1
|
Hilligan KL, Darrah PA, Seder RA, Sher A. Deconvoluting the interplay of innate and adaptive immunity in BCG-induced nonspecific and TB-specific host resistance. J Exp Med 2025; 222:e20240496. [PMID: 40100096 PMCID: PMC11917170 DOI: 10.1084/jem.20240496] [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: 10/28/2024] [Revised: 01/23/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025] Open
Abstract
BCG is the oldest vaccine in continuous use. While current intradermal vaccination regimens confer limited protection outside the context of pediatric extrapulmonary tuberculosis (TB), promising new data indicate that when administered mucosally or intravenously at a higher dose, BCG can induce sterilizing immunity against pulmonary TB in nonhuman primates. BCG is also known to promote nonspecific host resistance against a variety of unrelated infections and is a standard immunotherapy for bladder cancer, suggesting that this innate immune function may contribute to its protective role against TB. Here, we propose that both the mycobacterial-specific and off-target effects of BCG depend on the interplay of adaptive and innate cells and the cytokines they produce, and that the elucidation of this interaction should be a major strategy in the development of more effective BCG-based vaccines and immunotherapies.
Collapse
Affiliation(s)
| | - Patricia A. Darrah
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Robert A. Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alan Sher
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Warrick KA, Vallez CN, Meibers HE, Pasare C. Bidirectional Communication Between the Innate and Adaptive Immune Systems. Annu Rev Immunol 2025; 43:489-514. [PMID: 40279312 DOI: 10.1146/annurev-immunol-083122-040624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
Effective bidirectional communication between the innate and adaptive immune systems is crucial for tissue homeostasis and protective immunity against infections. The innate immune system is responsible for the early sensing of and initial response to threats, including microbial ligands, toxins, and tissue damage. Pathogen-related information, detected primarily by the innate immune system via dendritic cells, is relayed to adaptive immune cells, leading to the priming and differentiation of naive T cells into effector and memory lineages. Memory T cells that persist long after pathogen clearance are integral for durable protective immunity. In addition to rapidly responding to reinfections, memory T cells also directly instruct the interacting myeloid cells to induce innate inflammation, which resembles microbial inflammation. As such, memory T cells act as newly emerging activators of the innate immune system and function independently of direct microbial recognition. While T cell-mediated activation of the innate immune system likely evolved as a protective mechanism to combat reinfections by virulent pathogens, the detrimental outcomes of this mechanism manifest in the forms of autoimmunity and other T cell-driven pathologies. Here, we review the complexities and layers of regulation at the interface between the innate and adaptive immune systems to highlight the implications of adaptive instruction of innate immunity in health and disease.
Collapse
Affiliation(s)
- Kathrynne A Warrick
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| | - Charles N Vallez
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| | - Hannah E Meibers
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| | - Chandrashekhar Pasare
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| |
Collapse
|
3
|
Freitas IL, Macedo MF, Oliveira L, Oliveira P, do Vale A, dos Santos NM. AIP56, an AB toxin secreted by Photobacterium damselae subsp. piscicida, has tropism for myeloid cells. Front Immunol 2025; 15:1527088. [PMID: 39872526 PMCID: PMC11769971 DOI: 10.3389/fimmu.2024.1527088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 12/17/2024] [Indexed: 01/30/2025] Open
Abstract
Introduction The AB-type toxin AIP56 is a key virulence factor of Photobacterium damselae subsp. piscicida (Phdp), inducing apoptosis in fish immune cells. The discovery of AIP56-like and AIP56-related toxins in diverse organisms, including human-associated Vibrio strains, highlights the evolutionary conservation of this toxin family, suggesting that AIP56 and its homologs may share conserved receptors across species. These toxins have potential for biotechnological applications, such as therapeutic protein delivery and immune modulation. Methods Herein, the cell specificity of AIP56 for immune cells was characterized. The tropism of AIP56 for cells of the sea bass, mouse and human immune system was analyzed by following toxin internalization by flow cytometry and arrival of the toxin in the cytosol by evaluating the cleavage of NF-kB p65 by western blotting. Results Only a small population of sea bass neutrophils internalized AIP56, indicating that most of the neutrophilic destruction during Phdp infection and/or AIP56 intoxication does not result from the direct action of the toxin. Moreover, the cellular tropism of AIP56 for myeloid cells was observed in the three species, including its preference for macrophages. Further, mouse and human M0 and M2-like macrophages internalized more toxin than M1-like macrophages. Despite the limited interaction of lymphoid cells with AIP56, mouse B1-cells were able to internalize the toxin, possibly due to its myeloid features. Conclusion AIP56 has tropism for sea bass, mouse and human myeloid cells, with greater affinity for macrophages. This points to an evolutionary conservation of its receptor(s) and mechanism of action across species, raising the possibility that AIP56-like and -related toxins may also play a role in pathogenesis. These findings are relevant for both pathogenicity and biomedical contexts.
Collapse
Affiliation(s)
- Inês Lua Freitas
- Fish Immunology and Vaccinology Group, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Fish Immunology and Vaccinology Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- McBiology Doctoral Program, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Maria Fátima Macedo
- Cell Activation and Gene Expression, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Cell Activation and Gene Expression, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Liliana Oliveira
- Cell Activation and Gene Expression, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Cell Activation and Gene Expression, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Pedro Oliveira
- EPIUnit, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Ana do Vale
- Fish Immunology and Vaccinology Group, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Fish Immunology and Vaccinology Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Nuno M.S. dos Santos
- Fish Immunology and Vaccinology Group, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Fish Immunology and Vaccinology Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| |
Collapse
|
4
|
Ortega MA, Fraile-Martinez O, García-Montero C, Diaz-Pedrero R, Lopez-Gonzalez L, Monserrat J, Barrena-Blázquez S, Alvarez-Mon MA, Lahera G, Alvarez-Mon M. Understanding immune system dysfunction and its context in mood disorders: psychoneuroimmunoendocrinology and clinical interventions. Mil Med Res 2024; 11:80. [PMID: 39681901 DOI: 10.1186/s40779-024-00577-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/01/2024] [Indexed: 12/18/2024] Open
Abstract
Mood disorders include a set of psychiatric manifestations of increasing prevalence in our society, being mainly represented by major depressive disorder (MDD) and bipolar disorder (BD). The etiopathogenesis of mood disorders is extremely complex, with a wide spectrum of biological, psychological, and sociocultural factors being responsible for their appearance and development. In this sense, immune system dysfunction represents a key mechanism in the onset and pathophysiology of mood disorders, worsening mainly the central nervous system (neuroinflammation) and the periphery of the body (systemic inflammation). However, these alterations cannot be understood separately, but as part of a complex picture in which different factors and systems interact with each other. Psychoneuroimmunoendocrinology (PNIE) is the area responsible for studying the relationship between these elements and the impact of mind-body integration, placing the immune system as part of a whole. Thus, the dysfunction of the immune system is capable of influencing and activating different mechanisms that promote disruption of the psyche, damage to the nervous system, alterations to the endocrine and metabolic systems, and disruption of the microbiota and intestinal ecosystem, as well as of other organs and, in turn, all these mechanisms are responsible for inducing and enhancing the immune dysfunction. Similarly, the clinical approach to these patients is usually multidisciplinary, and the therapeutic arsenal includes different pharmacological (for example, antidepressants, antipsychotics, and lithium) and non-pharmacological (i.e., psychotherapy, lifestyle, and electroconvulsive therapy) treatments. These interventions also modulate the immune system and other elements of the PNIE in these patients, which may be interesting to understand the therapeutic success or failure of these approaches. In this sense, this review aims to delve into the relationship between immune dysfunction and mood disorders and their integration in the complex context of PNIE. Likewise, an attempt will be made to explore the effects on the immune system of different strategies available in the clinical approach to these patients, in order to identify the mechanisms described and their possible uses as biomarkers.
Collapse
Affiliation(s)
- Miguel A Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcalá de Henares, Spain.
- Ramón y Cajal Institute of Sanitary Research IRYCIS, 28034, Madrid, Spain.
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research IRYCIS, 28034, Madrid, Spain
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcalá de Henares, Spain.
- Ramón y Cajal Institute of Sanitary Research IRYCIS, 28034, Madrid, Spain.
| | - Raul Diaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research IRYCIS, 28034, Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
| | - Laura Lopez-Gonzalez
- Ramón y Cajal Institute of Sanitary Research IRYCIS, 28034, Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
| | - Jorge Monserrat
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research IRYCIS, 28034, Madrid, Spain
| | - Silvestra Barrena-Blázquez
- Ramón y Cajal Institute of Sanitary Research IRYCIS, 28034, Madrid, Spain
- Department of Nursing and Physiotherapy, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcalá de Henares, Spain
| | - Miguel Angel Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research IRYCIS, 28034, Madrid, Spain
- Department of Psychiatry and Mental Health, Hospital Universitario Infanta Leonor, 28031, Madrid, Spain
| | - Guillermo Lahera
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research IRYCIS, 28034, Madrid, Spain
- Psychiatry Service, Center for Biomedical Research in the Mental Health Network, University Hospital Príncipe de Asturias, 28806, Alcalá de Henares, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research IRYCIS, 28034, Madrid, Spain
- Immune System Diseases-Rheumatology and Internal Medicine Service, University Hospital Príncipe de Asturias, CIBEREHD, 28806, Alcalá de Henares, Spain
| |
Collapse
|
5
|
Leveque E, Battut L, Petitfils C, Valitutti S, Cenac N, Dietrich G, Espinosa E. Alternative activation of mast cells by CD4+ T helper cells. J Leukoc Biol 2024; 116:1127-1141. [PMID: 38916515 DOI: 10.1093/jleuko/qiae139] [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: 12/30/2023] [Revised: 05/16/2024] [Accepted: 06/19/2024] [Indexed: 06/26/2024] Open
Abstract
Effector CD4+ T (Teff) lymphocytes infiltrate sites of inflammation and orchestrate the immune response by instructing local leukocytes. Mast cells (MCs) are tissue sentinel cells strategically located near blood vessels and T cell-rich areas. MC/Teff cell interactions shape Teff cell responses, but in turn, Teff cell action on MCs is still poorly understood. Here, we analyzed the human MC/Teff cell interplay through both the application of RNA sequencing and functional assays. We showed that activated Teff cells induce a specific transcriptomic program in MCs including production of both inflammatory cytokines and chemokines, prostaglandin, and a FcεRI-dependent degranulation facilitation, thereby driving them toward an inflammatory phenotype. Moreover, Teff cells induce in MCs the capacity to interact with CD4+ T cells through a wide range of dedicated soluble and membrane ligands and to play the role of antigen-presenting cells.
Collapse
Affiliation(s)
- Edouard Leveque
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche U1037, Centre de Recherche en Cancérologie de Toulouse, 2 Avenue H. Curien, F-31037, France
- Université Toulouse III - Paul Sabatier, 118 route de Narbone, Toulouse F-31062, France
| | - Louise Battut
- Université Toulouse III - Paul Sabatier, 118 route de Narbone, Toulouse F-31062, France
- Institut National de la Santé et de la Recherche Médicale, U1220, Institut de Recherche en Santé Digestive, Institut National de la Recherche Agronomique, Institut National Polytechnique de Toulouse-École Nationale Vétérinaire de Toulouse, CHU Purpan place du Dr Baylac CS 60039, Toulouse F-31024, France
| | - Camille Petitfils
- Université Toulouse III - Paul Sabatier, 118 route de Narbone, Toulouse F-31062, France
- Institut National de la Santé et de la Recherche Médicale, U1220, Institut de Recherche en Santé Digestive, Institut National de la Recherche Agronomique, Institut National Polytechnique de Toulouse-École Nationale Vétérinaire de Toulouse, CHU Purpan place du Dr Baylac CS 60039, Toulouse F-31024, France
| | - Salvatore Valitutti
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche U1037, Centre de Recherche en Cancérologie de Toulouse, 2 Avenue H. Curien, F-31037, France
- Université Toulouse III - Paul Sabatier, 118 route de Narbone, Toulouse F-31062, France
- Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse, Centre Hospitalier Universitaire de Toulouse, 1 avenue Irène Joliot-Curie, Toulouse F-31059, France
| | - Nicolas Cenac
- Université Toulouse III - Paul Sabatier, 118 route de Narbone, Toulouse F-31062, France
- Institut National de la Santé et de la Recherche Médicale, U1220, Institut de Recherche en Santé Digestive, Institut National de la Recherche Agronomique, Institut National Polytechnique de Toulouse-École Nationale Vétérinaire de Toulouse, CHU Purpan place du Dr Baylac CS 60039, Toulouse F-31024, France
| | - Gilles Dietrich
- Université Toulouse III - Paul Sabatier, 118 route de Narbone, Toulouse F-31062, France
- Institut National de la Santé et de la Recherche Médicale, U1220, Institut de Recherche en Santé Digestive, Institut National de la Recherche Agronomique, Institut National Polytechnique de Toulouse-École Nationale Vétérinaire de Toulouse, CHU Purpan place du Dr Baylac CS 60039, Toulouse F-31024, France
| | - Eric Espinosa
- Université Toulouse III - Paul Sabatier, 118 route de Narbone, Toulouse F-31062, France
- Institut National de la Santé et de la Recherche Médicale, U1220, Institut de Recherche en Santé Digestive, Institut National de la Recherche Agronomique, Institut National Polytechnique de Toulouse-École Nationale Vétérinaire de Toulouse, CHU Purpan place du Dr Baylac CS 60039, Toulouse F-31024, France
| |
Collapse
|
6
|
Song HA, Jang SY, Park MJ, Kim SW, Kang CG, Lee JH, Kim HJ, Kim J, Lee JK, Chung KS, Lee KT. Immunostimulation Signaling via Toll-like Receptor 2 Activation: A Molecular Mechanism of Lactococcus lactis OTG1204 In Vitro and In Vivo. Nutrients 2024; 16:3629. [PMID: 39519462 PMCID: PMC11547582 DOI: 10.3390/nu16213629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
INTRODUCTION The immune system's defense against pathogens involves innate and adaptive responses, crucial in maintaining overall health. Immunosuppressed states render individuals more susceptible to potential diseases, indicating the need for effective strategies to bolster immune functions. OBJECTIVES Although the immunostimulatory effects of various probiotics have been studied, the specific effects and molecular mechanisms of Lactococcus lactis OTG1204 (OTG1204) remain unknown. In this study, the aim was to investigate the molecular mechanisms of OTG1204 in RAW 264.7 macrophages, the key effector cells of the innate immune system involved in host defense and inflammatory responses. Additionally, in this study, the effects of OTG1204 on cyclophosphamide (CTX)-induced immunosuppression states were investigated, thereby demonstrating its potential as an immune stimulant. METHODS To assess the macrophage activation ability and underlying mechanisms of OTG1204, RAW 264.7 cells were utilized with transfection, enzyme-linked immunosorbent assay, and quantitative real-time PCR analyses. Furthermore, to evaluate the immunostimulatory effects under immunosuppressed conditions, CTX-induced immunosuppression mice model was employed, and analyses were performed using hematoxylin and eosin staining, flow cytometry, and microbiota examination. RESULTS OTG1204 activated RAW 264.7 macrophages, leading to increased production of nitric oxide, prostaglandin E2, and cytokines. This immune activation was mediated through the upregulation of toll-like receptor 2, which subsequently activated the nuclear factor-κB (NF-kB) and mitogen-activated protein kinase (MAPK)/activator protein 1 (AP-1) pathways, thereby stimulating the immune response. In CTX-treated mice, OTG1204 recovered body weight, spleen, and mesenteric lymph node indices, and natural killer cell activity. It re-established populations of innate and adaptive immune cells and activated T cells to secrete cytokines. We also examined the gut barrier integrity and microbiota composition to assess OTG1204's impact on intestinal health, as these factors play a significant role in immune enhancement. OTG1204 enhanced gut barrier integrity by upregulating mucin 2 and tight junction proteins and modulated the gut microbiota by restoring the Firmicutes/Bacteroidetes balance and reducing the abundance of Actinobacteria and Tenericutes. CONCLUSION These results suggest that OTG1204 may serve as an effective probiotic for immune enhancement and gut health management by targeting the NF-κB and MAPK/AP-1 pathways, with minimal side effects.
Collapse
Affiliation(s)
- Hyeon-A Song
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (H.-A.S.); (S.-Y.J.); (M.-J.P.); (K.-S.C.)
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Seo-Yun Jang
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (H.-A.S.); (S.-Y.J.); (M.-J.P.); (K.-S.C.)
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Min-Ji Park
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (H.-A.S.); (S.-Y.J.); (M.-J.P.); (K.-S.C.)
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Seung Wook Kim
- Ottogi Research Center, Anyang 14060, Republic of Korea; (S.W.K.); (C.G.K.); (J.H.L.); (H.-J.K.); (J.K.)
| | - Choon Gil Kang
- Ottogi Research Center, Anyang 14060, Republic of Korea; (S.W.K.); (C.G.K.); (J.H.L.); (H.-J.K.); (J.K.)
| | - Joo Hyun Lee
- Ottogi Research Center, Anyang 14060, Republic of Korea; (S.W.K.); (C.G.K.); (J.H.L.); (H.-J.K.); (J.K.)
| | - Hye-Jin Kim
- Ottogi Research Center, Anyang 14060, Republic of Korea; (S.W.K.); (C.G.K.); (J.H.L.); (H.-J.K.); (J.K.)
| | - Jiheon Kim
- Ottogi Research Center, Anyang 14060, Republic of Korea; (S.W.K.); (C.G.K.); (J.H.L.); (H.-J.K.); (J.K.)
| | - Jong Kil Lee
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Kyung-Sook Chung
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (H.-A.S.); (S.-Y.J.); (M.-J.P.); (K.-S.C.)
| | - Kyung-Tae Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (H.-A.S.); (S.-Y.J.); (M.-J.P.); (K.-S.C.)
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| |
Collapse
|
7
|
Kang ZP, Xiao QP, Huang JQ, Wang MX, Huang J, Wei SY, Cheng N, Wang HY, Liu DY, Zhong YB, Zhao HM. Curcumin Attenuates Dextran Sodium Sulfate Induced Colitis in Obese Mice. Mol Nutr Food Res 2024; 68:e2300598. [PMID: 39380356 DOI: 10.1002/mnfr.202300598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 06/01/2024] [Indexed: 10/10/2024]
Abstract
SCOPE Curcumin (Cur), with diverse pharmacological properties, shows anti-obesity, immunomodulatory, and anti-inflammatory effects. Its role in ulcerative colitis complicated by obesity remains unclear. METHODS AND RESULTS Here, colitis is induced in obese mice using dextran sulfate sodium (DSS), followed by administration of Cur at a dosage of 100 mg kg-1 for 14 days. Cur effectively alleviates DSS-induced colitis in obese mice, accompanied by an increase in body weight and survival rate, reduction in disease activity index, elongation of the colon, decrease in colonic weight, and improvements in ulcer formation and inflammatory cell infiltration in colonic tissues. Additionally, Cur effectively improves lipid metabolism and the composition of the gut microbiota, and enhances mucosal integrity and boosts anti-oxidative stress capacity in obese mice with colitis. Importantly, Cur is effective in improving the homeostasis of memory T cells in obese mice with colitis. Furthermore, Cur regulates inflammatory cytokines expression and inhibits activation of the JAK2/STAT signaling pathway in colonic tissues of obese mice with colitis. CONCLUSIONS Cur alleviates colitis in obese mice through a comprehensive mechanism that improves lipid metabolism, modulates gut microbiota composition, enhances mucosal integrity and anti-oxidative stress, balances memory T cell populations, regulates inflammatory cytokines, and suppresses the JAK2/STAT signaling pathway.
Collapse
Affiliation(s)
- Zeng-Ping Kang
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| | - Qiu-Ping Xiao
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| | - Jia-Qi Huang
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| | - Meng-Xue Wang
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| | - Jie Huang
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| | - Si-Yi Wei
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| | - Nian Cheng
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| | - Hai-Yan Wang
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
- Formula-Pattern Research Center of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| | - Duan-Yong Liu
- Formula-Pattern Research Center of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| | - You-Bao Zhong
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
- Formula-Pattern Research Center of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| | - Hai-Mei Zhao
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
- Formula-Pattern Research Center of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| |
Collapse
|
8
|
Assefa M, Girmay G. Mycobacterium tuberculosis Biofilms: Immune Responses, Role in TB Pathology, and Potential Treatment. Immunotargets Ther 2024; 13:335-342. [PMID: 38974843 PMCID: PMC11227863 DOI: 10.2147/itt.s455744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 06/28/2024] [Indexed: 07/09/2024] Open
Abstract
Tuberculosis (TB) is a major public health problem worldwide, and the burden of drug-resistant TB is rapidly increasing. Although there are literatures about the Mtb biofilms, their impact on immune responses has not yet been summarized. This review article provides recent knowledge on Mycobacterium tuberculosis (Mtb) biofilm-immunity interactions, their importance in pulmonary TB pathology, and immune-based therapy targeting Mtb biofilms. Pellicle/biofilm formation in Mtb contributes to drug resistance, persistence, chronicity, surface attachment, transfer of resistance genes, and modulation of the immune response, including reduced complement activation, changes in the expression of antigenic proteins, enhanced activation of T-lymphocytes, elevated local IFNγ+ T cells, and strong antibody production. The combination of anti-TB drugs and anti-biofilm agents has recently become an effective strategy to improve TB treatment. Additionally, immune-targeted therapy and biofilm-based vaccines are crucial for TB prevention.
Collapse
Affiliation(s)
- Muluneh Assefa
- Department of Medical Microbiology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Getu Girmay
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
9
|
Morrison AI, Sjoerds MJ, Vonk LA, Gibbs S, Koning JJ. In vitro immunity: an overview of immunocompetent organ-on-chip models. Front Immunol 2024; 15:1373186. [PMID: 38835750 PMCID: PMC11148285 DOI: 10.3389/fimmu.2024.1373186] [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: 01/19/2024] [Accepted: 04/30/2024] [Indexed: 06/06/2024] Open
Abstract
Impressive advances have been made to replicate human physiology in vitro over the last few years due to the growth of the organ-on-chip (OoC) field in both industrial and academic settings. OoCs are a type of microphysiological system (MPS) that imitates functional and dynamic aspects of native human organ biology on a microfluidic device. Organoids and organotypic models, ranging in their complexity from simple single-cell to complex multi-cell type constructs, are being incorporated into OoC microfluidic devices to better mimic human physiology. OoC technology has now progressed to the stage at which it has received official recognition by the Food and Drug Administration (FDA) for use as an alternative to standard procedures in drug development, such as animal studies and traditional in vitro assays. However, an area that is still lagging behind is the incorporation of the immune system, which is a critical element required to investigate human health and disease. In this review, we summarise the progress made to integrate human immunology into various OoC systems, specifically focusing on models related to organ barriers and lymphoid organs. These models utilise microfluidic devices that are either commercially available or custom-made. This review explores the difference between the use of innate and adaptive immune cells and their role for modelling organ-specific diseases in OoCs. Immunocompetent multi-OoC models are also highlighted and the extent to which they recapitulate systemic physiology is discussed. Together, the aim of this review is to describe the current state of immune-OoCs, the limitations and the future perspectives needed to improve the field.
Collapse
Affiliation(s)
- Andrew I. Morrison
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
| | - Mirthe J. Sjoerds
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Leander A. Vonk
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Susan Gibbs
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, Netherlands
| | - Jasper J. Koning
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
| |
Collapse
|
10
|
Hussen J, AL-Jabr OA, Alkuwayti MA, Alrabiah NA, Falemban B, Alouffi A, Al Salim WS, Kamyingkird K, Desquesnes M. A Flow Cytometry Study of the Binding and Stimulation Potential of Inactivated Trypanosoma evansi toward Dromedary Camel Leukocytes. Pathogens 2023; 13:21. [PMID: 38251329 PMCID: PMC10820945 DOI: 10.3390/pathogens13010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 01/23/2024] Open
Abstract
Surra, a wasting disease caused by Trypanosoma evansi, is one of the major animal health burdens in camel-rearing countries, imposing significant economic losses due to reduced fertility and high mortality rates. The present study used inactivated T. evansi (from the Card Agglutination Test for Trypanosomes/Trypanosoma evansi; CATT/T. evansi) and flow cytometry to investigate their binding and activation potential toward camel leukocyte subsets. Labeling T. evansi with propidium iodide (PI) enabled their flow cytometric enumeration and identification with forward scatter (FSC; indicative for cell size) and side scatter (SSC; indicative for cell internal complexity) characteristics that are comparable with values reported for Trypanosoma cruzi. The incubation of PI-labeled non-opsonized T. evansi with camel leukocyte populations revealed that camel monocytes have the highest potential to bind T. evansi, followed by granulocytes and lymphocytes. The identification of pattern recognition receptors (PRRs) on camel immune cells and the pathogen-associated molecular patterns (PAMPs) in T. evansi that are responsible for this different binding capacity requires further studies. Stimulation of camel neutrophils with Trypanosoma evansi induced shape change, reactive oxygen species (ROS) production, and neutrophil extracellular traps (NET)-formation. To ensure that T. evansi, in the parasite concentration used in this study, is not apoptotic or necrotic to camel leukocytes, we evaluated cell apoptosis and necrosis after stimulation with T. evansi. The results revealed no impact of T. evansi stimulation for 2 h on the cell viability of camel leukocytes. Subsequent work may focus on the diagnostic employment of labeled T. evansi and flow cytometry for the detection of anti-Trypanosoma antibodies in camel serum. In addition, more efforts should be deployed to investigate the host-pathogen interaction mechanisms and the escape mechanisms of T. evansi in camels. To complete these data, further studies using the living or freshly killed parasites could also be implemented in camels and/or horses.
Collapse
Affiliation(s)
- Jamal Hussen
- Department of Microbiology, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (O.A.A.-J.); (B.F.)
- Central Veterinary Laboratory, Ministry of Environment, Water and Agriculture, Riyadh 11195, Saudi Arabia; (A.A.); (W.S.A.S.)
| | - Omar A. AL-Jabr
- Department of Microbiology, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (O.A.A.-J.); (B.F.)
| | - Mayyadah Abdullah Alkuwayti
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (M.A.A.); (N.A.A.)
| | - Noof Abdulrahman Alrabiah
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (M.A.A.); (N.A.A.)
| | - Baraa Falemban
- Department of Microbiology, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (O.A.A.-J.); (B.F.)
| | - Abdulaziz Alouffi
- Central Veterinary Laboratory, Ministry of Environment, Water and Agriculture, Riyadh 11195, Saudi Arabia; (A.A.); (W.S.A.S.)
- King Abdulaziz City for Science and Technology, Riyadh 12354, Saudi Arabia
| | - Waleed S. Al Salim
- Central Veterinary Laboratory, Ministry of Environment, Water and Agriculture, Riyadh 11195, Saudi Arabia; (A.A.); (W.S.A.S.)
- Ministry of Environment, Water and Agriculture, Riyadh 11195, Saudi Arabia
| | - Ketsarin Kamyingkird
- Department of Parasitology, Faculty of Veterinary Medicine, Kasetsart University, Ladyao, Chatuchak, Bangkok 10900, Thailand;
| | - Marc Desquesnes
- CIRAD, UMR INTERTRYP, Ecole Nationale Vétérinaire de Toulouse (ENVT), 31300 Toulouse, France;
- Interactions Hosts-Vectors-Parasites-Environment in the Tropical Neglected Disease due to Trypanosoma-Tids (INTERTRYP), University Montpellier, 34398 Montpellier, France
| |
Collapse
|
11
|
Sawada K, Chung H, Softic S, Moreno-Fernandez ME, Divanovic S. The bidirectional immune crosstalk in metabolic dysfunction-associated steatotic liver disease. Cell Metab 2023; 35:1852-1871. [PMID: 37939656 PMCID: PMC10680147 DOI: 10.1016/j.cmet.2023.10.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is an unabated risk factor for end-stage liver diseases with no available therapies. Dysregulated immune responses are critical culprits of MASLD pathogenesis. Independent contributions from either the innate or adaptive arms of the immune system or their unidirectional interplay are commonly studied in MASLD. However, the bidirectional communication between innate and adaptive immune systems and its impact on MASLD remain insufficiently understood. Given that both innate and adaptive immune cells are indispensable for the development and progression of inflammation in MASLD, elucidating pathogenic contributions stemming from the bidirectional interplay between these two arms holds potential for development of novel therapeutics for MASLD. Here, we review the immune cell types and bidirectional pathways that influence the pathogenesis of MASLD and highlight potential pharmacologic approaches to combat MASLD based on current knowledge of this bidirectional crosstalk.
Collapse
Affiliation(s)
- Keisuke Sawada
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - Hak Chung
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Samir Softic
- Department of Pediatrics and Gastroenterology, University of Kentucky, Lexington, KY 40536, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Maria E Moreno-Fernandez
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
12
|
Liu X, Vigorito M, Huang W, Khan MAS, Chang SL. The Impact of Alcohol-Induced Dysbiosis on Diseases and Disorders of the Central Nervous System. J Neuroimmune Pharmacol 2022; 17:131-151. [PMID: 34843074 DOI: 10.1007/s11481-021-10033-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/11/2021] [Indexed: 12/29/2022]
Abstract
The human digestive tract contains a diverse and abundant microbiota that is important for health. Excessive alcohol use can disrupt the balance of these microbes (known as dysbiosis), leading to elevated blood endotoxin levels and systemic inflammation. Using QIAGEN Ingenuity Pathway Analysis (IPA) bioinformatics tool, we have confirmed that peripheral endotoxin (lipopolysaccharide) mediates various cytokines to enhance the neuroinflammation signaling pathway. The literature has identified alcohol-mediated neuroinflammation as a possible risk factor for the onset and progression of neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD), and psychiatric disorders such as addiction to alcohol and other drugs. In this review, we discuss alcohol-use-induced dysbiosis in the gut and other body parts as a causal factor in the progression of Central Nervous System (CNS) diseases including neurodegenerative disease and possibly alcohol use disorder.
Collapse
Affiliation(s)
- Xiangqian Liu
- Institute of Neuroimmune Pharmacology, Seton Hall University, South Orange, NJ, 07079, USA
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, P.R. China
| | - Michael Vigorito
- Institute of Neuroimmune Pharmacology, Seton Hall University, South Orange, NJ, 07079, USA
- Department of Psychology, Seton Hall University, South Orange, NJ, 07079, USA
| | - Wenfei Huang
- Institute of Neuroimmune Pharmacology, Seton Hall University, South Orange, NJ, 07079, USA
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, 07079, USA
| | - Mohammed A S Khan
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, Boston, MA, 02114, USA.
| | - Sulie L Chang
- Institute of Neuroimmune Pharmacology, Seton Hall University, South Orange, NJ, 07079, USA.
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, 07079, USA.
| |
Collapse
|
13
|
Liang W, Meng X, Zhen Y, Zhang Y, Hu X, Zhang Q, Zhou X, Liu B. Integration of Transcriptome and Proteome in Lymph Nodes Reveal the Different Immune Responses to PRRSV Between PRRSV-Resistant Tongcheng Pigs and PRRSV-Susceptible Large White Pigs. Front Genet 2022; 13:800178. [PMID: 35154273 PMCID: PMC8829461 DOI: 10.3389/fgene.2022.800178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/07/2022] [Indexed: 11/13/2022] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is an infectious disease that seriously affects the swine industry worldwide. Understanding the interaction between the host immune response and PRRS virus (PRRSV) can provide insight into the PRRSV pathogenesis, as well as potential clues to control PRRSV infection. Here, we examined the transcriptome and proteome differences of lymph nodes between PRRSV-resistant Tongcheng (TC) pigs and PRRSV-susceptible Large White (LW) pigs in response to PRRSV infection. 2245 and 1839 differentially expressed genes (DEGs) were detected in TC and LW pigs upon PRRSV infection, respectively. Transcriptome analysis revealed genetic differences in antigen presentation and metabolism between TC pigs and LW pigs, which may lead to different immune responses to PRRSV infection. Furthermore, 678 and 1000 differentially expressed proteins (DEPs) were identified in TC and LW pigs, and DEPs were mainly enriched in the metabolism pathways. Integrated analysis of transcriptome and proteome datasets revealed antigen recognition capacity, immune activation, cell cycles, and cell metabolism are important for PRRSV clearance. In conclusion, this study provides important resources on transcriptomic and proteomic levels in lymph nodes for further revealing the interaction between the host immune response and PRRSV, which would give us new insight into molecular mechanisms related to genetic complexity against PRRSV.
Collapse
Affiliation(s)
- Wan Liang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Animal Husbandry and Veterinary Institute, Hubei Academy of Agricultural Science, Wuhan, China
| | - Xiangge Meng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yueran Zhen
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yu Zhang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xueying Hu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qingde Zhang
- Laboratory Animal Center, College of Animal Science and Technology and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiang Zhou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Bang Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
14
|
Deets KA, Nichols Doyle R, Rauch I, Vance RE. Inflammasome activation leads to cDC1-independent cross-priming of CD8 T cells by epithelial cell-derived antigen. eLife 2021; 10:e72082. [PMID: 34939932 PMCID: PMC8719880 DOI: 10.7554/elife.72082] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 12/21/2021] [Indexed: 12/17/2022] Open
Abstract
The innate immune system detects pathogens and initiates adaptive immune responses. Inflammasomes are central components of the innate immune system, but whether inflammasomes provide sufficient signals to activate adaptive immunity is unclear. In intestinal epithelial cells (IECs), inflammasomes activate a lytic form of cell death called pyroptosis, leading to epithelial cell expulsion and the release of cytokines. Here, we employed a genetic system to show that simultaneous antigen expression and inflammasome activation specifically in IECs is sufficient to activate CD8+ T cells. By genetic elimination of direct T cell priming by IECs, we found that IEC-derived antigens were cross-presented to CD8+ T cells. However, cross-presentation of IEC-derived antigen to CD8+ T cells only partially depended on IEC pyroptosis. In the absence of inflammasome activation, cross-priming of CD8+ T cells required Batf3+ dendritic cells (conventional type one dendritic cells [cDC1]), whereas cross-priming in the presence of inflammasome activation required a Zbtb46+ but Batf3-independent cDC population. These data suggest the existence of parallel inflammasome-dependent and inflammasome-independent pathways for cross-presentation of IEC-derived antigens.
Collapse
Affiliation(s)
- Katherine A Deets
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Randilea Nichols Doyle
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Isabella Rauch
- Department of Molecular Microbiology and Immunology, Oregon Health and Science UniversityPortlandUnited States
| | - Russell E Vance
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
- Cancer Research Laboratory, University of California, BerkeleyBerkeleyUnited States
- Howard Hughes Medical Institute, University of California, BerkeleyBerkeleyUnited States
| |
Collapse
|
15
|
Abstract
Elderly individuals with chronic disorders tend to develop inflammaging, a condition associated with elevated levels of blood inflammatory markers, and increased susceptibility to chronic disease progression. Native and adaptive immunity are both involved in immune system senescence, kidney fibrosis and aging. The innate immune system is characterized by a limited number of receptors, constantly challenged by self and non-self stimuli. Circulating and kidney resident myeloid and lymphoid cells are all equipped with pattern recognition receptors (PRRs). Recent reports on PRRs show kidney overexpression of toll-like receptors (TLRs) in inflammaging autoimmune renal diseases, vasculitis, acute kidney injury and kidney transplant rejection. TLR upregulation leads to proinflammatory cytokine induction, fibrosis, and chronic kidney disease progression. TLR2 blockade in a murine model of renal ischemia reperfusion injury prevented the escape of natural killer cells and neutrophils by inflammaging kidney injury. Tumor necrosis factor-α blockade in endothelial cells with senescence-associated secretory phenotype significantly reduced interleukin-6 release. These findings should encourage experimental and translational clinical trials aimed at modulating renal inflammaging by native immunity blockade.
Collapse
|