1
|
Bahl A, Pandey S, Rakshit R, Kant S, Tripathi D. Infection-induced trained immunity: a twist in paradigm of innate host defense and generation of immunological memory. Infect Immun 2025; 93:e0047224. [PMID: 39655962 PMCID: PMC11784091 DOI: 10.1128/iai.00472-24] [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: 02/01/2025] Open
Abstract
In contrast to adaptive immunity, which relies on memory T and B cells for long-term pathogen-specific responses, trained immunity involves the enhancement of innate immune responses through cellular reprogramming. Experimental evidence from animal models and human studies supports the concept of trained immunity and its potential therapeutic applications in the development of personalized medicine. However, there remains a huge gap in understanding the mechanisms, identifying specific microbial triggers responsible for the induction of trained immunity. This underscores the importance of investigating the potential role of trained immunity in redefining host defense and highlights future research directions. This minireview will provide a comprehensive summary of the new paradigm of trained immunity or innate memory pathways. It will shed light on infection-induced pathways through non-specific stimulation within macrophages and natural killer cells, which will be further elaborated in multiple disease perspectives caused by infectious agents such as bacteria, fungi, and viruses. The article further elaborates on the biochemical and cellular basis of trained immunity and its impact on disease status during recurrent exposures. The review concludes with a perspective segment discussing potential therapeutic benefits, limitations, and future challenges in this area of study. The review also sheds light upon potential risks involved in the induction of trained immunity.
Collapse
Affiliation(s)
- Aayush Bahl
- Microbial Pathogenesis and Microbiome Lab, Department of Microbiology, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Saurabh Pandey
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, Delhi, India
| | - Roopshali Rakshit
- Microbial Pathogenesis and Microbiome Lab, Department of Microbiology, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Sashi Kant
- Bacterial Pathogenesis, Boehringer Ingelheim Animal Health USA Inc, Ames, Iowa, USA
| | - Deeksha Tripathi
- Microbial Pathogenesis and Microbiome Lab, Department of Microbiology, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| |
Collapse
|
2
|
Song X, Lei T, Cui N, Jin X, Huang Y, Shi Y, Zhao Z. A preliminary investigation on the protective effects of β-glucan and mannan induced trained immunity in pufferfish Takifugu obscurus. FISH & SHELLFISH IMMUNOLOGY 2025; 156:110035. [PMID: 39577788 DOI: 10.1016/j.fsi.2024.110035] [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: 08/25/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
Immune stimuli are able to trigger long-term protective effects through mechanisms of trained immunity, which has attracted increasing attention. Although the existence of trained immunity has evidenced in teleost fish, while there were no such reports in pufferfish (Takifugu obscurus) so far. Therefore, the present study aimed to evaluate the induction of β-glucan and mannan on the trained immunity and their protective efficacy against Vibrio harveyi re-stimulation in pufferfish. β-glucan and mannan induction of trained immunity in head-kidney primary leukocytes is accompanied by a strong increase in immediate ROS burst, cumulative NO production and lactate concentrations after V. harveyi re-stimulation. In addition, β-glucan and mannan-treated pufferfish exhibited reduced bacterial loads in multiple tissues, a rapid and long-term elevated inflammatory response in head kidney during secondary V. harveyi infection. Notably, immune receptors dectin-1 and dectin-2, and cytokines tnfsf14 and il-1β exhibited comparatively upregulation to the β-glucan training, while NK-lysin and faslg showed stronger response to the mannan training post V. harveyi stimulation, implying the different signaling pathway activated post β-glucan and mannan training. Subsequent markers for immune training including abundance of genes encoding glycolytic enzymes (hk1, pfkla, and ldha) and transcription factors (mtor and hif-1α), as well as increased acetylation levels were elevated in the β-glucan and mannan trained pufferfish, depicting heightened glycolysis following β-glucan and mannan training. These results collectively demonstrated that β-glucan and mannan both induced protective responses against V. harveyi infection probably through mediating distinct signaling pathway in pufferfish, and studies are underway to harness its potential applicability for prime and boost vaccination strategies.
Collapse
Affiliation(s)
- Xiaorui Song
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing, 210024, China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Tianying Lei
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing, 210024, China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Nan Cui
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing, 210024, China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Xingkun Jin
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing, 210024, China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Ying Huang
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing, 210024, China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Yan Shi
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing, 210024, China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Zhe Zhao
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing, 210024, China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China.
| |
Collapse
|
3
|
Di Pietrantonio N, Sánchez-Ceinos J, Shumliakivska M, Rakow A, Mandatori D, Di Tomo P, Formoso G, Bonfini T, Baldassarre MPA, Sennström M, Almahmeed W, Pandolfi A, Cosentino F. The inflammatory and oxidative phenotype of gestational diabetes is epigenetically transmitted to the offspring: role of methyltransferase MLL1-induced H3K4me3. Eur Heart J 2024; 45:5171-5185. [PMID: 39471481 DOI: 10.1093/eurheartj/ehae688] [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: 02/08/2024] [Revised: 06/16/2024] [Accepted: 09/25/2024] [Indexed: 11/01/2024] Open
Abstract
BACKGROUND AND AIMS Hyperglycaemia during gestational diabetes (GD) predisposes women and their offspring to later cardiometabolic disease. The hyperglycaemia-mediated epigenetic changes remain to be elucidated. Methyltransferase MLL1-induced trimethylation of histone 3 at lysine 4 (H3K4me3) activates inflammatory and oxidative phenotype. This epigenetic mark in GD women and its transmission to the offspring were investigated. METHODS Peripheral blood mononuclear cells (PBMC) were collected from GD and control (C) women and also from adolescents born to women of both groups. Endothelial human umbilical vein endothelial cells (HUVEC) and cord blood mononuclear cells (CBMC) were from umbilical cords. The NF-κBp65 and NOX4 expressions were investigated by reverse transcription quantitative polymerase chain reaction and immunofluorescence (IF). MLL1 and H3K4me3 were investigated by immunoblotting and IF. H3K4me3 on NF-κBp65 and NOX4 promoters was studied by chromatin immunoprecipitation. Superoxide anion generation was measured by electron spin resonance spectroscopy. Plasma cytokines were measured by enzyme-linked immunosorbent assay. To investigate the role of MLL1, HUVEC were exposed to inhibitor MM102 or siRNA transfection. RESULTS PBMC, CBMC, and HUVEC showed an increase of NF-κBp65, IL-6, ICAM-1, MCP-1, and VCAM-1 mRNAs. These findings were associated with H3K4me3 enrichment in the promoter of NF-κBp65. Elevated H3K4me3 and cytokine levels were observed in GD adolescents. MLL1 drives H3K4me3 not only on NF-kB p65, but also on NOX4 promoter. Inhibition of MLL1 blunted NF-κBp65 and NOX4 by modulating inflammatory and oxidative phenotype. CONCLUSIONS Such proof-of-concept study shows persistence of MLL1-dependent H3K4me3 in offspring born to GD women, suggesting an epigenetic-driven transmission of maternal phenotype. These findings may pave the way for pharmacological reprogramming of adverse histone modifications to mitigate abnormal phenotypes underlying early ASCVD.
Collapse
Affiliation(s)
- Nadia Di Pietrantonio
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm 171 76, Sweden
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology-CAST, University G. D'Annunzio of Chieti-Pescara, Chieti 66100, Italy
| | - Julia Sánchez-Ceinos
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm 171 76, Sweden
| | - Mariana Shumliakivska
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm 171 76, Sweden
| | - Alexander Rakow
- Department of Neonatology, Karolinska University Hospital, Stockholm, Sweden
| | - Domitilla Mandatori
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology-CAST, University G. D'Annunzio of Chieti-Pescara, Chieti 66100, Italy
| | - Pamela Di Tomo
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology-CAST, University G. D'Annunzio of Chieti-Pescara, Chieti 66100, Italy
| | - Gloria Formoso
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology-CAST, University G. D'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Tiziana Bonfini
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Maria Pompea Antonia Baldassarre
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology-CAST, University G. D'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Maria Sennström
- Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Wael Almahmeed
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE
| | - Assunta Pandolfi
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology-CAST, University G. D'Annunzio of Chieti-Pescara, Chieti 66100, Italy
| | - Francesco Cosentino
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm 171 76, Sweden
- Heart and Vascular Theme, Karolinska University Hospital, Stockholm 171 76, Sweden
| |
Collapse
|
4
|
Netea MG, Joosten LAB. Trained innate immunity: Concept, nomenclature, and future perspectives. J Allergy Clin Immunol 2024; 154:1079-1084. [PMID: 39278362 DOI: 10.1016/j.jaci.2024.09.005] [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: 07/08/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/18/2024]
Abstract
During the past decade, compelling evidence has accumulated demonstrating that innate immune cells can mount adaptive characteristics, leading to long-term changes in their function. This de facto innate immune memory has been termed trained immunity. Trained immunity, which is mediated through extensive metabolic rewiring and epigenetic modifications, has important effects in human diseases. Although the upregulation of trained immunity by certain vaccines provides heterologous protection against infections, the inappropriate activation of trained immunity by endogenous stimuli contributes to the pathogenesis of inflammatory and neurodegenerative disorders. Development of vaccines that can induce both classical adaptive immunity and trained immunity may lead to a new generation of vaccines with increased efficacy. Activation of trained immunity can also lead to novel strategies for the treatment of cancer, whereas modulation of trained immunity can provide new approaches to the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands; Department for Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany.
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
5
|
Schaefer Z, Iradukunda J, Lumngwena EN, Basso KB, Blackburn JM, Parker IK. Multilevel Proteomics Reveals Epigenetic Signatures in BCG-Mediated Macrophage Activation. Mol Cell Proteomics 2024; 23:100851. [PMID: 39366656 PMCID: PMC11585779 DOI: 10.1016/j.mcpro.2024.100851] [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: 04/08/2024] [Revised: 09/17/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024] Open
Abstract
The bacillus Calmette-Guérin BCG vaccine (Mycobacterium bovis) is primarily used to prevent tuberculosis (TB) infections but has wide-ranging immunogenic effects. One of its most notable properties is its ability to induce trained immunity, a memory-like response in innate immune cells such as macrophages. Through targeted analyses of well-established histone marks, prior research has shown that these changes are generated through epigenetic modification. Mass spectrometry-based proteomic approaches provide a way to globally profile various aspects of the proteome, providing data to further identify unexplored mechanisms of BCG-mediated immunomodulation. Here we use multi-level proteomics (total, histone, and phospho to identify networks and potential mechanisms that mediate BCG-induced immunomodulation in macrophages. Histone-focused proteomics and total proteomics were performed at the University of Cape Town (data available via ProteomeXchange with identifier PXD051187), while phosphoproteomics data was retrieved from the ProteomeXchange Repository (identifier PXD013171). We identify several epigenetic mechanisms that may drive BCG-induced training phenotypes. Evidence across the proteomics and histone-focused proteomics data set pair 6 epigenetic effectors (NuA4, NuRD, NSL, Sin3A, SIRT2, SIRT6) and their substrates.
Collapse
Affiliation(s)
- Zoe Schaefer
- Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - John Iradukunda
- Division of Chemical & Systems Biology, University of Cape Town, Cape Town, South Africa
| | - Evelyn N Lumngwena
- School of Clinical Medicine, University of The Witwatersrand, Johannesburg, South Africa; Center for the Study of Emerging and Re-emerging Infections (CREMER), Institute for Medical Research and Medicinal Plant Studies (IMPM), Ministry of Scientific Research and Innovation, Yaounde, Cameroon
| | - Kari B Basso
- Mass Spectrometry Research and Education Center, Department of Chemistry, University of Florida, Gainesville, Florida, USA
| | - Jonathan M Blackburn
- Division of Chemical & Systems Biology, University of Cape Town, Cape Town, South Africa
| | - Ivana K Parker
- Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA.
| |
Collapse
|
6
|
Park HJ, Kim SM, Choi UY, Kim LK. Multifaceted roles of trained immunity in diverse pathological contexts. BMB Rep 2024; 57:431-440. [PMID: 38835118 PMCID: PMC11524827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/23/2024] [Accepted: 05/13/2024] [Indexed: 06/06/2024] Open
Abstract
Trained immunity, an innate immune response characterized by enhanced cellular responsiveness, exhibits a profound memory akin to adaptive immunity. This phenomenon involves intricate metabolic and epigenetic reprogramming triggered by stimuli such as β-glucan and BCG, shaping innate immune memory. Following elucidation of the background on trained immunity, it is important to explore its multifaceted roles in various pathological contexts. In this review, we delve into the specific contributions of trained immunity in the intricate landscape of viral infections, tumorigenesis, and diverse inflammatory diseases, shedding light on its potential as a therapeutic target, and offering comprehensive understanding of its broader immunological implications. [BMB Reports 2024; 57(10): 431-440].
Collapse
Affiliation(s)
- Hyo Jin Park
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06230, Korea
| | - Su Min Kim
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06230, Korea
| | - Un Yung Choi
- Department of Microbiology, Konkuk University School of Medicine, Chungju 27478, Korea
- KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Korea
| | - Lark Kyun Kim
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06230, Korea
| |
Collapse
|
7
|
Vuscan P, Kischkel B, Joosten LAB, Netea MG. Trained immunity: General and emerging concepts. Immunol Rev 2024; 323:164-185. [PMID: 38551324 DOI: 10.1111/imr.13326] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 03/11/2024] [Indexed: 05/18/2024]
Abstract
Over the past decade, compelling evidence has unveiled previously overlooked adaptive characteristics of innate immune cells. Beyond their traditional role in providing short, non-specific protection against pathogens, innate immune cells can acquire antigen-agnostic memory, exhibiting increased responsiveness to secondary stimulation. This long-term de-facto innate immune memory, also termed trained immunity, is mediated through extensive metabolic rewiring and epigenetic modifications. While the upregulation of trained immunity proves advantageous in countering immune paralysis, its overactivation contributes to the pathogenesis of autoinflammatory and autoimmune disorders. In this review, we present the latest advancements in the field of innate immune memory followed by a description of the fundamental mechanisms underpinning trained immunity generation and different cell types that mediate it. Furthermore, we explore its implications for various diseases and examine current limitations and its potential therapeutic targeting in immune-related disorders.
Collapse
Affiliation(s)
- Patricia Vuscan
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Brenda Kischkel
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| |
Collapse
|
8
|
Lian X, Li Y, Wang W, Zuo J, Yu T, Wang L, Song L. The Modification of H3K4me3 Enhanced the Expression of CgTLR3 in Hemocytes to Increase CgIL17-1 Production in the Immune Priming of Crassostrea gigas. Int J Mol Sci 2024; 25:1036. [PMID: 38256110 PMCID: PMC10816183 DOI: 10.3390/ijms25021036] [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: 12/05/2023] [Revised: 01/04/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Increasing evidence confirms that histone modification plays a critical role in preserving long-term immunological memory. Immune priming is a novel form of immunological memory recently verified in invertebrates. Toll-like receptor (TLR) signaling and cytokines have been reported to be involved in the immune priming of the Pacific oyster Crassostrea gigas. In the present study, the expression of Toll-like receptor 3 (CgTLR3), myeloid differentiation factor 88-2 (CgMyd88-2) and interleukin 17-1 (CgIL17-1) was found to be elevated in the hemocytes of C. gigas at 6 h after the secondary stimulation with Vibrio splendidus, which was significantly higher than that at 6 h after the primary stimulation (p < 0.05). A significant increase in histone H3 lysine 4 trimethylation (H3K4me3) enrichment was detected in the promoter region of the CgTLR3 gene at 7 d after the primary stimulation with inactivated V. splendidus (p < 0.05). After the treatment with a histone methyltransferase inhibitor (5'-methylthioadenosine, MTA), the level of H3K4me3 at the promoter of the CgTLR3 gene decreased significantly at 7 d after the primary stimulation with inactivated V. splendidus (p < 0.05), and the expression of CgTLR3, CgMyD88-2 and CgIL17-1 was significantly repressed at 6 h after the secondary stimulation with V. splendidus (p < 0.05). Conversely, the treatment with monomethyl fumarate (MEF, an inhibitor of histone demethylases) resulted in a significant increase in H3K4me3 enrichment levels at the CgTLR3 promoter at 7 d after the primary stimulation (p < 0.05), and the expression of CgTLR3, CgMyD88-2 and CgIL17-1 was observed to increase significantly at 6 h after the secondary stimulation (p < 0.05). These results suggested that H3K4me3 regulated MyD88-dependent TLR signaling in the hemocytes of C. gigas, which defined the role of histone modifications in invertebrate immune priming.
Collapse
Affiliation(s)
- Xingye Lian
- School of Life Science, Liaoning Normal University, Dalian 116029, China; (X.L.); (Y.L.)
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (W.W.); (J.Z.); (T.Y.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Yinan Li
- School of Life Science, Liaoning Normal University, Dalian 116029, China; (X.L.); (Y.L.)
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (W.W.); (J.Z.); (T.Y.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Weilin Wang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (W.W.); (J.Z.); (T.Y.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Jiajun Zuo
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (W.W.); (J.Z.); (T.Y.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Tianqi Yu
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (W.W.); (J.Z.); (T.Y.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (W.W.); (J.Z.); (T.Y.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (W.W.); (J.Z.); (T.Y.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| |
Collapse
|
9
|
Wang Z, You X, Zhang Y, Liu Q, Yang D. Poly(I:C) induces anti-inflammatory response against secondary LPS challenge in zebrafish larvae. FISH & SHELLFISH IMMUNOLOGY 2024; 144:109285. [PMID: 38092095 DOI: 10.1016/j.fsi.2023.109285] [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: 10/17/2023] [Revised: 11/23/2023] [Accepted: 12/07/2023] [Indexed: 12/17/2023]
Abstract
Poly(I:C) is known as an agonist of the TLR3 receptor which could prime inflammation and elicit the host immune response, which is widely applied as adjuvant or antivirus treatment. However, the negative effects of poly(I:C) on regulating immune response to protect the host from inflammatory diseases remain largely unknown. Here, we establish an in vivo model to pre-treat zebrafish larvae with poly(I:C) at 2 dpf, then challenge them with LPS at 6 dpf, and find that poly(I:C) training could significantly alleviate the LPS challenge-induced septic shock and inflammatory phenotypes. Moreover, the poly(I:C)-trained larvae exhibit decreased number of macrophages, but not neutrophils, after secondary LPS challenge. Furthermore, training the larvae with poly(I:C) could elevate the transcripts of mTOR signaling and heighten the H3K4me3-mediated epigenetic modifications. And interestingly, we find that inhibiting the H3K4me3 modification, rather than mTOR signaling, could recover the number of macrophages in poly(I:C)-trained larvae, which is consistent with the observations of inflammatory phenotypes. Taken together, these results suggest that poly(I:C) training could induce epigenetic rewiring to mediate the anti-inflammatory response against secondary LPS challenge-induced septic shock through decreasing macrophages' number in vivo, which might expand our understanding of poly(I:C) in regulating fish immune response.
Collapse
Affiliation(s)
- Zhuang Wang
- State Key Laboratory of Bioreactor Engineering, Laboratory for Aquatic Animal Diseases, East China University of Science and Technology, Shanghai, 200237, China
| | - Xinwei You
- State Key Laboratory of Bioreactor Engineering, Laboratory for Aquatic Animal Diseases, East China University of Science and Technology, Shanghai, 200237, China
| | - Yuanxing Zhang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, 200237, China
| | - Qin Liu
- State Key Laboratory of Bioreactor Engineering, Laboratory for Aquatic Animal Diseases, East China University of Science and Technology, Shanghai, 200237, China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, 200237, China
| | - Dahai Yang
- State Key Laboratory of Bioreactor Engineering, Laboratory for Aquatic Animal Diseases, East China University of Science and Technology, Shanghai, 200237, China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, 200237, China.
| |
Collapse
|
10
|
Yang F, Nagahawatta DP, Yang HW, Ryu B, Lee HG, Je JG, Heo MS, Jeon YJ. In vitro and in vivo immuno-enhancing effect of fucoidan isolated from non-edible brown seaweed Sargassum thunbergii. Int J Biol Macromol 2023; 253:127212. [PMID: 37802428 DOI: 10.1016/j.ijbiomac.2023.127212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 08/12/2023] [Accepted: 10/01/2023] [Indexed: 10/09/2023]
Abstract
Fucoidan has been reported to have various biological activities, such as antioxidant, antitumor and anticoagulant, with various health benefits. However, few studies have been conducted to extract fucoidan from Sargassum thunbergii in terms of its immuno-enhancing activities. This aim of this study was to investigate the immuno-enhancing effect of fucoidan (S3) isolated from Sargassum thunbergii through water extraction and ethanol precipitation in RAW 264.7 macrophages and zebrafish. The results showed that S3 contained a relatively high content of fucose and sulfated polysaccharide. Fourier-transform infrared spectroscopy (FTIR) results show that the characteristic peaks at 845 cm-1 and 1220-1270 cm-1 indicate that S3 contains sulfate groups. In vitro, S3 effectively enhanced nitric oxide (NO) production and phagocytic activity. In addition, the results of the study demonstrated that the secretion of tumor necrosis factor-α, interleukin (IL)-6, IL-1β, and IL-10 was upregulated by nuclear factor kappa B (NF-κB) signaling pathway in a dose-dependent manner. In vivo, S3 activates zebrafish immune responses by promoting secretion of NO and activating the NF-κB pathway. Overall, these results suggest that S3 could be used as a functional ingredient added to nutritional supplements and functional foods.
Collapse
Affiliation(s)
- Fengqi Yang
- Department of Marine Life Sciences, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province 63333, Republic of Korea
| | - D P Nagahawatta
- Department of Marine Life Sciences, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea
| | - Hye-Won Yang
- Department of Marine Life Sciences, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea
| | - Bomi Ryu
- Major of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea
| | - Hyo-Geun Lee
- Department of Marine Life Sciences, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea
| | - Jun-Geon Je
- Department of Marine Life Sciences, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea
| | - Moon-Soo Heo
- Department of Marine Life Sciences, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea.
| | - You-Jin Jeon
- Department of Marine Life Sciences, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province 63333, Republic of Korea.
| |
Collapse
|
11
|
Zhang S, Yang H, Wang M, Mantovani D, Yang K, Witte F, Tan L, Yue B, Qu X. Immunomodulatory biomaterials against bacterial infections: Progress, challenges, and future perspectives. Innovation (N Y) 2023; 4:100503. [PMID: 37732016 PMCID: PMC10507240 DOI: 10.1016/j.xinn.2023.100503] [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] [Received: 05/12/2023] [Accepted: 08/24/2023] [Indexed: 09/22/2023] Open
Abstract
Bacterial infectious diseases are one of the leading causes of death worldwide. Even with the use of multiple antibiotic treatment strategies, 4.95 million people died from drug-resistant bacterial infections in 2019. By 2050, the number of deaths will reach 10 million annually. The increasing mortality may be partly due to bacterial heterogeneity in the infection microenvironment, such as drug-resistant bacteria, biofilms, persister cells, intracellular bacteria, and small colony variants. In addition, the complexity of the immune microenvironment at different stages of infection makes biomaterials with direct antimicrobial activity unsatisfactory for the long-term treatment of chronic bacterial infections. The increasing mortality may be partly attributed to the biomaterials failing to modulate the active antimicrobial action of immune cells. Therefore, there is an urgent need for effective alternatives to treat bacterial infections. Accordingly, the development of immunomodulatory antimicrobial biomaterials has recently received considerable interest; however, a comprehensive review of their research progress is lacking. In this review, we focus mainly on the research progress and future perspectives of immunomodulatory antimicrobial biomaterials used at different stages of infection. First, we describe the characteristics of the immune microenvironment in the acute and chronic phases of bacterial infections. Then, we highlight the immunomodulatory strategies for antimicrobial biomaterials at different stages of infection and their corresponding advantages and disadvantages. Moreover, we discuss biomaterial-mediated bacterial vaccines' potential applications and challenges for activating innate and adaptive immune memory. This review will serve as a reference for future studies to develop next-generation immunomodulatory biomaterials and accelerate their translation into clinical practice.
Collapse
Affiliation(s)
- Shutao Zhang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
| | - Hongtao Yang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Minqi Wang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
| | - Diego Mantovani
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Department of Min-Met-Materials Engineering, Research Center of CHU de Quebec, Division of Regenerative Medicine, Laval University, Quebec City, QC G1V 0A6, Canada
| | - Ke Yang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, China
| | - Frank Witte
- Department of Prosthodontics, Geriatric Dentistry and Craniomandibular Disorders, Charite Medical University, Assmannshauser Strasse 4–6, 14197 Berlin, Germany
| | - Lili Tan
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, China
| | - Bing Yue
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
| | - Xinhua Qu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
| |
Collapse
|
12
|
Di Pietrantonio N, Cappellacci I, Mandatori D, Baldassarre MPA, Pandolfi A, Pipino C. Role of Epigenetics and Metabolomics in Predicting Endothelial Dysfunction in Type 2 Diabetes. Adv Biol (Weinh) 2023; 7:e2300172. [PMID: 37616517 DOI: 10.1002/adbi.202300172] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/15/2023] [Indexed: 08/26/2023]
Abstract
Type 2 diabetes (T2D) is a worldwide health problem and cardiovascular disease (CVD) is a leading cause of morbidity and mortality in T2D patients, making the prevention of CVD onset a major priority. It is therefore crucial to optimize diagnosis and treatment to reduce this burden. Endothelial dysfunction is one of the most important prognostic factors for CVD progression, thus novel approaches to identify the early phase of endothelial dysfunction may lead to specific preventive measures to reduce the occurrence of CVD. Nowadays, multiomics approaches have provided unprecedented opportunities to stratify T2D patients into endotypes, improve therapeutic treatment and outcome and amend the survival prediction. Among omics strategies, epigenetics and metabolomics are gaining increasing interest. Recently, a dynamic correlation between metabolic pathways and gene expression through chromatin remodeling, such as DNA methylation, has emerged, indicating new perspectives on the regulatory networks impacting cellular processes. Thus, a better understanding of epigenetic-metabolite relationships can provide insight into the physiological processes altered early in the endothelium that ultimately head to disease development. Here, recent studies on epigenetics and metabolomics related to CVD prevention potentially useful to identify disease biomarkers, as well as new therapies hopefully targeting the early phase of endothelial dysfunction are highlighted.
Collapse
Affiliation(s)
- Nadia Di Pietrantonio
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
- Center for Advanced Studies and Technology-CAST, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
| | - Ilaria Cappellacci
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
- Center for Advanced Studies and Technology-CAST, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
| | - Domitilla Mandatori
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
- Center for Advanced Studies and Technology-CAST, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
| | - Maria Pompea Antonia Baldassarre
- Center for Advanced Studies and Technology-CAST, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
- Department of Medicine and Aging Sciences, "G. d'Annunzio" University Chieti-Pescara, Chieti, 66100, Italy
| | - Assunta Pandolfi
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
- Center for Advanced Studies and Technology-CAST, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
| | - Caterina Pipino
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
- Center for Advanced Studies and Technology-CAST, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
| |
Collapse
|
13
|
Chen PC, Hsieh MH, Kuo WS, Wu LSH, Wang JY. Trained immunity and macrophage reprogramming in allergic disorders. Cell Mol Immunol 2023; 20:1084-1086. [PMID: 37012398 PMCID: PMC10468505 DOI: 10.1038/s41423-023-01005-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 03/11/2023] [Indexed: 04/05/2023] Open
Affiliation(s)
- Pei-Chi Chen
- Center for Allergy, Immunology, and Microbiome (A.I.M.), China Medical University Hospital, Taichung, Taiwan, China
- Department of Nursing, National Tainan Junior College of Nursing, Tainan, Taiwan, China
| | - Miao-Hsi Hsieh
- Center for Allergy, Immunology, and Microbiome (A.I.M.), China Medical University Hospital, Taichung, Taiwan, China
| | - Wen-Shuo Kuo
- Center for Allergy, Immunology, and Microbiome (A.I.M.), China Medical University Hospital, Taichung, Taiwan, China
- School of Chemistry and Materials Science, Nanjing University of Information Science and Technology, Nanjing, China
| | - Lawrence Shih-Hsin Wu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, China
| | - Jiu-Yao Wang
- Center for Allergy, Immunology, and Microbiome (A.I.M.), China Medical University Hospital, Taichung, Taiwan, China.
- Department of Allergy, Immunology, and Rheumatology (AIR), China Medical University Children's Hospital, Taichung, Taiwan, China.
| |
Collapse
|
14
|
Abstract
The inflammaging concept was introduced in 2000 by Prof. Franceschi. This was an evolutionary or rather a revolutionary conceptualization of the immune changes in response to a lifelong stress. This conceptualization permitted to consider the lifelong proinflammatory process as an adaptation which could eventually lead to either beneficial or detrimental consequences. This dichotomy is influenced by both the genetics and the environment. Depending on which way prevails in an individual, the outcome may be healthy longevity or pathological aging burdened with aging-related diseases. The concept of inflammaging has also revealed the complex, systemic nature of aging. Thus, this conceptualization opens the way to consider age-related processes in their complexity, meaning that not only the process but also all counter-processes should be considered. It has also opened the way to add new concepts to the original one, leading to better understanding of the nature of inflammaging and of aging itself. Finally, it showed the way towards potential multimodal interventions involving a holistic approach to optimize the aging process towards a healthy longevity.
Collapse
|
15
|
Movassagh H, Prunicki M, Kaushik A, Zhou X, Dunham D, Smith EM, He Z, Aleman Muench GR, Shi M, Weimer AK, Cao S, Andorf S, Feizi A, Snyder MP, Soroosh P, Mellins ED, Nadeau KC. Proinflammatory polarization of monocytes by particulate air pollutants is mediated by induction of trained immunity in pediatric asthma. Allergy 2023. [PMID: 36929161 DOI: 10.1111/all.15692] [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: 11/09/2022] [Revised: 01/10/2023] [Accepted: 01/24/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND The impact of exposure to air pollutants, such as fine particulate matter (PM), on the immune system and its consequences on pediatric asthma, are not well understood. We investigated whether ambient levels of fine PM with aerodynamic diameter ≤2.5 microns (PM2.5 ) are associated with alterations in circulating monocytes in children with or without asthma. METHODS Monocyte phenotyping was performed by cytometry time-of-flight (CyTOF). Cytokines were measured using cytometric bead array and Luminex assay. ChIP-Seq was utilized to address histone modifications in monocytes. RESULTS Increased exposure to ambient PM2.5 was linked to specific monocyte subtypes, particularly in children with asthma. Mechanistically, we hypothesized that innate trained immunity is evoked by a primary exposure to fine PM and accounts for an enhanced inflammatory response after secondary stimulation in vitro. We determined that the trained immunity was induced in circulating monocytes by fine particulate pollutants, and it was characterized by the upregulation of proinflammatory mediators, such as TNF, IL-6, and IL-8, upon stimulation with house dust mite or lipopolysaccharide. This phenotype was epigenetically controlled by enhanced H3K27ac marks in circulating monocytes. CONCLUSION The specific alterations of monocytes after ambient pollution exposure suggest a possible prognostic immune signature for pediatric asthma, and pollution-induced trained immunity may provide a potential therapeutic target for asthmatic children living in areas with increased air pollution.
Collapse
Affiliation(s)
- Hesam Movassagh
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Palo Alto, California, USA
| | - Mary Prunicki
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Palo Alto, California, USA
| | - Abhinav Kaushik
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Palo Alto, California, USA
| | - Xiaoying Zhou
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Palo Alto, California, USA
| | - Diane Dunham
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Palo Alto, California, USA
| | - Eric M Smith
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Palo Alto, California, USA
| | - Ziyuan He
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Palo Alto, California, USA
| | | | - Minyi Shi
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
- Center for Genomics and Personalized Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Annika K Weimer
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
- Center for Genomics and Personalized Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Shu Cao
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Palo Alto, California, USA
| | - Sandra Andorf
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Palo Alto, California, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Divisions of Biomedical Informatics and Allergy & Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
- Center for Genomics and Personalized Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Pejman Soroosh
- Janssen Research & Development, LLC, San Diego, California, USA
| | - Elizabeth D Mellins
- Department of Pediatrics, Stanford Program in Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Kari C Nadeau
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Palo Alto, California, USA
| |
Collapse
|
16
|
Martín-Cruz L, Sevilla-Ortega C, Angelina A, Domínguez-Andrés J, Netea MG, Subiza JL, Palomares O. From trained immunity in allergy to trained immunity-based allergen vaccines. Clin Exp Allergy 2023; 53:145-155. [PMID: 36494877 DOI: 10.1111/cea.14261] [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: 08/16/2022] [Revised: 11/04/2022] [Accepted: 11/20/2022] [Indexed: 12/14/2022]
Abstract
Innate immune cells experience long lasting metabolic and epigenetic changes after an encounter with specific stimuli. This facilitates enhanced immune responses upon secondary exposition to both the same and unrelated pathogens, a process termed trained immunity. Trained immunity-based vaccines (TIbV) are vaccines able to induce innate immune memory, thus conferring heterologous protection against a broad range of pathogens. While trained immunity has been well documented in the context of infections and multiple immune-mediated diseases, the role of innate immune memory and its contribution to the initiation and maintenance of chronic allergic diseases remains poorly understood. Over the last years, different studies attempting to uncover the role of trained immunity in allergy have emerged. Exposition to environmental factors impacting allergy development such as allergens or viruses induces the reprogramming of innate immune cells to acquire a more pro-inflammatory phenotype in the context of asthma or food allergy. Several studies have convincingly demonstrated that prevention of viral infections using TIbV contributes to reduce wheezing attacks in children, which represent a high-risk factor for asthma development later in life. Innate immune cells trained with specific stimuli might also acquire anti-inflammatory features and promote tolerance, which may have important implications for chronic inflammatory diseases such as allergies. Recent findings showed that allergoid-mannan conjugates, which are next generation vaccines for allergen-specific immunotherapy (AIT), are able to reprogram monocytes into tolerogenic dendritic cells by mechanisms depending on metabolic and epigenetic rewiring. A better understanding of the underlying mechanisms of trained immunity in allergy will pave the way for the design of novel trained immunity-based allergen vaccines as potential alternative strategies for the prevention and treatment of allergic diseases.
Collapse
Affiliation(s)
- Leticia Martín-Cruz
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Carmen Sevilla-Ortega
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Alba Angelina
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Jorge Domínguez-Andrés
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, The Netherlands.,Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, The Netherlands.,Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Immunology and Metabolism, Life & Medical Sciences Institute, University of Bonn, Bonn, Germany
| | | | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
17
|
Effects of Newcastle Disease/Infectious Bronchitis Vaccine and Feeding Yeast Products on the Innate Immune System in the Proventriculus and Ileum of Broiler Chicks. J Poult Sci 2023; 60:2023005. [PMID: 36756044 PMCID: PMC9884637 DOI: 10.2141/jpsa.2023005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/06/2022] [Indexed: 01/25/2023] Open
Abstract
The aim of this study was to determine whether Newcastle disease/infectious bronchitis (ND/IB) vaccination and yeast product diet supplementation modulate the expression of innate immune molecules in the proventriculus and ileum of broiler chicks. One-day-old male broiler chicks were divided into four groups (V-Y- (control), V-Y+, V+Y-, and V+Y+ groups, where V and Y represent vaccination and yeast product supplementation, respectively). Chicks in the V+Y- and V+Y+ groups were immunized with the live ND/IB vaccine, whereas chicks in the V-Y- and V-Y+ groups were not. Chicks in the V-Y+ and V+Y+ groups received feed containing yeast products from day 4, whereas chicks in the V-Y- and V+Y- groups did not. The proventriculus and ileum were collected on day 7 to analyze the expression of seven Toll-like receptors (TLRs) and Dectin-1. In the proventriculus, compared with those of the V-Y- control group, the TLR7 and TLR21 expression levels were higher in the V+Y- group; however, there were no differences in the expression levels of any TLR or Dectin-1 in the ileum. There were also no differences in the expression of avian β-defensins and cathelicidin-1 in the proventriculus and ileum between the control and treatment groups. The expression of granzyme in cytotoxic cells and interleukin (IL)-1B was upregulated by ND/IB vaccination in the proventriculus. Supplementation with yeast products upregulated only granzyme expression in the ileum and downregulated IL-6 expression in the proventriculus in chicks immunized with the ND/IB vaccine. Thus, we concluded that ND/IB vaccination is effective at enhancing the innate immune system in the proventriculus of chicks, at least until day 7 post-hatching, whereas the effects of diet supplementation with yeast products may be limited, at least under the present study conditions.
Collapse
|
18
|
Di Simone SK, Rudloff I, Nold-Petry CA, Forster SC, Nold MF. Understanding respiratory microbiome-immune system interactions in health and disease. Sci Transl Med 2023; 15:eabq5126. [PMID: 36630485 DOI: 10.1126/scitranslmed.abq5126] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Interactions between the developing microbiome and maturing immune system in early life are critical for establishment of a homeostasis beneficial to both host and commensals. The lung harbors a diverse community of microbes associated with health and local or systemic disease. We discuss how early life colonization and community changes correlate with immune development and health and disease throughout infancy, childhood, and adult life. We highlight key advances in microbiology, immunology, and computational biology that allow investigation of the functional relevance of interactions between the respiratory microbiome and host immune system, which may unlock the potential for microbiome-based therapeutics.
Collapse
Affiliation(s)
- Sara K Di Simone
- Department of Paediatrics, Monash University, Melbourne 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne 3168, Australia.,Centre for Innate Immunity and Infectious Disease, Hudson Institute of Medical Research, Melbourne 3168, Australia
| | - Ina Rudloff
- Department of Paediatrics, Monash University, Melbourne 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne 3168, Australia
| | - Claudia A Nold-Petry
- Department of Paediatrics, Monash University, Melbourne 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne 3168, Australia
| | - Samuel C Forster
- Centre for Innate Immunity and Infectious Disease, Hudson Institute of Medical Research, Melbourne 3168, Australia.,Department of Molecular and Translational Science, Monash University, Melbourne 3168, Australia
| | - Marcel F Nold
- Department of Paediatrics, Monash University, Melbourne 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne 3168, Australia.,Monash Newborn, Monash Children's Hospital, Melbourne 3168, Australia
| |
Collapse
|
19
|
Beignon AS, Galeotti C, Menager MM, Schvartz A. Trained immunity as a possible newcomer in autoinflammatory and autoimmune diseases pathophysiology. Front Med (Lausanne) 2023; 9:1085339. [PMID: 36743677 PMCID: PMC9896524 DOI: 10.3389/fmed.2022.1085339] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/20/2022] [Indexed: 01/12/2023] Open
Abstract
Autoimmune disorders have been well characterized over the years and many pathways-but not all of them-have been found to explain their pathophysiology. Autoinflammatory disorders, on the other hand, are still hiding most of their molecular and cellular mechanisms. During the past few years, a newcomer has challenged the idea that only adaptive immunity could display memory response. Trained immunity is defined by innate immune responses that are faster and stronger to a second stimulus than to the first one, being the same or not. In response to the trained immunity inducer, and through metabolic and epigenetic changes of hematopoietic stem and progenitor cells in the bone marrow that are transmitted to their cellular progeny (peripheral trained immunity), or directly of tissue-resident cells (local innate immunity), innate cells responsiveness and functions upon stimulation are improved in the long-term. Innate immunity can be beneficial, but it could also be detrimental when maladaptive. Here, we discuss how trained immunity could contribute to the physiopathology of autoimmune and autoinflammatory diseases.
Collapse
Affiliation(s)
- Anne-Sophie Beignon
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases/Infectious Diseases Models and Innovative Technologies (IMVA-HB/IDMIT), U1184, Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France
| | - Caroline Galeotti
- Department of Pediatric Rheumatology, Reference Center for AutoInflammatory Diseases and Amyloidosis (CEREMAIA), Hôpital Bicêtre, AP-HP, Le Kremlin-Bicêtre, France
| | - Mickael M. Menager
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases/Infectious Diseases Models and Innovative Technologies (IMVA-HB/IDMIT), U1184, Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France
| | - Adrien Schvartz
- Department of Pediatric Rheumatology, Reference Center for AutoInflammatory Diseases and Amyloidosis (CEREMAIA), Hôpital Bicêtre, AP-HP, Le Kremlin-Bicêtre, France,*Correspondence: Adrien Schvartz,
| |
Collapse
|
20
|
Kloc M, Kubiak JZ, Zdanowski R, Ghobrial RM. Memory Macrophages. Int J Mol Sci 2022; 24:ijms24010038. [PMID: 36613481 PMCID: PMC9819859 DOI: 10.3390/ijms24010038] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/12/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Immunological memory is a crucial part of the immune defense that allows organisms to respond against previously encountered pathogens or other harmful factors. Immunological memory is based on the establishment of epigenetic modifications of the genome. The ability to memorize encounters with pathogens and other harmful factors and mount enhanced defense upon subsequent encounters is an evolutionarily ancient mechanism operating in all animals and plants. However, the term immunological memory is usually restricted to the organisms (invertebrates and vertebrates) possessing the immune system. The mammalian immune system, with innate and adaptive branches, is the most sophisticated among vertebrates. The concept of innate memory and memory macrophages is relatively new and thus understudied. We introduce the concept of immunological memory and describe types of memory in different species and their evolutionary status. We discuss why the traditional view of innate immune cells as the first-line defenders is too restrictive and how the innate immune cells can accumulate and retain immunologic memory. We describe how the initial priming leads to chromatin remodeling and epigenetic changes, which allow memory macrophage formation. We also summarize what is currently known about the mechanisms underlying development of memory macrophages; their molecular and metabolic signature and surface markers; and how they may contribute to immune defense, diseases, and organ transplantation.
Collapse
Affiliation(s)
- Malgorzata Kloc
- The Houston Methodist Research Institute, Transplant Immunology, Houston, TX 77030, USA
- Department of Surgery, The Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Genetics, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA
- Correspondence:
| | - Jacek Z. Kubiak
- Dynamics and Mechanics of Epithelia Group, Faculty of Medicine, Institute of Genetics and Development of Rennes, University of Rennes, CNRS, UMR 6290, 35043 Rennes, France
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine National Research Institute, Szaserow 128, 04-141 Warsaw, Poland
| | - Robert Zdanowski
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine National Research Institute, Szaserow 128, 04-141 Warsaw, Poland
| | - Rafik M. Ghobrial
- The Houston Methodist Research Institute, Transplant Immunology, Houston, TX 77030, USA
- Department of Surgery, The Houston Methodist Hospital, Houston, TX 77030, USA
| |
Collapse
|
21
|
Laudanski K, Liu D, Hajj J, Ghani D, Szeto WY. Serum level of total histone 3, H3K4me3, and H3K27ac after non-emergent cardiac surgery suggests the persistence of smoldering inflammation at 3 months in an adult population. Clin Epigenetics 2022; 14:112. [PMID: 36068552 PMCID: PMC9446722 DOI: 10.1186/s13148-022-01331-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/24/2022] [Indexed: 11/29/2022] Open
Abstract
Background Despite clinical relevance of immunological activation due to histone leakage into the serum following cardiac surgery, long-term data describing their longitudinal dynamic are lacking. Therefore, this study examines the serum levels of histone 3 (tH3) and its modifications (H3K4me3 and H3K27ac) alongside immune system activation during the acute and convalescence phases of cardiac surgery. Methods Blood samples from fifty-nine individuals were collected before non-emergent cardiac surgery (tpre-op) and 24 h (t24hr), seven days (t7d), and three months (t3m) post-procedure to examine serum levels of tH3, H3K4me3, and H3K27ac. Serum heat shock protein-60 (HSP-60) was a surrogate of the cellular damage marker. Serum C-reactive protein (CRP) and interleukin 6 (IL-6) assessed smoldering inflammation. TNFα and IL-6 production by whole blood in response to lipopolysaccharide (LPS) evaluated immunological activation. Electronic medical records provided demographic, peri-operative, and clinical information. Paired longitudinal analyses were employed with data expressed as mean and standard deviation (X ± SD) or median and interquartile range (Me[IQ25; 75%]. Results Compared to pre-operative levels (tH3Pre-op = 1.6[0.33;2.4]), post-operative serum tH3 significantly (p > 0.0001) increased after heart surgery (tH324hr = 2.2[0.3;28]), remained elevated at 7 days (tH37d = 2.4[0.37;5.3]), and at 3 months (tH33m = 2.0[0.31;2.9]). Serum H3K27ac was elevated at 24 h (H3K27ac24hr = 0.66 ± 0.51; p = 0.025) and seven days (H3K27ac7d = 0.94 ± 0.95; p = 0.032) as compared to baseline hours (H3K27acPre-op = 0.55 ± 0.54). Serum H3K4me3 was significantly diminished at three months (H3K4me3Pre-op = 0.94 ± 0.54 vs. H3K27ac3m = 0.59 ± 0.89; p = 0.008). tH3 correlated significantly with the duration of anesthesia (r2 = 0.38). In contrast, HSP-60 normalized seven days after surgery. Peri-operative intake of acetaminophen, but no acetylsalicylic acid (ASA), acid, ketorolac or steroids, resulted in the significant depression of serum H3K4me3 at 24 h (H3K4me3acetom- = 1.26[0.71; 3.21] vs H3K4me3acetom+ = 0.54[0.07;1.01]; W[50] = 2.26; p = 0.021). CRP, but not IL-6, remained elevated at 3 months compared to pre-surgical levels and correlated with tH324hrs (r2 = 0.43), tH37d (r2 = 0.71; p < 0.05), H3K4me37d (r2 = 0.53), and H3K27ac7d (r2 = 0.49). Production of TNFα by whole blood in response to LPS was associated with serum tH324hrs (r2 = 0.67). Diminished H3K4me324hrs, H3K27ac24hrs, and H3K27ac3m, accompanied the emergence of liver failure. Conclusions We demonstrated a prolonged elevation in serum histone 3 three months after cardiac surgery. Furthermore, histone 3 modifications had a discrete time evolution indicating differential immune activation.
Collapse
Affiliation(s)
- Krzysztof Laudanski
- Department of Anesthesiology and Critical Care, University of Pennsylvania, JMB 127, 3620 Hamilton Walk, Philadelphia, PA, 19146, USA. .,Department of Neurology, University of Pennsylvania, JMB 127, 3620 Hamilton Walk, Philadelphia, PA, 19146, USA. .,Leonard Davis Institute for Health Economics, University of Pennsylvania, JMB 127, 3620 Hamilton Walk, Philadelphia, PA, 19146, USA.
| | - Da Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Jihane Hajj
- School of Nursing, Widener University, Philadelphia, PA, USA
| | - Danyal Ghani
- Department of Cardiac Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Wilson Y Szeto
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
22
|
Juste RA, Ferreras-Colino E, de la Fuente J, Domínguez M, Risalde MA, Domínguez L, Cabezas-Cruz A, Gortázar C. Heat inactivated mycobacteria, alpha-gal and zebra fish: insights gained from experiences with two promising trained immunity inductors and a validated animal model. Immunol Suppl 2022; 167:139-153. [PMID: 35752944 DOI: 10.1111/imm.13529] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/22/2022] [Indexed: 11/30/2022]
Abstract
Trained immunity (TRAIM) may be defined as a form of memory where innate immune cells such as monocytes, macrophages, dendritic and natural killer (NK) cells undergo an epigenetic reprogramming that enhances their primary defensive capabilities. Cross-pathogen protective TRAIM can be triggered in different hosts by exposure to live microbes or microbe-derived products such as heat-inactivated Mycobacterium bovis or with the glycan α-Gal to elicit protective responses against several pathogens. We review the TRAIM paradigm using two models representing distinct scales of immune sensitization: the whole bacterial cell and one of its building blocks, the polysaccharides or glycans. Observations point out to macrophage lytic capabilities and cytokine regulation as two key components in nonspecific innate immune responses against infections. The study of the TRAIM response deserves attention to better characterize the evolution of host-pathogen cooperation both for identifying the etiology of some diseases and for finding new therapeutic strategies. In this field, the zebrafish provides a convenient and complete biological system that could help to deepen in the knowledge of TRAIM-mediated mechanisms in pathogen-host interactions. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ramón A Juste
- Animal Health Department, NEIKER-Instituto Vasco de Investigación y Desarrollo Agrario, Derio, Bizkaia, Spain.,NySA. Servicio Regional de Investigación y Desarrollo Agroalimentario (SERIDA), Villaviciosa, Asturias, Spain
| | - Elisa Ferreras-Colino
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, Ciudad Real, Spain
| | - José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, Ciudad Real, Spain.,Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, USA
| | - Mercedes Domínguez
- Unidad de Inmunología Microbiana, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Carretera Pozuelo-Majadahonda km 2, 28220 Majadahonda, Madrid, Spain
| | - María A Risalde
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, Facultad de Veterinaria, Universidad de Córdoba (UCO), Córdoba, Spain.,CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Lucas Domínguez
- VISAVET Health Surveillance Centre, Complutense University of Madrid, 28040, Madrid, Spain.,Department of Animal Health, Faculty of Veterinary Medicine, Complutense University of Madrid, Madrid, Spain
| | - Alejandro Cabezas-Cruz
- UMR BIPAR, INRAE, ANSES, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort, France
| | - Christian Gortázar
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, Ciudad Real, Spain
| |
Collapse
|
23
|
Lepoittevin M, Giraud S, Kerforne T, Barrou B, Badet L, Bucur P, Salamé E, Goumard C, Savier E, Branchereau J, Battistella P, Mercier O, Mussot S, Hauet T, Thuillier R. Preservation of Organs to Be Transplanted: An Essential Step in the Transplant Process. Int J Mol Sci 2022; 23:ijms23094989. [PMID: 35563381 PMCID: PMC9104613 DOI: 10.3390/ijms23094989] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 12/23/2022] Open
Abstract
Organ transplantation remains the treatment of last resort in case of failure of a vital organ (lung, liver, heart, intestine) or non-vital organ (essentially the kidney and pancreas) for which supplementary treatments exist. It remains the best alternative both in terms of quality-of-life and life expectancy for patients and of public health expenditure. Unfortunately, organ shortage remains a widespread issue, as on average only about 25% of patients waiting for an organ are transplanted each year. This situation has led to the consideration of recent donor populations (deceased by brain death with extended criteria or deceased after circulatory arrest). These organs are sensitive to the conditions of conservation during the ischemia phase, which have an impact on the graft’s short- and long-term fate. This evolution necessitates a more adapted management of organ donation and the optimization of preservation conditions. In this general review, the different aspects of preservation will be considered. Initially done by hypothermia with the help of specific solutions, preservation is evolving with oxygenated perfusion, in hypothermia or normothermia, aiming at maintaining tissue metabolism. Preservation time is also becoming a unique evaluation window to predict organ quality, allowing repair and/or optimization of recipient choice.
Collapse
Affiliation(s)
- Maryne Lepoittevin
- Biochemistry Department, CHU Poitiers, 86021 Poitiers, France; (M.L.); (S.G.); (R.T.)
- Faculty of Medicine and Pharmacy, University of Poitiers, 86073 Poitiers, France;
- INSERM U1313, IRMETIST, 86021 Poitiers, France; (B.B.); (L.B.)
| | - Sébastien Giraud
- Biochemistry Department, CHU Poitiers, 86021 Poitiers, France; (M.L.); (S.G.); (R.T.)
- Faculty of Medicine and Pharmacy, University of Poitiers, 86073 Poitiers, France;
- INSERM U1313, IRMETIST, 86021 Poitiers, France; (B.B.); (L.B.)
| | - Thomas Kerforne
- Faculty of Medicine and Pharmacy, University of Poitiers, 86073 Poitiers, France;
- INSERM U1313, IRMETIST, 86021 Poitiers, France; (B.B.); (L.B.)
- Cardio-Thoracic and Vascular Surgery Intensive Care Unit, Coordination of P.M.O., CHU Poitiers, 86021 Poitiers, France
| | - Benoit Barrou
- INSERM U1313, IRMETIST, 86021 Poitiers, France; (B.B.); (L.B.)
- Sorbonne Université Campus Pierre et Marie Curie, Faculté de Médecine, 75005 Paris, France
- Service Médico-Chirurgical de Transplantation Rénale, APHP, Hôpital Pitié-Salpêtrière, 75013 Paris, France
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
| | - Lionel Badet
- INSERM U1313, IRMETIST, 86021 Poitiers, France; (B.B.); (L.B.)
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
- Faculté de Médecine, Campus Lyon Santé Est, Université Claude Bernard, 69622 Lyon, France
- Service d’Urologie et Transplantation, Hospices Civils de Lyon, Hôpital Edouard-Herriot, 69003 Lyon, France
| | - Petru Bucur
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
- Service de Chirurgie Digestive et Endocrinienne, Transplantation Hépatique, CHU de Tours, 37170 Chambray les Tours, France
- Groupement d’Imagerie Médicale, CHU de Tours, 37000 Tours, France
- University Hospital Federation SUPORT Tours Poitiers Limoges, 86021 Poitiers, France
| | - Ephrem Salamé
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
- Service de Chirurgie Digestive et Endocrinienne, Transplantation Hépatique, CHU de Tours, 37170 Chambray les Tours, France
- Groupement d’Imagerie Médicale, CHU de Tours, 37000 Tours, France
- University Hospital Federation SUPORT Tours Poitiers Limoges, 86021 Poitiers, France
| | - Claire Goumard
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
- Service de Chirurgie Digestive, Hépato-Bilio-Pancréatique et Transplantation Hépatique, APHP, Hôpital Pitié-Salpêtrière, 75013 Paris, France
| | - Eric Savier
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
- Service de Chirurgie Digestive, Hépato-Bilio-Pancréatique et Transplantation Hépatique, APHP, Hôpital Pitié-Salpêtrière, 75013 Paris, France
| | - Julien Branchereau
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
- Service d’Urologie et de Transplantation, CHU de Nantes, 44000 Nantes, France
| | - Pascal Battistella
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
- Service de Cardiologie et Maladies Vasculaires, CHU de Montpellier, CEDEX 5, 34295 Montpellier, France
| | - Olaf Mercier
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
- Service de Chirurgie Thoracique et Cardio-Vasculaire, Centre Chirurgical Marie LANNELONGUE, 92350 Le Plessis Robinson, France
| | - Sacha Mussot
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
- Service de Chirurgie Thoracique et Cardio-Vasculaire, Centre Chirurgical Marie LANNELONGUE, 92350 Le Plessis Robinson, France
| | - Thierry Hauet
- Biochemistry Department, CHU Poitiers, 86021 Poitiers, France; (M.L.); (S.G.); (R.T.)
- Faculty of Medicine and Pharmacy, University of Poitiers, 86073 Poitiers, France;
- INSERM U1313, IRMETIST, 86021 Poitiers, France; (B.B.); (L.B.)
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
- University Hospital Federation SUPORT Tours Poitiers Limoges, 86021 Poitiers, France
- Correspondence:
| | - Raphael Thuillier
- Biochemistry Department, CHU Poitiers, 86021 Poitiers, France; (M.L.); (S.G.); (R.T.)
- Faculty of Medicine and Pharmacy, University of Poitiers, 86073 Poitiers, France;
- INSERM U1313, IRMETIST, 86021 Poitiers, France; (B.B.); (L.B.)
| |
Collapse
|
24
|
Rigo R, Chelbi R, Agopian J, Letard S, Griffon A, Ghamlouch H, Vernerey J, Ladopoulos V, Voisset E, De Sepulveda P, Guittard G, Nunès JA, Bidaut G, Göttgens B, Weber M, Bernard OA, Dubreuil P, Soucie E. TET2 regulates immune tolerance in chronically activated mast cells. JCI Insight 2022; 7:154191. [PMID: 35393954 PMCID: PMC9057605 DOI: 10.1172/jci.insight.154191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 02/18/2022] [Indexed: 11/17/2022] Open
Abstract
Mutation of the TET2 DNA-hydroxymethylase has been associated with a number of immune pathologies. The disparity in phenotype and clinical presentation among these pathologies leads to questions regarding the role of TET2 mutation in promoting disease evolution in different immune cell types. Here we show that, in primary mast cells, Tet2 expression is induced in response to chronic and acute activation signals. In TET2-deficient mast cells, chronic activation via the oncogenic KITD816V allele associated with mastocytosis, selects for a specific epigenetic signature characterized by hypermethylated DNA regions (HMR) at immune response genes. H3K27ac and transcription factor binding is consistent with priming or more open chromatin at both HMR and non-HMR in proximity to immune genes in these cells, and this signature coincides with increased pathological inflammation signals. HMR are also associated with a subset of immune genes that are direct targets of TET2 and repressed in TET2-deficient cells. Repression of these genes results in immune tolerance to acute stimulation that can be rescued with vitamin C treatment or reiterated with a Tet inhibitor. Overall, our data support a model where TET2 plays a direct role in preventing immune tolerance in chronically activated mast cells, supporting TET2 as a viable target to reprogram the innate immune response for innovative therapies.
Collapse
Affiliation(s)
- Riccardo Rigo
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Institut Paoli-Calmettes, Equipe Labélisée Ligue Nationale Contre le Cancer, Marseille, France
| | - Rabie Chelbi
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Institut Paoli-Calmettes, Equipe Labélisée Ligue Nationale Contre le Cancer, Marseille, France.,Inovarion, Paris, France
| | - Julie Agopian
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Institut Paoli-Calmettes, Equipe Labélisée Ligue Nationale Contre le Cancer, Marseille, France
| | - Sebastien Letard
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Institut Paoli-Calmettes, Equipe Labélisée Ligue Nationale Contre le Cancer, Marseille, France
| | - Aurélien Griffon
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Institut Paoli-Calmettes, Equipe Labélisée Ligue Nationale Contre le Cancer, Marseille, France
| | - Hussein Ghamlouch
- INSERM, Mixed Research Unit (UMR) 1170, Institut Gustave Roussy, Facility of Medicine, Paris-Sud University, Paris-Saclay University, Equipe Labélisée Ligue Nationale Contre le Cancer, Villejuif, France
| | - Julien Vernerey
- CRCM, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille University, Marseille, France
| | - Vasileios Ladopoulos
- Department of Haematology, Cambridge Institute for Medical Research, and.,Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Edwige Voisset
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Institut Paoli-Calmettes, Equipe Labélisée Ligue Nationale Contre le Cancer, Marseille, France
| | - Paulo De Sepulveda
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Institut Paoli-Calmettes, Equipe Labélisée Ligue Nationale Contre le Cancer, Marseille, France
| | - Geoffrey Guittard
- CRCM, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille University, Marseille, France
| | - Jacques A Nunès
- CRCM, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille University, Marseille, France
| | - Ghislain Bidaut
- CRCM, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille University, Marseille, France
| | - Berthold Göttgens
- Department of Haematology, Cambridge Institute for Medical Research, and.,Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Michael Weber
- CNRS, University of Strasbourg, UMR7242 Biotechnology and Cell Signaling, Illkirch, France
| | - Olivier A Bernard
- INSERM, Mixed Research Unit (UMR) 1170, Institut Gustave Roussy, Facility of Medicine, Paris-Sud University, Paris-Saclay University, Equipe Labélisée Ligue Nationale Contre le Cancer, Villejuif, France
| | - Patrice Dubreuil
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Institut Paoli-Calmettes, Equipe Labélisée Ligue Nationale Contre le Cancer, Marseille, France
| | - Erinn Soucie
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Institut Paoli-Calmettes, Equipe Labélisée Ligue Nationale Contre le Cancer, Marseille, France
| |
Collapse
|
25
|
Netea MG, Domínguez-Andrés J, van de Veerdonk FL, van Crevel R, Pulendran B, van der Meer JWM. Natural resistance against infections: focus on COVID-19. Trends Immunol 2022; 43:106-116. [PMID: 34924297 PMCID: PMC8648669 DOI: 10.1016/j.it.2021.12.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 12/27/2022]
Abstract
Not all individuals exposed to a pathogen develop illness: some are naturally resistant whereas others develop an asymptomatic infection. Epidemiological studies suggest that there is similar variability in susceptibility to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections. We propose that natural resistance is part of the disease history in some individuals exposed to this new coronavirus. Epidemiological arguments for natural resistance to SARS-CoV-2 are the lower seropositivity of children compared to adults, studies on closed environments of ships with outbreaks, and prevalence studies in some developing countries. Potential mechanisms of natural resistance include host genetic variants, viral interference, cross-protective natural antibodies, T cell immunity, and highly effective innate immune responses. Better understanding of natural resistance can help to advance preventive and therapeutic measures against infections for improved preparedness against potential future pandemics.
Collapse
Affiliation(s)
- Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany.
| | - Jorge Domínguez-Andrés
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frank L van de Veerdonk
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Reinout van Crevel
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Bali Pulendran
- Institute for Immunology, Transplantation and Infectious Diseases, Department of Pathology and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jos W M van der Meer
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
26
|
Al-Moussawy M, Abdelsamed HA, Lakkis FG. Immunoglobulin-like receptors and the generation of innate immune memory. Immunogenetics 2022; 74:179-195. [PMID: 35034136 PMCID: PMC10074160 DOI: 10.1007/s00251-021-01240-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 11/25/2021] [Indexed: 12/22/2022]
Abstract
Host immunity is classically divided into "innate" and "adaptive." While the former has always been regarded as the first, rapid, and antigen-nonspecific reaction to invading pathogens, the latter represents the more sophisticated and antigen-specific response that has the potential to persist and generate memory. Recent work however has challenged this dogma, where murine studies have successfully demonstrated the ability of innate immune cells (monocytes and macrophages) to acquire antigen-specific memory to allogeneic major histocompatibility complex (MHC) molecules. The immunoreceptors so far identified that mediate innate immune memory are the paired immunoglobulin-like receptors (PIRs) in mice, which are orthologous to human leukocyte immunoglobulin-like receptors (LILRs). These receptor families are mainly expressed by the myelomonocytic cell lineage, suggesting an important role in the innate immune response. In this review, we will discuss the role of immunoglobulin-like receptors in the development of innate immune memory across species.
Collapse
Affiliation(s)
- Mouhamad Al-Moussawy
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, USA.
| | - Hossam A Abdelsamed
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, USA. .,Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, USA.
| | - Fadi G Lakkis
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, USA. .,Department of Immunology, University of Pittsburgh, Pittsburgh, USA. .,Department of Medicine, University of Pittsburgh, Pittsburgh, USA.
| |
Collapse
|
27
|
Pellon A, Barriales D, Peña-Cearra A, Castelo-Careaga J, Palacios A, Lopez N, Atondo E, Pascual-Itoiz MA, Martín-Ruiz I, Sampedro L, Gonzalez-Lopez M, Bárcena L, Martín-Mateos T, Landete JM, Prados-Rosales R, Plaza-Vinuesa L, Muñoz R, de las Rivas B, Rodríguez JM, Berra E, Aransay AM, Abecia L, Lavín JL, Rodríguez H, Anguita J. The commensal bacterium Lactiplantibacillus plantarum imprints innate memory-like responses in mononuclear phagocytes. Gut Microbes 2022; 13:1939598. [PMID: 34224309 PMCID: PMC8259724 DOI: 10.1080/19490976.2021.1939598] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Gut microbiota is a constant source of antigens and stimuli to which the resident immune system has developed tolerance. However, the mechanisms by which mononuclear phagocytes, specifically monocytes/macrophages, cope with these usually pro-inflammatory signals are poorly understood. Here, we show that innate immune memory promotes anti-inflammatory homeostasis, using as model strains of the commensal bacterium Lactiplantibacillus plantarum. Priming of monocytes/macrophages with bacteria, especially in its live form, enhances bacterial intracellular survival and decreases the release of pro-inflammatory signals to the environment, with lower production of TNF and higher levels of IL-10. Analysis of the transcriptomic landscape of these cells shows downregulation of pathways associated with the production of reactive oxygen species (ROS) and the release of cytokines, chemokines and antimicrobial peptides. Indeed, the induction of ROS prevents memory-induced bacterial survival. In addition, there is a dysregulation in gene expression of several metabolic pathways leading to decreased glycolytic and respiratory rates in memory cells. These data support commensal microbe-specific metabolic changes in innate immune memory cells that might contribute to homeostasis in the gut.
Collapse
Affiliation(s)
- Aize Pellon
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Diego Barriales
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Ainize Peña-Cearra
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain,Faculty of Medicine and Nursing, Universidad Del Pais Vasco (UPV/EHU), Leioa, Spain
| | - Janire Castelo-Careaga
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Ainhoa Palacios
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Nerea Lopez
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Estibaliz Atondo
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Miguel Angel Pascual-Itoiz
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Itziar Martín-Ruiz
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Leticia Sampedro
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | | | - Laura Bárcena
- Genomic Analysis Platform, CIC bioGUNE-BRTA, Derio, Spain
| | - Teresa Martín-Mateos
- Physiopathology of the Hypoxia-signaling Pathway Laboratory, CIC bioGUNE-BRTA, Derio, Spain
| | - Jose María Landete
- Departamento De Tecnología De Alimentos, Instituto Nacional De Investigación Y Tecnología Agraria Y Alimentaria (INIA), Madrid, Spain
| | - Rafael Prados-Rosales
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain,Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, United Kingdom; RPR: Department of Preventive Medicine and Public Health and Microbiology, Universidad Autónoma De Madrid, Madrid 28029, Spain; JLL: Applied Mathematics Department, Bioinformatics Unit, NEIKER-BRTA, Parque Tecnológico De Bizkaia, Derio, Spain
| | - Laura Plaza-Vinuesa
- Laboratorio De Biotecnología Bacteriana, Instituto De Ciencia Y Tecnología De Alimentos Y Nutrición (ICTAN-CSIC), Madrid, Spain
| | - Rosario Muñoz
- Laboratorio De Biotecnología Bacteriana, Instituto De Ciencia Y Tecnología De Alimentos Y Nutrición (ICTAN-CSIC), Madrid, Spain
| | - Blanca de las Rivas
- Laboratorio De Biotecnología Bacteriana, Instituto De Ciencia Y Tecnología De Alimentos Y Nutrición (ICTAN-CSIC), Madrid, Spain
| | - Juan Miguel Rodríguez
- Department of Nutrition and Food Science, Universidad Complutense De Madrid, Madrid, Spain
| | - Edurne Berra
- Physiopathology of the Hypoxia-signaling Pathway Laboratory, CIC bioGUNE-BRTA, Derio, Spain
| | - Ana M. Aransay
- Genomic Analysis Platform, CIC bioGUNE-BRTA, Derio, Spain,CIBERehd, ISCIII, Madrid, Spain
| | - Leticia Abecia
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain,Faculty of Medicine and Nursing, Universidad Del Pais Vasco (UPV/EHU), Leioa, Spain
| | - Jose Luis Lavín
- Bioinformatics Unit, CIC bioGUNE-BRTA, Derio, Spain,Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, United Kingdom; RPR: Department of Preventive Medicine and Public Health and Microbiology, Universidad Autónoma De Madrid, Madrid 28029, Spain; JLL: Applied Mathematics Department, Bioinformatics Unit, NEIKER-BRTA, Parque Tecnológico De Bizkaia, Derio, Spain
| | - Hector Rodríguez
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain,Hector Rodríguez Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio48160, Spain
| | - Juan Anguita
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain,Ikerbasque, Basque Foundation for Science, Bilbao, Bizkaia, Spain,CONTACT Juan Anguita Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio48160, Spain
| |
Collapse
|
28
|
Mata E, Tarancon R, Guerrero C, Moreo E, Moreau F, Uranga S, Gomez AB, Marinova D, Domenech M, Gonzalez-Camacho F, Monzon M, Badiola J, Dominguez-Andres J, Yuste J, Anel A, Peixoto A, Martin C, Aguilo N. Pulmonary BCG induces lung-resident macrophage activation and confers long-term protection against tuberculosis. Sci Immunol 2021; 6:eabc2934. [PMID: 34559551 DOI: 10.1126/sciimmunol.abc2934] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Elena Mata
- Grupo de Genética de Micobacterias, IIS Aragón, Facultad de Medicina, Universidad de Zaragoza, C/ Domingo Miral s/n, 50009 Zaragoza, Spain.,CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Raquel Tarancon
- Grupo de Genética de Micobacterias, IIS Aragón, Facultad de Medicina, Universidad de Zaragoza, C/ Domingo Miral s/n, 50009 Zaragoza, Spain.,CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Claudia Guerrero
- Grupo de Genética de Micobacterias, IIS Aragón, Facultad de Medicina, Universidad de Zaragoza, C/ Domingo Miral s/n, 50009 Zaragoza, Spain.,CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Eduardo Moreo
- Grupo de Genética de Micobacterias, IIS Aragón, Facultad de Medicina, Universidad de Zaragoza, C/ Domingo Miral s/n, 50009 Zaragoza, Spain.,CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Flavie Moreau
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Santiago Uranga
- Grupo de Genética de Micobacterias, IIS Aragón, Facultad de Medicina, Universidad de Zaragoza, C/ Domingo Miral s/n, 50009 Zaragoza, Spain.,CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Belen Gomez
- Grupo de Genética de Micobacterias, IIS Aragón, Facultad de Medicina, Universidad de Zaragoza, C/ Domingo Miral s/n, 50009 Zaragoza, Spain.,CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Dessislava Marinova
- Grupo de Genética de Micobacterias, IIS Aragón, Facultad de Medicina, Universidad de Zaragoza, C/ Domingo Miral s/n, 50009 Zaragoza, Spain.,CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Miriam Domenech
- CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - Fernando Gonzalez-Camacho
- CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Monzon
- Research Centre for Encephalopathies and Transmissible Emerging Diseases, Universidad de Zaragoza, Zaragoza, Spain
| | - Juan Badiola
- Research Centre for Encephalopathies and Transmissible Emerging Diseases, Universidad de Zaragoza, Zaragoza, Spain
| | - Jorge Dominguez-Andres
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Center, Nijmegen, Netherlands
| | - Jose Yuste
- CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - Alberto Anel
- Grupo Apoptosis, Inmunidad y Cáncer, IIS Aragón, Dpto. Bioquímica y Biología Molecular y Celular, Fac. Ciencias, Universidad de Zaragoza, Zaragoza, Spain
| | - Antonio Peixoto
- Research Centre for Encephalopathies and Transmissible Emerging Diseases, Universidad de Zaragoza, Zaragoza, Spain
| | - Carlos Martin
- Grupo de Genética de Micobacterias, IIS Aragón, Facultad de Medicina, Universidad de Zaragoza, C/ Domingo Miral s/n, 50009 Zaragoza, Spain.,CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Servicio de Microbiología, Hospital Universitario Miguel Servet, ISS Aragón, Paseo, Isabel la Católica 1-3, 50009 Zaragoza, Spain
| | - Nacho Aguilo
- Grupo de Genética de Micobacterias, IIS Aragón, Facultad de Medicina, Universidad de Zaragoza, C/ Domingo Miral s/n, 50009 Zaragoza, Spain.,CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
29
|
Jiang M, Xu K, Ren H, Wang M, Hou X, Cao J. Role of lincRNA-Cox2 targeting miR-150 in regulating the viability of chondrocytes in osteoarthritis. Exp Ther Med 2021; 22:800. [PMID: 34093756 PMCID: PMC8170664 DOI: 10.3892/etm.2021.10232] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 04/14/2021] [Indexed: 12/22/2022] Open
Abstract
Osteoarthritis (OA) is a joint disease characterised by progressive cartilage degradation and inflammation, but the detailed pathogenesis of OA remains unclear. The present study aimed to investigate the role of long intergenic non-coding RNA (lincRNA)-Cox2 in OA progression and the potential mechanism. An OA mouse model was used for in vivo experiments, and IL-1β-induced injury of mouse chondrocytes was conducted for in vitro experiments. Small interfering (si)-Cox2 was transfected into chondrocytes to elucidate the effect of lincRNA-Cox2 on OA. Quantitative reverse transcription PCR assays were conducted to detect the expression of lincRNA-Cox2 and microRNA (miR)-150. Cell proliferation and apoptosis were analysed based on an MTT assay and annexin V/propidium iodide staining, respectively. Western blotting was performed to evaluate the protein expression levels of Ki-67, PCNA, Bax, cleaved (c)-Caspase-3, c-Caspase-9 and Wnt/β-catenin pathway-associated proteins in chondrocytes. High levels of lincRNA-Cox2 were observed in cartilage tissues of the OA mouse model in vivo. In the in vitro experiments, the expression of lincRNA-Cox2 was increased in IL-1β-treated chondrocytes. Knockdown of lincRNA-Cox2 promoted the proliferation and inhibited the apoptosis of chondrocytes. Mechanistically, lincRNA-Cox2 was found to directly target miR-150, acting as a competing endogenous RNA, and the effect of si-Cox2 on the proliferation and apoptosis of chondrocytes was reversed by miR-150 inhibitors. Moreover, lincRNA-Cox2 activated the Wnt/β-catenin pathway to regulate chondrocyte proliferation and apoptosis. The present study demonstrated that silencing lincRNA-Cox2 expression plays a protective role in OA by enhancing the proliferation and suppressing the apoptosis of chondrocytes, which is related to increased miR-150 expression and activation of the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Meng Jiang
- Department of Orthopaedics, Qingdao No. 6 People's Hospital, Qingdao, Shandong 266033, P.R. China
| | - Kai Xu
- Department of Orthopaedics, Qingdao No. 6 People's Hospital, Qingdao, Shandong 266033, P.R. China
| | - Huafeng Ren
- Department of Functional Examination, Qingdao Haici Medical Group, Qingdao, Shandong 266033, P.R. China
| | - Mingmin Wang
- Department of Orthopaedics, Qingdao No. 6 People's Hospital, Qingdao, Shandong 266033, P.R. China
| | - Ximin Hou
- Department of Orthopaedics, Qingdao No. 6 People's Hospital, Qingdao, Shandong 266033, P.R. China
| | - Jianping Cao
- Department of Anesthesiology, Qingdao No. 6 People's Hospital, Qingdao, Shandong 266033, P.R. China
| |
Collapse
|
30
|
Non-coding RNAs in endodontic disease. Semin Cell Dev Biol 2021; 124:82-84. [PMID: 34257038 DOI: 10.1016/j.semcdb.2021.07.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/27/2021] [Accepted: 07/04/2021] [Indexed: 12/22/2022]
Abstract
The immunocompetence and regeneration potential of the dental pulp and its surrounding apical tissues have been investigated extensively in the field of endodontics. While research on the role of non-coding RNAs in these tissues is still in its infancy, it is envisioned that improved understanding of the regulatory function of ncRNAs in pulpal and periapical immune response will help prevent or treat endodontic disease. Of particular importance is the role of these RNAs in regenerating the dentin-pulp complex. In this review, we highlight recent progress on the role of non-coding RNAs in the immune response to endodontic infection as well as the repair and regenerative response to injury.
Collapse
|
31
|
Munawara U, Catanzaro M, Xu W, Tan C, Hirokawa K, Bosco N, Dumoulin D, Khalil A, Larbi A, Lévesque S, Ramassamy C, Barron AE, Cunnane S, Beauregard PB, Bellenger JP, Rodrigues S, Desroches M, Witkowski JM, Laurent B, Frost EH, Fulop T. Hyperactivation of monocytes and macrophages in MCI patients contributes to the progression of Alzheimer's disease. IMMUNITY & AGEING 2021; 18:29. [PMID: 34154615 PMCID: PMC8215492 DOI: 10.1186/s12979-021-00236-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/09/2021] [Indexed: 02/07/2023]
Abstract
Background Alzheimer’s disease (AD) is the most common neurodegenerative disease ultimately manifesting as clinical dementia. Despite considerable effort and ample experimental data, the role of neuroinflammation related to systemic inflammation is still unsettled. While the implication of microglia is well recognized, the exact contribution of peripheral monocytes/macrophages is still largely unknown, especially concerning their role in the various stages of AD. Objectives AD develops over decades and its clinical manifestation is preceded by subjective memory complaints (SMC) and mild cognitive impairment (MCI); thus, the question arises how the peripheral innate immune response changes with the progression of the disease. Therefore, to further investigate the roles of monocytes/macrophages in the progression of AD we assessed their phenotypes and functions in patients at SMC, MCI and AD stages and compared them with cognitively healthy controls. We also conceptualised an idealised mathematical model to explain the functionality of monocytes/macrophages along the progression of the disease. Results We show that there are distinct phenotypic and functional changes in monocyte and macrophage populations as the disease progresses. Higher free radical production upon stimulation could already be observed for the monocytes of SMC patients. The most striking results show that activation of peripheral monocytes (hyperactivation) is the strongest in the MCI group, at the prodromal stage of the disease. Monocytes exhibit significantly increased chemotaxis, free radical production, and cytokine production in response to TLR2 and TLR4 stimulation. Conclusion Our data suggest that the peripheral innate immune system is activated during the progression from SMC through MCI to AD, with the highest levels of activation being in MCI subjects and the lowest in AD patients. Some of these parameters may be used as biomarkers, but more holistic immune studies are needed to find the best period of the disease for clinical intervention. Supplementary Information The online version contains supplementary material available at 10.1186/s12979-021-00236-x.
Collapse
Affiliation(s)
- Usma Munawara
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001, 12th Avenue North, Sherbrooke, Quebec, J1H 5N4, Canada
| | - Michael Catanzaro
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001, 12th Avenue North, Sherbrooke, Quebec, J1H 5N4, Canada.,Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Weili Xu
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, Biopolis, Singapore, Singapore
| | - Crystal Tan
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, Biopolis, Singapore, Singapore
| | - Katsuiku Hirokawa
- Department of Diagnostic Pathology, Institute of Health and Life Science, Tokyo Med. Dent. University, Tokyo and Nitobe Memorial Nakanosogo Hospital, Tokyo, Japan
| | - Nabil Bosco
- Nestlé Research, Nestlé Institute of Health Sciences, Department of Cell Biology, Cellular Metabolism, EPFL Innovation Park, CH-1015, Lausanne, Switzerland
| | - David Dumoulin
- Department of Biology, Faculty of Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Abdelouahed Khalil
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001, 12th Avenue North, Sherbrooke, Quebec, J1H 5N4, Canada
| | - Anis Larbi
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001, 12th Avenue North, Sherbrooke, Quebec, J1H 5N4, Canada.,Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, Biopolis, Singapore, Singapore
| | - Simon Lévesque
- Department of Microbiology and Infectiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Charles Ramassamy
- INRS-Centre Armand-Frappier Santé-biotechnologie, Montréal, Québec, Canada
| | - Annelise E Barron
- Department of Bioengineering, Stanford School of Medicine, Stanford, California, USA
| | - Stephen Cunnane
- Research Center on Aging, Endocrinology Division, Department of Medicine, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Pascale B Beauregard
- Department of Biology, Faculty of Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Jean-Pierre Bellenger
- Department of Chemistry, Faculty of Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Serafim Rodrigues
- Ikerbasque, The Basque Foundation for Science, Bilbao, Spain. .,Basque Center for Applied Mathematics, Mathematical, Computational and Experimental Neuroscience research group, Alameda de Mazarredo 14, 48009, Bilbao, Bizkaia, Basque-Country, Spain.
| | - Mathieu Desroches
- MathNeuro Team, Inria Sophia Antipolis Méditerranée, Valbonne, France.,Université Côte d'Azur, Nice, France
| | - Jacek M Witkowski
- Department of Pathophysiology, Medical University of Gdansk, Gdansk, Poland
| | - Benoit Laurent
- Research Center on Aging, Department of Biochemistry, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Eric H Frost
- Department of Microbiology and Infectiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Tamas Fulop
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001, 12th Avenue North, Sherbrooke, Quebec, J1H 5N4, Canada.
| |
Collapse
|
32
|
Friščić J, Böttcher M, Reinwald C, Bruns H, Wirth B, Popp SJ, Walker KI, Ackermann JA, Chen X, Turner J, Zhu H, Seyler L, Euler M, Kirchner P, Krüger R, Ekici AB, Major T, Aust O, Weidner D, Fischer A, Andes FT, Stanojevic Z, Trajkovic V, Herrmann M, Korb-Pap A, Wank I, Hess A, Winter J, Wixler V, Distler J, Steiner G, Kiener HP, Frey B, Kling L, Raza K, Frey S, Kleyer A, Bäuerle T, Hughes TR, Grüneboom A, Steffen U, Krönke G, Croft AP, Filer A, Köhl J, Klein K, Buckley CD, Schett G, Mougiakakos D, Hoffmann MH. The complement system drives local inflammatory tissue priming by metabolic reprogramming of synovial fibroblasts. Immunity 2021; 54:1002-1021.e10. [PMID: 33761330 DOI: 10.1016/j.immuni.2021.03.003] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 12/23/2020] [Accepted: 03/05/2021] [Indexed: 12/14/2022]
Abstract
Arthritis typically involves recurrence and progressive worsening at specific predilection sites, but the checkpoints between remission and persistence remain unknown. Here, we defined the molecular and cellular mechanisms of this inflammation-mediated tissue priming. Re-exposure to inflammatory stimuli caused aggravated arthritis in rodent models. Tissue priming developed locally and independently of adaptive immunity. Repeatedly stimulated primed synovial fibroblasts (SFs) exhibited enhanced metabolic activity inducing functional changes with intensified migration, invasiveness and osteoclastogenesis. Meanwhile, human SF from patients with established arthritis displayed a similar primed phenotype. Transcriptomic and epigenomic analyses as well as genetic and pharmacological targeting demonstrated that inflammatory tissue priming relies on intracellular complement C3- and C3a receptor-activation and downstream mammalian target of rapamycin- and hypoxia-inducible factor 1α-mediated metabolic SF invigoration that prevents activation-induced senescence, enhances NLRP3 inflammasome activity, and in consequence sensitizes tissue for inflammation. Our study suggests possibilities for therapeutic intervention abrogating tissue priming without immunosuppression.
Collapse
Affiliation(s)
- Jasna Friščić
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Martin Böttcher
- Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Department of Medicine 5 for Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Christiane Reinwald
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Heiko Bruns
- Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Department of Medicine 5 for Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Benjamin Wirth
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Samantha-Josefine Popp
- Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Department of Medicine 5 for Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Kellie Irene Walker
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Jochen A Ackermann
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Xi Chen
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Jason Turner
- Institute for Inflammation and Ageing, University of Birmingham, NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, B15 2TT Birmingham, United Kingdom
| | - Honglin Zhu
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Lisa Seyler
- Institute of Radiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) ands Universitäts-klinikum Erlangen, 91054, Erlangen, Germany
| | - Maximilien Euler
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Philipp Kirchner
- Institute of Human Genetics, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - René Krüger
- Institute of Human Genetics, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Arif B Ekici
- Institute of Human Genetics, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Triin Major
- Institute for Inflammation and Ageing, University of Birmingham, NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, B15 2TT Birmingham, United Kingdom
| | - Oliver Aust
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Daniela Weidner
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Anita Fischer
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, 1090 Vienna, Austria; Ludwig Boltzmann Institute for Arthritis and Rehabilitation, 1090 Vienna, Austria
| | - Fabian T Andes
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Zeljka Stanojevic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Vladimir Trajkovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Martin Herrmann
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Adelheid Korb-Pap
- Institute of Musculoskeletal Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, D3, 48149 Muenster, Germany
| | - Isabel Wank
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Andreas Hess
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Johnathan Winter
- Division of Infection and Immunity, School of Medicine, Cardiff University, CF10 3AT, Cardiff, UK
| | - Viktor Wixler
- Institute of Molecular Virology (IMV), Centre for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms University Muenster, 48149 Muenster, Germany
| | - Jörg Distler
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Günter Steiner
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, 1090 Vienna, Austria; Ludwig Boltzmann Institute for Arthritis and Rehabilitation, 1090 Vienna, Austria
| | - Hans P Kiener
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, 1090 Vienna, Austria
| | - Benjamin Frey
- Department of Radiation Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Lasse Kling
- Innovations-Institut für Nanotechnologie und korrelative Mikroskopie, 91301 Forchheim, Germany
| | - Karim Raza
- Institute for Inflammation and Ageing, University of Birmingham, NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, B15 2TT Birmingham, United Kingdom; Department of Rheumatology, City Hospital, Sandwell and West Birmingham, B18 7QH Birmingham, UK
| | - Silke Frey
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Arnd Kleyer
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Tobias Bäuerle
- Institute of Radiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) ands Universitäts-klinikum Erlangen, 91054, Erlangen, Germany
| | - Timothy R Hughes
- Division of Infection and Immunity, School of Medicine, Cardiff University, CF10 3AT, Cardiff, UK
| | - Anika Grüneboom
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Ulrike Steffen
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Adam P Croft
- Institute for Inflammation and Ageing, University of Birmingham, NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, B15 2TT Birmingham, United Kingdom
| | - Andrew Filer
- Institute for Inflammation and Ageing, University of Birmingham, NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, B15 2TT Birmingham, United Kingdom
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany; Division of Immunobiology, Cincinnati Childrens Hospital Medical Center and University of Cincinnati College of Medicine, 45229-3026 Cincinnati, OH, USA
| | - Kerstin Klein
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Christopher D Buckley
- Institute for Inflammation and Ageing, University of Birmingham, NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, B15 2TT Birmingham, United Kingdom; Kennedy Institute of Rheumatology, University of Oxford, OX3 7FY Oxford, UK
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Dimitrios Mougiakakos
- Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Department of Medicine 5 for Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Markus H Hoffmann
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum fuer Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany.
| |
Collapse
|
33
|
Barbosa MMF, Kanno AI, Farias LP, Madej M, Sipos G, Sbrana S, Romani L, Boraschi D, Leite LCC, Italiani P. Primary and Memory Response of Human Monocytes to Vaccines: Role of Nanoparticulate Antigens in Inducing Innate Memory. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:931. [PMID: 33917456 PMCID: PMC8067467 DOI: 10.3390/nano11040931] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/25/2021] [Accepted: 04/01/2021] [Indexed: 01/09/2023]
Abstract
Innate immune cells such as monocytes and macrophages are activated in response to microbial and other challenges and mount an inflammatory defensive response. Exposed cells develop the so-called innate memory, which allows them to react differently to a subsequent challenge, aiming at better protection. In this study, using human primary monocytes in vitro, we have assessed the memory-inducing capacity of two antigenic molecules of Schistosoma mansoni in soluble form compared to the same molecules coupled to outer membrane vesicles of Neisseria lactamica. The results show that particulate challenges are much more efficient than soluble molecules in inducing innate memory, which is measured as the production of inflammatory and anti-inflammatory cytokines (TNFα, IL-6, IL-10). Controls run with LPS from Klebsiella pneumoniae compared to the whole bacteria show that while LPS alone has strong memory-inducing capacity, the entire bacteria are more efficient. These data suggest that microbial antigens that are unable to induce innate immune activation can nevertheless participate in innate activation and memory when in a particulate form, which is a notion that supports the use of nanoparticulate antigens in vaccination strategies for achieving adjuvant-like effects of innate activation as well as priming for improved reactivity to future challenges.
Collapse
Affiliation(s)
- Mayra M. Ferrari Barbosa
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, SP 05503-900, Brazil; (M.M.F.B.); (A.I.K.)
| | - Alex Issamu Kanno
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, SP 05503-900, Brazil; (M.M.F.B.); (A.I.K.)
| | - Leonardo Paiva Farias
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, BA 40296-710, Brazil;
| | - Mariusz Madej
- Istituto di Biochimica e Biologia Cellulare, Consiglio Nazionale delle Ricerche, 80131 Napoli, Italy; (M.M.); (G.S.)
| | - Gergö Sipos
- Istituto di Biochimica e Biologia Cellulare, Consiglio Nazionale delle Ricerche, 80131 Napoli, Italy; (M.M.); (G.S.)
| | - Silverio Sbrana
- Istituto di Fisiologia Clinica, Consiglio Nazionale delle Ricerche, 54100 Massa, Italy;
| | - Luigina Romani
- Dipartimento di Medicina e Chirurgia, University of Perugia, 06132 Perugia, Italy;
| | - Diana Boraschi
- Istituto di Biochimica e Biologia Cellulare, Consiglio Nazionale delle Ricerche, 80131 Napoli, Italy; (M.M.); (G.S.)
- Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy
| | - Luciana C. C. Leite
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, SP 05503-900, Brazil; (M.M.F.B.); (A.I.K.)
| | - Paola Italiani
- Istituto di Biochimica e Biologia Cellulare, Consiglio Nazionale delle Ricerche, 80131 Napoli, Italy; (M.M.); (G.S.)
- Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy
| |
Collapse
|
34
|
Immunological memory in rheumatic inflammation - a roadblock to tolerance induction. Nat Rev Rheumatol 2021; 17:291-305. [PMID: 33824526 DOI: 10.1038/s41584-021-00601-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2021] [Indexed: 12/20/2022]
Abstract
Why do we still have no cure for chronic inflammatory diseases? One reason could be that current therapies are based on the assumption that chronic inflammation is driven by persistent 'acute' immune reactions. Here we discuss a paradigm shift by suggesting that beyond these reactions, chronic inflammation is driven by imprinted, pathogenic 'memory' cells of the immune system. This rationale is based on the observation that in patients with chronic inflammatory rheumatic diseases refractory to conventional immunosuppressive therapies, therapy-free remission can be achieved by resetting the immune system; that is, by ablating immune cells and regenerating the immune system from stem cells. The success of this approach identifies antigen-experienced and imprinted immune cells as essential and sufficient drivers of inflammation. The 'dark side' of immunological memory primarily involves memory plasma cells secreting pathogenic antibodies and memory T lymphocytes secreting pathogenic cytokines and chemokines, but can also involve cells of innate immunity. New therapeutic strategies should address the persistence of these memory cells. Selective targeting of pathogenic immune memory cells could be based on their specificity, which is challenging, or on their lifestyle, which differs from that of protective immune memory cells, in particular for pathogenic T lymphocytes. The adaptations of such pathogenic memory cells to chronic inflammation offers entirely new therapeutic options for their selective ablation and the regeneration of immunological tolerance.
Collapse
|
35
|
Choudhury RP, Edgar L, Rydén M, Fisher EA. Diabetes and Metabolic Drivers of Trained Immunity. Arterioscler Thromb Vasc Biol 2021; 41:1284-1290. [PMID: 33657881 PMCID: PMC10069665 DOI: 10.1161/atvbaha.120.314211] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Accumulating evidence shows how diverse physiological functions, such as metabolism, immunity, tissue homeostasis, and hematopoiesis, are intricately and profoundly intertwined at multiple levels. This brief review will present evidence from a rapidly expanding field of immunometabolism, highlighting how cells that are relevant to processes at play in determining vascular health and disease can be programmed by changes in their metabolic environment. It will focus on how such changes can be imprinted or trained, particularly through epigenetic modifications, such that adaptations driven by metabolic signals can cause persistent changes in cell function, even after the original stimulus has been corrected or removed. Recognition of these processes and elucidation of the mechanisms underlying them stand to have far-reaching implications for the diagnosis and treatment of diabetes and related metabolic states.
Collapse
Affiliation(s)
- Robin P. Choudhury
- Radcliffe Department of Medicine, University of Oxford, United Kingdom (R.P.C., L.E.)
| | - Laurienne Edgar
- Radcliffe Department of Medicine, University of Oxford, United Kingdom (R.P.C., L.E.)
- Novo Nordisk A/S, Gatwick, United Kingdom (L.E.)
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institute, C2-94, Karolinska University Hospital, Huddinge, Stockholm, Sweden (M.R.)
| | - Edward A. Fisher
- Department of Medicine, NYU Grossman School of Medicine, NY (E.A.F.)
| |
Collapse
|
36
|
Zhou J, Lv J, Carlson C, Liu H, Wang H, Xu T, Wu F, Song C, Wang X, Wang T, Qian Z. Trained immunity contributes to the prevention of Mycobacterium tuberculosis infection, a novel role of autophagy. Emerg Microbes Infect 2021; 10:578-588. [PMID: 33666534 PMCID: PMC8018485 DOI: 10.1080/22221751.2021.1899771] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Mycobacterium tuberculosis (M. tuberculosis) is the pathogen which causes tuberculosis (TB), a significant human public health threat. Co-infection of M. tuberculosis and the human immunodeficiency virus (HIV), emergence of drug resistant M. tuberculosis, and failure to develop highly effective TB vaccines have limited control of the TB epidemic. Trained immunity is an enhanced innate immune response which functions independently of the adaptive/acquired immune system and responds non-specifically to reinfection with invading agents. Recently, several studies have found trained immunity has the capability to control and eliminate M. tuberculosis infection. Over the past decades, however, the consensus was adaptive immunity is the only protective mechanism by which hosts inhibit M. tuberculosis growth. Furthermore, autophagy plays an essential role in the development of trained immunity. Further investigation of trained immunity, M. tuberculosis infection, and the role of autophagy in this process provide new possibilities for vaccine development. In this review, we present the general characteristics of trained immunity and autophagy. We additionally summarize several examples where initiation of trained immunity contributes to the prevention of M. tuberculosis infection and propose future directions for research in this area.
Collapse
Affiliation(s)
- Jie Zhou
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical College, Bengbu, People's Republic of China
| | - Jingzhu Lv
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical College, Bengbu, People's Republic of China
| | - Chelsea Carlson
- Department of Internal Medicine, University of Arizona, Phoenix, AZ, USA
| | - Hui Liu
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical College, Bengbu, People's Republic of China
| | - Hongtao Wang
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical College, Bengbu, People's Republic of China
| | - Tao Xu
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical College, Bengbu, People's Republic of China
| | - Fengjiao Wu
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical College, Bengbu, People's Republic of China
| | - Chuanwang Song
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical College, Bengbu, People's Republic of China
| | - Xiaojing Wang
- Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, Department of Respiration, First Affiliated Hospital, Bengbu Medical College, Bengbu, People's Republic of China
| | - Ting Wang
- Department of Internal Medicine, University of Arizona, Phoenix, AZ, USA
| | - Zhongqing Qian
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical College, Bengbu, People's Republic of China
| |
Collapse
|
37
|
Fulop T, Tripathi S, Rodrigues S, Desroches M, Bunt T, Eiser A, Bernier F, Beauregard PB, Barron AE, Khalil A, Plotka A, Hirokawa K, Larbi A, Bocti C, Laurent B, Frost EH, Witkowski JM. Targeting Impaired Antimicrobial Immunity in the Brain for the Treatment of Alzheimer's Disease. Neuropsychiatr Dis Treat 2021; 17:1311-1339. [PMID: 33976546 PMCID: PMC8106529 DOI: 10.2147/ndt.s264910] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and aging is the most common risk factor for developing the disease. The etiology of AD is not known but AD may be considered as a clinical syndrome with multiple causal pathways contributing to it. The amyloid cascade hypothesis, claiming that excess production or reduced clearance of amyloid-beta (Aβ) and its aggregation into amyloid plaques, was accepted for a long time as the main cause of AD. However, many studies showed that Aβ is a frequent consequence of many challenges/pathologic processes occurring in the brain for decades. A key factor, sustained by experimental data, is that low-grade infection leading to production and deposition of Aβ, which has antimicrobial activity, precedes the development of clinically apparent AD. This infection is chronic, low grade, largely clinically silent for decades because of a nearly efficient antimicrobial immune response in the brain. A chronic inflammatory state is induced that results in neurodegeneration. Interventions that appear to prevent, retard or mitigate the development of AD also appear to modify the disease. In this review, we conceptualize further that the changes in the brain antimicrobial immune response during aging and especially in AD sufferers serve as a foundation that could lead to improved treatment strategies for preventing or decreasing the progression of AD in a disease-modifying treatment.
Collapse
Affiliation(s)
- Tamas Fulop
- Research Center on Aging, Geriatric Division, Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Shreyansh Tripathi
- Cluster Innovation Centre, North Campus, University of Delhi, Delhi, 110007, India.,Ikerbasque, The Basque Foundation for Science, Bilbao, Spain
| | - Serafim Rodrigues
- Ikerbasque, The Basque Foundation for Science, Bilbao, Spain.,Mathematical Computational and Experimental Neuroscience (MCEN), BCAM - The Basque Center for Applied Mathematics, Bilbao, Spain
| | - Mathieu Desroches
- MathNeuro Team, Inria Sophia Antipolis Méditerranée, Sophia Antipolis, France.,Department of Mathematics, Université Côte d'Azur, Nice, France
| | - Ton Bunt
- Izumi Biosciences, Inc., Lexington, MA, USA
| | - Arnold Eiser
- Leonard Davis Institute, University of Pennsylvania, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Francois Bernier
- Morinaga Milk Industry Co., Ltd, Next Generation Science Institute, Kanagawa, Japan
| | - Pascale B Beauregard
- Department of Biology, Faculty of Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Annelise E Barron
- Department of Bioengineering, Stanford School of Medicine, Stanford, CA, USA
| | - Abdelouahed Khalil
- Research Center on Aging, Geriatric Division, Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Adam Plotka
- Department of Pathophysiology, Medical University of Gdansk, Gdansk, Poland
| | - Katsuiku Hirokawa
- Institute of Health and Life Science, Tokyo Med. Dent. University, Tokyo and Nito-Memory Nakanosogo Hospital, Department of Pathology, Tokyo, Japan
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (ASTAR), Immunos Building, Biopolis, Singapore, Singapore
| | - Christian Bocti
- Research Center on Aging, Department of Medicine, Division of Neurology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Benoit Laurent
- Research Center on Aging, Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Eric H Frost
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Jacek M Witkowski
- Department of Pathophysiology, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
38
|
Choi Y, Park C, Kwon D, Lee H, Hong S, Kim GY, Cha HJ, Kim DH, Kim S, Kim HS, Hwang HJ. Immunostimulatory effect of ethanol extract of Chondracanthus tenellus in RAW 264.7 macrophages in vitro. Asian Pac J Trop Biomed 2021. [DOI: 10.4103/2221-1691.314052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
39
|
Karstensen KT, Schein A, Petri A, Bøgsted M, Dybkær K, Uchida S, Kauppinen S. Long Non-Coding RNAs in Diffuse Large B-Cell Lymphoma. Noncoding RNA 2020; 7:1. [PMID: 33379241 PMCID: PMC7838888 DOI: 10.3390/ncrna7010001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 12/12/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoid malignancy in adults. Although significant progress has been made in recent years to treat DLBCL patients, 30%-40% of the patients eventually relapse or are refractory to first line treatment, calling for better therapeutic strategies for DLBCL. Long non-coding RNAs (lncRNAs) have emerged as a highly diverse group of non-protein coding transcripts with intriguing molecular functions in human disease, including cancer. Here, we review the current understanding of lncRNAs in the pathogenesis and progression of DLBCL to provide an overview of the field. As the current knowledge of lncRNAs in DLBCL is still in its infancy, we provide molecular signatures of lncRNAs in DLBCL cell lines to assist further lncRNA research in DLBCL.
Collapse
Affiliation(s)
- Kasper Thystrup Karstensen
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, DK-2450 Copenhagen, Denmark; (K.T.K.); (A.S.); (A.P.)
| | - Aleks Schein
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, DK-2450 Copenhagen, Denmark; (K.T.K.); (A.S.); (A.P.)
| | - Andreas Petri
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, DK-2450 Copenhagen, Denmark; (K.T.K.); (A.S.); (A.P.)
| | - Martin Bøgsted
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, DK-9000 Aalborg, Denmark; (M.B.); (K.D.)
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, DK-9000 Aalborg, Denmark
| | - Karen Dybkær
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, DK-9000 Aalborg, Denmark; (M.B.); (K.D.)
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, DK-9000 Aalborg, Denmark
| | - Shizuka Uchida
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, DK-2450 Copenhagen, Denmark; (K.T.K.); (A.S.); (A.P.)
| | - Sakari Kauppinen
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, DK-2450 Copenhagen, Denmark; (K.T.K.); (A.S.); (A.P.)
| |
Collapse
|
40
|
Fernández-García V, González-Ramos S, Martín-Sanz P, Castrillo A, Boscá L. Contribution of Extramedullary Hematopoiesis to Atherosclerosis. The Spleen as a Neglected Hub of Inflammatory Cells. Front Immunol 2020; 11:586527. [PMID: 33193412 PMCID: PMC7649205 DOI: 10.3389/fimmu.2020.586527] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/06/2020] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular diseases (CVDs) incidence is becoming higher. This fact is promoted by metabolic disorders such as obesity, and aging. Atherosclerosis is the underlying cause of most of these pathologies. It is a chronic inflammatory disease that begins with the progressive accumulation of lipids and fibrotic materials in the blood-vessel wall, which leads to massive leukocyte recruitment. Rupture of the fibrous cap of the atherogenic cusps is responsible for tissue ischemic events, among them myocardial infarction. Extramedullary hematopoiesis (EMH), or blood cell production outside the bone marrow (BM), occurs when the normal production of these cells is impaired (chronic hematological and genetic disorders, leukemia, etc.) or is altered by metabolic disorders, such as hypercholesterolemia, or after myocardial infarction. Recent studies indicate that the main EMH tissues (spleen, liver, adipose and lymph nodes) complement the hematopoietic function of the BM, producing circulating inflammatory cells that infiltrate into the atheroma. Indeed, the spleen, which is a secondary lymphopoietic organ with high metabolic activity, contains a reservoir of myeloid progenitors and monocytes, constituting an important source of inflammatory cells to the atherosclerotic lesion. Furthermore, the spleen also plays an important role in lipid homeostasis and immune-cell selection. Interestingly, clinical evidence from splenectomized subjects shows that they are more susceptible to developing pathologies, such as dyslipidemia and atherosclerosis due to the loss of immune selection. Although CVDs represent the leading cause of death worldwide, the mechanisms involving the spleen-atherosclerosis-heart axis cross-talk remain poorly characterized.
Collapse
Affiliation(s)
- Victoria Fernández-García
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Silvia González-Ramos
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Paloma Martín-Sanz
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Antonio Castrillo
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
- Unidad de Biomedicina, (Unidad Asociada al CSIC), Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM) and Universidad de Las Palmas, Gran Canaria, Spain
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias, Grupo de Investigación Medio Ambiente y Salud, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Unidad de Biomedicina, (Unidad Asociada al CSIC), Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM) and Universidad de Las Palmas, Gran Canaria, Spain
| |
Collapse
|
41
|
Paris S, Chapat L, Martin-Cagnon N, Durand PY, Piney L, Cariou C, Bergamo P, Bonnet JM, Poulet H, Freyburger L, De Luca K. β-Glucan as Trained Immunity-Based Adjuvants for Rabies Vaccines in Dogs. Front Immunol 2020; 11:564497. [PMID: 33162977 PMCID: PMC7580252 DOI: 10.3389/fimmu.2020.564497] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/14/2020] [Indexed: 12/16/2022] Open
Abstract
The mechanisms of trained immunity have been extensively described in vitro and the beneficial effects are starting to be deciphered in in vivo settings. Prototypical compounds inducing trained immunity, such as β-glucans, act through epigenetic reprogramming and metabolic changes of innate immune cells. The recent advances in this field have opened new areas for the development of Trained immunity-based adjuvants (TIbAs). In this study, we assessed in dogs the potential immune training effects of β-glucans as well as their capacity to enhance the adaptive immune response of an inactivated rabies vaccine (Rabisin®). Injection of β-glucan from Euglena gracilis was performed 1 month before vaccination with Rabisin® supplemented or not with the same β-glucan used as adjuvant. Trained innate immunity parameters were assessed during the first month of the trial. The second phase of the study was focused on the ability of β-glucan to enhance adaptive immune responses measured by multiple immunological parameters. B and T-cell specific responses were monitored to evaluate the immunogenicity of the rabies vaccine adjuvanted with β-glucan or not. Our preliminary results support that adjuvantation of Rabisin® vaccine with β-glucan elicit a higher B-lymphocyte immune response, the prevailing factor of protection against rabies. β-glucan also tend to stimulate the T cell response as shown by the cytokine secretion profile of PBMCs re-stimulated ex vivo. Our data are providing new insights on the impact of trained immunity on the adaptive immune response to vaccines in dogs. The administration of β-glucan, 1 month before or simultaneously to Rabisin® vaccination give promising results for the generation of new TIbA candidates and their potential to provide increased immunogenicity of specific vaccines.
Collapse
Affiliation(s)
- Simon Paris
- Boehringer Ingelheim Animal Health, R&D, Lyon, France
- Université de Lyon, APCSe, Pulmonary and Cardiovascular Agression in Sepsis, VetAgro Sup-Campus Vétérinaire de Lyon, Marcy l’Etoile, France
- Département Biologie, Faculté des Sciences et Techniques, Université Claude Bernard Lyon 1, Villeurbanne, France
| | | | | | | | | | - Carine Cariou
- Boehringer Ingelheim Animal Health, R&D, Lyon, France
| | | | - Jeanne-Marie Bonnet
- Université de Lyon, APCSe, Pulmonary and Cardiovascular Agression in Sepsis, VetAgro Sup-Campus Vétérinaire de Lyon, Marcy l’Etoile, France
| | - Hervé Poulet
- Boehringer Ingelheim Animal Health, R&D, Lyon, France
| | - Ludovic Freyburger
- Université de Lyon, APCSe, Pulmonary and Cardiovascular Agression in Sepsis, VetAgro Sup-Campus Vétérinaire de Lyon, Marcy l’Etoile, France
| | | |
Collapse
|
42
|
Italiani P, Della Camera G, Boraschi D. Induction of Innate Immune Memory by Engineered Nanoparticles in Monocytes/Macrophages: From Hypothesis to Reality. Front Immunol 2020; 11:566309. [PMID: 33123137 PMCID: PMC7573069 DOI: 10.3389/fimmu.2020.566309] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/24/2020] [Indexed: 12/11/2022] Open
Abstract
The capacity of engineered nanoparticles to activate cells of the innate immune system, in particular monocytes and macrophages, is considered at the basis of their toxic/inflammatory effects. It is, however, evident that even nanoparticles that do not directly induce inflammatory activation, and are therefore considered as safe, can nevertheless induce epigenetic modifications and affect metabolic pathways in monocytes and macrophages. Since epigenetic and metabolic changes are the main mechanisms of innate memory, we had previously proposed that nanoparticles can induce/modulate innate memory, that is, have the ability of shaping the secondary response to inflammatory challenges. In light of new data, it is now possible to support the original hypothesis and show that different types of nanoparticles can both directly induce innate memory, priming macrophages for a more potent response to subsequent stimuli, and modulate bacteria-induced memory by attenuating the priming-induced enhancement. This evidence raises two important issues. First, in addition to overt toxic/inflammatory effects, we should consider evaluating the capacity to induce innate memory and the related epigenetic and metabolic changes in the immunosafety assessment of nanomaterials, since modulation of innate memory may be at the basis of long-term unwanted immunological effects. The other important consideration is that this capacity of nanomaterials could open a new avenue in immunomodulation and the possibility of using engineered nanomaterials for improving immune responses to vaccines and resistance to infections, and modulate anomalous immune/inflammatory reactions in chronic inflammatory diseases, autoimmunity, and a range of other immune-related pathologies.
Collapse
Affiliation(s)
- Paola Italiani
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Naples, Italy
| | - Giacomo Della Camera
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Naples, Italy
| | - Diana Boraschi
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Naples, Italy
| |
Collapse
|
43
|
Csaba G. Possible contribution of trained immunity in faulty hormonal imprinting and DOHaD: Review and hypothesis. Acta Microbiol Immunol Hung 2020; 67:143-147. [PMID: 32997645 DOI: 10.1556/030.2020.01178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 04/18/2020] [Indexed: 11/19/2022]
Abstract
The faulty hormonal imprinting theory (published in 1980) and the DOHaD (Developmental Origin of Health and Disease theory (published in 1986) are twin-concepts: both justify the manifestation after long time (in adults) diseases which had been provoked in differentiating cells (e.g. during gestation). This was demonstrated using animal experiments as well, as comparative statistical methods (in human cases). However, there is no explanation for the tools of memorization (even after decades) of the early adversity and the tools of execution (manifestation) in adult age. It seems likely that immune memory is involved to the memorization of early adversity, up to the manifestation of the result (non-communicable diseases). Nevertheless, the relatively short timespan of adaptive immune memory makes this system insuitable for this function, however the newly recognized trained memory of the innate immune system seems to be theoretically suitable for the storage of the records and handling the sequalae, which is the epigenetic reprogramming in the time of provocation, without changes in base sequences (mutation). The flawed (damaged) program is manifested later, in adult age. Evidences are incomplete, so further animal experiments and human observations are needed for justifying the theory.
Collapse
Affiliation(s)
- György Csaba
- Department of Genetics, Cell-and Immunobiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
44
|
D'Agostino MR, Lai R, Afkhami S, Khera A, Yao Y, Vaseghi-Shanjani M, Zganiacz A, Jeyanathan M, Xing Z. Airway Macrophages Mediate Mucosal Vaccine-Induced Trained Innate Immunity against Mycobacterium tuberculosis in Early Stages of Infection. THE JOURNAL OF IMMUNOLOGY 2020; 205:2750-2762. [PMID: 32998983 DOI: 10.4049/jimmunol.2000532] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/07/2020] [Indexed: 12/20/2022]
Abstract
Mycobacterium tuberculosis, the causative agent of pulmonary tuberculosis (TB), is responsible for millions of infections and deaths annually. Decades of TB vaccine development have focused on adaptive T cell immunity, whereas the importance of innate immune contributions toward vaccine efficacy has only recently been recognized. Airway macrophages (AwM) are the predominant host cell during early pulmonary M. tuberculosis infection and, therefore, represent attractive targets for vaccine-mediated immunity. We have demonstrated that respiratory mucosal immunization with a viral-vectored vaccine imprints AwM, conferring enhanced protection against heterologous bacterial challenge. However, it is unknown if innate immune memory also protects against M. tuberculosis In this study, by using a murine model, we detail whether respiratory mucosal TB vaccination profoundly alters the airway innate immune landscape associated with AwM prior to M. tuberculosis exposure and whether such AwM play a critical role in host defense against M. tuberculosis infection. Our study reveals an important role of AwM in innate immune protection in early stages of M. tuberculosis infection in the lung.
Collapse
Affiliation(s)
- Michael R D'Agostino
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4L8, Canada; and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Rocky Lai
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4L8, Canada; and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Sam Afkhami
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4L8, Canada; and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Amandeep Khera
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4L8, Canada; and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Yushi Yao
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4L8, Canada; and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Maryam Vaseghi-Shanjani
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4L8, Canada; and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Anna Zganiacz
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4L8, Canada; and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Mangalakumari Jeyanathan
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4L8, Canada; and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Zhou Xing
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4L8, Canada; and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| |
Collapse
|
45
|
Campit SE, Meliki A, Youngson NA, Chandrasekaran S. Nutrient Sensing by Histone Marks: Reading the Metabolic Histone Code Using Tracing, Omics, and Modeling. Bioessays 2020; 42:e2000083. [PMID: 32638413 PMCID: PMC11426192 DOI: 10.1002/bies.202000083] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/23/2020] [Indexed: 12/19/2022]
Abstract
Several metabolites serve as substrates for histone modifications and communicate changes in the metabolic environment to the epigenome. Technologies such as metabolomics and proteomics have allowed us to reconstruct the interactions between metabolic pathways and histones. These technologies have shed light on how nutrient availability can have a dramatic effect on various histone modifications. This metabolism-epigenome cross talk plays a fundamental role in development, immune function, and diseases like cancer. Yet, major challenges remain in understanding the interactions between cellular metabolism and the epigenome. How the levels and fluxes of various metabolites impact epigenetic marks is still unclear. Discussed herein are recent applications and the potential of systems biology methods such as flux tracing and metabolic modeling to address these challenges and to uncover new metabolic-epigenetic interactions. These systems approaches can ultimately help elucidate how nutrients shape the epigenome of microbes and mammalian cells.
Collapse
Affiliation(s)
- Scott E. Campit
- Program in Chemical Biology, University of Michigan, Ann Arbor, MI, USA 48109
| | - Alia Meliki
- Center for Bioinformatics and Computational Medicine, Ann Arbor, MI, USA 48109
| | - Neil A. Youngson
- Institute of Hepatology, Foundation for Liver Research, London, UK
- Faculty of Life Sciences and Medicine, King’s College London, London, UK
- School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Sriram Chandrasekaran
- Program in Chemical Biology, University of Michigan, Ann Arbor, MI, USA 48109
- Center for Bioinformatics and Computational Medicine, Ann Arbor, MI, USA 48109
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA 48109
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA 48109
| |
Collapse
|
46
|
Park C, HwangBo H, Lee H, Kim GY, Cha HJ, Choi SH, Kim S, Kim HS, Yun SJ, Kim WJ, Jeon YJ, Choi YH. The immunostimulatory effect of indole-6-carboxaldehyde isolated from Sargassum thunbergii (Mertens) Kuntze in RAW 264.7 macrophages. Anim Cells Syst (Seoul) 2020; 24:233-241. [PMID: 33029301 PMCID: PMC7473310 DOI: 10.1080/19768354.2020.1808529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Indole-6-carboxaldehyde (I6CA), an indole derivative isolated from the marine brown algae Sargassum thunbergii, is known to have several beneficial effects, but no studies on immune regulation have been conducted. In this study, the immunomodulatory properties of I6CA on murine RAW 264.7 monocyte/macrophage cells were evaluated. As the concentration of I6CA increased, the morphology of RAW 264.7 cells changed to a typical active macrophage shape, and the phagocytic activity increased significantly. I6CA effectively enhanced the production and secretion of immunomodulatory mediators and cytokines due to increased expression of their respective genes. Additionally, I6CA markedly stimulated the expression of Toll-like receptor 4 (TLR4) and its adapter molecule, myeloid differentiation factor 88 (Myd88), and increased TLR4 complexed with Myd88. Furthermore, I6CA promoted the nuclear translocation of nuclear factor-kappa B (NF-κB) by increasing the degradation of the inhibitor of NF-κB-α. Meanwhile, similar trends were also found in lipopolysaccharide-treated cells as a positive control. Furthermore, molecular docking simulation showed that I6CA interacted with TLR4-myeloid differentiation 2 complex. Taken together, the results support the concept that I6CA may increase the activity of the TLR4/NF-κB signaling pathway in order to enhance the immunomodulatory activity of RAW 264.7 cells.
Collapse
Affiliation(s)
- Cheol Park
- Division of Basic Sciences, College of Liberal Studies, Dong-eui University, Busan, Republic of Korea
| | - Hyun HwangBo
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea.,Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan, Republic of Korea
| | - Hyesook Lee
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea.,Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju, Republic of Korea
| | - Hee-Jae Cha
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan, Republic of Korea
| | - Sung Hyun Choi
- Department of System Management, Korea Lift College, Geochang, Republic of Korea
| | - Suhkmann Kim
- Department of Chemistry, College of Natural Sciences, Pusan National University, Busan, Republic of Korea
| | - Heui-Soo Kim
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan, Republic of Korea
| | - Seok Joong Yun
- Department of Urology, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Wun-Jae Kim
- Department of Urology, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - You-Jin Jeon
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju, Republic of Korea
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea.,Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan, Republic of Korea
| |
Collapse
|