1
|
Lee KM, Kim KH. Long-term and non-specific immune memory induced by muramyl dipeptide and its protective effect against Vibrio anguillarum in rainbow trout (Oncorhynchus mykiss). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 167:105386. [PMID: 40334803 DOI: 10.1016/j.dci.2025.105386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/12/2025] [Accepted: 05/03/2025] [Indexed: 05/09/2025]
Abstract
Muramyl dipeptide (MDP), a minimal bioactive peptidoglycan motif derived from the bacterial cell wall, is known to trigger a wide range of signaling cascades related to inflammatory and antibacterial responses, thereby enhancing the antimicrobial function of various innate immune cells including monocytes, macrophages, neutrophils, natural killer (NK) cells, and lymphocytes. To date, several studies have demonstrated that MDP has the capacity to stimulate non-specific immunity in fish. However, the long-term effect of MDP on fish remains unclear. In this study, we aimed to investigate the long-term protective effect of MDP against Vibrio anguillarum in rainbow trout (Oncorhynchus mykiss). MDP was administered to the rainbow trout by intraperitoneal (IP) injection, and the bacterial challenge was conducted at 4 weeks post-administration. The MDP-injected fish exhibited significantly higher survival rates than fish injected with PBS. Following bacterial infection, significantly reduced bacterial loads were shown in the head kidney of MDP-injected fish, accompanied by elevated expression levels of TNF-α, IL-1β, IL-6, IL-12, and HIF-1α. Furthermore, a significant increase of acetylation of histone 3 at lysine 27 (H3K27ac) was evident at the promoter regions of TNF-α and IL-1β in the fish of the MDP group at 4 weeks post-administration. These results suggest that MDP-induced histone acetylation in immune-related genes' promoters enhanced immune gene expression upon infection, possibly contributing to the observed long-term antibacterial effect and protection against V. anguillarum.
Collapse
Affiliation(s)
- Kyung Min Lee
- Department of Aquatic Life Medicine, Pukyong National University, Busan, 48513, South Korea
| | - Ki Hong Kim
- Department of Aquatic Life Medicine, Pukyong National University, Busan, 48513, South Korea.
| |
Collapse
|
2
|
Lian X, Wang W, Li Y, Zhou X, Li X, Zuo J, Song L, Wang L, Song L. Histone lysine demethylase 5 regulates haemocyte proliferation during immune priming in the oyster Crassostreagigas. FISH & SHELLFISH IMMUNOLOGY 2025; 160:110236. [PMID: 40010616 DOI: 10.1016/j.fsi.2025.110236] [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: 11/27/2024] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 02/28/2025]
Abstract
Histone lysine demethylase 5 (KDM5) is a ketoglutarate-dependent dioxygenase in histone lysine demethylation, playing a vital role in immunological memory by modulating H3K4me3. Investigating on the role of invertebrate KDM5 in the immune priming, a novel form of immunological memory recently verified in invertebrates, will further our knowledge of epigenetic regulation for innate immune memory. In the present study, a KDM5A was identified in Pacific oyster Crassostrea gigas, and its role in regulating haemocyte proliferation during immune priming was assessed. The deduced amino acid sequence of CgKDM5A harbors a complete JmjC domain featuring a conserved αKG binding site. The mRNA expression of CgKDM5A in the haemolymph was significantly higher than that in the tested tissues of the mature oysters. After Vibrio splendidus stimulation, CgKDM5A transcripts and KDM5 enzymatic activity in haemocytes significantly decreased, accompanying with increased H3K4me3 levels. Moreover, H3K4me3 modifications at the CgBMP7 and CgGATA2/3 promoters were elevated at 7 d after V. splendidus stimulation (p < 0.05), and the haemocyte proliferation index increased significantly at 12 h after the secondary stimulation (p < 0.05). Treatment with KDM5 activator DM-αKG further led to a significant increase in H3K4me3 enrichment levels at the CgBMP7 and CgGATA2/3 promoters at 7 d after the primary stimulation (p < 0.05). Subsequently, the expression of CgBMP7 and CgGATA2/3, as well as the haemocyte proliferation index decreased significantly after the secondary stimulation (p < 0.05). In contrast, CgKDM5A-RNAi oysters exhibited an enriched H3K4me3 at the CgBMP7 and CgGATA2/3 promoters at 7 d after the primary stimulation and an increased haemocyte proliferation index at 12 h after the secondary stimulation (p < 0.05). These findings suggest that CgKDM5A plays a critical role in haemocyte proliferation by affect H3K4me3 enrichment at the CgBMP7 and CgGATA2/3 promoters during immune priming in C. gigas, highlighting the potential of epigenetic regulation in innate immune memory of mollusks.
Collapse
Affiliation(s)
- Xingye Lian
- School of Life Science, Liaoning Normal University, Dalian, 116029, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, 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; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, 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; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Xiaoxu Zhou
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Xuesong Li
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, 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; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Lingyuan Song
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, 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; Functional Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, 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; Functional Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| |
Collapse
|
3
|
Russell DG, Simwela NV, Mattila JT, Flynn J, Mwandumba HC, Pisu D. How macrophage heterogeneity affects tuberculosis disease and therapy. Nat Rev Immunol 2025; 25:370-384. [PMID: 39774813 DOI: 10.1038/s41577-024-01124-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2024] [Indexed: 01/11/2025]
Abstract
Macrophages are the primary host cell type for infection by Mycobacterium tuberculosis in vivo. Macrophages are also key immune effector cells that mediate the control of bacterial growth. However, the specific macrophage phenotypes that are required for optimal immune control of M. tuberculosis infection in vivo remain poorly defined. There are two distinct macrophage lineages in the lung, comprising embryonically derived, tissue-resident alveolar macrophages and recruited, blood monocyte-derived interstitial macrophages. Recent studies have shown that these lineages respond divergently to similar immune environments within the tuberculosis granuloma. Here, we discuss how the differing responses of macrophage lineages might affect the control or progression of tuberculosis disease. We suggest that the ability to reprogramme macrophage responses appropriately, through immunological or chemotherapeutic routes, could help to optimize vaccines and drug regimens for tuberculosis.
Collapse
Affiliation(s)
- David G Russell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.
| | - Nelson V Simwela
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Joshua T Mattila
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - JoAnne Flynn
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Henry C Mwandumba
- Malawi Liverpool Wellcome Research Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Davide Pisu
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
- Department of Microbial Pathogenesis and Immunology, Texas A&M School of Medicine, Bryan, TX, USA
| |
Collapse
|
4
|
Monreal-Escalante E, Angulo M, Ramos-Vega A, Trujillo E, Angulo C. Plant-made trained immunity-based vaccines: Beyond one approach. Int J Pharm 2025; 675:125572. [PMID: 40204041 DOI: 10.1016/j.ijpharm.2025.125572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/14/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
Plant-made vaccines and trained immunity-based vaccines (TIbV or TRAIMbV) represent two strategies for enhancing immunity against diseases. Plants provide an effective and cost-efficient vaccine production platform, while TIbV induces innate immune memory that can protect against both homologous and heterologous diseases. Both strategies are generally compatible; however, they have not been explored in a transdisciplinary manner. Despite their strengths in vaccinology, each faces limitations that hinder widespread adoption and health benefits. This review revisits both strategies, discussing their fundamental knowledge alongside practical and experimental examples, ultimately highlighting their limitations and perspectives to pave the way for a unified approach to combat diseases. Future scenarios are envisioned and presented if research on plant-made trained immunity-based vaccines is adopted.
Collapse
Affiliation(s)
- Elizabeth Monreal-Escalante
- Immunology & Vaccinology Group and Laboratorio Nacional CONAHCYT (SECIHTI) de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD). Centro de Investigaciones Biológicas del Noroeste, S.C. Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico; SECIHTI-Centro de Investigaciones Biológicas del Noroeste, S.C. Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico
| | - Miriam Angulo
- Immunology & Vaccinology Group and Laboratorio Nacional CONAHCYT (SECIHTI) de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD). Centro de Investigaciones Biológicas del Noroeste, S.C. Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico
| | - Abel Ramos-Vega
- Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada (CICATA) Unidad Morelos del Instituto Politécnico Nacional (IPN), Dirección: Boulevard de la Tecnología No.1036, Código Postal 62790 Xochitepec, Morelos, Mexico
| | - Edgar Trujillo
- Immunology & Vaccinology Group and Laboratorio Nacional CONAHCYT (SECIHTI) de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD). Centro de Investigaciones Biológicas del Noroeste, S.C. Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico
| | - Carlos Angulo
- Immunology & Vaccinology Group and Laboratorio Nacional CONAHCYT (SECIHTI) de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD). Centro de Investigaciones Biológicas del Noroeste, S.C. Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico.
| |
Collapse
|
5
|
Liatsos GD, Mariolis I, Hadziyannis E, Bamias A, Vassilopoulos D. Review of BCG immunotherapy for bladder cancer. Clin Microbiol Rev 2025; 38:e0019423. [PMID: 39932308 PMCID: PMC11905372 DOI: 10.1128/cmr.00194-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025] Open
Abstract
SUMMARYFor several decades, intravesical Bacillus Calmette-Guérin (iBCG) immunotherapy has been the gold standard adjuvant treatment for high-risk and selected intermediate-risk patients with non-muscle-invasive bladder cancer (NMIBC). In this review, the mechanisms of iBCG immune-mediated anti-cancer activity and resistance are presented. Furthermore, a literature review of short-term and systemic iBCG-related side effects was performed. A high incidence (75.5%) of iBCG-related short-term, self-limiting adverse events was observed, while more severe iBCG-related local/systemic complications (iBCG-rL/SCs) that required medical treatment or hospitalization occurred at a lower rate (2.35%). Disseminated was the most common form of iBCG-rSCs, while two-thirds of the cases were classified as infectious. The implementation of molecular-based techniques resulted in significantly higher diagnostic rates. Anti-tuberculous treatment (ATT) is the mainstay of treatment, while in patients with any iBCG-rL/SC form involving the vasculature, ATT should be combined with surgery. Local and osteoarticular forms have the lowest mortality, but their management necessitates severe and debilitating surgical procedures. The overall iBCG-attributed mortality in patients with iBCG-rL/SC was 7.4%, with disseminated, vascular, and lung involvements exhibiting the highest rates. Given the global shortage of BCG for the last two decades, as well as the paucity of effective options for iBCG-refractory or relapsing NMIBC patients, new therapeutic strategies are being tested with promising early results.
Collapse
Affiliation(s)
- George D. Liatsos
- 2nd Department of Medicine and Laboratory, National and Kapodistrian University of Athens, School of Medicine, General Hospital of Athens "Hippokration", Athens, Greece
| | - Ilias Mariolis
- 2nd Department of Medicine and Laboratory, National and Kapodistrian University of Athens, School of Medicine, General Hospital of Athens "Hippokration", Athens, Greece
| | - Emilia Hadziyannis
- 2nd Department of Medicine and Laboratory, National and Kapodistrian University of Athens, School of Medicine, General Hospital of Athens "Hippokration", Athens, Greece
| | - Aristotelis Bamias
- 2nd Propaedeutic Department of Internal Medicine, National and Kapodistrian University of Athens, School of Medicine, Attikon University General Hospital, Athens, Greece
| | - Dimitrios Vassilopoulos
- 2nd Department of Medicine and Laboratory, National and Kapodistrian University of Athens, School of Medicine, General Hospital of Athens "Hippokration", Athens, Greece
| |
Collapse
|
6
|
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
|
7
|
Wang X, Yu G. Advancing veterinary vaccines design through trained immunity insights. Front Vet Sci 2025; 11:1524668. [PMID: 39881716 PMCID: PMC11776093 DOI: 10.3389/fvets.2024.1524668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Trained immunity, characterized by long-term functional reprogramming of innate immune cells, offers promising new directions for veterinary vaccine development. This perspective examines how trained immunity can be integrated into veterinary vaccine design through metabolic reprogramming and epigenetic modifications. We analyze key molecular mechanisms, including the shift to aerobic glycolysis and sustained epigenetic changes, that enable enhanced immune responses. Strategic approaches for vaccine optimization are proposed, focusing on selecting effective trained immunity inducers, developing innovative adjuvant systems, and achieving synergistic enhancement of immune responses. While implementation challenges exist, including individual response variations and safety considerations, trained immunity-based vaccines show potential for providing broader protection against emerging pathogens. This approach could revolutionize veterinary vaccinology by offering enhanced efficacy and cross-protection against heterologous infections, particularly valuable for zoonotic disease control.
Collapse
Affiliation(s)
- Xin Wang
- College of Life Science, Longyan University, Longyan, China
- Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnolog, Longyan, China
- Key Laboratory of Preventive Veterinary Medicine and Biotechnology, Longyan University, Longyan, China
- Chinese International College, Dhurakij Pundit University, Bangkok, Thailand
| | - Guohua Yu
- College of Life Science, Longyan University, Longyan, China
- Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnolog, Longyan, China
- Key Laboratory of Preventive Veterinary Medicine and Biotechnology, Longyan University, Longyan, China
| |
Collapse
|
8
|
Szabó K, Balogh F, Romhányi D, Erdei L, Toldi B, Gyulai R, Kemény L, Groma G. Epigenetic Regulatory Processes Involved in the Establishment and Maintenance of Skin Homeostasis-The Role of Microbiota. Int J Mol Sci 2025; 26:438. [PMID: 39859154 PMCID: PMC11764776 DOI: 10.3390/ijms26020438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
Epigenetic mechanisms are central to the regulation of all biological processes. This manuscript reviews the current understanding of diverse epigenetic modifications and their role in the establishment and maintenance of normal skin functions. In healthy skin, these mechanisms allow for the precise control of gene expression, facilitating the dynamic balance between cell proliferation and differentiation necessary for effective barrier function. Furthermore, as the skin ages, alterations in epigenetic marks can lead to impaired regenerative capacity and increased susceptibility to environmental stressors. The interaction between skin microbiota and epigenetic regulation will also be explored, highlighting how microbial communities can influence skin health by modulating the host gene expression. Future research should focus on the development of targeted interventions to promote skin development, resilience, and longevity, even in an ever-changing environment. This underscores the need for integrative approaches to study these complex regulatory networks.
Collapse
Affiliation(s)
- Kornélia Szabó
- HUN-REN-SZTE Dermatological Research Group, 6720 Szeged, Hungary (L.K.); (G.G.)
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
- HCEMM-USZ Skin Research Group, 6720 Szeged, Hungary
| | - Fanni Balogh
- HUN-REN-SZTE Dermatological Research Group, 6720 Szeged, Hungary (L.K.); (G.G.)
| | - Dóra Romhányi
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| | - Lilla Erdei
- HUN-REN-SZTE Dermatological Research Group, 6720 Szeged, Hungary (L.K.); (G.G.)
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
- HCEMM-USZ Skin Research Group, 6720 Szeged, Hungary
| | - Blanka Toldi
- HUN-REN-SZTE Dermatological Research Group, 6720 Szeged, Hungary (L.K.); (G.G.)
| | - Rolland Gyulai
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| | - Lajos Kemény
- HUN-REN-SZTE Dermatological Research Group, 6720 Szeged, Hungary (L.K.); (G.G.)
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
- HCEMM-USZ Skin Research Group, 6720 Szeged, Hungary
| | - Gergely Groma
- HUN-REN-SZTE Dermatological Research Group, 6720 Szeged, Hungary (L.K.); (G.G.)
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| |
Collapse
|
9
|
Angulo M, Angulo C. Trained immunity-based vaccines: A vision from the one health initiative. Vaccine 2025; 43:126505. [PMID: 39520776 DOI: 10.1016/j.vaccine.2024.126505] [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: 08/16/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Trained immunity-based vaccines (TIbV or TRIMbV) represent a novel approach to combating infectious diseases. The innate immune system in animals, including humans, exhibits "memory-like" functions. Remarkably, the immunological mechanisms -both epigenetic and metabolic-) underlying this memory enables immune cells to develop defensive and protective outcomes against unspecific pathogenic infections. Under this context, the One Health initiative promotes integrative efforts to combat zoonotic (and anthropozoonotic) diseases, which is critical because 3 of 4 animal infections are transmitted to humans. Therefore, TIbV constitutes a potential affordable approach to control zoonotic pathologies, especially under pandemic scenarios. This review describes the state-of-the-art TIbV and their hurdles, opportunities, and prospects for the One Health initiative to prevent, control, and treat infectious diseases.
Collapse
Affiliation(s)
- Miriam Angulo
- Immunology & Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico.; Laboratorio Nacional CONAHCYT de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD), Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C. S., C.P. 23096, Mexico
| | - Carlos Angulo
- Immunology & Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico.; Laboratorio Nacional CONAHCYT de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD), Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C. S., C.P. 23096, Mexico.
| |
Collapse
|
10
|
Ali A, Azmat U, Ji Z, Khatoon A, Murtaza B, Akbar K, Irshad U, Raza R, Su Z. Beyond Genes: Epiregulomes as Molecular Commanders in Innate Immunity. Int Immunopharmacol 2024; 142:113149. [PMID: 39278059 DOI: 10.1016/j.intimp.2024.113149] [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: 06/02/2024] [Revised: 08/09/2024] [Accepted: 09/08/2024] [Indexed: 09/17/2024]
Abstract
The natural fastest way to deal with pathogens or danger signals is the innate immune system. This system prevents too much inflammation and tissue damage and efficiently eliminates pathogens. The epiregulome is the chromatin structure influenced by epigenetic factors and linked to cis-regulatory elements (CREs). The epiregulome helps to end the inflammatory response and also assists innate immune cells to show specific action by making cell-specific gene expression patterns. This inspection unfolds two concepts: (1) how epiregulomes are shaped by switching the expression levels of genes, manoeuvre enzyme activity and earmark of chromatin modifiers on specific genes; during and after the infection, and (2) how the expression of specific genes (aids in prompt management of innate cell growth, or the reaction to aggravation and illness) command by epiregulomes that formed during the above process. In this review, the consequences of intrinsic immuno-metabolic remodelling on epiregulomes and potential difficulties in identifying the master epiregulome that regulates innate immunity and inflammation have been discussed.
Collapse
Affiliation(s)
- Ashiq Ali
- Department of Histology and Embryology, Shantou University Medical College, China.
| | - Urooj Azmat
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, Pakistan
| | - Ziyi Ji
- Department of Histology and Embryology, Shantou University Medical College, China
| | - Aisha Khatoon
- Department of Pathology, University of Agriculture Faisalabad, Pakistan
| | - Bilal Murtaza
- School of Bioengineering, Dalian University of Science and Technology, Dalian, China
| | - Kaynaat Akbar
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, Pakistan
| | - Urooj Irshad
- Department Biological Sciences, Faculty of Sciences, Superior University Lahore, Punjab, Pakistan
| | - Rameen Raza
- Department of Pathology, University of Agriculture Faisalabad, Pakistan
| | - Zhongjing Su
- Department of Histology and Embryology, Shantou University Medical College, China.
| |
Collapse
|
11
|
Suárez LJ, González-Duarte W, Torrez-Velasco R, Salinas V, Roa-Molina NS, Muñoz SM, Rodríguez LS, Arce RM, Shibli JA, Rodríguez-Ciodaro A. Increased Resolvin E1 Production by Peripheral Blood Mononuclear Cells in Periodontitis Patients: Pilot Study. Braz Dent J 2024; 35:e245861. [PMID: 39476108 PMCID: PMC11506237 DOI: 10.1590/0103-6440202405861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/06/2024] [Indexed: 11/03/2024] Open
Abstract
This study quantified the production of the pro-resolving agent Resolvin E1 by peripheral blood mononuclear cells (PBMC) from 20 systemically healthy volunteers with and without periodontitis after stimulation with lipopolysaccharide (LPS) from Porphyromonas gingivalis (Pg). Ten periodontitis patients and 10 healthy volunteers (30-50 years old), matched by age and sex, were recruited. Peripheral blood mononuclear cells were isolated and stimulated in culture plates for 24 hours with Pg LPS. Resolvin E1 levels were measured in the supernatants by enzyme-linked immunosorbent assay. Significantly higher production of Resolvin E1 was observed in both groups when stimulated with LPS compared to baseline levels (p<0.001). A significant increase in Resolvin E1 was observed in the presence of Lipopolysaccharide in the patients with periodontitis compared to the healthy group (p=0.0019). Resolvin E1 levels may reflect a measure of resolution of inflammation that warrants further clinical investigation.
Collapse
Affiliation(s)
- Lina J Suárez
- Universidad Nacional de Colombia, Departamento de ciencias básicas y medicina oral, Facultad de Odontología, Bogotá, Colombia
| | - Wilmer González-Duarte
- Universidad Nacional de Colombia, Departamento de ciencias básicas y medicina oral, Facultad de Odontología, Bogotá, Colombia
| | - Rodrigo Torrez-Velasco
- Universidad Nacional de Colombia, Departamento de ciencias básicas y medicina oral, Facultad de Odontología, Bogotá, Colombia
| | - Viviana Salinas
- Universidad Nacional de Colombia, Departamento de ciencias básicas y medicina oral, Facultad de Odontología, Bogotá, Colombia
| | - Nelly S Roa-Molina
- Pontificia Universidad Javeriana, Centro de investigaciones odontológicas, Facultad de Odontología, Bogotá, Colombia
| | - Sindy M Muñoz
- Pontificia Universidad Javeriana, Instituto de genética Humana, Facultad de Medicina, Bogotá, Colombia
| | - Luz-Stella Rodríguez
- Pontificia Universidad Javeriana, Instituto de genética Humana, Facultad de Medicina, Bogotá, Colombia
| | - Roger M Arce
- Department of Periodontics and Dental Hygiene, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jamil A Shibli
- Guarulhos University, Department of Periodontology, Dental Research Division, Guarulhos, Brasil
| | - Adriana Rodríguez-Ciodaro
- Pontificia Universidad Javeriana, Centro de investigaciones odontológicas, Facultad de Odontología, Bogotá, Colombia
| |
Collapse
|
12
|
Du P, Li J, Hua M, Zhu L, Chen C, Zeng H. Potential Contributions of Human Endogenous Retroviruses in Innate Immune Memory. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1225-1233. [PMID: 39230265 DOI: 10.4049/jimmunol.2300411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/08/2024] [Indexed: 09/05/2024]
Abstract
The phenomenon wherein innate immune cells adopt long-term inflammatory phenotypes following the first stimuli is named trained immunity and can improve host defense against infections. Transcriptional and epigenetic reprogramming are critical mechanisms of trained immunity; however, the regulatory networks are not entirely clear at present. The human endogenous retroviruses (HERVs) provide large amounts of transcriptional regulators in the regulatory pathways. In this study, we analyzed published large omics data to explore the roles of such "dark matter" of the human genome in trained and tolerant macrophages. We collected 80 RNA sequencing data and 62 sequencing data to detect histone modifications and active regulatory regions from nine published studies on trained and tolerant macrophages. By analyzing the characteristics of transcription and epigenetic modification of HERVs, as well as their association with gene expression, we found that 15.3% of HERVs were transcribed nonrandomly from noncoding regions and enriched in specific HERV families and specific chromosomes, such as chromosomes 11, 15, 17, and 19, and they were highly related with the expression of adjacent genes. We found that 295 differentially expressed HERVs are located in 50-kbp flanking regions of 142 differentially expressed genes. We found epigenetic changes of these HERVs and that overlap with predicted enhancers and identified 35 enhancer-like HERVs. The related genes were highly involved in the activation and inflammatory responses, such as the TLR pathway. Other pathways including phosphoinositide signaling and transport of folate and K+ might be also related with trained immunity, which require further study. These results demonstrated that HERVs might play important roles in trained immunity.
Collapse
Affiliation(s)
- Pengcheng Du
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China; and
| | - Jiarui Li
- Biomedical Innovation Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Mingxi Hua
- Biomedical Innovation Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Liuluan Zhu
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China; and
| | - Chen Chen
- Biomedical Innovation Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Hui Zeng
- Biomedical Innovation Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
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
|
14
|
Jacobs MME, Maas RJF, Jonkman I, Negishi Y, Tielemans Zamora W, Yanginlar C, van Heck J, Matzaraki V, Martens JHA, Baltissen M, Vermeulen M, Morla-Folch J, Ranzenigo A, Wang W, Umali M, Ochando J, van der Vlag J, Hilbrands LB, Joosten LAB, Netea MG, Mulder WJM, van Leent MMT, Mhlanga MM, Teunissen AJP, Rother N, Duivenvoorden R. Trained immunity is regulated by T cell-induced CD40-TRAF6 signaling. Cell Rep 2024; 43:114664. [PMID: 39178113 PMCID: PMC11536040 DOI: 10.1016/j.celrep.2024.114664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/08/2024] [Accepted: 08/06/2024] [Indexed: 08/25/2024] Open
Abstract
Trained immunity is characterized by histone modifications and metabolic changes in innate immune cells following exposure to inflammatory signals, leading to heightened responsiveness to secondary stimuli. Although our understanding of the molecular regulation of trained immunity has increased, the role of adaptive immune cells herein remains largely unknown. Here, we show that T cells modulate trained immunity via cluster of differentiation 40-tissue necrosis factor receptor-associated factor 6 (CD40-TRAF6) signaling. CD40-TRAF6 inhibition modulates functional, transcriptomic, and metabolic reprogramming and modifies histone 3 lysine 4 trimethylation associated with trained immunity. Besides in vitro studies, we reveal that single-nucleotide polymorphisms in the proximity of CD40 are linked to trained immunity responses in vivo and that combining CD40-TRAF6 inhibition with cytotoxic T lymphocyte antigen 4-immunoglobulin (CTLA4-Ig)-mediated co-stimulatory blockade induces long-term graft acceptance in a murine heart transplantation model. Combined, our results reveal that trained immunity is modulated by CD40-TRAF6 signaling between myeloid and adaptive immune cells and that this can be leveraged for therapeutic purposes.
Collapse
Affiliation(s)
- Maaike M E Jacobs
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rianne J F Maas
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Inge Jonkman
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Yutaka Negishi
- Department of Cell Biology, Faculty of Science, Radboud University, Nijmegen, the Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Willem Tielemans Zamora
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cansu Yanginlar
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Julia van Heck
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Vasiliki Matzaraki
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Joost H A Martens
- Department of Molecular Biology, Faculty of Science, Oncode Institute, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Marijke Baltissen
- Department of Molecular Biology, Faculty of Science, Oncode Institute, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Oncode Institute, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Judit Morla-Folch
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Ranzenigo
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - William Wang
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Martin Umali
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jordi Ochando
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Transplant Immunology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Johan van der Vlag
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Luuk B Hilbrands
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - 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, University of Medicine and Pharmacy, Iuliu Haţieganu, Cluj-Napoca, Romania
| | - 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
| | - Willem J M Mulder
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Mandy M T van Leent
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Musa M Mhlanga
- Department of Cell Biology, Faculty of Science, Radboud University, Nijmegen, the Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Abraham J P Teunissen
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nils Rother
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Raphaël Duivenvoorden
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands; BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
15
|
Cheng M, Dai Q, Liu Z, Wang Y, Zhou C. New progress in pediatric allergic rhinitis. Front Immunol 2024; 15:1452410. [PMID: 39351215 PMCID: PMC11439695 DOI: 10.3389/fimmu.2024.1452410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024] Open
Abstract
The prevalence of allergic rhinitis (AR) in children is steadily increasing, and its onset is closely associated with genetic factors, living environment, and exposure to allergens. In recent years, an increasing number of diagnostic methods have been employed to assist in diagnosing AR. In addition to pharmaceutical treatments, personalized approaches such as environmental control and allergen-specific immunotherapy are gradually gaining popularity. In this article, we reviewed recent research on the etiology, diagnostic classification, treatment methods, and health management of AR in children. These insights will benefit the implementation of personalized diagnosis and treatment for children with AR, promoting health management strategies that improve symptoms and quality of life.
Collapse
Affiliation(s)
- Miao Cheng
- Department of Ophthalmology and Otolaryngology, Jingmen Centra Hospital, Jingmen Central Hospital Affiliated to Jingchu University of Technology, Jingmen, Hubei, China
| | - Qianqian Dai
- Department of Infectious Disease, Jingmen Central Hospital, Jingmen Central Hospital Affiliated to Jingchu University of Technology, Jingmen, Hubei, China
| | - Zhi Liu
- Department of Pediatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yulin Wang
- Department of Pediatrics, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, Hubei, China
| | - Cuiyun Zhou
- Department of Ophthalmology and Otolaryngology, Jingmen Centra Hospital, Jingmen Central Hospital Affiliated to Jingchu University of Technology, Jingmen, Hubei, China
| |
Collapse
|
16
|
Liu Y, Zhou J, Yang Y, Chen X, Chen L, Wu Y. Intestinal Microbiota and Its Effect on Vaccine-Induced Immune Amplification and Tolerance. Vaccines (Basel) 2024; 12:868. [PMID: 39203994 PMCID: PMC11359036 DOI: 10.3390/vaccines12080868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 09/03/2024] Open
Abstract
This review provides the potential of intestinal microbiota in vaccine design and application, exploring the current insights into the interplay between the intestinal microbiota and the immune system, with a focus on its intermediary function in vaccine efficacy. It summarizes families and genera of bacteria that are part of the intestinal microbiota that may enhance or diminish vaccine efficacy and discusses the foundational principles of vaccine sequence design and the application of gut microbial characteristics in vaccine development. Future research should further investigate the use of multi-omics technologies to elucidate the interactive mechanisms between intestinal microbiota and vaccine-induced immune responses, aiming to optimize and improve vaccine design.
Collapse
Affiliation(s)
- Yixin Liu
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China;
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; (J.Z.); (L.C.)
| | - Jianfeng Zhou
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; (J.Z.); (L.C.)
| | - Yushang Yang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; (J.Z.); (L.C.)
| | - Xiangzheng Chen
- Department of Liver Surgery & Liver Transplantation, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Longqi Chen
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; (J.Z.); (L.C.)
| | - Yangping Wu
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China;
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Chengdu 610041, China
| |
Collapse
|
17
|
Sui Y, Berzofsky JA. Trained immunity inducers in cancer immunotherapy. Front Immunol 2024; 15:1427443. [PMID: 39081326 PMCID: PMC11286386 DOI: 10.3389/fimmu.2024.1427443] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024] Open
Abstract
While most of the cancer immunotherapy strategies engage adaptive immunity, especially tumor-associated T cells, the small fraction of responding patients and types of cancers amenable, and the possibility of severe adverse effects limit its usage. More effective and general interventions are urgently needed. Recently, a de facto innate immune memory, termed 'trained immunity', has become a new research focal point, and promises to be a powerful tool for achieving long-term therapeutic benefits against cancers. Trained immunity-inducing agents such as BCG and fungal glucan have been shown to be able to avert the suppressive tumor microenvironment (TME), enhance T cell responses, and eventually lead to tumor regression. Here, we review the current understating of trained immunity induction and highlight the critical roles of emergency granulopoiesis, interferon γ and tissue-specific induction. Preclinical and clinical studies that have exploited trained immunity inducers for cancer immunotherapy are summarized, and repurposed trained immunity inducers from other fields are proposed. We also outline the challenges and opportunities for trained immunity in future cancer immunotherapies. We envisage that more effective cancer vaccines will combine the induction of trained immunity with T cell therapies.
Collapse
Affiliation(s)
- Yongjun Sui
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | | |
Collapse
|
18
|
Perea L, Faner R, Chalmers JD, Sibila O. Pathophysiology and genomics of bronchiectasis. Eur Respir Rev 2024; 33:240055. [PMID: 38960613 PMCID: PMC11220622 DOI: 10.1183/16000617.0055-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/02/2024] [Indexed: 07/05/2024] Open
Abstract
Bronchiectasis is a complex and heterogeneous inflammatory chronic respiratory disease with an unknown cause in around 30-40% of patients. The presence of airway infection together with chronic inflammation, airway mucociliary dysfunction and lung damage are key components of the vicious vortex model that better describes its pathophysiology. Although bronchiectasis research has significantly increased over the past years and different endotypes have been identified, there are still major gaps in the understanding of the pathophysiology. Genomic approaches may help to identify new endotypes, as has been shown in other chronic airway diseases, such as COPD.Different studies have started to work in this direction, and significant contributions to the understanding of the microbiome and proteome diversity have been made in bronchiectasis in recent years. However, the systematic application of omics approaches to identify new molecular insights into the pathophysiology of bronchiectasis (endotypes) is still limited compared with other respiratory diseases.Given the complexity and diversity of these technologies, this review describes the key components of the pathophysiology of bronchiectasis and how genomics can be applied to increase our knowledge, including the study of new techniques such as proteomics, metabolomics and epigenomics. Furthermore, we propose that the novel concept of trained innate immunity, which is driven by microbiome exposures leading to epigenetic modifications, can complement our current understanding of the vicious vortex. Finally, we discuss the challenges, opportunities and implications of genomics application in clinical practice for better patient stratification into new therapies.
Collapse
Affiliation(s)
- Lidia Perea
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Rosa Faner
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias M.P. (CIBERES), Barcelona, Spain
| | - James D Chalmers
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Oriol Sibila
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias M.P. (CIBERES), Barcelona, Spain
- Respiratory Department, Hospital Clínic, University of Barcelona, Barcelona, Spain
| |
Collapse
|
19
|
Nelson BN, Friedman JE. Developmental Programming of the Fetal Immune System by Maternal Western-Style Diet: Mechanisms and Implications for Disease Pathways in the Offspring. Int J Mol Sci 2024; 25:5951. [PMID: 38892139 PMCID: PMC11172957 DOI: 10.3390/ijms25115951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Maternal obesity and over/undernutrition can have a long-lasting impact on offspring health during critical periods in the first 1000 days of life. Children born to mothers with obesity have reduced immune responses to stimuli which increase susceptibility to infections. Recently, maternal western-style diets (WSDs), high in fat and simple sugars, have been associated with skewing neonatal immune cell development, and recent evidence suggests that dysregulation of innate immunity in early life has long-term consequences on metabolic diseases and behavioral disorders in later life. Several factors contribute to abnormal innate immune tolerance or trained immunity, including changes in gut microbiota, metabolites, and epigenetic modifications. Critical knowledge gaps remain regarding the mechanisms whereby these factors impact fetal and postnatal immune cell development, especially in precursor stem cells in bone marrow and fetal liver. Components of the maternal microbiota that are transferred from mothers consuming a WSD to their offspring are understudied and identifying cause and effect on neonatal innate and adaptive immune development needs to be refined. Tools including single-cell RNA-sequencing, epigenetic analysis, and spatial location of specific immune cells in liver and bone marrow are critical for understanding immune system programming. Considering the vital role immune function plays in offspring health, it will be important to understand how maternal diets can control developmental programming of innate and adaptive immunity.
Collapse
Affiliation(s)
- Benjamin N. Nelson
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Department of Physiology and Biochemistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Pediatrics, Section of Diabetes and Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
20
|
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: 26] [Impact Index Per Article: 26.0] [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
|
21
|
Ariyanto EF, Wijaya I, Pradian ZA, Bhaskara APM, Rahman PHA, Oktavia N. Recent Updates on Epigenetic-Based Pharmacotherapy for Atherosclerosis. Diabetes Metab Syndr Obes 2024; 17:1867-1878. [PMID: 38706808 PMCID: PMC11068051 DOI: 10.2147/dmso.s463221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/19/2024] [Indexed: 05/07/2024] Open
Abstract
Atherosclerosis is one of the most dominant pathological processes responsible in cardiovascular diseases (CVD) caused by cholesterol accumulation accompanied by inflammation in the arteries which will subsequently lead to further complications, including myocardial infarction and stroke. Although the incidence of atherosclerosis is decreasing in some countries, it is still considered the leading cause of death worldwide. Atherosclerosis is a vascular pathological process that is chronically inflammatory and is characterized by the invasion of inflammatory cells and cytokines. Many reports have unraveled the pivotal roles of epigenetics such as DNA methylation, post-translational histone modifications, and non-coding RNAs (ncRNAs) in atherogenesis, which regulate the expression of numerous genes related to various responsible pathways. Many studies have been conducted to develop new therapeutical approaches based on epigenetic changes for combating atherosclerosis. This review elaborates on recent updates on the development of new atherosclerosis drugs whose mechanism of action is associated with the modulation of DNA methylation, posttranslational histone modifications, and ncRNA-based gene regulation.
Collapse
Affiliation(s)
- Eko Fuji Ariyanto
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, Indonesia
| | - Ibnu Wijaya
- Faculty of Medicine, Universitas Padjadjaran, Sumedang, Indonesia
| | | | | | | | - Nandina Oktavia
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, Indonesia
| |
Collapse
|
22
|
Shaku MT, Um PK, Ocius KL, Apostolos AJ, Pires MM, Bishai WR, Kana BD. A modified BCG with depletion of enzymes associated with peptidoglycan amidation induces enhanced protection against tuberculosis in mice. eLife 2024; 13:e89157. [PMID: 38639995 PMCID: PMC11132681 DOI: 10.7554/elife.89157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 04/17/2024] [Indexed: 04/20/2024] Open
Abstract
Mechanisms by which Mycobacterium tuberculosis (Mtb) evades pathogen recognition receptor activation during infection may offer insights for the development of improved tuberculosis (TB) vaccines. Whilst Mtb elicits NOD-2 activation through host recognition of its peptidoglycan-derived muramyl dipeptide (MDP), it masks the endogenous NOD-1 ligand through amidation of glutamate at the second position in peptidoglycan side-chains. As the current BCG vaccine is derived from pathogenic mycobacteria, a similar situation prevails. To alleviate this masking ability and to potentially improve efficacy of the BCG vaccine, we used CRISPRi to inhibit expression of the essential enzyme pair, MurT-GatD, implicated in amidation of peptidoglycan side-chains. We demonstrate that depletion of these enzymes results in reduced growth, cell wall defects, increased susceptibility to antibiotics, altered spatial localization of new peptidoglycan and increased NOD-1 expression in macrophages. In cell culture experiments, training of a human monocyte cell line with this recombinant BCG yielded improved control of Mtb growth. In the murine model of TB infection, we demonstrate that depletion of MurT-GatD in BCG, which is expected to unmask the D-glutamate diaminopimelate (iE-DAP) NOD-1 ligand, yields superior prevention of TB disease compared to the standard BCG vaccine. In vitro and in vivo experiments in this study demonstrate the feasibility of gene regulation platforms such as CRISPRi to alter antigen presentation in BCG in a bespoke manner that tunes immunity towards more effective protection against TB disease.
Collapse
Affiliation(s)
- Moagi Tube Shaku
- DST/NRF Centre of Excellence for Biomedical TB Research, Faculty of Health Sciences, University of the Witwatersrand, National Health Laboratory ServiceJohannesburgSouth Africa
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins School of MedicineBaltimoreUnited States
| | - Peter K Um
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins School of MedicineBaltimoreUnited States
| | - Karl L Ocius
- Department of Chemistry, University of VirginiaCharlottesvilleUnited States
| | - Alexis J Apostolos
- Department of Chemistry, University of VirginiaCharlottesvilleUnited States
| | - Marcos M Pires
- Department of Chemistry, University of VirginiaCharlottesvilleUnited States
| | - William R Bishai
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins School of MedicineBaltimoreUnited States
| | - Bavesh D Kana
- DST/NRF Centre of Excellence for Biomedical TB Research, Faculty of Health Sciences, University of the Witwatersrand, National Health Laboratory ServiceJohannesburgSouth Africa
| |
Collapse
|
23
|
Bhargavi G, Subbian S. The causes and consequences of trained immunity in myeloid cells. Front Immunol 2024; 15:1365127. [PMID: 38665915 PMCID: PMC11043514 DOI: 10.3389/fimmu.2024.1365127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Conventionally, immunity in humans has been classified as innate and adaptive, with the concept that only the latter type has an immunological memory/recall response against specific antigens or pathogens. Recently, a new concept of trained immunity (a.k.a. innate memory response) has emerged. According to this concept, innate immune cells can exhibit enhanced responsiveness to subsequent challenges, after initial stimulation with antigen/pathogen. Thus, trained immunity enables the innate immune cells to respond robustly and non-specifically through exposure or re-exposure to antigens/infections or vaccines, providing enhanced resistance to unrelated pathogens or reduced infection severity. For example, individuals vaccinated with BCG to protect against tuberculosis were also protected from malaria and SARS-CoV-2 infections. Epigenetic modifications such as histone acetylation and metabolic reprogramming (e.g. shift towards glycolysis) and their inter-linked regulations are the key factors underpinning the immune activation of trained cells. The integrated metabolic and epigenetic rewiring generates sufficient metabolic intermediates, which is crucial to meet the energy demand required to produce proinflammatory and antimicrobial responses by the trained cells. These factors also determine the efficacy and durability of trained immunity. Importantly, the signaling pathways and regulatory molecules of trained immunity can be harnessed as potential targets for developing novel intervention strategies, such as better vaccines and immunotherapies against infectious (e.g., sepsis) and non-infectious (e.g., cancer) diseases. However, aberrant inflammation caused by inappropriate onset of trained immunity can lead to severe autoimmune pathological consequences, (e.g., systemic sclerosis and granulomatosis). In this review, we provide an overview of conventional innate and adaptive immunity and summarize various mechanistic factors associated with the onset and regulation of trained immunity, focusing on immunologic, metabolic, and epigenetic changes in myeloid cells. This review underscores the transformative potential of trained immunity in immunology, paving the way for developing novel therapeutic strategies for various infectious and non-infectious diseases that leverage innate immune memory.
Collapse
Affiliation(s)
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| |
Collapse
|
24
|
Zhang N, Wang X, Feng M, Li M, Wang J, Yang H, He S, Xia Z, Shang L, Jiang X, Sun M, Wu Y, Ren C, Zhang X, Li J, Gao F. Early-life exercise induces immunometabolic epigenetic modification enhancing anti-inflammatory immunity in middle-aged male mice. Nat Commun 2024; 15:3103. [PMID: 38600123 PMCID: PMC11006929 DOI: 10.1038/s41467-024-47458-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 04/03/2024] [Indexed: 04/12/2024] Open
Abstract
Exercise is usually regarded to have short-term beneficial effects on immune health. Here we show that early-life regular exercise exerts long-term beneficial effects on inflammatory immunity. Swimming training for 3 months in male mice starting from 1-month-old curbs cytokine response and mitigates sepsis when exposed to lipopolysaccharide challenge, even after an 11-month interval of detraining. Metabolomics analysis of serum and liver identifies pipecolic acid, a non-encoded amino acid, as a pivotal metabolite responding to early-life regular exercise. Importantly, pipecolic acid reduces inflammatory cytokines in bone marrow-derived macrophages and alleviates sepsis via inhibiting mTOR complex 1 signaling. Moreover, early-life exercise increases histone 3 lysine 4 trimethylation at the promoter of Crym in the liver, an enzyme responsible for catalyzing pipecolic acid production. Liver-specific knockdown of Crym in adult mice abolishes this early exercise-induced protective effects. Our findings demonstrate that early-life regular exercise enhances anti-inflammatory immunity during middle-aged phase in male mice via epigenetic immunometabolic modulation, in which hepatic pipecolic acid production has a pivotal function.
Collapse
Affiliation(s)
- Nini Zhang
- Key Laboratory of Aerospace Medicine, Ministry of Education; School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
- Department of Pediatrics, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Xinpei Wang
- Key Laboratory of Aerospace Medicine, Ministry of Education; School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| | - Mengya Feng
- Key Laboratory of Aerospace Medicine, Ministry of Education; School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
- Center for Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Min Li
- Key Laboratory of Aerospace Medicine, Ministry of Education; School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| | - Jing Wang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hongyan Yang
- Key Laboratory of Aerospace Medicine, Ministry of Education; School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| | - Siyu He
- Key Laboratory of Aerospace Medicine, Ministry of Education; School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| | - Ziqi Xia
- Key Laboratory of Aerospace Medicine, Ministry of Education; School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| | - Lei Shang
- Key Laboratory of Hazard Assessment and Control in Special Operational Environment, Ministry of Education; Department of Health Statistics, School of Public Health, Fourth Military Medical University, Xi'an, China
| | - Xun Jiang
- Department of Pediatrics, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Mao Sun
- Department of Biochemistry and Molecular Biology, Center for DNA Typing, Fourth Military Medical University, Xi'an, China
| | - Yuanming Wu
- Department of Biochemistry and Molecular Biology, Center for DNA Typing, Fourth Military Medical University, Xi'an, China
| | - Chaoxue Ren
- School of Sport and Health Science, Xi'an Physical Education University, Xi'an, China
| | - Xing Zhang
- Key Laboratory of Aerospace Medicine, Ministry of Education; School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| | - Jia Li
- Key Laboratory of Aerospace Medicine, Ministry of Education; School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China.
- Key Laboratory of Hazard Assessment and Control in Special Operational Environment, Ministry of Education; Department of Health Statistics, School of Public Health, Fourth Military Medical University, Xi'an, China.
| | - Feng Gao
- Key Laboratory of Aerospace Medicine, Ministry of Education; School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
25
|
Wu Y, Caldwell B, Wang J, Zhang Y, Li L. Alleviation of monocyte exhaustion by BCG derivative mycolic acid. iScience 2024; 27:108978. [PMID: 38323001 PMCID: PMC10845070 DOI: 10.1016/j.isci.2024.108978] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/27/2023] [Accepted: 01/16/2024] [Indexed: 02/08/2024] Open
Abstract
Monocyte exhaustion with sustained pathogenic inflammation and immune-suppression, a hallmark of sepsis resulting from systemic infections, presents a challenge with limited therapeutic solutions. This study identified Methoxy-Mycolic Acid (M-MA), a branched mycolic acid derived from Mycobacterium bovis Bacillus Calmette-Guérin (BCG), as a potent agent in alleviating monocyte exhaustion and restoring immune homeostasis. Co-treatment of monocytes with M-MA effectively blocked the expansion of Ly6Chi/CD38hi/PD-L1hi monocytes induced by LPS challenges and restored the expression of immune-enhancing CD86. M-MA treatment restored mitochondrial functions of exhausted monocytes and alleviated their suppressive activities on co-cultured T cells. Independent of TREM2, M-MA blocks Src-STAT1-mediated inflammatory polarization and reduces the production of immune suppressors TAX1BP1 and PLAC8. Whole genome methylation analyses revealed M-MA's ability to erase the methylation memory of exhausted monocytes, particularly restoring Plac8 methylation. Together, our data suggest M-MA as an effective agent in restoring monocyte homeostasis with a therapeutic potential for treating sepsis.
Collapse
Affiliation(s)
- Yajun Wu
- Department of Biological Sciences, Virginia Tech; Blacksburg, VA 24061-0910, USA
| | - Blake Caldwell
- Department of Biological Sciences, Virginia Tech; Blacksburg, VA 24061-0910, USA
| | - Jing Wang
- Department of Biological Sciences, Virginia Tech; Blacksburg, VA 24061-0910, USA
| | - Yao Zhang
- Department of Biological Sciences, Virginia Tech; Blacksburg, VA 24061-0910, USA
| | - Liwu Li
- Department of Biological Sciences, Virginia Tech; Blacksburg, VA 24061-0910, USA
| |
Collapse
|
26
|
Greco A, Mul K, Jaeger MH, Dos Santos JC, Koenen H, de Jong L, Mann R, Fütterer J, Netea MG, Pruijn GJM, van Engelen BGM, Joosten LAB. IL-6 and TNF are Potential Inflammatory Biomarkers in Facioscapulohumeral Muscular Dystrophy. J Neuromuscul Dis 2024; 11:327-347. [PMID: 38250782 DOI: 10.3233/jnd-230063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Background FSHD is a highly prevalent inherited myopathy with a still poorly understood pathology. Objective To investigate whether proinflammatory cytokines are associated with FSHD and which specific innate immune cells are involved in its pathology. Methods First, we measured circulating cytokines in serum samples: IL-6 (FSHD, n = 150; HC, n = 98); TNF (FSHD, n = 150; HC, n = 59); IL-1α (FSHD, n = 150; HC, n = 66); IL-1β (FSHD, n = 150; HC, n = 98); MCP-1 (FSHD, n = 14; HC, n = 14); VEGF-A (FSHD, n = 14; HC, n = 14). Second, we tested trained immunity in monocytes (FSHD, n = 15; HC, n = 15) and NK cells (FSHD, n = 11; HC, n = 11). Next, we explored the cytokine production capacity of NK cells in response to different stimuli (FSHD, n = 39; HC, n = 22). Lastly, we evaluated the cytokine production of ex vivo stimulated MRI guided inflamed (TIRM+) and paired MRI guided non inflamed (TIRM-) muscle biopsies of 21 patients and of 8 HC muscle biopsies. Results We included a total of 190 FSHD patients (N = 190, 48±14 years, 49% men) and of 135 HC (N = 135, 44±15 years, 47% men). We found that FSHD patients had higher concentrations of IL-6 and TNF measured (a) in the circulation, (b) after ex-vivo stimulation of NK cells, and (c) in muscle specimens. Besides, IL-6 circulating concentrations, as well as its production by NK cells and IL-6 content of FSHD muscle specimens, showed a mild correlation with disease duration, disease severity, and muscle weakness. Conclusion These results show that IL-6 and TNF may contribute to FSHD pathology and suggest novel therapeutic targets. Additionally, the activation of NK cells in FSHD may be a novel pathway contributing to FSHD pathology.
Collapse
Affiliation(s)
- Anna Greco
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Karlien Mul
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martin H Jaeger
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jéssica C Dos Santos
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hans Koenen
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Leon de Jong
- Department of Radiology, Nuclear Medicine and Anatomy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ritse Mann
- Department of Radiology, Nuclear Medicine and Anatomy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jurgen Fütterer
- Department of Radiology, Nuclear Medicine and Anatomy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ger J M Pruijn
- Department of Biomolecular Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Baziel G M van Engelen
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
27
|
Piret J, Boivin G. The impact of trained immunity in respiratory viral infections. Rev Med Virol 2024; 34:e2510. [PMID: 38282407 DOI: 10.1002/rmv.2510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/20/2023] [Accepted: 12/27/2023] [Indexed: 01/30/2024]
Abstract
Epidemic peaks of respiratory viruses that co-circulate during the winter-spring seasons can be synchronous or asynchronous. The occurrence of temporal patterns in epidemics caused by some respiratory viruses suggests that they could negatively interact with each other. These negative interactions may result from a programme of innate immune memory, known as trained immunity, which may confer broad protective effects against respiratory viruses. It is suggested that stimulation of innate immune cells by a vaccine or a pathogen could induce their long-term functional reprogramming through an interplay between metabolic and epigenetic changes, which influence the transcriptional response to a secondary challenge. During the coronavirus disease 2019 pandemic, the circulation of most respiratory viruses was prevented by non-pharmacological interventions and then resumed at unusual periods once sanitary measures were lifted. With time, respiratory viruses should find again their own ecological niches. This transition period provides an opportunity to study the interactions between respiratory viruses at the population level.
Collapse
Affiliation(s)
- Jocelyne Piret
- Research Center of the CHU de Québec-Université Laval, Quebec City, Quebec, Canada
| | - Guy Boivin
- Research Center of the CHU de Québec-Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|
28
|
Liu QJ, Yuan W, Yang P, Shao C. Role of glycolysis in diabetic atherosclerosis. World J Diabetes 2023; 14:1478-1492. [PMID: 37970130 PMCID: PMC10642412 DOI: 10.4239/wjd.v14.i10.1478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/16/2023] [Accepted: 09/14/2023] [Indexed: 10/09/2023] Open
Abstract
Diabetes mellitus is a kind of typical metabolic disorder characterized by elevated blood sugar levels. Atherosclerosis (AS) is one of the most common complications of diabetes. Modern lifestyles and trends that promote overconsumption and unhealthy practices have contributed to an increase in the annual incidence of diabetic AS worldwide, which has created a heavy burden on society. Several studies have shown the significant effects of glycolysis-related changes on the occurrence and development of diabetic AS, which may serve as novel thera-peutic targets for diabetic AS in the future. Glycolysis is an important metabolic pathway that generates energy in various cells of the blood vessel wall. In particular, it plays a vital role in the physiological and pathological activities of the three important cells, Endothelial cells, macrophages and vascular smooth muscle cells. There are lots of similar mechanisms underlying diabetic and common AS, the former is more complex. In this article, we describe the role and mechanism underlying glycolysis in diabetic AS, as well as the therapeutic targets, such as trained immunity, microRNAs, gut microbiota, and associated drugs, with the aim to provide some new perspectives and potentially feasible programs for the treatment of diabetic AS in the foreseeable future.
Collapse
Affiliation(s)
- Qian-Jia Liu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, Jiangsu Province, China
| | - Wei Yuan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, Jiangsu Province, China
| | - Ping Yang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, Jiangsu Province, China
| | - Chen Shao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, Jiangsu Province, China
| |
Collapse
|
29
|
Zhu J, Liu J, Yan C, Wang D, Pan W. Trained immunity: a cutting edge approach for designing novel vaccines against parasitic diseases? Front Immunol 2023; 14:1252554. [PMID: 37868995 PMCID: PMC10587610 DOI: 10.3389/fimmu.2023.1252554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/25/2023] [Indexed: 10/24/2023] Open
Abstract
The preventive situation of parasitosis, a global public health burden especially for developing countries, is not looking that good. Similar to other infections, vaccines would be the best choice for preventing and controlling parasitic infection. However, ideal antigenic molecules for vaccine development have not been identified so far, resulting from the complicated life history and enormous genomes of the parasites. Furthermore, the suppression or down-regulation of anti-infectious immunity mediated by the parasites or their derived molecules can compromise the effect of parasitic vaccines. Comparing the early immune profiles of several parasites in the permissive and non-permissive hosts, a robust innate immune response is proposed to be a critical event to eliminate the parasites. Therefore, enhancing innate immunity may be essential for designing novel and effective parasitic vaccines. The newly emerging trained immunity (also termed innate immune memory) has been increasingly recognized to provide a novel perspective for vaccine development targeting innate immunity. This article reviews the current status of parasitic vaccines and anti-infectious immunity, as well as the conception, characteristics, and mechanisms of trained immunity and its research progress in Parasitology, highlighting the possible consideration of trained immunity in designing novel vaccines against parasitic diseases.
Collapse
Affiliation(s)
- Jinhang Zhu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiaxi Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chao Yan
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dahui Wang
- Liangshan College (Li Shui) China, Lishui University, Lishui, Zhejiang, China
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
30
|
Gupta MK, Peng H, Li Y, Xu CJ. The role of DNA methylation in personalized medicine for immune-related diseases. Pharmacol Ther 2023; 250:108508. [PMID: 37567513 DOI: 10.1016/j.pharmthera.2023.108508] [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: 11/01/2022] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
Epigenetics functions as a bridge between host genetic & environmental factors, aiding in human health and diseases. Many immune-related diseases, including infectious and allergic diseases, have been linked to epigenetic mechanisms, particularly DNA methylation. In this review, we summarized an updated overview of DNA methylation and its importance in personalized medicine, and demonstrated that DNA methylation has excellent potential for disease prevention, diagnosis, and treatment in a personalized manner. The future implications and limitations of the DNA methylation study have also been well-discussed.
Collapse
Affiliation(s)
- Manoj Kumar Gupta
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - He Peng
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Yang Li
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany; Department of Internal Medicine and Radboud Institute for Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Cheng-Jian Xu
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany; Department of Internal Medicine and Radboud Institute for Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
31
|
Morroni J, Benedetti A, Esposito L, De Bardi M, Borsellino G, Riera CS, Giordani L, Bouche M, Lozanoska-Ochser B. Injury-experienced satellite cells retain long-term enhanced regenerative capacity. Stem Cell Res Ther 2023; 14:246. [PMID: 37697344 PMCID: PMC10496398 DOI: 10.1186/s13287-023-03492-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 09/06/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND Inflammatory memory or trained immunity is a recently described process in immune and non-immune tissue resident cells, whereby previous exposure to inflammation mediators leads to a faster and stronger responses upon secondary challenge. Whether previous muscle injury is associated with altered responses to subsequent injury by satellite cells (SCs), the muscle stem cells, is not known. METHODS We used a mouse model of repeated muscle injury, in which intramuscular cardiotoxin (CTX) injections were administered 50 days apart in order to allow for full recovery of the injured muscle before the second injury. The effect of prior injury on the phenotype, proliferation and regenerative potential of satellite cells following a second injury was examined in vitro and in vivo by immunohistochemistry, RT-qPCR and histological analysis. RESULTS We show that SCs isolated from muscle at 50 days post-injury (injury-experienced SCs (ieSCs)) enter the cell cycle faster and form bigger myotubes when cultured in vitro, compared to control SCs isolated from uninjured contralateral muscle. Injury-experienced SCs were characterized by the activation of the mTORC 1 signaling pathway, suggesting they are poised to activate sooner following a second injury. Consequently, upon second injury, SCs accumulate in greater numbers in muscle at 3 and 10 days after injury. These changes in SC phenotype and behavior were associated with accelerated muscle regeneration, as evidenced by an earlier appearance of bigger fibers and increased number of myonuclei per fiber at day 10 after the second injury. CONCLUSIONS Overall, we show that skeletal muscle injury has a lasting effect on SC function priming them to respond faster to a subsequent injury. The ieSCs have long-term enhanced regenerative properties that contribute to accelerated regeneration following a secondary challenge.
Collapse
Affiliation(s)
- Jacopo Morroni
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, Rome, Italy
- COU of Neurology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Anna Benedetti
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, Rome, Italy
- Translational Oncology Research Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Lorenza Esposito
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, Rome, Italy
| | - Marco De Bardi
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | | | - Carles Sanchez Riera
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, Rome, Italy
| | - Lorenzo Giordani
- Sorbonne Université, INSERM UMRS 974, Association Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Marina Bouche
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, Rome, Italy
| | - Biliana Lozanoska-Ochser
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, Rome, Italy.
- Department of Medicine and Surgery, LUM University, Casamassima, Bari, Italy.
| |
Collapse
|
32
|
Al B, Suen TK, Placek K, Netea MG. Innate (learned) memory. J Allergy Clin Immunol 2023; 152:551-566. [PMID: 37385546 DOI: 10.1016/j.jaci.2023.06.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 07/01/2023]
Abstract
With the growing body of evidence, it is now clear that not only adaptive immune cells but also innate immune cells can mount a more rapid and potent nonspecific immune response to subsequent exposures. This process is known as trained immunity or innate (learned) immune memory. This review discusses the different immune and nonimmune cell types of the central and peripheral immune systems that can develop trained immunity. This review highlights the intracellular signaling and metabolic and epigenetic mechanisms underlying the formation of innate immune memory. Finally, this review explores the health implications together with the potential therapeutic interventions harnessing trained immunity.
Collapse
Affiliation(s)
- Burcu Al
- Department of Molecular Immunology and Cell Biology, Life and Medical Sciences Institute, University of Bonn
| | - Tsz K Suen
- Department of Molecular Immunology and Cell Biology, Life and Medical Sciences Institute, University of Bonn
| | - Katarzyna Placek
- Department of Molecular Immunology and Cell Biology, Life and Medical Sciences Institute, University of Bonn
| | - Mihai G Netea
- Department of Molecular Immunology and Cell Biology, Life and Medical Sciences Institute, University of Bonn; Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen.
| |
Collapse
|
33
|
Xu K, Saaoud F, Shao Y, Lu Y, Wu S, Zhao H, Chen K, Vazquez-Padron R, Jiang X, Wang H, Yang X. Early hyperlipidemia triggers metabolomic reprogramming with increased SAH, increased acetyl-CoA-cholesterol synthesis, and decreased glycolysis. Redox Biol 2023; 64:102771. [PMID: 37364513 PMCID: PMC10310484 DOI: 10.1016/j.redox.2023.102771] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023] Open
Abstract
To identify metabolomic reprogramming in early hyperlipidemia, unbiased metabolome was screened in four tissues from ApoE-/- mice fed with high fat diet (HFD) for 3 weeks. 30, 122, 67, and 97 metabolites in the aorta, heart, liver, and plasma, respectively, were upregulated. 9 upregulated metabolites were uremic toxins, and 13 metabolites, including palmitate, promoted a trained immunity with increased syntheses of acetyl-CoA and cholesterol, increased S-adenosylhomocysteine (SAH) and hypomethylation and decreased glycolysis. The cross-omics analysis found upregulation of 11 metabolite synthetases in ApoE‾/‾ aorta, which promote ROS, cholesterol biosynthesis, and inflammation. Statistical correlation of 12 upregulated metabolites with 37 gene upregulations in ApoE‾/‾ aorta indicated 9 upregulated new metabolites to be proatherogenic. Antioxidant transcription factor NRF2-/- transcriptome analysis indicated that NRF2 suppresses trained immunity-metabolomic reprogramming. Our results have provided novel insights on metabolomic reprogramming in multiple tissues in early hyperlipidemia oriented toward three co-existed new types of trained immunity.
Collapse
Affiliation(s)
- Keman Xu
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Fatma Saaoud
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Ying Shao
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Yifan Lu
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Sheng Wu
- Metabolic Disease Research, Thrombosis Research, Departments of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Huaqing Zhao
- Medical Education and Data Science, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | - Kaifu Chen
- Computational Biology Program, Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Roberto Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33125, USA
| | - Xiaohua Jiang
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA; Metabolic Disease Research, Thrombosis Research, Departments of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Hong Wang
- Metabolic Disease Research, Thrombosis Research, Departments of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Xiaofeng Yang
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA; Metabolic Disease Research, Thrombosis Research, Departments of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA.
| |
Collapse
|
34
|
Mol JQ, van Tuijl J, Bekkering S, van der Heijden CD, Damen SA, Cossins BC, van Emst L, Nielen TM, Rodwell L, Li Y, Pop GA, Netea MG, van Royen N, Riksen NP, El Messaoudi S. Peripheral blood mononuclear cell hyperresponsiveness in patients with premature myocardial infarction without traditional risk factors. iScience 2023; 26:107183. [PMID: 37456854 PMCID: PMC10338301 DOI: 10.1016/j.isci.2023.107183] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 04/14/2023] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
An increasing number of patients develop an atherothrombotic myocardial infarction (MI) in the absence of standard modifiable risk factors (SMuRFs). Monocytes and macrophages regulate the development of atherosclerosis, and monocytes can adopt a long-term hyperinflammatory phenotype by epigenetic reprogramming, which can contribute to atherogenesis (called "trained immunity"). We assessed circulating monocyte phenotype and function and specific histone marks associated with trained immunity in SMuRFless patients with MI and matched healthy controls. Even in the absence of systemic inflammation, monocytes from SMuRFless patients with MI had an increased overall cytokine production capacity, with the strongest difference for LPS-induced interleukin-10 production, which was associated with an enrichment of the permissive histone marker H3K4me3 at the promoter region. Considering the lack of intervenable risk factors in these patients, trained immunity could be a promising target for future therapy.
Collapse
Affiliation(s)
- Jan-Quinten Mol
- Department of Cardiology, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Julia van Tuijl
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Siroon Bekkering
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | | | - Sander A.J. Damen
- Department of Cardiology, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Benjamin C. Cossins
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Liesbeth van Emst
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Tim M. Nielen
- Department of Cardiology, Canisius Wilhelmina Hospital, 6532 SZ Nijmegen, the Netherlands
| | - Laura Rodwell
- Section Biostatistics, Department for Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Yang Li
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM) & TWINCORE, joint ventures between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), 30625 Hannover, Germany
| | - Gheorghe A.M. Pop
- Department of Cardiology, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Mihai G. Netea
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - Niels van Royen
- Department of Cardiology, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Niels P. Riksen
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Saloua El Messaoudi
- Department of Cardiology, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| |
Collapse
|
35
|
Antunes MDSM, Sugiyama FHC, Gravina HD, Castro RC, Mercado FJR, de Lima JO, Fontanari C, Frantz FG. COVID-19 inactivated and non-replicating viral vector vaccines induce regulatory training phenotype in human monocytes under epigenetic control. Front Cell Infect Microbiol 2023; 13:1200789. [PMID: 37520439 PMCID: PMC10382685 DOI: 10.3389/fcimb.2023.1200789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/26/2023] [Indexed: 08/01/2023] Open
Abstract
Background Trained immunity is the enhanced innate immune response resulting from exposure to pathogens or vaccines against an unrelated pathogen stimulus. Certain vaccines induce a memory like response in monocytes and NK cells, leading to modulation in cytokine production, metabolic changes, and modifications in histone patterns. Here, we hypothesized that vaccination against SARS-CoV-2 could induce the training of monocytes in addition to stimulating the adaptive immune response. Methods Therefore, we aimed to investigate the immunophenotyping, cytokine and metabolic profile of monocytes from individuals who were completely immunized with two doses of inactivated COVID-19 vaccine or non-replicating viral vector vaccine. Subsequently, we investigated the epigenetic mechanisms underlying monocyte immune training. As a model of inflammatorychallenge, to understand if the monocytes were trained by vaccination and how they were trained, cells were stimulated in vitro with the endotoxin LPS, an unrelated stimulus that would provoke the effects of training. Results When challenged in vitro, monocytes from vaccinated individuals produced less TNF-α and those who received inactivated vaccine produced less IL-6, whereas vaccination with non-replicating viral vector vaccine induced more IL-10. Inactivated vaccine increased classical monocyte frequency, and both groups showed higher CD163 expression, a hallmark of trained immunity. We observed increased expression of genes involved in glycolysis and reduced IRG1 expression in vaccinated subjects, a gene associated with the tolerance phenotype in monocytes. We observed that both vaccines reduced the chromatin accessibility of genes associated with the inflammatory response, the inactivated COVID-19 vaccine trained monocytes to a regulatory phenotype mediated by histone modifications in the IL6 and IL10 genes, while the non-replicating viral vector COVID-19 vaccine trained monocytes to a regulatory phenotype, mediated by histone modifications in the IL6, IL10, TNF, and CCL2 genes. Conclusions Our findings support the recognized importance of adopting vaccination against SARS CoV-2, which has been shown to be effective in enhancing the adaptive immune response against the virus and reducing mortality and morbidity rates. Here, we provide evidence that vaccination also modulates the innate immune response by controlling the detrimental inflammatory response to unrelated pathogen stimulation.
Collapse
|
36
|
Gonciarz W, Piątczak E, Chmiela M. The influence of Salvia cadmica Boiss. extracts on the M1/M2 polarization of macrophages primed with Helicobacter pylori lipopolysaccharide in conjunction with NF-kappa B activation, production of cytokines, phagocytic activity and total DNA methylation. JOURNAL OF ETHNOPHARMACOLOGY 2023; 310:116386. [PMID: 36921911 DOI: 10.1016/j.jep.2023.116386] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/23/2023] [Accepted: 03/09/2023] [Indexed: 05/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The large number of secondary derivatives have been isolated from the genus Salvia with about 700 species, and used in the pharmacopoeia throughout the world. Various biological properties of Salvia formulations have been reported including as antioxidant, antimicrobial, hypotensive, anti-hyperglycemia, anti-hyperlipidemia, anti-cancer, and skin curative. Salvia cadmica Boiss. root and aerial part extracts enriched with polyphenols are bactericidal towards gastric pathogen Helicobacter pylori (Hp) and diminish deleterious effects induced by Hp lipopolysaccharide (LPS) towards gastric epithelial cells. AIM OF THIS STUDY To examine the influence of S. cadmica extracts on the M1/M2 polarization of macrophages primed with Hp LPS vs standard LPS Escherichia coli (Ec), and the macrophage cytokine as well as phagocytic activity, which are affected during Hp infection. MATERIAL AND METHODS Macrophages derived from THP-1 human monocytes primed with LPS Hp/Ec and/or S. cadmica extracts, were examined for the biomarkers of activation (surface, cytoplasmic or soluble), and phagocytic capacity. The bone marrow macrophages of Caviaporcellus were used to determine the engulfment of Hp. RESULTS Priming of THP-1 cells (24h) with LPS Hp/Ec resulted in polarization of M1 macrophages, activation of nuclear factor kappa B, secretion of tumor necrosis factor (TNF)-α, interleukin (IL)-1 beta, macrophage chemotactic protein (MCP)-1, immunoregulatory IL-10, and production of reactive oxygen species. These effects were diminished after restimulation of cells with S. cadmica extracts. THP-1 macrophages exposed to studied extracts showed an increased phagocytic capacity, in conjunction with elevated CD11b/CD11d expression and enhanced production of inducible nitric oxide synthase. They also increased Hp engulfment by bone marrow macrophages. These effects were not related to a global DNA methylation. CONCLUSIONS S. cadmica extracts possess an immunomodulating activity, which might be useful in control of H. pylori LPS driven activity of macrophages.
Collapse
Affiliation(s)
- Weronika Gonciarz
- Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, 12/16 Banacha St., 90-237, Lodz, Poland.
| | - Ewelina Piątczak
- Department of Pharmaceutical Biotechnology, Medical University of Lodz, Muszyńskiego 1 St., 90-151, Lodz, Poland.
| | - Magdalena Chmiela
- Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, 12/16 Banacha St., 90-237, Lodz, Poland.
| |
Collapse
|
37
|
Guo C, Zhao M, Sui X, Balsara Z, Zhai S, Ahdoot M, Zhang Y, Lam CM, Zhu P, Li X. Targeting the PRC2-dependent epigenetic program alleviates urinary tract infections. iScience 2023; 26:106925. [PMID: 37332606 PMCID: PMC10272480 DOI: 10.1016/j.isci.2023.106925] [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: 09/15/2022] [Revised: 04/08/2023] [Accepted: 05/15/2023] [Indexed: 06/20/2023] Open
Abstract
Urinary tract infection (UTI) is a pervasive health problem worldwide. Patients with a history of UTIs suffer increased risk of recurrent infections, a major risk of antibiotic resistance. Here, we show that bladder infections induce expression of Ezh2 in bladder urothelial cells. Ezh2 is the methyltransferase of polycomb repressor complex 2 (PRC2)-a potent epigenetic regulator. Urothelium-specific inactivation of PRC2 results in reduced urine bacterial burden, muted inflammatory response, and decreased activity of the NF-κB signaling pathway. PRC2 inactivation also facilitates proper regeneration after urothelial damage from UTIs, by attenuating basal cell hyperplasia and increasing urothelial differentiation. In addition, treatment with Ezh2-specific small-molecule inhibitors improves outcomes of the chronic and severe bladder infections in mice. These findings collectively suggest that the PRC2-dependent epigenetic reprograming controls the amplitude of inflammation and severity of UTIs and that Ezh2 inhibitors may be a viable non-antibiotic strategy to manage chronic and severe UTIs.
Collapse
Affiliation(s)
- Chunming Guo
- Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis 3089, Los Angeles, CA 90048, USA
- Departments of Urology and Surgery, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Mingyi Zhao
- Departments of Urology and Surgery, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
- Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Pathogenesis, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, Guangdong 510100, China
| | - Xinbing Sui
- Departments of Urology and Surgery, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Zarine Balsara
- Departments of Urology and Surgery, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Songhui Zhai
- Departments of Urology and Surgery, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Michael Ahdoot
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis 3089, Los Angeles, CA 90048, USA
| | - Yingsheng Zhang
- Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis 3089, Los Angeles, CA 90048, USA
| | - Christa M. Lam
- Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis 3089, Los Angeles, CA 90048, USA
- Departments of Urology and Surgery, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Pathogenesis, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, Guangdong 510100, China
| | - Xue Li
- Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis 3089, Los Angeles, CA 90048, USA
- Departments of Urology and Surgery, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
38
|
Tang C, Kurata S, Fuse N. Re-recognition of innate immune memory as an integrated multidimensional concept. Microbiol Immunol 2023. [PMID: 37311618 DOI: 10.1111/1348-0421.13083] [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: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/15/2023]
Abstract
In the past decade, the concept of immunological memory, which has long been considered a phenomenon observed in the adaptive immunity of vertebrates, has been extended to the innate immune system of various organisms. This de novo immunological memory is mainly called "innate immune memory", "immune priming", or "trained immunity" and has received increased attention because of its potential for clinical and agricultural applications. However, research on different species, especially invertebrates and vertebrates, has caused controversy regarding this concept. Here we discuss the current studies focusing on this immunological memory and summarize several mechanisms underlying it. We propose "innate immune memory" as a multidimensional concept as an integration between the seemingly different immunological phenomena.
Collapse
Affiliation(s)
- Chang Tang
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Shoichiro Kurata
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Naoyuki Fuse
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| |
Collapse
|
39
|
Chaumond E, Peron S, Daniel N, Le Gouar Y, Guédon É, Williams DL, Le Loir Y, Jan G, Berkova N. Development of innate immune memory by non-immune cells during Staphylococcus aureus infection depends on reactive oxygen species. Front Immunol 2023; 14:1138539. [PMID: 37325649 PMCID: PMC10264681 DOI: 10.3389/fimmu.2023.1138539] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 05/19/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction The mechanisms underlying innate immune memory (trained immunity) comprise epigenetic reprogramming of transcriptional pathways associated with alterations of intracellular metabolism. While the mechanisms of innate immune memory carried out by immune cells are well characterized, such processes in non-immune cells, are poorly understood. The opportunistic pathogen, Staphylococcus aureus, is responsible for a multitude of human diseases, including pneumonia, endocarditis and osteomyelitis, as well as animal infections, including chronic cattle mastitis that are extremely difficult to treat. An induction of innate immune memory may be considered as a therapeutic alternative to fight S. aureus infection. Methods In the current work, we demonstrated the development of innate immune memory in non-immune cells during S. aureus infection employing a combination of techniques including Enzyme-linked immunosorbent assay (ELISA), microscopic analysis, and cytometry. Results We observed that training of human osteoblast-like MG-63 cells and lung epithelial A549 cells with β-glucan increased IL-6 and IL-8 production upon a stimulation with S. aureus, concomitant with histones modifications. IL-6 and IL-8 production was positively correlated with an acetylation of histone 3 at lysine 27 (H3K27), thus suggesting epigenetic reprogramming in these cells. An addition of the ROS scavenger N-Acetylcysteine, NAC, prior to β-glucan pretreatment followed by an exposure to S. aureus, resulted in decreased IL-6 and IL-8 production, thereby supporting the involvement of ROS in the induction of innate immune memory. Exposure of cells to Lactococcus lactis resulted in increased IL-6 and IL-8 production by MG-63 and A549 cells upon a stimulation with S. aureus that was correlated with H3K27 acetylation, suggesting the ability of this beneficial bacterium to induce innate immune memory. Discussion This work improves our understanding of innate immune memory in non-immune cells in the context of S. aureus infection. In addition to known inducers, probiotics may represent good candidates for the induction of innate immune memory. Our findings may help the development of alternative therapeutic approaches for the prevention of S. aureus infection.
Collapse
Affiliation(s)
- Emmanuel Chaumond
- l'Institut national de recherche pour l'agriculture, l'alimentation et l'environnement (INRAE), Institut Agro, Science et Technologie du Lait et de l'Oeuf (STLO), Rennes, France
| | - Sandrine Peron
- l'Institut national de recherche pour l'agriculture, l'alimentation et l'environnement (INRAE), Institut Agro, Science et Technologie du Lait et de l'Oeuf (STLO), Rennes, France
| | - Nathalie Daniel
- l'Institut national de recherche pour l'agriculture, l'alimentation et l'environnement (INRAE), Institut Agro, Science et Technologie du Lait et de l'Oeuf (STLO), Rennes, France
| | - Yann Le Gouar
- l'Institut national de recherche pour l'agriculture, l'alimentation et l'environnement (INRAE), Institut Agro, Science et Technologie du Lait et de l'Oeuf (STLO), Rennes, France
| | - Éric Guédon
- l'Institut national de recherche pour l'agriculture, l'alimentation et l'environnement (INRAE), Institut Agro, Science et Technologie du Lait et de l'Oeuf (STLO), Rennes, France
| | - David L. Williams
- Department of Surgery and Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson, TN, United States
| | - Yves Le Loir
- l'Institut national de recherche pour l'agriculture, l'alimentation et l'environnement (INRAE), Institut Agro, Science et Technologie du Lait et de l'Oeuf (STLO), Rennes, France
| | - Gwénaël Jan
- l'Institut national de recherche pour l'agriculture, l'alimentation et l'environnement (INRAE), Institut Agro, Science et Technologie du Lait et de l'Oeuf (STLO), Rennes, France
| | - Nadia Berkova
- l'Institut national de recherche pour l'agriculture, l'alimentation et l'environnement (INRAE), Institut Agro, Science et Technologie du Lait et de l'Oeuf (STLO), Rennes, France
| |
Collapse
|
40
|
Williams H, Mack C, Baraz R, Marimuthu R, Naralashetty S, Li S, Medbury H. Monocyte Differentiation and Heterogeneity: Inter-Subset and Interindividual Differences. Int J Mol Sci 2023; 24:ijms24108757. [PMID: 37240103 DOI: 10.3390/ijms24108757] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
The three subsets of human monocytes, classical, intermediate, and nonclassical, show phenotypic heterogeneity, particularly in their expression of CD14 and CD16. This has enabled researchers to delve into the functions of each subset in the steady state as well as in disease. Studies have revealed that monocyte heterogeneity is multi-dimensional. In addition, that their phenotype and function differ between subsets is well established. However, it is becoming evident that heterogeneity also exists within each subset, between health and disease (current or past) states, and even between individuals. This realisation casts long shadows, impacting how we identify and classify the subsets, the functions we assign to them, and how they are examined for alterations in disease. Perhaps the most fascinating is evidence that, even in relative health, interindividual differences in monocyte subsets exist. It is proposed that the individual's microenvironment could cause long-lasting or irreversible changes to monocyte precursors that echo to monocytes and through to their derived macrophages. Here, we will discuss the types of heterogeneity recognised in monocytes, the implications of these for monocyte research, and most importantly, the relevance of this heterogeneity for health and disease.
Collapse
Affiliation(s)
- Helen Williams
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW 2145, Australia
- Sydney Medical School, The University of Sydney, Westmead, NSW 2145, Australia
| | - Corinne Mack
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW 2145, Australia
- Sydney Medical School, The University of Sydney, Westmead, NSW 2145, Australia
| | - Rana Baraz
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW 2145, Australia
- Sydney Medical School, The University of Sydney, Westmead, NSW 2145, Australia
| | - Rekha Marimuthu
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW 2145, Australia
- Sydney Medical School, The University of Sydney, Westmead, NSW 2145, Australia
| | - Sravanthi Naralashetty
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW 2145, Australia
- Sydney Medical School, The University of Sydney, Westmead, NSW 2145, Australia
| | - Stephen Li
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW 2145, Australia
- Chemical Pathology, NSW Health Pathology, Westmead Hospital and Institute of Clinical Pathology and Medical Research, Westmead, NSW 2145, Australia
- . Blacktown/Mt Druitt Clinical School, Blacktown Hospital, Western Sydney University, Blacktown, NSW 2148, Australia
| | - Heather Medbury
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW 2145, Australia
- Sydney Medical School, The University of Sydney, Westmead, NSW 2145, Australia
| |
Collapse
|
41
|
Saaoud F, Shao Y, Cornwell W, Wang H, Rogers TJ, Yang X. Cigarette Smoke Modulates Inflammation and Immunity via Reactive Oxygen Species-Regulated Trained Immunity and Trained Tolerance Mechanisms. Antioxid Redox Signal 2023; 38:1041-1069. [PMID: 36017612 PMCID: PMC10171958 DOI: 10.1089/ars.2022.0087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/22/2022] [Indexed: 12/14/2022]
Abstract
Significance: Cigarette smoke (CS) is a prominent cause of morbidity and death and poses a serious challenge to the current health care system worldwide. Its multifaceted roles have led to cardiovascular, respiratory, immunological, and neoplastic diseases. Recent Advances: CS influences both innate and adaptive immunity and regulates immune responses by exacerbating pathogenic immunological responses and/or suppressing defense immunity. There is substantial evidence pointing toward a critical role of CS in vascular immunopathology, but a comprehensive and up-to-date review is lacking. Critical Issues: This review aims to synthesize novel conceptual advances on the immunomodulatory action of CS with a focus on the cardiovascular system from the following perspectives: (i) the signaling of danger-associated molecular pattern (DAMP) receptors contributes to CS modulation of inflammation and immunity; (ii) CS reprograms immunometabolism and trained immunity-related metabolic pathways in innate immune cells and T cells, which can be sensed by the cytoplasmic (cytosolic and non-nuclear organelles) reactive oxygen species (ROS) system in vascular cells; (iii) how nuclear ROS drive CS-promoted DNA damage and cell death pathways, thereby amplifying inflammation and immune responses; and (iv) CS induces endothelial cell (EC) dysfunction and vascular inflammation to promote cardiovascular diseases (CVDs). Future Directions: Despite significant progress in understanding the cellular and molecular mechanisms linking CS to immunity, further investigations are warranted to elucidate novel mechanisms responsible for CS-mediated immunopathology of CVDs; in particular, the research in redox regulation of immune functions of ECs and their fate affected by CS is still in its infancy.
Collapse
Affiliation(s)
- Fatma Saaoud
- Cardiovascular Research Center, Department of Cardiovascular Sciences, Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ying Shao
- Cardiovascular Research Center, Department of Cardiovascular Sciences, Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - William Cornwell
- Center for Inflammation and Lung Research, Department of Microbiology, Immunology & Inflammation, Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Hong Wang
- Metabolic Disease Research and Thrombosis Research Centers, Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Thomas J. Rogers
- Center for Inflammation and Lung Research, Department of Microbiology, Immunology & Inflammation, Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Xiaofeng Yang
- Cardiovascular Research Center, Department of Cardiovascular Sciences, Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
- Metabolic Disease Research and Thrombosis Research Centers, Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
42
|
Nash MJ, Dobrinskikh E, Soderborg TK, Janssen RC, Takahashi DL, Dean TA, Varlamov O, Hennebold JD, Gannon M, Aagaard KM, McCurdy CE, Kievit P, Bergman BC, Jones KL, Pietras EM, Wesolowski SR, Friedman JE. Maternal diet alters long-term innate immune cell memory in fetal and juvenile hematopoietic stem and progenitor cells in nonhuman primate offspring. Cell Rep 2023; 42:112393. [PMID: 37058409 PMCID: PMC10570400 DOI: 10.1016/j.celrep.2023.112393] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/30/2023] [Accepted: 03/30/2023] [Indexed: 04/15/2023] Open
Abstract
Maternal overnutrition increases inflammatory and metabolic disease risk in postnatal offspring. This constitutes a major public health concern due to increasing prevalence of these diseases, yet mechanisms remain unclear. Here, using nonhuman primate models, we show that maternal Western-style diet (mWSD) exposure is associated with persistent pro-inflammatory phenotypes at the transcriptional, metabolic, and functional levels in bone marrow-derived macrophages (BMDMs) from 3-year-old juvenile offspring and in hematopoietic stem and progenitor cells (HSPCs) from fetal and juvenile bone marrow and fetal liver. mWSD exposure is also associated with increased oleic acid in fetal and juvenile bone marrow and fetal liver. Assay for transposase-accessible chromatin with sequencing (ATAC-seq) profiling of HSPCs and BMDMs from mWSD-exposed juveniles supports a model in which HSPCs transmit pro-inflammatory memory to myeloid cells beginning in utero. These findings show that maternal diet alters long-term immune cell developmental programming in HSPCs with proposed consequences for chronic diseases featuring altered immune/inflammatory activation across the lifespan.
Collapse
Affiliation(s)
- Michael J Nash
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Evgenia Dobrinskikh
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Taylor K Soderborg
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Rachel C Janssen
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Diana L Takahashi
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Tyler A Dean
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Oleg Varlamov
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Jon D Hennebold
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Maureen Gannon
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN 37235, USA
| | - Kjersti M Aagaard
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Carrie E McCurdy
- Department of Human Physiology, University of Oregon, Eugene, OR 97403, USA
| | - Paul Kievit
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Bryan C Bergman
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kenneth L Jones
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Eric M Pietras
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Stephanie R Wesolowski
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jacob E Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
43
|
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
|
44
|
Dagenais A, Villalba-Guerrero C, Olivier M. Trained immunity: A “new” weapon in the fight against infectious diseases. Front Immunol 2023; 14:1147476. [PMID: 36993966 PMCID: PMC10040606 DOI: 10.3389/fimmu.2023.1147476] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 02/23/2023] [Indexed: 03/18/2023] Open
Abstract
Innate immune cells can potentiate the response to reinfection through an innate form of immunological memory known as trained immunity. The potential of this fast-acting, nonspecific memory compared to traditional adaptive immunological memory in prophylaxis and therapy has been a topic of great interest in many fields, including infectious diseases. Amidst the rise of antimicrobial resistance and climate change—two major threats to global health—, harnessing the advantages of trained immunity compared to traditional forms of prophylaxis and therapy could be game-changing. Here, we present recent works bridging trained immunity and infectious disease that raise important discoveries, questions, concerns, and novel avenues for the modulation of trained immunity in practice. By exploring the progress in bacterial, viral, fungal, and parasitic diseases, we equally highlight future directions with a focus on particularly problematic and/or understudied pathogens.
Collapse
Affiliation(s)
- Amy Dagenais
- Department of Microbiology and Immunology, Faculty of Medicine, Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Carlos Villalba-Guerrero
- Department of Microbiology and Immunology, Faculty of Medicine, Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Martin Olivier
- Department of Microbiology and Immunology, Faculty of Medicine, Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada
- Department of Medicine, Faculty of Medicine, Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada
- *Correspondence: Martin Olivier,
| |
Collapse
|
45
|
Mycobacterium bovis BCG increase the selected determinants of monocyte/macrophage activity, which were diminished in response to gastric pathogen Helicobacter pylori. Sci Rep 2023; 13:3107. [PMID: 36813949 PMCID: PMC9944772 DOI: 10.1038/s41598-023-30250-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
High antibiotic resistance of gastric pathogen Helicobacter pylori (Hp) and the ability to escape the host immune response prompt searching for therapeutic immunomodulators. Bacillus Calmette-Guerin (BCG) vaccine with Mycobacterium bovis (Mb) is a candidate for modulation the activity of immunocompetent cells, and onco-BCG formulation was successfully used in immunotherapy of bladder cancer. We determined the influence of onco-BCG on the phagocytic capacity of human THP-1 monocyte/macrophage cells, using the model of Escherichia coli bioparticles and Hp fluorescently labeled. Deposition of cell integrins CD11b, CD11d, CD18, membrane/soluble lipopolysaccharide (LPS) receptors, CD14 and sCD14, respectively, and the production of macrophage chemotactic protein (MCP)-1 were determined. Furthermore, a global DNA methylation, was also assessed. Human THP-1 monocytes/macrophages (TIB 202) primed or primed and restimulated with onco-BCG or Hp, were used for assessment of phagocytosis towards E. coli or Hp, surface (immunostaining) or soluble activity determinants, and global DNA methylation (ELISA). THP-1 monocytes/macrophages primed/restimulated with BCG showed increased phagocytosis capacity towards E. coli fluorescent particles, elevated expression of CD11b, CD11d, CD18, CD14, sCD14, increased MCP-1 secretion and DNA methylation. Preliminary results indicate that BCG mycobacteria may also induce the phagocytosis of H. pylori by THP-1 monocytes. Priming or priming and restimulation of monocytes/macrophages with BCG resulted in an increased activity of these cells, which was negatively modulated by Hp.
Collapse
|
46
|
Immunity in Sea Turtles: Review of a Host-Pathogen Arms Race Millions of Years in the Running. Animals (Basel) 2023; 13:ani13040556. [PMID: 36830343 PMCID: PMC9951749 DOI: 10.3390/ani13040556] [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: 11/30/2022] [Revised: 01/05/2023] [Accepted: 01/20/2023] [Indexed: 02/08/2023] Open
Abstract
The immune system of sea turtles is not completely understood. Sea turtles (as reptiles) bridge a unique evolutionary gap, being ectothermic vertebrates like fish and amphibians and amniotes like birds and mammals. Turtles are ectotherms; thus, their immune system is influenced by environmental conditions like temperature and season. We aim to review the turtle immune system and note what studies have investigated sea turtles and the effect of the environment on the immune response. Turtles rely heavily on the nonspecific innate response rather than the specific adaptive response. Turtles' innate immune effectors include antimicrobial peptides, complement, and nonspecific leukocytes. The antiviral defense is understudied in terms of the diversity of pathogen receptors and interferon function. Turtles also mount adaptive responses to pathogens. Lymphoid structures responsible for lymphocyte activation and maturation are either missing in reptiles or function is affected by season. Turtles are a marker of health for their marine environment, and their immune system is commonly dysregulated because of disease or contaminants. Fibropapillomatosis (FP) is a tumorous disease that afflicts sea turtles and is thought to be caused by a virus and an environmental factor. We aim, by exploring the current understanding of the immune system in turtles, to aid the investigation of environmental factors that contribute to the pathogenesis of this disease and provide options for immunotherapy.
Collapse
|
47
|
Polcz VE, Rincon JC, Hawkins RB, Barrios EL, Efron PA, Moldawer LL, Larson SD. TRAINED IMMUNITY: A POTENTIAL APPROACH FOR IMPROVING HOST IMMUNITY IN NEONATAL SEPSIS. Shock 2023; 59:125-134. [PMID: 36383390 PMCID: PMC9957873 DOI: 10.1097/shk.0000000000002054] [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: 11/17/2022]
Abstract
ABSTRACT Sepsis, a dysregulated host immune response to infection, is one of the leading causes of neonatal mortality worldwide. Improved understanding of the perinatal immune system is critical to improve therapies to both term and preterm neonates at increased risk of sepsis. Our narrative outlines the known and unknown aspects of the human immune system through both the immune tolerant in utero period and the rapidly changing antigen-rich period after birth. We will highlight the key differences in innate and adaptive immunity noted through these developmental stages and how the unique immune phenotype in early life contributes to the elevated risk of overwhelming infection and dysregulated immune responses to infection upon exposure to external antigens shortly after birth. Given an initial dependence on neonatal innate immune host responses, we will discuss the concept of innate immune memory, or "trained immunity," and describe several potential immune modulators, which show promise in altering the dysregulated immune response in newborns and improving resilience to sepsis.
Collapse
Affiliation(s)
- Valerie E Polcz
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | | | | | | | | | | | | |
Collapse
|
48
|
Huang M, Malovic E, Ealy A, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG. Microglial immune regulation by epigenetic reprogramming through histone H3K27 acetylation in neuroinflammation. Front Immunol 2023; 14:1052925. [PMID: 37033967 PMCID: PMC10073546 DOI: 10.3389/fimmu.2023.1052925] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
Epigenetic reprogramming is the ability of innate immune cells to form memories of environmental stimuli (priming), allowing for heightened responses to secondary stressors. Herein, we explored microglial epigenetic marks using the known inflammagen LPS as a memory priming trigger and Parkinsonian-linked environmental neurotoxic stressor manganese (Mn) as the secondary environmental trigger. To mimic physiological responses, the memory priming trigger LPS treatment was removed by triple-washing to allow the cells' acute inflammatory response to reset back before applying the secondary insult. Our results show that after the secondary Mn insult, levels of key proinflammatory markers, including nitrite release, iNOS mRNA and protein expression, Il-6, Il-α and cytokines were exaggerated in LPS-primed microglia. Our paradigm implies primed microglia retain immune memory that can be reprogrammed to augment inflammatory response by secondary environmental stress. To ascertain the molecular underpinning of this neuroimmune memory, we further hypothesize that epigenetic reprogramming contributes to the retention of a heightened immune response. Interestingly, Mn-exposed, LPS-primed microglia showed enhanced deposition of H3K27ac and H3K4me3 along with H3K4me1. We further confirmed the results using a PD mouse model (MitoPark) and postmortem human PD brains, thereby adding clinical relevance to our findings. Co-treatment with the p300/H3K27ac inhibitor GNE-049 reduced p300 expression and H3K27ac deposition, decreased iNOS, and increased ARG1 and IRF4 levels. Lastly, since mitochondrial stress is a driver of environmentally linked Parkinson's disease (PD) progression, we examined the effects of GNE-049 on primary trigger-induced mitochondrial stress. GNE-049 reduced mitochondrial superoxide, mitochondrial circularity and stress, and mitochondrial membrane depolarization, suggesting beneficial consequences of GNE-049 on mitochondrial function. Collectively, our findings demonstrate that proinflammatory primary triggers can shape microglial memory via the epigenetic mark H3K27ac and that inhibiting H3K27ac deposition can prevent primary trigger immune memory formation and attenuate subsequent secondary inflammatory responses.
Collapse
Affiliation(s)
- Minhong Huang
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
| | - Emir Malovic
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
| | - Alyssa Ealy
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States
| | - Huajun Jin
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States
| | - Vellareddy Anantharam
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States
| | - Arthi Kanthasamy
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States
| | - Anumantha G. Kanthasamy
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States
- *Correspondence: Anumantha G. Kanthasamy,
| |
Collapse
|
49
|
Abstract
Macrophages have been recognized as the primary mediators of innate immunity starting from embryonic/fetal development. Macrophage-mediated defenses may not be as antigen-specific as adaptive immunity, but increasing information suggests that these responses do strengthen with repeated immunological triggers. The concept of innate memory in macrophages has been described as "trained immunity" or "innate immune memory (IIM)." As currently understood, this cellular memory is rooted in epigenetic and metabolic reprogramming. The recognition of IIM may be particularly important in the fetus and the young neonate who are yet to develop protective levels of adaptive immunity, and could even be of preventive/therapeutic importance in many disorders. There may also be a possibility of therapeutic enhancement with targeted vaccination. This article presents a review of the properties, mechanisms, and possible clinical significance of macrophage-mediated IIM.
Collapse
Affiliation(s)
- Akhil Maheshwari
- Founding Chairman, Global Newborn Society, Clarksville, Maryland, United States of America
| |
Collapse
|
50
|
Laupèze B, Doherty TM. Maintaining a 'fit' immune system: the role of vaccines. Expert Rev Vaccines 2023; 22:256-266. [PMID: 36864769 DOI: 10.1080/14760584.2023.2185223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
INTRODUCTION Conventionally, vaccines are thought to induce a specific immune response directed against a target pathogen. Long recognized but poorly understood nonspecific benefits of vaccination, such as reduced susceptibility to unrelated diseases or cancer, are now being investigated and may be due in part to "trained immunity'. AREAS COVERED We discuss 'trained immunity' and whether vaccine-induced 'trained immunity' could be leveraged to prevent morbidity due to a broader range of causes. EXPERT OPINION The prevention of infection i.e. maintaining homeostasis by preventing the primary infection and resulting secondary illnesses, is the pivotal strategy used to direct vaccine design and may have long-term, positive impacts on health at all ages. In the future, we anticipate that vaccine design will change to not only prevent the target infection (or related infections) but to generate positive modifications to the immune response that could prevent a wider range of infections and potentially reduce the impact of immunological changes associated with aging. Despite changing demographics, adult vaccination has not always been prioritized. However, the SARS-CoV-2 pandemic has demonstrated that adult vaccination can flourish given the right circumstances, demonstrating that harnessing the potential benefits of life-course vaccination is achievable for all.
Collapse
|