1
|
Gautam S, Kumar S, Dada R. Transcription Factor Analysis to Investigate Immunosenescence in Rheumatoid Arthritis Patients. Methods Mol Biol 2025; 2857:79-87. [PMID: 39348056 DOI: 10.1007/978-1-0716-4128-6_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Rheumatoid arthritis (RA) is linked to various signs of advanced aging, such as premature immunosenescence which occurs due to decline in regenerative ability of T cells. RA T cells develop a unique aggressive inflammatory senescent phenotype with an imbalance of Th17/T regulatory (Treg) cell homeostasis and presence of CD28- T cells. The phenotypic analysis and characterization of T cell subsets become necessary to ascertain if any functional deficiencies exist within with the help of transcription factor (TF) analysis. These subset-specific TFs dictate the functional characteristics of T-cell populations, leading to the production of distinct effector cytokines and functions. Examining the expression, activity, regulation, and genetic sequence of TFs not only aids researchers in determining their importance in disease processes but also aids in immunological monitoring of patients enrolled in clinical trials, particularly in evaluating various T-cell subsets [Th17 (CD3+CD4+IL17+RORγt+) cells and T regulatory (Treg) (CD3+CD4+CD25+CD127-FOXP3+) cells], markers of T-cell aging [aged Th17 cells (CD3+CD4+IL17+RORγt+CD28-), and aged Treg cells (CD3+CD4+CD25+CD127-FOXP3+CD28-)]. In this context, we propose and outline the protocols for assessing the expression of TFs in aged Th17 and Treg cells, highlighting the crucial aspects of this cytometric approach.
Collapse
Affiliation(s)
- Surabhi Gautam
- Department of Anatomy, Molecular Reproduction and Genetics Facility, All India Institute of Medical Sciences (AIIMS), New Delhi, India
- Department of Orthopaedics, Emory Musculoskeletal Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Sanjeev Kumar
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Rima Dada
- Department of Anatomy, Molecular Reproduction and Genetics Facility, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| |
Collapse
|
2
|
Lubin R, Patel AA, Mackerodt J, Zhang Y, Gvili R, Mulder K, Dutertre CA, Jalali P, Glanville JR, Hazan I, Sridharan N, Rivkin G, Akarca A, Marafioti T, Gilroy DW, Kandel L, Mildner A, Wilensky A, Asquith B, Ginhoux F, Macallan D, Yona S. The lifespan and kinetics of human dendritic cell subsets and their precursors in health and inflammation. J Exp Med 2024; 221:e20220867. [PMID: 39417994 PMCID: PMC11488382 DOI: 10.1084/jem.20220867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/16/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024] Open
Abstract
Dendritic cells (DC) are specialized mononuclear phagocytes that link innate and adaptive immunity. They comprise two principal subsets: plasmacytoid DC (pDC) and conventional DC (cDC). Understanding the generation, differentiation, and migration of cDC is critical for immune homeostasis. Through human in vivo deuterium-glucose labeling, we observed the rapid appearance of AXL+ Siglec6+ DC (ASDC) in the bloodstream. ASDC circulate for ∼2.16 days, while cDC1 and DC2 circulate for ∼1.32 and ∼2.20 days, respectively, upon release from the bone marrow. Interestingly, DC3, a cDC subset that shares several similarities with monocytes, exhibits a labeling profile closely resembling that of DC2. In a human in vivo model of cutaneous inflammation, ASDC were recruited to the inflammatory site, displaying a distinctive effector signature. Taken together, these results quantify the ephemeral circulating lifespan of human cDC and propose functions of cDC and their precursors that are rapidly recruited to sites of inflammation.
Collapse
Affiliation(s)
- Ruth Lubin
- The Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Amit A. Patel
- Division of Medicine, University College London, London, UK
| | - Jonas Mackerodt
- Department of Infectious Disease, Imperial College London, London, UK
| | - Yan Zhang
- Institute for Infection and Immunity, St. George’s, University of London, London, UK
| | - Rotem Gvili
- The Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Kevin Mulder
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée—Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, France
| | - Charles-Antoine Dutertre
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée—Ligue Nationale Contre le Cancer, Villejuif, France
| | | | | | - Idit Hazan
- The Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Nikhila Sridharan
- The Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Gurion Rivkin
- Department of Orthopaedic Surgery, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | - Leonid Kandel
- Department of Orthopaedic Surgery, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Alexander Mildner
- MediCity Research Laboratory, University of Turku, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Asaf Wilensky
- Department of Periodontology, Faculty of Dental Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Becca Asquith
- Department of Infectious Disease, Imperial College London, London, UK
| | - Florent Ginhoux
- Gustave Roussy Cancer Campus, Villejuif, France
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong, University School of Medicine, Shanghai, China
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore
| | - Derek Macallan
- Institute for Infection and Immunity, St. George’s, University of London, London, UK
- St. George’s University Hospitals NHS Foundation Trust, London, UK
| | - Simon Yona
- The Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| |
Collapse
|
3
|
Becerro-Rey L, Martín-Cano FE, Fabres Robaina Sancler-Silva Y, Gil MC, Ortega-Ferrusola C, Aparicio IM, Gaitskell-Phillips G, da Silva-Álvarez E, Peña FJ. In vitro, the aging of stallion spermatozoa at 22 °C is linked to alteration in Ca 2+ and redox homeostasis and may be slowed by regulating metabolism. Theriogenology 2024; 229:127-137. [PMID: 39178614 DOI: 10.1016/j.theriogenology.2024.08.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 08/11/2024] [Accepted: 08/17/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND Conservation of equine semen in the liquid state is a central procedure in horse breeding and constitutes the basis of associated reproductive technologies. The intense mitochondrial activity of the stallion spermatozoa increases oxidative stress along the storage period, leading to sperm demise within 24-48 h of storage, particularly when maintained at room temperature. Recently, the relationship between metabolism and oxidative stress has been revealed. The study aimed to extend the period of conservation of equine semen, at room temperature through modification of the metabolites present in the media. MATERIAL AND METHODS Processed ejaculates (n = 9) by single-layer colloid centrifugation were split in different aliquots and extended in Tyrode's basal media, or modified Tyrode's consisting of 1 mM glucose, 1 mM glucose 10 mM pyruvate, 40 mM glucose, 40 mM Glucose 10 mM pyruvate, 67 mM glucose and 67 mM glucose 10 mM pyruvate. At time 0h, and after 24 and 96 h of storage, motility was evaluated by CASA, while mitochondrial production of Reactive oxygen species (ROS), and intracellular Ca2+ concentrations were determined via flow cytometry using Mitosox Red and Fluo-4 respectively. ROS and Ca2+ were estimated as Relative Fluorescence Units (RFU) in compensated, arcsin-transformed data in the live sperm population. RESULTS After 48 h of incubation, motility was greater in all the 10 mM pyruvate-based media, with the poorest result in the 40 mM glucose (41 ± 1.1 %) while the highest motility was yielded in the 40 mM glucose 10 mM pyruvate aliquot (60.3 ± 3.5 %; P < 0.001); after 96 h of storage highest motility values were observed in the 40 mM glucose 10 mM pyruvate media (23.0 ± 6.2 %) while the lowest was observed in the 1 mM glucose media was 9.2 ± 2.0 % (P < 0.05). Mitochondrial ROS was lower in the 40 mM glucose 10 mM pyruvate group compared to the 40 mM glucose (P < 0.01). Over time Ca2+ increased in all treatment groups compared to time 0h. DISCUSSION AND CONCLUSION Viable spermatozoa may experience oxidative stress and alterations in Ca2+ homeostasis during prolonged storage, however, these effects can be reduced by regulating metabolism. The 40 mM glucose- 10 mM pyruvate group yielded the highest sperm quality parameters.
Collapse
Affiliation(s)
- Laura Becerro-Rey
- Laboratory of Equine Reproduction and Equine Spermatology, Veterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| | - Francisco Eduardo Martín-Cano
- Laboratory of Equine Reproduction and Equine Spermatology, Veterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| | - Yame Fabres Robaina Sancler-Silva
- Laboratory of Equine Reproduction and Equine Spermatology, Veterinary Teaching Hospital, University of Extremadura, Cáceres, Spain; Department of Animal Science, Universidade Federal de Viçosa, Viçosa, 36570-900, Brazil
| | - María Cruz Gil
- Laboratory of Equine Reproduction and Equine Spermatology, Veterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| | - Cristina Ortega-Ferrusola
- Laboratory of Equine Reproduction and Equine Spermatology, Veterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| | - Inés M Aparicio
- Department of Anatomy, University of Extremadura, Cáceres, Spain
| | - Gemma Gaitskell-Phillips
- Laboratory of Equine Reproduction and Equine Spermatology, Veterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| | - Eva da Silva-Álvarez
- Laboratory of Equine Reproduction and Equine Spermatology, Veterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| | - Fernando J Peña
- Laboratory of Equine Reproduction and Equine Spermatology, Veterinary Teaching Hospital, University of Extremadura, Cáceres, Spain.
| |
Collapse
|
4
|
Duan H, Shao C, Luo Z, Wang T, Tong L, Liu H, Yao X, Lei J, Zhao J, Gao Y, Jiang T, Yan X. Perioperative sintilimab and neoadjuvant anlotinib plus chemotherapy for resectable non-small-cell lung cancer: a multicentre, open-label, single-arm, phase 2 trial (TD-NeoFOUR trial). Signal Transduct Target Ther 2024; 9:296. [PMID: 39465257 PMCID: PMC11514280 DOI: 10.1038/s41392-024-01992-0] [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: 03/06/2024] [Revised: 09/10/2024] [Accepted: 09/26/2024] [Indexed: 10/29/2024] Open
Abstract
This open-label, single-arm, phase 2 trial evaluated the efficacy and safety of neoadjuvant sintilimab combined with anlotinib and chemotherapy, followed by adjuvant sintilimab, for resectable NSCLC. Forty-five patients received anlotinib (10 mg, QD, PO, days 1-14), sintilimab (200 mg, day 1), and platinum-based chemotherapy of each three-week cycle for 3 cycles, followed by surgery within 4-6 weeks. Adjuvant sintilimab (200 mg) was administered every 3 weeks. The primary endpoint was achieving a pathological complete response (pCR). From June 10, 2021 through October 10, 2023, 45 patients were enrolled and composed the intention-to-treat population. Twenty-six patients (57.8%) achieved pCR, and 30 (66.7%) achieved major pathological response (MPR). Forty-one patients underwent surgery. In the per-protocol set (PP set), 63.4% (26/41) achieved pCR, and 73.2% achieved MPR. The median event-free survival was not attained (95% CI, 25.1-NE). During the neoadjuvant treatment phase, grade 3 or 4 treatment-related adverse events were observed in 25 patients (55.6%), while immune-related adverse events were reported in 7 patients (15.6%). We assessed vascular normalization and infiltration of immune-related cells by detecting the expression of relevant cell markers in NSCLC tissues with mIHC. Significant tumor microenvironment changes were observed in pCR patients, including reduced VEGF+ cells and CD4+Foxp3+ Treg cells, and increased perivascular CD4+ T cells, CD39+CD8+ T cells, and M1 macrophages. In conclusion, perioperative sintilimab and neoadjuvant anlotinib plus chemotherapy achieved pCR in a notable proportion of patients with resectable NSCLC and were associated with profound changes in the tumour microenvironment (ClinicalTrials.gov NCT05400070).
Collapse
Affiliation(s)
- Hongtao Duan
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, No. 1, Xinsi Road, Xi'an, Shaanxi, China
| | - Changjian Shao
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, No. 1, Xinsi Road, Xi'an, Shaanxi, China
| | - Zhilin Luo
- Department of Thoracic Surgery, Third Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Tianhu Wang
- Department of Thoracic Surgery, Third Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Liping Tong
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, No. 1, Xinsi Road, Xi'an, Shaanxi, China
| | - Honggang Liu
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, No. 1, Xinsi Road, Xi'an, Shaanxi, China
| | - Xin Yao
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, No. 1, Xinsi Road, Xi'an, Shaanxi, China
| | - Jie Lei
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, No. 1, Xinsi Road, Xi'an, Shaanxi, China
| | - Jinbo Zhao
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, No. 1, Xinsi Road, Xi'an, Shaanxi, China
| | - Yuan Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, Air Force Medical University, No. 169, Changle West Road, Xi'an, Shaanxi, China.
| | - Tao Jiang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, No. 1, Xinsi Road, Xi'an, Shaanxi, China.
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, No. 1, Xinsi Road, Xi'an, Shaanxi, China.
| |
Collapse
|
5
|
Chirullo B, Fruscione F, Del Zotto G, Dell'Anno F, Tarantino M, Porcellato I, Petrucci P, De Ciucis CG, Bufalari A, Guardone L, Cappelli K, Moretti G, Mecocci S, Monti E, De Paolis L, Razzuoli E. Evaluation of attenuated Salmonella Typhimurium (STMΔznuABC) anticancer activity on canine mammary cancer-associated fibroblasts. Res Vet Sci 2024; 180:105438. [PMID: 39447298 DOI: 10.1016/j.rvsc.2024.105438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/23/2024] [Accepted: 10/13/2024] [Indexed: 10/26/2024]
Abstract
Bacteria-mediated treatments gained increasing attention as alternative therapies against tumors. An attenuated mutant strain of Salmonella enterica serovar Typhimurium (STMΔznuABC) has recently been considered as a potential new anti-cancer strategy. However, it is unclear whether this activity is tumor-induced or species-specific, and no data are available regarding STMΔznuABC on canine mammary tumors (CMTs). This study aimed to investigate the ability of STMΔznuABC in modulating the response of CMTs, focusing on cancer-associated fibroblasts. Four CMT cell lines (CF33, TM51, TM52 TM53) were treated with STMΔznuABC. Then, antiproliferative activity (MTT assay), bacterial invasion, and CMT cell lines gene expression analysis (RT-qPCR) of genes involved in immune response and cancer aggressiveness were evaluated. STMΔznuABC penetrated in TM51, TM52, TM53, and CF33 cell lines, causing a significant reduction of cell viability. Moreover, the expression of several genes was significantly modulated in all CMT cell lines: STMΔznuABC infection determined a significant up-regulation of CXCL8, IL18, IL10, TLR4 and RAD51, while CD14, IL6, CXCR4, P53, PTEN, STAT5, TLR5 and TGFB1 were downregulated in TM53. In CF33, CXCL8 and P53 were upregulated, while MYD88, MD2, IL18, TLR4,5, TGFB1 were downregulated. In TM52, CXCL8, CD44 and MD2 were upregulated and PTEN was downregulated, while in TM51 CXCL8, CD44 and ErbB2 were downregulated. We demonstrated the anti-proliferative and immuno-modulatory activity of STMΔznuABC in CMTs, paving the way for potential new anti-cancer treatments.
Collapse
Affiliation(s)
- Barbara Chirullo
- Unit of Emerging Zoonoses, Department of Food Safety, Nutrition and Veterinary Public Health Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Floriana Fruscione
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy.
| | | | - Filippo Dell'Anno
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy.
| | - Michela Tarantino
- Unit of Emerging Zoonoses, Department of Food Safety, Nutrition and Veterinary Public Health Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Ilaria Porcellato
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Paola Petrucci
- Unit of Emerging Zoonoses, Department of Food Safety, Nutrition and Veterinary Public Health Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Chiara Grazia De Ciucis
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy; Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Via Forlanini 2, 27100 Pavia, Italy.
| | - Antonello Bufalari
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Lisa Guardone
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy; Department of Veterinary Sciences, University of Pisa, Viale delle Piagge 2, 56124 Pisa, Italy.
| | - Katia Cappelli
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Giulia Moretti
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Samanta Mecocci
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Eleonora Monti
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Livia De Paolis
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy; Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Elisabetta Razzuoli
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy.
| |
Collapse
|
6
|
Song Y, Lee Y. Brief guide to flow cytometry. Mol Cells 2024:100129. [PMID: 39426684 DOI: 10.1016/j.mocell.2024.100129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024] Open
Abstract
Flow cytometry is a powerful analytical technique for measuring the physical and chemical properties of cells or particles as they flow through a beam of light in a fluid stream. It is widely used in several research fields for a wide variety of purposes. This article provides a concise guide to the principles and utilization of flow cytometry. Step-by-step instructions, from instrument setup to data analysis are intended to help researchers successfully apply flow cytometry in their studies.
Collapse
Affiliation(s)
- Youngkwon Song
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Yoontae Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, Republic of Korea.
| |
Collapse
|
7
|
Bonilla DL, Paul A, Gil-Pulido J, Park LM, Jaimes MC. The Power of Reagent Titration in Flow Cytometry. Cells 2024; 13:1677. [PMID: 39451195 PMCID: PMC11506663 DOI: 10.3390/cells13201677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/13/2024] [Accepted: 09/28/2024] [Indexed: 10/26/2024] Open
Abstract
Flow cytometry facilitates the detection of multiple cell parameters simultaneously with a high level of resolution and throughput, enabling in-depth immunological evaluations. High data resolution in flow cytometry depends on multiple factors, including the concentration of reagents used in the staining protocol, and reagent validation and titration should be the first step in any assay optimization. Titration is the process of finding the concentration of the reagent that best resolves a positive signal from the background, with the saturation of all binding sites, and minimal antibody excess. The titration process involves the evaluation of serial reagent dilutions in cells expressing the antigen target for the tested antibody. The concentration of antibody that provides the highest signal to noise ratio is calculated by plotting the percentage of positive cells and the intensity of the fluorescence of the stained cells with respect to the negative events, in a concentration-response curve. The determination of the optimal antibody concentration is necessary to ensure reliable and reproducible results and is required for each sample type, reagent clone and lot, as well as the methods used for cell collection, staining, and storage conditions. If the antibody dilution is too low, the signal will be too weak to be accurately determined, leading to suboptimal data resolution, high variability across measurements, and the underestimation of the frequency of cells expressing a specific marker. The use of excess antibodies could lead to non-specific binding, reagent misuse, and detector overloading with the signal off scale and higher spillover spreading. In this publication, we summarized the titration fundamentals and best practices, and evaluated the impact of using a different instrument, sample, staining, acquisition, and analysis conditions in the selection of the optimal titer and population resolution.
Collapse
Affiliation(s)
- Diana L. Bonilla
- Scientific Commercialization, Cytek Biosciences, Inc., 47215 Lakeview Boulevard, Fremont, CA 94538, USA
| | - Alberta Paul
- Scientific Commercialization, Cytek Biosciences, Inc., 47215 Lakeview Boulevard, Fremont, CA 94538, USA
| | - Jesus Gil-Pulido
- Customer Education, Cytek Biosciences, Inc., 47215 Lakeview Boulevard, Fremont, CA 94538, USA
| | - Lily M. Park
- Scientific Commercialization, Cytek Biosciences, Inc., 47215 Lakeview Boulevard, Fremont, CA 94538, USA
| | - Maria C. Jaimes
- Scientific Commercialization, Cytek Biosciences, Inc., 47215 Lakeview Boulevard, Fremont, CA 94538, USA
| |
Collapse
|
8
|
Yuan H, He Y, Zhang Y, Min H, Chen J, Li C. Crystalline silica-induced endoplasmic reticulum stress promotes the pathogenesis of silicosis by augmenting proinflammatory interstitial pulmonary macrophages. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 946:174299. [PMID: 38936737 DOI: 10.1016/j.scitotenv.2024.174299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
Crystalline silica (CS) particles are ubiquitously present in the environment, particularly in occupational settings, and exposure to respirable CS causes silicosis, imposing a significant disease burden. However, the pathogenesis of silicosis remains unclear. Exposure to external stimuli, such as CS, leads to the accumulation of unfolded proteins and triggers endoplasmic reticulum (ER) stress, disrupting tissue immune homeostasis and accelerating pathological progression. While pulmonary macrophages phagocytose CS particles to initiate the immune response, the role of ER stress in this process is unknown. Herein, we used a murine model of silicosis to simulate the pathological progression from acute inflammation to fibrosis in silicosis and conducted in vivo pharmacological inhibition of ER stress to explore the underlying mechanism. Using flow cytometry, we further classified pulmonary macrophages into monocyte-like macrophages (monocytes), interstitial macrophages (IMs), and alveolar macrophages (AMs). Our results showed that CS-induced ER stress primarily contributed to the augmentation of IMs and thereby exerted a significant impact on pulmonary macrophages. Despite coexpressing M1- and M2-like markers, IMs predominantly exhibited an M1-like polarization state and played a proinflammatory role by expressing the cytokines pro-IL-1β and TNF-α during the pathological progression of silicosis. Additionally, IMs recruited by CS-induced ER stress also exhibited high expression of MHCII and exerted active immunomodulatory effects. Overall, our study demonstrates that ER stress induced by CS particles triggers a proinflammatory immune microenvironment dominated by IMs and reveals novel insights into the pulmonary toxicological effects of CS particles.
Collapse
Affiliation(s)
- 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
| | - Yuting Zhang
- 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
|
9
|
Saggau C, Bacher P, Esser D, Rasa M, Meise S, Mohr N, Kohlstedt N, Hutloff A, Schacht SS, Dargvainiene J, Martini GR, Stürner KH, Schröder I, Markewitz R, Hartl J, Hastermann M, Duchow A, Schindler P, Becker M, Bautista C, Gottfreund J, Walter J, Polansky JK, Yang M, Naghavian R, Wendorff M, Schuster EM, Dahl A, Petzold A, Reinhardt S, Franke A, Wieczorek M, Henschel L, Berger D, Heine G, Holtsche M, Häußler V, Peters C, Schmidt E, Fillatreau S, Busch DH, Wandinger KP, Schober K, Martin R, Paul F, Leypoldt F, Scheffold A. Autoantigen-specific CD4 + T cells acquire an exhausted phenotype and persist in human antigen-specific autoimmune diseases. Immunity 2024; 57:2416-2432.e8. [PMID: 39226901 DOI: 10.1016/j.immuni.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/14/2024] [Accepted: 08/07/2024] [Indexed: 09/05/2024]
Abstract
Pro-inflammatory autoantigen-specific CD4+ T helper (auto-Th) cells are central orchestrators of autoimmune diseases (AIDs). We aimed to characterize these cells in human AIDs with defined autoantigens by combining human leukocyte antigen (HLA)-tetramer-based and activation-based multidimensional ex vivo analyses. In aquaporin4-antibody-positive neuromyelitis optica spectrum disorder (AQP4-NMOSD) patients, auto-Th cells expressed CD154, but proliferative capacity and pro-inflammatory cytokines were strongly reduced. Instead, exhaustion-associated co-inhibitory receptors were expressed together with FOXP3, the canonical regulatory T cell (Treg) transcription factor. Auto-Th cells responded in vitro to checkpoint inhibition and provided potent B cell help. Cells with the same exhaustion-like (ThEx) phenotype were identified in soluble liver antigen (SLA)-antibody-autoimmune hepatitis and BP180-antibody-positive bullous pemphigoid, AIDs of the liver and skin, respectively. While originally described in cancer and chronic infection, our data point to T cell exhaustion as a common mechanism of adaptation to chronic (self-)stimulation across AID types and link exhausted CD4+ T cells to humoral autoimmune responses, with implications for therapeutic targeting.
Collapse
Affiliation(s)
- Carina Saggau
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | - Petra Bacher
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany; Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Daniela Esser
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Mahdi Rasa
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany; Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Silja Meise
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | - Nicola Mohr
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | - Nora Kohlstedt
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | - Andreas Hutloff
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany; Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Sarah-Sophie Schacht
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | - Justina Dargvainiene
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Gabriela Rios Martini
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany; Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Klarissa H Stürner
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Lübeck, Germany; Department of Neurology, University Hospital Schleswig-Holstein Kiel, Kiel, Germany
| | - Ina Schröder
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Robert Markewitz
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Johannes Hartl
- Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Maria Hastermann
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Ankelien Duchow
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Patrick Schindler
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Mareike Becker
- Institute of Experimental Dermatology, Lübeck, Germany; Department of Pediatric Dermatology, Catholic Children's Hospital Wilhelmstift, Hamburg, Germany
| | - Carolin Bautista
- Department of Dermatology, Allergy and Venerology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Judith Gottfreund
- Department of Genetics and Epigenetics, Saarland University, Saarbrücken, Germany
| | - Jörn Walter
- Department of Genetics and Epigenetics, Saarland University, Saarbrücken, Germany
| | - Julia K Polansky
- Berlin Institute of Health (BIH) at Charité Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany; German Rheumatism Research Centre, a Leibniz Institute (DRFZ), Charité Platz 1, 10117 Berlin, Germany
| | - Mingxing Yang
- Berlin Institute of Health (BIH) at Charité Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Reza Naghavian
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, Zurich, Switzerland; Cellerys AG, Wagistrasse 21, 8952 Schlieren, Switzerland
| | - Mareike Wendorff
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany; Leibniz Institute for Science and Mathematics Education, Kiel, Germany
| | - Ev-Marie Schuster
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstr. 3/5, 91054 Erlangen, Germany
| | - Andreas Dahl
- DRESDEN-concept Genome Center, Technology Platform at the Center for Molecular and Cellular Bioengineering (CMCB), Technical University of Dresden, Dresden, Germany
| | - Andreas Petzold
- DRESDEN-concept Genome Center, Technology Platform at the Center for Molecular and Cellular Bioengineering (CMCB), Technical University of Dresden, Dresden, Germany
| | - Susanne Reinhardt
- DRESDEN-concept Genome Center, Technology Platform at the Center for Molecular and Cellular Bioengineering (CMCB), Technical University of Dresden, Dresden, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Marek Wieczorek
- Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Straße 68, 51429 Bergisch Gladbach, Germany
| | - Lea Henschel
- Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Straße 68, 51429 Bergisch Gladbach, Germany
| | - Daniel Berger
- Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Straße 68, 51429 Bergisch Gladbach, Germany
| | - Guido Heine
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Maike Holtsche
- Institute of Experimental Dermatology, University of Lübeck, Department of Dermatology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Vivien Häußler
- Clinic and Polyclinic for Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Christian Peters
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | - Enno Schmidt
- Institute of Experimental Dermatology, University of Lübeck, Department of Dermatology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Simon Fillatreau
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, 75015 Paris, France; Université Paris Cité, Faculté de Médecine, Paris, France; AP-HP, Hôpital Necker-Enfants Malades, Paris, France
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany
| | - Klaus-Peter Wandinger
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Kilian Schober
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstr. 3/5, 91054 Erlangen, Germany; Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054 Erlangen, Germany
| | - Roland Martin
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, Zurich, Switzerland; Cellerys AG, Wagistrasse 21, 8952 Schlieren, Switzerland; Institute of Experimental Immunology, University of Zurich, Wintherturerstrasse 191, 8057 Zurich, Switzerland; Department of Clinical Neuroscience, Karolinska Institute, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Friedemann Paul
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Frank Leypoldt
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Lübeck, Germany; Department of Neurology, University Hospital Schleswig-Holstein Kiel, Kiel, Germany
| | - Alexander Scheffold
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany.
| |
Collapse
|
10
|
Dabbah-Krancher G, Ruchinskas A, Kallarakal MA, Lee KP, Bauman BM, Epstein B, Yin H, Krappmann D, Schaefer BC, Snow AL. A20 intrinsically influences human effector T-cell survival and function by regulating both NF-κB and JNK signaling. Eur J Immunol 2024:e2451245. [PMID: 39359035 DOI: 10.1002/eji.202451245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024]
Abstract
A20 is a dual-function ubiquitin-editing enzyme that maintains immune homeostasis by restraining inflammation. Although A20 serves a similar negative feedback function for T-cell receptor (TCR) signaling, the molecular mechanisms utilized and their ultimate impact on human T-cell function remain unclear. TCR engagement triggers the assembly of the CARD11-BCL10-MALT1 (CBM) protein complex, a signaling platform that governs the activation of downstream transcription factors including NF-κB and c-Jun/AP-1. Utilizing WT and A20 knockout Jurkat T cells, we found that A20 is required to negatively regulate NF-κB and JNK. Utilizing a novel set of A20 mutants in NF-κB and AP-1-driven reporter systems, we discovered the ZnF7 domain is crucial for negative regulatory capacity, while deubiquitinase activity is dispensable. Successful inactivation of A20 in human primary effector T cells congruently conferred sustained NF-κB and JNK signaling, including enhanced upregulation of activation markers, and increased secretion of several cytokines including IL-9. Finally, loss of A20 in primary human T cells resulted in decreased sensitivity to restimulation-induced cell death and increased sensitivity to cytokine withdrawal-induced death. These findings demonstrate the importance of A20 in maintaining T-cell homeostasis via negative regulation of both NF-κB and JNK signaling.
Collapse
Affiliation(s)
- Gina Dabbah-Krancher
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, C-2013 Bethesda, MD 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Allison Ruchinskas
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, C-2013 Bethesda, MD 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Melissa A Kallarakal
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, C-2013 Bethesda, MD 20814, USA
| | - Katherine P Lee
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, C-2013 Bethesda, MD 20814, USA
| | - Bradly M Bauman
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, C-2013 Bethesda, MD 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Benjamin Epstein
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, C-2013 Bethesda, MD 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Hongli Yin
- Research Unit Signaling and Translation, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Daniel Krappmann
- Research Unit Signaling and Translation, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Brian C Schaefer
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Andrew L Snow
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, C-2013 Bethesda, MD 20814, USA
| |
Collapse
|
11
|
Schroderus AM, Pitkänen V, Ekman I, Stevens D, Rytkönen-Nissinen M, Rintamäki R, Pihlajamäki J, Knip M, Veijola R, Toppari J, Ilonen J, Lempainen J, Kinnunen T. Temporal Alterations in CD8+ T Cells During the Progression From Stage 1 to Stage 3 Type 1 Diabetes. Diabetes 2024; 73:1705-1715. [PMID: 38967999 DOI: 10.2337/db24-0159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/06/2024] [Indexed: 07/07/2024]
Abstract
CD8+ T cells are perceived to play a major role in the pathogenesis of type 1 diabetes (T1D). In this study, we characterized the function and phenotype of circulating CD8+ memory T cells in samples from individuals at different stages of T1D progression using flow cytometry and single-cell multiomics. We observed two distinct CD8+ T-cell signatures during progression of T1D within the highly differentiated CD27-CD8+ memory T-cell subset. A proinflammatory signature, with an increased frequency of IFN-γ+TNF-α+ CD27-CD8+ memory T cells, was observed in children with newly diagnosed T1D (stage 3) and correlated with the level of dysglycemia at diagnosis. In contrast, a coinhibitory signature, with an increased frequency of KLRG1+TIGIT+ CD27-CD8+ memory T cells, was observed in islet autoantibody-positive children who later progressed to T1D (stage 1). No alterations within CD27-CD8+ memory T cells were observed in adults with established T1D or in children during the initial seroconversion to islet autoantibody positivity. Single-cell multiomics analyses suggested that CD27-CD8+ T cells expressing the IFNG+TNF+ proinflammatory signature may be distinct from those expressing the KLRG1+TIGIT+ coinhibitory signature at the single-cell level. Collectively, our findings suggest that distinct blood CD8+ T-cell signatures could be employed as potential biomarkers of T1D progression. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Anna-Mari Schroderus
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Viola Pitkänen
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Ilse Ekman
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Daniella Stevens
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Marja Rytkönen-Nissinen
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Reeta Rintamäki
- Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
| | - Jussi Pihlajamäki
- Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Mikael Knip
- Tampere Center for Child Health Research, Tampere University Hospital, Tampere, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Riitta Veijola
- Research Unit of Clinical Medicine, Department of Pediatrics, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Jorma Toppari
- Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland
- Research Centre for Integrative Physiology and Pharmacology, InFLAMES Research Flagship, Institute of Biomedicine, University of Turku, Turku, Finland
- Centre for Population Health Research, InFLAMES Research Flagship, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Johanna Lempainen
- Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Tuure Kinnunen
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- ISLAB Laboratory Centre, Kuopio, Finland
| |
Collapse
|
12
|
Matsuura K, Hashioka S, Takata K. Sorting differentiated mammalian cells using deterministic lateral displacement microfluidic devices. ANAL SCI 2024; 40:1801-1807. [PMID: 39060754 DOI: 10.1007/s44211-024-00634-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/14/2024] [Indexed: 07/28/2024]
Abstract
Separation of differentiated and undifferentiated cells without labeling is required for cell analyses and clinical application of cultured differentiated cells in vitro. To proceed with the passive separation of differentiated cells inside a clean bench, we developed a system of deterministic lateral displacement (DLD) microfluidic devices and applied this system to sort differentiated cells in vitro. The fluid flow is driven by compressed air to the buffer. Priming and sorting can be completed by air pressure control. We use this system to separate C2C12 mononuclear myocytes from multinuclear myotubes. Additionally, using a DLD microfluidic channel of Dc = 20 μm, multinuclear myotubes can be effectively sorted as larger particles. We prepared differentiated adipocytes from mouse embryonic fibroblast (MEF) cells and sorted those containing lipid droplets. The diameters of these sorted adipocytes considered larger particles, exceeded 20 μm, similar to the Dc of the DLD microfluidic channel. Differentiated cell sorting by cell size will contribute to single-cell analyses and in vitro tissue model preparation for drug discovery.
Collapse
Affiliation(s)
- Koji Matsuura
- Department of Biosciences, Faculty of Life Science, Okayama University of Science, Okayama, Japan.
| | - Shingi Hashioka
- Medical & Life Science Studio, R&D Center, Zeon Corporation, Toyama, Japan
| | - Koji Takata
- Life Materials Development Section, Human Life Technology Research Institute, Toyama Industrial Technology Research and Development Center, Toyama, Japan
| |
Collapse
|
13
|
Wei X, Wu D, Li J, Wu M, Li Q, Che Z, Cheng X, Cheng Q, Yin F, Zhang H, Wang X, Abtahi S, Zuo L, Hang L, Ma L, Kuo WT, Liu X, Turner JR, Wang H, Xiao J, Wang F. Myeloid beta-arrestin 2 depletion attenuates metabolic dysfunction-associated steatohepatitis via the metabolic reprogramming of macrophages. Cell Metab 2024; 36:2281-2297.e7. [PMID: 39305895 DOI: 10.1016/j.cmet.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/27/2024] [Accepted: 08/26/2024] [Indexed: 10/04/2024]
Abstract
Macrophage-mediated inflammation has been implicated in the pathogenesis of metabolic dysfunction-associated steatohepatitis (MASH); however, the immunometabolic program underlying the regulation of macrophage activation remains unclear. Beta-arrestin 2, a multifunctional adaptor protein, is highly expressed in bone marrow tissues and macrophages and is involved in metabolism disorders. Here, we observed that β-arrestin 2 expression was significantly increased in the liver macrophages and circulating monocytes of patients with MASH compared with healthy controls and positively correlated with the severity of metabolic dysfunction-associated steatotic liver disease (MASLD). Global or myeloid Arrb2 deficiency prevented the development of MASH in mice. Further study showed that β-arrestin 2 acted as an adaptor protein and promoted ubiquitination of immune responsive gene 1 (IRG1) to prevent increased itaconate production in macrophages, which resulted in enhanced succinate dehydrogenase activity, thereby promoting the release of mitochondrial reactive oxygen species and M1 polarization. Myeloid β-arrestin 2 depletion may be a potential approach for MASH.
Collapse
Affiliation(s)
- Xiaoli Wei
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dongqing Wu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jing Li
- College of Life Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Miaomiao Wu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; College of Pharmacy, Anhui Medical University, Hefei, China
| | - Qianhui Li
- Division of Gastroenterology, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Zhaodi Che
- Clinical Medicine Research Institute and Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xu Cheng
- Department of Cardiology, First Affiliated Hospital, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Qianying Cheng
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fan Yin
- Department of Pharmacy, Huainan First People's Hospital, The First Affiliated Hospital of Anhui University of Science and Technology, Huainan, China
| | - Hao Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xuefu Wang
- College of Pharmacy, Anhui Medical University, Hefei, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Shabnam Abtahi
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA; Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Li Zuo
- School of Basic Medical Sciences, Molecular Biology Laboratory, Anhui Medical University, Hefei, China; Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
| | - Lei Hang
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
| | - Lili Ma
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
| | - Wei-Ting Kuo
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA; Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Xiaoying Liu
- College of Life Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Jerrold R Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA; Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China; Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China.
| | - Jia Xiao
- Clinical Medicine Research Institute and Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China.
| | - Fei Wang
- Division of Gastroenterology, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
14
|
Valenzano G, Russell SN, Go S, O'Neill E, Jones KI. Using Spectral Flow Cytometry to Characterize Anti-Tumor Immunity in Orthotopic and Subcutaneous Mouse Models of Cancer. Curr Protoc 2024; 4:e70032. [PMID: 39432378 DOI: 10.1002/cpz1.70032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Mouse models remain at the forefront of immuno-oncology research, providing invaluable insights into the complex interactions between the immune system and developing tumors. While several flow cytometry panels have been developed to study cancer immunity in mice, most are limited in their capacity to address the complexity of anti-cancer immune responses. For example, many of the panels developed to date focus on a restricted number of leukocyte populations (T cells or antigen-presenting cells), failing to include the multitude of other subsets that participate in anti-cancer immunity. In addition, these panels were developed using blood or splenic leukocytes. While the immune composition of the blood or spleen can provide information on systemic immune responses to cancer, it is in the tumor microenvironment (TME) that local immunity takes place. Therefore, we optimized this spectral flow cytometry panel to identify the chief cell types that take part in cancer immunity using immune cells from cancer tissue. We used pancreatic tumors implanted both orthotopically and subcutaneously to demonstrate the panel's flexibility and suitability in diverse mouse models. The panel was also validated in peripheral immune districts (the blood, spleen, and liver of tumor-bearing mice) to allow comparisons between local and systemic anti-tumor immunity. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Tumor induction-Orthotopic Alternate Protocol: Tumor induction-Subcutaneous Basic Protocol 2: Preparation of single-cell suspensions from the tumor, spleen, liver, and blood of tumor-bearing mice Basic Protocol 3: Staining single-cell suspensions from the tumor, spleen, liver, and blood of tumor-bearing mice.
Collapse
Affiliation(s)
| | | | - Simei Go
- Department of Oncology, University of Oxford, Oxford, UK
| | - Eric O'Neill
- Department of Oncology, University of Oxford, Oxford, UK
| | - Keaton I Jones
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
15
|
Li Y, Du X, Hu Y, Wang D, Duan L, Zhang H, Zhang R, Xu Y, Zhou R, Zhang X, Zhang M, Liu J, Lv Z, Chen Y, Wang W, Sun Y, Cui Y. Iron-laden macrophage-mediated paracrine profibrotic signaling induces lung fibroblast activation. Am J Physiol Cell Physiol 2024; 327:C979-C993. [PMID: 39183565 DOI: 10.1152/ajpcell.00675.2023] [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: 12/06/2023] [Revised: 07/22/2024] [Accepted: 08/15/2024] [Indexed: 08/27/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating condition characterized by progressive lung scarring and uncontrolled fibroblast proliferation, inevitably leading to organ dysfunction and mortality. Although elevated iron levels have been observed in patients and animal models of lung fibrosis, the mechanisms linking iron dysregulation to lung fibrosis pathogenesis, particularly the role of macrophages in orchestrating this process, remain poorly elucidated. Here we evaluate iron metabolism in macrophages during pulmonary fibrosis using both in vivo and in vitro approaches. In murine bleomycin- and amiodarone-induced pulmonary fibrosis models, we observed significant iron deposition and lipid peroxidation in pulmonary macrophages. Intriguingly, the ferroptosis regulator glutathione peroxidase 4 (GPX4) was upregulated in pulmonary macrophages following bleomycin instillation, a finding corroborated by single-cell RNA sequencing analysis. Moreover, macrophages isolated from fibrotic mouse lungs exhibited increased transforming growth factor (TGF)-β1 expression that correlated with lipid peroxidation. In vitro, iron overload in bone marrow-derived macrophages triggered lipid peroxidation and TGF-β1 upregulation, which was effectively suppressed by ferroptosis inhibitors. When cocultured with iron-overloaded macrophages, lung fibroblasts exhibited heightened activation, evidenced by increased α-smooth muscle actin and fibronectin expression. Importantly, this profibrotic effect was attenuated by treating macrophages with a ferroptosis inhibitor or blocking TGF-β receptor signaling in fibroblasts. Collectively, our study elucidates a novel mechanistic paradigm in which the accumulation of iron within macrophages initiates lipid peroxidation, thereby amplifying TGF-β1 production, subsequently instigating fibroblast activation through paracrine signaling. Thus, inhibiting iron overload and lipid peroxidation warrants further exploration as a strategy to suppress fibrotic stimulation by disease-associated macrophages. NEW & NOTEWORTHY This study investigates the role of iron in pulmonary fibrosis, specifically focusing on macrophage-mediated mechanisms. Iron accumulation in fibrotic lung macrophages triggers lipid peroxidation and an upregulation of transforming growth factor (TGF)-β1 expression. Coculturing iron-laden macrophages activates lung fibroblasts in a TGF-β1-dependent manner, which can be mitigated by ferroptosis inhibitors. These findings underscore the potential of targeting iron overload and lipid peroxidation as a promising strategy to alleviate fibrotic stimulation provoked by disease-associated macrophages.
Collapse
Affiliation(s)
- Yunqi Li
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Xinqian Du
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Yue Hu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Dan Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Luo Duan
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Hanxiao Zhang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Ruoyang Zhang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People's Republic of China
- National Center for Respiratory Medicine, Beijing, People's Republic of China
| | - Yingjie Xu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Ruonan Zhou
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Xinyu Zhang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Muzhi Zhang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Jie Liu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Zhe Lv
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Yan Chen
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Wei Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Ying Sun
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Ye Cui
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
16
|
Liang J, Ma M, Feng W, Xu Q, Chen D, Lai J, Chen J. Anti-PD-L1 blockade facilitates antitumor effects of radiofrequency ablation by improving tumor immune microenvironment in hepatocellular carcinoma. Apoptosis 2024:10.1007/s10495-024-02019-3. [PMID: 39327353 DOI: 10.1007/s10495-024-02019-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2024] [Indexed: 09/28/2024]
Abstract
Hepatocellular carcinoma (HCC) is a complex disease with advanced presentation that significantly affects survival rates. Therefore, novel therapeutic strategies are needed. In this study, we investigate the tumor microenvironment (TME) in HCC by analyzing 13 HCC samples at single cell level. We identified key cell populations, including CD8 + T cells, Tregs, M1/M2 macrophages, and CD4 + memory T cells, and explored their roles and interactions. Our research revealed an early enrichment of CD8 + T cells, which could potentially lead to their exhaustion and facilitate tumor progression. We also investigated the impact of percutaneous radiofrequency ablation (RFA) on the immune microenvironment. Using a dual tumor mouse model, we demonstrated that RFA induces necrosis, enhancing antigen presentation and altering immune responses. Our results indicate that RFA increases PD-L1 expression in residual liver tissue, suggesting potential immune escape mechanisms. Furthermore, the combination of RFA and anti-PD-L1 therapy in the mouse model resulted in significant improvements in immune modulation. This included increased CD8 + T cell efficacy and decreased Treg infiltration. This combination shows promise as an approach to counteract HCC progression by altering the immune landscape. This study highlights the critical interaction within the TME of HCC and suggests the possibility of improving patient outcomes by targeting immune evasion mechanisms through combined therapeutic strategies.
Collapse
Affiliation(s)
- Jiahua Liang
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Medicine II, Medical Faculty Mannheim, Universitätsmedizin Mannheim, Heidelberg University, Mannheim, Germany
| | - Mingjian Ma
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Wei Feng
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qiongcong Xu
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Dong Chen
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jiaming Lai
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Jiancong Chen
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
17
|
Amon L, Seichter A, Vurnek D, Tchitashvili G, Heß I, Heger L, Lehmann CHK, Dudziak D. Protocol for mapping the heterogeneous dendritic cell network across the murine tissue landscape via high-dimensional flow cytometry. STAR Protoc 2024; 5:103151. [PMID: 38990726 PMCID: PMC11295986 DOI: 10.1016/j.xpro.2024.103151] [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: 04/04/2024] [Revised: 05/11/2024] [Accepted: 06/05/2024] [Indexed: 07/13/2024] Open
Abstract
Dendritic cells (DCs) populate nearly all tissues and represent the central orchestrators of immunity. Here, we present a protocol for the mild but efficient preparation of single-cell suspensions from multiple murine tissues and the downstream analysis of the DC network via high-parameter flow cytometry. Additionally, we provide evaluation strategies that facilitate the stringent separation of the DC family from other myeloid cells, particularly macrophages and monocytes, and include an in-depth assessment of DC-intrinsic heterogeneity. For complete details on the use and execution of this protocol, please refer to Amon et al.1.
Collapse
Affiliation(s)
- Lukas Amon
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052 Erlangen, Germany
| | - Anna Seichter
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052 Erlangen, Germany
| | - Damir Vurnek
- Institute of Immunology, Jena University Hospital, Friedrich-Schiller-University Jena, 07743 Jena, Germany
| | - Giorgi Tchitashvili
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052 Erlangen, Germany
| | - Isabel Heß
- University Hospital Erlangen, 91054 Erlangen, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052 Erlangen, Germany
| | - Christian H K Lehmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052 Erlangen, Germany; Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany; FAU Profile Center Immunomedicine (FAU I-MED), 91054 Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany; Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCCER-EMN), 91054 Erlangen, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052 Erlangen, Germany; Institute of Immunology, Jena University Hospital, Friedrich-Schiller-University Jena, 07743 Jena, Germany; FAU Profile Center Immunomedicine (FAU I-MED), 91054 Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany; Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCCER-EMN), 91054 Erlangen, Germany; Comprehensive Cancer Center Central Germany Jena/Leipzig (CCCG), 07743 Jena, Germany.
| |
Collapse
|
18
|
Arlt E, Kindermann A, Fritsche AK, Navarrete Santos A, Kielstein H, Bazwinsky-Wutschke I. A Flow Cytometry-Based Examination of the Mouse White Blood Cell Differential in the Context of Age and Sex. Cells 2024; 13:1583. [PMID: 39329764 PMCID: PMC11430320 DOI: 10.3390/cells13181583] [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: 07/31/2024] [Revised: 09/10/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
Analysis of the white blood cell differential as part of a flow cytometry-based approach is a common routine diagnostic tool used in clinics and research. For human blood, the methodological approach, suitable markers, and gating strategies are well-established. However, there is a lack of information regarding the mouse blood count. In this article, we deliver a fast and easy protocol for reprocessing mouse blood for the purpose of flow cytometric analysis, as well as suitable markers and gating strategies. We also present two possible applications: for the analysis of the whole blood count, with blood from a cardiac puncture, and for the analysis of a certain leukocyte subset at multiple time points in the framework of a mouse experiment, using blood from the facial vein. Additionally, we provide orientation values by applying the method to 3-month-old and 24-month-old male and female C57BL/6J mice. Our analyses demonstrate differences in the leukocyte fractions depending on age and sex. We discuss the influencing factors and limitations that can affect the results and that, therefore, need to be considered when applying this method. The present study fills the gap in the knowledge related to the rare information on flow cytometric analysis of mouse blood and, thus, lays the foundation for further investigations in this area.
Collapse
Affiliation(s)
- Elise Arlt
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle (Saale), Germany; (A.K.); (A.-K.F.); (H.K.); (I.B.-W.)
| | - Andrea Kindermann
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle (Saale), Germany; (A.K.); (A.-K.F.); (H.K.); (I.B.-W.)
| | - Anne-Kristin Fritsche
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle (Saale), Germany; (A.K.); (A.-K.F.); (H.K.); (I.B.-W.)
- Institute of Anatomy, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany
| | - Alexander Navarrete Santos
- Core Facility Flow Cytometry, Center for Basic Medical Research, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle (Saale), Germany;
| | - Heike Kielstein
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle (Saale), Germany; (A.K.); (A.-K.F.); (H.K.); (I.B.-W.)
| | - Ivonne Bazwinsky-Wutschke
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle (Saale), Germany; (A.K.); (A.-K.F.); (H.K.); (I.B.-W.)
| |
Collapse
|
19
|
Zakeri A, Zhang K, Wang LL. Protocol for analyzing arginase I expression in tumor-associated myeloid-derived suppressor cells from murine colon cancer using flow cytometry. STAR Protoc 2024; 5:103222. [PMID: 39088325 PMCID: PMC11342165 DOI: 10.1016/j.xpro.2024.103222] [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: 03/28/2024] [Revised: 05/27/2024] [Accepted: 07/03/2024] [Indexed: 08/03/2024] Open
Abstract
Arginase1 (ARG1) is a metabolic enzyme that is highly expressed in tumor-associated myeloid-derived suppressor cells (MDSCs) and causes the dysfunction of tumor-reactive T cells. Here, we present a protocol for detecting ARG1 expression in tumor MDSCs from a murine model of colon cancer using flow cytometry. We describe steps for tumor tissue processing, antibody staining, and data acquisition. We then detail procedures for identifying MDSC subsets and detecting ARG1 expression using a precise gating strategy. For complete details on the use and execution of this protocol, please refer to Zhang et al.1.
Collapse
Affiliation(s)
- Amin Zakeri
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Keman Zhang
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Li Lily Wang
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA; Case Comprehensive Cancer Center, Cleveland, OH, USA.
| |
Collapse
|
20
|
Bocanegra-Zamora F, Espinosa-Bautista F, Jiménez-Rodríguez GM, Masso F, Paez A, Gonzalez-Pacheco H, Patlán M, Eid-Lidt G, Amezcua-Guerra LM. Senescent CD4+ T-Cell Phenotypes and Inflammatory Milieu in the Coronary and Systemic Circulation in ST-Elevation Myocardial Infarction: An Exploratory Study. J Vasc Res 2024; 61:260-266. [PMID: 39293415 DOI: 10.1159/000541069] [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: 04/17/2024] [Accepted: 08/20/2024] [Indexed: 09/20/2024] Open
Abstract
INTRODUCTION In ST-elevation myocardial infarction (STEMI), inflammation is pivotal, with early senescent CD4+CD28null cells implicated in its pathogenesis. However, the functional phenotype of these cells within the coronary circulation remains unclear. METHODS We examined CD4+ cell subpopulations in blood samples from the coronary sinus and vena cava of 24 STEMI patients and the cephalic vein of seven healthy controls. RESULTS Our findings revealed reduced CD4+ cell counts in STEMI patients compared to controls (1,998, 1,275-3,268 vs. 4,278, 3,595-4,449), alongside an increased proportion of CD4+ cells lacking CD28 expression (20.1 vs. 6.1%). These CD4+CD28null cells in STEMI predominantly exhibited a Th1 phenotype (47.8% vs. 6.6%). Intriguingly, no significant differences were detected in CD4+CD28null cells between coronary sinus and vena cava, and cytokine levels in these compartments remained similar. CONCLUSION CD4+CD28null cells are increased in STEMI, mainly polarized toward a Th1 phenotype, and distributed equally between the different vascular beds.
Collapse
Affiliation(s)
| | - Fernanda Espinosa-Bautista
- Immunology Department, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
- Master in Chemobiological Sciences, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Gian M Jiménez-Rodríguez
- Interventional Cardiology Department, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Felipe Masso
- UNAM/INC Translational Research Unit, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Araceli Paez
- UNAM/INC Translational Research Unit, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | | | - Mariana Patlán
- Basic Research Sub directorate, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Guering Eid-Lidt
- Interventional Cardiology Department, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Luis M Amezcua-Guerra
- Immunology Department, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
- Health Care Department, Universidad Autónoma Metropolitana-Xochimilco, Mexico City, Mexico
| |
Collapse
|
21
|
Manfroi B, Cuc BT, Sokal A, Vandenberghe A, Temmam S, Attia M, El Behi M, Camaglia F, Nguyen NT, Pohar J, Salem-Wehbe L, Pottez-Jouatte V, Borzakian S, Elenga N, Galeotti C, Morelle G, de Truchis de Lays C, Semeraro M, Romain AS, Aubart M, Ouldali N, Mahuteau-Betzer F, Beauvineau C, Amouyal E, Berthaud R, Crétolle C, Arnould MD, Faye A, Lorrot M, Benoist G, Briand N, Courbebaisse M, Martin R, Van Endert P, Hulot JS, Blanchard A, Tartour E, Leite-de-Moraes M, Lezmi G, Ménager M, Luka M, Reynaud CA, Weill JC, Languille L, Michel M, Chappert P, Mora T, Walczak AM, Eloit M, Bacher P, Scheffold A, Mahévas M, Sermet-Gaudelus I, Fillatreau S. Preschool-age children maintain a distinct memory CD4 + T cell and memory B cell response after SARS-CoV-2 infection. Sci Transl Med 2024; 16:eadl1997. [PMID: 39292802 DOI: 10.1126/scitranslmed.adl1997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 07/19/2024] [Indexed: 09/20/2024]
Abstract
The development of the human immune system lasts for several years after birth. The impact of this maturation phase on the quality of adaptive immunity and the acquisition of immunological memory after infection at a young age remains incompletely defined. Here, using an antigen-reactive T cell (ARTE) assay and multidimensional flow cytometry, we profiled circulating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-reactive CD3+CD4+CD154+ T cells in children and adults before infection, during infection, and 11 months after infection, stratifying children into separate age groups and adults according to disease severity. During SARS-CoV-2 infection, children younger than 5 years old displayed a lower antiviral CD4+ T cell response, whereas children older than 5 years and adults with mild disease had, quantitatively and phenotypically, comparable virus-reactive CD4+ T cell responses. Adults with severe disease mounted a response characterized by higher frequencies of virus-reactive proinflammatory and cytotoxic T cells. After SARS-CoV-2 infection, preschool-age children not only maintained neutralizing SARS-CoV-2-reactive antibodies postinfection comparable to adults but also had phenotypically distinct memory T cells displaying high inflammatory features and properties associated with migration toward inflamed sites. Moreover, preschool-age children had markedly fewer circulating virus-reactive memory B cells compared with the other cohorts. Collectively, our results reveal unique facets of antiviral immunity in humans at a young age and indicate that the maturation of adaptive responses against SARS-CoV-2 toward an adult-like profile occurs in a progressive manner.
Collapse
Affiliation(s)
- Benoît Manfroi
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
| | - Bui Thi Cuc
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
| | - Aurélien Sokal
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, F-75015 Paris, France
- Service de Médecine interne, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris (AP-HP), 92110 Clichy, France
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
| | - Alexis Vandenberghe
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, F-75015 Paris, France
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
- INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
| | - Sarah Temmam
- Pathogen Discovery Laboratory, Institut Pasteur, Université Paris Cité, and Institut Pasteur, the WOAH Collaborating Center for the Detection and Identification in Humans of Emerging Animal Pathogens, Université Paris Cité, 75015 Paris, France
| | - Mikaël Attia
- Molecular Genetics of RNA Viruses, Department of Virology, Institut Pasteur, Université Paris-Cité, CNRS UMR 3569, 75015 Paris, France
| | - Mohamed El Behi
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
| | - Francesco Camaglia
- Laboratoire de physique de l'École normale supérieure, CNRS, Paris Sciences et Lettres (PSL) University, Sorbonne Université, and Université de Paris, 75005 Paris, France
| | - Ngan Thu Nguyen
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
| | - Jelka Pohar
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Immunology and Cellular Immunotherapy (ICI) Group, Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, 1000 Ljubljana, Slovenia
| | - Layale Salem-Wehbe
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
| | - Valentine Pottez-Jouatte
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
| | - Sibyline Borzakian
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- CNRS UMR 9187, INSERM U1196, Chemistry and Modeling for the Biological of Cancer, Institut Curie, PSL Research University, 91405 Orsay, France
- Université Paris-Saclay, 91405 Orsay, France
| | - Narcisse Elenga
- Service de Pédiatrie, Centre Hospitalier de Cayenne, 97300 French Guiana
| | - Caroline Galeotti
- Department of Pediatric Rheumatology, Bicêtre Hospital, AP-HP, Paris-Saclay University, 94275 Le Kremlin-Bicêtre, France
| | - Guillaume Morelle
- Department of General Paediatrics, Hôpital Bicêtre, AP-HP, University of Paris Saclay, 94275 Le Kremlin-Bicêtre, France
| | - Camille de Truchis de Lays
- Service de Pédiatrie. Hôpital Jean-Verdier, AP-HP, Hôpitaux Universitaires Paris Seine-Saint-Denis, 93140 Bondy, France
| | - Michaela Semeraro
- University of Paris Cité, and Clinical Investigation Center, Clinical Research Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Anne-Sophie Romain
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Trousseau Hospital, General Paediatrics Department, 75012 Paris, France
| | - Mélodie Aubart
- INSERM U1163, Genetic Predisposition to Infectious Diseases, Imagine Institute, Université Paris Cité, Paris F-75015, France
- Pediatric Neurology Department, Necker-Enfants Malades Universitary Hospital, AP-HP, Paris-Cité University, 75015 Paris, France
| | - Naim Ouldali
- Department of General Pediatrics, Pediatric Infectious Disease and Internal Medicine, Robert Debré University Hospital, Assistance Publique-Hôpitaux de Paris, 75019 Paris, France
- Paris Cité University, INSERM UMR 1137, Infection, Antimicrobials, Modelling, Evolution (IAME), 75018 Paris, France
| | - Florence Mahuteau-Betzer
- CNRS UMR 9187, INSERM U1196, Chemistry and Modeling for the Biological of Cancer, Institut Curie, PSL Research University, 91405 Orsay, France
- Université Paris-Saclay, 91405 Orsay, France
| | - Claire Beauvineau
- CNRS UMR 9187, INSERM U1196, Chemistry and Modeling for the Biological of Cancer, Institut Curie, PSL Research University, 91405 Orsay, France
- Université Paris-Saclay, 91405 Orsay, France
| | - Elsa Amouyal
- SIREDO Pediatric Oncology Center, Institut Curie, Paris-Science Lettres University, 75005 Paris, France
| | - Romain Berthaud
- Pediatric Nephrology, Maladies Rénales Héréditaires de l'Enfant et de l'Adulte (MARHEA) Reference Center, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
- Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), 75015 Paris, France
| | - Célia Crétolle
- Département de Pédiatrie, Service de Chirurgie viscérale pédiatrique, Hôpital Universitaire Necker-Enfants Malades, GH Paris Centre, 75015 Paris, France
| | - Marc Duval Arnould
- Department of General Paediatrics, Hôpital Bicêtre, AP-HP, University of Paris Saclay, 94275 Le Kremlin-Bicêtre, France
| | - Albert Faye
- Pediatric Neurology Department, Necker-Enfants Malades Universitary Hospital, AP-HP, Paris-Cité University, 75015 Paris, France
| | - Mathie Lorrot
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Trousseau Hospital, General Paediatrics Department, 75012 Paris, France
| | - Grégoire Benoist
- Service de pédiatrie générale et hôpital de jour allergologie, CHU Ambroise-Paré, AP-HP, 92100 Boulogne-Billancourt, France
| | - Nelly Briand
- University of Paris Cité, and Clinical Investigation Center, Clinical Research Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Marie Courbebaisse
- Faculté de Médecine, Université Paris Cité, 75015 Paris, France
- Explorations fonctionnelles rénales, Physiologie, Hôpital européen Georges-Pompidou, Assistance Publique-Hôpitaux de Paris, 75908 Paris Cedex 15, France
| | - Roland Martin
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
- Therapeutic Immune Design, Center for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institute, 171 76 Stockholm, Sweden
| | - Peter Van Endert
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Service Immunologie Biologique, AP-HP, Hôpital Universitaire Necker-Enfants Malades, F-75015 Paris, France
| | - Jean-Sébastien Hulot
- PARCC, INSERM, Université Paris Cité, 75015 Paris, France
- Centre d'Investigation Clinique, AP-HP, INSERM CIC-1418, Européen Georges Pompidou Hospital, 75015 Paris, France
| | - Anne Blanchard
- Centre d'Investigation Clinique, AP-HP, INSERM CIC-1418, Européen Georges Pompidou Hospital, 75015 Paris, France
- Sorbonne Paris Cité, Paris Descartes University, 75015 Paris, France
| | - Eric Tartour
- Pediatric Nephrology, Maladies Rénales Héréditaires de l'Enfant et de l'Adulte (MARHEA) Reference Center, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
- PARCC, INSERM, Université Paris Cité, 75015 Paris, France
- Department of Immunology, Hôpital Européen Georges-Pompidou, AP-HP, CEDEX 15, 75908 Paris, France
| | - Maria Leite-de-Moraes
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
| | - Guillaume Lezmi
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- AP-HP, Hôpital Necker-Enfants Malades, Service de Pneumologie et Allergologie Pédiatriques, 75015 Paris, France
| | - Mickael Ménager
- Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, Université Paris Cité, Imagine Institute, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, 75015 Paris, France
| | - Marine Luka
- Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, Université Paris Cité, Imagine Institute, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, 75015 Paris, France
| | - Claude-Agnès Reynaud
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, F-75015 Paris, France
| | - Jean-Claude Weill
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, F-75015 Paris, France
| | - Laetitia Languille
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
| | - Marc Michel
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
| | - Pascal Chappert
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, F-75015 Paris, France
- INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
| | - Thierry Mora
- Laboratoire de physique de l'École normale supérieure, CNRS, Paris Sciences et Lettres (PSL) University, Sorbonne Université, and Université de Paris, 75005 Paris, France
| | - Aleksandra M Walczak
- Laboratoire de physique de l'École normale supérieure, CNRS, Paris Sciences et Lettres (PSL) University, Sorbonne Université, and Université de Paris, 75005 Paris, France
| | - Marc Eloit
- Pathogen Discovery Laboratory, Institut Pasteur, Université Paris Cité, and Institut Pasteur, the WOAH Collaborating Center for the Detection and Identification in Humans of Emerging Animal Pathogens, Université Paris Cité, 75015 Paris, France
- Ecole Nationale Vétérinaire d'Alfort, University of Paris-Est, 94700 Maisons-Alfort, France
| | - Petra Bacher
- Institute of Immunology, Christian-Albrecht Universität zu Kiel and UKSH Schleswig-Holstein, 24105 Kiel, Germany
- Institute of Clinical Molecular Biology, Christian-Albrecht University of Kiel and UKSH Schleswig-Holstein, 24105 Kiel, Germany
| | - Alexander Scheffold
- Institute of Immunology, Christian-Albrecht Universität zu Kiel and UKSH Schleswig-Holstein, 24105 Kiel, Germany
| | - Matthieu Mahévas
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, F-75015 Paris, France
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
- INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
| | - Isabelle Sermet-Gaudelus
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Reference Center for Rare Diseases: Cystic Fibrosis and Other Epithelial Respiratory Protein Misfolding Diseases, Hôpital Necker-Enfants Malades, AP-HP Centre Université Paris Cité, 75015 Paris, France
| | - Simon Fillatreau
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), 75015 Paris, France
- Faculté de Médecine, Université Paris Cité, 75015 Paris, France
- Service Immunologie Biologique, AP-HP, Hôpital Universitaire Necker-Enfants Malades, F-75015 Paris, France
| |
Collapse
|
22
|
Kanno H, Hiramatsu K, Mikami H, Nakayashiki A, Yamashita S, Nagai A, Okabe K, Li F, Yin F, Tominaga K, Bicer OF, Noma R, Kiani B, Efa O, Büscher M, Wazawa T, Sonoshita M, Shintaku H, Nagai T, Braun S, Houston JP, Rashad S, Niizuma K, Goda K. High-throughput fluorescence lifetime imaging flow cytometry. Nat Commun 2024; 15:7376. [PMID: 39231964 PMCID: PMC11375057 DOI: 10.1038/s41467-024-51125-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 07/31/2024] [Indexed: 09/06/2024] Open
Abstract
Flow cytometry is a vital tool in biomedical research and laboratory medicine. However, its accuracy is often compromised by undesired fluctuations in fluorescence intensity. While fluorescence lifetime imaging microscopy (FLIM) bypasses this challenge as fluorescence lifetime remains unaffected by such fluctuations, the full integration of FLIM into flow cytometry has yet to be demonstrated due to speed limitations. Here we overcome the speed limitations in FLIM, thereby enabling high-throughput FLIM flow cytometry at a high rate of over 10,000 cells per second. This is made possible by using dual intensity-modulated continuous-wave beam arrays with complementary modulation frequency pairs for fluorophore excitation and acquiring fluorescence lifetime images of rapidly flowing cells. Moreover, our FLIM system distinguishes subpopulations in male rat glioma and captures dynamic changes in the cell nucleus induced by an anti-cancer drug. FLIM flow cytometry significantly enhances cellular analysis capabilities, providing detailed insights into cellular functions, interactions, and environments.
Collapse
Affiliation(s)
- Hiroshi Kanno
- Department of Chemistry, The University of Tokyo, Tokyo, Japan.
- Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, Miyagi, Japan.
| | - Kotaro Hiramatsu
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
- Department of Chemistry, Kyushu University, Fukuoka, Japan
| | - Hideharu Mikami
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
- Research Institute for Electronic Science, Hokkaido University, Hokkaido, Japan
| | - Atsushi Nakayashiki
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Shota Yamashita
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Arata Nagai
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Kohki Okabe
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Fan Li
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | - Fei Yin
- Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Keita Tominaga
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | | | - Ryohei Noma
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, Osaka, Japan
| | - Bahareh Kiani
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Olga Efa
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Martin Büscher
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Tetsuichi Wazawa
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, Osaka, Japan
| | | | - Hirofumi Shintaku
- Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Takeharu Nagai
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, Osaka, Japan
| | - Sigurd Braun
- Institute for Genetics, Justus-Liebig-University Giessen, Giessen, Germany
| | - Jessica P Houston
- Department of Chemical and Materials Engineering, New Mexico State University, Las Cruces, NM, USA
| | - Sherif Rashad
- Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Department of Neurosurgical Engineering and Translational Neuroscience Graduate School of Biomedical Engineering, Tohoku University, Miyagi, Japan
| | - Kuniyasu Niizuma
- Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Department of Neurosurgical Engineering and Translational Neuroscience Graduate School of Biomedical Engineering, Tohoku University, Miyagi, Japan
| | - Keisuke Goda
- Department of Chemistry, The University of Tokyo, Tokyo, Japan.
- Institute of Technological Sciences, Wuhan University, Hubei, China.
- Department of Bioengineering, University of California, Los Angeles, CA, USA.
| |
Collapse
|
23
|
Yu Z, Sasidharan-Nair V, Buchta T, Bonifacius A, Khan F, Pietzsch B, Ahmadi H, Beckstette M, Niemz J, Hilgendorf P, Mausberg P, Keller A, Falk C, Busch DH, Schober K, Cicin-Sain L, Müller F, Brinkmann MM, Eiz-Vesper B, Floess S, Huehn J. DNA methylation profiling identifies TBKBP1 as potent amplifier of cytotoxic activity in CMV-specific human CD8+ T cells. PLoS Pathog 2024; 20:e1012581. [PMID: 39325839 PMCID: PMC11460711 DOI: 10.1371/journal.ppat.1012581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 10/08/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024] Open
Abstract
Epigenetic mechanisms stabilize gene expression patterns during CD8+ T cell differentiation. Although adoptive transfer of virus-specific T cells is clinically applied to reduce the risk of virus infection or reactivation in immunocompromised individuals, the DNA methylation pattern of virus-specific CD8+ T cells is largely unknown. Hence, we here performed whole-genome bisulfite sequencing of cytomegalovirus-specific human CD8+ T cells and found that they display a unique DNA methylation pattern consisting of 79 differentially methylated regions (DMRs) when compared to memory CD8+ T cells. Among the top demethylated DMRs in cytomegalovirus-specific CD8+ T cells was TBKBP1, coding for TBK-binding protein 1 that can interact with TANK-binding kinase 1 (TBK1) and mediate pro-inflammatory responses in innate immune cells downstream of intracellular virus sensing. Since TBKBP1 has not yet been reported in T cells, we aimed to unravel its role in virus-specific CD8+ T cells. TBKBP1 demethylation in terminal effector CD8+ T cells correlated with higher TBKBP1 expression at both mRNA and protein level, independent of alternative splicing of TBKBP1 transcripts. Notably, the distinct DNA methylation patterns in CD8+ T cell subsets was stable upon long-term in vitro culture. TBKBP1 overexpression resulted in enhanced TBK1 phosphorylation upon stimulation of CD8+ T cells and significantly improved their virus neutralization capacity. Collectively, our data demonstrate that TBKBP1 modulates virus-specific CD8+ T cell responses and could be exploited as therapeutic target to improve adoptive T cell therapies.
Collapse
Affiliation(s)
- Zheng Yu
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Varun Sasidharan-Nair
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Thalea Buchta
- Institute of Genetics, Technische Universität Braunschweig, Braunschweig, Germany
- Research Group Virology and Innate Immunity, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Agnes Bonifacius
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Thematical Translation Unit-Immunocompromised Host (TTU-IICH), partner site Hannover-Braunschweig, Germany
| | - Fawad Khan
- Department Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine (CIIM), a joint venture of HZI and Hannover Medical School, Hannover, Germany
| | - Beate Pietzsch
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Hosein Ahmadi
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Michael Beckstette
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jana Niemz
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Philipp Hilgendorf
- Mikrobiologisches Institut–Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Philip Mausberg
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Andreas Keller
- Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Saarland University, Saarbrücken, Germany
| | - Christine Falk
- German Center for Infection Research (DZIF), Thematical Translation Unit-Immunocompromised Host (TTU-IICH), partner site Hannover-Braunschweig, Germany
- Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany
| | - Dirk H. Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University Munich (TUM), Munich, Germany
- German Center for Infection Research (DZIF), Thematical Translation Unit-Immunocompromised Host (TTU-IICH), partner site Munich, Germany
| | - Kilian Schober
- Mikrobiologisches Institut–Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- FAU Profile Center Immunomedicine, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Luka Cicin-Sain
- German Center for Infection Research (DZIF), Thematical Translation Unit-Immunocompromised Host (TTU-IICH), partner site Hannover-Braunschweig, Germany
- Department Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine (CIIM), a joint venture of HZI and Hannover Medical School, Hannover, Germany
| | - Fabian Müller
- Integrative Cellular Biology and Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Melanie M. Brinkmann
- Institute of Genetics, Technische Universität Braunschweig, Braunschweig, Germany
- Research Group Virology and Innate Immunity, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Britta Eiz-Vesper
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Thematical Translation Unit-Immunocompromised Host (TTU-IICH), partner site Hannover-Braunschweig, Germany
| | - Stefan Floess
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jochen Huehn
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Cluster of Excellence Resolving Infection Susceptibility (RESIST; EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
24
|
Ekman I, Schroderus AM, Vuorinen T, Knip M, Veijola R, Toppari J, Ilonen J, Lempainen J, Kinnunen T. The effect of early life cytomegalovirus infection on the immune profile of children. Clin Immunol 2024; 266:110330. [PMID: 39067678 DOI: 10.1016/j.clim.2024.110330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/05/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
Cytomegalovirus (CMV) infection has a life-long impact on the immune system, particularly on memory T cells. However, the effect of early life CMV infection on the phenotype and functionality of T cells in infants and especially longitudinal changes occurring during childhood have not been explored in detail. The phenotype and functionality of peripheral blood CD8+ and CD4+ T cells from children infected with CMV in early life (< 6 months of age) was analyzed using high-dimensional flow cytometry. Samples from CMV IgG-seropositive (CMV+) children were collected at 6 months and 6 years of age and compared to samples from CMV-seronegative (CMV-) children. Early life CMV infection caused multiple alterations within T cells. These include downregulation of CD28 expression and upregulation of CD57 expression within both CD27+ early and CD27- late effector memory CD8+ and CD4+ T-cells at 6 months of age. Of these changes, only alterations within the highly differentiated late effector memory compartment persisted at the age of 6 years. Early life CMV-infection has a distinct impact on developing CD8+ and CD4+ memory T cell compartments. It appears to induce both temporary as well as longer-lasting alterations, which may affect the functionality of the immune system throughout life.
Collapse
Affiliation(s)
- Ilse Ekman
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Anna-Mari Schroderus
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Tytti Vuorinen
- Institute of Biomedicine, University of Turku, Turku, Finland; Department of Clinical Microbiology, Turku University Hospital, Turku, Finland
| | - Mikael Knip
- Tampere Center for Child Health Research, Tampere University Hospital, Tampere, Finland; Research Unit of Clinical Medicine, Department of Pediatrics, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Riitta Veijola
- PEDEGO Research Unit, Department of Pediatrics, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Jorma Toppari
- Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Research Centre for Integrative Physiology and Pharmacology, and Centre for Population Health Research, InFLAMES Research Flagship, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Johanna Lempainen
- Department of Clinical Microbiology, Turku University Hospital, Turku, Finland; Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Tuure Kinnunen
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland; ISLAB Laboratory Centre, Kuopio, Finland.
| |
Collapse
|
25
|
Ionescu LI, Blydt-Hansen T, Foster BJ, Allen U, Birk PE, Hamiwka L, Phan V, Min S, Ivison S, Levings M, West LJ, Mital S, Urschel S. Immune phenotyping in a pediatric multicenter transplant study: Suitability of a preformulated dry-antibody panel system. Hum Immunol 2024; 85:110837. [PMID: 39013208 DOI: 10.1016/j.humimm.2024.110837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/18/2024]
Abstract
Flow-cytometric immune phenotyping is influenced by cryopreservation and inter-laboratory variability limiting comparability in multicenter studies. We assessed a system of optimized, pre-mixed dry-antibody panel tubes requiring small amounts of whole blood for validity, reliability and challenges in a Canadian multicenter study (POSITIVE) with long-distance sample shipping, using standardized protocols. Thirty-seven children awaiting solid-organ transplant were enrolled for parallel immune-phenotyping with both validated, optimized in-house panels and the dry-antibody system. Samples were collected before, 3 and 12 months post-transplant. Quality-assurance measures and congruence of phenotypes were compared using Bland-Altman comparisons, linear regression and group comparisons. Samples showed excellent lymphocyte viability (mean 94.8 %) and recovery when processed within 30 h. Comparing staining methods, significant correlations (Spearman correlation coefficient >0.6, p < 0.05), mean difference <5 % and variation 2SD <25 % were found for natural-killer, T and B cells, including many immunologically important cell subsets (CD8+, naïve, memory CD4+ T; switched-memory, transitional B). Some subgroups (plasmablasts, CD1d+CD5hi B cells) showed weak correlations, limiting interpretation reliability. The dry-antibody system provides a reliable method for standardized analysis of many immune phenotypes after long-distance shipping when processed within 30 h, rendering the system attractive for pediatric studies due to small blood amounts required and highly standardized processing and analysis.
Collapse
Affiliation(s)
- Lavinia I Ionescu
- Division of Pediatric Cardiology, University of Alberta, Edmonton, Alberta, Canada; Canadian Donation and Transplant Research Program, Edmonton, Alberta, Canada
| | - Tom Blydt-Hansen
- Canadian Donation and Transplant Research Program, Edmonton, Alberta, Canada; Division of Pediatric Nephrology, University of British Columbia, Vancouver, Canada
| | - Bethany J Foster
- Canadian Donation and Transplant Research Program, Edmonton, Alberta, Canada; Division of Nephrology, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada
| | - Upton Allen
- Canadian Donation and Transplant Research Program, Edmonton, Alberta, Canada; Division of Infectious Diseases, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Patricia E Birk
- Canadian Donation and Transplant Research Program, Edmonton, Alberta, Canada; Department of Pediatrics and Child Health, Health Sciences Centre Winnipeg, Winnipeg, Manitoba, Canada
| | - Lorraine Hamiwka
- Canadian Donation and Transplant Research Program, Edmonton, Alberta, Canada; Division of Nephrology, Alberta Children's Hospital, University of Calgary, Calgary, Canada
| | - Veronique Phan
- Division of Nephrology, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Montreal, Quebec, Canada
| | - Sandar Min
- Canadian Donation and Transplant Research Program, Edmonton, Alberta, Canada; Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | | - Lori J West
- Division of Pediatric Cardiology, University of Alberta, Edmonton, Alberta, Canada; Canadian Donation and Transplant Research Program, Edmonton, Alberta, Canada
| | - Seema Mital
- Canadian Donation and Transplant Research Program, Edmonton, Alberta, Canada; Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada; Division of Cardiology, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Simon Urschel
- Division of Pediatric Cardiology, University of Alberta, Edmonton, Alberta, Canada; Canadian Donation and Transplant Research Program, Edmonton, Alberta, Canada.
| |
Collapse
|
26
|
Fiz-López A, De Prado Á, Arribas-Rodríguez E, García-Alonso FJ, Izquierdo S, Martín-Muñoz Á, Garrote JA, Arranz E, Barrio J, Fernández-Salazar L, Bernardo D. Biological variability of human intraepithelial lymphocytes throughout the human gastrointestinal tract in health and coeliac disease. Eur J Clin Invest 2024:e14304. [PMID: 39210517 DOI: 10.1111/eci.14304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Intraepithelial lymphocytes are the first line of defence of the human intestinal immune system. Besides, their composition is altered on patients with coeliac disease (CD), so they are considered as biomarkers with utility on their diagnose and/or monitoring. Our aim is to address their variability through the human gastrointestinal tract in health and characterized them in further depth in the coeliac duodenum. METHODS Intraepithelial lymphocytes were isolated from human gastric, duodenal, ileal and colonic biopsies, then stained with specific antibodies and acquired by flow cytometry. RESULTS Our results confirmed that the profile of Intraepithelial lymphocytes change through the length of the human gastrointestinal tract. Besides and given the central role that Interleukin-15 (IL-15) elicits on CD pathogenesis; we also assessed the expression of its receptor revealing that there was virtually no functional IL-15 receptor on duodenal Intraepithelial lymphocytes. Nevertheless and contrary to our expectations, the active IL-15 receptor was not increased either on Intraepithelial lymphocytes from CD patients. CONCLUSIONS IL-15 might require additional stimulus to activate intraepithelial lymphocytes. These findings may provide novel tools to aid on a CD diagnosis and/or monitoring, at the time that provide the bases to perform functional studies in order of getting a deeper insight in the specific function that Intraepithelial lymphocytes elicit on CD pathogenesis.
Collapse
Affiliation(s)
- Aida Fiz-López
- Mucosal Immunology Lab, Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
| | - Ángel De Prado
- Mucosal Immunology Lab, Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
- Servicio de Gastroenterología, Hospital Universitario Río Hortega, Valladolid, Spain
| | - Elisa Arribas-Rodríguez
- Mucosal Immunology Lab, Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
| | | | - Sandra Izquierdo
- Servicio de Gastroenterología, Hospital Clínico Universitario, Valladolid, Spain
| | - Álvaro Martín-Muñoz
- Cytometry Facility, Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM, Universidad de Valladolid-CSIC), Valladolid, Spain
| | - José A Garrote
- Mucosal Immunology Lab, Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
| | - Eduardo Arranz
- Mucosal Immunology Lab, Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
| | - Jesús Barrio
- Servicio de Gastroenterología, Hospital Universitario Río Hortega, Valladolid, Spain
| | - Luis Fernández-Salazar
- Servicio de Gastroenterología, Hospital Clínico Universitario, Valladolid, Spain
- Departamento de Medicina, Dermatología y Toxicología, Universidad de Valladolid, Valladolid, Spain
| | - David Bernardo
- Mucosal Immunology Lab, Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| |
Collapse
|
27
|
Jin K, Yao Q, Sun B. The phenotypic characteristics of polymorphonuclear neutrophils and their correlation with B cell and CD4+T cell subsets in thyroid-associated ophthalmopathy. Front Immunol 2024; 15:1413849. [PMID: 39234250 PMCID: PMC11371595 DOI: 10.3389/fimmu.2024.1413849] [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: 04/08/2024] [Accepted: 07/19/2024] [Indexed: 09/06/2024] Open
Abstract
Introduction Thyroid-associated ophthalmopathy (TAO) is considered to be an organ-specific autoimmune disease. Polymorphonuclear neutrophils (PMN) have been implicated in the pathogenesis of TAO. However, little is known about the role of PMN in the development of TAO, much less the relationship between PMN with B cells and CD4+T cells in TAO. Objective This study aims to investigate the phenotypic characteristics of PMN and the relationship between PMN with CD4+T cell and B cell subsets in the pathogenesis of TAO. Methods Blood routine information was collected from 135 TAO patients, 95 Grave's disease without TAO (GD) patients, and 116 normal controls (NC), while surface marker expression of PMN and the level of CD4+T cell and B cell subsets in peripheral blood from 40 TAO patients, 17 GD patients, and 45 NC was assessed by flow cytometry. Result The level of PMN, CD62L+PMN, CD54+PMN, CD4+T cells, and Th17 cells displayed an increase in TAO patients than NC, while Treg cells were lower in the TAO group compared to NC. There was no statistical difference in Th1 and plasma cells among the groups. PMN were positively correlated with Th17 cells, but not the Th1, Treg, and plasma cells. Conclusion In the present study, we found that the percentage of PMN and PMN subset cells was significantly higher in TAO than in NC, and PMN were positively correlated with Th17 cells. It suggests that PMN may be involved in the immunopathogenesis of TAO and modulate the Th17 cell response during this process.
Collapse
Affiliation(s)
- Ke Jin
- Department of Ophthalmology, Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Qian Yao
- Department of Ophthalmology, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Bin Sun
- Department of Ophthalmology, Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| |
Collapse
|
28
|
De Biasi S, Ciobanu AL, Santacroce E, Lo Tartaro D, Degliesposti G, D’Angerio M, Leccese M, Cardi M, Trenti T, Cuccorese M, Gibellini L, Ferraro D, Cossarizza A. SARS-CoV-2 Vaccination Responses in Anti-CD20-Treated Progressive Multiple Sclerosis Patients Show Immunosenescence in Antigen-Specific B and T Cells. Vaccines (Basel) 2024; 12:924. [PMID: 39204047 PMCID: PMC11360119 DOI: 10.3390/vaccines12080924] [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: 07/01/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 09/03/2024] Open
Abstract
Clinical, pathological, and imaging evidence in multiple sclerosis (MS) shows that inflammation starts early and progresses with age. B cells play a central role in this process, contributing to cytokine production, defective regulatory functions, and abnormal immunoglobulin production, even in the central nervous system. Anti-CD20 (aCD20) therapies, which deplete CD20+ B cells, are largely used in the treatment of both relapsing remitting (RR) and progressive (PR) forms of MS. Although effective against MS symptoms and lesions detectable by magnetic resonance imaging, aCD20 therapies can reduce the immune response to COVID-19 vaccination. By using high-parameter flow cytometry, we examined the antigen-specific (Ag+) immune response six months post-third COVID-19 mRNA vaccination in MS patients with RR and PR forms on aCD20 therapy. Despite lower Ag+ B cell responses and lower levels of anti-SARS-CoV2, both total and neutralizing antibodies, RR and PR patients developed strong Ag+ T cell responses. We observed similar percentages and numbers of Ag+ CD4+ T cells and a high proportion of Ag+ CD8+ T cells, with slight differences in T cell phenotype and functionality; this, however, suggested the presence of differences in immune responses driven by age and disease severity.
Collapse
Affiliation(s)
- Sara De Biasi
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy (A.C.)
| | - Alin Liviu Ciobanu
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy (A.C.)
| | - Elena Santacroce
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy (A.C.)
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy (A.C.)
| | - Gianluca Degliesposti
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy (A.C.)
| | - Miriam D’Angerio
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy (A.C.)
| | - Maristella Leccese
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy (A.C.)
| | - Martina Cardi
- AOU Policlinico di Modena, Neurology Unit, Department of Biomedical, Metabolic and Neuroscience, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Tommaso Trenti
- AOU Policlinico di Modena, Diagnostic Hematology and Clinical Genomics, Department of Laboratory Medicine and Pathology, 41124 Modena, Italy
| | - Michela Cuccorese
- AOU Policlinico di Modena, Diagnostic Hematology and Clinical Genomics, Department of Laboratory Medicine and Pathology, 41124 Modena, Italy
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy (A.C.)
| | - Diana Ferraro
- AOU Policlinico di Modena, Neurology Unit, Department of Biomedical, Metabolic and Neuroscience, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy (A.C.)
- National Institute for Cardiovascular Research, 40126 Bologna, Italy
| |
Collapse
|
29
|
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
|
30
|
Christensen LM, Akimova T, Wang L, Han R, Samanta A, Di Giorgio E, Hancock WW. T-regulatory cells require Sin3a for stable expression of Foxp3. Front Immunol 2024; 15:1444937. [PMID: 39156895 PMCID: PMC11327135 DOI: 10.3389/fimmu.2024.1444937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 07/08/2024] [Indexed: 08/20/2024] Open
Abstract
Histone deacetylases 1 and 2 play a major role in the transcriptional regulation of T-regulatory (Treg) cells via interactions with a myriad of coregulatory factors. Sin3a has been well established as a Hdac1/2 cofactor, while its role within Tregs has not been established. In this study, the effects of conditional deletion of Sin3a within Foxp3+ Tregs were evaluated. Developmental deletion of Sin3a from Foxp3+ Tregs resulted in the rapid onset of fatal autoimmunity. Treg numbers were greatly reduced, while residual Tregs had impaired suppressive function. Mice also showed effector T-cell activation, autoantibody production, and widespread tissue injury. Mechanistically, Sin3a deletion resulted in decreased transcription of Foxp3 with a complete lack of CNS2 CpG demethylation. In addition, Foxp3 protein stability was impaired with an increased ex-Treg population. Thus, Sin3a plays a critical role in the maintenance of Treg identity and function and is essential for the expression and stability of Foxp3.
Collapse
Affiliation(s)
- Lanette M. Christensen
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Tatiana Akimova
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Liqing Wang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Rongxiang Han
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Arabinda Samanta
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | | | - Wayne W. Hancock
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
31
|
Kuhn TM, Paulsen M, Cuylen-Haering S. Accessible high-speed image-activated cell sorting. Trends Cell Biol 2024; 34:657-670. [PMID: 38789300 DOI: 10.1016/j.tcb.2024.04.007] [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: 09/06/2023] [Revised: 04/15/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024]
Abstract
Over the past six decades, fluorescence-activated cell sorting (FACS) has become an essential technology for basic and clinical research by enabling the isolation of cells of interest in high throughput. Recent technological advancements have started a new era of flow cytometry. By combining the spatial resolution of microscopy with high-speed cell sorting, new instruments allow cell sorting based on simple image-derived parameters or sophisticated image analysis algorithms, thereby greatly expanding the scope of applications. In this review, we discuss the systems that are commercially available or have been described in enough methodological and engineering detail to allow their replication. We summarize their strengths and limitations and highlight applications that have the potential to transform various fields in basic life science research and clinical settings.
Collapse
Affiliation(s)
- Terra M Kuhn
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Malte Paulsen
- Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Sara Cuylen-Haering
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
| |
Collapse
|
32
|
Ya X, Liu C, Ma L, Ge P, Xu X, Zheng Z, Mou S, Wang R, Zhang Q, Ye X, Zhang D, Zhang Y, Wang W, Li H, Zhao J. Single-cell atlas of peripheral blood by CyTOF revealed peripheral blood immune cells metabolic alterations and neutrophil changes in intracranial aneurysm rupture. MedComm (Beijing) 2024; 5:e637. [PMID: 39015556 PMCID: PMC11247334 DOI: 10.1002/mco2.637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 05/12/2024] [Accepted: 05/30/2024] [Indexed: 07/18/2024] Open
Abstract
Previous studies have found that the peripheral immune environment is closely related to the occurrence and development of intracranial aneurysms. However, it remains unclear how the metabolism of peripheral blood mononuclear cells (PBMCs) and the composition of polymorphonuclear leukocytes (PMNs) changes in the process of intracranial aneurysm rupture. This study utilized cytometry by time of flight technology to conduct single-cell profiling analysis of PBMCs and PMNs from 72 patients with IAs. By comparing the expression differences of key metabolic enzymes in PBMCs between patients with ruptured intracranial aneurysms (RIAs) and unruptured intracranial aneurysms, we found that most PBMCs subsets from RIA group showed upregulation of rate-limiting enzymes related to the glycolytic pathway. By comparing the composition of PMNs, it was found that the proinflammatory CD101+HLA DR+ subsets were increased in the RIA group, accompanied by a decrease in the anti-inflammatory polymorphonuclear myeloid-derived suppressor cells. In conclusion, this study showed the changes in the peripheral immune profile of RIAs, which is helpful for our understanding of the mechanisms underlying peripheral changes and provides a direction for future related research.
Collapse
Affiliation(s)
- Xiaolong Ya
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Chenglong Liu
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Long Ma
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Peicong Ge
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Xiaoxue Xu
- Department of Core Facility CenterCapital Medical UniversityBeijingChina
| | - Zhiyao Zheng
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Department of Neurosurgery, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Siqi Mou
- Medical SchoolUniversity of Chinese Academy of SciencesBeijingChina
| | - Rong Wang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Qian Zhang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Xun Ye
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Dong Zhang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Department of NeurosurgeryBeijing HospitalBeijingChina
| | - Yan Zhang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Wenjing Wang
- Beijing Institute of Hepatology, Beijing YouAn HospitalCapital Medical UniversityBeijingChina
| | - Hao Li
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Jizong Zhao
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| |
Collapse
|
33
|
Dolfi B, Gallerand A, Caillot Z, Castiglione A, Zair FN, Leporati L, Giacchero M, Goës E, Strazzulla A, Dombrowicz D, Guinamard RR, Bertola A, Ivanov S. Sex-specific impact of psychosocial stress on hematopoiesis and blood leukocytes. Eur J Immunol 2024; 54:e2350851. [PMID: 38803021 DOI: 10.1002/eji.202350851] [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: 10/20/2023] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024]
Abstract
Stress exposure has been shown to modulate innate and adaptive immune responses. Indeed, stress favors myelopoiesis and monocyte generation and contributes to cardiovascular disease development. As sex hormones regulate innate and adaptive immune responses, we decided to investigate whether stress exposure leads to a different immune response in female and male mice. Our data demonstrated that psychosocial stressinduced neutrophilia in male, but not female mice. Importantly, we identified that B-cell numbers were reduced in female, but not male mice upon exposure to stress. Thus, our study revealed that the stress-induced immune alterations are sex-dependent, and this is an important feature to consider for future investigations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Eloïse Goës
- Université Côte d'Azur, CNRS, LP2M, Nice, France
| | | | - David Dombrowicz
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | | | | | | |
Collapse
|
34
|
Macchia I, Iacobone F, Urbani F. Optimization and intra-assay validation of a multiparametric flow cytometric test for monitoring circulating TREGs. Methods Cell Biol 2024; 189:169-188. [PMID: 39393882 DOI: 10.1016/bs.mcb.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
Abstract
Multiparametric flow cytometry (MFC) represents an essential tool for immune monitoring, and validation of MFC panels is a fundamental prerequisite in routine laboratory settings as well as for translational and clinical research purposes. Regulatory T cells (TREGs) constitute a subset of CD4+ effector T cells that modulate the immune response in numerous settings, including autoimmune disease, allergy, microbial infection, tumor immunity, transplantation, and more. These cells comprise a small fraction of total CD4+ T cells in human peripheral blood and mouse spleen. In oncology, TREG cells are highly relevant, as they are involved in the suppression of the anti-tumor response in many types of cancer, to the extent that the first immune checkpoint inhibitor approved for clinical use in humans was a monoclonal antibody directed against CTLA-4, a molecule functionally associated with TREGs. Due to all these factors, robust assays are mandatory to accurately determine TREG cell frequency and function. Here, we describe the validation of an 8-color flow-cytometry protocol for TREG detection and analysis in a real-world laboratory scenario. The entire process includes the workflow plan and the standard operating procedure resembling each phase, from the panel design to the staining, acquisition, and analysis steps. Validation is planned to be performed in replicates on fresh whole blood samples derived from multiple healthy subjects. The analytical validity of the TREG cell assay is ensured by testing the intra-assay accuracy. The detailed procedure for the entire process is accompanied by important troubleshooting suggestions and other useful tips.
Collapse
Affiliation(s)
- Iole Macchia
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità (ISS), Rome, Italy.
| | - Floriana Iacobone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità (ISS), Rome, Italy
| | - Francesca Urbani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità (ISS), Rome, Italy
| |
Collapse
|
35
|
Chiu H, Weinstein KN, Spath S, Hu A, Varela S, Obata-Ninomiya K, Ziegler SF. SKI Regulates Medullary Thymic Epithelial Cell Differentiation to Control Peripheral T Cell Responses in Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:52-62. [PMID: 38767415 PMCID: PMC11182718 DOI: 10.4049/jimmunol.2300262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 04/15/2024] [Indexed: 05/22/2024]
Abstract
The thymus is an important site for the establishment of an appropriate immune response through positive and negative selection of developing T cells. During selection, developing T cells interact with cortical and medullary thymic epithelial cells (TECs), termed cTECs and mTECs, respectively. Using a Foxn1Cre+/-SKIfl/fl mouse model, we found that TEC-specific deletion of SKI reduced the mTEC compartment in the thymus and that tissue-restricted Ag expression in mTECs was altered. This decrease in the medullary area led to a decrease in CD4 thymocyte cellularity; however, mature CD4 cellularity in the spleen remained normal. Interestingly, naive CD4 T cells purified from SKI-deleted mice showed a defect in proliferation in vitro after global TCR stimulation, and these mice were significantly protected from developing experimental autoimmune encephalomyelitis compared with the control mice. Overall, our findings suggest that SKI signaling in the thymus regulates mTEC differentiation and function as well as downstream peripheral T cell responses and provide evidence for targeting SKI in T cell-driven autoimmune diseases such as multiple sclerosis.
Collapse
|
36
|
Vetter C, Schieb J, Vedder N, Lange T, Brunn T, van Geffen C, Gercke P, Kolahian S. The impact of IL-10 and IL-17 on myeloid-derived suppressor cells in vitro and in vivo in a murine model of asthma. Eur J Immunol 2024; 54:e2350785. [PMID: 38654479 DOI: 10.1002/eji.202350785] [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: 09/23/2023] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/26/2024]
Abstract
Myeloid-derived suppressor cells (MDSCs) hold promise for clinical applications due to their immunosuppressive properties, particularly in the context of inflammation. In the present study, the number and immunosuppressive activity of MDSCs isolated from naïve Il10-/-, Il17-/-, and WT mice as control, as well as from house dust mite extract (HDM)-induced asthmatic Il10-/- and Il17-/- mice, were investigated. IL-10 deficiency increased the number of polymorphonuclear (PMN)-MDSCs in the lung, spleen, and bone marrow, without concurrent impairment of their suppressive activity in vitro. In the asthma model, the IL-17 knockout was concomitant with a lower number and activity of monocytic (M)-MDSCs and an altered inflammatory reaction with impaired lung function. Additionally, we found a higher baseline inflammation of the Il17-/- mice in the lung, manifested in increased airway resistance. We conclude that the impact of IL-10 and IL-17 deficiency on MDSCs differs in the context of inflammation. Accordingly, the in vitro experiments demonstrated an increased number of PMN-MDSCs across tissues in Il10-/- mice, which indicates that IL-10 might serve a pivotal role in preserving immune homeostasis under physiological circumstances. In the context of HDM-induced airway inflammation, IL-17 was found to be an important player in the suppression of pulmonary inflammation and regulation of M-MDSCs.
Collapse
Affiliation(s)
- Charlotte Vetter
- Institute of Laboratory Medicine, member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), Philipps University Marburg, Marburg, Germany
| | - Jakob Schieb
- Institute of Laboratory Medicine, member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), Philipps University Marburg, Marburg, Germany
| | - Nora Vedder
- Institute of Laboratory Medicine, member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), Philipps University Marburg, Marburg, Germany
| | - Tim Lange
- Institute of Laboratory Medicine, member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), Philipps University Marburg, Marburg, Germany
| | - Tobias Brunn
- Institute of Laboratory Medicine, member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), Philipps University Marburg, Marburg, Germany
| | - Chiel van Geffen
- Institute of Laboratory Medicine, member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), Philipps University Marburg, Marburg, Germany
| | - Philipp Gercke
- Institute of Laboratory Medicine, member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), Philipps University Marburg, Marburg, Germany
| | - Saeed Kolahian
- Institute of Laboratory Medicine, member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), Philipps University Marburg, Marburg, Germany
- Small Animal Imaging Core Facility, Center for Tumor Biology and Immunology (ZTI), Philipps University Marburg, Marburg, Germany
| |
Collapse
|
37
|
Madan U, Verma B, Awasthi A. Cenicriviroc, a CCR2/CCR5 antagonist, promotes the generation of type 1 regulatory T cells. Eur J Immunol 2024; 54:e2350847. [PMID: 38643381 DOI: 10.1002/eji.202350847] [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: 10/20/2023] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 04/22/2024]
Abstract
Cenicriviroc, a dual CCR2/CCR5 antagonist, initially developed as an anti-HIV drug, has shown promising results in nonalcoholic steatohepatitis phase 2 clinical trials. It inhibits the infiltration and activation of CCR2+/CCR5+ monocytes and macrophages to the site of liver injury, preventing liver fibrosis. However, the role of Cenicriviroc in the modulation of helper T cell differentiation and functions remains to be explored. In inflamed colons of Crohn's disease patients, CCR2+ and CCR5+ CD4+ T cells are enriched. Considering the role of CCR2+ and CCR5+ T cells in IBD pathogenesis, we investigated the potential role of Cenicriviroc in colitis. Our in vitro studies revealed that Cenicriviroc inhibits Th1-, Th2-, and Th17-cell differentiation while promoting the generation of type 1 regulatory T cells (Tr1), known for preventing inflammation through induction of IL-10. This study is the first to report that Cenicriviroc promotes Tr1 cell generation by up-regulating the signature of Tr1 cell transcription factors such as c-Maf, Prdm1, Irf-1, Batf, and EGR-2. Cenicriviroc displayed a protective effect in experimental colitis models by preventing body weight loss and intestinal inflammation and preserving epithelial barrier integrity. We show that Cenicriviroc induced IL-10 and inhibited the generation of pro-inflammatory cytokines IFN-γ, IL-17, IL-6, and IL-1β during colitis. Based on our data, we propose Cenicriviroc as a potential therapeutic in controlling tissue inflammation by inhibiting the generation and functions of effector T cells and promoting the induction of anti-inflammatory Tr1 cells.
Collapse
Affiliation(s)
- Upasna Madan
- Centre for Immuno-biology and Immunotherapy, NCR-Biotech Science Cluster, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Bhawna Verma
- Centre for Immuno-biology and Immunotherapy, NCR-Biotech Science Cluster, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Amit Awasthi
- Centre for Immuno-biology and Immunotherapy, NCR-Biotech Science Cluster, Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Immunology-Core Lab, NCR Biotech Science Cluster, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| |
Collapse
|
38
|
Yang JC, Hsu TH, Chen CS, Yu JH, Lin KI, Chen YJ. Enhanced Proteomic Coverage in Tissue Microenvironment by Immune Cell Subtype Library-Assisted DIA-MS. Mol Cell Proteomics 2024; 23:100792. [PMID: 38810695 PMCID: PMC11260568 DOI: 10.1016/j.mcpro.2024.100792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/30/2024] [Accepted: 05/26/2024] [Indexed: 05/31/2024] Open
Abstract
Immune cells that infiltrate the tumor microenvironment (TME) play crucial roles in shaping cancer development and influencing clinical outcomes and therapeutic responses. However, obtaining a comprehensive proteomic snapshot of tumor-infiltrating immunity in clinical specimens is often hindered by small sample amounts and a low proportion of immune infiltrating cells in the TME. To enable in-depth and highly sensitive profiling of microscale tissues, we established an immune cell-enriched library-assisted strategy for data-independent acquisition mass spectrometry (DIA-MS). Firstly, six immune cell subtype-specific spectral libraries were established from sorted cluster of differentiation markers, CD8+, CD4+ T lymphocytes, B lymphocytes, natural killer cells, dendritic cells, and macrophages in murine mesenteric lymph nodes (MLNs), covering 7815 protein groups with surface markers and immune cell-enriched proteins. The feasibility of microscale immune proteomic profiling was demonstrated on 1 μg tissue protein from the tumor of murine colorectal cancer (CRC) models using single-shot DIA; the immune cell-enriched library increased coverage to quantify 7419 proteins compared to directDIA analysis (6978 proteins). The enhancement enabled the mapping of 841 immune function-related proteins and exclusive identification of many low-abundance immune proteins, such as CD1D1, and CD244, demonstrating high sensitivity for immune landscape profiling. This approach was used to characterize the MLNs in CRC models, aiming to elucidate the mechanism underlying their involvement in cancer development within the TME. Even with a low percentage of immune cell infiltration (0.25-3%) in the tumor, our results illuminate downregulation in the adaptive immune signaling pathways (such as C-type lectin receptor signaling, and chemokine signaling), T cell receptor signaling, and Th1/Th2/Th17 cell differentiation, suggesting an immunosuppressive status in MLNs of CRC model. The DIA approach using the immune cell-enriched libraries showcased deep coverage and high sensitivity that can facilitate illumination of the immune proteomic landscape for microscale samples.
Collapse
Affiliation(s)
- Jhih-Ci Yang
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan; Sustainable Chemical Science and Technology, Taiwan International Graduate Program, Academia Sinica and National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Applied Chemistry, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Tzi-Hui Hsu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | | | - Jou-Hui Yu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Kuo-I Lin
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.
| | - Yu-Ju Chen
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan; Sustainable Chemical Science and Technology, Taiwan International Graduate Program, Academia Sinica and National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Chemistry, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
39
|
Mei HE, Schulz AR, Hirseland H, Diekmann LM, Habermann E, Ten Hagen A, Albach F, Burmester GR, Grützkau A, Biesen R. Prophylactic antibodies inhibit spike-specific T and B cell responses after COVID-19 vaccination. J Med Virol 2024; 96:e29778. [PMID: 38965882 DOI: 10.1002/jmv.29778] [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: 12/28/2023] [Revised: 04/29/2024] [Accepted: 06/25/2024] [Indexed: 07/06/2024]
Abstract
Active and passive immunization is used in high-risk patients to prevent severe courses of COVID-19, but the impact of prophylactic neutralizing antibodies on the immune reaction to the mRNA vaccines has remained enigmatic. Here we show that CD4 T and B cell responses to Spikevax booster immunization are suppressed by the therapeutic antibodies Casirivimab and Imdevimab. B cell and T cell responses were significantly induced in controls but not in antibody-treated patients. The data indicates that humoral immunity, i. e. high levels of antibodies, negatively impacts reactive immunity, resulting in blunted cellular responses upon boosting. This argues for temporal separation of vaccination efforts; with active vaccination preferably applied before prophylactic therapeutic antibody treatment.
Collapse
MESH Headings
- Humans
- COVID-19/prevention & control
- COVID-19/immunology
- B-Lymphocytes/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- COVID-19 Vaccines/immunology
- SARS-CoV-2/immunology
- Middle Aged
- Male
- Female
- Vaccination
- Adult
- Aged
- CD4-Positive T-Lymphocytes/immunology
- T-Lymphocytes/immunology
- Immunization, Secondary
- Immunity, Humoral
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/therapeutic use
Collapse
Affiliation(s)
- Henrik E Mei
- Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Axel Ronald Schulz
- Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Heike Hirseland
- Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Lisa-Marie Diekmann
- Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Elisa Habermann
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Alexander Ten Hagen
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Fredrik Albach
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Gerd-Rüdiger Burmester
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andreas Grützkau
- Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Robert Biesen
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
40
|
Glehr G, Riquelme P, Kronenberg K, Lohmayer R, López-Madrona VJ, Kapinsky M, Schlitt HJ, Geissler EK, Spang R, Haferkamp S, Hutchinson JA. Restricting datasets to classifiable samples augments discovery of immune disease biomarkers. Nat Commun 2024; 15:5417. [PMID: 38926389 PMCID: PMC11208602 DOI: 10.1038/s41467-024-49094-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 05/14/2024] [Indexed: 06/28/2024] Open
Abstract
Immunological diseases are typically heterogeneous in clinical presentation, severity and response to therapy. Biomarkers of immune diseases often reflect this variability, especially compared to their regulated behaviour in health. This leads to a common difficulty that frustrates biomarker discovery and interpretation - namely, unequal dispersion of immune disease biomarker expression between patient classes necessarily limits a biomarker's informative range. To solve this problem, we introduce dataset restriction, a procedure that splits datasets into classifiable and unclassifiable samples. Applied to synthetic flow cytometry data, restriction identifies biomarkers that are otherwise disregarded. In advanced melanoma, restriction finds biomarkers of immune-related adverse event risk after immunotherapy and enables us to build multivariate models that accurately predict immunotherapy-related hepatitis. Hence, dataset restriction augments discovery of immune disease biomarkers, increases predictive certainty for classifiable samples and improves multivariate models incorporating biomarkers with a limited informative range. This principle can be directly extended to any classification task.
Collapse
Affiliation(s)
- Gunther Glehr
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Paloma Riquelme
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | | | - Robert Lohmayer
- Algorithmic Bioinformatics Research Group, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | | | | | - Hans J Schlitt
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Edward K Geissler
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Rainer Spang
- Department of Statistical Bioinformatics, University of Regensburg, Regensburg, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - James A Hutchinson
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany.
| |
Collapse
|
41
|
Swatler J, Targońska A, Turos-Korgul L, Mosieniak G, Piwocka K. Protocol for isolation of tumor-derived extracellular vesicles and functional studies on human T cell subsets. STAR Protoc 2024; 5:103011. [PMID: 38613780 PMCID: PMC11026832 DOI: 10.1016/j.xpro.2024.103011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/06/2024] [Accepted: 03/26/2024] [Indexed: 04/15/2024] Open
Abstract
Extracellular vesicles (EVs) enable communication between cells and tissues and are implicated in modulation of tumor immunosuppression. Here, we present a protocol for isolating tumor-derived EVs and assessing their functional influence in cultures with different subsets of human T cells. We describe steps for differential ultracentrifugation, size exclusion chromatography, EVs quantification, and fluorescence-activated cell sorting of human T cells. We then detail procedures for culturing T cells with EVs and using high-resolution spectral flow cytometry phenotyping for the analysis thereof. For complete details on the use and execution of this protocol, please refer to Swatler et al.1 and Swatler et al.2.
Collapse
Affiliation(s)
- Julian Swatler
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, 02093 Warsaw, Poland; Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy.
| | - Alicja Targońska
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, 02093 Warsaw, Poland
| | - Laura Turos-Korgul
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, 02093 Warsaw, Poland
| | - Grażyna Mosieniak
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, 02093 Warsaw, Poland
| | - Katarzyna Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, 02093 Warsaw, Poland.
| |
Collapse
|
42
|
Rivero-Pino F, Casquete M, Castro MJ, Redondo del Rio P, Gutierrez E, Mayo-Iscar A, Nocito M, Corell A. Prospective, Randomized, Double-Blind Parallel Group Nutritional Study to Evaluate the Effects of Routine Intake of Fresh vs. Pasteurized Yogurt on the Immune System in Healthy Adults. Nutrients 2024; 16:1969. [PMID: 38931322 PMCID: PMC11206341 DOI: 10.3390/nu16121969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/28/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
The immune system is affected by the dietary products humans intake. Immune system regulation by nutrition has uses in the clinical context, but it can also benefit healthy populations by delaying or preventing the emergence of immune-mediated chronic illnesses. In this study, the purpose was to describe and compare the modulator effects on the immune system of the routine ingestion of fresh vs. pasteurized yogurt. A unicentral, prospective, randomized, double-blind, parallel group 8-week nutritional study was carried out comparing the ingestion of 125 g of the products in healthy adults three times a day. A complete battery of in vitro tests on the activity of the immune system, processes and phenomena was performed. Exclusive immune-modulatory effects of fresh yogurt with respect to base line were found in terms of increased systemic IgM (primary immune responses), increased synthesis of IFN-gamma upon stimulation (Th1) and increased peripheral T cells (mainly "naive" CD4s). In the three interventions, we observed an increased phagocytic activity and burst test in granulocytes, together with increased secretion of IL-6, IL-1 β and IL-8 (pro-inflammatory) and increased CD16 expression (FcR favoring phagocytosis) in granulocytes. Overall, it is concluded that regardless of bacteria being alive or thermally inactivated, yogurt has common effects on the innate system, but the presence of live bacteria is necessary to achieve a potentiating effect on the specific immune response.
Collapse
Affiliation(s)
- Fernando Rivero-Pino
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocío, CSIC, University of Seville, 41013 Seville, Spain
| | - Mar Casquete
- Departamento de Pediatría, Inmunología, Obstetricia-Ginecología, Nutrición-Bromatología, Universidad de Valladolid, 47005 Valladolid, Spain
| | - Maria José Castro
- Departamento de Enfermería, Universidad de Valladolid, 47003 Valladolid, Spain
| | - Paz Redondo del Rio
- Departamento de Pediatría, Inmunología, Obstetricia-Ginecología, Nutrición-Bromatología, Universidad de Valladolid, 47005 Valladolid, Spain
| | - Eloina Gutierrez
- Departamento de Pediatría, Inmunología, Obstetricia-Ginecología, Nutrición-Bromatología, Universidad de Valladolid, 47005 Valladolid, Spain
| | - Agustín Mayo-Iscar
- Departamento de Estadística e Investigación Operativa & IMUVA, Universidad de Valladolid, 47011 Valladolid, Spain
| | - Mercedes Nocito
- Inmunología, Hospital Clínico de Zaragoza, 50009 Zaragoza, Spain
| | - Alfredo Corell
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocío, CSIC, University of Seville, 41013 Seville, Spain
- Departamento de Pediatría, Inmunología, Obstetricia-Ginecología, Nutrición-Bromatología, Universidad de Valladolid, 47005 Valladolid, Spain
| |
Collapse
|
43
|
Baude J, Ladjohounlou R, Limagne E, Froidurot L, Morgand V, Mirjolet C. Characterization of Natural Killer cells infiltrating irradiated murine tumors through flow cytometry. Methods Cell Biol 2024; 189:117-133. [PMID: 39393879 DOI: 10.1016/bs.mcb.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
Abstract
The radiation-induced immune response is increasingly well documented. However, some aspects remain unclear, notably the role of Natural Killer (NK) cells, a subgroup of innate lymphoid cells involved in the antitumor response, in the response to RT. It therefore seems necessary to better characterize NK cells infiltrating irradiated tumors in order to better understand the mechanisms of action of RT, enabling its subsequent optimization and combination with other immunomodulatory treatments. A key technology for studying intratumoral immune cells is flow cytometry, which can simultaneously quantify and analyze the phenotype of numerous cells. Here, we propose a method for phenotyping intratumoral NK cells through flow cytometry in mice bearing colorectal tumors treated with radiotherapy. This procedure can also be used to study the radiation-induced NK cell response in a wide range of solid tumors.
Collapse
Affiliation(s)
- Jérémy Baude
- Department of Radiation Oncology, Centre Georges-François Leclerc, Dijon, France; Preclinical Radiation Therapy and Radiobiology Unit, Centre Georges-François Leclerc, Dijon, France
| | - Riad Ladjohounlou
- Preclinical Radiation Therapy and Radiobiology Unit, Centre Georges-François Leclerc, Dijon, France
| | - Emeric Limagne
- University of Burgundy, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France; Genetic and Immunology Medical Institute, Dijon, France; INSERM LNC-UMR1231, Dijon, France
| | - Lisa Froidurot
- Preclinical Radiation Therapy and Radiobiology Unit, Centre Georges-François Leclerc, Dijon, France
| | - Véronique Morgand
- Preclinical Radiation Therapy and Radiobiology Unit, Centre Georges-François Leclerc, Dijon, France
| | - Céline Mirjolet
- Department of Radiation Oncology, Centre Georges-François Leclerc, Dijon, France; Preclinical Radiation Therapy and Radiobiology Unit, Centre Georges-François Leclerc, Dijon, France; TIReCS Team, UMR INSERM 1231, Dijon, France.
| |
Collapse
|
44
|
Aublin-Gex A, Jacolin F, Diaz O, Jacquemin C, Marçais A, Walzer T, Lotteau V, Vidalain PO, Perrin-Cocon L. Tethering of hexokinase 2 to mitochondria promotes resistance of liver cancer cells to natural killer cell cytotoxicity. Eur J Immunol 2024:e2350954. [PMID: 38837415 DOI: 10.1002/eji.202350954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/07/2024]
Abstract
Hexokinases (HKs) control the first step of glucose catabolism. A switch of expression from liver HK (glucokinase, GCK) to the tumor isoenzyme HK2 is observed in hepatocellular carcinoma progression. Our prior work revealed that HK isoenzyme switch in hepatocytes not only regulates hepatic metabolic functions but also modulates innate immunity and sensitivity to Natural Killer (NK) cell cytotoxicity. This study investigates the impact of HK2 expression and its mitochondrial binding on the resistance of human liver cancer cells to NK-cell-induced cytolysis. We have shown that HK2 expression induces resistance to NK cell cytotoxicity in a process requiring mitochondrial binding of HK2. Neither HK2 nor GCK expression affects target cells' ability to activate NK cells. In contrast, mitochondrial binding of HK2 reduces effector caspase 3/7 activity both at baseline and upon NK-cell activation. Furthermore, HK2 tethering to mitochondria enhances their resistance to cytochrome c release triggered by tBID. These findings indicate that HK2 mitochondrial binding in liver cancer cells is an intrinsic resistance factor to cytolysis and an escape mechanism from immune surveillance.
Collapse
Affiliation(s)
- Anne Aublin-Gex
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Florentine Jacolin
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Olivier Diaz
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Clémence Jacquemin
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Antoine Marçais
- CIRI, Centre International de Recherche en Infectiologie, Team Lymphocyte activation and signaling, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Thierry Walzer
- CIRI, Centre International de Recherche en Infectiologie, Team Lymphocyte activation and signaling, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Vincent Lotteau
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Pierre-Olivier Vidalain
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Laure Perrin-Cocon
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| |
Collapse
|
45
|
Hein Y, Zipfel F, O'Connor G, Macdonald R, Hussels M. Flow cytometer for a dilution-free measurement approach with sample recollection. THE REVIEW OF SCIENTIFIC INSTRUMENTS 2024; 95:065101. [PMID: 38829217 DOI: 10.1063/5.0187052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/12/2024] [Indexed: 06/05/2024]
Abstract
Blood testing using flow cytometry is a common and rapid method for the initial screening and diagnosis of patients. Measurements are often combined with other scientific techniques, and analyzed samples are commonly diluted and discarded afterward. When the sample is recollected instead, sample dilution is a challenge when the sample is intended or needed for additional measurements. Therefore, it is advantageous to recollect the undiluted sample. In order to enable measurements of the same undiluted sample aliquot, we designed and constructed a purpose-built flow cytometer. Our instrument employs syringes, acoustic focusing, and an open fluidics system to recollect and reuse the unadulterated sample. The cytometer is compact, which reduces sample consumption. It acquires forward, sideward, and fluorescence signals, offering opportunities for diverse measurement approaches. In particular, our cytometer has been designed to be ready for additional downstream analysis of cells, e.g., applying mass spectrometry, magnetic resonance spectroscopy, or other analytical tools. This study presents results on instrument performance, a comparison with a cytometer that uses standard hydrodynamic focusing, and a proof of concept for multiple measurements.
Collapse
|
46
|
Konecny AJ, Mage P, Tyznik AJ, Prlic M, Mair F. OMIP-102: 50-color phenotyping of the human immune system with in-depth assessment of T cells and dendritic cells. Cytometry A 2024; 105:430-436. [PMID: 38634730 PMCID: PMC11178442 DOI: 10.1002/cyto.a.24841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/27/2024] [Accepted: 04/02/2024] [Indexed: 04/19/2024]
Abstract
We report the development of an optimized 50-color spectral flow cytometry panel designed for the in-depth analysis of the immune system in human blood and tissues, with the goal of maximizing the amount of information that can be collected using currently available flow cytometry platforms. We established and tested this panel using peripheral blood mononuclear cells (PBMCs), but included CD45 to enable its future use for the analysis of human tissue samples. The panel contains lineage markers for all major immune cell subsets, and an extensive set of phenotyping markers focused on the activation and differentiation status of the T cell and dendritic cell (DC) compartment. We outline the biological insight that can be gained from the simultaneous measurement of such a large number of proteins and propose that this approach provides a unique opportunity for the comprehensive exploration of the immune status in human samples with a limited number of cells. Of note, we tested the panel to be compatible with cell sorting for further downstream applications. Furthermore, to facilitate the wide-spread implementation of such a panel across different cohorts and samples, we established a trimmed-down 45-color version which can be used with different spectral cytometry platforms. Finally, to generate this panel, we utilized not only existing panel design guidelines, but also developed new metrics to systematically identify the optimal combination of 50 fluorochromes and evaluate fluorochrome-specific resolution in the context of a 50-color unmixing matrix.
Collapse
Affiliation(s)
- Andrew J. Konecny
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle WA, 98109, USA
- Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Peter Mage
- Advanced Technology Group, BD Biosciences, San Jose, CA 95131, USA
| | - Aaron J. Tyznik
- Applied Research & Technology, Medical and Scientific Affairs, BD Biosciences, San Diego, CA 92037, USA
| | - Martin Prlic
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle WA, 98109, USA
- Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Florian Mair
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle WA, 98109, USA
- Flow Cytometry Core Facility, Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
47
|
Rau CN, Severin ME, Lee PW, Deffenbaugh JL, Liu Y, Murphy SP, Petersen-Cherubini CL, Lovett-Racke AE. MicroRNAs targeting TGF-β signaling exacerbate central nervous system autoimmunity by disrupting regulatory T cell development and function. Eur J Immunol 2024; 54:e2350548. [PMID: 38634287 PMCID: PMC11156541 DOI: 10.1002/eji.202350548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 04/19/2024]
Abstract
Transforming growth factor beta (TGF-β) signaling is essential for a balanced immune response by mediating the development and function of regulatory T cells (Tregs) and suppressing autoreactive T cells. Disruption of this balance can result in autoimmune diseases, including multiple sclerosis (MS). MicroRNAs (miRNAs) targeting TGF-β signaling have been shown to be upregulated in naïve CD4 T cells in MS patients, resulting in a limited in vitro generation of human Tregs. Utilizing the murine model experimental autoimmune encephalomyelitis, we show that perinatal administration of miRNAs, which target the TGF-β signaling pathway, enhanced susceptibility to central nervous system (CNS) autoimmunity. Neonatal mice administered with these miRNAs further exhibited reduced Treg frequencies with a loss in T cell receptor repertoire diversity following the induction of experimental autoimmune encephalomyelitis in adulthood. Exacerbated CNS autoimmunity as a result of miRNA overexpression in CD4 T cells was accompanied by enhanced Th1 and Th17 cell frequencies. These findings demonstrate that increased levels of TGF-β-associated miRNAs impede the development of a diverse Treg population, leading to enhanced effector cell activity, and contributing to an increased susceptibility to CNS autoimmunity. Thus, TGF-β-targeting miRNAs could be a risk factor for MS, and recovering optimal TGF-β signaling may restore immune homeostasis in MS patients.
Collapse
Affiliation(s)
- Christina N Rau
- Department of Microbial Infection and Immunity, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
| | - Mary E Severin
- Department of Microbial Infection and Immunity, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio, USA
| | - Priscilla W Lee
- Department of Microbial Infection and Immunity, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
- Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, Ohio, USA
| | - Joshua L Deffenbaugh
- Department of Microbial Infection and Immunity, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
| | - Yue Liu
- Department of Microbial Infection and Immunity, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
| | - Shawn P Murphy
- Department of Microbial Infection and Immunity, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
| | - Cora L Petersen-Cherubini
- Department of Microbial Infection and Immunity, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
- Neuroscience Graduate Program, The Ohio State University, Columbus, Ohio, USA
| | - Amy E Lovett-Racke
- Department of Microbial Infection and Immunity, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
48
|
Núñez SY, Trotta A, Regge MV, Amarilla MS, Secchiari F, Sierra JM, Santilli MC, Gantov M, Rovegno A, Richards N, Ameri C, Ríos Pita H, Rico L, Mieggi M, Vitagliano G, Blas L, Friedrich AD, Domaica CI, Fuertes MB, Zwirner NW. Tumor-associated macrophages impair NK cell IFN-γ production and contribute to tumor progression in clear cell renal cell carcinoma. Eur J Immunol 2024; 54:e2350878. [PMID: 38581345 DOI: 10.1002/eji.202350878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 04/08/2024]
Abstract
Tumor-associated macrophages (TAM) are abundant in several tumor types and usually correlate with poor prognosis. Previously, we demonstrated that anti-inflammatory macrophages (M2) inhibit NK cell effector functions. Here, we explored the impact of TAM on NK cells in the context of clear-cell renal cell carcinoma (ccRCC). Bioinformatics analysis revealed that an exhausted NK cell signature strongly correlated with an M2 signature. Analysis of TAM from human ccRCC samples confirmed that they exhibited an M2-skewed phenotype and inhibited IFN-γ production by NK cells. Moreover, human M0 macrophages cultured with conditioned media from ccRCC cell lines generated macrophages with an M2-skewed phenotype (TAM-like), which alike TAM, displayed suppressive activity on NK cells. Moreover, TAM depletion in the mouse Renca ccRCC model resulted in delayed tumor growth and reduced volume, accompanied by an increased frequency of IFN-γ-producing tumor-infiltrating NK cells that displayed heightened expression of T-bet and NKG2D and reduced expression of the exhaustion-associated co-inhibitory molecules PD-1 and TIM-3. Therefore, in ccRCC, the tumor microenvironment polarizes TAM toward an immunosuppressive profile that promotes tumor-infiltrating NK cell dysfunction, contributing to tumor progression. In addition, immunotherapy strategies targeting TAM may result in NK cell reinvigoration, thereby counteracting tumor progression.
Collapse
Affiliation(s)
- Sol Yanel Núñez
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Fundación IBYME, Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina
| | - Aldana Trotta
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Fundación IBYME, Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina
| | - María Victoria Regge
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Fundación IBYME, Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina
| | - María Sofía Amarilla
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Fundación IBYME, Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina
| | - Florencia Secchiari
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Fundación IBYME, Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina
| | - Jessica Mariel Sierra
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Fundación IBYME, Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina
| | - María Cecilia Santilli
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Fundación IBYME, Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina
| | - Mariana Gantov
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Fundación IBYME, Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina
| | - Agustín Rovegno
- Centro de Educación Médica e Investigaciones Clínicas "Norberto Quirno" (CEMIC), Servicio de Urología, Buenos Aires, Argentina
| | - Nicolás Richards
- Centro de Educación Médica e Investigaciones Clínicas "Norberto Quirno" (CEMIC), Servicio de Urología, Buenos Aires, Argentina
| | - Carlos Ameri
- Hospital Alemán, Servicio de Urología, Buenos Aires, Argentina
| | | | - Luis Rico
- Hospital Alemán, Servicio de Urología, Buenos Aires, Argentina
| | - Mauro Mieggi
- Hospital Alemán, Servicio de Urología, Buenos Aires, Argentina
| | | | - Leandro Blas
- Hospital Alemán, Servicio de Urología, Buenos Aires, Argentina
| | - Adrián David Friedrich
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Fundación IBYME, Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina
| | - Carolina Inés Domaica
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Fundación IBYME, Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina
| | - Mercedes Beatriz Fuertes
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Fundación IBYME, Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina
| | - Norberto Walter Zwirner
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Fundación IBYME, Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
49
|
Mandracchia B, Zheng C, Rajendran S, Liu W, Forghani P, Xu C, Jia S. High-speed optical imaging with sCMOS pixel reassignment. Nat Commun 2024; 15:4598. [PMID: 38816394 PMCID: PMC11139943 DOI: 10.1038/s41467-024-48987-7] [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: 08/11/2023] [Accepted: 05/13/2024] [Indexed: 06/01/2024] Open
Abstract
Fluorescence microscopy has undergone rapid advancements, offering unprecedented visualization of biological events and shedding light on the intricate mechanisms governing living organisms. However, the exploration of rapid biological dynamics still poses a significant challenge due to the limitations of current digital camera architectures and the inherent compromise between imaging speed and other capabilities. Here, we introduce sHAPR, a high-speed acquisition technique that leverages the operating principles of sCMOS cameras to capture fast cellular and subcellular processes. sHAPR harnesses custom fiber optics to convert microscopy images into one-dimensional recordings, enabling acquisition at the maximum camera readout rate, typically between 25 and 250 kHz. We have demonstrated the utility of sHAPR with a variety of phantom and dynamic systems, including high-throughput flow cytometry, cardiomyocyte contraction, and neuronal calcium waves, using a standard epi-fluorescence microscope. sHAPR is highly adaptable and can be integrated into existing microscopy systems without requiring extensive platform modifications. This method pushes the boundaries of current fluorescence imaging capabilities, opening up new avenues for investigating high-speed biological phenomena.
Collapse
Affiliation(s)
- Biagio Mandracchia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- E.T.S.I. Telecomunicación, Universidad de Valladolid, Valladolid, Spain
| | - Corey Zheng
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Suraj Rajendran
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Wenhao Liu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Parvin Forghani
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Chunhui Xu
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Shu Jia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
50
|
Zou M, Pezoldt J, Mohr J, Philipsen L, Leufgen A, Cerovic V, Wiechers C, Pils M, Ortiz D, Hao L, Yang J, Beckstette M, Dupont A, Hornef M, Dersch P, Strowig T, Müller AJ, Raila J, Huehn J. Early-life vitamin A treatment rescues neonatal infection-induced durably impaired tolerogenic properties of celiac lymph nodes. Cell Rep 2024; 43:114153. [PMID: 38687643 DOI: 10.1016/j.celrep.2024.114153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 11/23/2023] [Accepted: 04/10/2024] [Indexed: 05/02/2024] Open
Abstract
Gut-draining mesenteric and celiac lymph nodes (mLNs and celLNs) critically contribute to peripheral tolerance toward food and microbial antigens by supporting the de novo induction of regulatory T cells (Tregs). These tolerogenic properties of mLNs and celLNs are stably imprinted within stromal cells (SCs) by microbial signals and vitamin A (VA), respectively. Here, we report that a single, transient gastrointestinal infection in the neonatal, but not adult, period durably abrogates the efficient Treg-inducing capacity of celLNs by altering the subset composition and gene expression profile of celLNSCs. These cells carry information about the early-life pathogen encounter until adulthood and durably instruct migratory dendritic cells entering the celLN with reduced tolerogenic properties. Mechanistically, transiently reduced VA levels cause long-lasting celLN functional impairment, which can be rescued by early-life treatment with VA. Together, our data highlight the therapeutic potential of VA to prevent sequelae post gastrointestinal infections in infants.
Collapse
Affiliation(s)
- Mangge Zou
- Department Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Joern Pezoldt
- Department Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; Laboratory of Systems Biology and Genetics, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Juliane Mohr
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Lars Philipsen
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; Multi-Parametric Bioimaging and Cytometry (MPBIC) Platform, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Andrea Leufgen
- Institute of Molecular Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Vuk Cerovic
- Institute of Molecular Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Carolin Wiechers
- Department Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Marina Pils
- Mouse Pathology Platform, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Diego Ortiz
- Department Microbial Immune Regulation, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Lianxu Hao
- Department Microbial Immune Regulation, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Juhao Yang
- Department Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Michael Beckstette
- Department Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Aline Dupont
- Institute of Medical Microbiology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Mathias Hornef
- Institute of Medical Microbiology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Petra Dersch
- Institute for Infectiology, University of Münster, 48149 Münster, Germany; German Center for Infection Research (DZIF), Associated Site University of Münster, 48149 Münster, Germany
| | - Till Strowig
- Department Microbial Immune Regulation, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany
| | - Andreas J Müller
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; Multi-Parametric Bioimaging and Cytometry (MPBIC) Platform, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; Intravital Microscopy in Infection and Immunity, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Jens Raila
- Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany
| | - Jochen Huehn
- Department Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany.
| |
Collapse
|