhanced mitophagy. Therapeutically, we demonstrated abolishing GSDMD-dependent pyroptosis by disulfiram attenuated CS-induced silicosis. Collectively, our data demonstrated that macrophage pyroptosis interacting with mitophagy contributes to pulmonary fibrosis via modulating mitochondria homeostasis, which may provide potential therapeutic targets.
Collapse
Affiliation(s)
- Yungeng Wei
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Yichuan You
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Jiarui Zhang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, Liaoning, China; Experimental Teaching Center, School of Public health, China Medical University, Shenyang 110122, Liaoning, China
| | - Jiaqi Ban
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Hui Min
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang 110122, Liaoning, China
| | - Chao Li
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China.
| | - Jie Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China.
| |
Collapse
|
25
|
Drummond RA. What fungal CNS infections can teach us about neuroimmunology and CNS-specific immunity. Semin Immunol 2023; 67:101751. [PMID: 36989541 DOI: 10.1016/j.smim.2023.101751] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Indexed: 03/29/2023]
Abstract
Immunity to fungal infections of the central nervous system (CNS) is one of the most poorly understood subjects within the field of medical mycology. Yet, the majority of deaths from invasive fungal infections are caused by brain-tropic fungi. In recent years, there have been several significant discoveries in the regulation of neuroinflammation and the role of the immune system in tissue homeostasis within the CNS. In this review, I highlight five important advances in the neuroimmunology field over the last decade and discuss how we should capitalise on these discoveries to better understand the pathogenesis of fungal CNS infections. In addition, the latest insights into fungal invasion tactics, microglia-astrocyte crosstalk and regulation of antifungal adaptive immune responses are summarised in the context of our contemporary understanding of CNS-specific immunity.
Collapse
|
26
|
Wang Q, Zhong Y, Chen N, Chen J. From the immune system to mood disorders especially induced by Toxoplasma gondii: CD4+ T cell as a bridge. Front Cell Infect Microbiol 2023; 13:1078984. [PMID: 37077528 PMCID: PMC10106765 DOI: 10.3389/fcimb.2023.1078984] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 03/23/2023] [Indexed: 04/05/2023] Open
Abstract
Toxoplasma gondii (T. gondii), a ubiquitous and obligatory intracellular protozoa, not only alters peripheral immune status, but crosses the blood-brain barrier to trigger brain parenchymal injury and central neuroinflammation to establish latent cerebral infection in humans and other vertebrates. Recent findings underscore the strong correlation between alterations in the peripheral and central immune environment and mood disorders. Th17 and Th1 cells are important pro-inflammatory cells that can drive the pathology of mood disorders by promoting neuroinflammation. As opposed to Th17 and Th1, regulatory T cells have inhibitory inflammatory and neuroprotective functions that can ameliorate mood disorders. T. gondii induces neuroinflammation, which can be mediated by CD4+ T cells (such as Tregs, Th17, Th1, and Th2). Though the pathophysiology and treatment of mood disorder have been currently studied, emerging evidence points to unique role of CD4+ T cells in mood disorder, especially those caused by T. gondii infection. In this review, we explore some recent studies that extend our understanding of the relationship between mood disorders and T. gondii.
Collapse
|
27
|
Matsuno SY, Pandori WJ, Lodoen MB. Capers with caspases: Toxoplasma gondii tales of inflammation and survival. Curr Opin Microbiol 2023; 72:102264. [PMID: 36791673 DOI: 10.1016/j.mib.2023.102264] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/23/2022] [Accepted: 12/30/2022] [Indexed: 02/15/2023]
Abstract
Intracellular pathogens strike a delicate balance between maintaining their survival within infected cells, while also activating host defense mechanisms. Toxoplasma gondii is a protozoan parasite that initiates a variety of host signaling pathways as it invades host cells and establishes residence in a parasitophorous vacuole. Recent work has highlighted the interplay between T. gondii infection and innate immune pathways that lead to inflammation, several of which converge on caspases. This family of cysteine proteases function at the crossroads of inflammation and cell death and serve as a key target for parasite manipulation. This review focuses on the interaction of T. gondii with caspase-dependent inflammatory and cell death pathways and the role of parasite effector proteins in modulating these processes.
Collapse
Affiliation(s)
- Stephanie Y Matsuno
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92617 USA
| | - William J Pandori
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92617 USA
| | - Melissa B Lodoen
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92617 USA.
| |
Collapse
|
28
|
Clark JT, Weizman OE, Aldridge DL, Shallberg LA, Eberhard J, Lanzar Z, Wasche D, Huck JD, Zhou T, Ring AM, Hunter CA. IL-18BP mediates the balance between protective and pathological immune responses to Toxoplasma gondii. Cell Rep 2023; 42:112147. [PMID: 36827187 PMCID: PMC10131179 DOI: 10.1016/j.celrep.2023.112147] [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: 02/14/2022] [Revised: 12/02/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
Interleukin-18 (IL-18) promotes natural killer (NK) and T cell production of interferon (IFN)-γ, a key factor in resistance to Toxoplasma gondii, but previous work has shown a limited role for endogenous IL-18 in control of this parasite. Although infection with T. gondii results in release of IL-18, the production of IFN-γ induces high levels of the IL-18 binding protein (IL-18BP). Antagonism of IL-18BP with a "decoy-to-the-decoy" (D2D) IL-18 construct that does not signal but rather binds IL-18BP results in enhanced innate lymphoid cell (ILC) and T cell responses and improved parasite control. In addition, the use of IL-18 resistant to IL-18BP ("decoy-resistant" IL-18 [DR-18]) is more effective than exogenous IL-18 at promoting innate resistance to infection. DR-18 enhances CD4+ T cell production of IFN-γ but results in CD4+ T cell-mediated pathology. Thus, endogenous IL-18BP restrains aberrant immune pathology, and this study highlights strategies that can be used to tune this regulatory pathway for optimal anti-pathogen responses.
Collapse
Affiliation(s)
- Joseph T Clark
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Orr-El Weizman
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06519, USA
| | - Daniel L Aldridge
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Lindsey A Shallberg
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Julia Eberhard
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Zachary Lanzar
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Devon Wasche
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06519, USA
| | - John D Huck
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06519, USA
| | - Ting Zhou
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06519, USA
| | - Aaron M Ring
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06519, USA.
| | - Christopher A Hunter
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
29
|
Wei Y, Wang K, Zhang Y, Duan Y, Tian Y, Yin H, Fu X, Ma Z, Zhou J, Yu M, Ni Q, Tang W. Potent anti-inflammatory responses: Role of hydrogen in IL-1α dominated early phase systemic inflammation. Front Pharmacol 2023; 14:1138762. [PMID: 37007020 PMCID: PMC10063881 DOI: 10.3389/fphar.2023.1138762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/08/2023] [Indexed: 03/19/2023] Open
Abstract
Introduction: It has been proven that hydrogen has obvious anti-inflammatory effects in animal experiments and clinical practice. However, the early dynamic process of the inflammatory response caused by lipopolysaccharide (LPS) and the anti-inflammatory effect of hydrogen has not been definitively reported. Methods: Inflammation in male C57/BL6J mice or RAW264.7 cells was induced with LPS, for which hydrogen was immediately administered until samples were taken. Pathological changes in lung tissue were assessed using hematoxylin and eosin (HE) staining. Levels of inflammatory factors in serum were determined using liquid protein chip. The mRNA levels of chemotactic factors in lung tissues, leukocytes, and peritoneal macrophages were measured by qRT-PCR. The expression levels of IL-1α and HIF-1α were measured by immunocytochemistry. Results: Hydrogen alleviated LPS-induced inflammatory infiltration in the lung tissues of mice. Among the 23 inflammatory factors screened, LPS-induced upregulation of IL-1α etc. was significantly inhibited by hydrogen within 1 hour. The mRNA expression of MCP-1, MIP-1α, G-CSF, and RANTES was inhibited obviously by hydrogen at 0.5 and 1 h in mouse peritoneal macrophages. In addition, hydrogen significantly blocked LPS or H2O2-induced upregulation of HIF-1α, and IL-1α in 0.5 h in RAW264.7 cells. Discussion: The results suggested that hydrogen is potentially inhibitive against inflammation by inhibiting HIF-1α and IL-1α release at early occurrence. The target of the inhibitive LPS-induced-inflammatory action of hydrogen is chemokines in macrophages in the peritoneal cavity. This study provides direct experimental evidence for quickly controlling inflammation with the translational application of a hydrogen-assisted protocol.
Collapse
Affiliation(s)
- Youzhen Wei
- Research Institute of Heart Failure, Research Center for Translational Medicine & Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, China
- Hydrogen Medicine Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
- Research Center for Translational Medicine, Jinan People’s Hospital, Shandong First Medical University, Jinan, Shandong, China
| | - Kun Wang
- Office of Academic Research, Taishan Vocational College of Nursing, Taian, Shandong, China
| | - Yafang Zhang
- Department of Neonatology and NICU, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| | - Yi Duan
- Research Institute of Heart Failure, Research Center for Translational Medicine & Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yan Tian
- Research Institute of Heart Failure, Research Center for Translational Medicine & Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hongling Yin
- Research Institute of Heart Failure, Research Center for Translational Medicine & Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xuelian Fu
- Research Institute of Heart Failure, Research Center for Translational Medicine & Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zuan Ma
- Research Institute of Heart Failure, Research Center for Translational Medicine & Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianjun Zhou
- Research Institute of Heart Failure, Research Center for Translational Medicine & Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Min Yu
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medicine, Fudan University, Shanghai, China
| | - Qingbin Ni
- Hydrogen Medicine Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
- *Correspondence: Wenjie Tang, ; Qingbin Ni,
| | - Wenjie Tang
- Research Institute of Heart Failure, Research Center for Translational Medicine & Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, China
- Research Institute of Regenerative Medicine, East Hospital, Tongji University, Shanghai, China
- *Correspondence: Wenjie Tang, ; Qingbin Ni,
| |
Collapse
|
30
|
Kumar S, Budhathoki S, Oliveira CB, Kahle AD, Calhan OY, Lukens JR, Deppmann CD. Role of the caspase-8/RIPK3 axis in Alzheimer's disease pathogenesis and Aβ-induced NLRP3 inflammasome activation. JCI Insight 2023; 8:157433. [PMID: 36602874 PMCID: PMC9977425 DOI: 10.1172/jci.insight.157433] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 12/27/2022] [Indexed: 01/06/2023] Open
Abstract
The molecular mediators of cell death and inflammation in Alzheimer's disease (AD) have yet to be fully elucidated. Caspase-8 is a critical regulator of several cell death and inflammatory pathways; however, its role in AD pathogenesis has not yet been examined in detail. In the absence of caspase-8, mice are embryonic lethal due to excessive receptor interacting protein kinase 3-dependent (RIPK3-dependent) necroptosis. Compound RIPK3 and caspase-8 mutants rescue embryonic lethality, which we leveraged to examine the roles of these pathways in an amyloid β-mediated (Aβ-mediated) mouse model of AD. We found that combined deletion of caspase-8 and RIPK3, but not RIPK3 alone, led to diminished Aβ deposition and microgliosis in the mouse model of AD carrying human presenilin 1 and amyloid precursor protein with 5 familial AD mutations (5xFAD). Despite its well-known role in cell death, caspase-8 did not appear to affect cell loss in the 5xFAD model. In contrast, we found that caspase-8 was a critical regulator of Aβ-driven inflammasome gene expression and IL-1β release. Interestingly, loss of RIPK3 had only a modest effect on disease progression, suggesting that inhibition of necroptosis or RIPK3-mediated cytokine pathways is not critical during midstages of Aβ amyloidosis. These findings suggest that therapeutics targeting caspase-8 may represent a novel strategy to limit Aβ amyloidosis and neuroinflammation in AD.
Collapse
Affiliation(s)
- Sushanth Kumar
- Department of Biology and,Neuroscience Graduate Program, School of Medicine, and
| | | | | | | | | | - John R. Lukens
- Neuroscience Graduate Program, School of Medicine, and,Center for Brain Immunology and Glia (BIG), Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | | |
Collapse
|
31
|
Zhu D, Zhang J, Hashem J, Gao F, Chen C. Inhibition of 2-arachidonoylglycerol degradation enhances glial immunity by single-cell transcriptomic analysis. J Neuroinflammation 2023; 20:17. [PMID: 36717883 PMCID: PMC9885699 DOI: 10.1186/s12974-023-02701-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 01/17/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND 2-Arachidonoylglycerol (2-AG) is the most abundant endogenous cannabinoid. Inhibition of 2-AG metabolism by inactivation of monoacylglycerol lipase (MAGL), the primary enzyme that degrades 2-AG in the brain, produces anti-inflammatory and neuroprotective effects in neurodegenerative diseases. However, the molecular mechanisms underlying these beneficial effects are largely unclear. METHODS Hippocampal and cortical cells were isolated from cell type-specific MAGL knockout (KO) mice. Single-cell RNA sequencing was performed by 10 × Genomics platform. Cell Ranger, Seurat (v3.2) and CellChat (1.1.3) packages were used to carry out data analysis. RESULTS Using single-cell RNA sequencing analysis, we show here that cell type-specific MAGL KO mice display distinct gene expression profiles in the brain. Inactivation of MAGL results in robust changes in expression of immune- and inflammation-related genes in microglia and astrocytes. Remarkably, upregulated expression of chemokines in microglia is more pronounced in mice lacking MAGL in astrocytes. In addition, expression of genes that regulate other cellular functions and Wnt signaling in astrocytes is altered in MAGL KO mice. CONCLUSIONS Our results provide transcriptomic evidence that cell type-specific inactivation of MAGL induces differential expression of immune-related genes and other fundamental cellular pathways in microglia and astrocytes. Upregulation of the immune/inflammatory genes suggests that tonic levels of immune/inflammatory vigilance are enhanced in microglia and astrocytes, particularly in microglia, by inhibition of 2-AG metabolism, which likely contribute to anti-inflammatory and neuroprotective effects produced by inactivation of MAGL in neurodegenerative diseases.
Collapse
Affiliation(s)
- Dexiao Zhu
- grid.267309.90000 0001 0629 5880Department of Cellular and Integrative Physiology, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229 USA
| | - Jian Zhang
- grid.267309.90000 0001 0629 5880Department of Cellular and Integrative Physiology, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229 USA
| | - Jack Hashem
- grid.267309.90000 0001 0629 5880Department of Cellular and Integrative Physiology, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229 USA
| | - Fei Gao
- grid.267309.90000 0001 0629 5880Department of Cellular and Integrative Physiology, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229 USA
| | - Chu Chen
- grid.267309.90000 0001 0629 5880Department of Cellular and Integrative Physiology, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229 USA ,grid.267309.90000 0001 0629 5880Center for Biomedical Neuroscience, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA
| |
Collapse
|
32
|
Cai S, Lin J, Li Z, Liu S, Feng Z, Zhang Y, Zhang Y, Huang J, Chen Q. Alterations in intestinal microbiota and metabolites in individuals with Down syndrome and their correlation with inflammation and behavior disorders in mice. Front Microbiol 2023; 14:1016872. [PMID: 36910172 PMCID: PMC9998045 DOI: 10.3389/fmicb.2023.1016872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
The intestinal microbiota and fecal metabolome have been shown to play a vital role in human health, and can be affected by genetic and environmental factors. We found that individuals with Down syndrome (DS) had abnormal serum cytokine levels indicative of a pro-inflammatory environment. We investigated whether these individuals also had alterations in the intestinal microbiome. High-throughput sequencing of bacterial 16S rRNA gene in fecal samples from 17 individuals with DS and 23 non-DS volunteers revealed a significantly higher abundance of Prevotella, Escherichia/Shigella, Catenibacterium, and Allisonella in individuals with DS, which was positively associated with the levels of pro-inflammatory cytokines. GC-TOF-MS-based fecal metabolomics identified 35 biomarkers (21 up-regulated metabolites and 14 down-regulated metabolites) that were altered in the microbiome of individuals with DS. Metabolic pathway enrichment analyses of these biomarkers showed a characteristic pattern in DS that included changes in valine, leucine, and isoleucine biosynthesis and degradation; synthesis and degradation of ketone bodies; glyoxylate and dicarboxylate metabolism; tyrosine metabolism; lysine degradation; and the citrate cycle. Treatment of mice with fecal bacteria from individuals with DS or Prevotella copri significantly altered behaviors often seen in individuals with DS, such as depression-associated behavior and impairment of motor function. These studies suggest that changes in intestinal microbiota and the fecal metabolome are correlated with chronic inflammation and behavior disorders associated with DS.
Collapse
Affiliation(s)
- Shaoli Cai
- Biomedical Research Center of South China, Fujian Normal University, Fuzhou, Fujian, China.,Fujian Key Laboratory of Innate Immune Biology, Fujian Normal University, Fuzhou, Fujian, China.,College of Life Sciences, Fujian Normal University, Fuzhou, Fujian, China
| | - Jinxin Lin
- Biomedical Research Center of South China, Fujian Normal University, Fuzhou, Fujian, China.,Fujian Key Laboratory of Innate Immune Biology, Fujian Normal University, Fuzhou, Fujian, China.,College of Life Sciences, Fujian Normal University, Fuzhou, Fujian, China
| | - Zhaolong Li
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, Fujian, China
| | - Songnian Liu
- Biomedical Research Center of South China, Fujian Normal University, Fuzhou, Fujian, China.,Fujian Key Laboratory of Innate Immune Biology, Fujian Normal University, Fuzhou, Fujian, China
| | - Zhihua Feng
- Biomedical Research Center of South China, Fujian Normal University, Fuzhou, Fujian, China.,Fujian Key Laboratory of Innate Immune Biology, Fujian Normal University, Fuzhou, Fujian, China.,College of Life Sciences, Fujian Normal University, Fuzhou, Fujian, China
| | - Yangfan Zhang
- Biomedical Research Center of South China, Fujian Normal University, Fuzhou, Fujian, China.,Fujian Key Laboratory of Innate Immune Biology, Fujian Normal University, Fuzhou, Fujian, China.,College of Life Sciences, Fujian Normal University, Fuzhou, Fujian, China
| | - Yanding Zhang
- College of Life Sciences, Fujian Normal University, Fuzhou, Fujian, China
| | - Jianzhong Huang
- College of Life Sciences, Fujian Normal University, Fuzhou, Fujian, China
| | - Qi Chen
- Biomedical Research Center of South China, Fujian Normal University, Fuzhou, Fujian, China.,Fujian Key Laboratory of Innate Immune Biology, Fujian Normal University, Fuzhou, Fujian, China.,College of Life Sciences, Fujian Normal University, Fuzhou, Fujian, China
| |
Collapse
|
33
|
Ennerfelt H, Frost EL, Shapiro DA, Holliday C, Zengeler KE, Voithofer G, Bolte AC, Lammert CR, Kulas JA, Ulland TK, Lukens JR. SYK coordinates neuroprotective microglial responses in neurodegenerative disease. Cell 2022; 185:4135-4152.e22. [PMID: 36257314 PMCID: PMC9617784 DOI: 10.1016/j.cell.2022.09.030] [Citation(s) in RCA: 156] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 05/05/2022] [Accepted: 09/23/2022] [Indexed: 11/09/2022]
Abstract
Recent studies have begun to reveal critical roles for the brain's professional phagocytes, microglia, and their receptors in the control of neurotoxic amyloid beta (Aβ) and myelin debris accumulation in neurodegenerative disease. However, the critical intracellular molecules that orchestrate neuroprotective functions of microglia remain poorly understood. In our studies, we find that targeted deletion of SYK in microglia leads to exacerbated Aβ deposition, aggravated neuropathology, and cognitive defects in the 5xFAD mouse model of Alzheimer's disease (AD). Disruption of SYK signaling in this AD model was further shown to impede the development of disease-associated microglia (DAM), alter AKT/GSK3β-signaling, and restrict Aβ phagocytosis by microglia. Conversely, receptor-mediated activation of SYK limits Aβ load. We also found that SYK critically regulates microglial phagocytosis and DAM acquisition in demyelinating disease. Collectively, these results broaden our understanding of the key innate immune signaling molecules that instruct beneficial microglial functions in response to neurotoxic material.
Collapse
Affiliation(s)
- Hannah Ennerfelt
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA 22908, USA; Neuroscience Graduate Program, UVA, Charlottesville, VA 22908, USA; Cell and Molecular Biology Graduate Training Program, UVA, Charlottesville, VA 22908, USA
| | - Elizabeth L Frost
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA 22908, USA
| | - Daniel A Shapiro
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA 22908, USA
| | - Coco Holliday
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA 22908, USA
| | - Kristine E Zengeler
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA 22908, USA; Neuroscience Graduate Program, UVA, Charlottesville, VA 22908, USA; Cell and Molecular Biology Graduate Training Program, UVA, Charlottesville, VA 22908, USA
| | - Gabrielle Voithofer
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA 22908, USA
| | - Ashley C Bolte
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA 22908, USA; Department of Microbiology, Immunology and Cancer Biology, UVA, Charlottesville, VA 22908, USA; Medical Scientist Training Program, UVA, Charlottesville, VA 22908, USA
| | - Catherine R Lammert
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA 22908, USA; Neuroscience Graduate Program, UVA, Charlottesville, VA 22908, USA
| | - Joshua A Kulas
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA 22908, USA
| | - Tyler K Ulland
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI 53705, USA
| | - John R Lukens
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA 22908, USA; Neuroscience Graduate Program, UVA, Charlottesville, VA 22908, USA; Cell and Molecular Biology Graduate Training Program, UVA, Charlottesville, VA 22908, USA; Department of Microbiology, Immunology and Cancer Biology, UVA, Charlottesville, VA 22908, USA; Medical Scientist Training Program, UVA, Charlottesville, VA 22908, USA.
| |
Collapse
|
34
|
Cowan MN, Kovacs MA, Sethi I, Babcock IW, Still K, Batista SJ, O’Brien CA, Thompson JA, Sibley LA, Labuzan SA, Harris TH. Microglial STAT1-sufficiency is required for resistance to toxoplasmic encephalitis. PLoS Pathog 2022; 18:e1010637. [PMID: 36067217 PMCID: PMC9481170 DOI: 10.1371/journal.ppat.1010637] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/16/2022] [Accepted: 08/11/2022] [Indexed: 12/02/2022] Open
Abstract
Toxoplasma gondii is a ubiquitous intracellular protozoan parasite that establishes a life-long chronic infection largely restricted to the central nervous system (CNS). Constant immune pressure, notably IFN-γ-STAT1 signaling, is required for preventing fatal pathology during T. gondii infection. Here, we report that abrogation of STAT1 signaling in microglia, the resident immune cells of the CNS, is sufficient to induce a loss of parasite control in the CNS and susceptibility to toxoplasmic encephalitis during the early stages of chronic infection. Using a microglia-specific genetic labeling and targeting system that discriminates microglia from blood-derived myeloid cells that infiltrate the brain during infection, we find that, contrary to previous in vitro reports, microglia do not express inducible nitric-oxide synthase (iNOS) during T. gondii infection in vivo. Instead, transcriptomic analyses of microglia reveal that STAT1 regulates both (i) a transcriptional shift from homeostatic to “disease-associated microglia” (DAM) phenotype conserved across several neuroinflammatory models, including T. gondii infection, and (ii) the expression of anti-parasitic cytosolic molecules that are required for eliminating T. gondii in a cell-intrinsic manner. Further, genetic deletion of Stat1 from microglia during T. gondii challenge leads to fatal pathology despite largely equivalent or enhanced immune effector functions displayed by brain-infiltrating immune populations. Finally, we show that microglial STAT1-deficiency results in the overrepresentation of the highly replicative, lytic tachyzoite form of T. gondii, relative to its quiescent, semi-dormant bradyzoite form typical of chronic CNS infection. Our data suggest an overall protective role of CNS-resident microglia against T. gondii infection, illuminating (i) general mechanisms of CNS-specific immunity to infection (ii) and a clear role for IFN-STAT1 signaling in regulating a microglial activation phenotype observed across diverse neuroinflammatory disease states. The brain, an immune-privileged organ, can be invaded and colonized by pathogens such as the opportunistic parasite, Toxoplasma gondii. How microglia, the resident immune cells of the brain, provide resistance to infection is an active area of investigation. In this study, we used a genetic approach to generate and study mice with microglia that lack STAT1, a critical transcription factor that confers protection against intracellular pathogens in both humans and mice. We find that despite robust activation and recruitment of immune cells from the blood to the brain during infection, STAT1 deficiency in microglia leads to increased brain parasite burden and uniform lethality in mice when challenged with T. gondii. Our bioinformatic analyses also indicate that STAT1 in microglia regulates (i) the expression of large families of genes associated with parasite killing and (ii) a microglial activation state that has been classically seen in neurodegeneration. Our findings identify mechanisms by which microglia contribute to parasite control and contribute to a greater understanding of their cellular physiology during neuroinflammation.
Collapse
Affiliation(s)
- Maureen N. Cowan
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Michael A. Kovacs
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Ish Sethi
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Isaac W. Babcock
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Katherine Still
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Samantha J. Batista
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Carleigh A. O’Brien
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jeremy A. Thompson
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Lydia A. Sibley
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Sydney A. Labuzan
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Tajie H. Harris
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
35
|
Johnson HJ, Koshy AA. Understanding neuroinflammation through central nervous system infections. Curr Opin Neurobiol 2022; 76:102619. [PMID: 35985075 PMCID: PMC10147316 DOI: 10.1016/j.conb.2022.102619] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/09/2022] [Accepted: 07/08/2022] [Indexed: 11/28/2022]
Abstract
Neuroinflammation is now recognized to compound many central nervous system (CNS) pathologies, from stroke to dementia. As immune responses evolved to handle infections, studying CNS infections can offer unique insights into the CNS immune response and address questions such as: What defenses and strategies do CNS parenchymal cells deploy in response to a dangerous pathogen? How do CNS cells interact with each other and infiltrating immune cells to control microbes? What pathways are beneficial for the host or for the pathogen? Here, we review recent studies that use CNS-tropic infections in combination with cutting-edge techniques to delve into the complex relationships between microbes, immune cells, and cells of the CNS.
Collapse
Affiliation(s)
- Hannah J Johnson
- Neuroscience Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, USA
| | - Anita A Koshy
- Neuroscience Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, USA; Department of Neurology, University of Arizona, Tucson, AZ, USA; BIO5 Institute, University of Arizona, Tucson, AZ, USA; Department of Immunobiology, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
36
|
Bai RX, Chen XZ, Ren JF, Hu L, Li H, Wang H, He C. Toxoplasma gondii rhoptry protein (TgROP18) enhances the expression of pro-inflammatory factor in LPS/IFN-γ-induced murine BV2 microglia cells via NF-κB signal pathway. Acta Trop 2022; 235:106650. [PMID: 35963313 DOI: 10.1016/j.actatropica.2022.106650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/09/2022] [Accepted: 08/09/2022] [Indexed: 11/25/2022]
Abstract
Toxoplasma gondii, an opportunistic pathogenic protozoan, exhibits a strong predilection to infect the brain, causing severe neurological diseases, such as toxoplasmic encephalitis (TE), in immunocompromised patients. Microglia, the resident immune cells in the brain, is reported to play important roles in regulating the neuroinflammation mediated by T. gondii infection. Here we demonstrated that the tachyzoites of T. gondii RH strain could significantly upregulate the expression levels of microglial M1 phenotype markers including IL-1β, IL-6, TNF-α, iNOS and IL18 in activated murine BV2 microglia cells, which were regulated by T. gondii rhoptry protein 18 (TgROP18). Moreover, we found that TgROP18 could enhance the expression of M1 phenotype markers in activated murine BV2 microglia cells via activating NF-κB signal pathway. Additionally, TgROP18 was suggested to interact with the host p65 in activated murine BV2 microglia cells and induce the phosphorylation of p65 at S536. In summary, the present study demonstrated that TgROP18 could promote the activated microglia to polarize to M1 phenotype and enhanced the expression of pro-inflammatory factors via activating NF-κB signal pathway, which could contribute to elucidating the mechanism underlying the neuroinflammation mediated by activated microglia in the brain with T. gondii infection.
Collapse
Affiliation(s)
- Rui-Xue Bai
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China
| | - Xin-Zhu Chen
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China
| | - Jin-Feng Ren
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China
| | - Lang Hu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China
| | - Hui Li
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China
| | - Hui Wang
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China.
| | - Cheng He
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China.
| |
Collapse
|
37
|
Primary Infection by E. multilocularis Induces Distinct Patterns of Cross Talk between Hepatic Natural Killer T Cells and Regulatory T Cells in Mice. Infect Immun 2022; 90:e0017422. [PMID: 35862712 PMCID: PMC9387288 DOI: 10.1128/iai.00174-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The larval stage of the helminthic cestode Echinococcus multilocularis can inflict tumor-like hepatic lesions that cause the parasitic disease alveolar echinococcosis in humans, with high mortality in untreated patients. Opportunistic properties of the disease have been established based on the increased incidence in immunocompromised patients and mouse models, indicating that an appropriate adaptive immune response is required for the control of the disease. However, cellular interactions and the kinetics of the local hepatic immune responses during the different stages of infection with E. multilocularis remain unknown. In a mouse model of oral infection that mimics the normal infection route in human patients, the networks of the hepatic immune response were assessed using single-cell RNA sequencing (scRNA-seq) of isolated hepatic CD3+ T cells at different infection stages. We observed an early and sustained significant increase in natural killer T (NKT) cells and regulatory T cells (Tregs). Early tumor necrosis factor (TNF)- and integrin-dependent interactions between these two cell types promote the formation of hepatic lesions. At late time points, downregulation of programmed cell death protein 1 (PD-1) and ectonucleoside triphosphate diphosphohydrolase 1 (ENTPD1)-dependent signaling suppress the resolution of parasite-induced pathology. The obtained data provide fresh insight into the adaptive immune responses and local regulatory pathways at different infection stages of E. multilocularis in mice.
Collapse
|
38
|
Immune responses to Toxoplasma gondii. Curr Opin Immunol 2022; 77:102226. [PMID: 35785567 DOI: 10.1016/j.coi.2022.102226] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/19/2022] [Accepted: 05/19/2022] [Indexed: 11/22/2022]
Abstract
Toxoplasma gondii is an obligate intracellular parasite that can cause severe complications in the newborn and immunocompromised individuals. The parasite evokes a strong innate immune response in the infected hosts which is followed by a robust adaptive immunity. In the last few years, importance of innate immune mechanisms dependent on the role of MyD-88 independent pathways, inflammatory monocytes and innate lymphocyte have been identified. However, notwithstanding the strong immune response to the parasite, the chronic infection persists in the host. The inability to prevent chronic infection can be attributed to aberration in the memory CD8 T cell response caused by an increased expression of inhibitory receptors that leads to their dysfunctionality.
Collapse
|
39
|
Wang S, Moreau F, Chadee K. Gasdermins in Innate Host Defense Against Entamoeba histolytica and Other Protozoan Parasites. Front Immunol 2022; 13:900553. [PMID: 35795683 PMCID: PMC9251357 DOI: 10.3389/fimmu.2022.900553] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 05/23/2022] [Indexed: 11/16/2022] Open
Abstract
Gasdermins (GSDMs) are a group of proteins that are cleaved by inflammatory caspases to induce pore formation in the plasma membrane to cause membrane permeabilization and lytic cell death or pyroptosis. All GSDMs share a conserved structure, containing a cytotoxic N-terminal (NT) pore-forming domain and a C-terminal (CT) repressor domain. Entamoeba histolytica (Eh) in contact with macrophages, triggers outside-in signaling to activate inflammatory caspase-4/1 via the noncanonical and canonical pathway to promote cleavage of gasdermin D (GSDMD). Cleavage of GSDMD removes the auto-inhibition that masks the active pore-forming NT domain in the full-length protein by interactions with GSDM-CT. The cleaved NT-GSDMD monomers then oligomerize to form pores in the plasma membrane to facilitate the release of IL-1β and IL-18 with a measured amount of pyroptosis. Pyroptosis is an effective way to counteract intracellular parasites, which exploit replicative niche to avoid killing. To date, most GSDMs have been verified to perform pore-forming activity and GSDMD-induced pyroptosis is rapidly emerging as a mechanism of anti-microbial host defence. Here, we review our comprehensive and current knowledge on the expression, activation, biological functions, and regulation of GSDMD cleavage with emphases on physiological scenario and related dysfunctions of each GSDM member as executioner of cell death, cytokine secretion and inflammation against Eh and other protozoan parasitic infections.
Collapse
Affiliation(s)
| | | | - Kris Chadee
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
40
|
Ma H, Cheng N, Zhang C. Schizophrenia and Alarmins. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58060694. [PMID: 35743957 PMCID: PMC9230958 DOI: 10.3390/medicina58060694] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/20/2022] [Accepted: 05/20/2022] [Indexed: 11/16/2022]
Abstract
Schizophrenia, consisting of a group of severe psychiatric disorders with a complex etiology, is a leading cause of disability globally. Due to the lack of objective indicators, accurate diagnosis and selection of effective treatments for schizophrenia remain challenging. The association between schizophrenia and alarmins levels has been proposed for many years, but without solid evidence. Alarmins are prestored molecules that do not require processing and can be released upon cell death or damage, making them an ideal candidate for an early initiator of inflammation. Immunological biomarkers seem to be related to disease progression and treatment effectiveness. Several studies suggest strong associations among the high-mobility group box 1 protein (HMGB1), interleukin-1α, interleukin-33, S100B, heat-shock proteins, and uric acid with schizophrenic disorders. The purpose of this review is to discuss the evidence of central and peripheral immune findings in schizophrenia, their potential causes, and the effects of immunomodulatory therapies on symptoms and outline potential applications of these markers in managing the illness. Although there are currently no effective markers for diagnosing or predicting treatment effects in patients with schizophrenia, we believe that screening immune-inflammatory biomarkers that are closely related to the pathological mechanism of schizophrenia can be used for early clinical identification, diagnosis, and treatment of schizophrenia, which may lead to more effective treatment options for people with schizophrenia.
Collapse
Affiliation(s)
- Huan Ma
- Department of Psychiatry, First Clinical College, Xuzhou Medical University, Xuzhou 221000, China; (H.M.); (N.C.)
| | - Ning Cheng
- Department of Psychiatry, First Clinical College, Xuzhou Medical University, Xuzhou 221000, China; (H.M.); (N.C.)
| | - Caiyi Zhang
- Department of Psychiatry, First Clinical College, Xuzhou Medical University, Xuzhou 221000, China; (H.M.); (N.C.)
- Department of Psychiatry, The Affiliated Xuzhou Oriental Hospital of Xuzhou Medical University, Xuzhou 221000, China
- Department of Medical Psychology, Second Clinical College, Xuzhou Medical University, Xuzhou 221000, China
- Correspondence: ; Tel.: +86-137-7588-9105
| |
Collapse
|
41
|
Sun R, Gao DS, Shoush J, Lu B. The IL-1 family in tumorigenesis and antitumor immunity. Semin Cancer Biol 2022; 86:280-295. [DOI: 10.1016/j.semcancer.2022.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/24/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022]
|
42
|
Zhang Y, Zhang C, Yang Y, Wang G, Wang Z, Liu J, Zhang L, Yu Y. Pyroptosis-Related Gene Signature Predicts Prognosis and Indicates Immune Microenvironment Infiltration in Glioma. Front Cell Dev Biol 2022; 10:862493. [PMID: 35547808 PMCID: PMC9081442 DOI: 10.3389/fcell.2022.862493] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/04/2022] [Indexed: 01/13/2023] Open
Abstract
Objective: Gliomas are the most common primary tumors in the central nervous system with a bad prognosis. Pyroptosis, an inflammatory form of regulated cell death, plays a vital role in the progression and occurrence of tumors. However, the value of pyroptosis related genes (PRGs) in glioma remains poorly understood. This study aims to construct a PRGs signature risk model and explore the correlation with clinical characteristics, prognosis, tumor microenviroment (TME), and immune checkpoints. Methods: RNA sequencing profiles and the relevant clinical data were obtained from the Chinese Glioma Genome Atlas (CGGA), the Cancer Genome Atlas (TCGA), the Repository of Molecular Brain Neoplasia Data (REMBRANDT), and the Genotype-Tissue Expression Project (GTEx-Brain). Then, the differentially expressed pyroptosis related genes (PRGs) were identified, and the least absolute shrinkage and selection operator (LASSO) and mutiCox regression model was generated using the TCGA-train dataset. Then the expression of mRNA and protein levels of PRGs signature was detected through qPCR and human protein atlas (HPA). Further, the predictive ability of the PRGs-signature, prognostic analysis, and stratification analysis were utilized and validated using TCGA-test, CGGA, and REMBRANDT datasets. Subsequently, we constructed the nomogram by combining the PRGs signature and other key clinical features. Moreover, we used gene set enrichment analysis (GSEA), GO, KEGG, the tumor immune dysfunction and exclusion (TIDE) single-sample GSEA (ssGSEA), and Immunophenoscore (IPS) to determine the relationship between PRGs and TME, immune infiltration, and predict the response of immune therapy in glioma. Results: A four-gene PRGs signature (CASP4, CASP9, GSDMC, IL1A) was identified and stratified patients into low- or high-risk group. Survival analysis, ROC curves, and stratified analysis revealed worse outcomes in the high-risk group than in the low-risk group. Correlation analysis showed that the risk score was correlated with poor disease features. Furthermore, GSEA and immune infiltrating and IPS analysis showed that the PRGs signature could potentially predict the TME, immune infiltration, and immune response in glioma. Conclusion: The newly identified four-gene PRGs signature is effective in diagnosis and could robustly predict the prognosis of glioma, and its impact on the TME and immune cell infiltrations may provide further guidance for immunotherapy.
Collapse
Affiliation(s)
- Yulian Zhang
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
| | - Chuanpeng Zhang
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
- Department of Neurosurgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Yanbo Yang
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
- Department of Neurosurgery, Graduate School of Peking Union Medical College, Beijing, China
| | - Guohui Wang
- Department of Radiotherapy, Tianjin First Center Hospital, Tianjin, China
| | - Zai Wang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Jiang Liu
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
| | - Li Zhang
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
- Department of Neurosurgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
- Department of Neurosurgery, Graduate School of Peking Union Medical College, Beijing, China
| | - Yanbing Yu
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
- Department of Neurosurgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
- Department of Neurosurgery, Graduate School of Peking Union Medical College, Beijing, China
- *Correspondence: Yanbing Yu,
| |
Collapse
|
43
|
Demarco B, Danielli S, Fischer FA, Bezbradica JS. How Pyroptosis Contributes to Inflammation and Fibroblast-Macrophage Cross-Talk in Rheumatoid Arthritis. Cells 2022; 11:1307. [PMID: 35455985 PMCID: PMC9028325 DOI: 10.3390/cells11081307] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/09/2022] [Accepted: 04/11/2022] [Indexed: 12/11/2022] Open
Abstract
About thirty years ago, a new form of pro-inflammatory lytic cell death was observed and termed pyroptosis. Only in 2015, gasdermins were defined as molecules that create pores at the plasma membrane and drive pyroptosis. Today, we know that gasdermin-mediated death is an important antimicrobial defence mechanism in bacteria, yeast and mammals as it destroys the intracellular niche for pathogen replication. However, excessive and uncontrolled cell death also contributes to immunopathology in several chronic inflammatory diseases, including arthritis. In this review, we discuss recent findings where pyroptosis contributes to tissue damage and inflammation with a main focus on injury-induced and autoimmune arthritis. We also review novel functions and regulatory mechanisms of the pyroptotic executors gasdermins. Finally, we discuss possible models of how pyroptosis may contribute to the cross-talk between fibroblast and macrophages, and also how this cross-talk may regulate inflammation by modulating inflammasome activation and pyroptosis induction.
Collapse
Affiliation(s)
- Benjamin Demarco
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK; (S.D.); (F.A.F.)
| | | | | | - Jelena S. Bezbradica
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK; (S.D.); (F.A.F.)
| |
Collapse
|
44
|
Life and death of microglia: mechanisms governing microglial states and fates. Immunol Lett 2022; 245:51-60. [PMID: 35413354 DOI: 10.1016/j.imlet.2022.04.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 12/16/2022]
|
45
|
Forrester JV, Mölzer C, Kuffova L. Immune Privilege Furnishes a Niche for Latent Infection. FRONTIERS IN OPHTHALMOLOGY 2022; 2:869046. [PMID: 38983514 PMCID: PMC11182092 DOI: 10.3389/fopht.2022.869046] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/15/2022] [Indexed: 07/11/2024]
Abstract
The microenvironment of the CNS (eye and brain) is fertile ground for infection if the barriers are breached. The result of pathogen invasion is often devastating destruction of tissues. In the eye, inflammation is broadly classified either as "infectious" (i.e. caused by infection) or "non-infectious". However, increasingly, forms of intraocular inflammation (IOI), which clinically appear to be "non-infectious" turn out to be initiated by infectious agents, suggesting that pathogens have been retained in latent or persistent form within ocular tissues and have reactivated to cause overt disease. A similar pathogenesis applies to latent infections in the brain. Not all CNS tissues provide an equally protective niche while different pathogens escape detection using different strategies. This review summarises how immune privilege (IP) in the CNS may be permissive for latent infection and allow the eye and the brain to act as a reservoir of pathogens which often remain undetected for the lifetime of the host but in states of immune deficiency may be activated to cause sight- and life-threatening inflammation.
Collapse
Affiliation(s)
- John V Forrester
- Ocular Immunology Group, Section of Infection and Immunity, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Christine Mölzer
- Ocular Immunology Group, Section of Infection and Immunity, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Lucia Kuffova
- Ocular Immunology Group, Section of Infection and Immunity, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
- Eye Clinic, Aberdeen Royal Infirmary, Aberdeen, United Kingdom
| |
Collapse
|
46
|
Nemeth DP, Liu X, McKim DB, DiSabato DJ, Oliver B, Herd A, Katta A, Negray CE, Floyd J, McGovern S, Pruden PS, Zhutang F, Smirnova M, Godbout JP, Sheridan J, Quan N. Dynamic Interleukin-1 Receptor Type 1 Signaling Mediates Microglia-Vasculature Interactions Following Repeated Systemic LPS. J Inflamm Res 2022; 15:1575-1590. [PMID: 35282272 PMCID: PMC8906862 DOI: 10.2147/jir.s350114] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/16/2022] [Indexed: 01/18/2023] Open
Abstract
Introduction Lipopolysaccharide (LPS) preconditioning involves repeated, systemic, and sub-threshold doses of LPS, which induces a neuroprotective state within the CNS, thus preventing neuronal death and functional losses. Recently, proinflammatory cytokine, Interleukin-1 (IL-1), and its primary signaling partner, interleukin-1 receptor type 1 (IL-1R1), have been associated with neuroprotection in the CNS. However, it is still unknown how IL-1/IL-1R1 signaling impacts the processes associated with neuroprotection. Methods Using our IL-1R1 restore genetic mouse model, mouse lines were generated to restrict IL-1R1 expression either to endothelia (Tie2-Cre-Il1r1r/r) or microglia (Cx3Cr1-Cre-Il1r1 r/r), in addition to either global ablation (Il1r1 r/r) or global restoration of IL-1R1 (Il1r1 GR/GR). The LPS preconditioning paradigm consisted of four daily i.p. injections of LPS at 1 mg/kg (4d LPS). 24 hrs following the final i.p. LPS injection, tissue was collected for qPCR analysis, immunohistochemistry, or FAC sorting. Results Following 4d LPS, we found multiple phenotypes that are dependent on IL-1R1 signaling such as microglia morphology alterations, increased microglial M2-like gene expression, and clustering of microglia onto the brain vasculature. We determined that 4d LPS induces microglial morphological changes, clustering at the vasculature, and gene expression changes are dependent on endothelial IL-1R1, but not microglial IL-1R1. A novel observation was the induction of microglial IL-1R1 (mIL-1R1) following 4d LPS. The induced mIL-1R1 permits a unique response to central IL-1β: the mIL-1R1 dependent induction of IL-1R1 antagonist (IL-1RA) and IL-1β gene expression. Analysis of RNA sequencing datasets revealed that mIL-1R1 is also induced in neurodegenerative diseases. Discussion Here, we have identified cell type-specific IL-1R1 mediated mechanisms, which may contribute to the neuroprotection observed in LPS preconditioning. These findings identify key cellular and molecular contributors in LPS-induced neuroprotection.
Collapse
Affiliation(s)
- Daniel P Nemeth
- College of Dentistry, The Ohio State University, Columbus, OH, USA,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA,Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA,Correspondence: Daniel P Nemeth; Ning Quan, 5353 Parkside Drive, Jupiter, FL, 33458, USA, Email ;
| | - Xiaoyu Liu
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Daniel B McKim
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Champaign, IL, USA
| | - Damon J DiSabato
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA,Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Braedan Oliver
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Anu Herd
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Asish Katta
- College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Christina E Negray
- College of Dentistry, The Ohio State University, Columbus, OH, USA,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - James Floyd
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Samantha McGovern
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Paige S Pruden
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Feiyang Zhutang
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Maria Smirnova
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Jonathan P Godbout
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA,Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - John Sheridan
- College of Dentistry, The Ohio State University, Columbus, OH, USA,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Ning Quan
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| |
Collapse
|
47
|
Song M, Wang J, Sun Y, Pang J, Li X, Liu Y, Zhou Y, Yang P, Fan T, Liu Y, Li Z, Qi X, Li B, Zhang X, Wang J, Wang C. Inhibition of gasdermin D-dependent pyroptosis attenuates the progression of silica-induced pulmonary inflammation and fibrosis. Acta Pharm Sin B 2022; 12:1213-1224. [PMID: 35530143 PMCID: PMC9069405 DOI: 10.1016/j.apsb.2021.10.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/12/2021] [Accepted: 09/30/2021] [Indexed: 12/13/2022] Open
Abstract
Silicosis is a leading cause of occupational disease-related morbidity and mortality worldwide, but the molecular basis underlying its development remains unclear. An accumulating body of evidence supports gasdermin D (GSDMD)-mediated pyroptosis as a key component in the development of various pulmonary diseases. However, there is little experimental evidence connecting silicosis and GSDMD-driven pyroptosis. In this work, we investigated the role of GSDMD-mediated pyroptosis in silicosis. Single-cell RNA sequencing of healthy and silicosis human and murine lung tissues indicated that GSDMD-induced pyroptosis in macrophages was relevant to silicosis progression. Through microscopy we then observed morphological alterations of pyroptosis in macrophages treated with silica. Measurement of interleukin-1β release, lactic dehydrogenase activity, and real-time propidium iodide staining further revealed that silica induced pyroptosis of macrophages. Additionally, we verified that both canonical (caspase-1-mediated) and non-canonical (caspase-4/5/11-mediated) signaling pathways mediated silica-induced pyroptosis activation, in vivo and in vitro. Notably, Gsdmd knockout mice exhibited dramatically alleviated silicosis phenotypes, which highlighted the pivotal role of pyroptosis in this disease. Taken together, our results demonstrated that macrophages underwent GSDMD-dependent pyroptosis in silicosis and inhibition of this process could serve as a viable clinical strategy for mitigating silicosis.
Collapse
|
48
|
Cowan MN, Sethi I, Harris TH. Microglia in CNS infections: insights from Toxoplasma gondii and other pathogens. Trends Parasitol 2022; 38:217-229. [PMID: 35039238 PMCID: PMC8852251 DOI: 10.1016/j.pt.2021.12.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 12/11/2022]
Abstract
Microglia, the resident immune cells of the central nervous system (CNS), are poised to respond to neuropathology. Microglia play multiple roles in maintaining homeostasis and promoting inflammation in numerous disease states. The study of microglial innate immune programs has largely focused on exploring neurodegenerative disease states with the use of genetic targeting approaches. Our understanding of how microglia participate in immune responses against pathogens is just beginning to take shape. Here, we review existing animal models of CNS infection, with a focus on how microglial physiology and inflammatory processes control protozoan and viral infections of the brain. We further discuss how microglial participation in over-exuberant immune responses can drive immunopathology that is detrimental to CNS health and homeostasis.
Collapse
Affiliation(s)
- Maureen N. Cowan
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA, United States
| | - Ish Sethi
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA, United States
| | - Tajie H. Harris
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA, United States,Correspondence: (T. H. Harris)
| |
Collapse
|
49
|
Wisuitiprot V, Ingkaninan K, Chakkavittumrong P, Wisuitiprot W, Neungchamnong N, Chantakul R, Waranuch N. Effects of Acanthus ebracteatus Vahl. extract and verbascoside on human dermal papilla and murine macrophage. Sci Rep 2022; 12:1491. [PMID: 35087085 PMCID: PMC8795396 DOI: 10.1038/s41598-022-04966-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/04/2022] [Indexed: 12/22/2022] Open
Abstract
Androgenic alopecia is a common type of hair loss, usually caused by testosterone metabolism generating dihydrotestosterone and hair follicular micro-inflammation. These processes induce dermal papilla cells to undergo apoptosis. Currently approved effective medications for alopecia are Finasteride, an oral 5α-reductase inhibitor, Minoxidil, a topical hair growth promoter, and Diclofenac, an anti-inflammatory agent, all of which, however, have several adverse side effects. In our study, we showed the bioactivity of Acanthus ebracteatus Vahl. (AE) extract performed by 95% ethanol, and verbascoside (VB), a biomarker of AE extract. Both AE extract and VB were studied for their effects on dermal papilla cell viability and the cell cycle by using MTT assay and flow cytometry. The effect of an anti-inflammatory activity of AE extract and VB on IL-1β, NO, and TNF-α, released from LPS induced RAW 264.7 cells, and IL-1α and IL-6 released from irradiated dermal papilla cells were detected using ELISA technique. The preventive effect on dermal papilla cell apoptosis induced by testosterone was determined by MTT assay. In controlled in vitro assays it was found that AE extract and VB at various concentrations induced dermal papilla cell proliferation which was indicated by an increase in the number of cells in the S and G2/M phases of the cell cycle. AE extract at 250 µg/mL concentration or VB at 62.50 µg/mL concentration prevented cell apoptosis induced by testosterone at a statistically significant level. In addition, both AE extract and VB greatly inhibited the release of pro-inflammatory cytokines from RAW 264.7 and dermal papilla cells. The release of IL-1β, TNF-α, and NO from RAW 264.7 cells, as well as IL-1α and IL-6 from dermal papilla cells, was also diminished by AE extract 250 µg/mL and VB 125 µg/mL. Our results indicate that AE extract and VB are promising ingredients for anti-hair loss applications. However, further clinical study is necessary to evaluate the effectiveness of AE extract and VB as treatment for actual hair loss.
Collapse
Affiliation(s)
- Vanuchawan Wisuitiprot
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences and Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok, 65000, Thailand
| | - Kornkanok Ingkaninan
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| | - Panlop Chakkavittumrong
- Division of Dermatology, Department of Medicine, Faculty of Medicine, Thammasat University, Khlong Luang, Pathumthani, 12121, Thailand
| | - Wudtichai Wisuitiprot
- Department of Thai Traditional Medicine, Sirindhorn College of Public Health, Phitsanulok, 65130, Thailand
| | - Nitra Neungchamnong
- Science Laboratory Centre, Faculty of Science, Naresuan University, Mueang, Phitsanulok, 65000, Thailand
| | - Ruttanaporn Chantakul
- Bioscreening Unit, Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| | - Neti Waranuch
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences and Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok, 65000, Thailand. .,Cosmetics and Natural Products Research Center, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, 65000, Thailand.
| |
Collapse
|
50
|
Jiang D, Wu S, Xu L, Xie G, Li D, Peng H. Anti-infection roles of miR-155-5p packaged in exosomes secreted by dendritic cells infected with Toxoplasma gondii. Parasit Vectors 2022; 15:3. [PMID: 34986898 PMCID: PMC8731220 DOI: 10.1186/s13071-021-05003-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 09/08/2021] [Indexed: 02/08/2023] Open
Abstract
Background Toxoplasma gondii is a zoonotic intracellular protozoon that is estimated to infect about 30% of the world’s population, resulting in toxoplasmosis in immunocompromised patients and adverse outcomes in cases of primary infection during pregnancy. Exosomes are tubular vesicles secreted by cells, and function in intercellular communication. It has been reported that the exosomes secreted by T. gondii-infected immune cells transmit infection signals to the uninfected cells. However, the mechanism and effect of the exosome transmission are still vague. We therefore investigated the function of the exosomes transmitted from DC2.4 cells infected with the T. gondii RH strain (Tg-DC-Exo) to the uninfected cells, as well as their roles in anti-infection. Methods We conducted exosome isolation and identification with ultracentrifugation, transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and western blot (WB) analysis. Exosome uptake by recipient cells was identified by PKH67 assay. The signal transmission and the abundance of miR-155-5p were determined using transwell assay and qRT-PCR. For detection of immune responses, cytokine secretion was evaluated. The T. gondii B1 gene was determined to evaluate tachyzoite proliferation. Results We observed that Toxoplasma infection upregulated miR-155-5p expression in DC2.4 cell-secreted exosomes, and those exosomes could be ingested by murine macrophage RAW264.7 cells. Tg-DC-Exo and miR-155-5p stimulated host proinflammatory immune responses including increased production of proinflammatory cytokines IL-6 and TNF-α, and proinflammatory marker-inducible nitric oxide synthase (iNOS). The NF-κB pathway was activated by downregulation of SOCS1, leading to inhibition of T. gondii tachyzoite proliferation in RAW264.7 cells. Conclusions Our findings provide a novel mechanism for how infected cells transmit infection signals to the uninfected cells through exosome secretion after T. gondii infection, followed by inflammatory responses and anti-infection reactions, which may help us develop a new strategy for toxoplasmosis prevention, especially in immunocompromised patients. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13071-021-05003-x.
Collapse
Affiliation(s)
- Dan Jiang
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Shuizhen Wu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Liqing Xu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Guantai Xie
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Dongliang Li
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Hongjuan Peng
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, Guangdong Province, China.
| |
Collapse
|