1
|
Lu Y, Ma Q, Yu L, Liu X, Chen P, Liu W. Circulating CD45RA -Foxp3 ++ Treg cells serve as a biomarker for predicting minimal clinical manifestations status of myasthenia gravis. Life Sci 2024; 358:123162. [PMID: 39433086 DOI: 10.1016/j.lfs.2024.123162] [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: 04/01/2024] [Revised: 07/28/2024] [Accepted: 10/17/2024] [Indexed: 10/23/2024]
Abstract
AIMS Regulatory T cells (Tregs) are key mediators of the induction of immune tolerance; however, the mechanisms by which they regulate myasthenia gravis (MG) are not fully understood. This study aimed to explore the characteristics of Tregs and their subpopulations in the peripheral blood of patients with minimal clinical manifestations (MM) of MG and identify biomarkers that predict MM-MG for treatment guidance. MATERIALS AND METHODS The clinical data of patients with general MG who visited our hospital were retrospectively analyzed. Age- and sex-matched volunteers were selected as healthy controls (HC). Flow cytometry was used to determine the proportion, function, and subpopulations of total Tregs. A correlation analysis was conducted for subpopulation proportions and MG disease severity. KEY FINDINGS A total of 27 cases of MM-MG, 40 cases of naїve-MG, and 33 cases of HC were included in this study. The number of total Tregs and the suppressive function of total Tregs were elevated in patients with MM-MG compared to those of patients with naїve-MG. Further analysis revealed that the frequency of CD45RA-Foxp3++ Tregs (a-Tregs) negatively correlated with quantitative myasthenia gravis (QMG) scores for patients with naїve-MG. In addition, the number of a-Tregs was significantly greater in patients with MM-MG than in patients with naїve-MG, and CD45RA-Foxp3+ Tregs expressed higher and lower levels of CTLA-4 and CXCR3, respectively. SIGNIFICANCE CD45RA-Foxp3++ Tregs were significantly more abundant and highly expressed surface inhibitory molecules in patients with MM-MG. This profile may serve as a predictive biomarker for MM-MG.
Collapse
Affiliation(s)
- Yaru Lu
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou 450003, Henan, China; Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou 510080, Guangdong, China
| | - Qian Ma
- Department of Neurology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Lu Yu
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Xiaoxi Liu
- Department of Neurology, Nanfang Hospital of Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Pei Chen
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou 510080, Guangdong, China
| | - Weibin Liu
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou 450003, Henan, China.
| |
Collapse
|
2
|
Schoberleitner I, Faserl K, Lackner M, Coraça-Huber DC, Augustin A, Imsirovic A, Sigl S, Wolfram D. Unraveling the Immune Web: Advances in SMI Capsular Fibrosis from Molecular Insights to Preclinical Breakthroughs. Biomolecules 2024; 14:1433. [PMID: 39595609 PMCID: PMC11592141 DOI: 10.3390/biom14111433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Breast implant surgery has evolved significantly, yet challenges such as capsular contracture remain a persistent concern. This review presents an in-depth analysis of recent advancements in understanding the immune mechanisms and clinical implications associated with silicone mammary implants (SMIs). The article systematically examines the complex interplay between immune responses and capsular fibrosis, emphasizing the pathophysiological mechanisms of inflammation in the etiology of this fibrotic response. It discusses innovations in biomaterial science, including the development of novel anti-biofilm coatings and immunomodulatory surfaces designed to enhance implant integration and minimize complications. Emphasis is placed on personalized risk assessment strategies, leveraging molecular insights to tailor interventions and improve patient outcomes. Emerging therapeutic targets, advancements in surgical techniques, and the refinement of post-operative care are also explored. Despite notable progress, challenges such as the variability in immune responses, the long-term efficacy of new interventions, and ethical considerations remain. Future research directions are identified, focusing on personalized medicine, advanced biomaterials, and bridging preclinical findings with clinical applications. As we advance from bench to bedside, this review illuminates the path forward, where interdisciplinary collaboration and continued inquiry weave together to enhance the art and science of breast implant surgery, transforming patient care into a realm of precision and excellence.
Collapse
Affiliation(s)
- Ines Schoberleitner
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, Müllerstraße 44, 6020 Innsbruck, Austria
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Klaus Faserl
- Protein Core Facility, Institute of Medical Chemistry, Biocenter, Medical University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Michaela Lackner
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Schöpfstraße 41, 6020 Innsbruck, Austria
| | - Débora C. Coraça-Huber
- BIOFILM Lab, Department of Orthopedics and Traumatology, Medical University of Innsbruck, Müllerstraße 44, 6020 Innsbruck, Austria
| | - Angela Augustin
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Anja Imsirovic
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Stephan Sigl
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Dolores Wolfram
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| |
Collapse
|
3
|
Liu M, Liu JP, Wang P, Fu YJ, Zhao M, Jiang YJ, Zhang ZN, Shang H. Approaches for Performance Verification Toward Standardization of Peripheral Blood Regulatory T-Cell Detection by Flow Cytometry. Arch Pathol Lab Med 2024; 148:1234-1243. [PMID: 38385871 DOI: 10.5858/arpa.2023-0284-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2023] [Indexed: 02/23/2024]
Abstract
CONTEXT.— Regulatory T-cell (Treg) detection in peripheral blood, based on flow cytometry, is invaluable for diagnosis and treatment of immune-mediated diseases. However, there is a lack of reliable methods to verify the performance, which is pivotal toward standardization of the Tregs assay. OBJECTIVE.— To conduct standardization studies and verify the performance of 3 commercially available reagent sets for the Tregs assay based on flow cytometry and agreement analysis for Treg detection across the different reagent sets. DESIGN.— The analytical performance of Tregs assay using reagent sets supplied by 3 manufacturers was evaluated after establishing the gating strategy and determining the optimal antibody concentration. Postcollection sample stability was evaluated, as well as the repeatability, reproducibility, reportable range, linearity, and assay carryover. Agreement between the different assays was assessed via Bland-Altman plots and linear regression analysis. The relationship between the frequency of CD4+CD25+CD127low/- Tregs and CD4+CD25+Foxp3+ Tregs was evaluated. RESULTS.— The postcollection sample stability was set at 72 hours after collection at room temperature. The accuracy, repeatability, reproducibility, and accuracy all met the requirements for clinical analysis. Excellent linearity, with R2 ≥0.9 and no assay carryover, was observed. For reportable range, a minimum of 1000 events in the CD3+CD4+ gate was required for Tregs assay. Moreover, the results for Tregs labeled by antibodies from the 3 manufacturers were in good agreement. The percentage of CD4+CD25+CD127low/- Tregs was closely correlated with CD4+CD25+Foxp3+ Tregs. CONCLUSIONS.— This is the first study to evaluate systematically the measurement performance of Tregs in peripheral blood by flow cytometry, which provides a practical solution to verifying the performance of flow cytometry-based immune monitoring projects in clinical practice.
Collapse
Affiliation(s)
- Mei Liu
- From NHC Key Laboratory of AIDS Immunology - China Medical University (M Liu, J-P Liu, Wang, Fu, Jiang, Zhang, Shang), National Clinical Research Center for Laboratory Medicine (M Liu, J-P Liu, Wang, Fu, Zhao, Jiang, Zhang, Shang), and the Department of Laboratory Medicine (Zhao, Shang), The First Hospital of China Medical University, Shenyang, China
- Units of Medical Laboratory (M Liu, J-P Liu, Wang, Fu, Jiang, Zhang, Shang) and Key Laboratory of AIDS Immunology (J-P Liu, Wang, Fu, Jiang, Zhang, Shang), Chinese Academy of Medical Sciences, Shenyang, China
| | - Jin-Peng Liu
- From NHC Key Laboratory of AIDS Immunology - China Medical University (M Liu, J-P Liu, Wang, Fu, Jiang, Zhang, Shang), National Clinical Research Center for Laboratory Medicine (M Liu, J-P Liu, Wang, Fu, Zhao, Jiang, Zhang, Shang), and the Department of Laboratory Medicine (Zhao, Shang), The First Hospital of China Medical University, Shenyang, China
- Units of Medical Laboratory (M Liu, J-P Liu, Wang, Fu, Jiang, Zhang, Shang) and Key Laboratory of AIDS Immunology (J-P Liu, Wang, Fu, Jiang, Zhang, Shang), Chinese Academy of Medical Sciences, Shenyang, China
| | - Pan Wang
- From NHC Key Laboratory of AIDS Immunology - China Medical University (M Liu, J-P Liu, Wang, Fu, Jiang, Zhang, Shang), National Clinical Research Center for Laboratory Medicine (M Liu, J-P Liu, Wang, Fu, Zhao, Jiang, Zhang, Shang), and the Department of Laboratory Medicine (Zhao, Shang), The First Hospital of China Medical University, Shenyang, China
- Units of Medical Laboratory (M Liu, J-P Liu, Wang, Fu, Jiang, Zhang, Shang) and Key Laboratory of AIDS Immunology (J-P Liu, Wang, Fu, Jiang, Zhang, Shang), Chinese Academy of Medical Sciences, Shenyang, China
| | - Ya-Jing Fu
- From NHC Key Laboratory of AIDS Immunology - China Medical University (M Liu, J-P Liu, Wang, Fu, Jiang, Zhang, Shang), National Clinical Research Center for Laboratory Medicine (M Liu, J-P Liu, Wang, Fu, Zhao, Jiang, Zhang, Shang), and the Department of Laboratory Medicine (Zhao, Shang), The First Hospital of China Medical University, Shenyang, China
- Units of Medical Laboratory (M Liu, J-P Liu, Wang, Fu, Jiang, Zhang, Shang) and Key Laboratory of AIDS Immunology (J-P Liu, Wang, Fu, Jiang, Zhang, Shang), Chinese Academy of Medical Sciences, Shenyang, China
| | - Min Zhao
- From NHC Key Laboratory of AIDS Immunology - China Medical University (M Liu, J-P Liu, Wang, Fu, Jiang, Zhang, Shang), National Clinical Research Center for Laboratory Medicine (M Liu, J-P Liu, Wang, Fu, Zhao, Jiang, Zhang, Shang), and the Department of Laboratory Medicine (Zhao, Shang), The First Hospital of China Medical University, Shenyang, China
| | - Yong-Jun Jiang
- From NHC Key Laboratory of AIDS Immunology - China Medical University (M Liu, J-P Liu, Wang, Fu, Jiang, Zhang, Shang), National Clinical Research Center for Laboratory Medicine (M Liu, J-P Liu, Wang, Fu, Zhao, Jiang, Zhang, Shang), and the Department of Laboratory Medicine (Zhao, Shang), The First Hospital of China Medical University, Shenyang, China
- Units of Medical Laboratory (M Liu, J-P Liu, Wang, Fu, Jiang, Zhang, Shang) and Key Laboratory of AIDS Immunology (J-P Liu, Wang, Fu, Jiang, Zhang, Shang), Chinese Academy of Medical Sciences, Shenyang, China
| | - Zi-Ning Zhang
- From NHC Key Laboratory of AIDS Immunology - China Medical University (M Liu, J-P Liu, Wang, Fu, Jiang, Zhang, Shang), National Clinical Research Center for Laboratory Medicine (M Liu, J-P Liu, Wang, Fu, Zhao, Jiang, Zhang, Shang), and the Department of Laboratory Medicine (Zhao, Shang), The First Hospital of China Medical University, Shenyang, China
- Units of Medical Laboratory (M Liu, J-P Liu, Wang, Fu, Jiang, Zhang, Shang) and Key Laboratory of AIDS Immunology (J-P Liu, Wang, Fu, Jiang, Zhang, Shang), Chinese Academy of Medical Sciences, Shenyang, China
| | - Hong Shang
- From NHC Key Laboratory of AIDS Immunology - China Medical University (M Liu, J-P Liu, Wang, Fu, Jiang, Zhang, Shang), National Clinical Research Center for Laboratory Medicine (M Liu, J-P Liu, Wang, Fu, Zhao, Jiang, Zhang, Shang), and the Department of Laboratory Medicine (Zhao, Shang), The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
4
|
Bekis Bozkurt H, Bayram Catak F, Sahin A, Yalcin Gungoren E, Gemici Karaarslan B, Yakici N, Yorgun Altunbas M, Catak MC, Can S, Amirov R, Bozkurt S, Ozturk N, Bilgic Eltan S, Kasap N, Bal Cetinkaya F, Orhan F, Arga M, Cavkaytar O, Kiykim A, Karakoc-Aydiner E, Ozen A, Baris S. Diverse Clinical and Immunological Profiles in Patients with IPEX Syndrome: a Multicenter Analysis from Turkey. J Clin Immunol 2024; 45:9. [PMID: 39283523 DOI: 10.1007/s10875-024-01791-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] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 08/19/2024] [Indexed: 12/06/2024]
Abstract
PURPOSE Immunodysregulation, Polyendocrinopathy, Enteropathy, and X-linked syndrome (IPEX), caused by pathogenic FOXP3 variants, is a rare autoimmune disorder with diverse clinical features, including early-onset diabetes, eczema, and enteropathy. Atypical cases show milder symptoms and unique signs, requiring different treatments. Therefore, there are ambiguities in the accurate diagnosis and management of IPEX. We sought to present clinical, genetic, and immunological assessments of 12 IPEX patients with long-term follow-up to facilitate the diagnosis and management of the disease. METHODS Clinical findings and treatment options of the patients were collected over time. Lymphocyte subpopulations, protein expressions, regulatory T (Treg) and circulating T follicular helper (cTFH) cells, and T-cell proliferation were analyzed. RESULTS Predominant presentations included autoimmunity (91.6%), failure to thrive (66.7%), and eczema (58.3%). There were four classical and eight atypical IPEX individuals. Allergic manifestations were more common in atypical patients. Notably, chronic diarrhea demonstrated heightened severity compared to other manifestations. Four patients (33.3%) demonstrated eosinophilia, and nine (75%) showed high serum IgE levels. Most patients exhibited normal percentages of Treg cells with reduced CD25, FOXP3, and CTLA-4 expressions, corrected after hematopoietic stem cell transplantation (HSCT). Compared to healthy controls, the TH2-like skewing accompanied by reduced TH17-like responses was observed in cTFH and Treg cells of patients. Overall, nine patients (75%) received immunosuppressants (ISs), and six (50%) underwent HSCT, which was the only treatment revealing sustained control. Sirolimus was used in six patients and showed better control than other ISs. CONCLUSIONS The first cohort from Turkey with long-term follow-up results, comparing typical and atypical cases, provides insights into the outcomes of different therapeutic modalities and T- cell subtype changes in IPEX syndrome.
Collapse
MESH Headings
- Humans
- Turkey
- Male
- Child, Preschool
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Genetic Diseases, X-Linked/diagnosis
- Genetic Diseases, X-Linked/genetics
- Genetic Diseases, X-Linked/immunology
- Genetic Diseases, X-Linked/therapy
- T-Lymphocytes, Regulatory/immunology
- Infant
- Female
- Child
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/diagnosis
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/congenital
- Immune System Diseases/diagnosis
- Immune System Diseases/genetics
- Immune System Diseases/therapy
- Immune System Diseases/congenital
- Autoimmunity
- Adolescent
- Diarrhea
Collapse
Affiliation(s)
- Hayrunnisa Bekis Bozkurt
- Faculty of Medicine, Department of Pediatric Allergy and Immunology, Istanbul Medeniyet University, Istanbul, Turkey
| | - Feyza Bayram Catak
- Faculty of Medicine, Department of Pediatric Allergy and Immunology, Marmara University, Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Ali Sahin
- Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Ezgi Yalcin Gungoren
- Faculty of Medicine, Department of Pediatric Allergy and Immunology, Marmara University, Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Betul Gemici Karaarslan
- Faculty of Medicine, Department of Pediatric Allergy and Immunology, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Nalan Yakici
- Faculty of Medicine, Department of Pediatric Allergy and Immunology, Karadeniz Technical University, Trabzon, Turkey
| | - Melek Yorgun Altunbas
- Faculty of Medicine, Department of Pediatric Allergy and Immunology, Marmara University, Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Mehmet Cihangir Catak
- Faculty of Medicine, Department of Pediatric Allergy and Immunology, Marmara University, Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Salim Can
- Faculty of Medicine, Department of Pediatric Allergy and Immunology, Marmara University, Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Razin Amirov
- Faculty of Medicine, Department of Pediatric Allergy and Immunology, Marmara University, Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Selcen Bozkurt
- Faculty of Medicine, Department of Pediatric Allergy and Immunology, Marmara University, Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Necmiye Ozturk
- Faculty of Medicine, Department of Pediatric Allergy and Immunology, Marmara University, Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Sevgi Bilgic Eltan
- Faculty of Medicine, Department of Pediatric Allergy and Immunology, Marmara University, Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Nurhan Kasap
- Faculty of Medicine, Department of Pediatric Allergy and Immunology, Istanbul Medeniyet University, Istanbul, Turkey
| | - Fatma Bal Cetinkaya
- Faculty of Medicine, Department of Pediatric Allergy and Immunology, Istanbul Medeniyet University, Istanbul, Turkey
| | - Fazil Orhan
- Faculty of Medicine, Department of Pediatric Allergy and Immunology, Karadeniz Technical University, Trabzon, Turkey
| | - Mustafa Arga
- Faculty of Medicine, Department of Pediatric Allergy and Immunology, Istanbul Medeniyet University, Istanbul, Turkey
| | - Ozlem Cavkaytar
- Faculty of Medicine, Department of Pediatric Allergy and Immunology, Istanbul Medeniyet University, Istanbul, Turkey
| | - Ayca Kiykim
- Faculty of Medicine, Department of Pediatric Allergy and Immunology, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Elif Karakoc-Aydiner
- Faculty of Medicine, Department of Pediatric Allergy and Immunology, Marmara University, Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Ahmet Ozen
- Faculty of Medicine, Department of Pediatric Allergy and Immunology, Marmara University, Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Safa Baris
- Faculty of Medicine, Department of Pediatric Allergy and Immunology, Marmara University, Istanbul, Turkey.
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey.
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey.
| |
Collapse
|
5
|
Wang L, Liang A, Huang J. Exendin-4-enriched exosomes from hUCMSCs alleviate diabetic nephropathy via gut microbiota and immune modulation. Front Microbiol 2024; 15:1399632. [PMID: 39282564 PMCID: PMC11392743 DOI: 10.3389/fmicb.2024.1399632] [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: 03/12/2024] [Accepted: 07/30/2024] [Indexed: 09/19/2024] Open
Abstract
Introduction Diabetic nephropathy (DN) presents a significant therapeutic challenge, compounded by complex pathophysiological mechanisms. Recent studies suggest Exendin-4 (Ex-4) as a potential ameliorative agent for DN, albeit with unclear mechanisms. This research investigates the effects and underlying mechanisms of Ex-4-enriched exosomes derived from human umbilical cord mesenchymal stem cells (hUCMSCs) on DN, focusing on their renoprotective properties and interactions with gut microbiota. Method Exosomes from hUCMSCs (hUCMSCs-Exo) were loaded with Ex-4 via electroporation. A streptozotocin (STZ) -induced DN mouse model was employed to assess the therapeutic impact of these engineered exosomes. The study further explored immune cell dynamics, mainly CD4+ regulatory T (Treg) cells, using bioinformatics, flow cytometry, and the influence of gut microbiota through antibiotic treatment and specific bacterial reintroduction. Results Treatment with hUCMSCs-Exo@Ex-4 significantly improved key DN markers, including blood glucose and proteinuria, alleviating kidney damage. A notable decrease in natural Treg cell infiltration in DN was observed, while Ex-4-loaded exosomes promoted CD4+ Treg cell induction. The therapeutic benefits of hUCMSCs-Exo@Ex-4 were diminished upon CD4+ Treg cell depletion, underscoring their role in this context. Notably, CD4+ Treg cell induction correlated with the presence of Prevotella species, and disruption of gut microbiota adversely affected these cells and the therapeutic efficacy of the treatment. However, the reintroduction of Prevotella strains counteracted these adverse effects. Discussion This study elucidates a novel therapeutic mechanism of Ex-4-loaded hUCMSCs exosomes in DN, highlighting the induction of CD4+ Treg cells mediated by specific gut microbiota components. These findings underscore the potential of leveraging gut microbiota and immune cell interplay in developing effective DN treatments.
Collapse
Affiliation(s)
- Liping Wang
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical, Beijing, China
| | - Aihua Liang
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical, Beijing, China
| | - Jukai Huang
- Department of Endocrinology, Beijing University of Chinese Medicine, Dongzhimen Hospital, Beijing, China
| |
Collapse
|
6
|
You Y, Wu X, Yuan H, He Y, Chen Y, Wang S, Min H, Chen J, Li C. Crystalline silica-induced recruitment and immuno-imbalance of CD4 + tissue resident memory T cells promote silicosis progression. Commun Biol 2024; 7:971. [PMID: 39122899 PMCID: PMC11316055 DOI: 10.1038/s42003-024-06662-z] [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: 03/29/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Occupational crystalline silica (CS) particle exposure leads to silicosis. The burden of CS-associated disease remains high, and treatment options are limited due to vague mechanisms. Here we show that pulmonary CD4+ tissue-resident memory T cells (TRM) accumulate in response to CS particles, mediating the pathogenesis of silicosis. The TRM cells are derived from peripheral lymphocyte recruitment and in situ expansion. Specifically, CD69+CD103+ TRM-Tregs depend more on circulating T cell replenishment. CD69 and CD103 can divide the TRM cells into functionally distinct subsets, mirroring the immuno-balance within CD4+ TRM cells. However, targeting CD103+ TRM-Tregs do not mitigate disease phenotype since the TRM subsets exert immunosuppressive but not pro-fibrotic roles. After identifying pathogenic CD69+CD103- subsets, we highlight IL-7 for their maintenance and function, that present a promising avenue for mitigating silicosis. Together, our findings highlight the distinct role of CD4+ TRM cells in mediating CS-induced fibrosis and provide potential therapeutic strategies.
Collapse
Affiliation(s)
- Yichuan You
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
| | - Xiulin Wu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
| | - Haoyang Yuan
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
| | - Yangyang He
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
| | - Yinghui Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
| | - Sisi Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
| | - Hui Min
- Department of Immunology, College of Basic Medical Sciences, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
| | - Jie Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China.
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China.
| | - Chao Li
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China.
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China.
| |
Collapse
|
7
|
Alhosseini MN, Ebadi P, Karimi MH, Migliorati G, Cari L, Nocentini G, Heidari M, Soleimanian S. Therapy with regulatory T-cell infusion in autoimmune diseases and organ transplantation: A review of the strengths and limitations. Transpl Immunol 2024; 85:102069. [PMID: 38844002 DOI: 10.1016/j.trim.2024.102069] [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: 11/10/2023] [Revised: 05/29/2024] [Accepted: 06/01/2024] [Indexed: 06/17/2024]
Abstract
In the last decade, cell therapies have revolutionized the treatment of some diseases, earning the definition of being the "third pillar" of therapeutics. In particular, the infusion of regulatory T cells (Tregs) is explored for the prevention and control of autoimmune reactions and acute/chronic allograft rejection. Such an approach represents a promising new treatment for autoimmune diseases to recover an immunotolerance against autoantigens, and to prevent an immune response to alloantigens. The efficacy of the in vitro expanded polyclonal and antigen-specific Treg infusion in the treatment of a large number of autoimmune diseases has been extensively demonstrated in mouse models. Similarly, experimental work documented the efficacy of Treg infusions to prevent acute and chronic allograft rejections. The Treg therapy has shown encouraging results in the control of type 1 diabetes (T1D) as well as Crohn's disease, systemic lupus erythematosus, autoimmune hepatitis and delaying graft rejection in clinical trials. However, the best method for Treg expansion and the advantages and pitfalls with the different types of Tregs are not fully understood in terms of how these therapeutic treatments can be applied in the clinical setting. This review provides an up-to-date overview of Treg infusion-based treatments in autoimmune diseases and allograft transplantation, the current technical challenges, and the highlights and disadvantages of this therapeutic approaches."
Collapse
Affiliation(s)
| | - Padideh Ebadi
- Islamic Azad University, Department of Biochemistry, Kazerun, Iran
| | | | - Graziella Migliorati
- University of Perugia, Department of Medicine and Surgery, Section of Pharmacology, Perugia, Italy
| | - Luigi Cari
- University of Perugia, Department of Medicine and Surgery, Section of Pharmacology, Perugia, Italy
| | - Giuseppe Nocentini
- University of Perugia, Department of Medicine and Surgery, Section of Pharmacology, Perugia, Italy
| | - Mozhdeh Heidari
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeede Soleimanian
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
8
|
Nogueira JDS, Gomes TR, Secco DA, de Almeida IS, da Costa ASMF, Cobas RA, Costa Dos Santos G, Gomes MB, Porto LC. Type 1 Diabetes Brazilian patients exhibit reduced frequency of recent thymic emigrants in regulatory CD4 +CD25 +Foxp3 +T cells. Immunol Lett 2024; 267:106857. [PMID: 38604551 DOI: 10.1016/j.imlet.2024.106857] [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: 11/28/2023] [Revised: 03/13/2024] [Accepted: 04/04/2024] [Indexed: 04/13/2024]
Abstract
To control immune responses, regulatory CD4+CD25+Foxp3+ T cells (Treg) maintain their wide and diverse repertoire through continuous arrival of recent thymic emigrants (RTE). However, during puberty, the activity of RTE starts to decline as a natural process of thymic involution, introducing consequences, not completely described, to the repertoire. Type 1 diabetes (T1D) patients show quantitative and qualitative impairments on the Treg cells. Our aim was to evaluate peripheral Treg and RTE cell frequencies, in T1D patients from two distinct age groups (young and adults) and verify if HLA phenotypes are concomitant associated. To this, blood samples from Brazilian twenty established T1D patients (12 young and 8 adults) and twenty-one healthy controls (11 young and 10 adults) were analyzed, by flow cytometry, to verify the percentages of CD4, Treg (CD4+CD25+Foxp3+) and the subsets of CD45RA+ (naive) and CD31+(RTE) within then. Furthermore, the HLA typing was also set. We observed that the young established T1D patients feature decreased frequencies in total Treg cells and naive RTE within Treg cells. Significant prevalence of HLA alleles, associated with risk, in T1D patients, was also identified. Performing a multivariate analysis, we confirmed that the cellular changes described offers significant variables that distinct T1D patients from the controls. Our data collectively highlight relevant aspects about homeostasis imbalances in the Treg cells of T1D patients, especially in young, and disease prognosis; that might contribute for future therapeutic strategies involving Treg cells manipulation.
Collapse
Affiliation(s)
- Jeane de Souza Nogueira
- Immunogenetic and Histocompatibility Laboratory (HLA-UERJ), Technologic core in Tissue Repair and Histocompatibility (TIXUS), Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Thamires Rodrigues Gomes
- Immunogenetic and Histocompatibility Laboratory (HLA-UERJ), Technologic core in Tissue Repair and Histocompatibility (TIXUS), Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Danielle Angst Secco
- Immunogenetic and Histocompatibility Laboratory (HLA-UERJ), Technologic core in Tissue Repair and Histocompatibility (TIXUS), Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Inez Silva de Almeida
- Nursing Faculty, Department of Nursing Fundamentals, Ambulatory of the Adolescent Health Studies Center (NESA), Pedro Ernesto University Hospital, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | | | - Roberta Arnoldi Cobas
- Ambulatory of Diabetes, Piquet Carneiro Polyclinic, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Gilson Costa Dos Santos
- Laboratory of Metabolomics (LabMet), IBRAG, Rio de Janeiro State University, Rio de Janeiro RJ Brazil
| | - Marília Brito Gomes
- Ambulatory of Diabetes, Piquet Carneiro Polyclinic, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Luís Cristóvão Porto
- Immunogenetic and Histocompatibility Laboratory (HLA-UERJ), Technologic core in Tissue Repair and Histocompatibility (TIXUS), Rio de Janeiro State University, Rio de Janeiro, Brazil.
| |
Collapse
|
9
|
Riaz MF, Garg G, Umeano L, Iftikhar S, Alhaddad SF, Paulsingh CN, Hamid P. Comparison of Low-Dose Interleukin 2 Therapy in Conjunction With Standard Therapy in Patients With Systemic Lupus Erythematosus vs Rheumatoid Arthritis: A Systematic Review. Cureus 2024; 16:e56704. [PMID: 38646383 PMCID: PMC11032642 DOI: 10.7759/cureus.56704] [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: 02/03/2024] [Accepted: 03/22/2024] [Indexed: 04/23/2024] Open
Abstract
This systematic review aims to compare the efficacy and safety of a novel immunotherapy with low-dose interleukin 2 (IL2) across two of the most prevalent autoimmune diseases i.e. systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Contemporary therapeutic practices have not been able to achieve complete remission from these autoimmune disorders. In contrast, low-dose IL2 has shown promise in achieving this therapeutic goal via inducing self-tolerance in patients with autoimmune diseases; however, due to variable irregularities among autoimmune processes of variable diseases, the benefit of low-dose IL2 could not be determined among different autoimmune diseases. Therefore, we conducted a study to compare low-dose IL2 therapy effects on SLE and RA. We systematically screened four databases: PubMed, Medical Literature Analysis and Retrieval System Online (MEDLINE), PubMed Central (PMC), and Google Scholar. Inclusion and exclusion criteria were implemented. Quality appraisal of studies chosen for the review was done using the Cochrane Risk-of-Bias (RoB) assessment tool for randomized controlled trials, and the Newcastle-Ottawa Scale (NOS) and JBI critical appraisal tool for non-randomized clinical trials. Information was gathered from seven articles: three randomized controlled trials and four non-randomized clinical trials. Our review concluded that low-dose IL2 therapy in conjunction with respective standard therapies for SLE and RA has a higher efficacy and safety profile as compared to standard therapy alone and the therapeutic effects were comparable in both SLE and RA patients treated with low-dose IL2; however, this novel intervention does not seem to have a significant corrective effect on the biomarkers of RA as it does for SLE biomarkers.
Collapse
Affiliation(s)
- Muhammad Faisal Riaz
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Oncology, Nuclear Medicine, Oncology and Radiotherapy Institute (NORI), Rawalpindi, PAK
| | - Gourav Garg
- Orthopaedics, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Orthopaedics, King's Mill Hospital, Sutton-in-Ashfield, GBR
| | - Lotanna Umeano
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sadaf Iftikhar
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sarah F Alhaddad
- Pediatrics, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Christian N Paulsingh
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Pathology, St. George's University School of Medicine, St. George's, GRD
| | - Pousette Hamid
- Neurology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
10
|
Sun Z, Gao Z, Xiang M, Feng Y, Wang J, Xu J, Wang Y, Liang J. Comprehensive analysis of lactate-related gene profiles and immune characteristics in lupus nephritis. Front Immunol 2024; 15:1329009. [PMID: 38455045 PMCID: PMC10917958 DOI: 10.3389/fimmu.2024.1329009] [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: 10/27/2023] [Accepted: 02/12/2024] [Indexed: 03/09/2024] Open
Abstract
Objectives The most frequent cause of kidney damage in systemic lupus erythematosus (SLE) is lupus nephritis (LN), which is also a significant risk factor for morbidity and mortality. Lactate metabolism and protein lactylation might be related to the development of LN. However, there is still a lack of relative research to prove the hypothesis. Hence, this study was conducted to screen the lactate-related biomarkers for LN and analyze the underlying mechanism. Methods To identify differentially expressed genes (DEGs) in the training set (GSE32591, GSE127797), we conducted a differential expression analysis (LN samples versus normal samples). Then, module genes were mined using WGCNA concerning LN. The overlapping of DEGs, critical module genes, and lactate-related genes (LRGs) was used to create the lactate-related differentially expressed genes (LR-DEGs). By using a machine-learning algorithm, ROC, and expression levels, biomarkers were discovered. We also carried out an immune infiltration study based on biomarkers and GSEA. Results A sum of 1259 DEGs was obtained between LN and normal groups. Then, 3800 module genes in reference to LN were procured. 19 LR-DEGs were screened out by the intersection of DEGs, key module genes, and LRGs. Moreover, 8 pivotal genes were acquired via two machine-learning algorithms. Subsequently, 3 biomarkers related to lactate metabolism were obtained, including COQ2, COQ4, and NDUFV1. And these three biomarkers were enriched in pathways 'antigen processing and presentation' and 'NOD-like receptor signaling pathway'. We found that Macrophages M0 and T cells regulatory (Tregs) were associated with these three biomarkers as well. Conclusion Overall, the results indicated that lactate-related biomarkers COQ2, COQ4, and NDUFV1 were associated with LN, which laid a theoretical foundation for the diagnosis and treatment of LN.
Collapse
Affiliation(s)
- Zhan Sun
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhanyan Gao
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Mengmeng Xiang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yang Feng
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jie Wang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinhua Xu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Yilun Wang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jun Liang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Peng X, Li H, Zhu L, Zhao S, Li Z, Li S, DongtingWu, Chen J, Zheng S, Su W. Single-cell sequencing of the retina shows that LDHA regulates pathogenesis of autoimmune uveitis. J Autoimmun 2024; 143:103160. [PMID: 38160538 DOI: 10.1016/j.jaut.2023.103160] [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: 10/31/2023] [Revised: 11/29/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024]
Abstract
Autoimmune uveitis (AU) is a severe disorder causing poor vision and blindness. However, the cellular dynamics and pathogenic mechanisms underlying retinal injury in uveitis remain unclear. In this study, single-cell RNA sequencing of the retina and cervical draining lymph nodes in experimental autoimmune uveitis mice was conducted to identify the cellular spatiotemporal dynamics and upregulation of the glycolysis-related gene LDHA. Suppression of LDHA can rescue the imbalance of T effector (Teff) cells/T regulator (Treg) cells under inflammation via downregulation of the glycolysis-PI3K signaling circuit and inhibition of the migration of CXCR4+ Teff cells towards retinal tissue. Furthermore, LDHA and CXCR4 are upregulated in the peripheral blood mononuclear cells of Vogt-Koyanagi-Harada patients. The LDHA inhibitor suppresses CD4+ T cell proliferation in humans. Therefore, our data indicate that the autoimmune environment of uveitis regulates Teff cell accumulation in the retina via glycolysis-associated LDHA. Modulation of this target may provide a novel therapeutic strategy for treating AU.
Collapse
Affiliation(s)
- Xuening Peng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - He Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Lei Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Sichen Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Zhaohuai Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Si Li
- Sun Yat-sen University, Guangzhou 510060, China
| | - DongtingWu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | | | - Songguo Zheng
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Jiaotong University School of Medicine, 201600, Shanghai, China.
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China.
| |
Collapse
|
12
|
Chen Y, Liang R, Shi X, Shen R, Liu L, Liu Y, Xue Y, Guo X, Dang J, Zeng D, Huang F, Sun J, Zhang J, Wang J, Olsen N, August A, Huang W, Pan Y, Zheng SG. Targeting kinase ITK treats autoimmune arthritis via orchestrating T cell differentiation and function. Biomed Pharmacother 2023; 169:115886. [PMID: 37992572 DOI: 10.1016/j.biopha.2023.115886] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/06/2023] [Accepted: 11/13/2023] [Indexed: 11/24/2023] Open
Abstract
IL-2 inducible T cell kinase (ITK) is critical in T helper subset differentiation and its inhibition has been suggested for the treatment of T cell-mediated inflammatory diseases. T follicular helper (Tfh), Th17 and regulatory T cells (Treg) also play important roles in the development of rheumatoid arthritis (RA), while the role of ITK in the development of RA and the intricate balance between effector T and regulatory T cells remains unclear. Here, we found that CD4+ T cells from RA patients presented with an elevated ITK activation. ITK inhibitor alleviated existing collagen-induced arthritis (CIA) and reduced antigen specific antibody production. Blocking ITK kinase activity interferes Tfh cell generation. Moreover, ITK inhibitor effectively rebalances Th17 and Treg cells by regulating Foxo1 translocation. Furthermore, we identified dihydroartemisinin (DHA) as a potential ITK inhibitor, which could inhibit PLC-γ1 phosphorylation and the progression of CIA by rebalancing Th17 and Treg cells. Out data imply that ITK activation is upregulated in RA patients, and therefore blocking ITK signal may provide an effective strategy to treat RA patients and highlight the role of ITK on the Tfh induction and RA progression.
Collapse
Affiliation(s)
- Ye Chen
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, PR China; Department of Immunology, School of Cell and Gene Therapy, Songjiang Research Institute, Shanghai Songjiang District Central Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 201600, China
| | - Rongzhen Liang
- Department of Immunology, School of Cell and Gene Therapy, Songjiang Research Institute, Shanghai Songjiang District Central Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 201600, China
| | - Xiaoyi Shi
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, PR China
| | - Rong Shen
- Department of Geriatrics, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, PR China
| | - Liu Liu
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250000, PR China
| | - Yan Liu
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, PR China
| | - Youqiu Xue
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, PR China
| | - Xinghua Guo
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, PR China
| | - Junlong Dang
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, PR China
| | - Donglan Zeng
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, PR China
| | - Feng Huang
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, PR China
| | - Jianbo Sun
- The first Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523710, China
| | - Jingwen Zhang
- Department of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Julie Wang
- Department of Immunology, School of Cell and Gene Therapy, Songjiang Research Institute, Shanghai Songjiang District Central Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 201600, China
| | - Nancy Olsen
- Division of Rheumatology, Department of Medicine at the Penn State University Hershey Medical Center, Hershey, PA, USA
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Weishan Huang
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA; Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Yunfeng Pan
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, PR China.
| | - Song Guo Zheng
- Department of Immunology, School of Cell and Gene Therapy, Songjiang Research Institute, Shanghai Songjiang District Central Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 201600, China.
| |
Collapse
|
13
|
Zickert A, Janković MR, Malmström V, Chemin K, Gunnarsson I. Occurrence and localization of FOXP3 + cells in kidney biopsies in lupus nephritis and ANCA-associated vasculitis. Clin Rheumatol 2023; 42:2889-2895. [PMID: 37368057 PMCID: PMC10497686 DOI: 10.1007/s10067-023-06676-8] [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: 03/19/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/28/2023]
Abstract
The study aims to increase the understanding regarding the role of regulatory T cells (Tregs) in lupus nephritis (LN) and ANCA-associated vasculitis (AAV) by comparing their localization in renal tissue and changes following immunosuppressive therapy. Kidney biopsies from 12 patients with LN and 7 patients with AAV were examined. Kidney biopsies had been performed both at active disease and following immunosuppressive treatment. Clinical data was collected at both biopsy occasions. Expression of Forkhead Box P 3 (Foxp3) in renal tissue was assessed by immunohistochemistry. An arbitrary scale was used to estimate the number of Foxp3+ cells. In LN, 8/12 (67%) had positive tissue staining for Foxp3 at baseline, most pronounced in inflammatory infiltrates, but also interstitially and in a peri-glomerular pattern. At second biopsies, after immunosuppressive treatment, 4/12 (33%) still had detectable Foxp3+ cells, found in persisting inflammatory infiltrates and some in the interstitium. Patients with a good clinical response to treatment had high grade of Foxp3+ cells in first biopsies. In AAV, only 2/7 (29%) had positive staining for Foxp3 at baseline, in inflammatory infiltrates and to a lesser extent in the interstitium, despite large areas of inflammatory infiltrates in all patients. At follow-up, 2/7 (29%) biopsies were positive for Foxp3. Our data show a higher presence of Foxp3+ cells in renal tissue from LN patients compared to AAV, suggesting that Tregs may be differently involved in the control of inflammatory mechanisms in these diseases. These findings could have further implication for therapeutic approaches aiming at restoring the immunological tolerance. Key Points • Foxp3+-cells are present in larger amount in renal tissue in lupus nephritis vs. ANCA-associated vasculitis. • Our data suggest that Foxp3+ regulatory T cells are involved in the control of inflammatory processes in lupus nephritis.
Collapse
Affiliation(s)
- Agneta Zickert
- Department of Medicine, Rheumatology Unit, Karolinska Institutet, Solna, Sweden.
- Rheumatology Unit, S-171 76, Karolinska University Hospital, Stockholm, Sweden.
| | - Marija Ratković Janković
- Department of Nephrology, Clinic of Pediatrics, University Clinical Center Nis, Medical Faculty, University of Nis, Nis, Serbia
| | - Vivianne Malmström
- Department of Medicine, Rheumatology Unit, Karolinska Institutet, Solna, Sweden
| | - Karine Chemin
- Department of Medicine, Rheumatology Unit, Karolinska Institutet, Solna, Sweden
| | - Iva Gunnarsson
- Department of Medicine, Rheumatology Unit, Karolinska Institutet, Solna, Sweden
- Rheumatology Unit, S-171 76, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
14
|
Khatun A, Wu X, Qi F, Gai K, Kharel A, Kudek MR, Fraser L, Ceicko A, Kasmani MY, Majnik A, Burns R, Chen Y, Salzman N, Taparowsky EJ, Fang D, Williams CB, Cui W. BATF is Required for Treg Homeostasis and Stability to Prevent Autoimmune Pathology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206692. [PMID: 37587835 PMCID: PMC10558681 DOI: 10.1002/advs.202206692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 07/17/2023] [Indexed: 08/18/2023]
Abstract
Regulatory T (Treg) cells are inevitable to prevent deleterious immune responses to self and commensal microorganisms. Treg function requires continuous expression of the transcription factor (TF) FOXP3 and is divided into two major subsets: resting (rTregs) and activated (aTregs). Continuous T cell receptor (TCR) signaling plays a vital role in the differentiation of aTregs from their resting state, and in their immune homeostasis. The process by which Tregs differentiate, adapt tissue specificity, and maintain stable phenotypic expression at the transcriptional level is still inconclusivei. In this work, the role of BATF is investigated, which is induced in response to TCR stimulation in naïve T cells and during aTreg differentiation. Mice lacking BATF in Tregs developed multiorgan autoimmune pathology. As a transcriptional regulator, BATF is required for Treg differentiation, homeostasis, and stabilization of FOXP3 expression in different lymphoid and non-lymphoid tissues. Epigenetically, BATF showed direct regulation of Treg-specific genes involved in differentiation, maturation, and tissue accumulation. Most importantly, FOXP3 expression and Treg stability require continuous BATF expression in Tregs, as it regulates demethylation and accessibility of the CNS2 region of the Foxp3 locus. Considering its role in Treg stability, BATF should be considered an important therapeutic target in autoimmune disease.
Collapse
Affiliation(s)
- Achia Khatun
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
- Versiti Blood Research InstituteVersiti WisconsinMilwaukeeWI53226USA
| | - Xiaopeng Wu
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
- Versiti Blood Research InstituteVersiti WisconsinMilwaukeeWI53226USA
| | - Fu Qi
- Children's Mercy Hospital in Kansas City2401 Gillham RdKansas CityMO64108USA
| | - Kexin Gai
- Department of PathologyFeinberg School of MedicineNorthwestern University303 E Chicago AveChicagoIL60611USA
| | - Arjun Kharel
- Department of PathologyFeinberg School of MedicineNorthwestern University303 E Chicago AveChicagoIL60611USA
| | - Matthew R. Kudek
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
- Versiti Blood Research InstituteVersiti WisconsinMilwaukeeWI53226USA
- Department of PediatricsMedical College of Wisconsin8701 Watertown Plank RoadMilwaukeeWI53226USA
| | - Lisa Fraser
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
| | - Ashley Ceicko
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
| | - Moujtaba Y. Kasmani
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
- Versiti Blood Research InstituteVersiti WisconsinMilwaukeeWI53226USA
| | - Amber Majnik
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
- Children's Mercy Hospital in Kansas City2401 Gillham RdKansas CityMO64108USA
| | - Robert Burns
- Versiti Blood Research InstituteVersiti WisconsinMilwaukeeWI53226USA
| | - Yi‐Guang Chen
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
- Max McGee National Research Center for Juvenile DiabetesMedical College of Wisconsin8701 Watertown Plank RoadMilwaukeeWI53226USA
| | - Nita Salzman
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
- Department of PediatricsMedical College of Wisconsin8701 Watertown Plank RoadMilwaukeeWI53226USA
| | | | - Dayu Fang
- Department of PathologyFeinberg School of MedicineNorthwestern University303 E Chicago AveChicagoIL60611USA
| | - Calvin B. Williams
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
- Department of PediatricsMedical College of Wisconsin8701 Watertown Plank RoadMilwaukeeWI53226USA
| | - Weiguo Cui
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI53226USA
- Versiti Blood Research InstituteVersiti WisconsinMilwaukeeWI53226USA
- Department of PathologyFeinberg School of MedicineNorthwestern University303 E Chicago AveChicagoIL60611USA
| |
Collapse
|
15
|
Sun H, Lee HS, Kim SHJ, Fernandes de Lima M, Gingras AR, Du Q, McLaughlin W, Ablack J, Lopez-Ramirez MA, Lagarrigue F, Fan Z, Chang JT, VanDyke D, Spangler JB, Ginsberg MH. IL-2 can signal via chemokine receptors to promote regulatory T cells' suppressive function. Cell Rep 2023; 42:112996. [PMID: 37598341 PMCID: PMC10564087 DOI: 10.1016/j.celrep.2023.112996] [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: 09/07/2022] [Revised: 04/18/2023] [Accepted: 08/02/2023] [Indexed: 08/22/2023] Open
Abstract
Canonical interleukin-2 (IL-2) signaling via the high-affinity CD25-containing IL-2 receptor-Janus kinase (JAK)1,3-signal transducer and activator of transcription 5 (STAT5) pathway is essential for development and maintenance of CD4+CD25HiFoxp3+ regulatory T cells (Tregs) that support immune homeostasis. Here, we report that IL-2 signaling via an alternative CD25-chemokine receptor pathway promotes the suppressive function of Tregs. Using an antibody against CD25 that biases IL-2 signaling toward this alternative pathway, we establish that this pathway increases the suppressive activity of Tregs and ameliorates murine experimental autoimmune encephalomyelitis (EAE). Furthermore, heparan sulfate, an IL-2-binding element of cell surfaces and extracellular matrix, or an engineered IL-2 immunocytokine can also direct IL-2 signaling toward this alternative pathway. Overall, these data reveal a non-canonical mechanism for IL-2 signaling that promotes suppressive functions of Tregs, further elucidates how IL-2 supports immune homeostasis, and suggests approaches to promote or suppress Treg functions.
Collapse
Affiliation(s)
- Hao Sun
- University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Ho-Sup Lee
- University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Sarah Hyun-Ji Kim
- University of California San Diego School of Medicine, La Jolla, CA, USA
| | | | | | - Qinyi Du
- University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Wilma McLaughlin
- University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Jailail Ablack
- University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Miguel A Lopez-Ramirez
- University of California San Diego School of Medicine, La Jolla, CA, USA; Department of Pharmacology, University of California, San Diego, La Jolla, La Jolla, CA, USA
| | | | - Zhichao Fan
- University of Connecticut School of Medicine, Farmington, CT, USA
| | - John T Chang
- University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Derek VanDyke
- Department of Chemical & Biomolecular Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jamie B Spangler
- Department of Chemical & Biomolecular Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Mark H Ginsberg
- University of California San Diego School of Medicine, La Jolla, CA, USA.
| |
Collapse
|
16
|
Chen J, Sun N, Li F, Li H, Tian J, Zheng S, Zhang L, Wang H, Luo Y. Carnosol Alleviates Collagen-Induced Arthritis by Inhibiting Th17-Mediated Immunity and Favoring Suppressive Activity of Regulatory T Cells. BIOMED RESEARCH INTERNATIONAL 2023; 2023:1179973. [PMID: 37415927 PMCID: PMC10322527 DOI: 10.1155/2023/1179973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/22/2023] [Accepted: 06/05/2023] [Indexed: 07/08/2023]
Abstract
Current approaches are incurable for rheumatoid arthritis (RA). Regulatory T (Treg) cells and T helper cells (Th1 and Th17) are crucial in controlling the process of RA, which is characterized by inflammatory cell infiltration and bone destruction. Carnosol is an orthodiphenolic diterpene that has been extensively applied in traditional medicine for the treatment of multiple autoimmune and inflammatory diseases. Herein, we indicate that administration of carnosol dramatically alleviated the severity of collagen-induced arthritis (CIA) model with a decreased clinical score and inflammation reduction. Cellular mechanistically, carnosol inhibits the Th17 cell differentiation and maintains Treg cell suppressive function in vitro and in vivo. Meanwhile, it also restrains Treg cells from transdifferentiation into Th17 cells under inflammatory milieu. Furthermore, carnosol modulates the function of Th17 and Treg cells possibly via limiting IL-6R (CD126) expression. Collectively, our results suggest that carnosol can alleviate the severity of CIA via hiding Th17 cell differentiation and maintain the stability of Treg cells. Administration of carnosol can be applied as a potential therapy for patients with RA.
Collapse
Affiliation(s)
- Jun Chen
- The Department of Neurology, The First Hospital of Lanzhou University, Lanzhou, 730000 Gansu, China
| | - Nianzhe Sun
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000 Gansu, China
| | - Fuhan Li
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000 Gansu, China
| | - Haolin Li
- Rheumatic Bone Disease Center, Gansu Provincial Hospital of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, 730000 Gansu, China
| | - Jiale Tian
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000 Gansu, China
| | - Songguo Zheng
- Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Li Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000 Gansu, China
| | - Haidong Wang
- Rheumatic Bone Disease Center, Gansu Provincial Hospital of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, 730000 Gansu, China
| | - Yang Luo
- The Department of Neurology, The First Hospital of Lanzhou University, Lanzhou, 730000 Gansu, China
- Key Laboratory of Biotherapy and Regenerative Medicine, Lanzhou, 730000 Gansu, China
| |
Collapse
|
17
|
Sun L, Su Y, Jiao A, Wang X, Zhang B. T cells in health and disease. Signal Transduct Target Ther 2023; 8:235. [PMID: 37332039 PMCID: PMC10277291 DOI: 10.1038/s41392-023-01471-y] [Citation(s) in RCA: 209] [Impact Index Per Article: 104.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 06/20/2023] Open
Abstract
T cells are crucial for immune functions to maintain health and prevent disease. T cell development occurs in a stepwise process in the thymus and mainly generates CD4+ and CD8+ T cell subsets. Upon antigen stimulation, naïve T cells differentiate into CD4+ helper and CD8+ cytotoxic effector and memory cells, mediating direct killing, diverse immune regulatory function, and long-term protection. In response to acute and chronic infections and tumors, T cells adopt distinct differentiation trajectories and develop into a range of heterogeneous populations with various phenotype, differentiation potential, and functionality under precise and elaborate regulations of transcriptional and epigenetic programs. Abnormal T-cell immunity can initiate and promote the pathogenesis of autoimmune diseases. In this review, we summarize the current understanding of T cell development, CD4+ and CD8+ T cell classification, and differentiation in physiological settings. We further elaborate the heterogeneity, differentiation, functionality, and regulation network of CD4+ and CD8+ T cells in infectious disease, chronic infection and tumor, and autoimmune disease, highlighting the exhausted CD8+ T cell differentiation trajectory, CD4+ T cell helper function, T cell contributions to immunotherapy and autoimmune pathogenesis. We also discuss the development and function of γδ T cells in tissue surveillance, infection, and tumor immunity. Finally, we summarized current T-cell-based immunotherapies in both cancer and autoimmune diseases, with an emphasis on their clinical applications. A better understanding of T cell immunity provides insight into developing novel prophylactic and therapeutic strategies in human diseases.
Collapse
Affiliation(s)
- Lina Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Yanhong Su
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Anjun Jiao
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Xin Wang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China.
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China.
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China.
| |
Collapse
|
18
|
Zhu Y, Chang D. Interactions between the lung microbiome and host immunity in chronic obstructive pulmonary disease. Chronic Dis Transl Med 2023; 9:104-121. [PMID: 37305112 PMCID: PMC10249200 DOI: 10.1002/cdt3.66] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/01/2023] [Accepted: 03/15/2023] [Indexed: 04/07/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common chronic respiratory disease and the third leading cause of death worldwide. Developments in next-generation sequencing technology have improved microbiome analysis, which is increasingly recognized as an important component of disease management. Similar to the gut, the lung is a biosphere containing billions of microbial communities. The lung microbiome plays an important role in regulating and maintaining the host immune system. The microbiome composition, metabolites of microorganisms, and the interactions between the lung microbiome and the host immunity profoundly affect the occurrence, development, treatment, and prognosis of COPD. In this review, we drew comparisons between the lung microbiome of healthy individuals and that of patients with COPD. Furthermore, we summarize the intrinsic interactions between the host and the overall lung microbiome, focusing on the underlying mechanisms linking the microbiome to the host innate and adaptive immune response pathways. Finally, we discuss the possibility of using the microbiome as a biomarker to determine the stage and prognosis of COPD and the feasibility of developing a novel, safe, and effective therapeutic target.
Collapse
Affiliation(s)
- Yixing Zhu
- Graduate School of The PLA General HospitalBeijingChina
| | - De Chang
- Department of Respiratory and Critical Care Medicine, Eighth Medical Center, Department of Respiratory and Critical Care Seventh Medical CenterChinese PLA General HospitalBeijingChina
| |
Collapse
|
19
|
Paparella R, Menghi M, Micangeli G, Leonardi L, Profeta G, Tarani F, Petrella C, Ferraguti G, Fiore M, Tarani L. Autoimmune Polyendocrine Syndromes in the Pediatric Age. CHILDREN 2023; 10:children10030588. [PMID: 36980146 PMCID: PMC10047132 DOI: 10.3390/children10030588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/13/2023] [Accepted: 03/17/2023] [Indexed: 03/22/2023]
Abstract
Autoimmune polyendocrine syndromes (APSs) encompass a heterogeneous group of rare diseases characterized by autoimmune activity against two or more endocrine or non-endocrine organs. Three types of APSs are reported, including both monogenic and multifactorial, heterogeneous disorders. The aim of this manuscript is to present the main clinical and epidemiological characteristics of APS-1, APS-2, and IPEX syndrome in the pediatric age, describing the mechanisms of autoimmunity and the currently available treatments for these rare conditions.
Collapse
Affiliation(s)
- Roberto Paparella
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Michela Menghi
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Ginevra Micangeli
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Lucia Leonardi
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Giovanni Profeta
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Francesca Tarani
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Carla Petrella
- Institute of Biochemistry and Cell Biology, IBBC-CNR, 00185 Rome, Italy
| | - Giampiero Ferraguti
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Marco Fiore
- Institute of Biochemistry and Cell Biology, IBBC-CNR, 00185 Rome, Italy
- Correspondence: (M.F.); (L.T.)
| | - Luigi Tarani
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
- Correspondence: (M.F.); (L.T.)
| |
Collapse
|
20
|
The link between rheumatic disorders and inborn errors of immunity. EBioMedicine 2023; 90:104501. [PMID: 36870198 PMCID: PMC9996386 DOI: 10.1016/j.ebiom.2023.104501] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/11/2022] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
Inborn errors of immunity (IEIs) are immunological disorders characterized by variable susceptibility to infections, immune dysregulation and/or malignancies, as a consequence of damaging germline variants in single genes. Though initially identified among patients with unusual, severe or recurrent infections, non-infectious manifestations and especially immune dysregulation in the form of autoimmunity or autoinflammation can be the first or dominant phenotypic aspect of IEIs. An increasing number of IEIs causing autoimmunity or autoinflammation, including rheumatic disease have been reported over the last decade. Despite their rarity, identification of those disorders provided insight into the pathomechanisms of immune dysregulation, which may be relevant for understanding the pathogenesis of systemic rheumatic disorders. In this review, we present novel IEIs primarily causing autoimmunity or autoinflammation along with their pathogenic mechanisms. In addition, we explore the likely pathophysiological and clinical relevance of IEIs in systemic rheumatic disorders.
Collapse
|
21
|
Qiuping L, Pan P, Zhenzhen L, Zhen Z, Xuezhu Z, Shuting L. Acupuncture regulates the Th17/Treg balance and improves cognitive deficits in a rat model of vascular dementia. Heliyon 2023; 9:e13346. [PMID: 36816326 PMCID: PMC9929319 DOI: 10.1016/j.heliyon.2023.e13346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 12/20/2022] [Accepted: 01/25/2023] [Indexed: 02/02/2023] Open
Abstract
Objective The present study was developed to explore the impact of acupuncture on the Th17/Treg balance in the brain and the periphery and associated changes in cognitive deficits in a rat model of vascular dementia (VD). Methods Male Wistar rats (8 weeks old) were randomly assigned to sham-operated (Gs, n = 10), and operation (n = 30) groups. A VD model was established for all rats in the operation group via the permanent bilateral occlusion of the common carotid artery. Behavioral screening of these rats was conducted via a hidden platform trial at 2 months post-operation. These operation group rats were then further subdivided into impaired (Gi) and acupuncture (Ga) groups (n = 10/group). Acupuncture was performed over a 21-day period for rats in the Ga group. A Morris water maze (MWM) test was used to assess cognitive function for rats in all groups. Flow cytometry and fluorescent staining were used to detect Th17 and Treg cells in samples from these animals based on IL-17/FoxP3 or CD4+FoxP3+/CD4+RORγt+ staining profiles. Results Relative to the Gs group, escape latency values for rats in the Gi group were significantly increased. Following treatment, rats in the Ga group exhibited significant reductions in escape latency values as compared to rats in the Gi group (P < 0.05). The relative Treg proportion in the peripheral blood and spleen additionally trended upwards in these Ga rats as compared to those in the Gi group (P > 0.05), whereas the frequency of Th17 cells in the peripheral blood and spleen of Ga group rats trended downward relative to the Gi group (P > 0.05). Significantly fewer CD4+RORγt+ and RORγt+ cells were detected in the Ga group relative to the Gi group, whereas CD4+FoxP3+ and FoxP3+ cell counts were increased (P < 0.01). Conclusion In summary, VD model rats exhibited dysregulated Th17/Treg homeostasis. Acupuncture treatment was sufficient to reduce the frequency and numbers of Th17 cells in these animals while increasing Treg cell levels, thereby alleviating cognitive deficits with respect to both spatial learning and memory impairment. Consequently, the therapeutic benefits of such acupuncture treatment may be attributable to the regulation of the Th17/Treg balance and associated improvements in cognitive function.
Collapse
Affiliation(s)
- Liu Qiuping
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300380, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300380, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Pan Pan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300380, China
- Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan Province, 650500, China
| | - Ling Zhenzhen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300380, China
- Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Zhang Zhen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300380, China
- Weifang Traditional Chinese Hospital, Shandong Province, 261031, China
| | - Zhang Xuezhu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300380, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300380, China
| | - Li Shuting
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300380, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300380, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| |
Collapse
|
22
|
Giancotta C, Colantoni N, Pacillo L, Santilli V, Amodio D, Manno EC, Cotugno N, Rotulo GA, Rivalta B, Finocchi A, Cancrini C, Diociaiuti A, El Hachem M, Zangari P. Tailored treatments in inborn errors of immunity associated with atopy (IEIs-A) with skin involvement. Front Pediatr 2023; 11:1129249. [PMID: 37033173 PMCID: PMC10073443 DOI: 10.3389/fped.2023.1129249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/03/2023] [Indexed: 04/11/2023] Open
Abstract
Inborn errors of immunity associated with atopy (IEIs-A) are a group of inherited monogenic disorders that occur with immune dysregulation and frequent skin involvement. Several pathways are involved in the pathogenesis of these conditions, including immune system defects, alterations of skin barrier and metabolism perturbations. Current technological improvements and the higher accessibility to genetic testing, recently allowed the identification of novel molecular pathways involved in IEIs-A, also informing on potential tailored therapeutic strategies. Compared to other systemic therapy for skin diseases, biologics have the less toxic and the best tolerated profile in the setting of immune dysregulation. Here, we review IEIs-A with skin involvement focusing on the tailored therapeutic approach according to their pathogenetic mechanism.
Collapse
Affiliation(s)
- Carmela Giancotta
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Nicole Colantoni
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Lucia Pacillo
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
- Department of Systems Medicine, University of Tor Vergata, Rome, Italy
| | - Veronica Santilli
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Donato Amodio
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Emma Concetta Manno
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Nicola Cotugno
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
- Department of Systems Medicine, University of Tor Vergata, Rome, Italy
| | - Gioacchino Andrea Rotulo
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Beatrice Rivalta
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
- Department of Systems Medicine, University of Tor Vergata, Rome, Italy
| | - Andrea Finocchi
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
- Department of Systems Medicine, University of Tor Vergata, Rome, Italy
| | - Caterina Cancrini
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
- Department of Systems Medicine, University of Tor Vergata, Rome, Italy
| | - Andrea Diociaiuti
- Dermatology Unit and Genodermatosis Unit, Genetics and Rare Diseases Research Division, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - May El Hachem
- Dermatology Unit and Genodermatosis Unit, Genetics and Rare Diseases Research Division, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Paola Zangari
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
- Correspondence: Paola Zangari
| |
Collapse
|
23
|
Klaus T, Wilson AS, Vicari E, Hadaschik E, Klein M, Helbich SSC, Kamenjarin N, Hodapp K, Schunke J, Haist M, Butsch F, Probst HC, Enk AH, Mahnke K, Waisman A, Bednarczyk M, Bros M, Bopp T, Grabbe S. Impaired Treg-DC interactions contribute to autoimmunity in leukocyte adhesion deficiency type 1. JCI Insight 2022; 7:162580. [PMID: 36346673 PMCID: PMC9869970 DOI: 10.1172/jci.insight.162580] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
Leukocyte adhesion deficiency type 1 (LAD-1) is a rare disease resulting from mutations in the gene encoding for the common β-chain of the β2-integrin family (CD18). The most prominent clinical symptoms are profound leukocytosis and high susceptibility to infections. Patients with LAD-1 are prone to develop autoimmune diseases, but the molecular and cellular mechanisms that result in coexisting immunodeficiency and autoimmunity are still unresolved. CD4+FOXP3+ Treg are known for their essential role in preventing autoimmunity. To understand the role of Treg in LAD-1 development and manifestation of autoimmunity, we generated mice specifically lacking CD18 on Treg (CD18Foxp3), resulting in defective LFA-1 expression. Here, we demonstrate a crucial role of LFA-1 on Treg to maintain immune homeostasis by modifying T cell-DC interactions and CD4+ T cell activation. Treg-specific CD18 deletion did not impair Treg migration into extralymphatic organs, but it resulted in shorter interactions of Treg with DC. In vivo, CD18Foxp3 mice developed spontaneous hyperplasia in lymphatic organs and diffuse inflammation of the skin and in multiple internal organs. Thus, LFA-1 on Treg is required for the maintenance of immune homeostasis.
Collapse
Affiliation(s)
- Tanja Klaus
- Department of Dermatology,,Research Center for Immunotherapy, and
| | - Alicia S. Wilson
- Research Center for Immunotherapy, and,Institute of Immunology, University of Mainz Medical Center, Mainz, Germany
| | - Elisabeth Vicari
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany
| | - Eva Hadaschik
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany.,Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Matthias Klein
- Research Center for Immunotherapy, and,Institute of Immunology, University of Mainz Medical Center, Mainz, Germany
| | | | - Nadine Kamenjarin
- Research Center for Immunotherapy, and,Institute of Immunology, University of Mainz Medical Center, Mainz, Germany
| | - Katrin Hodapp
- Research Center for Immunotherapy, and,Institute of Immunology, University of Mainz Medical Center, Mainz, Germany
| | - Jenny Schunke
- Department of Dermatology,,Research Center for Immunotherapy, and
| | - Maximilian Haist
- Department of Dermatology,,Research Center for Immunotherapy, and
| | | | - Hans Christian Probst
- Research Center for Immunotherapy, and,Institute of Immunology, University of Mainz Medical Center, Mainz, Germany
| | - Alexander H. Enk
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany
| | - Karsten Mahnke
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany
| | - Ari Waisman
- Research Center for Immunotherapy, and,Institute for Molecular Medicine, University of Mainz Medical Center, Mainz, Germany
| | | | - Matthias Bros
- Department of Dermatology,,Research Center for Immunotherapy, and
| | - Tobias Bopp
- Research Center for Immunotherapy, and,Institute of Immunology, University of Mainz Medical Center, Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology,,Research Center for Immunotherapy, and
| |
Collapse
|
24
|
Hu X, Li F, Zeng J, Zhou Z, Wang Z, Chen J, Cao D, Hong Y, Huang L, Chen Y, Xu J, Dong F, Yu R, Zheng H. Noninvasive Low-Frequency Pulsed Focused Ultrasound Therapy for Rheumatoid Arthritis in Mice. RESEARCH (WASHINGTON, D.C.) 2022; 2022:0013. [PMID: 39290964 PMCID: PMC11407525 DOI: 10.34133/research.0013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 10/31/2022] [Indexed: 10/16/2023]
Abstract
Rheumatoid arthritis (RA) is a common autoimmune disease characterized by chronic and progressive inflammation of the synovium. Focused ultrasound therapy is an increasingly attractive alternative for treating RA owing to its noninvasiveness; however, it remains unclear which immune subsets respond to ultrasound stimulation. In this study, we showed that spleen-targeted low-frequency pulsed focused ultrasound (LFPFU) effectively improved the severity of arthritis in an arthritis mouse model established in DBA/1J mice. Additionally, we performed in-depth immune profiling of spleen samples from RA mice, RA mice that underwent ultrasound therapy, and healthy controls using mass cytometry along with extensive antibody panels and identified the immune composition of 14 cell populations, including CD4+/CD8+ T cells, B cells, natural killer cells, and dendritic cells. Moreover, multidimensional analysis according to cell-surface markers and phenotypes helped in identifying 4 and 5 cell subpopulations among T and myeloid cells, respectively, with 6 T cell subsets and 3 myeloid cell subsets responsive to ultrasound therapy among the 3 groups. Of these cell subsets, CD8+ T cell subsets showed a unique response to ultrasound stimulation in RA mice. Specifically, CD8+ T cells show a noticeable correlation with the degree of arthritis progression and could serve as an indicator for spleen-focused ultrasound-based therapy. Furthermore, single-cell RNA sequencing of spleen cells revealed the importance of T, B, and myeloid cell populations in the anti-inflammatory pathway. These results elucidated the unique cell subsets and transcriptome of splenic cells responsive to LFPFU and demonstrated the potential of spleen-focused ultrasound stimulation in the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Xuqiao Hu
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen 518055, China
| | - Fei Li
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen 518055, China
| | - Jieying Zeng
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Zhenru Zhou
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Zhaoyang Wang
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Jing Chen
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Dongyan Cao
- Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Yifan Hong
- Institute of Molecular Physiology, Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, P.R. China
| | - Laixin Huang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen 518055, China
| | - Yongsheng Chen
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Jinfeng Xu
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Fajin Dong
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Rongmin Yu
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China
- Department of Pharmacology, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
- Biotechnological Institute of Chinese Materia Medica, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen 518055, China
| |
Collapse
|
25
|
Zhao X, Wang S, Wang S, Xie J, Cui D. mTOR signaling: A pivotal player in Treg cell dysfunction in systemic lupus erythematosus. Clin Immunol 2022; 245:109153. [DOI: 10.1016/j.clim.2022.109153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/03/2022]
|
26
|
A LILRB1 variant with a decreased ability to phosphorylate SHP-1 leads to autoimmune diseases. Sci Rep 2022; 12:15420. [PMID: 36104364 PMCID: PMC9474825 DOI: 10.1038/s41598-022-19334-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 08/29/2022] [Indexed: 11/24/2022] Open
Abstract
Inborn errors of immunity are known to cause not only immunodeficiencies and allergies but also autoimmunity. Leukocyte immunoglobulin-like receptor B1 (LILRB1) is a receptor on leukocytes playing a role in regulating immune responses. No phenotypes have been reported to be caused by germline mutations in LILRB1. We aimed to identify the causative variant in a three-generation family with nine members suffering from one of the three autoimmune diseases—Graves’ disease, Hashimoto's thyroiditis, or systemic lupus erythematosus. Whole-genome linkage study revealed a locus on chromosome 19q13.4 with the maximum LOD score of 2.71. Whole-exome sequencing identified a heterozygous missense variant, c.479G > A (p. G160E) in LILRB1, located within the chromosomal-linked region, in all nine affected members. The variant has never been previously reported. Jurkat cells transfected with the mutant LILRB1, compared with those with the wild-type LILRB1, showed decreased phosphorylation of both LILRB1 and its downstream protein, SHP-1. Flow cytometry was used to study immunophenotype and revealed that LILRB1 was significantly lower on the surface of activated regulatory T lymphocytes (Treg) cells of patients. Single-cell RNA sequencing showed substantially increased M1-like monocytes in peripheral blood mononuclear cells of affected individuals. This study, for the first time, implicates LILRB1 as a new disease gene for autoimmunity.
Collapse
|
27
|
Peripheral distributions of IL-4-producing CD4 + T cells and CD4 + CD25 + FoxP3 + T cells (Tregs) in rheumatoid arthritis patients with poor response to therapy are associated with HLA shared epitope alleles and ACPA status. Immunol Res 2022; 70:481-492. [PMID: 35445929 DOI: 10.1007/s12026-022-09281-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 04/12/2022] [Indexed: 11/05/2022]
Abstract
Specific profiling of CD4 + T cell subsets in the circulation and inflamed joints of rheumatoid arthritis (RA) patients may have therapeutic implications. This study aimed to evaluate the peripheral distributions of Th2 and Treg cells in relation to HLA-shared epitope (SE) alleles and anti-cyclic citrullinated peptide antibody (ACPAs) status in patients with good response (GR) and poor response (PR) to treatment. The frequencies of IL-4-producing CD4 + T cells (Th2) and CD4 + CD25 + Foxp3 + T cells (Tregs) were determined by flow cytometry in 167 RA patients including 114 GR and 53 PR cases. CD4 + T cell subsets were also analyzed based on HLA-SE and ACPAs statuses. One hundred nine of 167 patients were positives for HLA-SE, 63.4% for ACPAs, 43.7% for SE/ACPAs and 14.9% were negatives for SE/ACPAs. Higher frequencies of Th2 (P = 0.001) and Treg cells (P = 0.03) were found in the patients versus controls. Increased and decreased frequencies of Th2 and Tregs cells were observed in the PR versus GR patients respectively (P = 0.003 and P = 0.004). Higher proportions of Th2 cells were observed in the SE+RA versus SE-RA (P = 0.001), in ACPA+RA versus ACPA-RA (P = 0.005) and in the SE+ACPA+RA versus SE-ACPA-RA patients (P = 0.002). Treg cells frequencies decreased in the SE+RA versus SE-RA (P = 0.03) and in SE+ACPA+RA versus SE-ACPA-RA (P = 0.02). ACPA+GR and SE+PR patients showed higher proportions of Th2 cells than ACPA-GR and SE-PR patients respectively (P = 0.02 and P = 0.01). Analysis of the CD4 + T cell subsets profiles in conjunction with genetic background and autoantibodies patterns can be useful for precise therapeutic response monitoring in the RA patients.
Collapse
|
28
|
Liu HY, Shi ZY, Fan D, Zhang SX, Wu LX, Lu KY, Yang SY, Li WT, kang JF, Li CH, Cheng ZH, Xue Y, Wu ZF, Li XF, Li SJ. Absolute reduction in peripheral regulatory T cells in patients with Graves’ disease and post-treatment recovery. Mol Immunol 2022; 144:49-57. [DOI: 10.1016/j.molimm.2022.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 01/19/2022] [Accepted: 02/06/2022] [Indexed: 11/25/2022]
|
29
|
Jin K, Parreau S, Warrington KJ, Koster MJ, Berry GJ, Goronzy JJ, Weyand CM. Regulatory T Cells in Autoimmune Vasculitis. Front Immunol 2022; 13:844300. [PMID: 35296082 PMCID: PMC8918523 DOI: 10.3389/fimmu.2022.844300] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 01/28/2022] [Indexed: 12/14/2022] Open
Abstract
Blood vessels are indispensable for host survival and are protected from inappropriate inflammation by immune privilege. This protection is lost in patients with autoimmune vasculitides, a heterogeneous group of diseases causing damage to arteries, arterioles, and capillaries. Vasculitis leads to vascular wall destruction and/or luminal occlusion, resulting in hemorrhage and tissue ischemia. Failure in the quantity and quality of immunosuppressive regulatory T cells (Treg) has been implicated in the breakdown of the vascular immune privilege. Emerging data suggest that Treg deficiencies are disease-specific, affecting distinct pathways in distinct vasculitides. Mechanistic studies have identified faulty CD8+ Tregs in Giant Cell Arteritis (GCA), a vasculitis of the aorta and the large aortic branch vessels. Specifically, aberrant signaling through the NOTCH4 receptor expressed on CD8+ Treg cells leads to rerouting of intracellular vesicle trafficking and failure in the release of immunosuppressive exosomes, ultimately boosting inflammatory attack to medium and large arteries. In Kawasaki’s disease, a medium vessel vasculitis targeting the coronary arteries, aberrant expression of miR-155 and dysregulated STAT5 signaling have been implicated in undermining CD4+ Treg function. Explorations of mechanisms leading to insufficient immunosuppression and uncontrolled vascular inflammation hold the promise to discover novel therapeutic interventions that could potentially restore the immune privilege of blood vessels and pave the way for urgently needed innovations in vasculitis management.
Collapse
Affiliation(s)
- Ke Jin
- Department of Medicine, Mayo College of Medicine and Science, Rochester, MN, United States
| | - Simon Parreau
- Department of Medicine, Mayo College of Medicine and Science, Rochester, MN, United States
| | - Kenneth J. Warrington
- Department of Medicine, Mayo College of Medicine and Science, Rochester, MN, United States
| | - Matthew J. Koster
- Department of Medicine, Mayo College of Medicine and Science, Rochester, MN, United States
| | - Gerald J. Berry
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Jörg J. Goronzy
- Department of Medicine, Mayo College of Medicine and Science, Rochester, MN, United States
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Cornelia M. Weyand
- Department of Medicine, Mayo College of Medicine and Science, Rochester, MN, United States
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
- *Correspondence: Cornelia M. Weyand,
| |
Collapse
|
30
|
Castro C, Oyamada HAA, Cafasso MOSD, Lopes LM, Monteiro C, Sacramento PM, Alves-Leon SV, da Fontoura Galvão G, Hygino J, de Souza JPBM, Bento CAM. Elevated proportion of TLR2- and TLR4-expressing Th17-like cells and activated memory B cells was associated with clinical activity of cerebral cavernous malformations. J Neuroinflammation 2022; 19:28. [PMID: 35109870 PMCID: PMC8808981 DOI: 10.1186/s12974-022-02385-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 01/12/2022] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Recent evidences have suggested the involvement of toll-like receptor (TLR)-4 in the pathogenesis of cerebral cavernous malformations (CCM). Elevated frequency of TLR+T-cells has been associated with neurological inflammatory disorders. As T-cells and B-cells are found in CCM lesions, the objective of the present study was to evaluate the cytokine profile of T-cells expressing TLR2 and TLR4, as well as B-cell subsets, in asymptomatic (CCMAsympt) and symptomatic (CCMSympt) patients. METHODS For our study, the cytokine profile from TLR2+ and TLR4+ T-cell and B-cell subsets in CCMAsympt and CCMSympt patients was investigated using flow cytometry and ELISA. T-cells were stimulated in vitro with anti-CD3/anti-CD28 beads or TLR2 (Pam3C) and TLR4 (LPS) ligands. RESULTS CCMSymptc patients presented a higher frequency of TLR4+(CD4+ and CD8+) T-cells and greater density of TLR4 expression on these cells. With regard to the cytokine profile, the percentage of TLR2+ and TLR4+ Th17 cells was higher in CCMSympt patients. In addition, an elevated proportion of TLR4+ Tc-1 cells, as well as Tc-17 and Th17.1 cells expressing TLR2 and TLR4, was observed in the symptomatic patients. By contrast, the percentage of TLR4+ IL-10+CD4+ T cells was higher in the CCMAsympt group. Both Pam3C and LPS were more able to elevate the frequency of IL-6+CD4+T cells and Th17.1 cells in CCMSympt cell cultures. Furthermore, in comparison with asymptomatic patients, purified T-cells from the CCMSympt group released higher levels of Th17-related cytokines in response to Pam3C and, mainly, LPS, as well as after activation via TCR/CD28. Concerning the B-cell subsets, a higher frequency of memory and memory activated B-cells was observed in CCMSympt patients. CONCLUSIONS Our findings reveal an increase in circulating Th17/Tc-17 cell subsets expressing functional TLR2 and, mainly, TLR4 molecules, associated with an increase in memory B-cell subsets in CCM patients with clinical activity of the disease.
Collapse
Affiliation(s)
- Camilla Castro
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil
- Post-Graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Hugo A A Oyamada
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil
- Post-Graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcos Octávio S D Cafasso
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil
| | - Lana M Lopes
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil
- Post-Graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Clarice Monteiro
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil
- Post-Graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Priscila M Sacramento
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil
- Post-Graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Soniza Vieira Alves-Leon
- Post-Graduate Program in Neurology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Translational Neuroscience Laboratory (LabNet), University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gustavo da Fontoura Galvão
- Service of Neurosurgery, University Hospital of the Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Joana Hygino
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil
- Post-Graduate Program in Neurology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jorge Paes Barreto Marcondes de Souza
- Post-Graduate Program in Neurology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Service of Neurosurgery, University Hospital of the Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Post-Graduate Program of Surgical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cleonice A M Bento
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil.
- Post-Graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
- Post-Graduate Program in Neurology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
31
|
Francalanci P, Cafferata B, Alaggio R, de Angelis P, Diamanti A, Parente P, Granai M, Lazzi S. Pediatric autoimmune disorders with gastrointestinal expressions: from bench to bedside. Pathologica 2022; 114:32-39. [PMID: 34856606 PMCID: PMC9040544 DOI: 10.32074/1591-951x-339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 07/30/2021] [Indexed: 11/30/2022] Open
Abstract
The gastrointestinal (GI) tract may be involved in systemic autoimmune diseases or may be the target of organ-specific autoimmunity. Autoimmune enteropathy (AIE) is a rare disorder characterized by severe and protracted diarrhea, weight loss from malabsorption and immune-mediated damage to the intestinal mucosa, generally occurring in infants and young children, only rarely in adult. The salient histopathologic features of AIE are most prominent in the small intestine: villous blunting, crypt hyperplasia, mononuclear cell inflammatory expansion of the lamina propria with intraepithelial lymphocytosis, crypt apoptosis and absence of Paneth cells, goblet cells or both. Esophagus, stomach and colon are frequently also involved. Anti-enterocyte antibodies are identified in the majority of cases, and their presence, even if variable, can help confirming the diagnosis. The purpose of this review is to provide an overview of the latest immunological advances in AIE, as well as to offer a practical approach for histological diagnosis for 'general' pathologist.
Collapse
Affiliation(s)
- Paola Francalanci
- Unit of Pathology, Children’s Hospital Bambino Gesù IRCCS, Rome, Italy
| | - Barbara Cafferata
- Unit of Pathology, Children’s Hospital Bambino Gesù IRCCS, Rome, Italy
| | - Rita Alaggio
- Unit of Pathology, Children’s Hospital Bambino Gesù IRCCS, Rome, Italy
| | - Paola de Angelis
- Digestive Endoscopy and Surgery Unit, Children’s Hospital Bambino Gesù IRCCS, Rome, Italy
| | - Antonella Diamanti
- Gastroenterology and Nutritional Rehabilitation Unit, Children’s Hospital Bambino Gesù IRCCS, Rome, Italy
| | - Paola Parente
- Unit of Pathology, Fondazione IRCCS Ospedale Casa Sollievo Della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Massimo Granai
- Institut für Pathologie und Neuropathologie Abt. Allgemeine und Molekulare Pathologie und Pathologische Anatomie University of Tubingen, German
| | - Stefano Lazzi
- Section of Pathology, Department of Medical Biotechnology, University of Siena, Siena, Italy
| |
Collapse
|
32
|
Simsek A, Kizmaz MA, Cagan E, Dombaz F, Tezcan G, Asan A, Ibrahim Demir H, Haldun Bal S, Ermis DY, Dilektaslı AG, Kazak E, Halis Akalin E, Barbaros Oral H, Budak F. Assessment of CD39 expression in regulatory T cell subsets by disease severity in adult and juvenile COVID -19 cases. J Med Virol 2022; 94:2089-2101. [PMID: 35032133 PMCID: PMC9015412 DOI: 10.1002/jmv.27593] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 01/03/2022] [Accepted: 01/06/2022] [Indexed: 11/11/2022]
Abstract
COVID‐19 is a disease characterized by acute respiratory failure and is a major health problem worldwide. Here, we aimed to investigate the role of CD39 expression in Treg cell subsets in COVID‐19 immunopathogenesis and its relationship to disease severity. One hundred and ninety COVID‐19 patients (juveniles, adults) and 43 volunteers as healthy controls were enrolled in our study. Flow cytometric analysis was performed using a 10‐color monoclonal antibody panel from peripheral blood samples. In adult patients, CD39+ Tregs increased with disease severity. In contrast, CD39+ Tregs were decreased in juvenile patients in an age‐dependent manner. Overall, our study reveals an interesting profile of CD39‐expressing Tregs in adult and juvenile cases of COVID‐19. Our results provide a better understanding of the possible role of Tregs in the mechanism of immune response in COVID‐19 cases. CD39+ Tregs increased with disease severity in adult COVID‐19 cases. In addition, significant changes were also observed in other Treg subsets. Treg subsets in the juvenile COVID‐19 cases showed age‐related variability but were significantly lower than in the healthy control group. Consistent correlations were found between laboratory findings in adult COVID‐19 cases and Treg subsets.
Collapse
Affiliation(s)
- Abdurrahman Simsek
- Department of Immunology, Faculty of Medicine, Bursa Uludağ University, Bursa, Turkey.,Department of Immunology, Health Science Institute, Bursa Uludag University, Bursa, Turkey
| | - Muhammed Ali Kizmaz
- Department of Immunology, Faculty of Medicine, Bursa Uludağ University, Bursa, Turkey.,Department of Immunology, Health Science Institute, Bursa Uludag University, Bursa, Turkey
| | - Eren Cagan
- Department of Pediatric Infectious Diseases, University of Health Sciences, Bursa Yuksek Ihtisas Training and Research Hospital, Bursa, Turkey
| | - Fatma Dombaz
- Department of Immunology, Faculty of Medicine, Bursa Uludağ University, Bursa, Turkey.,Department of Immunology, Health Science Institute, Bursa Uludag University, Bursa, Turkey
| | - Gulcin Tezcan
- Department of Fundamental Science, Faculty of Dentistry, Bursa Uludağ University, Bursa, Turkey
| | - Ali Asan
- Department of Infectious Diseases and Clinical Microbiology, University of Health Sciences, Bursa Yuksek Ihtisas Training and Research Hospital, Bursa, Turkey
| | - H Ibrahim Demir
- Department of Immunology, Faculty of Medicine, Bursa Uludağ University, Bursa, Turkey.,Department of Immunology, Health Science Institute, Bursa Uludag University, Bursa, Turkey
| | - S Haldun Bal
- Department of Immunology, Faculty of Medicine, Bursa Uludağ University, Bursa, Turkey
| | - Digdem Yoyen Ermis
- Department of Immunology, Faculty of Medicine, Bursa Uludağ University, Bursa, Turkey
| | - Aslı Gorek Dilektaslı
- Department of Chest Diseases, Faculty of Medicine, Bursa Uludağ University, Bursa, Turkey
| | - Esra Kazak
- Department of Clinical Microbiology and Infection Diseases, Faculty of Medicine, Bursa Uludağ University, Bursa, Turkey
| | - E Halis Akalin
- Department of Clinical Microbiology and Infection Diseases, Faculty of Medicine, Bursa Uludağ University, Bursa, Turkey
| | - H Barbaros Oral
- Department of Immunology, Faculty of Medicine, Bursa Uludağ University, Bursa, Turkey
| | - Ferah Budak
- Department of Immunology, Faculty of Medicine, Bursa Uludağ University, Bursa, Turkey
| |
Collapse
|
33
|
Adriawan IR, Atschekzei F, Dittrich-Breiholz O, Garantziotis P, Hirsch S, Risser LM, Kosanke M, Schmidt RE, Witte T, Sogkas G. Novel aspects of regulatory T cell dysfunction as a therapeutic target in giant cell arteritis. Ann Rheum Dis 2022; 81:124-131. [PMID: 34583923 PMCID: PMC8762021 DOI: 10.1136/annrheumdis-2021-220955] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 09/15/2021] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Giant cell arteritis (GCA) is the most common primary vasculitis, preferentially affecting the aorta and its large-calibre branches. An imbalance between proinflammatory CD4+ T helper cell subsets and regulatory T cells (Tregs) is thought to be involved in the pathogenesis of GCA and Treg dysfunction has been associated with active disease. Our work aims to explore the aetiology of Treg dysfunction and the way it is affected by remission-inducing immunomodulatory regimens. METHODS A total of 41 GCA patients were classified into active disease (n=14) and disease in remission (n=27). GCA patients' and healthy blood donors' (HD) Tregs were sorted and subjected to transcriptome and phenotypic analysis. RESULTS Transcriptome analysis revealed 27 genes, which were differentially regulated between GCA-derived and HD-derived Tregs. Among those, we identified transcription factors, glycolytic enzymes and IL-2 signalling mediators. We confirmed the downregulation of forkhead box P3 (FOXP3) and interferon regulatory factor 4 (IRF4) at protein level and identified the ineffective induction of glycoprotein A repetitions predominant (GARP) and CD25 as well as the reduced T cell receptor (TCR)-induced calcium influx as correlates of Treg dysfunction in GCA. Inhibition of glycolysis in HD-derived Tregs recapitulated most identified dysfunctions of GCA Tregs, suggesting the central pathogenic role of the downregulation of the glycolytic enzymes. Separate analysis of the subgroup of tocilizumab-treated patients identified the recovery of the TCR-induced calcium influx and the Treg suppressive function to associate with disease remission. CONCLUSIONS Our findings suggest that low glycolysis and calcium signalling account for Treg dysfunction and inflammation in GCA.
Collapse
Affiliation(s)
- Ignatius Ryan Adriawan
- Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST, Hannover Medical School, Hannover, Germany
| | - Faranaz Atschekzei
- Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST, Hannover Medical School, Hannover, Germany
| | | | | | - Stefanie Hirsch
- Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | | | - Maike Kosanke
- Research Core Unit Genomics, Hannover Medical School, Hannover, Germany
| | - Reinhold Ernst Schmidt
- Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST, Hannover Medical School, Hannover, Germany
| | - Torsten Witte
- Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST, Hannover Medical School, Hannover, Germany
| | - Georgios Sogkas
- Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST, Hannover Medical School, Hannover, Germany
| |
Collapse
|
34
|
Shimojima Y, Kishida D, Ichikawa T, Takamatsu R, Nomura S, Sekijima Y. Oxidative Stress Promotes Instability of Regulatory T Cells in Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. Front Immunol 2021; 12:789740. [PMID: 34950150 PMCID: PMC8691772 DOI: 10.3389/fimmu.2021.789740] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/22/2021] [Indexed: 02/02/2023] Open
Abstract
We investigated the characteristics of regulatory T cells (Tregs), focusing on the relationship between their stability and reactive oxygen species (ROS), in antineutrophil cytoplasmic antibody-associated vasculitis (AAV). Intracellular expressions of effector cytokines, forkhead box protein 3 (FoxP3), ROS, phosphorylated mammalian target of rapamycin (mTOR), and sirtuin 1 (SIRT1) in Tregs from peripheral blood mononuclear cells (PBMCs) of patients with AAV and healthy controls (HC) were analyzed. The alterations in and functional ability of Tregs were compared before and after resveratrol (RVL) treatment of PBMCs in patients with AAV. Significantly higher expressions of interferon (IFN)-γ, interleukin (IL)-17, IL-4, ROS, and phosphorylated mTOR (pho-mTOR) and lower expression of SIRT1 in CD4+CD25+FoxP3+ cells were found in patients with AAV than in the HC. FoxP3 expression in CD4+CD25+ cells and suppressive function of Tregs were significantly lower in patients with AAV than in the HC. Tregs after RVL treatment demonstrated significant decreases in IFN-γ, ROS, and pho-mTOR levels and increases in FoxP3, SIRT1 levels, and functional activity. Conversely, the direct activation of SIRT1 by SRT1720 resulted in decreased FoxP3 expression, with no reduction in ROS levels. The pho-mTOR levels were significantly higher in Tregs after activation by SRT1720 than in those after RVL treatment. This study suggested that imbalanced changes in Tregs could be attributed to mTOR activation, in which ROS overproduction was predominantly implicated. Therefore, ROS is a key mediator for promoting Tregs instability in AAV.
Collapse
Affiliation(s)
- Yasuhiro Shimojima
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Dai Kishida
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Takanori Ichikawa
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Ryota Takamatsu
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Shun Nomura
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Yoshiki Sekijima
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
35
|
Induction of Foxp3 and activation of Tregs by HSP gp96 for treatment of autoimmune diseases. iScience 2021; 24:103445. [PMID: 34877502 PMCID: PMC8633978 DOI: 10.1016/j.isci.2021.103445] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/16/2021] [Accepted: 11/10/2021] [Indexed: 01/06/2023] Open
Abstract
Upregulation and stabilization of Foxp3 expression in Tregs are essential for regulating Treg function and immune homeostasis. In this study, gp96 immunization showed obvious therapeutic effects in a Lyn–/– mouse model of systemic lupus erythematosus. Moreover, gp96 alleviated the initiation and progression of MOG-induced experimental autoimmune encephalomyelitis. Immunization of gp96 increased Treg frequency, expansion, and suppressive function. Gene expression profiling identified the NF-κB family member p65 and c-Rel as the key transcription factors for enhanced Foxp3 expression in Treg by gp96. Mutant gp96 within its Toll-like receptor (TLR) binding domain, TLR2 knockout mice, and mice with cell-specific deletion of MyD88, were used to demonstrate that gp96 activated Tregs and induced Foxp3 expression via a TLR2-MyD88-mediated NF-κB signaling pathway. Taken together, these results show that gp96 immunization restricted antibody-induced and Th-induced autoimmune diseases by integrating Treg expansion and activation, indicating its potential clinical usefulness against autoimmune diseases. SLE symptoms in Lyn–/– mice are ameliorated by gp96 immunization Tregs expanded by gp96 provide potential in suppressing Th-mediated EAE Gp96 promotes Treg proliferation, stability, and suppressive function Gp96 binds to and activates Treg in a TLR2-MyD88-NF-кB-Foxp3 pathway
Collapse
|
36
|
Дудина МА, Догадин СА, Савченко АА, Беленюк ВД. [T-lymphocytes phenotypic composition of peripheral blood in patients with Graves' disease undergoing conservative therapy with thiamazole]. PROBLEMY ENDOKRINOLOGII 2021; 67:39-49. [PMID: 35018760 PMCID: PMC9753810 DOI: 10.14341/probl12812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/13/2021] [Accepted: 11/09/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Effective control of autoimmune inflammation in Graves' disease determines necessity to study the T helper (Th) and cytotoxic T-lymphocytes dysfunction, as well as the level of regulatory T-cells (Treg) activation in patients with Graves' disease on thyrostatic medication, which will clarify the immunomodulatory effects of long-term thiamazole treatment serve as targets for more specific therapies. AIM To study the phenotypic composition of T-lymphocytes in the peripheral blood of patients with Graves' disease to assess the direction of immune response depending on thimazole-induced euthyroidism duration. MATERIALS AND METHODS A single-center, cohort, continuous, open-label, controlled trial was conducted to assess the phenotypic composition of T-lymphocytes in peripheral blood in women with Graves' disease on long-term thiamazole treatment. The phenotypic composition of T-lymphocytes was determined by flow cytometry using direct immunofluorescence with conjugated FITC monoclonal antibodies depending on the duration of thimazole-induced euthyroidism of long-term thiamazole treatment. RESULTS The study included 135 women with Graves' disease, mean age 43.09±12.81 years, 120 (88.91%) with a relapse of the disease and 15 (11.09%) with newly diagnosed hyperthyroidism. An increase of activated CD3+CD4+CD25+ was found in patients with Graves' disease with a duration of thimazole-induced euthyroidism 5-8 months and 9-12 months, respectively, Me=0.94 (0.48-1.45), p=0.020) and Me=0.95 (0.41-1.80), p=0.025), in control group - Me=0.12 (0.03-0.68). Compared to the control an increase of CD4+CD25+CD127Low (Treg) was found in patients with a duration of thimazole-induced euthyroidism 5-8 and 9-12 months. The content of Treg in peripheral blood in Graves' disease patients with a duration of thimazole-induced euthyroidism more than 12 months decreases, but remains elevated relative to the control. CONCLUSION In patients with Graves' disease with a duration of thimazole-induced euthyroidism 5-8 months and 9-12 months the level of Treg has been increased. The increase of activated Th (CD3+CD4+CD25+) persists independently of thimazole-induced euthyroidism. In patients with Graves' disease with a duration of thimazole-induced euthyroidism for more than 12 months, there is a compensatory increase in regulatory T-lymphocyte, and the total number of T-helpers is restored to the control.
Collapse
Affiliation(s)
- М. А. Дудина
- Красноярский государственный медицинский университет имени профессора В.Ф. Войно-Ясенецкого;
Краевая клиническая больница
| | - С. А. Догадин
- Красноярский государственный медицинский университет имени профессора В.Ф. Войно-Ясенецкого;
Краевая клиническая больница
| | - А. А. Савченко
- Красноярский государственный медицинский университет имени профессора В.Ф. Войно-Ясенецкого;
Красноярский научный центр Сибирского отделения Российской академии наук», обособленное подразделение «НИИ медицинских проблем Севера»
| | - В. Д. Беленюк
- Красноярский научный центр Сибирского отделения Российской академии наук», обособленное подразделение «НИИ медицинских проблем Севера»
| |
Collapse
|
37
|
Roberts LB, Kapoor P, Howard JK, Shah AM, Lord GM. An update on the roles of immune system-derived microRNAs in cardiovascular diseases. Cardiovasc Res 2021; 117:2434-2449. [PMID: 33483751 PMCID: PMC8562329 DOI: 10.1093/cvr/cvab007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 01/08/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVD) are a leading cause of human death worldwide. Over the past two decades, the emerging field of cardioimmunology has demonstrated how cells of the immune system play vital roles in the pathogenesis of CVD. MicroRNAs (miRNAs) are critical regulators of cellular identity and function. Cell-intrinsic, as well as cell-extrinsic, roles of immune and inflammatory cell-derived miRNAs have been, and continue to be, extensively studied. Several 'immuno-miRNAs' appear to be specifically expressed or demonstrate greatly enriched expression within leucocytes. Identification of miRNAs as critical regulators of immune system signalling pathways has posed the question of whether and how targeting these molecules therapeutically, may afford opportunities for disease treatment and/or management. As the field of cardioimmunology rapidly continues to advance, this review discusses findings from recent human and murine studies which contribute to our understanding of how leucocytes of innate and adaptive immunity are regulated-and may also regulate other cell types, via the actions of the miRNAs they express, in the context of CVD. Finally, we focus on available information regarding miRNA regulation of regulatory T cells and argue that targeted manipulation of miRNA regulated pathways in these cells may hold therapeutic promise for the treatment of CVD and associated risk factors.
Collapse
Affiliation(s)
- Luke B Roberts
- School of Immunology and Microbial Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK
| | - Puja Kapoor
- School of Immunology and Microbial Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK
- School of Cardiovascular Medicine and Sciences, King’s British Heart Foundation Centre, King’s College London, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Jane K Howard
- School of Life Course Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK
| | - Ajay M Shah
- School of Cardiovascular Medicine and Sciences, King’s British Heart Foundation Centre, King’s College London, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Graham M Lord
- School of Immunology and Microbial Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK
- Faculty of Biology, Medicine and Health, University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| |
Collapse
|
38
|
Yang SY, Long J, Huang MX, Luo PY, Bian ZH, Xu YF, Wang CB, Yang SH, Li L, Selmi C, Gershwin ME, Zhao ZB, Lian ZX. Characterization of Organ-Specific Regulatory B Cells Using Single-Cell RNA Sequencing. Front Immunol 2021; 12:711980. [PMID: 34594327 PMCID: PMC8476928 DOI: 10.3389/fimmu.2021.711980] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 08/23/2021] [Indexed: 12/04/2022] Open
Abstract
Regulatory B cells (Breg) are considered as immunosuppressive cells. Different subsets of Breg cells have been identified both in human beings and in mice. However, there is a lack of unique markers to identify Breg cells, and the heterogeneity of Breg cells in different organs needs to be further illuminated. In this study, we performed high-throughput single-cell RNA sequencing (scRNA-seq) and single-cell B-cell receptor sequencing (scBCR-seq) of B cells from the murine spleen, liver, mesenteric lymph nodes, bone marrow, and peritoneal cavity to better define the phenotype of these cells. Breg cells were identified based on the expression of immunosuppressive genes and IL-10-producing B (B10) cell-related genes, to define B10 and non-B10 subsets in Breg cells based on the score of the B10 gene signatures. Moreover, we characterized 19 common genes significantly expressed in Breg cells, including Fcrl5, Zbtb20, Ccdc28b, Cd9, and Ptpn22, and further analyzed the transcription factor activity in defined Breg cells. Last, a BCR analysis was used to determine the clonally expanded clusters and the relationship of Breg cells across different organs. We demonstrated that Atf3 may potentially modulate the function of Breg cells as a transcription factor and that seven organ-specific subsets of Breg cells are found. Depending on gene expression and functional modules, non-B10 Breg cells exhibited activated the TGF-β pathway, thus suggesting that non-B10 Breg cells have specific immunosuppressive properties different from conventional B10 cells. In conclusion, our work provides new insights into Breg cells and illustrates their transcriptional profiles and BCR repertoire in different organs under physiological conditions.
Collapse
Affiliation(s)
- Si-Yu Yang
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jie Long
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Meng-Xing Huang
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, China
| | - Pan-Yue Luo
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, China
| | - Zhen-Hua Bian
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ya-Fei Xu
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, China
| | - Cheng-Bo Wang
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, China
| | - Shu-Han Yang
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, China
| | - Liang Li
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Carlo Selmi
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital IRCCS, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - M Eric Gershwin
- Division of Rheumatology, Allergy, and Clinical Immunology, University of California Davis, Davis, CA, United States
| | - Zhi-Bin Zhao
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhe-Xiong Lian
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
39
|
Su M, Zhao C, Luo S. Therapeutic potential of chimeric antigen receptor based therapies in autoimmune diseases. Autoimmun Rev 2021; 21:102931. [PMID: 34481941 DOI: 10.1016/j.autrev.2021.102931] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/26/2021] [Accepted: 08/31/2021] [Indexed: 11/02/2022]
Abstract
Chimeric antigen receptor (CAR) based therapies have been adopted as an option for treating autoimmune diseases from the field of blood malignancies by targeting immune cells or rebalancing the pro-inflammatory milieu. Important questions still remained about the efficacy and safety regarding the dynamic and complex autoimmune pathological networks. We here reviewed the emerged developments in basic, translational, and clinical studies of the CAR based therapies in a wide spectrum of autoimmune diseases. The primary goal of the study is to provide some future perspectives on how to optimize the performance of CAR based therapies. The fundamental strategy is to engineer the recognition domains in CAR products for precisely targeting the components in the pro-inflammatory milieu. The second strategy is to incorporate multiple CARs in one carrier, or use fluorescein isothiocyanate (FITC)-CAR T cells for enhancing the therapeutic efficacy. In addition, we reviewed the preclinical evidence in disease-specific context. Overall, we aim to attract more attention in the field of developing future precision CAR based therapies to tailor medial decisions in autoimmune diseases.
Collapse
Affiliation(s)
- Manqiqige Su
- Department of Neurology, Huashan hospital Fudan University, 200040 Shanghai, China
| | - Chongbo Zhao
- Department of Neurology, Huashan hospital Fudan University, 200040 Shanghai, China
| | - Sushan Luo
- Department of Neurology, Huashan hospital Fudan University, 200040 Shanghai, China.
| |
Collapse
|
40
|
Zaki MM, Lesha E, Said K, Kiaee K, Robinson-McCarthy L, George H, Hanna A, Appleton E, Liu S, Ng AHM, Khoshakhlagh P, Church GM. Cell therapy strategies for COVID-19: Current approaches and potential applications. SCIENCE ADVANCES 2021; 7:eabg5995. [PMID: 34380619 PMCID: PMC8357240 DOI: 10.1126/sciadv.abg5995] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 06/22/2021] [Indexed: 05/03/2023]
Abstract
Coronavirus disease 2019 (COVID-19) continues to burden society worldwide. Despite most patients having a mild course, severe presentations have limited treatment options. COVID-19 manifestations extend beyond the lungs and may affect the cardiovascular, nervous, and other organ systems. Current treatments are nonspecific and do not address potential long-term consequences such as pulmonary fibrosis, demyelination, and ischemic organ damage. Cell therapies offer great potential in treating severe COVID-19 presentations due to their customizability and regenerative function. This review summarizes COVID-19 pathogenesis, respective areas where cell therapies have potential, and the ongoing 89 cell therapy trials in COVID-19 as of 1 January 2021.
Collapse
Affiliation(s)
- Mark M Zaki
- GC Therapeutics Inc., Cambridge, MA 02139, USA
- Department of Neurosurgery, University of Michigan, 1500 E Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Emal Lesha
- GC Therapeutics Inc., Cambridge, MA 02139, USA
- Department of Neurosurgery, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Khaled Said
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Kiavash Kiaee
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | | | | | - Angy Hanna
- Department of Medicine, Beaumont Hospital, Royal Oak, MI, USA
| | - Evan Appleton
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA
| | - Songlei Liu
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA
| | - Alex H M Ng
- GC Therapeutics Inc., Cambridge, MA 02139, USA.
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA
| | - Parastoo Khoshakhlagh
- GC Therapeutics Inc., Cambridge, MA 02139, USA.
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA
| | - George M Church
- GC Therapeutics Inc., Cambridge, MA 02139, USA.
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA
| |
Collapse
|
41
|
Shimojima Y, Ichikawa T, Kishida D, Takamatsu R, Sekijima Y. Circulating regulatory T cells in adult-onset Still's disease: Focusing on their plasticity and stability. Clin Exp Immunol 2021; 206:184-195. [PMID: 34319596 DOI: 10.1111/cei.13648] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/23/2021] [Accepted: 07/21/2021] [Indexed: 11/28/2022] Open
Abstract
We investigated the characteristics of regulatory T cells in adult-onset Still's disease (AOSD) with a focus on their plasticity, stability and relationship to disease severity. The proportion of circulating CD4+ CD25+ forkhead box protein 3 (FoxP3+ ) cells (Tregs ) and intracellular expression of effector cytokines, including interferon (IFN)-γ, interleukin (IL)-17 and IL-4, was analysed in 27 untreated patients with AOSD (acute AOSD), 11 of the 27 patients after remission and 16 healthy controls (HC) using flow cytometry. The suppressive ability of Tregs was also evaluated. Regression analyses of the results were performed. The proportion of Tregs was significantly lower in patients with acute AOSD than in the HC. The expression levels of IFN-γ, IL-17 and IL-4 in Tregs were significantly increased in patients with acute AOSD. IFN-γ and IL-4 expression levels were inversely correlated with the proportion of Tregs and positively correlated with serum ferritin levels. Decreased expression of FoxP3 in CD4+ CD25+ cells, which was correlated with increased expression of IL-17, and impaired suppressive function were observed in Tregs in acute AOSD. However, these aberrant findings in Tregs , including the reduced circulating proportion and functional ability and altered intracellular expression levels of cytokines and FoxP3, were significantly improved after remission. In acute AOSD, Tregs show plastic changes, including effector cytokine production and reductions in their proportion and functional activity. IFN-γ and IL-4 expression levels in Tregs may be associated with disease severity. Also, down-regulation of FoxP3 may be related to IL-17 expression in Tregs . Importantly, the stability of Tregs can be restored in remission.
Collapse
Affiliation(s)
- Yasuhiro Shimojima
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Takanori Ichikawa
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Dai Kishida
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Ryota Takamatsu
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Yoshiki Sekijima
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
42
|
Byun JI, Bae J, Moon J, Lee ST, Jung KH, Park KI, Kim M, Lee SK, Chu K. Proportion of peripheral regulatory T cells in patients with autoimmune encephalitis. ENCEPHALITIS 2021; 1:68-72. [PMID: 37469844 PMCID: PMC10295879 DOI: 10.47936/encephalitis.2021.00052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/01/2021] [Accepted: 04/02/2021] [Indexed: 07/21/2023] Open
Abstract
PURPOSE Regulatory T cells (Tregs) play a crucial role in maintaining immune tolerance. Any deficiency or dysfunction of the Tregs can influence the pathogenesis of autoimmune disease. This study aimed to assess the role of Tregs among patients with autoimmune encephalitis (AE) with different autoantibody types and to evaluate their association with clinical features. METHODS This was a cross-sectional observational study involving 29 patients with AE. Peripheral blood was sampled from each patient for flow cytometric analysis. Proportions of CD4+CD25+ and CD4+CD25+Foxp3+ Tregs were calculated and compared between the antibody types (synaptic, paraneoplastic, and undetermined). Associations between the proportion of Tregs and clinical features were also evaluated. RESULTS Five patients had synaptic autoantibodies, five had paraneoplastic autoantibodies, and the others were of an undetermined type. The proportion of CD4+CD25+ Tregs tended to be higher in those with paraneoplastic antibodies than in those with synaptic antibodies (post-hoc p = 0.028) and undetermined antibody status (post-hoc p = 0.043). A significant negative correlation was found between the proportion of Tregs and the initial modified Rankin score (r = -0.391, p = 0.036). Those who received intravenous immunoglobulin had lower proportions of Tregs than those who did not. CONCLUSION The results of the present study suggest that Tregs may play different roles according to the type of AE and may be linked to disease severity.
Collapse
Affiliation(s)
- Jung-Ick Byun
- Department of Neurology, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul, Korea
| | - Ji‐Yeon Bae
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
- Program in Neuroscience, Seoul National University College of Medicine, Seoul, Korea
| | - Jangsup Moon
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
- Program in Neuroscience, Seoul National University College of Medicine, Seoul, Korea
- Rare Disease Center, Seoul National University Hospital, Seoul, Korea
| | - Soon-Tae Lee
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
- Program in Neuroscience, Seoul National University College of Medicine, Seoul, Korea
| | - Keun-Hwa Jung
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
- Program in Neuroscience, Seoul National University College of Medicine, Seoul, Korea
| | - Kyung-Il Park
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
- Department of Neurology, Seoul National University Hospital Healthcare System Gangnam Center, Seoul, Korea
| | - Manho Kim
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
- Program in Neuroscience, Seoul National University College of Medicine, Seoul, Korea
| | - Sang Kun Lee
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
- Program in Neuroscience, Seoul National University College of Medicine, Seoul, Korea
| | - Kon Chu
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
- Program in Neuroscience, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
43
|
Lin LN, Zhang QM, Ge YY, Luo B, Xie XX. A Review of miR-326 and Female Related Diseases. Acta Histochem Cytochem 2021; 54:79-86. [PMID: 34276101 PMCID: PMC8275862 DOI: 10.1267/ahc.20-00027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 03/07/2021] [Indexed: 12/16/2022] Open
Abstract
MicroRNA (miRNA), a non-coding single-stranded RNA molecule with 20–23 nucleotides encoded by endogenous genes, plays an essential role in maintaining normal cell function and regulating cell proliferation, differentiation, apoptosis, autophagy, and cell metabolism. The imbalance between miRNA and genes can cause a series of diseases, including malignancies. miRNA-326 (miR-326) is extensively known for its core regulation of various biological processes. This review presents an overview of the highlights of miR-326 in female-related diseases. To understand the impact of miR-326 on female disorders, we search all published studies about miR-326 having a high incidence in female conditions, including cervical cancer, endometrial cancer, breast cancer, intrauterine adhesion, and multiple autoimmune diseases. We aim to learn about the mutual regulation mechanism between miR-326 and related genes and signaling pathways, as well as to elaborate on the value of miR-326 as a potential biomarker and therapeutic target of female diseases. Our results provide reliable evidence and new strategies for treating female tumors and autoimmune diseases.
Collapse
Affiliation(s)
- Li-na Lin
- Department of Histology and Embryology, School of Pre-clinical Medicine, Guangxi Medical University
| | - Qing-mei Zhang
- Department of Histology and Embryology, School of Pre-clinical Medicine, Guangxi Medical University
- Key Laboratory Research of Preclinical Medicine of Guangxi Colleges and Universities, Guangxi Medical University
| | - Ying-ying Ge
- Department of Histology and Embryology, School of Pre-clinical Medicine, Guangxi Medical University
- Key Laboratory Research of Preclinical Medicine of Guangxi Colleges and Universities, Guangxi Medical University
| | - Bin Luo
- Department of Histology and Embryology, School of Pre-clinical Medicine, Guangxi Medical University
- Key Laboratory Research of Preclinical Medicine of Guangxi Colleges and Universities, Guangxi Medical University
| | - Xiao-xun Xie
- Department of Histology and Embryology, School of Pre-clinical Medicine, Guangxi Medical University
- Key Laboratory Research of Preclinical Medicine of Guangxi Colleges and Universities, Guangxi Medical University
- Key Laboratory of Early Prevention and Treatment of Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education
| |
Collapse
|
44
|
GŁaczynska M, Machcinska M, Donskow-Lysoniewska K. Effects of Different Media on Human T Regulatory Cells Phenotype. In Vivo 2021; 35:283-289. [PMID: 33402475 DOI: 10.21873/invivo.12257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/10/2020] [Accepted: 11/17/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND/AIM Functional and quantitative Treg cell defects have been identified in a variety of autoimmune diseases. Therefore, Tregs are a major pharmaceutical target for these disorders. In the last decades, studies have been mainly focused on the identification and experimental understanding of the activity of Tregs and their mechanisms of action. MATERIALS AND METHODS This study describes how overnight storage of isolated peripheral blood mononuclear cells in different media (PBS pH 7.3, PBS pH 7.3 containing 0.5% BSA, RPMI 1640 and RPMI 1640 containing 10% FBS) affects the viability and expression of the commonly used markers for Tregs identification: CD25, CD127, CTLA-4, GITR, PD-1, FoxP3 and Helios. RESULTS Incorrectly selected storage conditions (temperature, time, medium) may affect the expression of surface and intracellular markers, thus, compromising the quality of the obtained results. CONCLUSION Appropriate protocols of cell isolation and storage are important for providing appropriate conditions for cell growth. This is crucial when analyzing small cell populations like Tregs.
Collapse
Affiliation(s)
- Magdalena GŁaczynska
- Laboratory of Parasitology, General Karol Kaczkowski Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| | - Maja Machcinska
- Laboratory of Parasitology, General Karol Kaczkowski Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| | | |
Collapse
|
45
|
Sogkas G, Atschekzei F, Adriawan IR, Dubrowinskaja N, Witte T, Schmidt RE. Cellular and molecular mechanisms breaking immune tolerance in inborn errors of immunity. Cell Mol Immunol 2021; 18:1122-1140. [PMID: 33795850 PMCID: PMC8015752 DOI: 10.1038/s41423-020-00626-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 12/11/2020] [Indexed: 02/01/2023] Open
Abstract
In addition to susceptibility to infections, conventional primary immunodeficiency disorders (PIDs) and inborn errors of immunity (IEI) can cause immune dysregulation, manifesting as lymphoproliferative and/or autoimmune disease. Autoimmunity can be the prominent phenotype of PIDs and commonly includes cytopenias and rheumatological diseases, such as arthritis, systemic lupus erythematosus (SLE), and Sjogren's syndrome (SjS). Recent advances in understanding the genetic basis of systemic autoimmune diseases and PIDs suggest an at least partially shared genetic background and therefore common pathogenic mechanisms. Here, we explore the interconnected pathogenic pathways of autoimmunity and primary immunodeficiency, highlighting the mechanisms breaking the different layers of immune tolerance to self-antigens in selected IEI.
Collapse
Affiliation(s)
- Georgios Sogkas
- Department of Rheumatology and Immunology, Hannover Medical School, Hanover, Germany.
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hanover, Germany.
| | - Faranaz Atschekzei
- Department of Rheumatology and Immunology, Hannover Medical School, Hanover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hanover, Germany
| | - Ignatius Ryan Adriawan
- Department of Rheumatology and Immunology, Hannover Medical School, Hanover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hanover, Germany
| | - Natalia Dubrowinskaja
- Department of Rheumatology and Immunology, Hannover Medical School, Hanover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hanover, Germany
| | - Torsten Witte
- Department of Rheumatology and Immunology, Hannover Medical School, Hanover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hanover, Germany
| | - Reinhold Ernst Schmidt
- Department of Rheumatology and Immunology, Hannover Medical School, Hanover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hanover, Germany
| |
Collapse
|
46
|
Huang Z, Li W, Su W. Tregs in Autoimmune Uveitis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1278:205-227. [PMID: 33523450 DOI: 10.1007/978-981-15-6407-9_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Uveitis is a chronic disease with relapsing and remitting ocular attack, which requires corticosteroids and systemic immunosuppression to prevent severe vision loss. Classically, uveitis is referred to an autoimmune disease, mediated by pro-inflammatory Th17 cells and immunosuppressive CD4+CD25+FoxP3+ T-regulatory cells (Tregs). More and more evidence indicates that Tregs are involved in development, resolution, and remission of uveitis. Clinically, many researchers have conducted quantitative and functional analyses of peripheral blood from patients with different subtypes of uveitis, in an attempt to find the changing rules of Tregs. Consistently, using the experimental autoimmune uveitis (EAU) model, researchers have explored the development and resolution mechanism of uveitis in many aspects. In addition, many drug and Tregs therapy investigations have yielded encouraging results. In this chapter, we introduced the current understanding of Tregs, summarized the clinical changes in the number and function of patients with uveitis and the immune mechanism of Tregs involved in EAU model, as well as discussed the progress and shortcomings of Tregs-related drug therapy and Tregs therapy. Although the exact mechanism of Tregs-mediated uveitis protection remains to be elucidated, the strategy of Tregs regulation may provide a specific and meaningful way for the prevention and treatment of uveitis.
Collapse
Affiliation(s)
- Zhaohao Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Wenli Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
47
|
Barzaghi F, Passerini L. IPEX Syndrome: Improved Knowledge of Immune Pathogenesis Empowers Diagnosis. Front Pediatr 2021; 9:612760. [PMID: 33692972 PMCID: PMC7937806 DOI: 10.3389/fped.2021.612760] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/21/2021] [Indexed: 12/18/2022] Open
Abstract
Immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome is a rare monogenic autoimmune disease with variable clinical manifestations, ranging from early-onset severe autoimmunity, including enteropathy, eczema, and type 1 diabetes, to late-onset or atypical symptoms. Despite the clinical heterogeneity, the unifying feature of IPEX is mutation of the FOXP3 gene, which encodes a transcription factor essential for maintenance of thymus-derived regulatory T cells (Tregs). In IPEX patients, Tregs can be present, although unstable and impaired in function, unable to inhibit proliferation and cytokine production of effector T (Teff) cells. Mutated FOXP3 can also disrupt other compartments: FOXP3-deficient Teff cells proliferate more than the wild-type counterpart, display altered T-cell-receptor signaling response, a reduced T-naïve compartment and a skew toward a Th2 profile. Due to FOXP3 mutations, the frequency of autoreactive B cells is increased and the IgA and IgE production is altered, together with early emergence of tissue-specific autoantibodies. Recently, the awareness of the wide clinical spectrum of IPEX improved the diagnostic tools. In cases presenting with enteropathy, histological evaluation is helpful, although there are no pathognomonic signs of disease. On the other hand, the study of FOXP3 expression and in vitro Treg function, as well as the detection of specific circulating autoantibodies, is recommended to narrow the differential diagnosis. Nowadays, Sanger sequencing should be limited to cases presenting with the classical triad of symptoms; otherwise, next-generation sequencing is recommended, given the cost-effectiveness and the advantage of excluding IPEX-like syndromes. The latter approach could be time spearing in children with severe phenotypes and candidate to advanced therapies.
Collapse
Affiliation(s)
- Federica Barzaghi
- Department of Paediatric Immunohematology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Laura Passerini
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
48
|
Chodaczek G, Pagni PP, Christoffersson G, Ratliff SS, Toporkiewicz M, Wegrzyn AS, von Herrath M. The effect of Toll-like receptor stimulation on the motility of regulatory T cells. J Autoimmun 2021; 116:102563. [PMID: 33189487 DOI: 10.1016/j.jaut.2020.102563] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 10/23/2022]
Abstract
Regulatory T cells (Tregs) have suppressive functions and play an important role in controlling inflammation and autoimmunity. The migratory capacity of Tregs determines their location and their location determines whether they inhibit the priming of naïve lymphocytes in lymphoid tissues or the effector phase of immune responses at inflamed sites. Tregs generated or expanded in vitro are currently being tested in clinics for the treatment of autoimmune disorders, however, little is known about the factors controlling their migration towards therapeutically relevant locations. In this study, we have modulated Treg dynamics using Toll-like receptor (TLR) agonists. Dynamic imaging with confocal and two-photon microscopy revealed that Tregs generated in vitro and stimulated with P3C (a TLR2 agonist) but not with R848 (a TLR7 agonist) or LPS (a TLR4 agonist) showed enhanced cell migration within splenic white pulp or draining lymph node when transferred into mice intravenously or into the footpad, respectively. In summary, our data demonstrate that Tregs are more motile in response to direct TLR stimulation in particular towards TLR2 signals. This may have implications for efficient clinical Treg induction protocols.
Collapse
Affiliation(s)
- Grzegorz Chodaczek
- Type 1 Diabetes Center, La Jolla Institute for Immunology, La Jolla, CA, USA; Bioimaging Laboratory, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland.
| | - Philippe P Pagni
- Type 1 Diabetes Center, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Gustaf Christoffersson
- Type 1 Diabetes Center, La Jolla Institute for Immunology, La Jolla, CA, USA; Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Monika Toporkiewicz
- Bioimaging Laboratory, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Agnieszka S Wegrzyn
- Bioimaging Laboratory, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | | |
Collapse
|
49
|
The Association of Gut Microbiota and Treg Dysfunction in Autoimmune Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1278:191-203. [PMID: 33523449 PMCID: PMC9290759 DOI: 10.1007/978-981-15-6407-9_10] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Autoimmune conditions affect 23 million Americans or 7% of the US population. There are more than 100 autoimmune disorders, affecting every major organ system in humans. This chapter aims to further explain Treg dysfunction autoimmune disorders, including monogenic primary immune deficiency such as immune dysregulation polyendocrinopathy, enteropathy, X-linked inheritance (IPEX) syndrome, and polygenic autoimmune diseases with Treg dysfunction such as multiple sclerosis (MS), inflammatory bowel disease (IBD), and food allergy. These conditions are associated with an abnormal small intestinal and colonic microbiome. Some disorders clearly improve with therapies aimed at microbial modification, including probiotics and fecal microbiota transplantation (FMT). Approaches to prevent and treat these disorders will need to focus on the acquisition and maintenance of a healthy colonic microbiota, in addition to more focused approaches at immune suppression during acute disease exacerbations.
Collapse
|
50
|
Mukhatayev Z, Dellacecca ER, Cosgrove C, Shivde R, Jaishankar D, Pontarolo-Maag K, Eby JM, Henning SW, Ostapchuk YO, Cedercreutz K, Issanov A, Mehrotra S, Overbeck A, Junghans RP, Leventhal JR, Le Poole IC. Antigen Specificity Enhances Disease Control by Tregs in Vitiligo. Front Immunol 2020; 11:581433. [PMID: 33335528 PMCID: PMC7736409 DOI: 10.3389/fimmu.2020.581433] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/27/2020] [Indexed: 12/19/2022] Open
Abstract
Vitiligo is an autoimmune skin disease characterized by melanocyte destruction. Regulatory T cells (Tregs) are greatly reduced in vitiligo skin, and replenishing peripheral skin Tregs can provide protection against depigmentation. Ganglioside D3 (GD3) is overexpressed by perilesional epidermal cells, including melanocytes, which prompted us to generate GD3-reactive chimeric antigen receptor (CAR) Tregs to treat vitiligo. Mice received either untransduced Tregs or GD3-specific Tregs to test the hypothesis that antigen specificity contributes to reduced autoimmune reactivity in vitro and in vivo. CAR Tregs displayed increased IL-10 secretion in response to antigen, provided superior control of cytotoxicity towards melanocytes, and supported a significant delay in depigmentation compared to untransduced Tregs and vehicle control recipients in a TCR transgenic mouse model of spontaneous vitiligo. The latter findings were associated with a greater abundance of Tregs and melanocytes in treated mice versus both control groups. Our data support the concept that antigen-specific Tregs can be prepared, used, and stored for long-term control of progressive depigmentation.
Collapse
Affiliation(s)
- Zhussipbek Mukhatayev
- Department of Dermatology, Northwestern University, Chicago, IL, United States.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, United States.,Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty, Kazakhstan.,Laboratory of Molecular immunology and Immunobiotechnology, M.A. Aitkhozhin's Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | - Emilia R Dellacecca
- Department of Dermatology, Northwestern University, Chicago, IL, United States.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, United States
| | - Cormac Cosgrove
- Department of Dermatology, Northwestern University, Chicago, IL, United States.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, United States
| | - Rohan Shivde
- Department of Dermatology, Northwestern University, Chicago, IL, United States.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, United States
| | - Dinesh Jaishankar
- Department of Dermatology, Northwestern University, Chicago, IL, United States.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, United States
| | | | - Jonathan M Eby
- Oncology Research Institute, Loyola University, Maywood, IL, United States
| | - Steven W Henning
- Oncology Research Institute, Loyola University, Maywood, IL, United States
| | - Yekaterina O Ostapchuk
- Laboratory of Molecular immunology and Immunobiotechnology, M.A. Aitkhozhin's Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | - Kettil Cedercreutz
- Department of Dermatology, Northwestern University, Chicago, IL, United States
| | - Alpamys Issanov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Andreas Overbeck
- Department for Surgery of Pigment Disorders, Lumiderm, Madrid, Spain
| | - Richard P Junghans
- Department of Hematology/Oncology, Boston University, Boston MA, United States
| | - Joseph R Leventhal
- Comprehensive Transplant Center, Northwestern Memorial Hospital, Chicago, IL, United States
| | - I Caroline Le Poole
- Department of Dermatology, Northwestern University, Chicago, IL, United States.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, United States
| |
Collapse
|