1
|
Wu O, Gao J, Zhang X, Liu W, Zhang H, Khederzadeh S, Lu X, Wu Y. TLR5's Role in Obesity-related Hypertension: Updated Evidence and Prospects. Angiology 2025:33197251326384. [PMID: 40079382 DOI: 10.1177/00033197251326384] [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: 03/15/2025]
Abstract
Toll-like receptor 5 (TLR5), integral to the immune system as a primary sensor for flagellin, is central to the link between innate and adaptive immunity, modulating immune responses and cytokine production essential for defense against flagellated pathogens and immune tolerance. This review consolidates the understanding of TLR5's structural and signaling mechanisms and its interactions with flagellin, shedding light on its dual role in immune responses and its promise as a therapeutic target. It highlights TLR5's intricate role in the pathogenesis of obesity-related hypertension, a growing global health concern that correlates with rising obesity rates and is characterized by a complex interplay of immune responses and metabolic dysregulation. Despite the current understanding, the impact of TLR5 on obesity-related hypertension is marked by conflicting findings, indicating a need for further exploration. The review critically analyzes the existing literature, providing novel insights from rodent models and human studies that underscore TLR5's therapeutic potential, setting the stage for transformative research in managing obesity-related hypertension. It calls for deeper investigation into TLR5's multifaceted role, emphasizing its promise as a target for managing obesity-related hypertension and the necessity for future research to clarify its complexities and to innovate treatment strategies.
Collapse
Affiliation(s)
- Ou Wu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, P.R. China
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Jin Gao
- Clinical Laboratory, the Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, P.R. China
| | - Xingyu Zhang
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Wei Liu
- JFIntelligent Healthcare Technology Co. Ltd, Nanchang, Jiangxi, P.R. China
| | - Hu Zhang
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital Affiliated with Medical College of Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Saber Khederzadeh
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, P.R. China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, P.R. China
| | - Xi Lu
- Hangzhou Vocational and Technical College, Hangzhou, Zhejiang, P.R. China
| | - Ya Wu
- Anhui Medical University, Hefei, Anhui, P.R. China
| |
Collapse
|
2
|
Khunger S, Mewara A, Kaur U, Duseja A, Ray P, Kalra N, Sharma N, Sehgal R. Toll-like receptor upregulation in liver and peripheral blood mononuclear cells of patients with amoebic liver abscess. Immunobiology 2025; 230:152869. [PMID: 39919544 DOI: 10.1016/j.imbio.2025.152869] [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: 05/21/2024] [Revised: 12/24/2024] [Accepted: 01/08/2025] [Indexed: 02/09/2025]
Abstract
AIM We aimed to understand the host and microbe interactions at the time of infection and inflammatory response in amoebic liver abscess (ALA) patients based on toll-like receptor (TLR) expression (mRNA), cytokine and IgG subtypes levels. METHODS AND RESULTS Liver aspirates from 100 ALA patients and 11 liver autopsy samples were used as negative controls. Blood samples from 100 ALA and 41 healthy individuals were collected. mRNA expression of TLR 1 to 9 genes was measured using reverse transcriptase polymerase chain reaction (RT-PCR). Serum cytokines level was quantified by flow cytometry. In-house ELISA for the analysis of IgG and its subtypes in the serum samples was performed. A total of 7 TLR genes (TLR1, TLR2, TLR4, TLR6, TLR7, TLR8 and TLR9) and 6 TLR genes (TLR1, TLR2, TLR3, TLR4, TLR5 and TLR8) were found to be elevated in liver aspirates and PBMCs respectively. Increased serum cytokine levels were observed in ALA patients vs. healthy controls. Interestingly, a significant increase in IgG and its subtypes (IgG1, IgG3 and IgG4) was found in the serum of ALA patients. CONCLUSION Increased levels of TLR, pro- and anti-inflammatory cytokines, IgG and its subtypes, are possibly linked with early-stage infection in ALA patients. IMPACT STATEMENT The role of TLRs in association with ALA might provide insights into new therapeutic strategies.
Collapse
Affiliation(s)
- Sandhya Khunger
- Department of Medical Parasitology, Postgraduate Institute of Medical Education and Research, Chandigarh, India; Department of Microbiology (FAHS), Shree Guru Gobind Singh Tricentenary (SGT) University, Gurugram, (Haryana) 122505,India
| | - Abhishek Mewara
- Department of Medical Parasitology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Upninder Kaur
- Department of Medical Parasitology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ajay Duseja
- Department of Hepatology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Pallab Ray
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Naveen Kalra
- Department of Radiodiagnosis and Imaging, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Navneet Sharma
- Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rakesh Sehgal
- Department of Microbiology, Aarupadai Veed Medical College and Hospital, Vinayaka Mission's Research Foundation- DU, Pondicherry, India.
| |
Collapse
|
3
|
Heinl PV, Graulich E, Weigmann B, Wangorsch A, Ose R, Bellinghausen I, Khatri R, Raker VK, Scheurer S, Vieths S, Saloga J, Steinbrink K. IL-10-modulated dendritic cells from birch pollen- and hazelnut-allergic patients facilitate Treg-mediated allergen-specific and cross-reactive tolerance. Allergy 2024; 79:2826-2839. [PMID: 39073174 DOI: 10.1111/all.16255] [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: 11/16/2023] [Revised: 06/19/2024] [Accepted: 07/06/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Approximately 70% of individuals allergic to birch pollen (Bet v 1.01 [Bet]) develop a secondary food allergy (e.g., hazelnut: Cor a 1.04 [Cor]), due to allergen cross-reactivity. However, standard immunotherapy for type I allergies often does not improve the food allergy sufficiently. We analyzed the allergen-specific and cross-reactive suppressive capacity of primary human regulatory T cells (Treg) induced by autologous IL-10-modulated dendritic cells (IL-10 DC) in vitro and in vivo. METHODS CD4+ T cells of patients with birch pollen and associated hazelnut allergies were differentiated into Bet-specific or non-specific induced Treg (iTreg). After Bet- or Cor-specific restimulation the phenotype, proliferation, and suppressive capacity of iTreg subsets were analyzed. iTreg function was further investigated in humanized mouse models of airway and intestinal allergy, generated by engraftment of peripheral blood mononuclear cells from allergic donors into immunodeficient animals. RESULTS After IL-10 DC priming and allergen-specific restimulation (Bet or Cor), non-specific control iTreg remained anergic, whereas Bet-specific iTreg proliferated extensively and exhibited a regulatory phenotype (enhanced expression of CTLA-4, PD-1, TNFR2, IL-10). Accordingly, activated Bet-specific iTreg displayed a high capacity to suppress Bet- and Cor-induced responder Th2 cell responses in vitro, indicating induction of both allergen-specific (birch) and cross-reactive tolerance (hazelnut). In vivo, the beneficial effect of Bet-specific iTreg was verified in humanized mouse models of allergic airway and intestinal inflammation, resulting in reduced allergen-induced clinical symptoms, and immune responses. CONCLUSION Human IL-10 DC-induced iTreg facilitate allergen-specific and cross-reactive tolerance. Therefore, they are potential candidates for regulatory cell therapy in allergic and autoimmune diseases.
Collapse
Affiliation(s)
- Patricia Vanessa Heinl
- Department of Dermatology, Division for Experimental and Translational Research, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Edith Graulich
- Department of Dermatology, Division for Experimental and Translational Research, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Benno Weigmann
- Department of Internal Medicine, University Hospital Erlangen, University Erlangen-Nürnberg, Erlangen, Germany
| | | | - Robert Ose
- Department of Dermatology, University Medical Center of the Johannes, Gutenberg-University, Mainz, Germany
| | - Iris Bellinghausen
- Department of Dermatology, University Medical Center of the Johannes, Gutenberg-University, Mainz, Germany
| | - Rahul Khatri
- Department of Dermatology, University Medical Center of the Johannes, Gutenberg-University, Mainz, Germany
| | - Verena K Raker
- Department of Dermatology, University Hospital Münster, University of Münster, Münster, Germany
| | | | - Stefan Vieths
- Molecular Allergology, Paul-Ehrlich-Institute, Langen, Germany
| | - Joachim Saloga
- Department of Dermatology, University Medical Center of the Johannes, Gutenberg-University, Mainz, Germany
| | - Kerstin Steinbrink
- Department of Dermatology, University Hospital Münster, University of Münster, Münster, Germany
| |
Collapse
|
4
|
Medina-Rodríguez EM, Martínez-Raga J, Sanz Y. Intestinal Barrier, Immunity and Microbiome: Partners in the Depression Crime. Pharmacol Rev 2024; 76:956-969. [PMID: 39084934 DOI: 10.1124/pharmrev.124.001202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 08/02/2024] Open
Abstract
Depression is a highly prevalent disorder and a leading cause of disability worldwide. It has a major impact on the affected individual and on society as a whole. Regrettably, current available treatments for this condition are insufficient in many patients. In recent years, the gut microbiome has emerged as a promising alternative target for treating and preventing depressive disorders. However, the microbes that form this ecosystem do not act alone but are part of a complicated network connecting the gut and the brain that influences our mood. Host cells that are in intimate contact with gut microbes, such as the epithelial cells forming the gut barrier and the immune cells in their vicinity, play a key role in the process. These cells continuously shape immune responses to maintain healthy communication between gut microbes and the host. In this article, we review how the interplay among epithelial cells, the immune system, and gut microbes mediates gut-brain communication to influence mood. We also discuss how advances in our knowledge of the mechanisms underlying the gut-brain axis could contribute to addressing depression. SIGNIFICANCE STATEMENT: This review does not aim to systematically describe intestinal microbes that might be beneficial or detrimental for depression. We have adopted a novel point of view by focusing on potential mechanisms underlying the crosstalk between gut microbes and their intestinal environment to control mood. These pathways could be targeted by well defined and individually tailored dietary interventions, microbes, or microbial metabolites to ameliorate depression and decrease its important social and economic impact.
Collapse
Affiliation(s)
- Eva M Medina-Rodríguez
- Psychiatry Service, Doctor Peset University Hospital, FISABIO, Valencia, Spain (E.M.M.-R., J.M.-R.); Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain (E.M.M.-R., Y.S.); and University of Valencia, Valencia, Spain (J.M.-R.)
| | - José Martínez-Raga
- Psychiatry Service, Doctor Peset University Hospital, FISABIO, Valencia, Spain (E.M.M.-R., J.M.-R.); Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain (E.M.M.-R., Y.S.); and University of Valencia, Valencia, Spain (J.M.-R.)
| | - Yolanda Sanz
- Psychiatry Service, Doctor Peset University Hospital, FISABIO, Valencia, Spain (E.M.M.-R., J.M.-R.); Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain (E.M.M.-R., Y.S.); and University of Valencia, Valencia, Spain (J.M.-R.)
| |
Collapse
|
5
|
Wu Z, Liu X, Huang W, Chen J, Li S, Chao J, Xie J, Liu L, Yang Y, Wu X, Qiu H. CIRP increases Foxp3 + regulatory T cells and inhibits development of Th17 cells by enhancing TLR4-IL-2 signaling in the late phase of sepsis. Int Immunopharmacol 2024; 132:111924. [PMID: 38531201 DOI: 10.1016/j.intimp.2024.111924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/14/2024] [Accepted: 03/21/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND T helper (Th) cell imbalances have been associated with the pathophysiology of sepsis, including the Th1/Th2 and Th17/T regulatory cells (Treg) paradigms. Cold-inducible RNA-binding protein (CIRP), a novel damage-associated molecular pattern (DAMP) was reported that could induce T cell activation, and skew CD4+ T cells towards a Th1 profile. However, the effect and underlying mechanisms of CIRP on Th17/Treg differentiation in sepsis still remains unknown. METHODS A prospective exploratory study including patients with sepsis was conducted. Blood samples were collected from patients on days 0, 3 and 7 on admission. The serum CIRP and peripheral blood Treg/Th17 percentage was determined by ELISA and flow cytometry. CD4+ T cells from the spleen and lymph nodes of mice with experimental sepsis were collected after treatment with normal saline (NS), recombinant murine CIRP (rmCIRP) and C23 (an antagonist for CIRP-TLR4) at late stage of sepsis. RNA-seq was conducted to reveal the pivotal molecular mechanism of CIRP on Treg/Th17 differentiation. Naïve CD4+ T cell was isolated from the Tlr4 null and wildtype mice in the presence or absence rmCIRP and C23 to confirmed above findings. RESULTS A total of 19 patients with sepsis finally completed the study. Serum CIRP levels remained high in the majority of patients up to 1 week after admittance was closely associated with high Treg/Th17 ratio of peripheral blood and poor outcome. A univariate logistic analysis demonstrated that higher CIRP concentration at Day 7 is an independent risk factor for Treg/Th17 ratio increasing. CIRP promotes Treg development and suppresses Th17 differentiation was found both in vivo and in vitro. Pretreated with C23 not only alleviated the majority of negative effect of CIRP on Th17 differentiation, but also inhibited Treg differentiation, to some extent. Tlr4 deficiency could abolish almost all downstream effects of rmCIRP. Furthermore, IL-2 is proved a key downstream molecules of the effect CIRP, which also could amplify the activated CD4+ T lymphocytes. CONCLUSIONS Persistent high circulating CIRP level may lead to Treg/Th17 ratio elevated through TLR4 and subsequent active IL-2 signaling which contribute to immunosuppression during late phases of sepsis.
Collapse
Affiliation(s)
- Zongsheng Wu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xu Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Wei Huang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jing Chen
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Songli Li
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jie Chao
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jianfeng Xie
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Ling Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yi Yang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xiaojing Wu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Haibo Qiu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
6
|
Carroll SL, Pasare C, Barton GM. Control of adaptive immunity by pattern recognition receptors. Immunity 2024; 57:632-648. [PMID: 38599163 PMCID: PMC11037560 DOI: 10.1016/j.immuni.2024.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 04/12/2024]
Abstract
One of the most significant conceptual advances in immunology in recent history is the recognition that signals from the innate immune system are required for induction of adaptive immune responses. Two breakthroughs were critical in establishing this paradigm: the identification of dendritic cells (DCs) as the cellular link between innate and adaptive immunity and the discovery of pattern recognition receptors (PRRs) as a molecular link that controls innate immune activation as well as DC function. Here, we recount the key events leading to these discoveries and discuss our current understanding of how PRRs shape adaptive immune responses, both indirectly through control of DC function and directly through control of lymphocyte function. In this context, we provide a conceptual framework for how variation in the signals generated by PRR activation, in DCs or other cell types, can influence T cell differentiation and shape the ensuing adaptive immune response.
Collapse
Affiliation(s)
- Shaina L Carroll
- Division of Immunology & Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| | - Chandrashekhar Pasare
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH USA
| | - Gregory M Barton
- Division of Immunology & Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA; Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720 USA.
| |
Collapse
|
7
|
Malik JA, Kaur G, Agrewala JN. Revolutionizing medicine with toll-like receptors: A path to strengthening cellular immunity. Int J Biol Macromol 2023; 253:127252. [PMID: 37802429 DOI: 10.1016/j.ijbiomac.2023.127252] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 10/01/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023]
Abstract
Toll-like receptors play a vital role in cell-mediated immunity, which is crucial for the immune system's defense against pathogens and maintenance of homeostasis. The interaction between toll-like-receptor response and cell-mediated immunity is complex and essential for effectively eliminating pathogens and maintaining immune surveillance. In addition to pathogen recognition, toll-like receptors serve as adjuvants in vaccines, as molecular sensors, and recognize specific patterns associated with pathogens and danger signals. Incorporating toll-like receptor ligands into vaccines can enhance the immune response to antigens, making them potent adjuvants. Furthermore, they bridge the innate and adaptive immune systems and improve antigen-presenting cells' capacity to process and present antigens to T cells. The intricate signaling pathways and cross-talk between toll-like-receptor and T cell receptor (TCR) signaling emphasize their pivotal role in orchestrating effective immune responses against pathogens, thus facilitating the development of innovative vaccine strategies. This article provides an overview of the current understanding of toll-like receptor response and explores their potential clinical applications. By unraveling the complex mechanisms of toll-like-receptor signaling, we can gain novel insights into immune responses and potentially develop innovative therapeutic approaches. Ongoing investigations into the toll-like-receptor response hold promise in the future in enhancing our ability to combat infections, design effective vaccines, and improve clinical outcomes.
Collapse
Affiliation(s)
- Jonaid Ahmad Malik
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology, Ropar, Punjab 140001, India
| | - Gurpreet Kaur
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology, Ropar, Punjab 140001, India; Department of Biotechnology, Chandigarh Group of Colleges, Landran, Mohali, Punjab 140055, India
| | - Javed N Agrewala
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology, Ropar, Punjab 140001, India.
| |
Collapse
|
8
|
Danaher P, Hasle N, Nguyen ED, Hayward K, Rosenwasser N, Alpers CE, Reed RC, Okamura DM, Baxter SK, Jackson SW. Single cell spatial transcriptomic profiling of childhood-onset lupus nephritis reveals complex interactions between kidney stroma and infiltrating immune cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.09.566503. [PMID: 38014158 PMCID: PMC10680641 DOI: 10.1101/2023.11.09.566503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Children with systemic lupus erythematosus (SLE) are at increased risk of developing kidney disease, termed childhood-onset lupus nephritis (cLN). Single cell transcriptomics of dissociated kidney tissue has advanced our understanding of LN pathogenesis, but loss of spatial resolution prevents interrogation of in situ cellular interactions. Using a technical advance in spatial transcriptomics, we generated a spatially resolved, single cell resolution atlas of kidney tissue (>400,000 cells) from eight cLN patients and two controls. Annotated cells were assigned to 35 reference cell types, including major kidney subsets and infiltrating immune cells. Analysis of spatial distribution demonstrated that individual immune lineages localize to specific regions in cLN kidneys, including myeloid cells trafficking to inflamed glomeruli and B cells clustering within tubulointerstitial immune hotspots. Notably, gene expression varied as a function of tissue location, demonstrating how incorporation of spatial data can provide new insights into the immunopathogenesis of SLE. Alterations in immune phenotypes were accompanied by parallel changes in gene expression by resident kidney stromal cells. However, there was little correlation between histologic scoring of cLN disease activity and glomerular cell transcriptional signatures at the level of individual glomeruli. Finally, we identified modules of spatially-correlated gene expression with predicted roles in induction of inflammation and the development of tubulointerstitial fibrosis. In summary, single cell spatial transcriptomics allows unprecedented insights into the molecular heterogeneity of cLN, paving the way towards more targeted and personalized treatment approaches.
Collapse
Affiliation(s)
| | - Nicholas Hasle
- Department of Pediatrics, University of Washington School of Medicine; Seattle, WA, USA
| | - Elizabeth D. Nguyen
- Department of Pediatrics, University of Washington School of Medicine; Seattle, WA, USA
- Seattle Children’s Research Institute, Seattle, WA, USA
| | - Kristen Hayward
- Department of Pediatrics, University of Washington School of Medicine; Seattle, WA, USA
- Seattle Children’s Research Institute, Seattle, WA, USA
| | - Natalie Rosenwasser
- Department of Pediatrics, University of Washington School of Medicine; Seattle, WA, USA
- Seattle Children’s Research Institute, Seattle, WA, USA
| | - Charles E. Alpers
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine; Seattle, WA, USA
| | - Robyn C. Reed
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine; Seattle, WA, USA
| | - Daryl M. Okamura
- Department of Pediatrics, University of Washington School of Medicine; Seattle, WA, USA
- Seattle Children’s Research Institute, Seattle, WA, USA
| | - Sarah K. Baxter
- Department of Pediatrics, University of Washington School of Medicine; Seattle, WA, USA
- Sanofi US, Bridgewater, NJ, USA
| | - Shaun W. Jackson
- Department of Pediatrics, University of Washington School of Medicine; Seattle, WA, USA
- Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine; Seattle, WA, USA
| |
Collapse
|
9
|
Wan T, Wang Y, He K, Zhu S. Microbial sensing in the intestine. Protein Cell 2023; 14:824-860. [PMID: 37191444 PMCID: PMC10636641 DOI: 10.1093/procel/pwad028] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/04/2023] [Indexed: 05/17/2023] Open
Abstract
The gut microbiota plays a key role in host health and disease, particularly through their interactions with the immune system. Intestinal homeostasis is dependent on the symbiotic relationships between the host and the diverse gut microbiota, which is influenced by the highly co-evolved immune-microbiota interactions. The first step of the interaction between the host and the gut microbiota is the sensing of the gut microbes by the host immune system. In this review, we describe the cells of the host immune system and the proteins that sense the components and metabolites of the gut microbes. We further highlight the essential roles of pattern recognition receptors (PRRs), the G protein-coupled receptors (GPCRs), aryl hydrocarbon receptor (AHR) and the nuclear receptors expressed in the intestinal epithelial cells (IECs) and the intestine-resident immune cells. We also discuss the mechanisms by which the disruption of microbial sensing because of genetic or environmental factors causes human diseases such as the inflammatory bowel disease (IBD).
Collapse
Affiliation(s)
- Tingting Wan
- Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Institute of Immunology, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Yalong Wang
- Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Institute of Immunology, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Kaixin He
- Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Institute of Immunology, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Shu Zhu
- Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Institute of Immunology, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
- Department of Digestive Disease, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei 230001, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230601, China
| |
Collapse
|
10
|
Riaz F, Huang Z, Pan F. Targeting post-translational modifications of Foxp3: a new paradigm for regulatory T cell-specific therapy. Front Immunol 2023; 14:1280741. [PMID: 37936703 PMCID: PMC10626496 DOI: 10.3389/fimmu.2023.1280741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/09/2023] [Indexed: 11/09/2023] Open
Abstract
A healthy immune system is pivotal for the hosts to resist external pathogens and maintain homeostasis; however, the immunosuppressive tumor microenvironment (TME) damages the anti-tumor immunity and promotes tumor progression, invasion, and metastasis. Recently, many studies have found that Foxp3+ regulatory T (Treg) cells are the major immunosuppressive cells that facilitate the formation of TME by promoting the development of various tumor-associated cells and suppressing the activity of effector immune cells. Considering the role of Tregs in tumor progression, it is pivotal to identify new therapeutic drugs to target and deplete Tregs in tumors. Although several studies have developed strategies for targeted deletion of Treg to reduce the TME and support the accumulation of effector T cells in tumors, Treg-targeted therapy systematically affects the Treg population and may lead to the progression of autoimmune diseases. It has been understood that, nevertheless, in disease conditions, Foxp3 undergoes several definite post-translational modifications (PTMs), including acetylation, glycosylation, phosphorylation, ubiquitylation, and methylation. These PTMs not only elevate or mitigate the transcriptional activity of Foxp3 but also affect the stability and immunosuppressive function of Tregs. Various studies have shown that pharmacological targeting of enzymes involved in PTMs can significantly influence the PTMs of Foxp3; thus, it may influence the progression of cancers and/or autoimmune diseases. Overall, this review will help researchers to understand the advances in the immune-suppressive mechanisms of Tregs, the post-translational regulations of Foxp3, and the potential therapeutic targets and strategies to target the Tregs in TME to improve anti-tumor immunity.
Collapse
Affiliation(s)
| | | | - Fan Pan
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| |
Collapse
|
11
|
Jiwrajka N, Toothacre NE, Beethem ZT, Sting S, Forsyth KS, Dubin AH, Driscoll A, Stohl W, Anguera MC. Impaired dynamic X-chromosome inactivation maintenance in T cells is a feature of spontaneous murine SLE that is exacerbated in female-biased models. J Autoimmun 2023; 139:103084. [PMID: 37399593 PMCID: PMC11140471 DOI: 10.1016/j.jaut.2023.103084] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/14/2023] [Indexed: 07/05/2023]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is a highly female-biased systemic autoimmune disease, but the molecular basis for this female bias remains incompletely elucidated. B and T lymphocytes from patients with SLE and female-biased mouse models of SLE exhibit features of epigenetic dysregulation on the X chromosome which may contribute to this strong female bias. We therefore examined the fidelity of dynamic X-chromosome inactivation maintenance (dXCIm) in the pathogenesis of two murine models of spontaneous lupus-NZM2328 and MRL/lpr-with disparate levels of female-bias to determine whether impaired dXCIm contributes to the female bias of disease. METHODS CD23+ B cells and CD3+ T cells were purified from age-matched C57BL/6 (B6), MRL/lpr, and NZM2328 male and female mice, activated in vitro, and processed for Xist RNA fluorescence in situ hybridization, H3K27me3 immunofluorescence imaging, qPCR, and RNA sequencing analyses. RESULTS The dynamic relocalization of Xist RNA and the canonical heterochromatin mark, H3K27me3, to the inactive X chromosome was preserved in CD23+ B cells, but impaired in activated CD3+ T cells from the MRL/lpr model (p < 0.01 vs. B6), and even more impaired in the heavily female-biased NZM2328 model (p < 0.001 vs. B6; p < 0.05 vs. MRL/lpr). RNAseq of activated T cells from NZM2328 mice revealed the female-biased upregulation of 32 X-linked genes distributed broadly across the X chromosome, many of which have roles in immune function. Many genes encoding Xist RNA-interacting proteins were also differentially expressed and predominantly downregulated, which may account for the observed mislocalization of Xist RNA to the inactive X chromosome. CONCLUSIONS Although evident in T cells from both the MRL/lpr and NZM2328 models of spontaneous SLE, impaired dXCIm is more severe in the heavily female-biased NZM2328 model. The aberrant X-linked gene dosage in female NZM2328 mice may contribute towards the development of female-biased immune responses in SLE-prone hosts. These findings provide important insights into the epigenetic mechanisms contributing to female-biased autoimmunity.
Collapse
Affiliation(s)
- Nikhil Jiwrajka
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA; Division of Rheumatology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Natalie E Toothacre
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Zachary T Beethem
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Sarah Sting
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Katherine S Forsyth
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Aimee H Dubin
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Amanda Driscoll
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - William Stohl
- Division of Rheumatology, Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Montserrat C Anguera
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Carmona-Pérez L, Dagenais-Lussier X, Mai LT, Stögerer T, Swaminathan S, Isnard S, Rice MR, Barnes BJ, Routy JP, van Grevenynghe J, Stäger S. The TLR7/IRF-5 axis sensitizes memory CD4+ T cells to Fas-mediated apoptosis during HIV-1 infection. JCI Insight 2023; 8:e167329. [PMID: 37227774 PMCID: PMC10371351 DOI: 10.1172/jci.insight.167329] [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: 11/18/2022] [Accepted: 05/23/2023] [Indexed: 05/27/2023] Open
Abstract
HIV-1 infection is characterized by inflammation and a progressive decline in CD4+ T cell count. Despite treatment with antiretroviral therapy (ART), the majority of people living with HIV (PLWH) maintain residual levels of inflammation, a low degree of immune activation, and higher sensitivity to cell death in their memory CD4+ T cell compartment. To date, the mechanisms responsible for this high sensitivity remain elusive. We have identified the transcription factor IRF-5 to be involved in impairing the maintenance of murine CD4+ T cells during chronic infection. Here, we investigate whether IRF-5 also contributes to memory CD4+ T cell loss during HIV-1 infection. We show that TLR7 and IRF-5 were upregulated in memory CD4+ T cells from PLWH, when compared with naturally protected elite controllers and HIVfree participants. TLR7 was upstream of IRF-5, promoting Caspase 8 expression in CD4+ T cells from ART HIV-1+ but not from HIVfree donors. Interestingly, the TLR7/IRF-5 axis acted synergistically with the Fas/FasL pathway, suggesting that TLR7 and IRF-5 expression in ART HIV-1+ memory CD4+ T cells represents an imprint that predisposes cells to Fas-mediated apoptosis. This predisposition could be blocked using IRF-5 inhibitory peptides, suggesting IRF-5 blockade as a possible therapy to prevent memory CD4+ T cell loss in PLWH.
Collapse
Affiliation(s)
- Liseth Carmona-Pérez
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, and Infectiopôle-INRS, Laval, Quebec, Canada
| | - Xavier Dagenais-Lussier
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, and Infectiopôle-INRS, Laval, Quebec, Canada
| | - Linh T. Mai
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, and Infectiopôle-INRS, Laval, Quebec, Canada
| | - Tanja Stögerer
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, and Infectiopôle-INRS, Laval, Quebec, Canada
| | - Sharada Swaminathan
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, and Infectiopôle-INRS, Laval, Quebec, Canada
| | - Stéphane Isnard
- Division of Hematology and Chronic Viral Illness Service, McGill University Health Centre, Montreal, Quebec, Canada
| | - Matthew R. Rice
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Betsy J. Barnes
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Jean-Pierre Routy
- Division of Hematology and Chronic Viral Illness Service, McGill University Health Centre, Montreal, Quebec, Canada
| | - Julien van Grevenynghe
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, and Infectiopôle-INRS, Laval, Quebec, Canada
| | - Simona Stäger
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, and Infectiopôle-INRS, Laval, Quebec, Canada
| |
Collapse
|
13
|
Morita N, Hoshi M, Tezuka H, Ando T, Yoshida S, Sato F, Yokoi H, Ito H, Saito K. CD8+ Regulatory T Cells Induced by Lipopolysaccharide Improve Mouse Endotoxin Shock. Immunohorizons 2023; 7:353-363. [PMID: 37212786 PMCID: PMC10579971 DOI: 10.4049/immunohorizons.2200074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 05/03/2023] [Indexed: 05/23/2023] Open
Abstract
Sepsis is a systemic inflammatory disease caused by a bacterial infection that leads to severe mortality, especially in elderly patients, because of an excessive immune response and impaired regulatory functions. Antibiotic treatment is widely accepted as the first-line therapy for sepsis; however, its excessive use has led to the emergence of multidrug-resistant bacteria in patients with sepsis. Therefore, immunotherapy may be effective in treating sepsis. Although CD8+ regulatory T cells (Tregs) are known to have immunomodulatory effects in various inflammatory diseases, their role during sepsis remains unclear. In this study, we investigated the role of CD8+ Tregs in an LPS-induced endotoxic shock model in young (8-12 wk old) and aged (18-20 mo old) mice. The adoptive transfer of CD8+ Tregs into LPS-treated young mice improved the survival rate of LPS-induced endotoxic shock. Moreover, the number of CD8+ Tregs in LPS-treated young mice increased through the induction of IL-15 produced by CD11c+ cells. In contrast, LPS-treated aged mice showed a reduced induction of CD8+ Tregs owing to the limited production of IL-15. Furthermore, CD8+ Tregs induced by treatment with the rIL-15/IL-15Rα complex prevented LPS-induced body wight loss and tissue injury in aged mice. In this study, to our knowledge, the induction of CD8+ Tregs as novel immunotherapy or adjuvant therapy for endotoxic shock might reduce the uncontrolled immune response and ultimately improve the outcomes of endotoxic shock.
Collapse
Affiliation(s)
- Nanaka Morita
- Department of Disease Control and Prevention, Fujita Health University, Toyoake, Aichi, Japan
| | - Masato Hoshi
- Department of Disease Control and Prevention, Fujita Health University, Toyoake, Aichi, Japan
- Department of Informative Clinical Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Hiroyuki Tezuka
- Cellular Function Analysis, Research Promotion Headquarters, Fujita Health University, Toyoake, Aichi, Japan
| | - Tatsuya Ando
- Joint Research Laboratory of Clinical Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Sayaka Yoshida
- Department of Informative Clinical Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Fumiaki Sato
- Department of Informative Clinical Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Hiroyuki Yokoi
- Department of Informative Clinical Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Hiroyasu Ito
- Cellular Function Analysis, Research Promotion Headquarters, Fujita Health University, Toyoake, Aichi, Japan
| | - Kuniaki Saito
- Department of Disease Control and Prevention, Fujita Health University, Toyoake, Aichi, Japan
| |
Collapse
|
14
|
Znaidia M, de Souza-Angelo Y, Létoffé S, Staropoli I, Grzelak L, Ghigo JM, Schwartz O, Casartelli N. Exposure to Secreted Bacterial Factors Promotes HIV-1 Replication in CD4 + T Cells. Microbiol Spectr 2023; 11:e0431322. [PMID: 36853052 PMCID: PMC10100953 DOI: 10.1128/spectrum.04313-22] [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: 10/21/2022] [Accepted: 02/08/2023] [Indexed: 03/01/2023] Open
Abstract
Microbial translocation is associated with systemic immune activation in HIV-1 disease. Circulating T cells can encounter microbial products in the bloodstream and lymph nodes, where viral replication takes place. The mechanisms by which bacteria contribute to HIV-associated pathogenesis are not completely deciphered. Here, we examined how bacteria may impact T cell function and viral replication. We established cocultures between a panel of live bacteria and uninfected or HIV-1-infected activated peripheral blood CD4-positive (CD4+) T cells. We show that some bacteria, such as Escherichia coli and Acinetobacter baumannii, sustain lymphocyte activation and enhance HIV-1 replication. Bacteria secrete soluble factors that upregulate CD25 and ICAM-1 cell surface levels and activate NF-κB nuclear translocation. Our data also demonstrate that CD25 polarizes at the virological synapse, suggesting a previously unappreciated role of CD25 during viral replication. These findings highlight how interactions between bacterial factors and T cells may promote T cell activation and HIV-1 replication. IMPORTANCE People living with HIV suffer from chronic immune activation despite effective antiretroviral therapy. Early after infection, HIV-1 actively replicates in the gut, causing the breakage of the intestinal epithelial barrier and microbial translocation. Microbial translocation and chronic immune activation have been proven linked; however, gaps in our knowledge on how bacteria contribute to the development of HIV-related diseases remain. Whether T cells in the peripheral blood react to bacterial products and how this affects viral replication are unknown. We show that some bacteria enriched in people living with HIV activate T cells and favor HIV-1's spread. Bacteria release soluble factors that cause the overexpression of cellular molecules related to their activation state. T cells overexpressing these molecules also replicate HIV-1 more efficiently. These results help us learn more about how HIV-1, T cells, and bacteria interact with each other, as well as the mechanisms behind chronic immune activation.
Collapse
Affiliation(s)
- M. Znaidia
- Institut Pasteur, Université Paris-Cité, UMR CNRS 3569, Virus and Immunity Unit, Paris, France
| | - Y. de Souza-Angelo
- Institut Pasteur, Université Paris-Cité, UMR CNRS 3569, Virus and Immunity Unit, Paris, France
| | - S. Létoffé
- Institut Pasteur, Université Paris-Cité, UMR CNRS 6047, Genetics of Biofilms Laboratory, Paris, France
| | - I. Staropoli
- Institut Pasteur, Université Paris-Cité, UMR CNRS 3569, Virus and Immunity Unit, Paris, France
| | - L. Grzelak
- Institut Pasteur, Université Paris-Cité, UMR CNRS 3569, Virus and Immunity Unit, Paris, France
| | - J. M. Ghigo
- Institut Pasteur, Université Paris-Cité, UMR CNRS 6047, Genetics of Biofilms Laboratory, Paris, France
| | - O. Schwartz
- Institut Pasteur, Université Paris-Cité, UMR CNRS 3569, Virus and Immunity Unit, Paris, France
- Vaccine Research Institute, Créteil, France
| | - N. Casartelli
- Institut Pasteur, Université Paris-Cité, UMR CNRS 3569, Virus and Immunity Unit, Paris, France
| |
Collapse
|
15
|
vom Werth KL, Kemper B, Kampmeier S, Mellmann A. Application of Digital Holographic Microscopy to Analyze Changes in T-Cell Morphology in Response to Bacterial Challenge. Cells 2023; 12:cells12050762. [PMID: 36899897 PMCID: PMC10000559 DOI: 10.3390/cells12050762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/16/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
Abstract
Quantitative phase imaging (QPI) is a non-invasive, label-free technique used to detect aberrant cell morphologies caused by disease, thus providing a useful diagnostic approach. Here, we evaluated the potential of QPI to differentiate specific morphological changes in human primary T-cells exposed to various bacterial species and strains. Cells were challenged with sterile bacterial determinants, i.e., membrane vesicles or culture supernatants, derived from different Gram-positive and Gram-negative bacteria. Timelapse QPI by digital holographic microscopy (DHM) was applied to capture changes in T-cell morphology over time. After numerical reconstruction and image segmentation, we calculated single cell area, circularity and mean phase contrast. Upon bacterial challenge, T-cells underwent rapid morphological changes such as cell shrinkage, alterations of mean phase contrast and loss of cell integrity. Time course and intensity of this response varied between both different species and strains. The strongest effect was observed for treatment with S. aureus-derived culture supernatants that led to complete lysis of the cells. Furthermore, cell shrinkage and loss of circular shape was stronger in Gram-negative than in Gram-positive bacteria. Additionally, T-cell response to bacterial virulence factors was concentration-dependent, as decreases in cellular area and circularity were enhanced with increasing concentrations of bacterial determinants. Our findings clearly indicate that T-cell response to bacterial stress depends on the causative pathogen, and specific morphological alterations can be detected using DHM.
Collapse
Affiliation(s)
| | - Björn Kemper
- Biomedical Technology Center of the Medical Faculty, University of Münster, 48149 Münster, Germany
| | - Stefanie Kampmeier
- Institute of Hygiene, University Hospital Münster, 48149 Münster, Germany
| | - Alexander Mellmann
- Institute of Hygiene, University Hospital Münster, 48149 Münster, Germany
- Correspondence: ; Tel.: +49-251-83-55361
| |
Collapse
|
16
|
Irahara M, Yamamoto-Hanada K, Sato M, Saito-Abe M, Miyaji Y, Yang L, Nishizato M, Kumasaka N, Mezawa H, Ohya Y. Endotoxin concentration and persistent eczema in early childhood. J Dermatol 2022; 50:646-655. [PMID: 36578125 DOI: 10.1111/1346-8138.16686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 11/16/2022] [Accepted: 12/05/2022] [Indexed: 12/30/2022]
Abstract
Although endotoxin concentration in the environment is negatively associated with atopic dermatitis (AD) onset in early childhood, the association between endotoxin concentration in the environment and eczema resolution in children with preexisting eczema is unclear. The aim of this study was to evaluate the association between endotoxin concentration in house dust and eczema persistence in young children. The authors used data from children participating in JECS (Japan Environment and Children's Study). In children who had AD or AD-like lesions at the age of 1 year, the authors investigated the association between the prevalence of eczema at the age of 3 years and endotoxin concentration (categorized by quartiles) in the dust on children's mattresses at the ages of 1.5 and 3 years. This study included 605 children. Eczema was significantly less prevalent among children whose mattresses were in the second and third quartiles of endotoxin concentration when they were 18 months old than among children whose mattresses were in the first quartile (adjusted odds ratio, 0.57 [95% confidence interval, 0.35-0.93] and adjusted odds ratio, 0.49 [95% confidence interval, 0.29-0.83], respectively). Moreover, of the children with eczema at age 3 years, those whose mattresses had endotoxin concentrations in the first quartile had significantly worse sleep disturbance caused by itchy rash (>1 time per week) than did those whose mattresses were in the third and fourth quartiles (20.0% vs 3.3% and 3.7%, both p values < 0.01). The findings indicate that low endotoxin exposure is associated with a higher prevalence of persistent eczema during early childhood.
Collapse
Affiliation(s)
- Makoto Irahara
- Allergy Center, National Center for Child Health and Development, Tokyo, Japan.,Medical Support Center for the Japan Environment and Children's Study, National Center for Child Health and Development, Tokyo, Japan
| | - Kiwako Yamamoto-Hanada
- Allergy Center, National Center for Child Health and Development, Tokyo, Japan.,Medical Support Center for the Japan Environment and Children's Study, National Center for Child Health and Development, Tokyo, Japan
| | - Miori Sato
- Allergy Center, National Center for Child Health and Development, Tokyo, Japan.,Medical Support Center for the Japan Environment and Children's Study, National Center for Child Health and Development, Tokyo, Japan
| | - Mayako Saito-Abe
- Allergy Center, National Center for Child Health and Development, Tokyo, Japan.,Medical Support Center for the Japan Environment and Children's Study, National Center for Child Health and Development, Tokyo, Japan
| | - Yumiko Miyaji
- Allergy Center, National Center for Child Health and Development, Tokyo, Japan.,Medical Support Center for the Japan Environment and Children's Study, National Center for Child Health and Development, Tokyo, Japan
| | - Limin Yang
- Medical Support Center for the Japan Environment and Children's Study, National Center for Child Health and Development, Tokyo, Japan
| | - Minaho Nishizato
- Medical Support Center for the Japan Environment and Children's Study, National Center for Child Health and Development, Tokyo, Japan
| | - Natsuhiko Kumasaka
- Medical Support Center for the Japan Environment and Children's Study, National Center for Child Health and Development, Tokyo, Japan
| | - Hidetohi Mezawa
- Medical Support Center for the Japan Environment and Children's Study, National Center for Child Health and Development, Tokyo, Japan
| | - Yukihiro Ohya
- Allergy Center, National Center for Child Health and Development, Tokyo, Japan.,Medical Support Center for the Japan Environment and Children's Study, National Center for Child Health and Development, Tokyo, Japan
| | | |
Collapse
|
17
|
Talipova D, Smagulova A, Poddighe D. Toll-like Receptors and Celiac Disease. Int J Mol Sci 2022; 24:265. [PMID: 36613709 PMCID: PMC9820541 DOI: 10.3390/ijms24010265] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/10/2022] [Accepted: 12/13/2022] [Indexed: 12/28/2022] Open
Abstract
Celiac disease (CD) is an immune-mediated disorder triggered by dietary gluten intake in some genetically predisposed individuals; however, the additional non-HLA-related genetic factors implicated in CD immunopathogenesis are not well-defined. The role of the innate immune system in autoimmunity has emerged in the last few years. Genetic polymorphisms of some pattern-recognition receptors, including toll-like receptors (TLRs), have been associated with several autoimmune disorders. In this review, we summarize and discuss the evidence from basic research and clinical studies as regards the potential role of TLRs in CD immunopathogenesis. The evidence supporting the role of TLRs in CD immunopathogenesis is limited, especially in terms of basic research. However, differences in the expression and activation of TLRs between active CD patients from one side, and controls and treated CD patients from the other side, have been described in some clinical studies. Therefore, TLRs may be part of those non-HLA-related genetic factors implicated in CD etiopathogenesis, considering their potential role in the interaction between the host immune system and some environmental factors (including viral infections and gut microbiota), which are included in the list of candidate agents potentially contributing to the determination of CD risk in genetically predisposed individuals exposed to dietary gluten intake. Further basic research and clinical studies focused on TLRs in the context of CD and other gluten-related disorders are needed.
Collapse
Affiliation(s)
- Diana Talipova
- Department of Medicine, Nazarbayev University School of Medicine, Astana 010000, Kazakhstan
| | - Aiganym Smagulova
- Department of Medicine, Nazarbayev University School of Medicine, Astana 010000, Kazakhstan
| | - Dimitri Poddighe
- Department of Medicine, Nazarbayev University School of Medicine, Astana 010000, Kazakhstan
- Clinical Academic Department of Pediatrics, National Research Center for Maternal and Child Health, University Medical Center, Astana 010000, Kazakhstan
| |
Collapse
|
18
|
Madani J, Aghebati-Maleki L, Gharibeh N, Pourakbari R, Yousefi M. Fetus, as an allograft, evades the maternal immunity. Transpl Immunol 2022; 75:101728. [DOI: 10.1016/j.trim.2022.101728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 10/09/2022] [Accepted: 10/09/2022] [Indexed: 11/05/2022]
|
19
|
Pieren DKJ, Boer MC, de Wit J. The adaptive immune system in early life: The shift makes it count. Front Immunol 2022; 13:1031924. [PMID: 36466865 PMCID: PMC9712958 DOI: 10.3389/fimmu.2022.1031924] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/31/2022] [Indexed: 10/13/2023] Open
Abstract
Respiratory infectious diseases encountered early in life may result in life-threatening disease in neonates, which is primarily explained by the relatively naive neonatal immune system. Whereas vaccines are not readily available for all infectious diseases, vaccinations have greatly reduced childhood mortality. However, repeated vaccinations are required to reach protective immunity in infants and not all vaccinations are effective at young age. Moreover, protective adaptive immunity elicited by vaccination wanes more rapidly at young age compared to adulthood. The infant adaptive immune system has previously been considered immature but this paradigm has changed during the past years. Recent evidence shows that the early life adaptive immune system is equipped with a strong innate-like effector function to eliminate acute pathogenic threats. These strong innate-like effector capacities are in turn kept in check by a tolerogenic counterpart of the adaptive system that may have evolved to maintain balance and to reduce collateral damage. In this review, we provide insight into these aspects of the early life's adaptive immune system by addressing recent literature. Moreover, we speculate that this shift from innate-like and tolerogenic adaptive immune features towards formation of immune memory may underlie different efficacy of infant vaccination in these different phases of immune development. Therefore, presence of innate-like and tolerogenic features of the adaptive immune system may be used as a biomarker to improve vaccination strategies against respiratory and other infections in early life.
Collapse
Affiliation(s)
| | | | - Jelle de Wit
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| |
Collapse
|
20
|
Frąk M, Grenda A, Krawczyk P, Milanowski J, Kalinka E. Interactions between Dietary Micronutrients, Composition of the Microbiome and Efficacy of Immunotherapy in Cancer Patients. Cancers (Basel) 2022; 14:5577. [PMID: 36428677 PMCID: PMC9688200 DOI: 10.3390/cancers14225577] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/07/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
The effectiveness of immunotherapy in cancer patients depends on the activity of the host's immune system. The intestinal microbiome is a proven immune system modulator, which plays an important role in the development of many cancers and may affect the effectiveness of anti-cancer therapy. The richness of certain bacteria in the gut microbiome (e.g., Bifidobacterium spp., Akkermanisa muciniphila and Enterococcus hire) improves anti-tumor specific immunity and the response to anti-PD-1 or anti-PD-L1 immunotherapy by activating antigen-presenting cells and cytotoxic T cells within the tumor. Moreover, micronutrients affect directly the activities of the immune system or regulate their function by influencing the composition of the microbiome. Therefore, micronutrients can significantly influence the effectiveness of immunotherapy and the development of immunorelated adverse events. In this review, we describe the relationship between the supply of microelements and the abundance of various bacteria in the intestinal microbiome and the effectiveness of immunotherapy in cancer patients. We also point to the function of the immune system in the case of shifts in the composition of the microbiome and disturbances in the supply of microelements. This may in the future become a therapeutic target supporting the effects of immunotherapy in cancer patients.
Collapse
Affiliation(s)
- Małgorzata Frąk
- Chair and Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Anna Grenda
- Chair and Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Paweł Krawczyk
- Chair and Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Janusz Milanowski
- Chair and Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Ewa Kalinka
- Department of Oncology, Polish Mother’s Memorial Hospital—Research Institute, 93-338 Lodz, Poland
| |
Collapse
|
21
|
Ge J, Liu Y. Heat shock protein-70 is elevated in childhood primary immune thrombocytopenia. Eur J Med Res 2022; 27:244. [DOI: 10.1186/s40001-022-00868-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/25/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
Background
Immune thrombocytopenia (ITP) is an acquired autoimmune disorder characterized by the destruction of the platelets resulting from autoimmune recognition and subsequent attack. Heat shock proteins (HSPs) are directly associated with progression and pathogenesis in some specific autoimmune diseases. The aim of this study was to investigate the serum expression of HSP-70 in ITP children and healthy controls.
Materials and methods
A total of 86 children aged 1–6 years were enrolled in the study. The participants were divided into 20 newly diagnosed ITP (ndITP), 34 chronic ITP (cITP) patients and 32 healthy children. The white blood cells and platelet counts were determined and compared between the groups. HSP-70 serum levels were analyzed by sandwich ELISA. Data analysis was done using SPSS and the data variables assessment was done through histogram, probability plots and Shapiro–Wilk tests to determine normal distribution.
Results
The white blood cell counts were 8.9 (4.2–10.4) for new diagnosis ITP, 7.1(3.9–11.9) for the chronic ITP group and 7.0 (4.3–9.5) for the healthy controls. The platelet counts were significantly increased in the chronic ITP group, 83.5(31.7–297) compared to the ndITP group 27.4 (3.7–63.7), but significantly lower compared to the healthy controls 271(172–462) (P = 0.0009). There were significantly increased HSP-70 serum levels in cITP patients compared to the ndITP and the healthy group. In addition, there was a positive correlation between the serum HSP-70 level and the thrombocyte counts among the ITP children.
Conclusions
HSP-70 has a role in the progression of childhood ITP. Increased HSP-70 level is associated with the severity of childhood primary ITP.
Collapse
|
22
|
Contribution of T- and B-cell intrinsic toll-like receptors to the adaptive immune response in viral infectious diseases. Cell Mol Life Sci 2022; 79:547. [PMID: 36224474 PMCID: PMC9555683 DOI: 10.1007/s00018-022-04582-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/27/2022] [Accepted: 09/30/2022] [Indexed: 11/03/2022]
Abstract
Toll-like receptors (TLRs) comprise a class of highly conserved molecules that recognize pathogen-associated molecular patterns and play a vital role in host defense against multiple viral infectious diseases. Although TLRs are highly expressed on innate immune cells and play indirect roles in regulating antiviral adaptive immune responses, intrinsic expression of TLRs in adaptive immune cells, including T cells and B cells, cannot be ignored. TLRs expressed in CD4 + and CD8 + T cells play roles in enhancing TCR signal-induced T-cell activation, proliferation, function, and survival, serving as costimulatory molecules. Gene knockout of TLR signaling molecules has been shown to diminish antiviral adaptive immune responses and affect viral clearance in multiple viral infectious animal models. These results have highlighted the critical role of TLRs in the long-term immunological control of viral infection. This review summarizes the expression and function of TLR signaling pathways in T and B cells, focusing on the in vitro and vivo mechanisms and effects of intrinsic TLR signaling in regulating T- and B-cell responses during viral infection. The potential clinical use of TLR-based immune regulatory drugs for viral infectious diseases is also explored.
Collapse
|
23
|
Wang L, Jiang W, Wang X, Tong L, Song Y. Regulatory T cells in inflammation and resolution of acute lung injury. THE CLINICAL RESPIRATORY JOURNAL 2022; 16:587-595. [PMID: 35924374 PMCID: PMC9436906 DOI: 10.1111/crj.13527] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 07/03/2022] [Accepted: 07/13/2022] [Indexed: 12/03/2022]
Abstract
Introduction Acute respiratory distress syndrome (ARDS) is characterized by hypoxemia and increased lung permeability and would result in acute respiratory failure and with high mortality. In patients who survive from acute lung injury (ALI)/ARDS, it is an active process of the transition from injury to resolution depending on the coordinated immune system. The roles of regulatory CD4+T cells (Tregs) are now gradually being clarified during inflammation and resolution of ARDS. However, clear conclusions about roles of Tregs in ALI/ARDS are only a few. Objective This review provides an overview of phenotype, differentiation, and suppressive mechanisms of Tregs and focuses on keys of biology of Tregs in alveolar space during the inflammatory response and resolution of ALI/ARDS. Data Source Literature search of Web of Science, PubMed, and EMBASE was made to find relative articles about Tregs in ALI/ARDS. We used the following search terms: Tregs, ALI, ARDS, inflammation, and resolution. Conclusion More and more studies have indicated Tregs involved in the processes of inflammation and resolution of ALI/ARDS. A deep understanding of the roles of Tregs may indicate new treatments for patients of ARDS. Therapies aimed at expansion or adaptive transfer of Tregs could be an effective therapy to ARDS patients.
Collapse
Affiliation(s)
- Linlin Wang
- Department of Pulmonary Medicine, Zhongshan Hospital Fudan University Shanghai China
| | - Weipeng Jiang
- Department of Pulmonary Medicine, Zhongshan Hospital Fudan University Shanghai China
| | - Xiaocen Wang
- Department of Pulmonary Medicine, Zhongshan Hospital Fudan University Shanghai China
| | - Lin Tong
- Department of Pulmonary Medicine, Zhongshan Hospital Fudan University Shanghai China
| | - Yuanlin Song
- Department of Pulmonary Medicine, Zhongshan Hospital Fudan University Shanghai China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital Fudan University Shanghai China
- Shanghai Institute of Infectious Disease and Biosecurity Shanghai China
- Shanghai Respiratory Research Institute Shanghai China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital Fudan University Shanghai China
| |
Collapse
|
24
|
Evavold CL, Kagan JC. Diverse Control Mechanisms of the Interleukin-1 Cytokine Family. Front Cell Dev Biol 2022; 10:910983. [PMID: 35832789 PMCID: PMC9272893 DOI: 10.3389/fcell.2022.910983] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/17/2022] [Indexed: 11/15/2022] Open
Abstract
The majority of interleukin-1 (IL-1) family cytokines lack amino terminal secretion signals or transmembrane domains for secretion along the conventional biosynthetic pathway. Yet, these factors must be translocated from the cytoplasm across the plasma membrane into the extracellular space in order to regulate inflammation. Recent work has identified an array of mechanisms by which IL-1 family cytokines can be released into the extracellular space, with supramolecular organizing centers known as inflammasomes serving as dominant drivers of this process. In this review, we discuss current knowledge of the mechanisms of IL-1 family cytokine synthesis, processing, and release from cells. Using this knowledge, we propose a model whereby host metabolic state dictates the route of IL-1β secretion, with implications for microbial infection and sterile inflammation.
Collapse
Affiliation(s)
- Charles L. Evavold
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
- *Correspondence: Charles L. Evavold, ; Jonathan C. Kagan,
| | - Jonathan C. Kagan
- Division of Gastroenterology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, United States
- *Correspondence: Charles L. Evavold, ; Jonathan C. Kagan,
| |
Collapse
|
25
|
Duan T, Du Y, Xing C, Wang HY, Wang RF. Toll-Like Receptor Signaling and Its Role in Cell-Mediated Immunity. Front Immunol 2022. [PMID: 35309296 DOI: 10.3389/fimmu.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
Innate immunity is the first defense system against invading pathogens. Toll-like receptors (TLRs) are well-defined pattern recognition receptors responsible for pathogen recognition and induction of innate immune responses. Since their discovery, TLRs have revolutionized the field of immunology by filling the gap between the initial recognition of pathogens by innate immune cells and the activation of the adaptive immune response. TLRs critically link innate immunity to adaptive immunity by regulating the activation of antigen-presenting cells and key cytokines. Furthermore, recent studies also have shown that TLR signaling can directly regulate the T cell activation, growth, differentiation, development, and function under diverse physiological conditions. This review provides an overview of TLR signaling pathways and their regulators and discusses how TLR signaling, directly and indirectly, regulates cell-mediated immunity. In addition, we also discuss how TLR signaling is critically important in the host's defense against infectious diseases, autoimmune diseases, and cancer.
Collapse
Affiliation(s)
- Tianhao Duan
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Yang Du
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Changsheng Xing
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Helen Y Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Rong-Fu Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
26
|
Duan T, Du Y, Xing C, Wang HY, Wang RF. Toll-Like Receptor Signaling and Its Role in Cell-Mediated Immunity. Front Immunol 2022; 13:812774. [PMID: 35309296 PMCID: PMC8927970 DOI: 10.3389/fimmu.2022.812774] [Citation(s) in RCA: 392] [Impact Index Per Article: 130.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/08/2022] [Indexed: 12/13/2022] Open
Abstract
Innate immunity is the first defense system against invading pathogens. Toll-like receptors (TLRs) are well-defined pattern recognition receptors responsible for pathogen recognition and induction of innate immune responses. Since their discovery, TLRs have revolutionized the field of immunology by filling the gap between the initial recognition of pathogens by innate immune cells and the activation of the adaptive immune response. TLRs critically link innate immunity to adaptive immunity by regulating the activation of antigen-presenting cells and key cytokines. Furthermore, recent studies also have shown that TLR signaling can directly regulate the T cell activation, growth, differentiation, development, and function under diverse physiological conditions. This review provides an overview of TLR signaling pathways and their regulators and discusses how TLR signaling, directly and indirectly, regulates cell-mediated immunity. In addition, we also discuss how TLR signaling is critically important in the host's defense against infectious diseases, autoimmune diseases, and cancer.
Collapse
Affiliation(s)
- Tianhao Duan
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Yang Du
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Changsheng Xing
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Helen Y. Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Rong-Fu Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
27
|
Haegebaert RM, Kempers M, Ceelen W, Lentacker I, Remaut K. Nanoparticle mediated targeting of toll-like receptors to treat colorectal cancer. Eur J Pharm Biopharm 2022; 172:16-30. [PMID: 35074555 DOI: 10.1016/j.ejpb.2022.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/16/2021] [Accepted: 01/17/2022] [Indexed: 02/07/2023]
|
28
|
Whitehead GS, Thomas SY, Nakano K, Royer DJ, Burke CG, Nakano H, Cook DN. A neutrophil/TGF-β axis limits the pathogenicity of allergen-specific CD4+ T cells. JCI Insight 2022; 7:150251. [PMID: 35191395 PMCID: PMC8876454 DOI: 10.1172/jci.insight.150251] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 01/05/2022] [Indexed: 11/17/2022] Open
Abstract
The intensity and longevity of inflammatory responses to inhaled allergens is determined largely by the balance between effector and regulatory immune responses, but the mechanisms that determine the relative magnitudes of these opposing forces remain poorly understood. We have found that the type of adjuvant used during allergic sensitization has a profound effect on both the nature and longevity of the pulmonary inflammation triggered by subsequent reexposure to that same provoking allergen. TLR ligand adjuvants and house dust extracts primed immune responses characterized by a mixed neutrophilic and eosinophilic inflammation that was suppressed by multiple daily allergen challenges. During TLR ligand–mediated allergic sensitization, mice displayed transient airway neutrophilia, which triggered the release of TGF-β into the airway. This neutrophil-dependent production of TGF-β during sensitization had a delayed, suppressive effect on eosinophilic responses to subsequent allergen challenge. Neutrophil depletion during sensitization did not affect numbers of Foxp3+ Tregs but increased proportions of Gata3+CD4+ T cells, which, upon their transfer to recipient mice, triggered stronger eosinophilic inflammation. Thus, a neutrophil/TGF-β axis acts during TLR-mediated allergic sensitization to fine-tune the phenotype of developing allergen-specific CD4+ T cells and limit their pathogenicity, suggesting a novel immunotherapeutic approach to control eosinophilia in asthma.
Collapse
|
29
|
Induction of Endotoxin Tolerance Delays Acute Rejection in a Hindlimb Transplantation Model in Rats. Plast Reconstr Surg 2022; 149:216e-228e. [PMID: 35077416 DOI: 10.1097/prs.0000000000008794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Acute rejection is seen in 85 percent of composite vascular allogeneic transplants despite long-term immunosuppression. Recently, it was reported that the induction of endotoxin tolerance prolonged heart allograft survival in mice. However, it produced side effects in all the animals secondary to the inflammatory reaction. Galactomannan has shown endotoxin tolerance without this side effect in vitro. The authors hypothesized that galactomannan-induced endotoxin tolerance delays acute rejection in vascular allogeneic transplantation without the side effects produced by lipopolysaccharide. METHODS Twenty-four rat hindlimb transplants were divided into four groups according to the preconditioning received: control, lipopolysaccharide (0.16 ml/kg), galactomannan 72 hours before (galactomannan-72) (8 ml/kg), and galactomannan 24 hours before (galactomannan-24) (8 ml/kg). Median acute rejection time, weight loss, and diarrheal episodes were monitored. Blood samples were collected at 0, 7, 21, 30, 45, and 60 days. Plasma cytokines (i.e., tumor necrosis factor alpha, interferon gamma), peripheral chimerism, and lymphocyte percentages were analyzed. RESULTS Median allograft survival was 40 days (range, 40 to 44 days) in the control group, 68 days (range, 61 to 71 days) in the lipopolysaccharide group, and 70 days (range, 69 to 73 days) in both galactomannan groups (p = 0.001). Weight loss was higher in the lipopolysaccharide group (p < 0.001), as was the 83.3 percent rate of diarrheal episodes (control, 0 percent, p = 0.015; galactomannan-72, 0 percent, p = 0.015; and galactomannan-24, 16.7 percent, p = 0.02). Preconditioned rats had higher peripheral blood chimerism (lipopolysaccharide, 2.30 ± 0.13 percent; galactomannan-72, 2.63 ±1.46 percent; and galactomannan-24, 2.47 ± 0.19 percent) compared to the control group (2.06 ± 0.36 percent) (lipopolysaccharide, p = 0.04; galactomannan-72, p = 0.002; and galactomannan-24, p = 0.002). CONCLUSIONS Induction of endotoxin tolerance delays acute rejection in the rat hindlimb transplantation model. Galactomannan preconditioning has no lipopolysaccharide side effects and was equally effective in delaying acute rejection. CLINICAL RELEVANCE STATEMENT The contributions of this experimental work are very incipient. Although the use of galactomannan in clinical practice requires more studies to assess its safety, there is no doubt that immunomodulation may be one of the responses that solve the problem of long-term immunosuppression.
Collapse
|
30
|
Benoit-Lizon I, Jacquin E, Rivera Vargas T, Richard C, Roussey A, Dal Zuffo L, Martin T, Melis A, Vinokurova D, Shahoei SH, Baeza Garcia A, Pignol C, Giorgiutti S, Carapito R, Boidot R, Végran F, Flavell RA, Ryffel B, Nelson ER, Soulas-Sprauel P, Lawrence T, Apetoh L. CD4 T cell-intrinsic STING signaling controls the differentiation and effector functions of TH1 and TH9 cells. J Immunother Cancer 2022; 10:jitc-2021-003459. [PMID: 35091453 PMCID: PMC8804688 DOI: 10.1136/jitc-2021-003459] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2021] [Indexed: 12/20/2022] Open
Abstract
Background While stimulator of interferon genes (STING) activation in innate immune cells of the tumor microenvironment can result in CD8 T cell-dependent antitumor immunity, whether STING signaling affects CD4 T-cell responses remains elusive. Methods Here, we tested whether STING activation modulated the effector functions of CD4 T cells in vivo by analyzing tumor-infiltrating CD4 T cells and evaluating the contribution of the CD4 T cell-derived cytokines in the antitumor activity of the STING ligand 2′3′-cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) in two mouse tumor models. We performed ex vivo experiments to assess the impact of STING activation on CD4 T-cell differentiation and investigate the underlying molecular mechanisms. Finally, we tested whether STING activation enhances TH9 cell antitumor activity against mouse melanoma upon adoptive transfer. Results We found that activation of STING signaling cell-intrinsically enhances the differentiation and antitumor functions of TH1 and TH9 cells by increasing their respective production of interferon gamma (IFN-γ) and interleukin-9. IRF3 and type I interferon receptors (IFNARs) are required for the STING-driven enhancement of TH1 cell differentiation. However, STING activation favors TH9 cell differentiation independently of the IFNARs/IRF3 pathway but through mammalian target of rapamycin (mTOR) signaling, underscoring that STING activation differentially affects the fate of distinct CD4 T-cell subsets. The therapeutic effect of STING activation relies on TH1 and TH9-derived cytokines, and STING activation enhances the antitumor activity of TH9 cells upon adoptive transfer. Conclusion Our results reveal the STING signaling pathway as a therapeutic target to boost CD4 T-cell effector functions and antitumor immunity.
Collapse
Affiliation(s)
- Isis Benoit-Lizon
- INSERM, U1231, Dijon, France
- UFR Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
| | - Elise Jacquin
- INSERM, U1231, Dijon, France
- UFR Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
- INSERM, UMR-S 1193, Université Paris-Saclay, Châtenay-Malabry, France
| | - Thaiz Rivera Vargas
- INSERM, U1231, Dijon, France
- UFR Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
| | - Corentin Richard
- INSERM, U1231, Dijon, France
- UFR Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
| | - Aurélie Roussey
- INSERM, U1231, Dijon, France
- UFR Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
| | - Ludivine Dal Zuffo
- INSERM, U1231, Dijon, France
- UFR Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
| | - Tiffany Martin
- INSERM, U1231, Dijon, France
- UFR Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
| | - Andréa Melis
- INSERM, U1231, Dijon, France
- UFR Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
| | - Daria Vinokurova
- INSERM, U1231, Dijon, France
- UFR Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
| | - Sayyed Hamed Shahoei
- INSERM, U1231, Dijon, France
- UFR Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana Champaign, Urbana, IL, USA
| | - Alvaro Baeza Garcia
- INSERM, U1231, Dijon, France
- UFR Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
| | - Cassandre Pignol
- INSERM, U1231, Dijon, France
- UFR Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
| | - Stéphane Giorgiutti
- INSERM UMR - S1109, Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (CNR RESO), Tertiary Center for Primary Immunodeficiency, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Raphaël Carapito
- Laboratoire d'ImmunoRhumatologie Moléculaire, GENOMAX platform, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), LabEx TRANSPLANTEX, Strasbourg, France
| | - Romain Boidot
- INSERM, U1231, Dijon, France
- UFR Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
- Department of Biology and Pathology of Tumors, Centre Georges François Leclerc, Dijon, France
| | - Frédérique Végran
- INSERM, U1231, Dijon, France
- UFR Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
- Department of Biology and Pathology of Tumors, Centre Georges François Leclerc, Dijon, France
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Heaven, CT, USA
| | - Bernhard Ryffel
- UMR 7355, Experimental and Molecular Immunology and Neurogenetics, CNRS, Orléans, France
- Department of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Eric R Nelson
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana Champaign, Urbana, IL, USA
- Anticancer Discovery from Pets to People Theme, University of Illinois Urbana-Champaign, Cancer Center at Illinois, Urbana Champaign, Illinois, USA
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Pauline Soulas-Sprauel
- INSERM UMR-S1109, Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (CNR RESO), Tertiary Center for Primary Immunodeficiency, Faculty of Pharmacy, Université de Strasbourg, Strasbourg, France
| | - Toby Lawrence
- Centre d'Immunologie de Marseille-Luminy, Université Aix-Marseille, INSERM, CNRS, Marseille, France
| | - Lionel Apetoh
- INSERM, U1231, Dijon, France
- UFR Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
- INSERM, U1100, Tours, France
- Faculté de Médecine, Université de Tours, Tours, France
| |
Collapse
|
31
|
Analysis of N 15-rat growth hormone after incubation with rat subcutaneous tissue and immune cells using ultra-pressure chromatography-mass spectrometry. Anal Biochem 2021; 634:114425. [PMID: 34678250 DOI: 10.1016/j.ab.2021.114425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/22/2021] [Accepted: 10/14/2021] [Indexed: 11/21/2022]
Abstract
Therapeutic proteins (TPs) are exposed to various immune cells like macrophages and neutrophils, especially after subcutaneous (SC) administration. It is well known that the immune cells can generate reactive oxygen species (ROS) and this may lead to oxidation of TPs. The oxidation can occur in the SC tissue after SC administration, during distribution to the immune organs like lymph nodes and spleen, and even in the blood circulation. The oxidation can lead to alteration of their pharmacokinetics and efficacy. Therefore, it is important to study the oxidation of TPs in the biological matrices using ultra-pressure chromatography-mass spectrometry. Rat growth hormone (rGH) was selected as a test protein due to its similarity with human growth hormone (hGH), which is widely used for treatment of growth hormone deficiency. In this manuscript, we have summarized sample processing strategy and ultra-pressure chromatography-mass spectrometry methodology to identify rGH and its degradation products after ex-vivo incubation with rat SC tissue, and in vitro incubation with rat splenocytes and canine peripheral blood mononuclear cells (cPBMCs) as a model foreign host species. We did not observe oxidation of rGH in these biological matrices. This could be due to very minor yields of oxidation products, lack of sensitivity of the mass spectrometry method, loss of protein during sample processing, rapid turnover of oxidized protein or a combination of all factors.
Collapse
|
32
|
Vacharanukrauh P, Meephansan J, Tangtanatakul P, Soonthornchai W, Wongpiyabovorn J, Serirat O, Komine M. High-Throughput RNA Sequencing Reveals the Effect of NB-UVB Phototherapy on Major Inflammatory Molecules of Lesional Psoriasis. PSORIASIS (AUCKLAND, N.Z.) 2021; 11:133-149. [PMID: 34858799 PMCID: PMC8631988 DOI: 10.2147/ptt.s335913] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/28/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To identify the narrowband ultraviolet B (NB-UVB)-induced molecular mechanisms that may account for their anti-inflammatory efficacy, gene expression and transcriptome profiling, which were performed using advanced molecular techniques. METHODS This research was conducted on patients with moderate-to-severe plaque-type psoriasis who received NB-UVB treatment. RNA sequencing (RNA-Seq) was conducted to assay the transcriptomes and identify the differentially expressed transcripts that had been enriched during the major pathway analysis. RESULTS Clinical improvement of psoriasis by NB-UVB therapy is linked to the suppression of the "immunological signaling pathways" and "cell cycle regulatory, growth and proliferation pathways" which are critical to the pathogenesis of the disease. In addition, these results were further substantiated by demonstrating that NB-UVB therapy has a significant effect on keratinocyte differentiation and affects the regulation of genes and inflammatory mediators that are related to cell proliferation and apoptosis. Moreover, NB-UVB phototherapy is also involved with the downregulation of toll-like receptors signaling in lesional psoriasis. CONCLUSION NB-UVB is an effective treatment for psoriasis. Our study supports the conclusion that the clinical effectiveness of NB-UVB therapy is based on the suppression of a broad range of inflammatory signaling pathways, gene expression of inflammatory cytokines and increased expressions of anti-inflammatory signaling pathways in psoriatic skin. This is the first study that applied advanced molecular techniques to investigate phototherapy as a new key to unlock genetic knowledge and create novel information. Ultimately, the goal is to increase medical knowledge and improve the patient care of psoriasis.
Collapse
Affiliation(s)
- Pinyadapat Vacharanukrauh
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, 12120, Thailand
| | - Jitlada Meephansan
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, 12120, Thailand
| | - Pattarin Tangtanatakul
- Department of Transfusion Medicine and Clinical Microbiology, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | | | - Jongkonnee Wongpiyabovorn
- Division of Immunology, Department of Microbiology, Faculty of Medicine, Center of Excellence in Immunology and Immune Mediated Diseases, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Onsiri Serirat
- Division of Dermatology, Department of Medicine, Rajavithi Hospital, Ministry of Public Health, Bangkok, 10400, Thailand
| | - Mayumi Komine
- Department of Dermatology, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
33
|
Nouri Y, Weinkove R, Perret R. T-cell intrinsic Toll-like receptor signaling: implications for cancer immunotherapy and CAR T-cells. J Immunother Cancer 2021; 9:jitc-2021-003065. [PMID: 34799397 PMCID: PMC8606765 DOI: 10.1136/jitc-2021-003065] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2021] [Indexed: 02/06/2023] Open
Abstract
Toll-like receptors (TLRs) are evolutionarily conserved molecules that specifically recognize common microbial patterns, and have a critical role in innate and adaptive immunity. Although TLRs are highly expressed by innate immune cells, particularly antigen-presenting cells, the very first report of a human TLR also described its expression and function within T-cells. Gene knock-out models and adoptive cell transfer studies have since confirmed that TLRs function as important costimulatory and regulatory molecules within T-cells themselves. By acting directly on T-cells, TLR agonists can enhance cytokine production by activated T-cells, increase T-cell sensitivity to T-cell receptor stimulation, promote long-lived T-cell memory, and reduce the suppressive activity of regulatory T-cells. Direct stimulation of T-cell intrinsic TLRs may be a relevant mechanism of action of TLR ligands currently under clinical investigation as cancer immunotherapies. Finally, chimeric antigen receptor (CAR) T-cells afford a new opportunity to specifically exploit T-cell intrinsic TLR function. This can be achieved by expressing TLR signaling domains, or domains from their signaling partner myeloid differentiation primary response 88 (MyD88), within or alongside the CAR. This review summarizes the expression and function of TLRs within T-cells, and explores the relevance of T-cell intrinsic TLR expression to the benefits and risks of TLR-stimulating cancer immunotherapies, including CAR T-cells.
Collapse
Affiliation(s)
- Yasmin Nouri
- Cancer Immunotherapy Programme, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Robert Weinkove
- Cancer Immunotherapy Programme, Malaghan Institute of Medical Research, Wellington, New Zealand.,Department of Pathology & Molecular Medicine, University of Otago, Wellington, Wellington, New Zealand.,Wellington Blood & Cancer Centre, Capital and Coast District Health Board, Wellington, New Zealand
| | - Rachel Perret
- Cancer Immunotherapy Programme, Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
34
|
Puck A, Künig S, Modak M, May L, Fritz P, Battin C, Radakovics K, Steinberger P, Reipert BM, Crowe BA, Stöckl J. The soluble cytoplasmic tail of CD45 regulates T-cell activation via TLR4 signaling. Eur J Immunol 2021; 51:3176-3185. [PMID: 34626426 DOI: 10.1002/eji.202149227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 08/03/2021] [Accepted: 10/06/2021] [Indexed: 11/10/2022]
Abstract
The soluble cytoplasmic tail of CD45 (ct-CD45) is a cleavage fragment of CD45, that is generated during the activation of human phagocytes. Upon release to the extracellular space, ct-CD45 binds to human T cells and inhibits their activation in vitro. Here, we studied the potential role of TLR4 as a receptor for ct-CD45. Treatment of Jurkat TLR4/CD14 reporter cells with ct-CD45 induced the upregulation of the reporter gene NFκB-eGFP and could be blocked by inhibitors of TLR4 signaling. Conversely, ct-CD45 did not promote the NFκB-controlled eGFP induction in reporter cells expressing TLR1, TLR2, and TLR6 transgenes and did not lead to the activation of the transcription factors NFκB, AP-1, and NFAT in a Jurkat reporter cell line expressing endogenous TLR5. Moreover, ct-CD45 binds to recombinant TLR4 in an in vitro assay and this association was reduced in the presence of oxidized 1-palmitoyl-2-arachidonyl-sn-glycero-3-phosphorylcholine. Blockade of TLR4 with mAb HTA125 partially reversed the ct-CD45-mediated inhibition of T-cell proliferation. Interestingly, targeting of TLR4 with mAb W7C11 also suppressed T-cell proliferation. In summary, the results of this study demonstrate that ct-CD45 acts via a noncanonical TLR4 activation pathway on T cells, which modulates TCR signaling.
Collapse
Affiliation(s)
- Alexander Puck
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Sarojinidevi Künig
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Madhura Modak
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Lara May
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Pia Fritz
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Claire Battin
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Katharina Radakovics
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Steinberger
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Birgit M Reipert
- Department of Immunology, Drug Discovery Austria, Baxalta Innovations GmbH, Vienna, Austria
| | - Brian A Crowe
- Department of Immunology, Drug Discovery Austria, Baxalta Innovations GmbH, Vienna, Austria
| | - Johannes Stöckl
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
35
|
Abstract
The immune (innate and adaptive) system has evolved to protect the host from any danger present in the surrounding outer environment (microbes and associated MAMPs or PAMPs, xenobiotics, and allergens) and dangers originated within the host called danger or damage-associated molecular patterns (DAMPs) and recognizing and clearing the cells dying due to apoptosis. It also helps to lower the tissue damage during trauma and initiates the healing process. The pattern recognition receptors (PRRs) play a crucial role in recognizing different PAMPs or MAMPs and DAMPs to initiate the pro-inflammatory immune response to clear them. Toll-like receptors (TLRs) are first recognized PRRs and their discovery proved milestone in the field of immunology as it filled the gap between the first recognition of the pathogen by the immune system and the initiation of the appropriate immune response required to clear the infection by innate immune cells (macrophages, neutrophils, dendritic cells or DCs, and mast cells). However, in addition to their expression by innate immune cells and controlling their function, TLRs are also expressed by adaptive immune cells. We have identified 10 TLRs (TLR1-TLR10) in humans and 12 TLRs (TLR1-TLR13) in laboratory mice till date as TLR10 in mice is present only as a defective pseudogene. The present chapter starts with the introduction of innate immunity, timing of TLR evolution, and the evolution of adaptive immune system and its receptors (T cell receptors or TCRs and B cell receptors or BCRs). The next section describes the role of TLRs in the innate immune function and signaling involved in the generation of inflammation. The subsequent sections describe the expression and function of different TLRs in murine and human adaptive immune cells (B cells and different types of T cells, including CD4+T cells, CD8+T cells, CD4+CD25+Tregs, and CD8+CD25+Tregs, etc.). The modulation of TLRs expressed on T and B cells has a great potential to develop different vaccine candidates, adjuvants, immunotherapies to target various microbial infections, including current COVID-19 pandemic, cancers, and autoimmune and autoinflammatory diseases.
Collapse
Affiliation(s)
- Vijay Kumar
- Children's Health Queensland Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, Brisbane, QLD, Australia.
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center (UTHSC), Memphis, TN, USA.
| |
Collapse
|
36
|
Kayama H, Okumura R, Takeda K. Interaction Between the Microbiota, Epithelia, and Immune Cells in the Intestine. Annu Rev Immunol 2021; 38:23-48. [PMID: 32340570 DOI: 10.1146/annurev-immunol-070119-115104] [Citation(s) in RCA: 416] [Impact Index Per Article: 104.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The gastrointestinal tract harbors numerous commensal bacteria, referred to as the microbiota, that benefit host health by digesting dietary components and eliminating pathogens. The intestinal microbiota maintains epithelial barrier integrity and shapes the mucosal immune system, balancing host defense and oral tolerance with microbial metabolites, components, and attachment to host cells. To avoid aberrant immune responses, epithelial cells segregate the intestinal microbiota from immune cells by constructing chemical and physical barriers, leading to the establishment of host-commensal mutualism. Furthermore, intestinal immune cells participate in the maintenance of a healthy microbiota community and reinforce epithelial barrier functions. Perturbations of the microbiota composition are commonly observed in patients with autoimmune diseases and chronic inflammatory disorders. An understanding of the intimate interactions between the intestinal microbiota, epithelial cells, and immune cells that are crucial for the maintenance of intestinal homeostasis might promote advances in diagnostic and therapeutic approaches for various diseases.
Collapse
Affiliation(s)
- Hisako Kayama
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; , , .,WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan.,Institute for Advanced Co-Creation Studies, Osaka University, Suita, Osaka 565-0871, Japan
| | - Ryu Okumura
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; , , .,WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kiyoshi Takeda
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; , , .,WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
37
|
Pseudomonas aeruginosa outer membrane vesicles ameliorates lung ischemia-reperfusion injury by regulating the balance of regulatory T cells and Th17 cells through Tim-3 and TLR4/NF-κB pathway. Inflamm Res 2021; 70:891-902. [PMID: 34223915 DOI: 10.1007/s00011-021-01483-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 06/15/2021] [Accepted: 06/21/2021] [Indexed: 10/20/2022] Open
Abstract
OBJECTIVE Regulatory T cells (Tregs) and T helper (Th) 17 cells are two subsets of CD4 + T cells with opposite effects which play a crucial role in the pathogenesis of lung injury. In this study, we aim to investigate the protective effect of Pseudomonas aeruginosa outer membrane vesicles (OMVs) preconditioning on lung ischemia-reperfusion (I/R) injury and potential mechanisms. METHODS Pathogen-free C57BL/6 mice were randomly divided into four groups: control, Control + OMVs, I/R and I/R + OMVs groups. Bronchoalveolar lavage fluid (BALF), serum, and lung tissues were collected and analyzed for pathophysiology and immune mechanism. RESULTS OMVs not only attenuated tissue injury and respiratory physiologic function but also mediated the downregulation of lung wet-to-dry weight ratio and the reduction of total protein concentration. The numbers of total cells, macrophages, neutrophils, and lymphocytes were markedly decreased in the I/R mice following OMVs preconditioning. OMVs also decreased inflammatory cytokines associated with CD4 + T cells in both BALF and serum. In addition, the level of Tregs and its transcription factor forkhead box P3 (Foxp3) were significantly increased, while the level of Th17 cells and its transcription factor retinoid-related orphan receptor γ (RORγt) were significantly decreased following OMVs preconditioning. In the process of exploring the underlying protection mechanisms of OMVs, we found that OMVs preconditioning significantly reduced protein expression of Toll-like receptor 4 (TLR4), which in turn not only inactivated myeloid differentiation factor 88 (MyD88) and Phosphorylated nuclear factor kappa B (p-NF-κB), but also simultaneously increased the levels of T-cell immunoglobulin and mucin domain-containing protein 3 (Tim-3). CONCLUSIONS These results suggest that OMVs preconditioning may ameliorate lung I/R injury by regulating the balance of Tregs and Th17 cells through Tim-3 and TLR4/NF-κB pathway.
Collapse
|
38
|
Quirino MG, Macedo LC, Pagnano KBB, Pagliarini-E-Silva S, Sell AM, Visentainer JEL. Toll-like receptor gene polymorphisms in patients with myeloproliferative neoplasms. Mol Biol Rep 2021; 48:4995-5001. [PMID: 34191235 DOI: 10.1007/s11033-021-06238-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 02/16/2021] [Indexed: 12/26/2022]
Abstract
Toll-like receptors (TLRs) are a family of transmembrane receptors whose signaling control cellular processes of cell proliferation, survival, apoptosis, angiogenesis, remodeling, and repair of tissues. Polymorphisms in TLR genes can change the balance between pro and anti-inflammatory cytokines, modulating the risk of infection, chronic inflammation, and cancer. Although many studies have demonstrated the direct involvement of TLR signaling in the benefit of tumor cells in certain cancers, little is known about the influence of these gene polymorphisms on myeloproliferative neoplasms (MPNs). In this context, the objective of the study was to investigate a possible association between the TLR polymorphisms and the development of MPNs. 167 patients diagnosed with MPN and 222 healthy controls from the same region were evaluated. Genomic DNA was extracted and the TLR2 (rs5743708), TLR4 (rs4986790, rs4986791), TLR9 (rs5743836, rs187084) and JAK2V617F polymorphisms were genotyped by PCR-RFLP. The statistical analysis was performed by OpenEpi and SNPstat software. The JAK2V617F mutation was found in 68.32% of patients. TLR9-1486C/T CT genotype was less frequent in patients with polycythemia vera (PV) (OR 0.39, 95% CI 0.20-0.78, P = 0.025). When haplotype frequencies were analyzed, -1237T/-1486C (TLR9) was also less frequent in men (OR 0.58, 95% CI 0.36-0.94) and JAK negative men patients (OR 0.43, 95% CI 0.21-0.88). We can infer that the TLR9-1486 CT genotype could be associated with protection for PV and the TLR9-1237T/-1486C haplotype, protection for men, as well as for JAK negative men patients with MPN. There were no associations between TLR2 and TLR4 gene polymorphisms and MPN.
Collapse
Affiliation(s)
- Marília Gonçalves Quirino
- Graduate Program in Biosciences and Physiopathology of the State University of Maringá, Av. Colombo 5790, bloco T20, sala 109, Maringá, PR, CEP: 87020-900, Brazil
| | - Luciana Conci Macedo
- Graduate Program in Biosciences and Physiopathology of the State University of Maringá, Av. Colombo 5790, bloco T20, sala 109, Maringá, PR, CEP: 87020-900, Brazil
| | | | | | - Ana Maria Sell
- Graduate Program in Biosciences and Physiopathology of the State University of Maringá, Av. Colombo 5790, bloco T20, sala 109, Maringá, PR, CEP: 87020-900, Brazil
| | - Jeane Eliete Laguila Visentainer
- Graduate Program in Biosciences and Physiopathology of the State University of Maringá, Av. Colombo 5790, bloco T20, sala 109, Maringá, PR, CEP: 87020-900, Brazil.
| |
Collapse
|
39
|
Takahashi R, Shiohara T, Mizukawa Y. Monocyte-Independent and -Dependent Regulation of Regulatory T-Cell Development in Mycoplasma Infection. J Infect Dis 2021; 223:1733-1742. [PMID: 32946556 DOI: 10.1093/infdis/jiaa590] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 09/16/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Although Mycoplasma pneumoniae (MP) infection has been implicated in the pathogenesis of allergic diseases, the mechanism of this trigger remains unknown. We explored the mechanism for how MP infection could tilt the balance between regulatory T cells (Tregs) and Th17 cells. METHODS We analyzed the frequency, phenotype, and function of Tregs in patients at the different stages of MP and various virus infections over a period of more than 1 year. We examined the effect of monocytes to elucidate signals that can regulate the balance between Treg and Th17 cells. RESULTS The functional activity of Tregs was profoundly impaired during the acute stage of MP as well as viral infections. Upon resolution, however, the Treg function remained impaired even 1 year after MP infection. In the resolution stage, the impaired Treg function was associated with an increase in interleukin (IL) 17A+ Tregs and Th17 cells. Development of Th17 cells was dependent on the "aberrant" proinflammatory monocytes (pMOs), characterized by potent ability to produce IL-6 in a Toll-like receptor 2-dependent manner. CONCLUSIONS Depending on the prevalence of the pMOs, Tregs and Th17 cells could mutually regulate the number and function of the other. The pMOs/IL-6 could be crucial therapeutic targets against MP-induced allergic diseases.
Collapse
Affiliation(s)
- Ryo Takahashi
- Flow Cytometry Core Facility, Kyorin University Graduate School of Medicine, Mitaka, Tokyo, Japan
| | - Tetsuo Shiohara
- Flow Cytometry Core Facility, Kyorin University Graduate School of Medicine, Mitaka, Tokyo, Japan.,Department of Dermatology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Yoshiko Mizukawa
- Department of Dermatology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| |
Collapse
|
40
|
Aksel EG, Akyüz B. Effect of LPS and LTA stimulation on the expression of TLR-pathway genes in PBMCs of Akkaraman lambs in vivo. Trop Anim Health Prod 2021; 53:65. [PMID: 33392825 PMCID: PMC7779097 DOI: 10.1007/s11250-020-02491-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 12/02/2020] [Indexed: 01/30/2023]
Abstract
This is the first study investigating the changes in some gene expressions related to the TLR pathway in vivo in sheep. Lipopolysaccharide (LPS) and lipoteichoic acid (LTA) molecules were administrated separately and in combination to the Akkaraman lambs via intranasal route. For this purpose, 28 lambs were distributed into four groups (LPS, LTA, LPS + LTA, and control, n = 7). Blood samples were collected to isolate the peripheral blood mononuclear cells (PBMCs) at 24 h and on day 7. Expression levels of TLR2, TLR4, MyD88, TRAF6, TNF-α, IL-1ß, IL-6, IL-10, NF-κß, and IFN-γ genes were determined by qRT-PCR. Increases were determined in the expression data of TLR2 [LPS (P < 0.05) and LTA + LPS (P < 0.01)], TLR4 [LTA + LPS (P < 0.05)], TNF-α, IL-10 [LTA + LPS (P < 0.05)], and IFN-γ genes in all groups in the mRNA expression analysis of PBMCs isolated at 24 h whereas decreases were determined in the expression levels of these genes on day 7. The combination of LPS + LTA stimulated lamb PBMCs more effectively than separate administration of LPS and LTA at 24 h. Therefore, this article may contribute to the understanding the host-pathogen interaction of respiratory-transmitted bacterial diseases concerning PBMCs at 24 h and on day 7. Also this study may contribute to the dose adjustment for bacterial vaccine studies in sheep. Experimental application doses will be helpful for in vivo and in vitro drug and vaccine development studies in the fields of pharmacology and microbiology.
Collapse
Affiliation(s)
- Esma Gamze Aksel
- Department of Genetic, Faculty of Veterinary Medicine, Erciyes University, 38039, Kayseri, Turkey.
| | - Bilal Akyüz
- Department of Genetic, Faculty of Veterinary Medicine, Erciyes University, 38039, Kayseri, Turkey
| |
Collapse
|
41
|
Chodaczek G, Pagni PP, Christoffersson G, Ratliff SS, Toporkiewicz M, Wegrzyn AS, von Herrath M. The effect of Toll-like receptor stimulation on the motility of regulatory T cells. J Autoimmun 2021; 116:102563. [PMID: 33189487 DOI: 10.1016/j.jaut.2020.102563] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 10/23/2022]
Abstract
Regulatory T cells (Tregs) have suppressive functions and play an important role in controlling inflammation and autoimmunity. The migratory capacity of Tregs determines their location and their location determines whether they inhibit the priming of naïve lymphocytes in lymphoid tissues or the effector phase of immune responses at inflamed sites. Tregs generated or expanded in vitro are currently being tested in clinics for the treatment of autoimmune disorders, however, little is known about the factors controlling their migration towards therapeutically relevant locations. In this study, we have modulated Treg dynamics using Toll-like receptor (TLR) agonists. Dynamic imaging with confocal and two-photon microscopy revealed that Tregs generated in vitro and stimulated with P3C (a TLR2 agonist) but not with R848 (a TLR7 agonist) or LPS (a TLR4 agonist) showed enhanced cell migration within splenic white pulp or draining lymph node when transferred into mice intravenously or into the footpad, respectively. In summary, our data demonstrate that Tregs are more motile in response to direct TLR stimulation in particular towards TLR2 signals. This may have implications for efficient clinical Treg induction protocols.
Collapse
Affiliation(s)
- Grzegorz Chodaczek
- Type 1 Diabetes Center, La Jolla Institute for Immunology, La Jolla, CA, USA; Bioimaging Laboratory, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland.
| | - Philippe P Pagni
- Type 1 Diabetes Center, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Gustaf Christoffersson
- Type 1 Diabetes Center, La Jolla Institute for Immunology, La Jolla, CA, USA; Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Monika Toporkiewicz
- Bioimaging Laboratory, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Agnieszka S Wegrzyn
- Bioimaging Laboratory, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | | |
Collapse
|
42
|
Nakao M, Sugaya M, Fujita H, Miyagaki T, Morimura S, Shibata S, Asano Y, Sato S. TLR2 Deficiency Exacerbates Imiquimod-Induced Psoriasis-Like Skin Inflammation through Decrease in Regulatory T Cells and Impaired IL-10 Production. Int J Mol Sci 2020; 21:E8560. [PMID: 33202847 PMCID: PMC7696365 DOI: 10.3390/ijms21228560] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/07/2020] [Accepted: 11/10/2020] [Indexed: 12/30/2022] Open
Abstract
Emerging evidence has demonstrated that Toll-like receptors (TLRs) are associated with autoimmune diseases. In this study, we investigated the role of TLR2 in psoriasis using imiquimod-induced psoriasis-like dermatitis. Although TLR2 signaling is known to play a critical role in the induction of proinflammatory cytokines by immune cells, such as dendritic cells (DCs), macrophages, and monocytes, TLR2 deficiency unexpectedly exacerbated psoriasiform skin inflammation. Importantly, messenger RNA (mRNA) levels of Foxp-3 and IL-10 in the lesional skin were significantly decreased in TLR2 KO mice compared with wild-type mice. Furthermore, flow cytometric analysis of the lymph nodes revealed that the frequency of regulatory T cells (Tregs) among CD4-positive cells was decreased. Notably, stimulation with Pam3CSK4 (TLR2/1 ligand) or Pam2CSK4 (TLR2/6 ligand) increased IL-10 production from Tregs and DCs and the proliferation of Tregs. Finally, adoptive transfer of Tregs from wild-type mice reduced imiquimod-induced skin inflammation in TLR2 KO mice. Taken together, our results suggest that TLR2 signaling directly enhances Treg proliferation and IL-10 production by Tregs and DCs, suppressing imiquimod-induced psoriasis-like skin inflammation. Enhancement of TLR2 signaling may be a new therapeutic strategy for psoriasis.
Collapse
Affiliation(s)
- Momoko Nakao
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan; (M.N.); (H.F.); (T.M.); (S.M.); (S.S.); (Y.A.); (S.S.)
| | - Makoto Sugaya
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan; (M.N.); (H.F.); (T.M.); (S.M.); (S.S.); (Y.A.); (S.S.)
- Department of Dermatology, International University of Health and Welfare, Chiba 286-8520, Japan
| | - Hideki Fujita
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan; (M.N.); (H.F.); (T.M.); (S.M.); (S.S.); (Y.A.); (S.S.)
- Division of Cutaneous Science, Department of Dermatology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Tomomitsu Miyagaki
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan; (M.N.); (H.F.); (T.M.); (S.M.); (S.S.); (Y.A.); (S.S.)
- Department of Dermatology, St. Marianna University School of Medicine, Kanagawa 216-8511, Japan
| | - Sohshi Morimura
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan; (M.N.); (H.F.); (T.M.); (S.M.); (S.S.); (Y.A.); (S.S.)
- Department of Dermatology, International University of Health and Welfare, Chiba 286-8520, Japan
| | - Sayaka Shibata
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan; (M.N.); (H.F.); (T.M.); (S.M.); (S.S.); (Y.A.); (S.S.)
| | - Yoshihide Asano
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan; (M.N.); (H.F.); (T.M.); (S.M.); (S.S.); (Y.A.); (S.S.)
| | - Shinichi Sato
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan; (M.N.); (H.F.); (T.M.); (S.M.); (S.S.); (Y.A.); (S.S.)
| |
Collapse
|
43
|
Ge Y, Huang M, Dong N, Yao YM. Effect of Interleukin-36β on Activating Autophagy of CD4+CD25+ Regulatory T cells and Its Immune Regulation in Sepsis. J Infect Dis 2020; 222:1517-1530. [PMID: 32421784 DOI: 10.1093/infdis/jiaa258] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/11/2020] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND CD4+CD25+ regulatory T cells (Tregs) play an essential role in sepsis-induced immunosuppression. How, the effects of interleukin 36 (IL-36) cytokines on CD4+CD25+ Tregs and their underlying mechanism(s) in sepsis remain unknown. METHODS Our study was designed to investigate the impacts of IL-36 cytokines on murine CD4+CD25+ Tregs in presence of lipopolysaccharide (LPS) and in a mouse model of sepsis induced by cecal ligation and puncture (CLP). IL-36-activated autophagy was evaluated by autophagy markers, autophagosome formation, and autophagic flux. RESULTS IL-36α, IL-36β, and IL-36γ were expressed in murine CD4+CD25+ Tregs. Stimulation of CD4+CD25+ Tregs with LPS markedly up-regulated the expression of these cytokines, particularly IL-36β. IL-36β strongly suppressed CD4+CD25+ Tregs under LPS stimulation and in septic mice challenged with CLP, resulting in the amplification of T-helper 1 response and the proliferation of effector T cells. Mechanistic studies revealed that IL-36β triggered autophagy of CD4+CD25+ Tregs. These effects were significantly attenuated in the presence of the autophagy inhibitor 3-methyladenine or Beclin1 knockdown. In addition, early IL-36β administration reduced the mortality rate in mice subjected to CLP. Depletion of CD4+CD25+ Tregs before the onset of sepsis obviously abrogated IL-36β-mediated protection against sepsis. CONCLUSIONS These findings suggest that IL-36β diminishes the immunosuppressive activity of CD4+CD25+ Tregs by activating the autophagic process, thereby contributing to improvement of the host immune response and prognosis in sepsis.
Collapse
Affiliation(s)
- Yun Ge
- Department of General Intensive Care Unit, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Man Huang
- Department of General Intensive Care Unit, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Ning Dong
- Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Yong-Ming Yao
- Department of General Intensive Care Unit, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing, People's Republic of China.,State Key Laboratory of Kidney Disease, the Chinese PLA General Hospital, Beijing, People's Republic of China
| |
Collapse
|
44
|
Culos A, Tsai AS, Stanley N, Becker M, Ghaemi MS, McIlwain DR, Fallahzadeh R, Tanada A, Nassar H, Espinosa C, Xenochristou M, Ganio E, Peterson L, Han X, Stelzer IA, Ando K, Gaudilliere D, Phongpreecha T, Marić I, Chang AL, Shaw GM, Stevenson DK, Bendall S, Davis KL, Fantl W, Nolan GP, Hastie T, Tibshirani R, Angst MS, Gaudilliere B, Aghaeepour N. Integration of mechanistic immunological knowledge into a machine learning pipeline improves predictions. NAT MACH INTELL 2020; 2:619-628. [PMID: 33294774 PMCID: PMC7720904 DOI: 10.1038/s42256-020-00232-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 08/26/2020] [Indexed: 12/17/2022]
Abstract
The dense network of interconnected cellular signalling responses that are quantifiable in peripheral immune cells provides a wealth of actionable immunological insights. Although high-throughput single-cell profiling techniques, including polychromatic flow and mass cytometry, have matured to a point that enables detailed immune profiling of patients in numerous clinical settings, the limited cohort size and high dimensionality of data increase the possibility of false-positive discoveries and model overfitting. We introduce a generalizable machine learning platform, the immunological Elastic-Net (iEN), which incorporates immunological knowledge directly into the predictive models. Importantly, the algorithm maintains the exploratory nature of the high-dimensional dataset, allowing for the inclusion of immune features with strong predictive capabilities even if not consistent with prior knowledge. In three independent studies our method demonstrates improved predictions for clinically relevant outcomes from mass cytometry data generated from whole blood, as well as a large simulated dataset. The iEN is available under an open-source licence.
Collapse
Affiliation(s)
- Anthony Culos
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Sciences, Stanford University, Stanford, CA, USA
- These authors contributed equally: Anthony Culos, Amy S. Tsai
| | - Amy S Tsai
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- These authors contributed equally: Anthony Culos, Amy S. Tsai
| | - Natalie Stanley
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Sciences, Stanford University, Stanford, CA, USA
| | - Martin Becker
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Sciences, Stanford University, Stanford, CA, USA
| | - Mohammad S Ghaemi
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Sciences, Stanford University, Stanford, CA, USA
- Digital Technologies Research Centre, National Research Council Canada, Toronto, Ontario, Canada
| | - David R McIlwain
- Department of Microbiology and Immunology, Baxter Laboratory in Stem Cell Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ramin Fallahzadeh
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Sciences, Stanford University, Stanford, CA, USA
| | - Athena Tanada
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Sciences, Stanford University, Stanford, CA, USA
| | - Huda Nassar
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Sciences, Stanford University, Stanford, CA, USA
| | - Camilo Espinosa
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Sciences, Stanford University, Stanford, CA, USA
| | - Maria Xenochristou
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Sciences, Stanford University, Stanford, CA, USA
| | - Edward Ganio
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Laura Peterson
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiaoyuan Han
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ina A Stelzer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Kazuo Ando
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Dyani Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Thanaphong Phongpreecha
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Sciences, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ivana Marić
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Alan L Chang
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Sciences, Stanford University, Stanford, CA, USA
| | - Gary M Shaw
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - David K Stevenson
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Sean Bendall
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kara L Davis
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Wendy Fantl
- Department of Microbiology and Immunology, Baxter Laboratory in Stem Cell Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Garry P Nolan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Trevor Hastie
- Department of Biomedical Data Sciences, Stanford University, Stanford, CA, USA
- Department of Statistics, Stanford University, Stanford, CA, USA
| | - Robert Tibshirani
- Department of Biomedical Data Sciences, Stanford University, Stanford, CA, USA
- Department of Statistics, Stanford University, Stanford, CA, USA
| | - Martin S Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- These authors jointly supervised this work: Martin S. Angst, Brice Gaudilliere, Nima Aghaeepour
| | - Brice Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
- These authors jointly supervised this work: Martin S. Angst, Brice Gaudilliere, Nima Aghaeepour
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Sciences, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
- These authors jointly supervised this work: Martin S. Angst, Brice Gaudilliere, Nima Aghaeepour
| |
Collapse
|
45
|
Δ42PD1-TLR4 Augments γδ-T Cell Activation of the Transitional Memory Subset of CD4 + T Cells. iScience 2020; 23:101620. [PMID: 33089108 PMCID: PMC7567942 DOI: 10.1016/j.isci.2020.101620] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/25/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
TLR ligands can contribute to T cell immune responses by indirectly stimulating antigen presentation and cytokines and directly serving as co-stimulatory signals. We have previously reported that the human endogenous surface protein, Δ42PD1, is expressed primarily on (Vγ9)Vδ2 cells and can interact with TLR4. Since Vδ2 cells possess antigen presentation capacity, we sought to further characterize if the Δ42PD1-TLR4 interaction has a role in stimulating T cell responses. In this study, we found that stimulation of Vδ2 cells not only upregulated Δ42PD1 expression but also increased MHC class II molecules necessary for the antigen presentation. In a mixed leukocyte reaction assay, upregulation of Δ42PD1 on Vδ2 cells elevated subsequent T cell proliferation. Furthermore, the interaction between Δ42PD1-TLR4 augments Vδ2 cell stimulation of autologous CMV pp65-or TT-specific CD4+ T cell proliferation and IFN-γ responses, which was specifically and significantly reduced by blocking the Δ42PD1-TLR4 interaction. Furthermore, confocal microscopy analysis confirmed the interaction between Δ42PD1+HLA-DR+Vδ2 cells and TLR4+CD4 T cells. Interestingly, the subset of CD4+ T cells expressing TLR4 appears to be PD-1+ CD45RO+CD45RA+ transitional memory T cells and responded to Δ42PD1+HLA-DR+Vδ2 cells. Overall, this study demonstrated an important biological role of Δ42PD1 protein exhibited by Vδ2 antigen-presenting cells in augmenting T cell activation through TLR4, which may serve as an additional co-stimulatory signal. Δ42PD1 is co-expressed with MHC-II on activated Vδ2 cells Δ42PD1+MHC-II+Vδ2 cells interact directly with TLR4+CD4+T cells in 3D imaging TLR4 is highly expressed on the PD-1+CD45RO+CD45RA+CD4+T cell subset Δ42PD1-TLR4 selectively activates this subset of Ag-specific CD4+ T cells
Collapse
|
46
|
Lee HG, Cho MZ, Choi JM. Bystander CD4 + T cells: crossroads between innate and adaptive immunity. Exp Mol Med 2020; 52:1255-1263. [PMID: 32859954 PMCID: PMC8080565 DOI: 10.1038/s12276-020-00486-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/21/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022] Open
Abstract
T cells are the central mediators of both humoral and cellular adaptive immune responses. Highly specific receptor-mediated clonal selection and expansion of T cells assure antigen-specific immunity. In addition, encounters with cognate antigens generate immunological memory, the capacity for long-term, antigen-specific immunity against previously encountered pathogens. However, T-cell receptor (TCR)-independent activation, termed “bystander activation”, has also been found. Bystander-activated T cells can respond rapidly and secrete effector cytokines even in the absence of antigen stimulation. Recent studies have rehighlighted the importance of antigen-independent bystander activation of CD4+ T cells in infection clearance and autoimmune pathogenesis, suggesting the existence of a distinct innate-like immunological function performed by conventional T cells. In this review, we discuss the inflammatory mediators that activate bystander CD4+ T cells and the potential physiological roles of these cells during infection, autoimmunity, and cancer. Immune cells that become activated in the absence of antigen stimulation could be harnessed in the fight against infection, autoimmunity, and cancer. Je-Min Choi and colleagues from Hanyang University in Seoul, South Korea, review how the immune system can deploy helper T cells through an unusual process called bystander activation. Most T cells become activated only after receptors on their surface bind to specific cognate antigen. In contrast, bystander T cells are activated non-specifically in response to cytokines and other pro-inflammatory mediators. Studies have shown that this cell population has a variety of protective and pathogenic functions, for example, guarding against multiple sclerosis, aggravating the symptoms of parasitic infections and promoting antitumor immunity. A better understanding of these immune cells could lead to new therapeutic options for these diseases.
Collapse
Affiliation(s)
- Hong-Gyun Lee
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Min-Zi Cho
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea. .,Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea. .,Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul, Republic of Korea.
| |
Collapse
|
47
|
Stögerer T, Stäger S. Innate Immune Sensing by Cells of the Adaptive Immune System. Front Immunol 2020; 11:1081. [PMID: 32547564 PMCID: PMC7274159 DOI: 10.3389/fimmu.2020.01081] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 05/05/2020] [Indexed: 01/05/2023] Open
Abstract
Sensing of microbes or of danger signals has mainly been attributed to myeloid innate immune cells. However, T and B cells also express functional pattern recognition receptors (PRRs). In these cells, PRRs mediate signaling cascades that result in different functions depending on the cell's activation and/or differentiation status, on the environment, and on the ligand/agonist. Some of these functions are beneficial for the host; however, some are detrimental and are exploited by pathogens to establish persistent infections. In this review, we summarize the available literature on innate immune sensing by cells of the adaptive immune system and discuss possible implications for chronic infections.
Collapse
Affiliation(s)
- Tanja Stögerer
- INRS Centre Armand-Frappier Santé Biotechnologie, Laval, QC, Canada
| | - Simona Stäger
- INRS Centre Armand-Frappier Santé Biotechnologie, Laval, QC, Canada
| |
Collapse
|
48
|
Hernandes C, Gueuvoghlanian-Silva BY, Monnaka VU, Ribeiro NM, Pereira WDO, Podgaec S. Regulatory T cells isolated from endometriotic peritoneal fluid express a different number of Toll-like receptors. EINSTEIN-SAO PAULO 2020; 18:eAO5294. [PMID: 32236327 PMCID: PMC7087457 DOI: 10.31744/einstein_journal/2020ao5294] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 02/09/2020] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE To analyze and compare the expression of Toll-like receptors by regulatory T cells present in the peritoneal fluid of patients with and without endometriosis. METHODS Regulatory T cells were isolated from peritoneal fluid of women with and without endometriosis, collected during surgery, and mRNA was extracted for analysis of Toll-like receptors expression by reverse-transcriptase polymerase chain reaction. RESULTS Patients with endometriosis presented regulatory T cells expressing a larger number and variety of Toll-like receptors when compared to regulatory T cells from patients in the Control Group. Toll-like receptor-1 and Toll-like receptor-2 in regulatory T cells were expressed in both groups. All other expressed Toll-like receptors types were only found in regulatory T cells from the Endometriosis Group. CONCLUSION Patients with endometriosis had peritoneal regulatory T cells expressing various Toll-like receptors types.
Collapse
Affiliation(s)
- Camila Hernandes
- Hospital Israelita Albert EinsteinSão PauloSPBrazilHospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Bárbara Yasmin Gueuvoghlanian-Silva
- Instituto Israelita de Ensino e Pesquisa Albert EinsteinHospital Israelita Albert EinsteinSão PauloSPBrazilInstituto Israelita de Ensino e Pesquisa Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Vitor Ulisses Monnaka
- Faculdade Israelita de Ciências da Saúde Albert EinsteinHospital Israelita Albert EinsteinSão PauloSPBrazilFaculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Natalia Mazini Ribeiro
- Instituto Israelita de Ensino e Pesquisa Albert EinsteinHospital Israelita Albert EinsteinSão PauloSPBrazilInstituto Israelita de Ensino e Pesquisa Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Welbert de Oliveira Pereira
- Hospital Israelita Albert EinsteinSão PauloSPBrazilHospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Sérgio Podgaec
- Hospital Israelita Albert EinsteinSão PauloSPBrazilHospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| |
Collapse
|
49
|
Abstract
The prevalence of allergic disease has dramatically increased over the past 30 years in Westernized countries. It is unlikely that the rapid increase in the prevalence of allergic disease is the result of genetic changes, which highlights the importance of environmental factors in the development of allergic disease. The "hygiene hypothesis" was put forward in 1989 and focused attention on the notion that exposure to microbes and their products in early life can modify the risk for development of allergic disease. Infections were thought to polarize the immunological response toward a Th2-mediated immune response causing allergic disease. However, it is likely that the Th1/Th2 imbalance is too simplistic to explain the increased prevalence of allergic disease. Current research is focusing on understanding the role of T-regulatory cells in inducing a state of tolerance and the resulting modified Th2 response observed in natural and induced tolerance.
Collapse
Affiliation(s)
- Meinir G Jones
- Department of Occupational and Environmental Medicine, Imperial College London, London, UK.
| |
Collapse
|
50
|
Martínez-Blanco M, Pérez-Rodríguez L, Lozano-Ojalvo D, Molina E, López-Fandiño R. Ovalbumin-Derived Peptides Activate Retinoic Acid Signalling Pathways and Induce Regulatory Responses Through Toll-Like Receptor Interactions. Nutrients 2020; 12:nu12030831. [PMID: 32245005 PMCID: PMC7146383 DOI: 10.3390/nu12030831] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/03/2020] [Accepted: 03/17/2020] [Indexed: 12/22/2022] Open
Abstract
This study investigates the potential of a hydrolysate of ovalbumin with pepsin (OP) to preclude Th2-type immunity by the enhancement of tolerogenic dendritic cells (DCs) and regulatory T (Treg) cells. Through Toll-like receptor (TLR) stimulation, OP enhances the retinoic acid pathway on DCs by means of the induction of aldehyde dehydrogenase enzymes and transforming growth factor beta (TGF-β), and it confers upon DC the ability to upregulate interleukin 10 (IL-10) as well as other tolerance-promoting mediators downstream of TRL signalling, such as IL-27, IL-33, Notch ligands, OX40L, and the transcription factors IRF4 and IRF8. OP-conditioned DCs induce the expansion of Foxp3+ and Tr1 cells in co-culture with CD4+ T cells. Furthermore, OP directly conditions CD4+ T cells from naïve mice, without the mediation of DCs, to express aldehyde dehydrogenase (ALDH) enzymes and, in the presence of the Th2 cytokine IL-4 and exogenous TGF-β, it enhances Foxp3 expression. It is noteworthy that, on CD4+ T cells isolated from egg-allergic mice, OP significantly enriches the levels of Foxp3+ and Foxp3+ RORγt+ CD4+ T cells. In conclusion, we show that food peptides may work, analogously to microbial-driven signals, through TLRs, to promote a tolerogenic phenotype on cells of the innate and adaptive immune system, a property that is further enhanced in the context of a Th2 cytokine-rich environment.
Collapse
|