1
|
Cheah S, Lowe AJ, Afshar N, Bassett JK, Bruinsma FJ, Cozen W, Harrison SJ, Hopper JL, Jayasekara H, Prince HM, Vajdic CM, Doo NW, Giles GG, Dharmage SC, Milne RL. Allergic disease and risk of multiple myeloma: A case-control study. Cancer Epidemiol 2025; 97:102839. [PMID: 40378505 DOI: 10.1016/j.canep.2025.102839] [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: 12/22/2024] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/19/2025]
Abstract
BACKGROUND AND AIMS Multiple myeloma (MM) is responsible for significant morbidity and mortality, yet our knowledge regarding MM aetiology remains limited. We investigated whether a history of allergic conditions is associated with MM risk. METHODS Incident cases (n = 782) of MM were recruited via cancer registries in Victoria and NSW. Controls (n = 733) were siblings (n = 436) or spouses (n = 297) of cases. Unconditional logistic regression was used to estimate odds ratios (OR) and 95 % confidence intervals (CI) for associations between self-reported allergic conditions (asthma, eczema, food allergy, hay fever) and MM risk. RESULTS Eczema was inversely associated with MM risk (OR = 0.54, 95 %CI = 0.42-0.70), as was a combined history of food allergy and eczema (OR = 0.52, 95 %CI = 0.29-0.93). There was an inverse association between a history of any allergic condition (compared with none) and risk of MM (OR = 0.68, 95 %CI = 0.55-0.84). In the mean-centred dose-risk analysis the OR was 0.87 (95 %CI = 0.73-1.04) per additional allergic condition of interest. No notable associations were identified for food allergy, asthma, or hay fever alone. CONCLUSIONS AND FUTURE DIRECTIONS We found that a history of allergic disease, particularly eczema, was associated with reduced MM risk. Further research is recommended to confirm findings and investigate potential mechanisms.
Collapse
Affiliation(s)
- Simon Cheah
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - Adrian J Lowe
- Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia; Murdoch Children's Research Institute, Melbourne, Australia
| | - Nina Afshar
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - Julie K Bassett
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - Fiona J Bruinsma
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia; Burnet Institute, Melbourne, Australia
| | - Wendy Cozen
- University of California, Irvine, United States
| | - Simon J Harrison
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia; Clinical Haematology Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Parkville, Australia
| | - John L Hopper
- Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - Harindra Jayasekara
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - H Miles Prince
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia; Epworth Healthcare, Melbourne, Australia
| | | | - Nicole Wong Doo
- Concord Clinical School, University of Sydney, Sydney, Australia
| | - Graham G Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia; Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Melbourne, Australia
| | - Shyamali C Dharmage
- Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia; Murdoch Children's Research Institute, Melbourne, Australia
| | - Roger L Milne
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia; Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Melbourne, Australia.
| |
Collapse
|
2
|
Khan MA, Bhusal S, Lau CL, Krupnick AS. Bronchial anastomotic complications as a microvascular disruption in a mouse model of airway transplantation. Front Immunol 2025; 16:1567657. [PMID: 40438113 PMCID: PMC12116303 DOI: 10.3389/fimmu.2025.1567657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 04/23/2025] [Indexed: 06/01/2025] Open
Abstract
Lung transplantation (LTx) offers a last resort for patients battling end-stage lung disease. Even though short-term survival has improved, these patients still face several long-term challenges, such as chronic rejection and ischemic bronchial anastomosis. In lung transplant recipients, the bronchial anastomosis is prone to complications-such as poor wound healing, necrosis, stenosis, and dehiscence-due to the marginal blood supply at this site. During peri-LTx, hypoxia and ischemia stimulate fibrotic and inflammatory cytokines at anastomotic sites, leading to abnormal collagen production and excessive granulation, which impair wound healing. Despite meticulous techniques, bronchial anastomosis remains a major cause of morbidity and mortality among lung transplant recipients. After LTx, most bronchial complications are attributed to ischemic insult since normal bronchial blood flow is disrupted, and bronchial revascularization usually takes two to four weeks, making the anastomotic bronchial vessels dependent on pulmonary artery circulation. It is clear that hypoxia, inflammation, oxidative stress, and extracellular matrix remodeling play critical roles in bronchial complications, but there is no small animal model to study them. In the context of LTx, mouse tracheal models are essential tools for studying bronchial complications, particularly ischemia, fibrosis, and stenosis, as well as evaluating potential therapeutic interventions. A well-established mouse model of orthotopic tracheal transplantation (OTT) mimics the anastomosis of the bronchi and the subsequent microvascular injury, providing a pathological correlation with anastomotic complications. A series of previous studies using the OTT model explored the microvascularization, ischemia-reperfusion, airway epithelial injury, and fibrotic remodeling effects after airway anastomosis. This review describes OTT as a model of airway anastomotic complications, which is crucial for understanding the immunological and molecular pathways as seen in clinical bronchial anastomoses, as well as improving anastomotic healing and reducing complications through targeted therapeutic strategies.
Collapse
|
3
|
Choi S, Yoo SA, Ji KY, Jung DH, Lee S, Lee KG, Kim KM, Lee JY, Jung MA, Pyun BJ, Hur J, Choi JY, Rhee CK, Kim WU, Kim T. Asthma Alleviation by Ginsenoside Rb1 via Promotion of Treg Proliferation and Inflammatory T Cell Inhibition. Allergy 2025. [PMID: 40251907 DOI: 10.1111/all.16551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 01/09/2025] [Accepted: 02/20/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Regulatory T cells (Tregs) are living drugs with feasibility, tolerability, and therapeutic benefits. Although Tregs are linked to asthma prognosis through inflammation regulation, no therapeutic agents specifically designed to manage asthma by upregulating Tregs have been developed to date. METHODS We screened a library of 250 natural products using a cytometric bead array. Among the selected candidates, gRb1 was identified for further investigation. The effects of gRb1 on Treg and Th17 populations were evaluated in mouse asthma models and human PBMCs from both healthy donors and asthma patients using flow cytometry and cytokine analysis. RESULTS In inflammatory conditions, ginsenoside Rb1 (gRb1, a major ginseng component) increased IL-10- and TGF-β-expressing Treg populations and decreased the Th17 population; activated phospho-STAT5 and NFAT1 in Tregs; inhibited NFAT1 activation in conventional T cells (Tconvs); increased Treg proliferation and Tconv-Treg differentiation, inhibiting Tconv proliferation; and reduced inflammatory cytokine secretion by Tconvs. In asthma model mice, suppression of asthma symptoms by gRb1 was associated with elevated Treg and lower Th17, Th1, and Th2 counts. gRb1 treatment of stimulated PBMCs from patients with asthma and healthy donors increased IL-10- and TGF-β-expressing Treg populations and decreased IL-17A-, IL-22-, IFN-γ-, and TNF-α-expressing T-cell populations. CONCLUSIONS gRb1 alleviate asthma by shifting the Treg-inflammatory T cell balance. These findings suggest a strategy for enhancing Treg activity through treatment with gRb1. This may provide a novel therapeutic approach for asthma and related disorders.
Collapse
Affiliation(s)
- Susanna Choi
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Seung-Ah Yoo
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kon-Young Ji
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Dong Ho Jung
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Saseong Lee
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kang-Gu Lee
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ki-Myo Kim
- Department of Plastic and Reconstructive Surgery, Seoul National University College of Medicine, Seoul National University Boramae Hospital, Seoul, Republic of Korea
| | - Joo Young Lee
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Myung-A Jung
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Bo-Jeong Pyun
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Jung Hur
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Joon Young Choi
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Republic of Korea
| | - Chin Kook Rhee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Wan-Uk Kim
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Taesoo Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| |
Collapse
|
4
|
Wu S, Chen H, Yu R, Li H, Zhao J, Stanton C, Paul Ross R, Chen W, Yang B. Human milk oligosaccharides 2'-fucosyllactose and 3-fucosyllactose attenuate ovalbumin-induced food allergy through immunoregulation and gut microbiota modulation. Food Funct 2025; 16:1267-1283. [PMID: 39918321 DOI: 10.1039/d4fo04638b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Abstract
The prebiotic properties of human milk oligosaccharides (HMOs) and emerging evidence of immunomodulatory effects suggest their potential therapeutic value in allergy management. 2'-Fucosyllactose (2'-FL) has been reported to alleviate food allergies, while the effect of other fucosylated HMOs on food allergy remains unclear. In this study, we assess the effect of two HMOs, 2'-FL and 3-fucosyllactose (3-FL), on symptomatology and immunological responses in an ovalbumin (OVA)-sensitized mouse model of food allergy as well as their influence on gut microbiota. The assessment of allergic symptoms, specific immunoglobulin E (IgE), and related gene expression levels in sensitized mice indicated that 3-FL was as effective as 2'-FL in alleviating food allergy. 2'-FL and 3-FL significantly decreased serum levels of OVA-specific IgE, mouse mast cell protease (mMCP-1) and IL-4 while increasing the levels of IFN-γ. Additionally, 2'-FL and 3-FL down-regulated gene expression of allergy-related cytokines in the small intestine and improved intestinal barrier damage. Furthermore, both 2'-FL and 3-FL treatment positively influenced the gut microbial profiles, in particular by enhancing the proportion of beneficial bacteria such as Lactobacillus and Bifidobacterium and decreasing the percentage of Turicibacter and Lachnospiraceae NK4A136 group, thereby modulating the immune system. Therefore, this study can provide insights into 2'-FL and 3-FL to alleviate OVA-induced allergy.
Collapse
Affiliation(s)
- Siya Wu
- State Key Laboratory of Food Science and Resources, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Haiqin Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Renqiang Yu
- Department of Neonatology, Affiliated Women's Hospital of Jiangnan University, Wuxi 214002, China.
| | - Huizhen Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Laboratory for Maternal-Infant Microbiota and Health, Jiangnan University, Wuxi 214122, China
| | - Catherine Stanton
- International Joint Research Laboratory for Maternal-Infant Microbiota and Health, Jiangnan University, Wuxi 214122, China
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Co. Cork, Ireland
| | - R Paul Ross
- International Joint Research Laboratory for Maternal-Infant Microbiota and Health, Jiangnan University, Wuxi 214122, China
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Bo Yang
- State Key Laboratory of Food Science and Resources, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Laboratory for Maternal-Infant Microbiota and Health, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
5
|
Loperfido A, Cavaliere C, Fionda B, Bellocchi G, Masieri S, Caminati M. Narrative Review of Genetic and Immunological Mechanisms Involved in the Pathogenesis of Kimura's Disease: New Therapeutic Targets. Genes (Basel) 2025; 16:194. [PMID: 40004521 PMCID: PMC11855017 DOI: 10.3390/genes16020194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/29/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Kimura's disease (KD) is a rare, chronic inflammatory disorder that predominantly affects young men of East Asian descent. It is characterized by painless solid masses primarily localized to the deep subcutaneous tissues of the head and neck, eosinophilia, and elevated serum immunoglobulin E (IgE). While the exact cause remains unclear, the pathogenesis is thought to involve dysregulated immune responses, particularly those mediated by T-helper cells 2 (Th2), eosinophils, and IgE production. Advances in molecular biology have suggested that genetic factors play a significant role in the development and progression of this chronic inflammatory condition. Recent studies have implicated several genes and immune pathways in its development, and understanding these genetic components may provide insights into better diagnostic tools and therapeutic strategies for KD. In this regard, biological therapies, by targeting the immune mechanisms underlying KD, have been used to treat this challenging condition with promising results, contributing to a better understanding of the pathogenesis of this rare disorder. The aim of this study was to review the literature concerning the genetic factors and immune mechanisms that contribute to the pathogenesis of KD, with a special focus on the role of biological therapies.
Collapse
Affiliation(s)
- Antonella Loperfido
- Otolaryngology Unit, San Camillo Forlanini Hospital, Circonvallazione Gianicolense 87, 00152 Rome, Italy
| | - Carlo Cavaliere
- Department of Sense Organs, Sapienza University, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Bruno Fionda
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Gianluca Bellocchi
- Otolaryngology Unit, San Camillo Forlanini Hospital, Circonvallazione Gianicolense 87, 00152 Rome, Italy
| | - Simonetta Masieri
- Department of Oral and Maxillofacial Sciences, Sapienza University, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Marco Caminati
- Allergy Unit and Asthma Center, Verona Integrated University Hospital, 37134 Verona, Italy
- Department of Medicine, University of Verona, 37124 Verona, Italy
| |
Collapse
|
6
|
Rosenzwajg M, Gherasim A, Dietsch F, Beck M, Domis N, Lorenzon R, Chantran Y, Bellier B, Vicaut E, Soria A, de Blay F, Klatzmann D. Low-dose IL-2 in birch pollen allergy: A phase-2 randomized double-blind placebo-controlled trial. J Allergy Clin Immunol 2025; 155:650-655. [PMID: 39532189 DOI: 10.1016/j.jaci.2024.10.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Regulatory T (Treg) cells are pivotal in immune tolerance to allergens. Low-dose IL-2 (IL-2LD) activates Treg cells. OBJECTIVE Our aim was to assess IL-2LD efficacy for controlling clinical responses to allergen exposures. METHODS RHINIL-2 was a phase-2a, randomized, double-blind, placebo-controlled trial. Patients with allergic rhinitis to birch pollen (BP) were included; 66% of them had concomitant asthma. All had a total nasal symptom score (TNSS) of 5 or more following nasal exposure to BP in an environmental exposure chamber. Patients received 1 MUI per day of IL-2 (n = 12) or placebo (n = 12) for 5 days, followed by weekly injections for 4 weeks. Clinical responses to subsequent BP exposures in the environmental exposure chamber were evaluated by using TNSS, the rhinitis visual analog scale (VAS), and spirometry. The primary efficacy end point was the difference in TNSS area under the curve (AUC) between inclusion and day 40. RESULTS IL-2LD treatment induced a significant expansion of Treg cells. The difference in TNSS AUC between inclusion and day 40 AUC in the IL-2 and placebo groups was not significant. TNSS and visual analog scale AUCs were significantly reduced from baseline to day 40 in the IL-2LD group only (P = .04 and P = .01, respectively). The ratio of FEV1 to forced vital capacity (FEV1P) and the forced midexpiratory flow (FEF25%-75%) showed improvement in the IL-2LD-treated versus in the groups given placebo at day 40 (P = .04 and P = .04, respectively). However, the short treatment duration used in this study could not have effects on specific IgE or IgG4 levels given their half-life. There were no severe treatment-related adverse events. CONCLUSION IL-2LD is well tolerated in patients with allergy, even in those with asthma, thus clearing the path for further therapeutic development. Our work suggests that Treg cells can safely attenuate an ongoing allergic response. It paves the way for larger studies with longer treatment periods, which are needed to properly evaluate the therapeutic potential of IL-2 in allergy.
Collapse
Affiliation(s)
- Michelle Rosenzwajg
- Biotherapy Clinical Investigation Center (CIC-BTi) and Inflammation-Immunopathology-Immunotherapy Department (i2B), Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; Immunology-Immunopathology-Immunotherapy, Sorbonne Université, INSERM, UMR_S 959, Paris, France
| | - Alina Gherasim
- Centre de recherche clinique ALYATEC, Strasbourg, France
| | - Franck Dietsch
- Centre de recherche clinique ALYATEC, Strasbourg, France
| | - Marine Beck
- Centre de recherche clinique ALYATEC, Strasbourg, France
| | - Nathalie Domis
- Centre de recherche clinique ALYATEC, Strasbourg, France
| | - Roberta Lorenzon
- Biotherapy Clinical Investigation Center (CIC-BTi) and Inflammation-Immunopathology-Immunotherapy Department (i2B), Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; Immunology-Immunopathology-Immunotherapy, Sorbonne Université, INSERM, UMR_S 959, Paris, France
| | - Yannick Chantran
- Immunology Department, Saint Antoine Hospital, AP-HP, Paris, France
| | - Bertrand Bellier
- Immunology-Immunopathology-Immunotherapy, Sorbonne Université, INSERM, UMR_S 959, Paris, France
| | - Eric Vicaut
- Unité de recherche clinique, Saint Louis/Lariboisière Hospitals, AP-HP, and Université Paris 7, Paris, France
| | - Angele Soria
- Dermatology and Allergy Department, Tenon Hospital, AP-HP, Paris
| | | | - David Klatzmann
- Biotherapy Clinical Investigation Center (CIC-BTi) and Inflammation-Immunopathology-Immunotherapy Department (i2B), Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; Immunology-Immunopathology-Immunotherapy, Sorbonne Université, INSERM, UMR_S 959, Paris, France.
| |
Collapse
|
7
|
Chandwaskar R, Dalal R, Gupta S, Sharma A, Parashar D, Kashyap VK, Sohal JS, Tripathi SK. Dysregulation of T cell response in the pathogenesis of inflammatory bowel disease. Scand J Immunol 2024; 100:e13412. [PMID: 39394898 DOI: 10.1111/sji.13412] [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: 11/20/2023] [Revised: 08/26/2024] [Accepted: 09/17/2024] [Indexed: 10/14/2024]
Abstract
Inflammatory bowel disease (IBD), comprised of Crohn's disease (CD) and ulcerative colitis (UC), are gut inflammatory diseases that were earlier prevalent in the Western Hemisphere but now are on the rise in the East, with India standing second highest in the incidence rate in the world. Inflammation in IBD is a cause of dysregulated immune response, wherein helper T (Th) cell subsets and their cytokines play a major role in the pathogenesis of IBD. In addition, gut microbiota, environmental factors such as dietary factors and host genetics influence the outcome and severity of IBD. Dysregulation between effector and regulatory T cells drives gut inflammation, as effector T cells like Th1, Th17 and Th9 subsets Th cell lineages were found to be increased in IBD patients. In this review, we attempted to discuss the role of different Th cell subsets together with other T cells like CD8+ T cells, NKT and γδT cells in the outcome of gut inflammation in IBD. We also highlighted the potential therapeutic candidates for IBD.
Collapse
Affiliation(s)
- Rucha Chandwaskar
- Amity Institute of Microbial Technology (AIMT), Amity University Jaipur, Rajasthan, India
| | - Rajdeep Dalal
- Infection and Immunology Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, India
| | - Saurabh Gupta
- Centre for Vaccines and Diagnostic Research, GLA University, Mathura, Uttar Pradesh, India
| | - Aishwarya Sharma
- Sri Siddhartha Medical College and Research Center, Tumkur, Karnataka, India
| | - Deepak Parashar
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Vivek K Kashyap
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas, USA
| | - Jagdip Singh Sohal
- Centre for Vaccines and Diagnostic Research, GLA University, Mathura, Uttar Pradesh, India
| | - Subhash K Tripathi
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, Washington, USA
| |
Collapse
|
8
|
Swaney MH, Steidl OR, Tackett A, Fye S, Lee KE, Ong IM, Bendixsen C, Spicer G, DeLine J, Gern JE, Lucey J, Seroogy CM, Kalan L, Singh AM. Farm exposure is associated with human breast milk immune profile and microbiome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618271. [PMID: 39464075 PMCID: PMC11507701 DOI: 10.1101/2024.10.14.618271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Prenatal and early life farm exposure, and breastfeeding, are associated with protection from allergic diseases. We hypothesize that farm exposure influences the human breast milk microbiome and immune proteins. The immune protein profiles and microbial communities of 152 human breast milk samples were compared among three maternal farm exposure groups (traditional agrarian, farm, and non-farm) in rural Wisconsin to identify signatures associated with farm status and atopic disease. We found significant differences between farm groups for 23 immune proteins (p-adj<0.05), microbiome diversity (p=2.2E-05), and microbiome richness (p=8.0e-06). Traditional agrarian human breast milk had the highest immune protein levels and microbiome diversity and richness, followed by farm and non-farm human breast milk. Furthermore, Gram-positive bacterial species correlated with IL-23 mediated signaling events (p-adj<1.0E-05). These data suggest that increased farm exposures promotes human breast milk that is more microbially-diverse and rich in immune-associated proteins, ultimately influencing immune development in the infant.
Collapse
Affiliation(s)
- Mary Hannah Swaney
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Microbiology Doctoral Training Program, University of Wisconsin, Madison, WI, USA
| | - Olivia Rae Steidl
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Anastasia Tackett
- Department of Food Science, University of Wisconsin-Madison, Madison, WI, USA
| | - Samantha Fye
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Kristine E. Lee
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Irene M. Ong
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Center for Human Genomics and Precision Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Casper Bendixsen
- National Farm Medicine Center, Marshfield Clinic Research Institute, Marshfield, WI, USA
| | - Gretchen Spicer
- LaFarge Medical Clinic, Vernon Memorial Health Care, LaFarge, WI, USA
| | - James DeLine
- LaFarge Medical Clinic, Vernon Memorial Health Care, LaFarge, WI, USA
| | - James E Gern
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - John Lucey
- Department of Food Science, University of Wisconsin-Madison, Madison, WI, USA
| | - Christine M. Seroogy
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Lindsay Kalan
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- M.G. DeGroote Institute for Infectious Disease Research
- David Braley Centre for Antibiotic Discovery
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Anne Marie Singh
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
9
|
Georgia AN, Claudine NE, Carole SN, Loveline NN, Abel L, Flaurent TT, Martin S, Waffo AB, Okeke M, Esimone C, Park CG, Vittorio C, François-Xavier E, Godwin NW. Regulatory T cells modulate monocyte functions in immunocompetent antiretroviral therapy naive HIV-1 infected people. BMC Immunol 2024; 25:68. [PMID: 39402453 PMCID: PMC11472541 DOI: 10.1186/s12865-024-00654-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
We previously demonstrated that the overall number of regulatory T (Treg) cells decrease proportionately with helper CD4+ T cells and their frequencies increase in antiretroviral therapy (ART)-naive human immunodeficiency virus type-1 (HIV-1) infected individuals. The question now is whether the discrepancies in Treg cell numbers and frequencies are synonymous to an impairment of their functions. To address this, we purified Treg cells and assessed their ability to modulate autologous monocytes functions. We observed that Treg cells were able to down modulate autologous monocytes activation as well as interleukin 6 (IL-6) and tumor necrosis factor-alpha (TNF-α) production during stimulation with polyinosinic-polycytidylic acid stabilized with poly-L-lysine and carboxymethylcellulose (poly-ICLC). This activity of Treg cells has been shown to be influenced by immunocompetence including but not limited to helper CD4+ T cell counts, in individuals with HIV-1 infection. Compared to immunosuppressed participants (CD4 < 500 cells/µL), immunocompetent participants (CD4 ≥ 500 cells/µL) showed significantly higher levels of transforming growth factor beta (TGF-β) and IL-10 (p < 0.001 and p < 0.05, respectively), key cytokines used by Treg cells to exert their immunosuppressive functions. Our findings suggest the contribution of both TGF-β and IL-10 in the suppressive activity of Treg cells.
Collapse
Affiliation(s)
- Ambada N Georgia
- Laboratory of Vaccinology/Biobanking, The Chantal Biya International Reference Center (CIRCB) for Research on the Prevention and Management of HIV/AIDS, P.O. Box: 3077, Messa Yaoundé, Cameroon.
- Department of Animal Biology and Physiology, University of Yaoundé I, Yaoundé, Cameroon.
| | - Ntsama E Claudine
- Department of Animal Biology and Physiology, University of Yaoundé I, Yaoundé, Cameroon
| | - Sake N Carole
- Laboratory of Vaccinology/Biobanking, The Chantal Biya International Reference Center (CIRCB) for Research on the Prevention and Management of HIV/AIDS, P.O. Box: 3077, Messa Yaoundé, Cameroon
- Department of Microbiology, University of Yaoundé I, Yaoundé, Cameroon
| | - Ngu N Loveline
- Laboratory of Vaccinology/Biobanking, The Chantal Biya International Reference Center (CIRCB) for Research on the Prevention and Management of HIV/AIDS, P.O. Box: 3077, Messa Yaoundé, Cameroon
- Department of Biochemistry, University of Yaoundé I, Yaoundé, Cameroon
| | - Lissom Abel
- Laboratory of Vaccinology/Biobanking, The Chantal Biya International Reference Center (CIRCB) for Research on the Prevention and Management of HIV/AIDS, P.O. Box: 3077, Messa Yaoundé, Cameroon
- Faculty of Science, Department of Biological Science, University of Bamenda, Bamenda, Cameroon
| | - Tchouangeu T Flaurent
- Laboratory of Vaccinology/Biobanking, The Chantal Biya International Reference Center (CIRCB) for Research on the Prevention and Management of HIV/AIDS, P.O. Box: 3077, Messa Yaoundé, Cameroon
- Department of Biochemistry, University of Dschang, Dschang, Cameroon
| | - Sosso Martin
- Laboratory of Vaccinology/Biobanking, The Chantal Biya International Reference Center (CIRCB) for Research on the Prevention and Management of HIV/AIDS, P.O. Box: 3077, Messa Yaoundé, Cameroon
| | - Alain Bopda Waffo
- Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, USA
| | - Malachy Okeke
- Department of Natural and Environmental Sciences, Biomedical Science Concentration, School of Arts and Sciences, American University of Nigeria, 98 Lamido Zubairu Way, Yola, PMB, 2250, Nigeria
| | - Charles Esimone
- Department of Pharmaceutical Microbiology & Biotechnology, Nnamdi Azikiwe University, Awka, Nigeria
| | - Chae Gyu Park
- Laboratory of Immunology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | | | - Nchinda W Godwin
- Laboratory of Vaccinology/Biobanking, The Chantal Biya International Reference Center (CIRCB) for Research on the Prevention and Management of HIV/AIDS, P.O. Box: 3077, Messa Yaoundé, Cameroon
- Department of Pharmaceutical Microbiology & Biotechnology, Nnamdi Azikiwe University, Awka, Nigeria
| |
Collapse
|
10
|
Knott-Torcal C, de la Blanca NS, Serrano-Somavilla A, Hernández RM, Sampedro-Núñez M, Ruiz-Rosso B, Jiménez-Blanco S, González-Amaro R, González-Baranda L, Garcimartin A, Marazuela M. Quantitative analysis of Tr1 lymphocytes in patients with type 2 diabetes mellitus. J Endocrinol Invest 2024; 47:1447-1455. [PMID: 38183564 PMCID: PMC11142976 DOI: 10.1007/s40618-023-02250-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 11/13/2023] [Indexed: 01/08/2024]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is usually accompanied by a low-grade inflammatory phenomenon, which participates in the pathogenesis of different complications of this condition. The inflammatory response is under the regulation of different mechanisms, including T regulatory (Treg) lymphocytes. However, the possible role of type 1 T regulatory (Tr1) cells in T2DM has not been explored so far. AIM To carry out a quantitative analysis of Tr1 lymphocytes and other immune cell subsets in patients with T2DM and correlate these results with clinical findings and treatments. MATERIALS AND METHODS Sixty patients with T2DM and twenty-three healthy controls were included in the study. Biochemical and anthropometric variables were evaluated, and Tr1 lymphocytes (CD4+CD49+LAG-3+IL-10+) and other cell subsets (Th17, Th22 and Foxp3 + Treg cells) were analyzed in peripheral blood samples by multiparametric flow cytometry. RESULTS Significant increased levels of Tr1 cells were detected in patients with severe and mild disease, compared to healthy controls. In addition, CD4+IL-10+ lymphocytes were also increased in patients with T2DM. In contrast, similar levels of Foxp3+ Treg cells, Th17 and Th22 lymphocytes were observed in patients and controls. Likewise, no significant associations were detected between Tr1 cell levels and different clinical and laboratory parameters. However, those patients receiving glucagon-like peptide-1 receptor agonists (GLP-1-RA) showed similar levels of Tr1 cells than healthy controls, and significant lower numbers than untreated patients. CONCLUSION We observed an increase in Tr1 and CD4+IL10+ lymphocyte levels in T2DM. Moreover, GLP1-RA treatment was significantly associated with normalization of the Tr1 levels. This highlights another potential immune dysfunction in patients with T2DM, which could participate in the pathogenesis of this condition.
Collapse
Affiliation(s)
- C Knott-Torcal
- Department of Endocrinology and Nutrition, Health Research Institute, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, C/Diego de León 62, 28006, Madrid, Spain
- Faculty of Pharmacy, Universidad Complutense de Madrid, Av. Séneca, 2, 28040, Madrid, Spain
| | - N S de la Blanca
- Department of Endocrinology and Nutrition, Health Research Institute, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, C/Diego de León 62, 28006, Madrid, Spain
| | - A Serrano-Somavilla
- Department of Endocrinology and Nutrition, Health Research Institute, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, C/Diego de León 62, 28006, Madrid, Spain
| | - R M Hernández
- Department of Endocrinology and Nutrition, Health Research Institute, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, C/Diego de León 62, 28006, Madrid, Spain
| | - M Sampedro-Núñez
- Department of Endocrinology and Nutrition, Health Research Institute, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, C/Diego de León 62, 28006, Madrid, Spain
| | - B Ruiz-Rosso
- Faculty of Pharmacy, Universidad Complutense de Madrid, Av. Séneca, 2, 28040, Madrid, Spain
| | - S Jiménez-Blanco
- Department of Endocrinology and Nutrition, Health Research Institute, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, C/Diego de León 62, 28006, Madrid, Spain
| | - R González-Amaro
- Research Center of Health Sciences and Biomedicine (CICSaB), Universidad Autónoma de San Luis Potosí, SLP, México
| | - L González-Baranda
- Research Center of Health Sciences and Biomedicine (CICSaB), Universidad Autónoma de San Luis Potosí, SLP, México
| | - A Garcimartin
- Faculty of Pharmacy, Universidad Complutense de Madrid, Av. Séneca, 2, 28040, Madrid, Spain.
| | - M Marazuela
- Department of Endocrinology and Nutrition, Health Research Institute, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, C/Diego de León 62, 28006, Madrid, Spain.
| |
Collapse
|
11
|
Headen K, Jakaite V, Mesaric VA, Scotta C, Lombardi G, Nicolaides KH, Shangaris P. The Role of Regulatory T Cells and Their Therapeutic Potential in Hypertensive Disease of Pregnancy: A Literature Review. Int J Mol Sci 2024; 25:4884. [PMID: 38732104 PMCID: PMC11084408 DOI: 10.3390/ijms25094884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Hypertensive disorders of pregnancy (HDP), including preeclampsia (PE) and gestational hypertension (GH), are major causes of maternal and foetal morbidity and mortality. This review elucidates the role of regulatory T cells (Tregs) in the immunological aspects of HDP and explores their therapeutic potential. Tregs, which play a critical role in maintaining immune homeostasis, are crucial in pregnancy to prevent immune-mediated rejection of the foetus. The review highlights that Tregs contribute to immunological adaptation in normal pregnancy, ensuring foetal acceptance. In contrast, HDP is associated with Treg dysfunction, which is marked by decreased numbers and impaired regulatory capacity, leading to inadequate immune tolerance and abnormal placental development. This dysfunction is particularly evident in PE, in which Tregs fail to adequately modulate the maternal immune response against foetal antigens, contributing to the pathophysiology of the disorder. Therapeutic interventions aiming to modulate Treg activity represent a promising avenue for HDP management. Studies in animal models and limited clinical trials suggest that enhancing Treg functionality could mitigate HDP symptoms and improve pregnancy outcomes. However, given the multifactorial nature of HDP and the intricate regulatory mechanisms of Tregs, the review explores the complexities of translating in vitro and animal model findings into effective clinical therapies. In conclusion, while the precise role of Tregs in HDP is still being unravelled, their central role in immune regulation during pregnancy is indisputable. Further research is needed to fully understand the mechanisms by which Tregs contribute to HDP and to develop targeted therapies that can safely and effectively harness their regulatory potential for treating hypertensive diseases of pregnancy.
Collapse
Affiliation(s)
- Kyle Headen
- Department of Women and Children’s Health, School of Life Course Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE5 8AF, UK; (K.H.); (K.H.N.)
- Harris Birthright Research Centre for Fetal Medicine, King’s College Hospital, London SE5 8BB, UK; (V.J.); (V.A.M.)
| | - Vaidile Jakaite
- Harris Birthright Research Centre for Fetal Medicine, King’s College Hospital, London SE5 8BB, UK; (V.J.); (V.A.M.)
| | - Vita Andreja Mesaric
- Harris Birthright Research Centre for Fetal Medicine, King’s College Hospital, London SE5 8BB, UK; (V.J.); (V.A.M.)
| | - Cristiano Scotta
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 1UL, UK; (C.S.); (G.L.)
| | - Giovanna Lombardi
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 1UL, UK; (C.S.); (G.L.)
| | - Kypros H. Nicolaides
- Department of Women and Children’s Health, School of Life Course Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE5 8AF, UK; (K.H.); (K.H.N.)
- Harris Birthright Research Centre for Fetal Medicine, King’s College Hospital, London SE5 8BB, UK; (V.J.); (V.A.M.)
| | - Panicos Shangaris
- Department of Women and Children’s Health, School of Life Course Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE5 8AF, UK; (K.H.); (K.H.N.)
- Harris Birthright Research Centre for Fetal Medicine, King’s College Hospital, London SE5 8BB, UK; (V.J.); (V.A.M.)
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 1UL, UK; (C.S.); (G.L.)
- Immunoregulation Laboratory, Faculty of Life Sciences & Medicine, 5th Floor, Bermondsey Wing, Guy’s Hospital, London SE1 9RT, UK
| |
Collapse
|
12
|
Patwardhan RS, Gohil D, Singh B, Kumar BK, Purohit V, Thoh M, Checker R, Gardi N, Gota V, Kutala VK, Patwardhan S, Sharma D, Sandur SK. Mitochondrial-targeted curcumin inhibits T-cell activation via Nrf2 and inhibits graft-versus-host-disease in a mouse model. Phytother Res 2024; 38:1555-1573. [PMID: 38281735 DOI: 10.1002/ptr.8126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 12/31/2023] [Accepted: 01/07/2024] [Indexed: 01/30/2024]
Abstract
Anti-inflammatory and immune suppressive agents are required to moderate hyper-activation of lymphocytes under disease conditions or organ transplantation. However, selective disruption of mitochondrial redox has not been evaluated as a therapeutic strategy for suppression of T-cell-mediated pathologies. Using mitochondrial targeted curcumin (MitoC), we studied the effect of mitochondrial redox modulation on T-cell responses by flow cytometry, transmission electron microscopy, transcriptomics, and proteomics, and the role of Nrf2 was studied using Nrf2- /- mice. MitoC decreased mitochondrial TrxR activity, enhanced mitochondrial ROS (mROS) production, depleted mitochondrial glutathione, and suppressed activation-induced increase in mitochondrial biomass. This led to suppression of T-cell responses and metabolic reprogramming towards Treg differentiation. MitoC induced nuclear translocation and DNA binding of Nrf2, leading to upregulation of Nrf2-dependent genes and proteins. MitoC-mediated changes in mitochondrial redox and modulation of T-cell responses are abolished in Nrf2- /- mice. Restoration of mitochondrial thiols abrogated inhibition of T-cell responses. MitoC suppressed alloantigen-induced lymphoblast formation, inflammatory cytokines, morbidity, and mortality in acute graft-versus-host disease mice. Disruption of mitochondrial thiols but not mROS increase inculcates an Nrf2-dependent immune-suppressive disposition in T cells for the propitious treatment of graft-versus-host disease.
Collapse
Affiliation(s)
| | - Dievya Gohil
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Babita Singh
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Binita K Kumar
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Vaitashi Purohit
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Maikho Thoh
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Rahul Checker
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
| | - Nilesh Gardi
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Vikram Gota
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Vijay Kumar Kutala
- Department of Biochemistry, Nizam's Institute of Medical Sciences (NIMS), Hyderabad, India
| | - Sejal Patwardhan
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Deepak Sharma
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
| | - Santosh K Sandur
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
| |
Collapse
|
13
|
Kennedy-Batalla R, Acevedo D, Luo Y, Esteve-Solé A, Vlagea A, Correa-Rocha R, Seoane-Reula ME, Alsina L. Treg in inborn errors of immunity: gaps, knowns and future perspectives. Front Immunol 2024; 14:1278759. [PMID: 38259469 PMCID: PMC10800401 DOI: 10.3389/fimmu.2023.1278759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 12/13/2023] [Indexed: 01/24/2024] Open
Abstract
Regulatory T cells (Treg) are essential for immune balance, preventing overreactive responses and autoimmunity. Although traditionally characterized as CD4+CD25+CD127lowFoxP3hi, recent research has revealed diverse Treg subsets such as Tr1, Tr1-like, and CD8 Treg. Treg dysfunction leads to severe autoimmune diseases and immune-mediated inflammatory disorders. Inborn errors of immunity (IEI) are a group of disorders that affect correct functioning of the immune system. IEI include Tregopathies caused by genetic mutations affecting Treg development or function. In addition, Treg dysfunction is also observed in other IEIs, whose underlying mechanisms are largely unknown, thus requiring further research. This review provides a comprehensive overview and discussion of Treg in IEI focused on: A) advances and controversies in the evaluation of Treg extended subphenotypes and function; B) current knowledge and gaps in Treg disturbances in Tregopathies and other IEI including Treg subpopulation changes, genotype-phenotype correlation, Treg changes with disease activity, and available therapies, and C) the potential of Treg cell-based therapies for IEI with immune dysregulation. The aim is to improve both the diagnostic and the therapeutic approaches to IEI when there is involvement of Treg. We performed a non-systematic targeted literature review with a knowledgeable selection of current, high-quality original and review articles on Treg and IEI available since 2003 (with 58% of the articles within the last 6 years) in the PubMed database.
Collapse
Affiliation(s)
- Rebeca Kennedy-Batalla
- Laboratory of Immune-Regulation, Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
| | - Daniel Acevedo
- Clinical Immunology and Primary Immunodeficiencies Unit, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Yiyi Luo
- Clinical Immunology and Primary Immunodeficiencies Unit, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Ana Esteve-Solé
- Clinical Immunology and Primary Immunodeficiencies Unit, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Alexandru Vlagea
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Immunology Department, Biomedic Diagnostic Center (CDB), Hospital Clínic of Barcelona, Clinical Immunology Unit Hospital Sant Joan de Déu-Hospital Clínic de Barcelona, Barcelona, Spain
| | - Rafael Correa-Rocha
- Laboratory of Immune-Regulation, Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
| | - Ma Elena Seoane-Reula
- Laboratory of Immune-Regulation, Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
- Pediatric Immuno-Allergy Unit, Allergy Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Primary Immunodeficiencies Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Laia Alsina
- Clinical Immunology and Primary Immunodeficiencies Unit, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Department of Surgery and Surgical Specializations, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
14
|
Murao T, Kouzaki H, Arai H, Matsumoto K, Nakamura K, Kawakita K, Tojima I, Shimizu S, Yuta A, Shimizu T. Increase in the prevalence of follicular regulatory T cells correlates with clinical efficacy of sublingual immunotherapy with house dust mites. Int Forum Allergy Rhinol 2024; 14:57-67. [PMID: 37345377 DOI: 10.1002/alr.23221] [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: 01/21/2023] [Revised: 06/02/2023] [Accepted: 06/17/2023] [Indexed: 06/23/2023]
Abstract
BACKGROUND Allergic rhinitis (AR) impairs quality of life and affects nearly 40% of the Japanese population. Sublingual immunotherapy (SLIT) is the disease-modifying treatment for AR, but requires the selection of a biomarker associate with clinical efficacy in patients with AR who are treated with SLIT. The present study sought to examine objective biomarkers used for assessing the clinical efficacy of SLIT. METHODS The authors examined the effects of 1 year of SLIT treatment with house dust mites (HDMs) using peripheral blood mononuclear cells (PBMCs) and serum from patients with AR. The prevalences of follicular regulatory T (Tfr), type 2 follicular helper T (Tfh2), type 2 helper T (Th2), conventional regulatory T (Treg), and type 1 regulatory T (Tr1) cells were examined by flow cytometry. Serum concentrations of HDM-specific IgA, IgE, and IgG4 antibodies, and HDM-induced production of interleukin (IL) 5 and IL-10 from cultured PBMCs were evaluated by enzyme-linked immunosorbent assay. RESULTS Following 1 year of SLIT, the prevalences of Tfr, conventional Treg, and Tr1 cells were significantly increased, whereas that of Th2 cells and Tfh2 cells were significantly decreased; the serum concentration of HDM-specific IgG4 was significantly increased; and HDM-induced production of IL-5 from PBMCs was significantly decreased, while that of IL-10 was significantly increased. The increase in the prevalence of Tfr cells after SLIT correlated positively with the improvement of clinical symptom scores. CONCLUSION An increase in Tfr cells may play an important role in SLIT, and may be a useful indicator for the clinical efficacy of SLIT.
Collapse
Affiliation(s)
- Takuya Murao
- Department of Otorhinolaryngology-Head and Neck Surgery, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Hideaki Kouzaki
- Department of Otorhinolaryngology-Head and Neck Surgery, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Hiroyuki Arai
- Department of Otorhinolaryngology-Head and Neck Surgery, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Koji Matsumoto
- Department of Otorhinolaryngology-Head and Neck Surgery, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Keigo Nakamura
- Department of Otorhinolaryngology-Head and Neck Surgery, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Kento Kawakita
- Department of Otorhinolaryngology-Head and Neck Surgery, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Ichiro Tojima
- Department of Otorhinolaryngology-Head and Neck Surgery, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Shino Shimizu
- Department of Otorhinolaryngology-Head and Neck Surgery, Shiga University of Medical Science, Otsu, Shiga, Japan
| | | | - Takeshi Shimizu
- Department of Otorhinolaryngology-Head and Neck Surgery, Shiga University of Medical Science, Otsu, Shiga, Japan
| |
Collapse
|
15
|
Klein M, Plante S, Boulay MÈ, Boulet LP, Chakir J. Discrepancy in the suppressive function of regulatory T cells in allergic asthmatic vs. allergic rhinitis subjects upon low-dose allergen challenges. FRONTIERS IN ALLERGY 2023; 4:1296601. [PMID: 38106504 PMCID: PMC10722309 DOI: 10.3389/falgy.2023.1296601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/15/2023] [Indexed: 12/19/2023] Open
Abstract
Background Regulatory T cells (Tregs) contribute to the maintenance of immunological tolerance. There is evidence of impaired function of these cells in people with asthma and allergy. In this study, we evaluated and compared the function of Tregs in allergic asthmatic and allergic non-asthmatic patients, both before and after low-dose allergen challenges. Methods Three groups of subjects were recruited for a baseline evaluation: healthy controls without allergy or asthma, allergic asthmatic subjects, and allergic non-asthmatic subjects. All of them were subjected to expiratory flow measurements, sputum induction, and blood sampling. In addition, both groups of allergic subjects underwent low-dose allergen challenges. Tregs were isolated from whole blood using CD4+CD25high and CD127low staining. The suppression function was measured by flow cytometry. The levels of IL-10, IFN-γ, IgG4, IgA, and TGF-β were measured using ELISA, and sputum Foxp3 was evaluated using qRT-PCR. Results The suppressive function of Tregs in healthy controls was significantly higher than in allergic asthmatic or allergic non-asthmatic subjects. Repeated exposure to low doses of allergen increased the suppressor function of Tregs in allergic non-asthmatic subjects but decreased it in allergic asthmatic subjects. Foxp3 gene expression was increased in induced sputum in allergic non-asthmatic subjects, whereas it did not change in asthmatic subjects. Serum IL-10 level was decreased in allergic asthmatic subjects after allergen challenge but not in allergic non-asthmatic subjects. IFN-γ level increased upon allergen challenge in allergic non-asthmatic subjects. IgG4 level was higher in allergic non-asthmatic subjects than in allergic asthmatic subjects. Conclusions Low-dose allergen challenges stimulate the suppressor function of Tregs in non-asthmatic allergic subjects but not in allergic asthmatic subjects.
Collapse
Affiliation(s)
| | | | | | | | - Jamila Chakir
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
16
|
Chiarella SE, Barnes PJ. Endogenous inhibitory mechanisms in asthma. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2023; 2:100135. [PMID: 37781649 PMCID: PMC10509980 DOI: 10.1016/j.jacig.2023.100135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/16/2023] [Accepted: 05/21/2023] [Indexed: 10/03/2023]
Abstract
Endogenous inhibitory mechanisms promote resolution of inflammation, enhance tissue repair and integrity, and promote homeostasis in the lung. These mechanisms include steroid hormones, regulatory T cells, IL-10, prostaglandin E2, prostaglandin I2, lipoxins, resolvins, protectins, maresins, glucagon-like peptide-1 receptor, adrenomedullin, nitric oxide, and carbon monoxide. Here we review the most recent literature regarding these endogenous inhibitory mechanisms in asthma, which remain a promising target for the prevention and treatment of asthma.
Collapse
|
17
|
Auger N, Bégin P, Kang H, Lo E, Brousseau É, Healy-Profitós J, Potter BJ. Multisystem inflammatory syndrome in adults: Comparison with other inflammatory conditions during the Covid-19 pandemic. Respir Med 2023; 206:107084. [PMID: 36527990 PMCID: PMC9733296 DOI: 10.1016/j.rmed.2022.107084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/18/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Multisystem inflammatory syndrome in adults (MIS-A) is an increasingly recognized complication of Covid-19. We assessed risk factors, clinical characteristics, and outcomes of patients with MIS-A compared with other inflammatory conditions. METHODS We analyzed a cohort of patients ≥21 years hospitalized with MIS-A in Quebec, Canada between February 2020 and March 2021. We included comparison groups that share symptomatology or pathophysiology with MIS-A, including Kawasaki disease, toxic shock syndrome, and other Covid-19 complications. We examined characteristics of men and women at admission, and identified preexisting factors associated with MIS-A through odds ratios (OR) and 95% confidence intervals (CI) from adjusted logistic regression models. RESULTS Among 22,251 patients in this study, 52 had MIS-A, 90 Kawasaki disease, 500 toxic shock syndrome, and 21,609 other Covid-19 complications. MIS-A was associated with an elevated risk of respiratory failure compared with Kawasaki disease (OR 7.22, 95% CI 1.26-41.24), toxic shock syndrome (OR 4.41, 95% CI 1.73-11.23), and other Covid-19 complications (OR 3.03, 95% CI 1.67-5.50). Patients with MIS-A had a greater risk of cardiac involvement, renal failure, and mortality. The data pointed towards sex-specific differences in presentation, with more respiratory involvement in women and cardiac involvement in men compared with patients that had other Covid-19 complications. Except for allergic disorders and cancer, prior medical risk factors were not associated with a greater likelihood of MIS-A. CONCLUSIONS Patients with MIS-A have an elevated risk of mortality compared with other inflammatory conditions, with women having a predominance of respiratory complications and men cardiovascular complications.
Collapse
Affiliation(s)
- Nathalie Auger
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada; Institut national de santé publique du Québec, Montreal, Quebec, Canada; Department of Social and Preventive Medicine, School of Public Health, University of Montreal, Montreal, Quebec, Canada; Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Quebec, Canada.
| | - Philippe Bégin
- Sainte-Justine Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Harb Kang
- Department of Rheumatology, Cité-de-la-Santé Hospital, Laval, Quebec, Canada
| | - Ernest Lo
- Institut national de santé publique du Québec, Montreal, Quebec, Canada; Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Quebec, Canada
| | - Émilie Brousseau
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada; Institut national de santé publique du Québec, Montreal, Quebec, Canada
| | - Jessica Healy-Profitós
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada; Institut national de santé publique du Québec, Montreal, Quebec, Canada
| | - Brian J Potter
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada; Division of Cardiology, Department of Medicine, University of Montreal Hospital Centre, Montreal, Quebec, Canada
| |
Collapse
|
18
|
Protective role of T regulatory (Treg) cells in systemic lupus erythematosus patients with nephritis. THE EGYPTIAN RHEUMATOLOGIST 2023. [DOI: 10.1016/j.ejr.2022.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
19
|
Dai H, Fan Q, Wang C. Recent applications of immunomodulatory biomaterials for disease immunotherapy. EXPLORATION (BEIJING, CHINA) 2022; 2:20210157. [PMID: 37324799 PMCID: PMC10191059 DOI: 10.1002/exp.20210157] [Citation(s) in RCA: 136] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/25/2022] [Indexed: 06/16/2023]
Abstract
Immunotherapy is used to regulate systemic hyperactivation or hypoactivation to treat various diseases. Biomaterial-based immunotherapy systems can improve therapeutic effects through targeted drug delivery, immunoengineering, etc. However, the immunomodulatory effects of biomaterials themselves cannot be neglected. In this review, we outline biomaterials with immunomodulatory functions discovered in recent years and their applications in disease treatment. These biomaterials can treat inflammation, tumors, or autoimmune diseases by regulating immune cell function, exerting enzyme-like activity, neutralizing cytokines, etc. The prospects and challenges of biomaterial-based modulation of immunotherapy are also discussed.
Collapse
Affiliation(s)
- Huaxing Dai
- Institute of Functional Nano & Soft Materials (FUNSOM)Jiangsu Key Laboratory for Carbon‐Based Functional Materials & DevicesSoochow University199 Ren'ai RoadSuzhouJiangsuChina
| | - Qin Fan
- Key Laboratory for Organic Electronics & Information Displays (KLOEID)Jiangsu Key Laboratory for BiosensorsInstitute of Advanced Materials (IAM) and School of Materials Science and EngineeringNanjing University of Posts & TelecommunicationsNanjingChina
| | - Chao Wang
- Institute of Functional Nano & Soft Materials (FUNSOM)Jiangsu Key Laboratory for Carbon‐Based Functional Materials & DevicesSoochow University199 Ren'ai RoadSuzhouJiangsuChina
| |
Collapse
|
20
|
Muacevic A, Adler JR. Classic and Current Opinions in Human Organ and Tissue Transplantation. Cureus 2022; 14:e30982. [PMID: 36337306 PMCID: PMC9624478 DOI: 10.7759/cureus.30982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2022] [Indexed: 11/30/2022] Open
Abstract
Graft tolerance is a pathophysiological condition heavily reliant on the dynamic interaction of the innate and adaptive immune systems. Genetic polymorphism determines immune responses to tissue/organ transplantation, and intricate humoral and cell-mediated mechanisms control these responses. In transplantation, the clinician's goal is to achieve a delicate equilibrium between the allogeneic immune response, undesired effects of the immunosuppressive drugs, and the existing morbidities that are potentially life-threatening. Transplant immunopathology involves sensitization, effector, and apoptosis phases which recruit and engages immunological cells like natural killer cells, lymphocytes, neutrophils, and monocytes. Similarly, these cells are involved in the transfer of normal or genetically engineered T cells. Advances in tissue transplantation would involve a profound knowledge of the molecular mechanisms that underpin the respective immunopathology involved and the design of precision medicines that are safe and effective.
Collapse
|
21
|
Nozari P, Mokhtari P, Nemati M, Zainodini N, Taghipour Z, Asadi F, Ayoobi F, Jafarzadeh A. Investigation of the effect of IFN-γ/TNF-α-treated mesenchymal stem cells on Th9- and Treg cell-related parameters in a mouse model of ovalbumin-induced allergic asthma. Immunopharmacol Immunotoxicol 2022; 44:773-785. [PMID: 35620857 DOI: 10.1080/08923973.2022.2082977] [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] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Th9- and regulatory T (Treg) cells exert pro- and anti-allergic activity, respectively. Mesenchymal stem cell (MSC)-related immunomodulatory impacts can be enhanced by inflammatory cytokines. Here, the modulatory effects of IFN-γ/TNF-α-induced MSCs on Th9- and Treg cell-related parameters were investigated using an asthma model. METHODS Allergic asthma was induced in BALB/c mice using sensitized and challenging with ovalbumin (OVA). The asthmatic groups were treated intraperitoneally with PBS, MSCs, IFN-γ-induced MSCs, TNF-α-induced MSCs and "IFN-γ + TNF-α"-induced MSCs before the challenge phase. The mice were sacrificed 24 hours after challenge. The serum IL-9 and IL-35 levels, as well as gene expression of IL-9, PU.1, IL-35-EBI3 and FOXP3 in the lung tissues were assessed using ELISA and real time-PCR, respectively. RESULTS The differences of Th9 and Treg-related parameters were not significant between untreated asthmatic mice and those treated with non-induced MSCs. In comparison with untreated asthmatic group, treatment with IFN-γ-induced MSCs significantly reduced serum IL-9 levels, reduced lung expression of IL-9 and PU.1, while increasing serum IL-35 levels as well as lung expression of FOXP3; treatment with TNF-α-induced MSCs significantly reduced serum IL-9 levels as well as lung expression of IL-9, and treatment with "IFN-γ + TNF-α"-induced MSCs significantly modulated all investigated Th9 and Treg-related parameters. In comparison to mice treated with non-induced MSCs, serum IL-9 levels were remarkably decreased in mice treated with IFN-γ-induced and "IFN-γ + TNF-α"-induced MSCs. CONCLUSIONS IFN-γ-and "IFN-γ + TNF-α" treated MSCs exerted almost comparable impacts, but were more efficient than TNF-α-exposed MSCs. Thus, IFN-γ alone can be sufficient to promote immunomodulatory effects of MSCs.
Collapse
Affiliation(s)
- Parvin Nozari
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Pejman Mokhtari
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Nemati
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.,Department of Haematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Nahid Zainodini
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Zahra Taghipour
- Department of Histology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Fatemeh Asadi
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Fatemeh Ayoobi
- Non-Communicable Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.,Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
22
|
Maestre-Batlle D, Nygaard UC, Huff RD, Alexis NE, Tebbutt SJ, Turvey SE, Carlsten C, Kocbach Bølling A. Dibutyl phthalate exposure alters T-cell subsets in blood from allergen-sensitized volunteers. INDOOR AIR 2022; 32:e13026. [PMID: 35481934 DOI: 10.1111/ina.13026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/04/2022] [Accepted: 03/24/2022] [Indexed: 06/14/2023]
Abstract
Phthalates are ubiquitous environmental contaminants associated with allergic disease in epidemiological and animal studies. This investigation aims to support these associations by interrogating systemic immune effects in allergen-sensitized volunteers after controlled indoor air exposure to a known concentration of dibutyl phthalate (DBP). The phthalate-allergen immune response (PAIR) study enrolled 16 allergen-sensitized participants to a double-blinded, randomized, crossover exposure to two conditions (DBP or control air for 3 hr), each followed immediately by inhaled allergen challenge. Peripheral blood immune cell composition and activation along with inflammatory mediators were measured before and after exposure. DBP exposure prior to the inhaled allergen challenge increased the percentage of CD4+ T helper cells and decreased the percentage of regulatory T cells (3 hr and 20 hr post-exposure), while only modest overall effects were observed for inflammatory mediators. The cells and mediators affected by the phthalate exposure were generally not overlapping with the endpoints affected by allergen inhalation alone. Thus, in distinction to our previously published effects on lung function, DBP appears to alter endpoints in peripheral blood that are not necessarily enhanced by allergen alone. Further studies are needed to clarify the role of phthalate-induced systemic effects in disease pathogenesis.
Collapse
Affiliation(s)
- Danay Maestre-Batlle
- Department of Medicine, Air Pollution Exposure Lab and Legacy for Airway Health, University of British Columbia and Vancouver Coastal Health, Vancouver, Canada
| | - Unni C Nygaard
- Department of Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Ryan D Huff
- Department of Medicine, Air Pollution Exposure Lab and Legacy for Airway Health, University of British Columbia and Vancouver Coastal Health, Vancouver, Canada
| | - Neil E Alexis
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Scott J Tebbutt
- Department of Medicine, PROOF Centre of Excellence, & Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada
| | - Stuart E Turvey
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, Canada
| | - Christopher Carlsten
- Department of Medicine, Air Pollution Exposure Lab and Legacy for Airway Health, University of British Columbia and Vancouver Coastal Health, Vancouver, Canada
| | | |
Collapse
|
23
|
Ahmad HI, Jabbar A, Mushtaq N, Javed Z, Hayyat MU, Bashir J, Naseeb I, Abideen ZU, Ahmad N, Chen J. Immune Tolerance vs. Immune Resistance: The Interaction Between Host and Pathogens in Infectious Diseases. Front Vet Sci 2022; 9:827407. [PMID: 35425833 PMCID: PMC9001959 DOI: 10.3389/fvets.2022.827407] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/17/2022] [Indexed: 11/13/2022] Open
Abstract
The immune system is most likely developed to reduce the harmful impact of infections on the host homeostasis. This defense approach is based on the coordinated activity of innate and adaptive immune system components, which detect and target infections for containment, killing, or expulsion by the body's defense mechanisms. These immunological processes are responsible for decreasing the pathogen burden of an infected host to maintain homeostasis that is considered to be infection resistance. Immune-driven resistance to infection is connected with a second, and probably more important, defensive mechanism: it helps to minimize the amount of dysfunction imposed on host parenchymal tissues during infection without having a direct adverse effect on pathogens. Disease tolerance is a defensive approach that relies on tissue damage control systems to prevent infections from causing harm to the host. It also uncouples immune-driven resistance mechanisms from immunopathology and disease, allowing the body to fight infection more effectively. This review discussed the cellular and molecular processes that build disease tolerance to infection and the implications of innate immunity on those systems. In addition, we discuss how symbiotic relationships with microbes and their control by particular components of innate and adaptive immunity alter disease tolerance to infection.
Collapse
Affiliation(s)
- Hafiz Ishfaq Ahmad
- Department of Animal Breeding and Genetics, University of Veterinary and Animal Sciences, Lahore, Pakistan
- *Correspondence: Hafiz Ishfaq Ahmad
| | - Abdul Jabbar
- Department of Clinical Medicine, Faculty of Veterinary Science, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Nadia Mushtaq
- Department of Biological Sciences, Faculty of Fisheries and Wildlife, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Zainab Javed
- Institute of Pharmaceutical Sciences, Faculty of Biosciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Muhammad Umar Hayyat
- Institute of Pharmaceutical Sciences, Faculty of Biosciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Javaria Bashir
- Department of Medical Sciences, Sharif Medical and Dental Hospital, Lahore, Pakistan
| | - Iqra Naseeb
- Institute of Microbiology, Faculty of Veterinary Science, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Zain Ul Abideen
- Department of Zoology, Ghazi University, Dera Ghazi Khan, Pakistan
| | - Nisar Ahmad
- Department of Livestock Management, University of Veterinary and Animal Sciences, Pattoki, Pakistan
| | - Jinping Chen
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, China
- Jinping Chen
| |
Collapse
|
24
|
CAR Treg: A new approach in the treatment of autoimmune diseases. Int Immunopharmacol 2021; 102:108409. [PMID: 34863655 DOI: 10.1016/j.intimp.2021.108409] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 11/07/2021] [Accepted: 11/23/2021] [Indexed: 12/22/2022]
Abstract
Regulatory T cells (Tregs) have the role of regulating self-tolerance, and suppressing immune responses. Defects in Treg function and number can lead to in loss of tolerance or autoimmune disease. To treat or control autoimmune diseases, one of the options is to develop immune tolerance for Tregs cell therapy, which includes promotion and activation. Recently, cell-based treatment as a promising approach to increase cells function and number has been developed. Cell therapy by chimeric T antigen receptor (CAR-T) cells has shown significant efficacy in the treatment of leukemia, which has led researchers to use CAR-T cells in other diseases like autoimmune diseases. Here, we describe the existing treatments for autoimmune diseases and the available treatments based on Treg, their benefits and restrictions for implementation in clinical trials. We also discussed potential solutions to overcome these limitations. It seems novel designs of CARs to be new hope for autoimmune diseases and expected to be a potential cure option in a wide array of disease in the future. Therefore, it is very important to address this issue and increase information about it.
Collapse
|
25
|
Chulanetra M, Chaicumpa W. Revisiting the Mechanisms of Immune Evasion Employed by Human Parasites. Front Cell Infect Microbiol 2021; 11:702125. [PMID: 34395313 PMCID: PMC8358743 DOI: 10.3389/fcimb.2021.702125] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/25/2021] [Indexed: 12/14/2022] Open
Abstract
For the establishment of a successful infection, i.e., long-term parasitism and a complete life cycle, parasites use various diverse mechanisms and factors, which they may be inherently bestowed with, or may acquire from the natural vector biting the host at the infection prelude, or may take over from the infecting host, to outmaneuver, evade, overcome, and/or suppress the host immunity, both innately and adaptively. This narrative review summarizes the up-to-date strategies exploited by a number of representative human parasites (protozoa and helminths) to counteract the target host immune defense. The revisited information should be useful for designing diagnostics and therapeutics as well as vaccines against the respective parasitic infections.
Collapse
Affiliation(s)
- Monrat Chulanetra
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Wanpen Chaicumpa
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
26
|
An overview of human pericardial space and pericardial fluid. Cardiovasc Pathol 2021; 53:107346. [PMID: 34023529 DOI: 10.1016/j.carpath.2021.107346] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 12/24/2022] Open
Abstract
The pericardium is a double-layered fibro-serous sac that envelops the majority of the surface of the heart as well as the great vessels. Pericardial fluid is also contained within the pericardial space. Together, the pericardium and pericardial fluid contribute to a homeostatic environment that facilitates normal cardiac function. Different diseases and procedural interventions may disrupt this homeostatic space causing an imbalance in the composition of immune mediators or by mechanical stress. Inflammatory cells, cytokines, and chemokines are present in the pericardial space. How these specific mediators contribute to different diseases is the subject of debate and research. With the advent of highly specialized assays that can identify and quantify various mediators we can potentially establish specific and sensitive biomarkers that can be used to differentiate pathologies, and aid clinicians in improving clinical outcomes for patients.
Collapse
|
27
|
Treg sensitivity to FasL and relative IL-2 deprivation drive idiopathic aplastic anemia immune dysfunction. Blood 2021; 136:885-897. [PMID: 32294156 DOI: 10.1182/blood.2019001347] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 04/07/2020] [Indexed: 12/12/2022] Open
Abstract
Idiopathic aplastic anemia (AA) has 2 key characteristics: an autoimmune response against hematopoietic stem/progenitor cells and regulatory T-cells (Tregs) deficiency. We have previously demonstrated reduction in a specific subpopulation of Treg in AA, which predicts response to immunosuppression. The aims of the present study were to define mechanisms of Treg subpopulation imbalance and identify potential for therapeutic intervention. We have identified 2 mechanisms that lead to skewed Treg composition in AA: first, FasL-mediated apoptosis on ligand interaction; and, second, relative interleukin-2 (IL-2) deprivation. We have shown that IL-2 augmentation can overcome these mechanisms. Interestingly, when high concentrations of IL-2 were used for in vitro Treg expansion cultures, AA Tregs were able to expand. The expanded populations expressed a high level of p-BCL-2, which makes them resistant to apoptosis. Using a xenograft mouse model, the function and stability of expanded AA Tregs were tested. We have shown that these Tregs were able to suppress the macroscopic clinical features and tissue manifestations of T-cell-mediated graft-versus-host disease. These Tregs maintained their suppressive properties as well as their phenotype in a highly inflammatory environment. Our findings provide an insight into the mechanisms of Treg reduction in AA. We have identified novel targets with potential for therapeutic interventions. Supplementation of ex vivo expansion cultures of Tregs with high concentrations of IL-2 or delivery of IL-2 directly to patients could improve clinical outcomes in addition to standard immunosuppressive therapy.
Collapse
|
28
|
Cai Y, Leng S, Ma Y, Xu T, Chang D, Ju S. Dynamic change of MMP-9 in diabetic stroke visualized by optical imaging and treated with CD28 superagonist. Biomater Sci 2021; 9:2562-2570. [DOI: 10.1039/d0bm02014a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
By utilizing NIRF imaging, diabetic stroke mice were visualized with a trend of higher levels of MMP-9 than wild-type mice. CD28 SA down-regulated the expression of MMP-9 and represents a potential treatment to diabetic stroke.
Collapse
Affiliation(s)
- Yu Cai
- Jiangsu Key Laboratory of Molecular and Functional Imaging
- Department of Radiology
- Zhongda Hospital
- Medical School of Southeast University
- Nanjing 210009
| | - Shou Leng
- Jiangsu Key Laboratory of Molecular and Functional Imaging
- Department of Radiology
- Zhongda Hospital
- Medical School of Southeast University
- Nanjing 210009
| | - Yuanyuan Ma
- Jiangsu Key Laboratory of Molecular and Functional Imaging
- Department of Radiology
- Zhongda Hospital
- Medical School of Southeast University
- Nanjing 210009
| | - Tingting Xu
- Jiangsu Key Laboratory of Molecular and Functional Imaging
- Department of Radiology
- Zhongda Hospital
- Medical School of Southeast University
- Nanjing 210009
| | - Di Chang
- Jiangsu Key Laboratory of Molecular and Functional Imaging
- Department of Radiology
- Zhongda Hospital
- Medical School of Southeast University
- Nanjing 210009
| | - Shenghong Ju
- Jiangsu Key Laboratory of Molecular and Functional Imaging
- Department of Radiology
- Zhongda Hospital
- Medical School of Southeast University
- Nanjing 210009
| |
Collapse
|
29
|
Bilbao A, Pérez-Garay R, Rius I, Irurzun A, Terrén I, Orrantia A, Astarloa-Pando G, Borrego F, Zenarruzabeitia O. Increased Frequency of CTLA-4 and PD-1 Expressing Regulatory T Cells and Basophils With an Activating Profile in Infants With Moderate-to-Severe Atopic Dermatitis Hypersensitized to Food Allergens. Front Pediatr 2021; 9:734645. [PMID: 34912758 PMCID: PMC8667617 DOI: 10.3389/fped.2021.734645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 11/08/2021] [Indexed: 11/20/2022] Open
Abstract
Background: Infants with severe atopic dermatitis (AD) may be sensitized to foods that have not been introduced into their diet, posing a risk for developing an immediate hypersensitivity reaction on the first exposure to the food to which they are sensitized. The aim of this work was to perform an analysis of the sensitization profile in infants with moderate-to-severe AD and to identify cellular and molecular markers for food allergy (FA). Methods: Blood samples from healthy donors and children with moderate-to-severe AD were studied. Specific IgE to several allergens were determined using ImmunoCAP FEIA system and ISAC technology. Furthermore, using flow cytometry-based studies, basophils and regulatory T (Treg) cells were phenotypically characterized. Results: 90% of children with AD were sensitized to food antigens before introducing them into the diet, and 100% developed FA. Phenotypic analysis showed a significantly higher percentage of CTLA-4 and PD-1 expressing Treg cells in AD patients than in healthy controls. Basophils from patients exhibited a marked reduction in the expression of CD300a, higher expression of FcεRI and CXCR4, and to some extent higher expression of CD63 and CD300c. Conclusions: Infants with moderate-to-severe AD are at high risk of being sensitized to food allergens. Therefore, to avoid allergic reactions, broad-spectrum sensitization studies are necessary before introducing complementary diet. Increased expression of CTLA-4 and PD-1 suggests greater suppressive potential of Treg cells in infants with AD than healthy controls. Furthermore, our results suggest a role for CD300 molecules on circulating basophils as possible biomarkers for FA susceptibility.
Collapse
Affiliation(s)
- Agurtzane Bilbao
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.,Pediatrics Service, Cruces University Hospital, Barakaldo, Spain
| | - Raquel Pérez-Garay
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.,Clinical Analysis Service, Cruces University Hospital, Barakaldo, Spain
| | - Idoia Rius
- Pediatrics Service, Cruces University Hospital, Barakaldo, Spain
| | - Alex Irurzun
- Pediatrics Service, Cruces University Hospital, Barakaldo, Spain
| | - Iñigo Terrén
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Ane Orrantia
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | | | - Francisco Borrego
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Olatz Zenarruzabeitia
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| |
Collapse
|
30
|
Khan MA. Regulatory T cells mediated immunomodulation during asthma: a therapeutic standpoint. J Transl Med 2020; 18:456. [PMID: 33267824 PMCID: PMC7713035 DOI: 10.1186/s12967-020-02632-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 08/26/2020] [Indexed: 12/24/2022] Open
Abstract
Asthma is an inflammatory disease of the lung airway network, which is initiated and perpetuated by allergen-specific CD4+ T cells, IgE antibodies, and a massive release of Th2 cytokines. The most common clinical manifestations of asthma progression include airway inflammation, pathological airway tissue and microvascular remodeling, which leads to airway hyperresponsiveness (AHR), and reversible airway obstruction. In addition to inflammatory cells, a tiny population of Regulatory T cells (Tregs) control immune homeostasis, suppress allergic responses, and participate in the resolution of inflammation-associated tissue injuries. Preclinical and clinical studies have demonstrated a tremendous therapeutic potential of Tregs in allergic airway disease, which plays a crucial role in immunosuppression, and rejuvenation of inflamed airways. These findings supported to harness the immunotherapeutic potential of Tregs to suppress airway inflammation and airway microvascular reestablishment during the progression of the asthma disease. This review addresses the therapeutic impact of Tregs and how Treg mediated immunomodulation plays a vital role in subduing the development of airway inflammation, and associated airway remodeling during the onset of disease.
Collapse
Affiliation(s)
- Mohammad Afzal Khan
- Organ Transplant Research Section, Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.
| |
Collapse
|
31
|
Jafarzadeh A, Nemati M, Jafarzadeh S, Chauhan P, Saha B. The immunomodulatory potentials of interleukin-27 in airway allergies. Scand J Immunol 2020; 93:e12959. [PMID: 32797730 DOI: 10.1111/sji.12959] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/31/2020] [Accepted: 08/10/2020] [Indexed: 12/15/2022]
Abstract
Allergic airway disorders such as asthma and allergic rhinitis are mainly caused by inhaled allergen-induced improper activation and responses of immune and non-immune cells. One important response is the production of IL-27 by macrophages and dendritic cells (DCs) during the early stage of airway allergies. IL-27 exerts powerful modulatory influences on the cells of innate immunity [eg neutrophils, eosinophils, mast cells, monocytes, macrophages, dendritic cells (DCs), innate lymphoid cells (ILCs), natural killer (NK) cells and NKT cells)] and adaptive immunity (eg Th1, Th2, Th9, Th17, regulatory T, CD8+ cytotoxic T and B cells). The IL-27-mediated signalling pathways may be modulated to attenuate asthma and allergic rhinitis. In this review, a comprehensive discussion concerning the roles carried out by IL-27 in asthma and allergic rhinitis was provided, while evidences are presented favouring the use of IL-27 in the treatment of airway allergies.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, Kerman University of Medical Sciences, Kerman, Iran.,Department of Immunology, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Maryam Nemati
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.,Department of Hematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Sara Jafarzadeh
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Bhaskar Saha
- National Centre for Cell Science, Pune, India.,Trident Academy of Creative Technology, Bhubaneswar, India
| |
Collapse
|
32
|
Wang W, Lyu SC, Ji X, Gupta S, Manohar M, Dhondalay GKR, Chinthrajah S, Andorf S, Boyd SD, Tibshirani R, Galli SJ, Nadeau KC, Maecker HT. Transcriptional changes in peanut-specific CD4+ T cells over the course of oral immunotherapy. Clin Immunol 2020; 219:108568. [PMID: 32783912 DOI: 10.1016/j.clim.2020.108568] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 01/12/2023]
Abstract
Oral immunotherapy (OIT) can successfully desensitize allergic individuals to offending foods such as peanut. Our recent clinical trial (NCT02103270) of peanut OIT allowed us to monitor peanut-specific CD4+ T cells, using MHC-peptide Dextramers, over the course of OIT. We used a single-cell targeted RNAseq assay to analyze these cells at 0, 12, 24, 52, and 104 weeks of OIT. We found a transient increase in TGFβ-producing cells at 52 weeks in those with successful desensitization, which lasted until 117 weeks. We also performed clustering and identified 5 major clusters of Dextramer+ cells, which we tracked over time. One of these clusters appeared to be anergic, while another was consistent with recently described TFH13 cells. The other 3 clusters appeared to be Th2 cells by their coordinated production of IL-4 and IL-13, but they varied in their expression of STAT signaling proteins and other markers. A cluster with high expression of STAT family members also showed a possible transient increase at week 24 in those with successful desensitization. Single cell TCRαβ repertoire sequences were too diverse to track clones over time. Together with increased TGFβ production, these changes may be mechanistic predictors of successful OIT that should be further investigated.
Collapse
Affiliation(s)
- Weiqi Wang
- Institute for Immunity, Transplantation, Infection, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Shu-Chen Lyu
- Sean Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Xuhuai Ji
- Institute for Immunity, Transplantation, Infection, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Sheena Gupta
- Institute for Immunity, Transplantation, Infection, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Monali Manohar
- Sean Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Gopal K R Dhondalay
- Sean Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Sharon Chinthrajah
- Sean Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Sandra Andorf
- Sean Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Scott D Boyd
- Departments of Pathology and of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Robert Tibshirani
- Department of Biomedical Data Science, Department of Statistics, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Stephen J Galli
- Sean Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA 94305, United States of America; Departments of Pathology and of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Kari C Nadeau
- Sean Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Holden T Maecker
- Institute for Immunity, Transplantation, Infection, Stanford University School of Medicine, Stanford, CA 94305, United States of America.
| |
Collapse
|
33
|
Lou H, Huang Y, Ouyang Y, Zhang Y, Xi L, Chu X, Wang Y, Wang C, Zhang L. Artemisia annua-sublingual immunotherapy for seasonal allergic rhinitis: A randomized controlled trial. Allergy 2020; 75:2026-2036. [PMID: 32030780 DOI: 10.1111/all.14218] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 11/17/2019] [Accepted: 12/08/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Artemisia annua is an important autumnal pollen allergen for seasonal allergic rhinitis (SAR) in northern China. To date, no study has investigated allergen immunotherapy with A annua. We aimed to investigate the efficacy and mechanisms underlying A annua-sublingual immunotherapy (SLIT). METHODS This was a randomized, double-blind, placebo-controlled phase III clinical trial involving 71 SAR patients, randomized to SLIT with A annua extract (n = 47) or placebo (n = 24) for 32 weeks. Total nasal symptom score (TNSS; primary clinical end point) was evaluated at baseline (peak pollen phase (PPP) in the previous year), initiation of A annua-SLIT, 1st PPP during SLIT, end of SLIT and 2nd PPP during follow-up. Blood samples and nasal secretions were collected at beginning and after SLIT for assessment of T cells and inflammatory mediators. Safety was assessed according to adverse events (AEs) reported. RESULTS Artemisia annua-SLIT significantly reduced TNSS to a greater level from baseline (from 9.45 ± 1.68 to 6.16 ± 2.27) than placebo (from 9.29 ± 2.09 to 9.05 ± 2.40) at the 1st PPP (P < .001) and sustained the improvement in symptoms throughout to the 2nd PPP. Preseasonal A annua-SLIT for 16 weeks significantly decreased Th2 cells, increased nTreg and Tr1 cells in blood; and increased cystatin 1 (CST1) in nasal secretion after 16 and 32 weeks compared with pretreatment. Overall, 17/47 patients experienced mild local AEs and 2 patients mild systemic AEs, after A annua-SLIT. CONCLUSION Artemisia annua-SLIT is an efficacious and safe treatment in patients with A annua SAR.
Collapse
Affiliation(s)
- Hongfei Lou
- Department of Otolaryngology Head and Neck Surgery Beijing TongRen Hospital Capital Medical University Beijing China
- Beijing Key Laboratory of Nasal Disease Beijing Institute of Otolaryngology Beijing China
| | - Yanran Huang
- Department of Otolaryngology Head and Neck Surgery Beijing TongRen Hospital Capital Medical University Beijing China
- Beijing Key Laboratory of Nasal Disease Beijing Institute of Otolaryngology Beijing China
| | - Yuhui Ouyang
- Beijing Key Laboratory of Nasal Disease Beijing Institute of Otolaryngology Beijing China
- Department of Allergy Beijing TongRen Hospital Capital Medical University Beijing China
| | - Yuan Zhang
- Beijing Key Laboratory of Nasal Disease Beijing Institute of Otolaryngology Beijing China
- Department of Allergy Beijing TongRen Hospital Capital Medical University Beijing China
| | - Lin Xi
- Beijing Key Laboratory of Nasal Disease Beijing Institute of Otolaryngology Beijing China
- Department of Allergy Beijing TongRen Hospital Capital Medical University Beijing China
| | - Xiaohan Chu
- Department of Otolaryngology Head and Neck Surgery Beijing TongRen Hospital Capital Medical University Beijing China
- Beijing Key Laboratory of Nasal Disease Beijing Institute of Otolaryngology Beijing China
| | - Yang Wang
- Beijing Key Laboratory of Nasal Disease Beijing Institute of Otolaryngology Beijing China
| | - Chengshuo Wang
- Department of Otolaryngology Head and Neck Surgery Beijing TongRen Hospital Capital Medical University Beijing China
- Beijing Key Laboratory of Nasal Disease Beijing Institute of Otolaryngology Beijing China
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery Beijing TongRen Hospital Capital Medical University Beijing China
- Beijing Key Laboratory of Nasal Disease Beijing Institute of Otolaryngology Beijing China
- Department of Allergy Beijing TongRen Hospital Capital Medical University Beijing China
| |
Collapse
|
34
|
Singampalli KL, Balaji S, Wang X, Parikh UM, Kaul A, Gilley J, Birla RK, Bollyky PL, Keswani SG. The Role of an IL-10/Hyaluronan Axis in Dermal Wound Healing. Front Cell Dev Biol 2020; 8:636. [PMID: 32850791 PMCID: PMC7396613 DOI: 10.3389/fcell.2020.00636] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/24/2020] [Indexed: 12/12/2022] Open
Abstract
Scar formation is the typical endpoint of postnatal dermal wound healing, which affects more than 100 million individuals annually. Not only do scars cause a functional burden by reducing the biomechanical strength of skin at the site of injury, but they also significantly increase healthcare costs and impose psychosocial challenges. Though the mechanisms that dictate how dermal wounds heal are still not completely understood, they are regulated by extracellular matrix (ECM) remodeling, neovascularization, and inflammatory responses. The cytokine interleukin (IL)-10 has emerged as a key mediator of the pro- to anti-inflammatory transition that counters collagen deposition in scarring. In parallel, the high molecular weight (HMW) glycosaminoglycan hyaluronan (HA) is present in the ECM and acts in concert with IL-10 to block pro-inflammatory signals and attenuate fibrotic responses. Notably, high concentrations of both IL-10 and HMW HA are produced in early gestational fetal skin, which heals scarlessly. Since fibroblasts are responsible for collagen deposition, it is critical to determine how the concerted actions of IL-10 and HA drive their function to potentially control fibrogenesis. Beyond their independent actions, an auto-regulatory IL-10/HA axis may exist to modulate the magnitude of CD4+ effector T lymphocyte activation and enhance T regulatory cell function in order to reduce scarring. This review underscores the pathophysiological impact of the IL-10/HA axis as a multifaceted molecular mechanism to direct primary cell responders and regulators toward either regenerative dermal tissue repair or scarring.
Collapse
Affiliation(s)
- Kavya L Singampalli
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States.,Department of Bioengineering, Rice University, Houston, TX, United States.,Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, United States
| | - Swathi Balaji
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States
| | - Xinyi Wang
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States
| | - Umang M Parikh
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States
| | - Aditya Kaul
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States
| | - Jamie Gilley
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States.,Division of Neonatology, Department of Pediatrics, Texas Children's Hospital, Houston, TX, United States
| | | | - Paul L Bollyky
- Division of Infectious Diseases, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Sundeep G Keswani
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States
| |
Collapse
|
35
|
Cai Y, Xu TT, Lu CQ, Ma YY, Chang D, Zhang Y, Gu XC, Ju S. Endogenous Regulatory T Cells Promote M2 Macrophage Phenotype in Diabetic Stroke as Visualized by Optical Imaging. Transl Stroke Res 2020; 12:136-146. [PMID: 32240524 DOI: 10.1007/s12975-020-00808-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 03/10/2020] [Accepted: 03/12/2020] [Indexed: 01/04/2023]
Abstract
Regulatory T cells (Tregs) play an immunosuppressive role in various diseases, yet their function remains controversial in stroke and obscure in diabetic stroke. In the present study, Tregs were found downregulated in the peripheral blood of type 2 diabetes mellitus (T2DM) stroke models and patients compared with controls. In ischemic stroke mice (both T2DM and wild type), endogenous Tregs boosted by CD28SA increased CD206+ M2 macrophage/microglia cells, decreased infarct volumes, and improved neurological recovery. Our results demonstrated the potential of boosting Tregs for treating T2DM stroke. Furthermore, we utilized an optical imaging probe (IRD-αCD206) to target M2 macrophage/microglia cells and demonstrated its effect in visualizing M2 macrophage/microglia cells in vivo in ischemic brain tissue.
Collapse
Affiliation(s)
- Yu Cai
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Ding Jia Qiao Road, Nanjing, 210009, China
| | - Ting-Ting Xu
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Ding Jia Qiao Road, Nanjing, 210009, China
| | - Chun-Qiang Lu
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Ding Jia Qiao Road, Nanjing, 210009, China
| | - Yuan-Yuan Ma
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Ding Jia Qiao Road, Nanjing, 210009, China
| | - Di Chang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Ding Jia Qiao Road, Nanjing, 210009, China
| | - Yi Zhang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Ding Jia Qiao Road, Nanjing, 210009, China
| | - Xiao-Chun Gu
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Ding Jia Qiao Road, Nanjing, 210009, China
| | - Shenghong Ju
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Ding Jia Qiao Road, Nanjing, 210009, China.
| |
Collapse
|
36
|
Chen Z, Wang Y, Ding X, Zhang M, He M, Zhao Y, Hu S, Zhao F, Wang J, Xie B, Shi B. The proportion of peripheral blood Tregs among the CD4+ T cells of autoimmune thyroid disease patients: a meta-analysis. Endocr J 2020; 67:317-326. [PMID: 31827051 DOI: 10.1507/endocrj.ej19-0307] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Autoimmune thyroid disease (AITD) is characterized by a loss of self-tolerance to thyroid antigen. Tregs, whose proportions are controversial among CD4+ T cell from AITD patients (AITDs), are crucial in immune tolerance. Considering that drugs might affect Treg levels, we assumed that the differences originated from different treatment statuses. Thus, we performed a meta-analysis to explore proportions of Tregs in untreated and treated AITDs. PubMed, Embase and ISI Web of Knowledge were searched for relevant studies. Review Manager 5.3 and Stata 14.0 were used to conduct the meta-analysis. Subgroup analysis based on different diseases and cell surface markers was performed. Egger linear regression analysis was used to assess publication bias. Approximately 1,100 AITDs and healthy controls (HCs) from fourteen studies were included. Proportions of Tregs among CD4+ T cells of untreated AITDs were significantly lower than those in HCs (p = 0.002), but were not in treated patients (p = 0.40). Subgroup analysis revealed lower proportions of Tregs in untreated Graves' disease patients (GDs) (p = 0.001) but did not show obvious differences in untreated Hashimoto's thyroiditis patients (HTs) (p = 0.62). Furthermore, proportions of circulating FoxP3+ Tregs were reduced in untreated GDs (p < 0.00001) and HTs (p = 0.04). No publication bias was found. In this first meta-analysis exploring proportions of circulating Tregs among CD4+ T cells of AITDs with different treatment statuses, we found that Tregs potentially contribute to the pathogenesis of AITD but function differently in GD and HT. Remarkably, FoxP3+ Tregs, which were decreased in both diseases, might be promising targets for novel therapies.
Collapse
Affiliation(s)
- Ziyi Chen
- Department of Endocrinology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yue Wang
- Department of Endocrinology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xi Ding
- Department of Endocrinology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Meng Zhang
- Department of Endocrinology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Mingqian He
- Department of Endocrinology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yang Zhao
- Department of Endocrinology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Shiqian Hu
- Department of Endocrinology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Fengyi Zhao
- Department of Endocrinology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jingya Wang
- Department of Endocrinology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Baosong Xie
- Department of Endocrinology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Bingyin Shi
- Department of Endocrinology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
37
|
Erkan K, Bozkurt MK, Artaç H, Özdemir H, Ünlü A, Korucu EN, Elsürer Ç. The role of regulatory T cells in allergic rhinitis and their correlation with IL-10, IL-17 and neopterin levels in serum and nasal lavage fluid. Eur Arch Otorhinolaryngol 2020; 277:1109-1114. [PMID: 31993765 DOI: 10.1007/s00405-020-05811-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/18/2020] [Indexed: 11/27/2022]
Abstract
PURPOSE Allergic rhinitis (AR), is an IgE-mediated inflammation of the nose. Regulatory T cells (Tregs) and inflammatory cytokines have been shown to play a critical role in allergic airway inflammation. The aim of the study was to compare the levels of blood T lymphocyte subsets and IL-10, IL-17 and neopterin concentrations in serum and nasal lavage of patients with AR compared to healthy subjects. METHODS The study included 38 subjects with moderate-severe AR and 36 sex- and age-matched controls. Peripheral blood CD3+, CD3+CD4+ and CD4+CD25+Foxp3 percentages were evaluated using flow cytometry. Levels of IL-10, IL-17 and neopterin were measured both in serum and nasal lavage fluid with ELISA and HPLC, respectively. RESULTS No difference was found in the percentages of T lymphocyte subsets between the two groups (p > 0.05). Serum IL-10 levels were similar (p > 0.05), whereas nasal IL-10 was lower in AR subjects compared to control group (2.22 ± 0.91 and 3.12 ± 1.45 pg/ml, respectively) (p < 0.05). Mean serum and nasal IL-17 were higher in AR (107.7 ± 79.61 and 527.36 ± 738.7 pg/ml) than the control group (76.29 ± 28.94 and 328.9 ± 430.8 pg/ml) (p < 0.05 and p > 0.05). There were no significant differences in serum and nasal neopterin levels (p > 0.05). CONCLUSIONS Although there were no differences in the distribution of lymphocyte subsets between the AR and control groups, the finding of higher levels of serum and nasal IL-17 and lower levels of nasal IL-10 support the cytokine imbalance in the pathogenesis of AR.
Collapse
Affiliation(s)
- Kadriye Erkan
- Otolaryngology Department, Konya Egitim ve Arastirma Hastanesi, Konya, Turkey
| | - Mete K Bozkurt
- Otolaryngology Department, Selcuk University School of Medicine, Konya, Turkey.
| | - Hasibe Artaç
- Selcuk University School of Medicine, Pediatric Allergy and Immunology Dept, Konya, Turkey
| | - Hülya Özdemir
- Selcuk University School of Medicine, Pediatric Allergy and Immunology Dept, Konya, Turkey
| | - Ali Ünlü
- Selcuk University School of Medicine, Biochemistry Dept, Konya, Turkey
| | - Emine N Korucu
- Necmettin Erbakan University, Molecular Biology and Genetics, Konya, Turkey
| | - Çağdaş Elsürer
- Otolaryngology Department, Selcuk University School of Medicine, Konya, Turkey
| |
Collapse
|
38
|
Yadava K, Medina CO, Ishak H, Gurevich I, Kuipers H, Shamskhou EA, Koliesnik IO, Moon JJ, Weaver C, Nadeau KC, Bollyky PL. Natural Tr1-like cells do not confer long-term tolerogenic memory. eLife 2019; 8:e44821. [PMID: 31603425 PMCID: PMC6788856 DOI: 10.7554/elife.44821] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022] Open
Abstract
IL-10-producing Tr1 cells promote tolerance but their contributions to tolerogenic memory are unclear. Using 10BiT mice that carry a Foxp3-eGFP reporter and stably express CD90.1 following IL-10 production, we characterized the spatiotemporal dynamics of Tr1 cells in a house dust mite model of allergic airway inflammation. CD90.1+Foxp3-IL-10+ Tr1 cells arise from memory cells and rejoin the tissue-resident memory T-cell pool after cessation of IL-10 production. Persistent antigenic stimulation is necessary to sustain IL-10 production and Irf1 and Batf expression distinguishes CD90.1+Foxp3-IL-10+ Tr1 cells from CD90.1+Foxp3-IL-10- 'former' Tr1. Depletion of Tr1-like cells after primary sensitization exacerbates allergic airway inflammation. However, neither transfer nor depletion of former Tr1 cells influences either Tr1 numbers or the inflammatory response during subsequent allergen memory re-challenge weeks later. Together these data suggest that naturally-arising Tr1 cells do not necessarily give rise to more Tr1 upon allergen re-challenge or contribute to tolerogenic memory. This phenotypic instability may limit efforts to re-establish tolerance by expanding Tr1 in vivo.
Collapse
Affiliation(s)
- Koshika Yadava
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Beckman CenterStanford University School of MedicineStanfordUnited States
- Radcliffe Department of Medicine, Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUnited Kingdom
| | - Carlos Obed Medina
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Beckman CenterStanford University School of MedicineStanfordUnited States
| | - Heather Ishak
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Beckman CenterStanford University School of MedicineStanfordUnited States
| | - Irina Gurevich
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Beckman CenterStanford University School of MedicineStanfordUnited States
| | - Hedwich Kuipers
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Beckman CenterStanford University School of MedicineStanfordUnited States
- Department of Clinical NeurosciencesUniversity of CalgaryCalgaryCanada
| | - Elya Ali Shamskhou
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Beckman CenterStanford University School of MedicineStanfordUnited States
| | - Ievgen O Koliesnik
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Beckman CenterStanford University School of MedicineStanfordUnited States
| | - James J Moon
- Center for Immunology and Inflammatory DiseasesMassachusetts General Hospital, Harvard Medical SchoolCharlestownUnited States
- Division of Pulmonary and Critical Care MedicineMassachusetts General Hospital, Harvard Medical SchoolCharlestownUnited States
| | - Casey Weaver
- Bevill Biomedical Research BuildingThe University of Alabama at BirminghamBirminghamUnited States
| | - Kari Christine Nadeau
- Sean N Parker Center for Allergy & Asthma ResearchStanford UniversityMountain ViewUnited States
| | - Paul L Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Beckman CenterStanford University School of MedicineStanfordUnited States
| |
Collapse
|
39
|
Kong J, Wu K, Ji Y, Chen K, Zhang J, Sun H, Liang Y, Liang W, Chang Y, Cheng J, Tong J, Li J, Xing G, Chen G. Enhanced Bioavailability by Orally Administered Sirolimus Nanocrystals. ACS APPLIED BIO MATERIALS 2019; 2:4612-4621. [DOI: 10.1021/acsabm.9b00695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Jianglong Kong
- College of Food Science, ShiHezi University, ShiHezi 832000, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Kai Wu
- State Key Laboratory of Organic−Inorganic Composites, Beijing University of Chemical Technology, Beijing 100029, China
| | - Ying Ji
- The University of California’s Center for Environmental Implications of Nanotechnology, Los Angeles, California 90095, United States
| | - Kui Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Jiaxin Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Hui Sun
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Yuelan Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Wei Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Yanan Chang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Jenny Cheng
- The University of California’s Center for Environmental Implications of Nanotechnology, Los Angeles, California 90095, United States
| | - Junmao Tong
- College of Food Science, ShiHezi University, ShiHezi 832000, China
| | - Juan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Gengmei Xing
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Guogang Chen
- College of Food Science, ShiHezi University, ShiHezi 832000, China
| |
Collapse
|
40
|
Collier F, Ponsonby A, O'Hely M, Tang ML, Saffery R, Molloy J, Gray LE, Ranganathan S, Burgner D, Allen KJ, Brix S, Vuillermin PJ, Sly P, Harrison LC, Dwyer T. Naïve regulatory T cells in infancy: Associations with perinatal factors and development of food allergy. Allergy 2019; 74:1760-1768. [PMID: 30972786 DOI: 10.1111/all.13822] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 03/20/2019] [Accepted: 03/22/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND In previous studies, deficits in regulatory T-cell (Treg) number and function at birth have been linked with subsequent allergic disease. However, longitudinal studies that account for relevant perinatal factors are required. The aim of this study was to investigate the relationship between perinatal factors, naïve Treg (nTreg) over the first postnatal year and development of food allergy. METHODS In a birth cohort (n = 1074), the proportion of nTreg in the CD4+ T-cell compartment was measured by flow cytometry at birth (n = 463), 6 (n = 600) and 12 (n = 675) months. IgE-mediated food allergy was determined by food challenge at 1 year. Associations between perinatal factors (gestation, labour, sex, birth size), nTreg at each time point and food allergy at 1 year were examined by linear regression. RESULTS A higher proportion of nTreg at birth, larger birth size and male sex was each associated with higher nTreg in infancy. Exposure to labour, as compared to delivery by prelabour Caesarean section, was associated with a transient decrease nTreg. Infants that developed food allergy had decreased nTreg at birth, and the labour-associated decrease in nTreg at birth was more evident among infants with subsequent food allergy. Mode of birth was not associated with risk of food allergy, and there was no evidence that nTreg at either 6 or 12 months were related to food allergy. CONCLUSION The proportion of nTreg at birth is a major determinant of the proportion present throughout infancy, highlighting the importance of prenatal immune development. Exposure to the inflammatory stimulus of labour appears to reveal differences in immune function among infants at risk of food allergy.
Collapse
Affiliation(s)
- Fiona Collier
- Barwon Health Geelong Victoria Australia
- Deakin University Waurn Ponds Victoria Australia
- Murdoch Children’s Research Institute Parkville Victoria Australia
| | - Anne‐Louise Ponsonby
- Murdoch Children’s Research Institute Parkville Victoria Australia
- University of Melbourne Parkville Victoria Australia
| | - Martin O'Hely
- Deakin University Waurn Ponds Victoria Australia
- Murdoch Children’s Research Institute Parkville Victoria Australia
| | - Mimi L.K. Tang
- Murdoch Children’s Research Institute Parkville Victoria Australia
- University of Melbourne Parkville Victoria Australia
- The Royal Children’s Hospital Parkville Victoria Australia
| | - Richard Saffery
- Murdoch Children’s Research Institute Parkville Victoria Australia
- University of Melbourne Parkville Victoria Australia
| | - John Molloy
- Deakin University Waurn Ponds Victoria Australia
- Murdoch Children’s Research Institute Parkville Victoria Australia
| | - Lawrence E. Gray
- Barwon Health Geelong Victoria Australia
- Deakin University Waurn Ponds Victoria Australia
| | - Sarath Ranganathan
- Murdoch Children’s Research Institute Parkville Victoria Australia
- University of Melbourne Parkville Victoria Australia
- The Royal Children’s Hospital Parkville Victoria Australia
| | - David Burgner
- Murdoch Children’s Research Institute Parkville Victoria Australia
- University of Melbourne Parkville Victoria Australia
- The Royal Children’s Hospital Parkville Victoria Australia
| | - Katrina J. Allen
- Murdoch Children’s Research Institute Parkville Victoria Australia
- University of Melbourne Parkville Victoria Australia
- The Royal Children’s Hospital Parkville Victoria Australia
| | - Susanne Brix
- Technical University of Denmark Kongens Lyngby Denmark
| | - Peter J. Vuillermin
- Barwon Health Geelong Victoria Australia
- Deakin University Waurn Ponds Victoria Australia
- Murdoch Children’s Research Institute Parkville Victoria Australia
| | | | | | | | | |
Collapse
|
41
|
Bae JS, Kim SH, Kim JH, Kim EH, Lyu L, Chung PS, Mo JH. Effects of Low-Level Laser Irradiation in a Mouse Model of Allergic Rhinitis. Lasers Surg Med 2019; 52:347-357. [PMID: 31338850 DOI: 10.1002/lsm.23141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2019] [Indexed: 01/18/2023]
Abstract
BACKGROUND AND OBJECTIVES To evaluate the antiallergic effect of low-level laser irradiation (LLLI) at 650 nm in a mouse model of allergic rhinitis (AR), and to examine the underlying mechanisms. STUDY DESIGN/MATERIALS AND METHODS BALB/c mice were sensitized with ovalbumin (OVA) and alum and challenged intranasally with OVA. Straight- and diffusion-type LLLI were applied directly into the intranasal cavity of the mice once daily for 10 days (650 nm, 5 mW, 15 min/day) and multiple allergic parameters were evaluated. RESULTS LLLI reduced allergic symptoms, such as rubbing and sneezing, and suppressed the serum total immunoglobulin E (IgE), OVA-specific IgE, and OVA-specific IgG1 levels. Diffusion-type LLLI significantly reduced eosinophil infiltration of nasal mucosa and lymph nodes (LNs). LLLI reduced the expression of interleukin-4 (IL-4) and IL-17 in cervical LN and splenocyte culture supernatant, as well as their messenger RNA levels in nasal mucosa. However, the expression of interferonγ (IFN-γ) and IL-6 was unaffected by LLLI. The levels of reactive oxygen species (ROS) and nitric oxide (NO) in LN cells and the nasal mucosa, which were increased in the AR group, were reduced by LLLI, suggesting involvement of ROS and NO within their mechanism. CONCLUSIONS LLLI exerted an antiallergic effect by decreasing local and systemic IL-4, IL-17, and IgE levels, as well as eosinophilic infiltration into the nasal mucosa, in a mouse model of AR by modulating ROS and NO levels. Diffusion-type LLLI exhibited greater efficacy against AR than straight-type LLLI. Lasers Surg. Med. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jun-Sang Bae
- Department of Otorhinolaryngology, Dankook University College of Medicine, Cheonan, 31116, Republic of Korea.,Beckman Laser Institute Korea, Dankook University College of Medicine, Cheonan, 31116, Republic of Korea
| | - Sang Hyub Kim
- Beckman Laser Institute Korea, Dankook University College of Medicine, Cheonan, 31116, Republic of Korea
| | - Jie Hye Kim
- Department of Otorhinolaryngology, Dankook University College of Medicine, Cheonan, 31116, Republic of Korea.,Beckman Laser Institute Korea, Dankook University College of Medicine, Cheonan, 31116, Republic of Korea
| | - Eun Hee Kim
- Department of Otorhinolaryngology, Dankook University College of Medicine, Cheonan, 31116, Republic of Korea.,Beckman Laser Institute Korea, Dankook University College of Medicine, Cheonan, 31116, Republic of Korea
| | - Lele Lyu
- Department of Otorhinolaryngology, Dankook University College of Medicine, Cheonan, 31116, Republic of Korea.,Beckman Laser Institute Korea, Dankook University College of Medicine, Cheonan, 31116, Republic of Korea
| | - Phil Sang Chung
- Department of Otorhinolaryngology, Dankook University College of Medicine, Cheonan, 31116, Republic of Korea.,Beckman Laser Institute Korea, Dankook University College of Medicine, Cheonan, 31116, Republic of Korea.,Laser Translational Clinical Trial Center, Dankook University Hospital, Cheonan, 31116, Republic of Korea
| | - Ji-Hun Mo
- Department of Otorhinolaryngology, Dankook University College of Medicine, Cheonan, 31116, Republic of Korea.,Beckman Laser Institute Korea, Dankook University College of Medicine, Cheonan, 31116, Republic of Korea.,Laser Translational Clinical Trial Center, Dankook University Hospital, Cheonan, 31116, Republic of Korea
| |
Collapse
|
42
|
Abdeladhim M, Zhang AH, Kropp LE, Lindrose AR, Venkatesha SH, Mitre E, Scott DW. Engineered ovalbumin-expressing regulatory T cells protect against anaphylaxis in ovalbumin-sensitized mice. Clin Immunol 2019; 207:49-54. [PMID: 31325629 DOI: 10.1016/j.clim.2019.07.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 07/15/2019] [Indexed: 11/15/2022]
Abstract
Allergy is a major public health concern, the main treatment for which is symptomatic relief with anti-inflammatory drugs. A key clinical challenge is to induce specific tolerance in order to control allergen-specific memory B and T cells, and specifically block effector cell responses. Our lab recently developed antigen-specific regulatory T-cell (Treg) therapies as a treatment for adverse responses. Recently, we created a chimeric antigen receptor (CAR) approach in which we engineered a target protein antigen, ovalbumin (OVA), linked with the transmembrane and signal transduction domains, CD28-CD3ζ to directly target B cells and sensitized mast cells in an allergy model. We named this receptor "BAR" for B-cell Antibody Receptor. Murine or human Tregs, transduced with a BAR containing OVA or control Tregs expressing an unrelated antigen, were successfully expanded in vitro and tested in the murine OVA-alum allergy model with measurable titers of anti-OVA IgE. Because BAR Tregs express the target antigen and could interact with specific IgE on sensitized mast cells, we first demonstrated that intravenously injected OVA-BAR Tregs did not directly lead to a drop in temperature or release of mediators in plasma indicative of anaphylaxis. Forty-eight hours later, mice were challenged intraperitoneally with 200 μg OVA to induce an anaphylactic reaction, and temperature immediately measured for 30 min. We found that OVA-BAR Tregs protected mice from hypothermia, whereas mice given control BARs (expressing an unrelated antigen) or PBS showed substantial temperature drops indicative of anaphylaxis when systemically challenged with OVA. Importantly, this effect was also demonstrated in a passive anaphylaxis model in which mice that received anti-OVA IgE antibody were protected from hypothermia when treated with OVA-BAR Tregs prior to systemic OVA challenge. These results provide proof of principle that engineered allergen-specific T-regulatory cells can provide clinical protection against severe allergic reactions in individuals already IgE-sensitized to an allergen.
Collapse
Affiliation(s)
- Maha Abdeladhim
- Department of Medicine, Uniformed Services University, Bethesda, MD 20814, United States of America
| | - Ai-Hong Zhang
- Department of Medicine, Uniformed Services University, Bethesda, MD 20814, United States of America
| | - Laura E Kropp
- Department of Microbiology & Immunology, Uniformed Services University, Bethesda, MD 20814, United States of America
| | - Alyssa R Lindrose
- Department of Microbiology & Immunology, Uniformed Services University, Bethesda, MD 20814, United States of America
| | - Shivaprasad H Venkatesha
- Department of Medicine, Uniformed Services University, Bethesda, MD 20814, United States of America
| | - Edward Mitre
- Department of Microbiology & Immunology, Uniformed Services University, Bethesda, MD 20814, United States of America
| | - David W Scott
- Department of Medicine, Uniformed Services University, Bethesda, MD 20814, United States of America.
| |
Collapse
|
43
|
Zhang YL, Shin HJ, Lee JH, Lee J. Antiallergic Effect of Hizikia fusiformis in an Ovalbumin-Induced Allergic Rhinitis Mouse Model. Clin Exp Otorhinolaryngol 2019; 12:196-205. [PMID: 30942049 PMCID: PMC6453782 DOI: 10.21053/ceo.2019.00094] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 02/14/2019] [Indexed: 12/21/2022] Open
Abstract
Objectives The extract of Hizikia fusiformis is known to exhibit anticancer, antiatopic and antioxidant activities. We aimed to investigate the extract of H. fusiformis on allergic rhinitis inflammation in a mouse model. Methods The 4-week-old BALB/c mice were randomly assigned into four groups: group A, control group (n=9); group B, allergic rhinitis group (n=10); group C (n=10) received 300 mg/kg of H. fusiformis during nasal challenging period; group D (n=10) received 600 mg/kg of H. fusiformis during general sensitization period and 300 mg/kg of H. fusiformis during nasal challenging period. Allergic inflammation was made with ovalbumin (OVA) and alum then challenged intranasally with OVA. H. fusiformis was intraperitoneally administered 3 hours before the OVA administration. Allergic symptom score and the levels of immunoglobulin G1 (IgG1), IgG2a, OVA-specific IgE antibodies, levels of cytokines in the nasal mucosa and in spleen cell culture supernatant, such as tumor necrosis factor alpha (TNF-α), interleukin 4 (IL-4), IL-5, IL-13, and IL-10 were assessed. The percentage of regulatory T cell was analyzed by flow cytometry. Eosinophilic infiltration and goblet cell hyperplasia were also evaluated. Results H. fusiformis administered groups C and D showed significant inhibitory effects on nasal symptoms, IL-13 mRNA expression and eosinophil infiltration/goblet cell hyperplasia in the nasal tissue; OVA-specific IgE production in serum (P<0.05). In group D, H. fusiformis treatment downregulated IL-4, IL-5, IL-13, TNF-α, and IL-10 cytokine expression in splenocyte culture as well as significantly decreased IgG2a, IgG1 levels in serum compared with group B (P<0.05). However, the expressions of IL-5, interferon-γ and forkhead box P3 mRNA did not change in groups C and D. Conclusion H. fusiformis could induce antiallergic inflammation by suppressing the T-helper type 2 cytokine production (IL-13) locally and systemically, OVA-specific IgE formation, goblet cell hyperplasia, and eosinophilic infiltration in a mouse model of allergic rhinitis. Thus, H. fusiformis could be considered as a potential therapeutic agent in treating allergic rhinitis.
Collapse
Affiliation(s)
- Yu-Lian Zhang
- Center of Morphological Experiment, Medical College of Yanbian University, Yanji, China
| | - Hyun-Jae Shin
- Department of Biochemical and Polymer Engineering, Chosun University, Gwangju, Korea
| | - Jung-Heon Lee
- Department of Biochemical and Polymer Engineering, Chosun University, Gwangju, Korea
| | - Jieun Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Chosun University College of Medicine, Gwangju, Korea
| |
Collapse
|
44
|
Fu G, Zhao K, Chen H, Wang Y, Nie L, Wei H, Wan C. Effect of 3 lactobacilli on immunoregulation and intestinal microbiota in a β-lactoglobulin–induced allergic mouse model. J Dairy Sci 2019; 102:1943-1958. [DOI: 10.3168/jds.2018-15683] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/26/2018] [Indexed: 12/17/2022]
|
45
|
Oxaliplatin Treatment Alters Systemic Immune Responses. BIOMED RESEARCH INTERNATIONAL 2019; 2019:4650695. [PMID: 30906773 PMCID: PMC6398049 DOI: 10.1155/2019/4650695] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/22/2018] [Indexed: 12/14/2022]
Abstract
Purpose Oxaliplatin is a platinum-based chemotherapeutic agent demonstrating significant antitumor efficacy. Unlike conventional anticancer agents which are immunosuppressive, oxaliplatin has the capacity to stimulate immunological effects in response to the presentation of damage associated molecular patterns (DAMPs) elicited upon cell death. However, the effects of oxaliplatin treatment on systemic immune responses remain largely unknown. Aims of this study were to investigate the effects of oxaliplatin treatment on the proportions of (1) splenic T cells, B cells, macrophages, pro-/anti-inflammatory cytokines, gene expression of splenic cytokines, chemokines, and mediators; (2) double-positive and single-positive CD4+ and CD8+ T thymocytes; (3) bone-marrow hematopoietic stem and progenitor cells. Methods Male BALB/c mice received intraperitoneal injections of oxaliplatin (3mg/kg/d) or sterile water tri-weekly for 2 weeks. Leukocyte populations within the spleen, thymus, and bone-marrow were assessed using flow cytometry. RT-PCR was performed to characterise changes in splenic inflammation-associated genes. Results Oxaliplatin treatment reduced spleen size and cellularity (CD45+ cells), increased the proportion of CD4+, CD8+, and Treg cells, and elevated TNF-α expression. Oxaliplatin was selectively cytotoxic to B cells but had no effect on splenic macrophages. Oxaliplatin treatment altered the gene expression of several cytokines, chemokines, and cell mediators. Oxaliplatin did not deplete double-positive thymocytes but increased the single-positive CD8+ subset. There was also an increase in activated (CD69+) CD8+ T cells. Bone-marrow hematopoietic progenitor pool was demonstrably normal following oxaliplatin treatment when compared to the vehicle-treated cohort. Conclusion Oxaliplatin does not cause systemic immunosuppression and, instead, has the capacity to induce beneficial antitumor immune responses.
Collapse
|
46
|
Imam S, Prathibha R, Dar P, Almotah K, Al-Khudhair A, Hasan SAM, Salim N, Jilani TN, Mirmira RG, Jaume JC. eIF5A inhibition influences T cell dynamics in the pancreatic microenvironment of the humanized mouse model of Type 1 Diabetes. Sci Rep 2019; 9:1533. [PMID: 30733517 PMCID: PMC6367423 DOI: 10.1038/s41598-018-38341-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 12/17/2018] [Indexed: 12/14/2022] Open
Abstract
We have developed a transgenic mouse model of Type 1 Diabetes (T1D) in which human GAD65 is expressed in pancreatic β-cells, and human MHC-II is expressed on antigen presenting cells. Induced GAD65 antigen presentation activates T-cells, which initiates the downstream events leading to diabetes. In our humanized mice, we have shown downregulation of eukaryotic translation initiation factor 5 A (elF5A), expressed only in actively dividing mammalian cells. In-vivo inhibition of elF5A hypusination by deoxyhypusine synthase (DHS) inhibitor "GC7" was studied; DHS inhibitor alters the pathophysiology in our mouse model by catalyzing the crucial hypusination and the rate-limiting step of elF5A activation. In our mouse model, we have shown that inhibition of eIF5A resets the pro-inflammatory bias in the pancreatic microenvironment. There was: (a) reduction of Th1/Th17 response, (b) an increase in Treg numbers, (c) debase in IL17 and IL21 cytokines levels in serum, (d) lowering of anti-GAD65 antibodies, and (e) ablation of the ER stress that improved functionality of the β-cells, but minimal effect on the cytotoxic CD8 T-cell (CTL) mediated response. Conclusively, immune modulation, in the case of T1D, may help to manipulate inflammatory responses, decreasing disease severity, and may help manage T1D in early stages of disease. Our study also demonstrates that without manipulating the CTLs mediated response extensively, it is difficult to treat T1D.
Collapse
Affiliation(s)
- Shahnawaz Imam
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA.
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA.
| | - R Prathibha
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Pervaiz Dar
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Shuhama, Srinagar, 190006, Jammu and Kashmir, India
| | - Khalil Almotah
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Ahmed Al-Khudhair
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Syed Abdul-Moiz Hasan
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Nancy Salim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Talha Naser Jilani
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Raghavendra G Mirmira
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Juan Carlos Jaume
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA.
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA.
| |
Collapse
|
47
|
Melnik BC, Schmitz G. Exosomes of pasteurized milk: potential pathogens of Western diseases. J Transl Med 2019; 17:3. [PMID: 30602375 PMCID: PMC6317263 DOI: 10.1186/s12967-018-1760-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 12/21/2018] [Indexed: 12/16/2022] Open
Abstract
Milk consumption is a hallmark of western diet. According to common believes, milk consumption has beneficial effects for human health. Pasteurization of cow's milk protects thermolabile vitamins and other organic compounds including bioactive and bioavailable exosomes and extracellular vesicles in the range of 40-120 nm, which are pivotal mediators of cell communication via systemic transfer of specific micro-ribonucleic acids, mRNAs and regulatory proteins such as transforming growth factor-β. There is compelling evidence that human and bovine milk exosomes play a crucial role for adequate metabolic and immunological programming of the newborn infant at the beginning of extrauterine life. Milk exosomes assist in executing an anabolic, growth-promoting and immunological program confined to the postnatal period in all mammals. However, epidemiological and translational evidence presented in this review indicates that continuous exposure of humans to exosomes of pasteurized milk may confer a substantial risk for the development of chronic diseases of civilization including obesity, type 2 diabetes mellitus, osteoporosis, common cancers (prostate, breast, liver, B-cells) as well as Parkinson's disease. Exosomes of pasteurized milk may represent new pathogens that should not reach the human food chain.
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Am Finkenhügel 7A, 49076 Osnabrück, Germany
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, University of Regensburg, Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| |
Collapse
|
48
|
The Therapeutic Strategies of Regulatory T Cells in Malignancies and Stem Cell Transplantations. JOURNAL OF ONCOLOGY 2019; 2019:5981054. [PMID: 30693029 PMCID: PMC6332959 DOI: 10.1155/2019/5981054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 10/09/2018] [Accepted: 12/02/2018] [Indexed: 12/14/2022]
Abstract
Regulatory T cells (Treg cells) are considered one of the main dynamic cell types within the immune system. Because Treg cells suppress immune responses, they have potential roles in immunological self-tolerance and may help to maintain immune homeostasis. Promoting Treg cell function and increasing their numbers might be useful in treating autoimmune disorders, as well as preventing allograft rejection. However, studies of mice and humans demonstrate that Treg cells promote cancer progression and suppress antitumor immunity. Therefore, suppressing Treg cell function or reducing their numbers could support the immune system's response to pathogenic microorganisms and tumors. As a result, there is great interest in investigating the Treg cells role in the treatment of hematological and nonhematological malignancies. Consequently, Treg cells could be a fundamentally important target for pathologies of the immune system. Targeting effector Treg cells could help to distinguish and selectively decrease these cells while preserving other Treg cells needed to suppress autoimmunity. Currently, a promising way to treat malignancies and other autoimmune disorders is stem cell transplantation. Stem cell transplants (SCT) can help to manage the production of Treg cells and also may produce more efficient Treg cells, thereby suppressing clinical disease progression. Specifically, mature T cells within the engrafted stem cells mediate this SCT beneficial effect. During SCT, the recipient's immune system is replaced with a donor, which allows for improved immune system function. In addition, SCT can protect from disease relapse, as graft-versus-host disease (GvHD) in transplant patients can be protective against cancer recurrence. The current review will define the role of regulatory T cells in treatment of malignancy. Additionally, it will summarize current promising research regarding the utility of regulatory T cells in stem cell transplantation.
Collapse
|
49
|
IL-10 gene-modified dendritic cells-induced type 1 T regulatory cells inhibit graft-versus-host disease while preserving graft-versus-leukemia effect. Biochem Biophys Res Commun 2018; 507:122-127. [DOI: 10.1016/j.bbrc.2018.10.183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 10/29/2018] [Indexed: 11/22/2022]
|
50
|
Ahmad S, Azid NA, Boer JC, Lim J, Chen X, Plebanski M, Mohamud R. The Key Role of TNF-TNFR2 Interactions in the Modulation of Allergic Inflammation: A Review. Front Immunol 2018; 9:2572. [PMID: 30473698 PMCID: PMC6238659 DOI: 10.3389/fimmu.2018.02572] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 10/18/2018] [Indexed: 12/14/2022] Open
Abstract
Tumor necrosis factor-alpha (TNF) is a pleiotropic cytokine, which is thought to play a major role in the pathogenesis of inflammatory diseases, including allergy. TNF is produced at the early stage of allergen sensitization, and then continues to promote the inflammation cascade in the effector phase of allergic reactions. Consequently, anti-TNF treatment has been proposed as a potential therapeutic option. However, recent studies reveal anti-intuitive effects of TNF in the activation and proliferative expansion of immunosuppressive Tregs, tolerogenic DCs and MDSCs. This immunosuppressive effect of TNF is mediated by TNFR2, which is preferentially expressed by immunosuppressive cells. These findings redefine the role of TNF in allergic reaction, and suggest that targeting TNF-TNFR2 interaction itself may represent a novel strategy in the treatment of allergy.
Collapse
Affiliation(s)
- Suhana Ahmad
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Nor Azrini Azid
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Jennifer C Boer
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - JitKang Lim
- School of Chemical Engineering, Universiti Sains Malaysia, Pulau Pinang, Malaysia
| | - Xin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, China
| | | | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia.,Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Kelantan, Malaysia
| |
Collapse
|