1
|
Henriques PM, Almeida GG, Rimkute I, dos Santos LI, Liechti T, Marino AP, do Vale INPC, Vasconcelos‐Santos DV, Martins‐Filho OA, Gazzinelli RT, Roederer M, Sher A, Teixeira‐Carvalho A, Jankovic D, Antonelli LRDV. Cytotoxic Signature and IFN-γ Production Dominate CD4 + T-Cell Response During Human Toxoplasmosis. Immunology 2025; 175:151-164. [PMID: 40035468 PMCID: PMC12052435 DOI: 10.1111/imm.13912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 03/05/2025] Open
Abstract
Toxoplasma gondii is a highly versatile parasite that infects most warm-blooded animals and is a major cause of retinochoroiditis and uveitis in humans. The pathophysiology of these conditions remains poorly understood. Both parasite virulence and host inflammatory response contribute to the development of ocular disease. While CD4+ T cells play a critical role in host resistance to Toxoplasma infection, their kinetics and effector functions, as well as their contribution to the clinical outcome of the infection, including ocular involvement, remain poorly understood. To address this question, we investigated the immune response during acute and convalescent toxoplasmosis and stratified patients further based on the presence or absence of ocular disease. We found that T. gondii infection leads to decreased and increased proportions of central and effector memory CD4+ T cells, respectively. Applying unsupervised analysis, distinct CD4+ T-cell subsets were determined. Among 50 clusters, 10 produced cytotoxic proteins (granzyme B and perforin) and one produced cytokines upon antigen-specific stimulation. We observed that proportions of five CD4+ T-cell clusters out of 50 were different during acute disease between T. gondii-infected patients with and without ocular lesions. Interestingly, three of the five displayed a cytotoxic signature indicating their possible involvement in ocular immunopathology. Taken together, our results reveal that during T. gondii infection, CD4+ T cells not only develop a Th1 cytokine profile, but also acquire previously unappreciated cytotoxic capacity/function. These results, while underscoring the complexity of the CD4+ T-cell response to T. gondii, suggest that specific subsets may be involved in the development of pathology and provide possible targets for therapeutic intervention.
Collapse
Affiliation(s)
- Priscilla Miranda Henriques
- Biology and Immunology of Infectious and Parasitic Diseases Group, René Rachou InstituteOswaldo Cruz Foundation‐FIOCRUZBelo HorizonteMinas GeraisBrazil
| | - Gregório Guilherme Almeida
- Biology and Immunology of Infectious and Parasitic Diseases Group, René Rachou InstituteOswaldo Cruz Foundation‐FIOCRUZBelo HorizonteMinas GeraisBrazil
| | - Inga Rimkute
- Vaccine Research Center, National Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMarylandUSA
| | - Luara Isabela dos Santos
- Biology and Immunology of Infectious and Parasitic Diseases Group, René Rachou InstituteOswaldo Cruz Foundation‐FIOCRUZBelo HorizonteMinas GeraisBrazil
- Department of Basic Science, Faculty of Medical Sciences of Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Thomas Liechti
- Vaccine Research Center, National Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMarylandUSA
| | - Ana Paula Marino
- Biology and Immunology of Infectious and Parasitic Diseases Group, René Rachou InstituteOswaldo Cruz Foundation‐FIOCRUZBelo HorizonteMinas GeraisBrazil
| | | | - Daniel Vitor Vasconcelos‐Santos
- Department of Ophthalmology and Otorhinolaryngology, Faculty of MedicineFederal University of Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Olindo Assis Martins‐Filho
- Integrated Research Group in Biomarkers, René Rachou InstituteOswaldo Cruz Foundation‐FIOCRUZBelo HorizonteMinas GeraisBrazil
| | - Ricardo Tostes Gazzinelli
- Laboratory of Immunopathology, René Rachou InstituteOswaldo Cruz Foundation‐FIOCRUZBelo HorizonteMinas GeraisBrazil
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMarylandUSA
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMarylandUSA
| | - Andréa Teixeira‐Carvalho
- Integrated Research Group in Biomarkers, René Rachou InstituteOswaldo Cruz Foundation‐FIOCRUZBelo HorizonteMinas GeraisBrazil
| | - Dragana Jankovic
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMarylandUSA
| | - Lis Ribeiro do Valle Antonelli
- Biology and Immunology of Infectious and Parasitic Diseases Group, René Rachou InstituteOswaldo Cruz Foundation‐FIOCRUZBelo HorizonteMinas GeraisBrazil
| |
Collapse
|
2
|
Nian S, Wang K, Wang J, Wang S, Li C, Li N, Chen J. Causal Associations between Immune Cell Phenotypes and Varicose Veins: A Mendelian Randomization Analysis. Ann Vasc Surg 2025; 114:126-132. [PMID: 39884498 DOI: 10.1016/j.avsg.2025.01.030] [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: 12/04/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Varicose veins (VVs) are a common chronic venous disorder with a complex pathophysiology involving immune dysregulation, inflammation, and genetic predisposition. This study aims to identify immune-related causal factors in the pathogenesis of VVs using Mendelian randomization (MR). METHODS A 2-sample MR analysis was conducted to assess the causal relationships between immune cell phenotypes and VVs. Genetic variants were used as instrumental variables, and data were derived from the Finland and genome-wide association study catalog. Inverse variance weighting (IVW) was used as the primary method, supported by sensitivity analyses such as MR-Egger, weighted median, and weighted mode. RESULTS A total of 79 immunophenotypes were identified as significantly associated with VVs, including 19 related to B-cells, 6 to conventional dendritic cells, 6 to T-cell maturation stages, 6 to monocytes, 12 to myeloid cells, 11 to T cells, B cells, and natural killer cells, and 17 to regulatory T-cells (Tregs). Of these, 8 immunophenotypes remained significant after multiple testing correction (false discovery rate <0.05). Specifically, high expression of cluster of differentiation 86 (CD86) on myeloid dendritic cells, CD33 expression on myeloid cells (e.g., basophils and human leukocyte antigen DR + subsets), increased side scatter area (SSC-A) on lymphocytes, and associations with CD39+ Tregs showed significant correlations with VVs. CONCLUSION This study highlights the involvement of immune cells in the pathogenesis of VVs. High expression of CD86 on myeloid DCs and SSC-A on lymphocytes may serve as potential markers for predicting VVs risk, while the protective role of CD39+ Tregs suggests a new direction for immune modulation therapy. Further research is needed to validate these findings across diverse populations and explore their clinical applications.
Collapse
Affiliation(s)
- Sunqi Nian
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Kui Wang
- The First Clinical College of Shandong University, Jinan, Shandong, China
| | - Jiawei Wang
- Department of Critical Care Medicine, Jieyang Third People's Hospital, Jieyang, Guangdong, China
| | - Suijian Wang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Chengjin Li
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Na Li
- Department of Anesthesiology, 920th Hospital of the Joint Logistics Support Force, Kunming, Yunnan, China.
| | - Jiayu Chen
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China.
| |
Collapse
|
3
|
Amiri SP, Nejad FS, Karamigolbaghi M, Jafari E, Robat-Jazi B, Sadeghi A, Amiri SGP, Namdari H, Saboor-Yaraghi AA. Decreased circulating CD39+ regulatory T cell frequencies following non-traumatic brain death. Transpl Immunol 2025; 90:102219. [PMID: 40118258 DOI: 10.1016/j.trim.2025.102219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/09/2025] [Accepted: 03/15/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND AND PURPOSE Regulatory T cells (Tregs) are thought to modulate immune responses during Brain death (BD), However findings on their role remain controversial. This study aimed to assess the frequency of circulating Tregs in the peripheral blood of non-traumatic BD cases, specifically focusing on CD4+CD25+CD127low/-CD39+ Tregs and the levels of inflammatory cytokine mRNA in BD individuals. METHODS The percentage of CD4+CD25+CD127low/-CD39+ Tregs was measured using flow cytometry in BD patients upon admission and in control subjects. Additionally, mRNA expression levels of interleukin (IL)-1β, IL-6, IL-8, IL-17, tumor necrosis factor (TNF)-α and Interferon (IFN)-γ were quantified in peripheral blood mononuclear cells (PBMCs) from 28 BD individuals and 28 controls using real-time polymerase chain reaction. RESULTS CD39+ Tregs were significantly reduced in non-traumatic BD cases compared with control group (P < 0.0001). Moreover, the expression levels of IL-1β, IL-6, IL-8, IL-17a, IFN-ɣ, and TNF-α were significantly elevated in non-traumatic BD cases compared to the control group (P < 0.01, P < 0.05, P < 0.01, P < 0.0001, P < 0.0001, P < 0.001 respectively). CONCLUSION This study provides novel evidence of reduced CD39+ Tregs in the peripheral blood of non-traumatic BD patients, accompanied by increased inflammatory cytokine gene expression. Further investigations are needed to explore the underlying mechanisms and potential therapeutic implications.
Collapse
Affiliation(s)
- Sedighe Poursaleh Amiri
- Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran; Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Fattah Sotoudeh Nejad
- Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Karamigolbaghi
- Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Jafari
- Department of Stem Cells Technology and Tissue Regeneration, School of Science, Tehran University of Medical Sciences, Tehran, Iran
| | - Behrouz Robat-Jazi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Iran
| | - Ahmadreza Sadeghi
- Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Ghasem Poursaleh Amiri
- Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Haideh Namdari
- Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
4
|
Chung DC, Shakfa N, Vakharia J, Warner K, Jacquelot N, Sayad A, Han S, Ghaedi M, Garcia-Batres CR, Sotty J, Azarmina A, Nowlan F, Chen EL, Zon M, Elford AR, Wang BX, Nguyen LT, Mrkonjic M, Clarke BA, Bernardini MQ, Haibe-Kains B, Ferguson SE, Crome SQ, Jackson HW, Ohashi PS. CD103+CD56+ ILCs Are Associated with an Altered CD8+ T-cell Profile within the Tumor Microenvironment. Cancer Immunol Res 2025; 13:527-546. [PMID: 40084939 PMCID: PMC11962407 DOI: 10.1158/2326-6066.cir-24-0151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 10/10/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025]
Abstract
Immunotherapies have had unprecedented success in the treatment of multiple cancer types, albeit with variable response rates. Unraveling the complex network of immune cells within the tumor microenvironment (TME) may provide additional insights to enhance antitumor immunity and improve clinical response. Many studies have shown that NK cells or innate lymphoid cells (ILC) have regulatory capacity. Here, we identified CD103 as a marker that was found on CD56+ cells that were associated with a poor proliferative capacity of tumor-infiltrating lymphocytes in culture. We further demonstrated that CD103+CD56+ ILCs isolated directly from tumors represented a distinct ILC population that expressed unique surface markers (such as CD49a and CD101), transcription factor networks, and transcriptomic profiles compared with CD103-CD56+ NK cells. Using single-cell multiomic and spatial approaches, we found that these CD103+CD56+ ILCs were associated with CD8+ T cells with reduced expression of granzyme B. Thus, this study identifies a population of CD103+CD56+ ILCs with potentially inhibitory functions that are associated with a TME that includes CD8+ T cells with poor antitumor activity. Further studies focusing on these cells may provide additional insights into the biology of an inhibitory TME.
Collapse
Affiliation(s)
- Douglas C. Chung
- Department of Immunology, University of Toronto, Toronto, Canada
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Noor Shakfa
- Systems Biology Program, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Health System, Toronto, Canada
| | - Jehan Vakharia
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Kathrin Warner
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Nicolas Jacquelot
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Azin Sayad
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - SeongJun Han
- Department of Immunology, University of Toronto, Toronto, Canada
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Maryam Ghaedi
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Carlos R. Garcia-Batres
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Jules Sotty
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Arvin Azarmina
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Ferris Nowlan
- Systems Biology Program, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Health System, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Edward L.Y. Chen
- Systems Biology Program, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Health System, Toronto, Canada
| | - Michael Zon
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Medical Biophysics, University of Toronto, Toronto, Canada
- Structural Genomics Consortium, Toronto, Canada
- Vector Institute for Artificial Intelligence, Toronto, Canada
| | - Alisha R. Elford
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Ben X. Wang
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Linh T. Nguyen
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Miralem Mrkonjic
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada
| | - Blaise A. Clarke
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Division of Gynecologic Oncology, University Health Network, Toronto, Canada
| | - Marcus Q. Bernardini
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Division of Gynecologic Oncology, University Health Network, Toronto, Canada
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Medical Biophysics, University of Toronto, Toronto, Canada
- Structural Genomics Consortium, Toronto, Canada
- Vector Institute for Artificial Intelligence, Toronto, Canada
| | - Sarah E. Ferguson
- Division of Gynecologic Oncology, University Health Network, Toronto, Canada
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, Canada
| | - Sarah Q. Crome
- Department of Immunology, University of Toronto, Toronto, Canada
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Hartland W. Jackson
- Systems Biology Program, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Health System, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- Ontario Institute of Cancer Research, Toronto, Canada
| | - Pamela S. Ohashi
- Department of Immunology, University of Toronto, Toronto, Canada
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| |
Collapse
|
5
|
Villalba A, Nuño L, Benito-Miguel M, Nieto-Carvalhal B, Monjo I, Novella-Navarro M, Peiteado D, García-Carazo S, Balsa A, Miranda-Carús ME. Transiently increased circulating CD39+FoxP3+ Treg cells predicts the clinical response to methotrexate in early rheumatoid arthritis. Rheumatology (Oxford) 2025; 64:2282-2289. [PMID: 39141491 DOI: 10.1093/rheumatology/keae446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 07/18/2024] [Accepted: 08/03/2024] [Indexed: 08/16/2024] Open
Abstract
OBJECTIVES A subset of human circulating FoxP3+ regulatory T cells expresses CD39 (cTreg39+) and hydrolyses pro-inflammatory adenine nucleotides released at inflammatory foci, releasing the anti-inflammatory agent adenosine. Methotrexate (MTX), inhibiting 5-aminoimidazole-4-carboxamide ribonucleotide transformylase, enhances the extrusion of adenine nucleotides and may help Treg39+ cells control inflammation. Therefore, we examined the relation of cTreg39+ cells with the effect of MTX in early rheumatoid arthritis (eRA). METHODS Freshly isolated peripheral blood lymphocytes from 98 untreated eRA patients and 98 healthy controls (HC) were examined by cytometry. Twelve months (12 m) after initiating MTX, 82 patients were clinically re-evaluated and cytometry was repeated in 40 of them. The effect of MTX on Treg cell potency was assessed in Treg/Tresp cocultures. RESULTS The baseline (0 m) cTreg39+ cell frequency was elevated in eRA above HC levels. Patients who reached low disease activity at 12 months (12 m-LDA, DAS28-ESR ≤ 3.2, n = 51) had presented with a significantly higher 0 m cTreg39+ frequency vs those who did not (n = 31). The 0 m cTreg39+ cutoff for attaining 12 m-LDA was 42.0% (sensitivity = 90.4%, specificity = 96.8%). At 12 m, the cTreg39+ frequency was no longer elevated but its association with disease activity remained: it was still significantly higher in patients who had reached LDA vs those who had not. In vitro, MTX augmented the Treg39+ cell potency but had no effect on Treg39- cells. CONCLUSION MTX cooperates with Treg39+ cells and the baseline cTreg39+ frequency predicts the response to MTX in eRA. In addition, the transiently elevated baseline cTreg39+ frequency in eRA may provide a slot for prompt MTX initiation.
Collapse
Affiliation(s)
- Alejandro Villalba
- Department of Rheumatology, Hospital Universitario La Paz-IdiPaz, Madrid, Spain
| | - Laura Nuño
- Department of Rheumatology, Hospital Universitario La Paz-IdiPaz, Madrid, Spain
| | - Marta Benito-Miguel
- Fundación San Juan de Dios, Centro de Ciencias de la Salud San Rafael, Department of Physiology, Universidad de Nebrija, Madrid, Spain
| | | | - Irene Monjo
- Department of Rheumatology, Hospital Universitario La Paz-IdiPaz, Madrid, Spain
| | | | - Diana Peiteado
- Department of Rheumatology, Hospital Universitario La Paz-IdiPaz, Madrid, Spain
| | - Sara García-Carazo
- Department of Rheumatology, Hospital Universitario La Paz-IdiPaz, Madrid, Spain
| | - Alejandro Balsa
- Department of Rheumatology, Hospital Universitario La Paz-IdiPaz, Madrid, Spain
| | | |
Collapse
|
6
|
Attrill MH, Shinko D, Viveiros TM, Milighetti M, de Gruijter NM, Jebson B, Kartawinata M, Rosser EC, Wedderburn LR, Pesenacker AM. Treg fitness signatures as a biomarker for disease activity in Juvenile Idiopathic Arthritis. J Autoimmun 2025; 152:103379. [PMID: 39954509 DOI: 10.1016/j.jaut.2025.103379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/06/2025] [Accepted: 02/01/2025] [Indexed: 02/17/2025]
Abstract
Juvenile Idiopathic Arthritis (JIA) is an autoimmune condition characterised by flares of joint inflammation. However, no reliable biomarker exists to predict the erratic disease course. Normally, regulatory T cells (Tregs) maintain tolerance, with altered Tregs associated with autoimmunity. Treg signatures have shown promise in monitoring other conditions, therefore a Treg gene/protein signature could offer novel biomarker potential for predicting disease activity in JIA. Machine learning on our nanoString Treg 48-gene signature on peripheral blood (PB) Tregs generated a model to distinguish active JIA (active joint count, AJC≥1) Tregs from healthy controls (HC, AUC = 0.9875 on test data). Biomarker scores from this model successfully differentiated inactive (AJC = 0) from active JIA PB Tregs. Moreover, scores correlated with clinical activity scores (cJADAS), and discriminated subclinical disease (AJC = 0, cJADAS≥0.5) from remission (cJADAS<0.5). To investigate altered protein expression as a surrogate measure for Treg fitness in JIA, we utilised spectral flow cytometry and unbiased clustering analysis. Three Treg clusters were of interest in active JIA PB, including TIGIThighCD226highCD25low Teff-like Tregs, CD39-TNFR2-Helioshigh, and a 4-1BBlowTIGITlowID2intermediate Treg cluster predominated in inactive JIA PB (AJC = 0). The ratio of these Treg clusters correlated to cJADAS, and higher ratios could potentially predict inactive individuals that flared by 9-month follow-up. Thus, we demonstrate altered Treg signatures and subsets as an important factor, and useful biomarker, for disease progression versus remission in JIA, revealing genes and proteins contributing to Treg fitness. Ultimately, PB Treg fitness measures could serve as routine biomarkers to guide disease and treatment management to sustain remission in JIA.
Collapse
Affiliation(s)
- Meryl H Attrill
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, London, NW3 2PP, UK; Infection, Immunity & Inflammation Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, WC1N 1EH, UK
| | - Diana Shinko
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, London, NW3 2PP, UK
| | - Telma Martins Viveiros
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, London, NW3 2PP, UK
| | - Martina Milighetti
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, London, NW3 2PP, UK; Cancer Institute, UCL, London, WC1E 6DD, UK
| | - Nina M de Gruijter
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, WC1E 6JF, UK; Division of Medicine, UCL, London, WC1E 6JF, UK
| | - Bethany Jebson
- Infection, Immunity & Inflammation Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, WC1N 1EH, UK; Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, WC1E 6JF, UK
| | - Melissa Kartawinata
- Infection, Immunity & Inflammation Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, WC1N 1EH, UK; Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, WC1E 6JF, UK
| | - Elizabeth C Rosser
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, WC1E 6JF, UK; Division of Medicine, UCL, London, WC1E 6JF, UK
| | - Lucy R Wedderburn
- Infection, Immunity & Inflammation Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, WC1N 1EH, UK; Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, WC1E 6JF, UK; NIHR Biomedical Research Centre at GOSH, London, WC1N 1EH, UK
| | - Anne M Pesenacker
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, London, NW3 2PP, UK.
| |
Collapse
|
7
|
Capra ME, Aliverti V, Bellani AM, Berzieri M, Montani AG, Pisseri G, Sguerso T, Esposito S, Biasucci G. Breastfeeding and Non-Communicable Diseases: A Narrative Review. Nutrients 2025; 17:511. [PMID: 39940369 PMCID: PMC11819769 DOI: 10.3390/nu17030511] [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: 01/08/2025] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
INTRODUCTION Breastfeeding plays a fundamental role in newborns' and infants' health. Breast milk's protective power against malnutrition and its positive effect on neurological and physical development are well established and are reflected in the policy statements of all major pediatric health entities. However, breastfeeding also plays an important role in the prevention of so-called non-communicable diseases, such as obesity, hypertension, dyslipidemia, and autoimmune diseases. METHODS This narrative review aims to analyze the effect of breastfeeding and breast milk on the development of non-communicable diseases, with a special focus on weight excess, dyslipidemia, allergy, and gastrointestinal diseases. This narrative review was carried out through three steps: executing the search, examining abstracts and full texts, and analyzing results. To achieve this, the databases PubMed, EMBASE, Scopus, ScienceDirect, Web of Science, and Google Scholar were explored to collect and select publications from 1990 to 2024 to find pertinent studies in line with this review's development. The search included randomized placebo-controlled trials, controlled clinical trials, double-blind, randomized controlled studies, and systematic reviews. A total of 104 manuscripts were ultimately included in the analysis. RESULTS Breastfeeding is associated with a decreased vulnerability to early viral infections or chronic inflammatory conditions during preschool years, a reduced incidence of weight excess, and likely lower cholesterol concentration, besides having a small protective effect against systolic blood hypertension. CONCLUSIONS Pediatricians must promote breastfeeding, support the mother-infant dyad, and consider breast milk as a real "health voucher" that can last lifelong. However, further studies are needed to better define the extent and duration of breastfeeding's protective power in this context.
Collapse
Affiliation(s)
- Maria Elena Capra
- Pediatrics and Neonatology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy; (M.E.C.); (G.B.)
| | - Valentina Aliverti
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Arianna Maria Bellani
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Martina Berzieri
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Anna Giuseppina Montani
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Gianlorenzo Pisseri
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Tullia Sguerso
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Susanna Esposito
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Giacomo Biasucci
- Pediatrics and Neonatology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy; (M.E.C.); (G.B.)
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| |
Collapse
|
8
|
Lu Y, Wang Y, Ruan T, Wang Y, Ju L, Zhou M, Liu L, Yao D, Yao M. Immunometabolism of Tregs: mechanisms, adaptability, and therapeutic implications in diseases. Front Immunol 2025; 16:1536020. [PMID: 39917294 PMCID: PMC11798928 DOI: 10.3389/fimmu.2025.1536020] [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: 11/28/2024] [Accepted: 01/06/2025] [Indexed: 02/09/2025] Open
Abstract
Immunometabolism is an emerging field that explores the intricate interplay between immune cells and metabolism. Regulatory T cells (Tregs), which maintain immune homeostasis in immunometabolism, play crucial regulatory roles. The activation, differentiation, and function of Tregs are influenced by various metabolic pathways, such as the Mammalian targets of rapamycin (mTOR) pathway and glycolysis. Correspondingly, activated Tregs can reciprocally impact these metabolic pathways. Tregs also possess robust adaptive capabilities, thus enabling them to adapt to various microenvironments, including the tumor microenvironment (TME). The complex mechanisms of Tregs in metabolic diseases are intriguing, particularly in conditions like MASLD, where Tregs are significantly upregulated and contribute to fibrosis, while in diabetes, systemic lupus erythematosus (SLE), and rheumatoid arthritis (RA), they show downregulation and reduced anti-inflammatory capacity. These phenomena suggest that the differentiation and function of Tregs are influenced by the metabolic environment, and imbalances in either can lead to the development of metabolic diseases. Thus, moderate differentiation and inhibitory capacity of Tregs are critical for maintaining immune system balance. Given the unique immunoregulatory abilities of Tregs, the development of targeted therapeutic drugs may position them as novel targets in immunotherapy. This could contribute to restoring immune system balance, resolving metabolic dysregulation, and fostering innovation and progress in immunotherapy.
Collapse
|
9
|
Huang T, Li F, Wang Y, Gu J, Lu L. Tumor-infiltrating regulatory T cell: A promising therapeutic target in tumor microenvironment. Chin Med J (Engl) 2024; 137:2996-3009. [PMID: 39679474 PMCID: PMC11706582 DOI: 10.1097/cm9.0000000000003450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Indexed: 12/17/2024] Open
Abstract
ABSTRACT Regulatory T cell (Tregs) predominantly maintain the immune balance and prevent autoimmunity via their immunosuppressive functions. However, tumor-infiltrating Tregs (TI-Tregs) may mediate tumor immune tolerance in complex tumor microenvironments, resulting in poor prognosis. Distinguishing specific TI-Treg subpopulations from peripheral Tregs and intratumoral conventional T cells (Tconvs) has recently emerged as an important topic in antitumor therapy. In this review, we summarize novel therapeutic approaches targeting both the metabolic pathways and hallmarks of TI-Tregs in preclinical and clinical studies. Although the phenotypic and functional diversity of TI-Tregs remains unclear, our review provides new insights into TI-Treg-based therapies and facilitates precision medicine for tumor treatment.
Collapse
Affiliation(s)
- Tianning Huang
- Department of Plastic and Cosmetic Surgery of the Affiliated Friendship Plastic Surgery Hospital & Hepatobiliary Center of the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu 210029, China
| | - Fan Li
- Department of Plastic and Cosmetic Surgery of the Affiliated Friendship Plastic Surgery Hospital & Hepatobiliary Center of the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu 210029, China
| | - Yiming Wang
- Department of Plastic and Cosmetic Surgery of the Affiliated Friendship Plastic Surgery Hospital & Hepatobiliary Center of the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu 210029, China
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jian Gu
- Department of Plastic and Cosmetic Surgery of the Affiliated Friendship Plastic Surgery Hospital & Hepatobiliary Center of the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu 210029, China
| | - Ling Lu
- Department of Plastic and Cosmetic Surgery of the Affiliated Friendship Plastic Surgery Hospital & Hepatobiliary Center of the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu 210029, China
- Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221011, China
| |
Collapse
|
10
|
Cochrane RW, Robino RA, Granger B, Allen E, Vaena S, Romeo MJ, de Cubas AA, Berto S, Ferreira LM. High-affinity chimeric antigen receptor signaling induces an inflammatory program in human regulatory T cells. Mol Ther Methods Clin Dev 2024; 32:101385. [PMID: 39687729 PMCID: PMC11647616 DOI: 10.1016/j.omtm.2024.101385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 11/14/2024] [Indexed: 12/18/2024]
Abstract
Regulatory T cells (Tregs) are promising cellular therapies to induce immune tolerance in organ transplantation and autoimmune disease. The success of chimeric antigen receptor (CAR) T cell therapy for cancer has sparked interest in using CARs to generate antigen-specific Tregs. Here, we compared CAR with endogenous T cell receptor (TCR)/CD28 activation in human Tregs. Strikingly, CAR Tregs displayed increased cytotoxicity and diminished suppression of antigen-presenting cells and effector T (Teff) cells compared with TCR/CD28-activated Tregs. RNA sequencing revealed that CAR Tregs activate Teff cell gene programs. Indeed, CAR Tregs secreted high levels of inflammatory cytokines, with a subset of FOXP3+ CAR Tregs uniquely acquiring CD40L surface expression and producing IFN-γ. Interestingly, decreasing CAR antigen affinity reduced Teff cell gene expression and inflammatory cytokine production by CAR Tregs. Our findings showcase the impact of engineered receptor activation on Treg biology and support tailoring CAR constructs to Tregs for maximal therapeutic efficacy.
Collapse
Affiliation(s)
- Russell W. Cochrane
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Rob A. Robino
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Bryan Granger
- Bioinformatics Core, Medical University of South Carolina, Charleston, SC, USA
| | - Eva Allen
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Silvia Vaena
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Martin J. Romeo
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Aguirre A. de Cubas
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Stefano Berto
- Bioinformatics Core, Medical University of South Carolina, Charleston, SC, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Leonardo M.R. Ferreira
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
11
|
Attrill MH, Shinko D, Alexiou V, Kartawinata M, Wedderburn LR, Pesenacker AM. The immune landscape of the inflamed joint defined by spectral flow cytometry. Clin Exp Immunol 2024; 218:221-241. [PMID: 39101538 PMCID: PMC11557149 DOI: 10.1093/cei/uxae071] [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: 01/05/2024] [Revised: 06/10/2024] [Accepted: 08/02/2024] [Indexed: 08/06/2024] Open
Abstract
Cellular phenotype and function are altered in different microenvironments. For targeted therapies it is important to understand site-specific cellular adaptations. Juvenile idiopathic arthritis (JIA) is characterized by autoimmune joint inflammation, with frequent inadequate treatment responses. To comprehensively assess the inflammatory immune landscape, we designed a 37-parameter spectral flow cytometry panel delineating mononuclear cells from JIA synovial fluid (SF) of autoimmune inflamed joints, compared to JIA and healthy control blood. Synovial monocytes and NK cells (CD56bright) lack Fc-receptor CD16, suggesting antibody-mediated targeting may be ineffective. B cells and DCs, both in small frequencies in SF, undergo maturation with high 4-1BB, CD71, CD39 expression, supporting T-cell activation. SF effector and regulatory T cells were highly active with newly described co-receptor combinations that may alter function, and suggestion of metabolic reprogramming via CD71, TNFR2, and PD-1. Most SF effector phenotypes, as well as an identified CD4-Foxp3+ T-cell population, were restricted to the inflamed joint, yet specific SF-predominant CD4+ Foxp3+ Treg subpopulations were increased in blood of active but not inactive JIA, suggesting possible recirculation and loss of immunoregulation at distal sites. This first comprehensive dataset of the site-specific inflammatory landscape at protein level will inform functional studies and the development of targeted therapeutics to restore immunoregulatory balance and achieve remission in JIA.
Collapse
Affiliation(s)
- Meryl H Attrill
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
- UCL Great Ormond Street Institute of Child Health, Infection, Immunity, and Inflammation Research and Teaching Department, University College London, London, UK
| | - Diana Shinko
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Vicky Alexiou
- UCL Great Ormond Street Institute of Child Health, Infection, Immunity, and Inflammation Research and Teaching Department, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, UK
- Centre for Rheumatology, Division of Medicine, University College London, London, UK
| | - Melissa Kartawinata
- UCL Great Ormond Street Institute of Child Health, Infection, Immunity, and Inflammation Research and Teaching Department, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, UK
| | - Lucy R Wedderburn
- UCL Great Ormond Street Institute of Child Health, Infection, Immunity, and Inflammation Research and Teaching Department, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, UK
- NIHR Biomedical Research Centre at GOSH, London, UK
| | - Anne M Pesenacker
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|
12
|
Krüger J, Wellbrock J, Witt M, Kruppa N, Muschhammer J, Bokemeyer C, Modemann F, Fiedler W, Behrmann L, Brauneck F. Murine Regulatory CD4 + T Cells Are Increased in Leukemic Spleens and Show High Co-Expression of Co-Regulatory Molecules CD39, CD73, PD1, and TIGIT. Int J Mol Sci 2024; 25:11412. [PMID: 39518969 PMCID: PMC11546357 DOI: 10.3390/ijms252111412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/16/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
Comprehensive characterization of AML-associated T cells during disease progression is essential to identify relevant immune escape mechanisms and new immunotherapeutic approaches. Investigating the processes that lead to an immunosuppressive environment under progression of AML is difficult in humans, because by the time of diagnosis the disease is often progressed far beyond the initial stages. Therefore, to investigate T-cell phenotypes during progression a C57BL/6 mouse model was used. The CD3+ T cells were characterized by performing multiparametric flow analyses at different time points (day 0 = healthy mice, day 7, day 14, and day 21). The study revealed that the spleen is highly infiltrated by reg CD4+ T cells at day 21 of AML progression. These spleen-infiltrating reg CD4+ T cells mainly showed an effector memory differentiation with high expression and co-expression of the checkpoint molecules TIGIT, PD-1, OX40, and the two ectoenzymes CD39 and CD73. Substantial expression of the checkpoint molecules was restricted to the central memory and effector memory compartments. Furthermore, functional evaluation of TIGIT was performed. Blocking TIGIT resulted in a significantly increased lysis of C1498 AML cells in cocultures with AML-primed CD3+ T cells. Together these data confirm that the expression of the checkpoint receptor TIGIT is relevant for dysfunction of AML-associated T cells and, thus, represents a suitable target for future immunotherapeutic approaches.
Collapse
Affiliation(s)
| | - Jasmin Wellbrock
- Correspondence: (J.W.); (W.F.); Tel.: +49-(0)40-7410-55606 or +49-(0)40-7410-55931 (J.W.); +49-(0)40-7410-23051 (W.F.)
| | | | | | | | | | | | - Walter Fiedler
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (J.K.); (M.W.); (N.K.); (J.M.); (C.B.); (F.M.); (L.B.); (F.B.)
| | | | | |
Collapse
|
13
|
Poblano-Pérez LI, Monroy-García A, Fragoso-González G, Mora-García MDL, Castell-Rodríguez A, Mayani H, Álvarez-Pérez MA, Pérez-Tapia SM, Macías-Palacios Z, Vallejo-Castillo L, Montesinos JJ. Mesenchymal Stem/Stromal Cells Derived from Dental Tissues Mediate the Immunoregulation of T Cells through the Purinergic Pathway. Int J Mol Sci 2024; 25:9578. [PMID: 39273524 PMCID: PMC11395442 DOI: 10.3390/ijms25179578] [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: 06/04/2024] [Revised: 08/26/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Human dental tissue mesenchymal stem cells (DT-MSCs) constitute an attractive alternative to bone marrow-derived mesenchymal stem cells (BM-MSCs) for potential clinical applications because of their accessibility and anti-inflammatory capacity. We previously demonstrated that DT-MSCs from dental pulp (DP-MSCs), periodontal ligaments (PDL-MSCs), and gingival tissue (G-MSCs) show immunosuppressive effects similar to those of BM, but to date, the DT-MSC-mediated immunoregulation of T lymphocytes through the purinergic pathway remains unknown. In the present study, we compared DP-MSCs, PDL-MSCs, and G-MSCs in terms of CD26, CD39, and CD73 expression; their ability to generate adenosine (ADO) from ATP and AMP; and whether the concentrations of ADO that they generate induce an immunomodulatory effect on T lymphocytes. BM-MSCs were included as the gold standard. Our results show that DT-MSCs present similar characteristics among the different sources analyzed in terms of the properties evaluated; however, interestingly, they express more CD39 than BM-MSCs; therefore, they generate more ADO from ATP. In contrast to those produced by BM-MSCs, the concentrations of ADO produced by DT-MSCs from ATP inhibited the proliferation of CD3+ T cells and promoted the generation of CD4+CD25+FoxP3+CD39+CD73+ Tregs and Th17+CD39+ lymphocytes. Our data suggest that DT-MSCs utilize the adenosinergic pathway as an immunomodulatory mechanism and that this mechanism is more efficient than that of BM-MSCs.
Collapse
Affiliation(s)
- Luis Ignacio Poblano-Pérez
- Mesenchymal Stem Cell Laboratory, Oncology Research Unit, Oncology Hospital, Centro Médico Nacional SXXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Alberto Monroy-García
- Immunology and Cancer Laboratory, Oncology Research Unit, Oncology Hospital, Centro Médico Nacional SXXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Gladis Fragoso-González
- Institute of Biomedical Research, Department of Immunology, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - María de Lourdes Mora-García
- Immunobiology Laboratory, Cell Differentiation and Cancer Unit, Facultad de Estudios Superiores-Zaragoza, Universidad Nacional Autónoma de México, Mexico City 09230, Mexico
| | - Andrés Castell-Rodríguez
- Department of Cellular and Tissue Biology, Faculty of Medicine, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Héctor Mayani
- Hematopoietic Stem Cell Laboratory, Oncology Research Unit, Oncology Hospital, Centro Médico Nacional SXXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Marco Antonio Álvarez-Pérez
- Tissue Bioengineering Laboratory, Postgraduate Studies, Research Division, Faculty of Dentistry, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Sonia Mayra Pérez-Tapia
- Research and Development in Biotherapeutic Unit (UDIBI), National School of Biological Sciences, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- National Laboratory for Specialized Services of Investigation, Development and Innovation (I+D+i) for Pharma Chemicals and Biotechnological Products (LANSEIDI-FarBiotec-CONACyT), Instituto Politécnico Nacional, Mexico City 11340, Mexico
- Department of Immunology, National School of Biological Sciences, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Zaira Macías-Palacios
- Research and Development in Biotherapeutic Unit (UDIBI), National School of Biological Sciences, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- National Laboratory for Specialized Services of Investigation, Development and Innovation (I+D+i) for Pharma Chemicals and Biotechnological Products (LANSEIDI-FarBiotec-CONACyT), Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Luis Vallejo-Castillo
- Research and Development in Biotherapeutic Unit (UDIBI), National School of Biological Sciences, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- National Laboratory for Specialized Services of Investigation, Development and Innovation (I+D+i) for Pharma Chemicals and Biotechnological Products (LANSEIDI-FarBiotec-CONACyT), Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Juan José Montesinos
- Mesenchymal Stem Cell Laboratory, Oncology Research Unit, Oncology Hospital, Centro Médico Nacional SXXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| |
Collapse
|
14
|
Rodriguez E, Zwart ES, Affandi AA, Verhoeff J, de Kok M, Boyd LNC, Meijer LL, Le Large TYS, Olesek K, Giovannetti E, García-Vallejo JJ, Mebius RE, van Kooyk Y, Kazemier G. In-depth immune profiling of peripheral blood mononuclear cells in patients with pancreatic ductal adenocarcinoma reveals discriminative immune subpopulations. Cancer Sci 2024; 115:2170-2183. [PMID: 38686549 PMCID: PMC11247553 DOI: 10.1111/cas.16147] [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: 09/16/2023] [Revised: 02/05/2024] [Accepted: 02/13/2024] [Indexed: 05/02/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis with a 5-year survival of less than 10%. More knowledge of the immune response developed in patients with PDAC is pivotal to develop better combination immune therapies to improve clinical outcome. In this study, we used mass cytometry time-of-flight to undertake an in-depth characterization of PBMCs from patients with PDAC and examine the differences with healthy controls and patients with benign diseases of the biliary system or pancreas. Peripheral blood mononuclear cells from patients with PDAC or benign disease are characterized by the increase of pro-inflammatory cells, as CD86+ classical monocytes and memory T cells expressing CCR6+ and CXCR3+, associated with T helper 1 (Th1) and Th17 immune responses, respectively. However, PBMCs from patients with PDAC present also an increase of CD39+ regulatory T cells and CCR4+CCR6-CXCR3- memory T cells, suggesting Th2 and regulatory responses. Concluding, our results show PDAC develops a multifaceted immunity, where a proinflammatory component is accompanied by regulatory responses, which could inhibit potential antitumor mechanisms.
Collapse
Affiliation(s)
- Ernesto Rodriguez
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Eline S Zwart
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
| | - Alsya A Affandi
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Jan Verhoeff
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Mike de Kok
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
| | - Lenka N C Boyd
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
| | - Laura L Meijer
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
| | - Tessa Y S Le Large
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
| | - Katarzyna Olesek
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisana per la Scienza, Pisa, Italy
| | - Juan J García-Vallejo
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Reina E Mebius
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Geert Kazemier
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Honing DY, Luiten RM, Matos TR. Regulatory T Cell Dysfunction in Autoimmune Diseases. Int J Mol Sci 2024; 25:7171. [PMID: 39000278 PMCID: PMC11241405 DOI: 10.3390/ijms25137171] [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: 06/11/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Regulatory T cells (Tregs), a suppressive subpopulation of T cells, are potent mediators of peripheral tolerance, responsible for immune homeostasis. Many autoimmune diseases exhibit disruptions in Treg function or quantity, resulting in an imbalance between protective and pathogenic immune cells. Selective expansion or manipulation of Tregs is a promising therapeutic approach for autoimmune diseases. However, the extensive diversity of Treg subpopulations and the multiple approaches used for Treg identification leads to high complexity, making it difficult to develop a successful treatment capable of modulating Tregs. In this review, we describe the suppressive mechanisms, subpopulations, classification, and identification methodology for Tregs, and their role in the pathogenesis of autoimmune diseases.
Collapse
Affiliation(s)
- Dionne Y Honing
- Department of Dermatology, Netherlands Institute for Pigment Disorders, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, 1081 HV Amsterdam, The Netherlands
| | - Rosalie M Luiten
- Department of Dermatology, Netherlands Institute for Pigment Disorders, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, 1081 HV Amsterdam, The Netherlands
| | - Tiago R Matos
- Department of Dermatology, Netherlands Institute for Pigment Disorders, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Sanofi, 1105 BP Amsterdam, The Netherlands
| |
Collapse
|
16
|
Wu X, Chen PI, Whitener RL, MacDougall MS, Coykendall VMN, Yan H, Kim YB, Harper W, Pathak S, Iliopoulou BP, Hestor A, Saunders DC, Spears E, Sévigny J, Maahs DM, Basina M, Sharp SA, Gloyn AL, Powers AC, Kim SK, Jensen KP, Meyer EH. CD39 delineates chimeric antigen receptor regulatory T cell subsets with distinct cytotoxic & regulatory functions against human islets. Front Immunol 2024; 15:1415102. [PMID: 39007132 PMCID: PMC11239501 DOI: 10.3389/fimmu.2024.1415102] [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: 04/09/2024] [Accepted: 06/13/2024] [Indexed: 07/16/2024] Open
Abstract
Human regulatory T cells (Treg) suppress other immune cells. Their dysfunction contributes to the pathophysiology of autoimmune diseases, including type 1 diabetes (T1D). Infusion of Tregs is being clinically evaluated as a novel way to prevent or treat T1D. Genetic modification of Tregs, most notably through the introduction of a chimeric antigen receptor (CAR) targeting Tregs to pancreatic islets, may improve their efficacy. We evaluated CAR targeting of human Tregs to monocytes, a human β cell line and human islet β cells in vitro. Targeting of HLA-A2-CAR (A2-CAR) bulk Tregs to HLA-A2+ cells resulted in dichotomous cytotoxic killing of human monocytes and islet β cells. In exploring subsets and mechanisms that may explain this pattern, we found that CD39 expression segregated CAR Treg cytotoxicity. CAR Tregs from individuals with more CD39low/- Tregs and from individuals with genetic polymorphism associated with lower CD39 expression (rs10748643) had more cytotoxicity. Isolated CD39- CAR Tregs had elevated granzyme B expression and cytotoxicity compared to the CD39+ CAR Treg subset. Genetic overexpression of CD39 in CD39low CAR Tregs reduced their cytotoxicity. Importantly, β cells upregulated protein surface expression of PD-L1 and PD-L2 in response to A2-CAR Tregs. Blockade of PD-L1/PD-L2 increased β cell death in A2-CAR Treg co-cultures suggesting that the PD-1/PD-L1 pathway is important in protecting islet β cells in the setting of CAR immunotherapy. In summary, introduction of CAR can enhance biological differences in subsets of Tregs. CD39+ Tregs represent a safer choice for CAR Treg therapies targeting tissues for tolerance induction.
Collapse
Affiliation(s)
- Xiangni Wu
- Department of Medicine, Division of Blood and Bone Marrow Transplantation and Cell Therapy, Stanford University School of Medicine, Stanford, CA, United States
- Department of Internal Medicine, University of Missouri Kansas City, Kansas City, MO, United States
| | - Pin-I Chen
- Department of Medicine, Division of Blood and Bone Marrow Transplantation and Cell Therapy, Stanford University School of Medicine, Stanford, CA, United States
| | - Robert L. Whitener
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, United States
| | - Matthew S. MacDougall
- Departments of Medicine and of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Vy M. N. Coykendall
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, United States
| | - Hao Yan
- Department of Medicine, Division of Blood and Bone Marrow Transplantation and Cell Therapy, Stanford University School of Medicine, Stanford, CA, United States
| | - Yong Bin Kim
- Department of Medicine, Division of Blood and Bone Marrow Transplantation and Cell Therapy, Stanford University School of Medicine, Stanford, CA, United States
- Department of Chemical Engineering, Stanford University, Stanford, CA, United States
| | - William Harper
- Department of Medicine, Division of Blood and Bone Marrow Transplantation and Cell Therapy, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Diabetes Research Center (SDRC), Stanford University School of Medicine, Stanford, CA, United States
| | - Shiva Pathak
- Department of Medicine, Division of Blood and Bone Marrow Transplantation and Cell Therapy, Stanford University School of Medicine, Stanford, CA, United States
| | - Bettina P. Iliopoulou
- Department of Medicine, Division of Blood and Bone Marrow Transplantation and Cell Therapy, Stanford University School of Medicine, Stanford, CA, United States
| | - Allison Hestor
- Department of Medicine, Division of Blood and Bone Marrow Transplantation and Cell Therapy, Stanford University School of Medicine, Stanford, CA, United States
| | - Diane C. Saunders
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Erick Spears
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jean Sévigny
- Centre de recherche du centre hospitalier universitaire (CHU) de Québec – Université Laval, Québec City, QC, Canada
- Départment de Microbiologie-Infectiologie et d’Immunologie, Faculté de Médecine, Université Laval, Québec City, QC, Canada
| | - David M. Maahs
- Stanford Diabetes Research Center (SDRC), Stanford University School of Medicine, Stanford, CA, United States
- Department of Pediatrics, Division of Pediatric Endocrinology, Stanford University School of Medicine, Stanford, CA, United States
| | - Marina Basina
- Stanford Diabetes Research Center (SDRC), Stanford University School of Medicine, Stanford, CA, United States
- Department of Medicine, Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Stanford, CA, United States
| | - Seth A. Sharp
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Anna L. Gloyn
- Stanford Diabetes Research Center (SDRC), Stanford University School of Medicine, Stanford, CA, United States
- Department of Pediatrics, Division of Pediatric Endocrinology, Stanford University School of Medicine, Stanford, CA, United States
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Alvin C. Powers
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Veterans Affairs (VA) Tennessee Valley Healthcare System, Nashville, TN, United States
| | - Seung K. Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Diabetes Research Center (SDRC), Stanford University School of Medicine, Stanford, CA, United States
- Department of Medicine, Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Stanford, CA, United States
- The Juvenile Diabetes Research Foundation (JDRF) Northern California Center of Excellence, Stanford University School of Medicine, Stanford, CA, United States
| | - Kent P. Jensen
- Department of Medicine, Division of Blood and Bone Marrow Transplantation and Cell Therapy, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Diabetes Research Center (SDRC), Stanford University School of Medicine, Stanford, CA, United States
- The Juvenile Diabetes Research Foundation (JDRF) Northern California Center of Excellence, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Department of Medicine, Stanford, CA, United States
| | - Everett H. Meyer
- Department of Medicine, Division of Blood and Bone Marrow Transplantation and Cell Therapy, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Diabetes Research Center (SDRC), Stanford University School of Medicine, Stanford, CA, United States
- The Juvenile Diabetes Research Foundation (JDRF) Northern California Center of Excellence, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Department of Medicine, Stanford, CA, United States
- Department of Pediatrics, Division of Stem Cell Transplantation, Stanford University School of Medicine, Stanford, CA, United States
- Department of Surgery, Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
17
|
Li SY, Xue ST, Li ZR. Osteoporosis: Emerging targets on the classical signaling pathways of bone formation. Eur J Pharmacol 2024; 973:176574. [PMID: 38642670 DOI: 10.1016/j.ejphar.2024.176574] [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: 12/06/2023] [Revised: 03/30/2024] [Accepted: 04/10/2024] [Indexed: 04/22/2024]
Abstract
Osteoporosis is a multifaceted skeletal disorder characterized by reduced bone mass and structural deterioration, posing a significant public health challenge, particularly in the elderly population. Treatment strategies for osteoporosis primarily focus on inhibiting bone resorption and promoting bone formation. However, the effectiveness and limitations of current therapeutic approaches underscore the need for innovative methods. This review explores emerging molecular targets within crucial signaling pathways, including wingless/integrated (WNT), bone morphogenetic protein (BMP), hedgehog (HH), and Notch signaling pathway, to understand their roles in osteogenesis regulation. The identification of crosstalk targets between these pathways further enhances our comprehension of the intricate bone metabolism cycle. In summary, unraveling the molecular complexity of osteoporosis provides insights into potential therapeutic targets beyond conventional methods, offering a promising avenue for the development of new anabolic drugs.
Collapse
Affiliation(s)
- Si-Yan Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Si-Tu Xue
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Zhuo-Rong Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
18
|
Sann S, Heng B, Vo HTM, Arroyo Hornero R, Lay S, Sorn S, Ken S, Ou TP, Laurent D, Yay C, Ly S, Dussart P, Duong V, Sakuntabhai A, Kleinewietfeld M, Cantaert T. Increased frequencies of highly activated regulatory T cells skewed to a T helper 1-like phenotype with reduced suppressive capacity in dengue patients. mBio 2024; 15:e0006324. [PMID: 38752787 PMCID: PMC11237415 DOI: 10.1128/mbio.00063-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/08/2024] [Indexed: 06/13/2024] Open
Abstract
The pathogenesis of dengue involves a complex interplay between the viral factor and the host immune response. A mismatch between the infecting serotype and the adaptive memory response is hypothesized to lead to exacerbated immune responses resulting in severe dengue. Here, we aim to define in detail the phenotype and function of different regulatory T cell (Treg) subsets and their association with disease severity in a cohort of acute dengue virus (DENV)-infected Cambodian children. Treg frequencies and proliferation of Tregs are increased in dengue patients compared to age-matched controls. Tregs from dengue patients are skewed to a Th1-type Treg phenotype. Interestingly, Tregs from severe dengue patients produce more interleukin-10 after in vitro stimulation compared to Tregs from classical dengue fever patients. Functionally, Tregs from dengue patients have reduced suppressive capacity, irrespective of disease severity. Taken together, these data suggest that even though Treg frequencies are increased in the blood of acute DENV-infected patients, Tregs fail to resolve inflammation and thereby could contribute to the immunopathology of dengue. IMPORTANCE According to the World Health Organization, dengue is the fastest-spreading, epidemic-prone infectious disease. The extent of dengue virus infections increased over the years, mainly driven by globalization-including travel and trade-and environmental changes. Dengue is an immunopathology caused by an imbalanced immune response to a secondary heterotypic infection. As regulatory T cells (Tregs) are essential in maintaining immune homeostasis and dampening excessive immune activation, this study addressed the role of Tregs in dengue immunopathology. We show that Tregs from dengue patients are highly activated, skewed to a Th1-like Treg phenotype and less suppressive compared to healthy donor Tregs. Our data suggest that Tregs fail to resolve ongoing inflammation during dengue infection and hence contribute to the immunopathology of severe dengue disease. These data clarify the role of Tregs in dengue immunopathogenesis, emphasizing the need to develop T cell-based vaccines for dengue.
Collapse
Affiliation(s)
- Sotheary Sann
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
- VIB Laboratory of Translational Immunomodulation, Hasselt University, Diepenbeek, Belgium
- Department of Immunology, Hasselt University, Diepenbeek, Belgium
- University Multiple Sclerosis Center, Hasselt University, Diepenbeek, Belgium
| | - Borita Heng
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Hoa Thi My Vo
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Rebeca Arroyo Hornero
- VIB Laboratory of Translational Immunomodulation, Hasselt University, Diepenbeek, Belgium
- Department of Immunology, Hasselt University, Diepenbeek, Belgium
- University Multiple Sclerosis Center, Hasselt University, Diepenbeek, Belgium
| | - Sokchea Lay
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Sopheak Sorn
- Epidemiology and Public Health Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Sreymom Ken
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Tey Putita Ou
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Denis Laurent
- Kantha Bopha Children's Hospital, Phnom Penh, Cambodia
| | | | - Sowath Ly
- Epidemiology and Public Health Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Philippe Dussart
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Veasna Duong
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Anavaj Sakuntabhai
- Department of Global Health, Ecology and Emergence of Arthropod-borne Pathogens, Institut Pasteur, Université de Paris, Paris, France
- Université de Paris-Cité, CNRS UMR 2000, Paris, France
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE) USC 1510, Paris, France
| | - Markus Kleinewietfeld
- VIB Laboratory of Translational Immunomodulation, Hasselt University, Diepenbeek, Belgium
- Department of Immunology, Hasselt University, Diepenbeek, Belgium
- University Multiple Sclerosis Center, Hasselt University, Diepenbeek, Belgium
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| |
Collapse
|
19
|
Blinova VG, Zhdanov DD. Many Faces of Regulatory T Cells: Heterogeneity or Plasticity? Cells 2024; 13:959. [PMID: 38891091 PMCID: PMC11171907 DOI: 10.3390/cells13110959] [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: 04/23/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Regulatory T cells (Tregs) are essential for maintaining the immune balance in normal and pathological conditions. In autoimmune diseases and transplantation, they restrain the loss of self-tolerance and promote engraftment, whereas in cancer, an increase in Treg numbers is mostly associated with tumor growth and poor prognosis. Numerous markers and their combinations have been used to identify Treg subsets, demonstrating the phenotypic diversity of Tregs. The complexity of Treg identification can be hampered by the unstable expression of some markers, the decrease in the expression of a specific marker over time or the emergence of a new marker. It remains unclear whether such phenotypic shifts are due to new conditions or whether the observed changes are due to initially different populations. In the first case, cellular plasticity is observed, whereas in the second, cellular heterogeneity is observed. The difference between these terms in relation to Tregs is rather blurred. Considering the promising perspectives of Tregs in regenerative cell-based therapy, the existing confusing data on Treg phenotypes require further investigation and analysis. In our review, we introduce criteria that allow us to distinguish between the heterogeneity and plasticity of Tregs normally and pathologically, taking a closer look at their diversity and drawing the line between two terms.
Collapse
Affiliation(s)
- Varvara G. Blinova
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya st. 10/8, 119121 Moscow, Russia;
| | - Dmitry D. Zhdanov
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya st. 10/8, 119121 Moscow, Russia;
- Department of Biochemistry, People’s Friendship University of Russia Named after Patrice Lumumba (RUDN University), Miklukho-Maklaya st. 6, 117198 Moscow, Russia
| |
Collapse
|
20
|
Zhang QW, Zhu MX, Liu WF, Rui WW, Chen Y, Ding XY, Jiang YS, Wu ZY, Liu BB. Identification of clinically relevant subsets CD39 +PD-1 +CD8 + T cells and CD39 + regulatory T cells in intrahepatic cholangiocarcinoma using single-cell CyTOF. Transl Oncol 2024; 44:101954. [PMID: 38608405 PMCID: PMC11024660 DOI: 10.1016/j.tranon.2024.101954] [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: 01/21/2024] [Revised: 03/05/2024] [Accepted: 03/31/2024] [Indexed: 04/14/2024] Open
Abstract
Intrahepatic cholangiocarcinoma (iCCA) is an aggressive liver malignancy with limited treatment options and a dismal prognosis. The tumor immune microenvironment (TIME) is crucial for iCCA progression, yet its comprehensive characterization remains incomplete. This study utilized mass cytometry by time of flight (CyTOF) to comprehensively analyze immune cell populations in fresh iCCA tumor samples and adjacent peritumor liver tissues. Notably, NK cell percentages significantly decreased in iCCA lesions compared to peritumor liver tissues. Conversely, an enrichment of immunosuppressive CD39+Foxp3+CD4+ regulatory T cells (CD39+T-regs) and exhausted-like CD8+T cells (with pronounced CD39 and PD-1 expression) within TIME was identified and confirmed by multiplex immunofluorescence staining in an independent patient cohort (n = 140). Crucially, tumor-infiltrating CD39+T-regs and CD39+PD-1+CD8+T cells emerged as independent prognostic indicators associated with an unfavorable prognosis in iCCA. These findings unveil the intricate immune landscape within iCCA, offering valuable insights for disease management and novel cancer immunotherapies.
Collapse
Affiliation(s)
- Qi-Wei Zhang
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai 200025, China
| | - Meng-Xuan Zhu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wen-Feng Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Wei-Wei Rui
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yong Chen
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiao-Yi Ding
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai 200025, China.
| | - Yong-Sheng Jiang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai 200025, China; Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Zhi-Yuan Wu
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai 200025, China.
| | - Bin-Bin Liu
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| |
Collapse
|
21
|
Gootjes C, Zwaginga JJ, Roep BO, Nikolic T. Defining Human Regulatory T Cells beyond FOXP3: The Need to Combine Phenotype with Function. Cells 2024; 13:941. [PMID: 38891073 PMCID: PMC11172350 DOI: 10.3390/cells13110941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/18/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Regulatory T cells (Tregs) are essential to maintain immune homeostasis by promoting self-tolerance. Reduced Treg numbers or functionality can lead to a loss of tolerance, increasing the risk of developing autoimmune diseases. An overwhelming variety of human Tregs has been described, based on either specific phenotype, tissue compartment, or pathological condition, yet the bulk of the literature only addresses CD25-positive and CD127-negative cells, coined by naturally occurring Tregs (nTregs), most of which express the transcription factor Forkhead box protein 3 (FOXP3). While the discovery of FOXP3 was seminal to understanding the origin and biology of nTregs, there is evidence in humans that not all T cells expressing FOXP3 are regulatory, and that not all Tregs express FOXP3. Namely, the activation of human T cells induces the transient expression of FOXP3, irrespective of whether they are regulatory or inflammatory effectors, while some induced T cells that may be broadly defined as Tregs (e.g., Tr1 cells) typically lack demethylation and do not express FOXP3. Furthermore, it is unknown whether and how many nTregs exist without FOXP3 expression. Several other candidate regulatory molecules, such as GITR, Lag-3, GARP, GPA33, Helios, and Neuropilin, have been identified but subsequently discarded as Treg-specific markers. Multiparametric analyses have uncovered a plethora of Treg phenotypes, and neither single markers nor combinations thereof can define all and only Tregs. To date, only the functional capacity to inhibit immune responses defines a Treg and distinguishes Tregs from inflammatory T cells (Teffs) in humans. This review revisits current knowledge of the Treg universe with respect to their heterogeneity in phenotype and function. We propose that it is unavoidable to characterize human Tregs by their phenotype in combination with their function, since phenotype alone does not unambiguously define Tregs. There is an unmet need to align the expression of specific markers or combinations thereof with a particular suppressive function to coin functional Treg entities and categorize Treg diversity.
Collapse
Affiliation(s)
- Chelsea Gootjes
- Laboratory of Immunomodulation and Regenerative Cell Therapy, Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (J.J.Z.); (T.N.)
| | | | | | | |
Collapse
|
22
|
Kim CG, Hong MH, Kim D, Lee BH, Kim H, Ock CY, Kelly G, Bang YJ, Kim G, Lee JE, Kim C, Kim SH, Hong HJ, Park YM, Sim NS, Park H, Park JW, Lee CG, Kim KH, Park G, Jung I, Han D, Kim JH, Cha J, Lee I, Kang M, Song H, Oum C, Kim S, Kim S, Lim Y, Kim-Schulze S, Merad M, Yoon SO, Kim HJ, Koh YW, Kim HR. A Phase II Open-Label Randomized Clinical Trial of Preoperative Durvalumab or Durvalumab plus Tremelimumab in Resectable Head and Neck Squamous Cell Carcinoma. Clin Cancer Res 2024; 30:2097-2110. [PMID: 38457288 DOI: 10.1158/1078-0432.ccr-23-3249] [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: 10/25/2023] [Revised: 01/06/2024] [Accepted: 03/06/2024] [Indexed: 03/10/2024]
Abstract
PURPOSE Clinical implications of neoadjuvant immunotherapy in patients with locally advanced but resectable head and neck squamous cell carcinoma (HNSCC) remain largely unexplored. PATIENTS AND METHODS Patients with resectable HNSCC were randomized to receive a single dose of preoperative durvalumab (D) with or without tremelimumab (T) before resection, followed by postoperative (chemo)radiotherapy based on multidisciplinary discretion and 1-year D treatment. Artificial intelligence (AI)-powered spatial distribution analysis of tumor-infiltrating lymphocytes and high-dimensional profiling of circulating immune cells tracked dynamic intratumoral and systemic immune responses. RESULTS Of the 48 patients enrolled (D, 24 patients; D+T, 24 patients), 45 underwent surgical resection per protocol (D, 21 patients; D+T, 24 patients). D±T had a favorable safety profile and did not delay surgery. Distant recurrence-free survival (DRFS) was significantly better in patients treated with D+T than in those treated with D monotherapy. AI-powered whole-slide image analysis demonstrated that D+T significantly reshaped the tumor microenvironment toward immune-inflamed phenotypes, in contrast with the D monotherapy or cytotoxic chemotherapy. High-dimensional profiling of circulating immune cells revealed a significant expansion of T-cell subsets characterized by proliferation and activation in response to D+T therapy, which was rare following D monotherapy. Importantly, expansion of specific clusters in CD8+ T cells and non-regulatory CD4+ T cells with activation and exhaustion programs was associated with prolonged DRFS in patients treated with D+T. CONCLUSIONS Preoperative D±T is feasible and may benefit patients with resectable HNSCC. Distinct changes in the tumor microenvironment and circulating immune cells were induced by each treatment regimen, warranting further investigation.
Collapse
Affiliation(s)
- Chang Gon Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min Hee Hong
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dahee Kim
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Brian Hyohyoung Lee
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyunwook Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | - Geoffrey Kelly
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yoon Ji Bang
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Gamin Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jung Eun Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chaeyeon Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Se-Heon Kim
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyun Jun Hong
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Min Park
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Nam Suk Sim
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Heejung Park
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin Woo Park
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chang Geol Lee
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyung Hwan Kim
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Goeun Park
- Division of Biostatistics, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Inkyung Jung
- Division of Biostatistics, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dawoon Han
- Department of Dermatology and Cutaneous Biology Research Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong Hoon Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Junha Cha
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Insuk Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | | | - Heon Song
- Lunit Inc., Seoul, Republic of Korea
| | | | | | | | | | - Seunghee Kim-Schulze
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Miriam Merad
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, New York
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sun Och Yoon
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyun Je Kim
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Republic of Korea
- Genome Medicine Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Seoul National University Hospital, Seoul, Republic of Korea
| | - Yoon Woo Koh
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hye Ryun Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
23
|
Starskaia I, Valta M, Pietilä S, Suomi T, Pahkuri S, Kalim UU, Rasool O, Rydgren E, Hyöty H, Knip M, Veijola R, Ilonen J, Toppari J, Lempainen J, Elo LL, Lahesmaa R. Distinct cellular immune responses in children en route to type 1 diabetes with different first-appearing autoantibodies. Nat Commun 2024; 15:3810. [PMID: 38714671 PMCID: PMC11076468 DOI: 10.1038/s41467-024-47918-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 04/12/2024] [Indexed: 05/10/2024] Open
Abstract
Previous studies have revealed heterogeneity in the progression to clinical type 1 diabetes in children who develop islet-specific antibodies either to insulin (IAA) or glutamic acid decarboxylase (GADA) as the first autoantibodies. Here, we test the hypothesis that children who later develop clinical disease have different early immune responses, depending on the type of the first autoantibody to appear (GADA-first or IAA-first). We use mass cytometry for deep immune profiling of peripheral blood mononuclear cell samples longitudinally collected from children who later progressed to clinical disease (IAA-first, GADA-first, ≥2 autoantibodies first groups) and matched for age, sex, and HLA controls who did not, as part of the Type 1 Diabetes Prediction and Prevention study. We identify differences in immune cell composition of children who later develop disease depending on the type of autoantibodies that appear first. Notably, we observe an increase in CD161 expression in natural killer cells of children with ≥2 autoantibodies and validate this in an independent cohort. The results highlight the importance of endotype-specific analyses and are likely to contribute to our understanding of pathogenic mechanisms underlying type 1 diabetes development.
Collapse
Grants
- 1-SRA-2016-342-M-R, 1-SRA-2019-732-M-B, 3-SRA-2020-955-S-B JDRF
- BMH4-CT98-3314 European Commission (EC)
- Academy of Finland (292538, 292335, 294337, 319280, 31444, 319280, 329277, 331790, 310561, 314443, 329278, 335434, 335611 and 341342), Novo Nordisk Foundation, Centre of Excellence in Molecular Systems Immunology and Physiology Research 2012-2017 [Decision No 250114]; Special Research Funds for University Hospitals in Finland; Diabetes Research Foundation, Finland; European Foundation for the Study of Diabetes; Päivikki and Sakari Sohlberg Foundation; Pediatric Research Foundation. Business Finland, the Sigrid Jusélius Foundation, Jane and Aatos Erkko Foundation, the Finnish Cancer Foundation, InFLAMES Flagship Programme of the Academy of Finland, Diabetes Wellness Suomi, the Finnish cultural foundation. the European Research Council ERC (677943), the Finnish Medical Foundation, the Finnish Pediatric Research Foundation and the Hospital Districht of South-West Finland.
Collapse
Affiliation(s)
- Inna Starskaia
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Turku Doctoral Programme of Molecular Medicine, University of Turku, Turku, Finland
| | - Milla Valta
- Turku Doctoral Programme of Molecular Medicine, University of Turku, Turku, Finland
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Sami Pietilä
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Tomi Suomi
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Sirpa Pahkuri
- Turku Doctoral Programme of Molecular Medicine, University of Turku, Turku, Finland
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Ubaid Ullah Kalim
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Omid Rasool
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Emilie Rydgren
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Heikki Hyöty
- Faculty of Medicine and Health Technology, Tampere University, and Fimlab Laboratories, Tampere, Finland
| | - Mikael Knip
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Center for Child Health Research, Tampere University Hospital, Tampere, Finland
| | - Riitta Veijola
- Department of Pediatrics, Research Unit of Clinical Medicine, Medical Research Centre, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jorma Toppari
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland
| | - Johanna Lempainen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland.
- Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland.
- Clinical Microbiology, Turku University Hospital, Turku, Finland.
| | - Laura L Elo
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.
- Institute of Biomedicine, University of Turku, Turku, Finland.
| | - Riitta Lahesmaa
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.
- Institute of Biomedicine, University of Turku, Turku, Finland.
| |
Collapse
|
24
|
Ning Z, Liu K, Zhang H, Dong G, Wang X, Xiong H. Platelets induce CD39 expression in tumor cells to facilitate tumor metastasis. Br J Cancer 2024; 130:1542-1551. [PMID: 38461171 PMCID: PMC11058827 DOI: 10.1038/s41416-024-02640-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 03/11/2024] Open
Abstract
BACKGROUND Tumor cells continue to evolve the metastatic potential in response to signals provided by the external microenvironment during metastasis. Platelets closely interact with tumor cells during hematogenous metastasis and facilitate tumor development. However, the molecular mechanisms underlying this process are not fully understood. METHODS RNA-sequencing was performed to screen differentially expressed genes mediated by platelets. The effects of platelet and CD39 on tumor metastasis were determined by experimental metastasis models with WT, NCG and CD39-/- mice. RESULTS RNA-sequencing results showed that platelets significantly up-regulated CD39 expression in tumor cells. CD39 is a novel immune checkpoint molecule and a key driver of immunosuppression. Our data provided evidence that the expression of CD39 was enhanced by platelets in a platelet-tumor cell contact dependent manner. Although the role of CD39 expressed by immune cells is well established, the effect of CD39 expressed by tumor cells on tumor cell behavior, anti-tumor immunity and tumor metastasis is unclear. We found that CD39 promoted tumor cell invasion, but had no effect on proliferation and migration. Notably, we showed that the ability of platelets to prime tumor cells for metastasis depends on CD39 in the experimental tumor metastasis model. CD39 silencing resulted in fewer experimental metastasis formation, and this anti-metastasis effect was significantly reduced in platelet-depleted mice. Furthermore, overexpression of CD39 in tumor cells promoted metastasis. In order to eliminate the effect of CD39 expressed in cells other than tumor cells, we detected tumor metastasis in CD39-/- mice and obtained similar results. Moreover, overexpression of CD39 in tumor cells inhibited antitumor immunity. Finally, the data from human samples also supported our findings. CONCLUSIONS Our study shows that direct contact with platelets induces CD39 expression in tumor cells, leading to immune suppression and promotion of metastasis.
Collapse
Affiliation(s)
- Zhaochen Ning
- Jining Key Laboratory of Immunology, Jining Medical University, Jining, 272067, China
| | - Keyan Liu
- Department of Public Health, Jining Medical University, Jining, 272067, China
| | - Hui Zhang
- Jining Key Laboratory of Immunology, Jining Medical University, Jining, 272067, China
| | - Guanjun Dong
- Jining Key Laboratory of Immunology, Jining Medical University, Jining, 272067, China
| | - Xiaotong Wang
- Jining Key Laboratory of Immunology, Jining Medical University, Jining, 272067, China
| | - Huabao Xiong
- Jining Key Laboratory of Immunology, Jining Medical University, Jining, 272067, China.
| |
Collapse
|
25
|
Chauhan SK, Dunn C, Andresen NK, Røssevold AH, Skorstad G, Sike A, Gilje B, Raj SX, Huse K, Naume B, Kyte JA. Peripheral immune cells in metastatic breast cancer patients display a systemic immunosuppressed signature consistent with chronic inflammation. NPJ Breast Cancer 2024; 10:30. [PMID: 38653982 DOI: 10.1038/s41523-024-00638-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 04/13/2024] [Indexed: 04/25/2024] Open
Abstract
Immunotherapies blocking the PD-1/PD-L1 checkpoint show some efficacy in metastatic breast cancer (mBC) but are often hindered by immunosuppressive mechanisms. Understanding these mechanisms is crucial for personalized treatments, with peripheral blood monitoring representing a practical alternative to repeated biopsies. In the present study, we performed a comprehensive mass cytometry analysis of peripheral blood immune cells in 104 patients with HER2 negative mBC and 20 healthy donors (HD). We found that mBC patients had significantly elevated monocyte levels and reduced levels of CD4+ T cells and plasmacytoid dendritic cells, when compared to HD. Furthermore, mBC patients had more effector T cells and regulatory T cells, increased expression of immune checkpoints and other activation/exhaustion markers, and a shift to a Th2/Th17 phenotype. Furthermore, T-cell phenotypes identified by mass cytometry correlated with functionality as assessed by IFN-γ production. Additional analysis indicated that previous chemotherapy and CDK4/6 inhibition impacted the numbers and phenotype of immune cells. From 63 of the patients, fresh tumor samples were analyzed by flow cytometry. Paired PBMC-tumor analysis showed moderate correlations between peripheral CD4+ T and NK cells with their counterparts in tumors. Further, a CD4+ T cell cluster in PBMCs, that co-expressed multiple checkpoint receptors, was negatively associated with CD4+ T cell tumor infiltration. In conclusion, the identified systemic immune signatures indicate an immune-suppressed environment in mBC patients who had progressed/relapsed on standard treatments, and is consistent with ongoing chronic inflammation. These activated immuno-suppressive mechanisms may be investigated as therapeutic targets, and for use as biomarkers of response or treatment resistance.
Collapse
Affiliation(s)
- Sudhir Kumar Chauhan
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Claire Dunn
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Nikolai Kragøe Andresen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Andreas Hagen Røssevold
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gjertrud Skorstad
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Adam Sike
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Bjørnar Gilje
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Sunil Xavier Raj
- Department of Oncology, St Olav University Hospital, Trondheim, Norway
| | - Kanutte Huse
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Bjørn Naume
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Jon Amund Kyte
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway.
- Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway.
| |
Collapse
|
26
|
Vaughan J, Patel M, Suchard M, Gededzha M, Ranchod H, Howard W, Snyman T, Wiggill T. Derangements of immunological proteins in HIV-associated diffuse large B-cell lymphoma: the frequency and prognostic impact. Front Cell Infect Microbiol 2024; 14:1340096. [PMID: 38633747 PMCID: PMC11021765 DOI: 10.3389/fcimb.2024.1340096] [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: 11/17/2023] [Accepted: 03/12/2024] [Indexed: 04/19/2024] Open
Abstract
Introduction Diffuse large B-cell lymphoma (DLBCL) is an aggressive malignancy of B-cells frequently encountered among people living with HIV. Immunological abnormalities are common in immunocompetent individuals with DLBCL, and are often associated with poorer outcomes. Currently, data on derangements of immunological proteins, such as cytokines and acute phase reactants, and their impact on outcomes in HIV-associated DLBCL (HIV-DLBCL) is lacking. This study assessed the levels and prognostic relevance of interleukin (IL)-6, IL-10 and Transforming Growth Factor Beta (TGFβ), the acute phase proteins C-reactive protein (CRP) and ferritin; serum free light chains (SFLC) (elevation of which reflects a prolonged pro-inflammatory state); and the activity of the immunosuppressive enzyme Indoleamine 2,3-dioxygenase (IDO)in South African patients with DLBCL. Methods Seventy-six patients with incident DLBCL were enrolled, and peripheral blood IL-6, IL-10, TGFβ, SFLC and IDO-activity measured in selected patients. Additional clinical and laboratory findings (including ferritin and CRP) were recorded from the hospital records. Results Sixty-one (80.3%) of the included patients were people living with HIV (median CD4-count = 148 cells/ul), and survival rates were poor (12-month survival rate 30.0%). The majority of the immunological proteins, except for TGFβ and ferritin, were significantly higher among the people living with HIV. Elevation of IL-6, SFLC and IDO-activity were not associated with survival in HIV-DLBCL, while raised IL-10, CRP, ferritin and TGFβ were. On multivariate analysis, immunological proteins associated with survival independently from the International Prognostic Index (IPI) included TGFβ, ferritin and IL-10. Conclusion Derangements of immunological proteins are common in HIV-DLBCL, and have a differential association with survival compared to that reported elsewhere. Elevation of TGFβ, IL-10 and ferritin were associated with survival independently from the IPI. In view of the poor survival rates in this cohort, investigation of the directed targeting of these cytokines would be of interest in our setting.
Collapse
Affiliation(s)
- Jenifer Vaughan
- Department of Molecular Medicine and Haematology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- National Health Laboratory Services, Johannesburg, South Africa
| | - Moosa Patel
- Department of Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Clinical Haematology Unit, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa
| | - Melinda Suchard
- Department of Chemical Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Maemu Gededzha
- National Health Laboratory Services, Johannesburg, South Africa
- Department of Immunology, University of the Witwatersrand, Johannesburg, South Africa
| | - Heena Ranchod
- Department of Chemical Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases, Centre for Vaccines and Immunology, Johannesburg, South Africa
| | - Wayne Howard
- Department of Chemical Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases, Centre for Vaccines and Immunology, Johannesburg, South Africa
| | - Tracy Snyman
- National Health Laboratory Services, Johannesburg, South Africa
| | - Tracey Wiggill
- Department of Molecular Medicine and Haematology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- National Health Laboratory Services, Johannesburg, South Africa
| |
Collapse
|
27
|
Dold L, Kalthoff S, Frank L, Zhou T, Esser P, Lutz P, Strassburg CP, Spengler U, Langhans B. STAT activation in regulatory CD4 + T cells of patients with primary sclerosing cholangitis. Immun Inflamm Dis 2024; 12:e1248. [PMID: 38607233 PMCID: PMC11010953 DOI: 10.1002/iid3.1248] [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: 12/20/2023] [Revised: 03/25/2024] [Accepted: 03/31/2024] [Indexed: 04/13/2024] Open
Abstract
INTRODUCTION Regulatory CD4+ T cells (Tregs) are pivotal for inhibition of autoimmunity. Primary sclerosing cholangitis (PSC) is an autoimmune cholestatic liver disease of unknown etiology where contribution of Tregs is still unclear. Activation of the JAK-STAT pathway critically modifies functions of Tregs. In PSC, we studied activation of STAT proteins and Treg functions in response to cytokines. METHODS In 51 patients with PSC, 10 disease controls (chronic replicative hepatitis C), and 36 healthy controls we analyzed frequencies of Foxp3+CD25+CD127lowCD4+ Tregs, their expression of ectonucleotidase CD39, and cytokine-induced phosphorylation of STAT1, 3, 5, and 6 using phospho-flow cytometry. In parallel, we measured cytokines IFN-gamma, interleukin (IL)-6, IL-2, and IL-4 in serum via bead-based immunoassays. RESULTS In patients with PSC, ex vivo frequencies of peripheral Tregs and their expression of CD39 were significantly reduced (p < .05 each). Furthermore, serum levels of IFN-gamma, IL-6, IL-2, and IL-4 were markedly higher in PSC (p < .05 each). Unlike activation of STAT1, STAT5, and STAT6, IL-6 induced increased phosphorylation of STAT3 in Tregs of PSC-patients (p = .0434). Finally, STAT3 activation in Tregs correlated with leukocyte counts. CONCLUSIONS In PSC, we observed enhanced STAT3 responsiveness of CD4+ Tregs together with reduced CD39 expression probably reflecting inflammatory activity of the disease.
Collapse
Affiliation(s)
- Leona Dold
- Department of Internal Medicine IUniversity Hospital of BonnBonnGermany
- German Center for Infection Research (DZIF)Partner Site Cologne‐BonnBonnGermany
| | - Sandra Kalthoff
- Department of Internal Medicine IUniversity Hospital of BonnBonnGermany
| | - Leonie Frank
- Department of Internal Medicine IUniversity Hospital of BonnBonnGermany
| | - Taotao Zhou
- Department of Internal Medicine IUniversity Hospital of BonnBonnGermany
| | - Pia Esser
- Department of Internal Medicine IUniversity Hospital of BonnBonnGermany
| | - Philipp Lutz
- Department of Internal Medicine IUniversity Hospital of BonnBonnGermany
| | | | - Ulrich Spengler
- Department of Internal Medicine IUniversity Hospital of BonnBonnGermany
| | - Bettina Langhans
- Department of Internal Medicine IUniversity Hospital of BonnBonnGermany
- German Center for Infection Research (DZIF)Partner Site Cologne‐BonnBonnGermany
| |
Collapse
|
28
|
Araujo Furlan CL, Boccardo S, Rodriguez C, Mary VS, Gimenez CMS, Robson SC, Gruppi A, Montes CL, Acosta Rodríguez EV. CD39 expression by regulatory T cells participates in CD8+ T cell suppression during experimental Trypanosoma cruzi infection. PLoS Pathog 2024; 20:e1012191. [PMID: 38683845 PMCID: PMC11081507 DOI: 10.1371/journal.ppat.1012191] [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: 09/22/2023] [Revised: 05/09/2024] [Accepted: 04/12/2024] [Indexed: 05/02/2024] Open
Abstract
An imbalance between suppressor and effector immune responses may preclude cure in chronic parasitic diseases. In the case of Trypanosoma cruzi infection, specialized regulatory Foxp3+ T (Treg) cells suppress protective type-1 effector responses. Herein, we investigated the kinetics and underlying mechanisms behind the regulation of protective parasite-specific CD8+ T cell immunity during acute T. cruzi infection. Using the DEREG mouse model, we found that Treg cells play a role during the initial stages after T. cruzi infection, restraining the magnitude of CD8+ T cell responses and parasite control. Early Treg cell depletion increased the frequencies of polyfunctional short-lived, effector T cell subsets, without affecting memory precursor cell formation or the expression of activation, exhaustion and functional markers. In addition, Treg cell depletion during early infection minimally affected the antigen-presenting cell response but it boosted CD4+ T cell responses before the development of anti-parasite effector CD8+ T cell immunity. Crucially, the absence of CD39 expression on Treg cells significantly bolstered effector parasite-specific CD8+ T cell responses, preventing increased parasite replication in T. cruzi infected mice adoptively transferred with Treg cells. Our work underscores the crucial role of Treg cells in regulating protective anti-parasite immunity and provides evidence that CD39 expression by Treg cells represents a key immunomodulatory mechanism in this infection model.
Collapse
Affiliation(s)
- Cintia L. Araujo Furlan
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, CONICET, Córdoba, Argentina
| | - Santiago Boccardo
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, CONICET, Córdoba, Argentina
| | - Constanza Rodriguez
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, CONICET, Córdoba, Argentina
| | - Verónica S. Mary
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, CONICET, Córdoba, Argentina
| | - Camila M. S. Gimenez
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, CONICET, Córdoba, Argentina
| | - Simon C. Robson
- Center for Inflammation Research, Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Adriana Gruppi
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, CONICET, Córdoba, Argentina
| | - Carolina L. Montes
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, CONICET, Córdoba, Argentina
| | - Eva V. Acosta Rodríguez
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, CONICET, Córdoba, Argentina
| |
Collapse
|
29
|
Cochrane RW, Robino RA, Granger B, Allen E, Vaena S, Romeo MJ, de Cubas AA, Berto S, Ferreira LM. High affinity chimeric antigen receptor signaling induces an inflammatory program in human regulatory T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.31.587467. [PMID: 38617240 PMCID: PMC11014479 DOI: 10.1101/2024.03.31.587467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Regulatory T cells (Tregs) are promising cellular therapies to induce immune tolerance in organ transplantation and autoimmune disease. The success of chimeric antigen receptor (CAR) T-cell therapy for cancer has sparked interest in using CARs to generate antigen-specific Tregs. Here, we compared CAR with endogenous T cell receptor (TCR)/CD28 activation in human Tregs. Strikingly, CAR Tregs displayed increased cytotoxicity and diminished suppression of antigen-presenting cells and effector T (Teff) cells compared with TCR/CD28 activated Tregs. RNA sequencing revealed that CAR Tregs activate Teff cell gene programs. Indeed, CAR Tregs secreted high levels of inflammatory cytokines, with a subset of FOXP3+ CAR Tregs uniquely acquiring CD40L surface expression and producing IFNγ. Interestingly, decreasing CAR antigen affinity reduced Teff cell gene expression and inflammatory cytokine production by CAR Tregs. Our findings showcase the impact of engineered receptor activation on Treg biology and support tailoring CAR constructs to Tregs for maximal therapeutic efficacy.
Collapse
Affiliation(s)
- Russell W. Cochrane
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Rob A. Robino
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Bryan Granger
- Bioinformatics Core, Medical University of South Carolina, Charleston, SC, USA
| | - Eva Allen
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Silvia Vaena
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Martin J. Romeo
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Aguirre A. de Cubas
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Stefano Berto
- Bioinformatics Core, Medical University of South Carolina, Charleston, SC, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Leonardo M.R. Ferreira
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
30
|
Xie Z, Huang J, Sun G, He S, Luo Z, Zhang L, Li L, Yao M, Du C, Yu W, Feng Y, Yang D, Zhang J, Ge C, Li H, Geng M. Integrated multi-omics analysis reveals gut microbiota dysbiosis and systemic disturbance in major depressive disorder. Psychiatry Res 2024; 334:115804. [PMID: 38417224 DOI: 10.1016/j.psychres.2024.115804] [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: 08/11/2023] [Revised: 11/18/2023] [Accepted: 02/17/2024] [Indexed: 03/01/2024]
Abstract
Major depressive disorder (MDD) involves systemic changes in peripheral blood and gut microbiota, but the current understanding is incomplete. Herein, we conducted a multi-omics analysis of fecal and blood samples obtained from an observational cohort including MDD patients (n = 99) and healthy control (HC, n = 50). 16S rRNA sequencing of gut microbiota showed structural alterations in MDD, as characterized by increased Enterococcus. Metagenomics sequencing of gut microbiota showed substantial functional alterations including upregulation in the superpathway of the glyoxylate cycle and fatty acid degradation and downregulation in various metabolic pathways in MDD. Plasma metabolomics revealed decreased amino acids and bile acids, together with increased sphingolipids and cholesterol esters in MDD. Notably, metabolites involved in arginine and proline metabolism were decreased while sphingolipid metabolic pathway were increased. Mass cytometry analysis of blood immune cell subtypes showed rises in proinflammatory immune subsets and declines in anti-inflammatory immune subsets in MDD. Furthermore, our findings revealed disease severity-related factors of MDD. Interestingly, we classified MDD into two immune subtypes that were highly correlated with disease relapse. Moreover, we established discriminative signatures that differentiate MDD from HC. These findings contribute to a comprehensive understanding of the MDD pathogenesis and provide valuable resources for the discovery of biomarkers.
Collapse
Affiliation(s)
- Zuoquan Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jingjing Huang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Guangqiang Sun
- Green Valley (shanghai) pharmaceutical technology Co., Ltd., Shanghai 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Shen He
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Zhiyu Luo
- Green Valley (shanghai) pharmaceutical technology Co., Ltd., Shanghai 201203, China
| | - Linna Zhang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Liang Li
- Green Valley (shanghai) pharmaceutical technology Co., Ltd., Shanghai 201203, China
| | - Min Yao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Chen Du
- Green Valley (shanghai) pharmaceutical technology Co., Ltd., Shanghai 201203, China
| | - Wenjuan Yu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Yuan Feng
- Green Valley (shanghai) pharmaceutical technology Co., Ltd., Shanghai 201203, China
| | - Dabing Yang
- Green Valley (shanghai) pharmaceutical technology Co., Ltd., Shanghai 201203, China
| | - Jing Zhang
- Green Valley (shanghai) pharmaceutical technology Co., Ltd., Shanghai 201203, China
| | - Changrong Ge
- Green Valley (shanghai) pharmaceutical technology Co., Ltd., Shanghai 201203, China
| | - Huafang Li
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| | - Meiyu Geng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China.
| |
Collapse
|
31
|
Sligar C, Reilly E, Cuthbertson P, Vine KL, Bird KM, Elhage A, Alexander SI, Sluyter R, Watson D. Graft-versus-leukaemia immunity is retained following treatment with post-transplant cyclophosphamide alone or combined with tocilizumab in humanised mice. Clin Transl Immunology 2024; 13:e1497. [PMID: 38495918 PMCID: PMC10941522 DOI: 10.1002/cti2.1497] [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: 11/21/2023] [Revised: 02/06/2024] [Accepted: 03/01/2024] [Indexed: 03/19/2024] Open
Abstract
Objectives Donor haematopoietic stem cell transplantation treats leukaemia by inducing graft-versus-leukaemia (GVL) immunity. However, this benefit is often mitigated by graft-versus-host disease (GVHD), which is reduced by post-transplant cyclophosphamide (PTCy) alone or combined with tocilizumab (TOC) in humanised mice. This study established a preclinical humanised mouse model of GVL and investigated whether PTCy alone or combined with TOC impacts GVL immunity. Methods NOD-scid-IL2Rγnull mice were injected with 2 × 107 human peripheral blood mononuclear cells (hPBMCs) on day 0 and with 1 × 106 THP-1 acute myeloid leukaemia cells on day 14. In subsequent experiments, mice were also injected with PTCy (33 mg kg-1) or Dulbecco's phosphate buffered saline (PBS) on days 3 and 4, alone or combined with TOC or control antibody (25 mg kg-1) twice weekly for 28 days. Clinical signs of disease were monitored until day 42. Results Mice with hPBMCs from three different donors and THP-1 cells showed similar survival, clinical score and weight loss. hCD33+ leukaemia cells were minimal in mice reconstituted with hPBMCs from two donors but present in mice with hPBMCs from a third donor, suggesting donor-specific GVL responses. hPBMC-injected mice treated with PTCy alone or combined with TOC (PTCy + TOC) demonstrated prolonged survival compared to control mice. PTCy alone and PTCy + TOC-treated mice with hPBMCs showed minimal hepatic hCD33+ leukaemia cells, indicating sustained GVL immunity. Further, the combination of PTCy + TOC reduced histological damage in the lung and liver. Conclusion Collectively, this research demonstrates that PTCy alone or combined with TOC impairs GVHD without compromising GVL immunity.
Collapse
Affiliation(s)
- Chloe Sligar
- Molecular Horizons and School of Chemistry and Molecular BioscienceUniversity of WollongongWollongongNSWAustralia
- Illawarra Health and Medical Research InstituteWollongongNSWAustralia
| | - Ellie Reilly
- Molecular Horizons and School of Chemistry and Molecular BioscienceUniversity of WollongongWollongongNSWAustralia
- Illawarra Health and Medical Research InstituteWollongongNSWAustralia
| | - Peter Cuthbertson
- Molecular Horizons and School of Chemistry and Molecular BioscienceUniversity of WollongongWollongongNSWAustralia
- Illawarra Health and Medical Research InstituteWollongongNSWAustralia
| | - Kara L Vine
- Molecular Horizons and School of Chemistry and Molecular BioscienceUniversity of WollongongWollongongNSWAustralia
- Illawarra Health and Medical Research InstituteWollongongNSWAustralia
| | - Katrina M Bird
- Molecular Horizons and School of Chemistry and Molecular BioscienceUniversity of WollongongWollongongNSWAustralia
- Illawarra Health and Medical Research InstituteWollongongNSWAustralia
| | - Amal Elhage
- Molecular Horizons and School of Chemistry and Molecular BioscienceUniversity of WollongongWollongongNSWAustralia
- Illawarra Health and Medical Research InstituteWollongongNSWAustralia
| | | | - Ronald Sluyter
- Molecular Horizons and School of Chemistry and Molecular BioscienceUniversity of WollongongWollongongNSWAustralia
- Illawarra Health and Medical Research InstituteWollongongNSWAustralia
| | - Debbie Watson
- Molecular Horizons and School of Chemistry and Molecular BioscienceUniversity of WollongongWollongongNSWAustralia
- Illawarra Health and Medical Research InstituteWollongongNSWAustralia
| |
Collapse
|
32
|
de Silva TA, Apte S, Voisey J, Spann K, Tan M, Chambers D, O'Sullivan B. Immunological Landscapes in Lung Transplantation: Insights from T Cell Profiling in BAL and PBMC. Int J Mol Sci 2024; 25:2476. [PMID: 38473722 DOI: 10.3390/ijms25052476] [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: 11/03/2023] [Revised: 02/05/2024] [Accepted: 02/15/2024] [Indexed: 03/14/2024] Open
Abstract
Lung transplant recipients frequently encounter immune-related complications, including chronic lung allograft dysfunction (CLAD). Monitoring immune cells within the lung microenvironment is pivotal for optimizing post-transplant outcomes. This study examined the proportion of T cell subsets in paired bronchoalveolar lavage (BAL) and peripheral PBMC comparing healthy (n = 4) and lung transplantation patients (n = 6, no CLAD and n = 14 CLAD) using 14-color flow cytometry. CD4+ T cell proportions were reduced in CD3 cells in both PBMC and BAL, and positive correlations were discerned between T cell populations in peripheral PBMC and BAL, suggesting the prospect of employing less invasive PBMC sampling as a means of monitoring lung T cells. Furthermore, regulatory T cells (Tregs) were enriched in BAL when compared to peripheral PBMC for transplant recipients. A parallel positive correlation emerged between Treg proportions in BAL and peripheral PBMC, underscoring potential avenues for monitoring lung Tregs. Finally, the most promising biomarker was the Teff (CD8+Granzyme B+)-Treg ratio, which was higher in both the PBMC and BAL of transplant recipients compared to healthy individuals, and increased in the patients with CLAD compared to no CLAD and healthy patients. Conclusions: Distinct T cell profiles in BAL and peripheral PBMC underscore the significance of localized immune monitoring in lung transplantation. The Teff (CD8+granzyme B+)-Treg ratio, particularly within the context of CLAD, emerges as a promising blood and BAL biomarker reflective of inflammation and transplant-related complications. These findings emphasize the imperative need for personalized immune monitoring strategies that tailored to address the unique immunological milieu in post-transplant lungs.
Collapse
Affiliation(s)
- Tharushi Ayanthika de Silva
- Centre for Genomics and Personalised Health, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4001, Australia
- Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Brisbane, QLD 4001, Australia
| | - Simon Apte
- Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Brisbane, QLD 4001, Australia
- Facility of Clinical Medicine, The University of Queensland, Brisbane, QLD 4001, Australia
| | - Joanne Voisey
- Centre for Genomics and Personalised Health, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4001, Australia
| | - Kirsten Spann
- Centre for Immunology and Infection Control, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4001, Australia
| | - Maxine Tan
- Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Brisbane, QLD 4001, Australia
- Facility of Clinical Medicine, The University of Queensland, Brisbane, QLD 4001, Australia
| | - Daniel Chambers
- Centre for Genomics and Personalised Health, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4001, Australia
- Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Brisbane, QLD 4001, Australia
- Facility of Clinical Medicine, The University of Queensland, Brisbane, QLD 4001, Australia
| | - Brendan O'Sullivan
- Centre for Genomics and Personalised Health, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4001, Australia
- Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Brisbane, QLD 4001, Australia
- Facility of Clinical Medicine, The University of Queensland, Brisbane, QLD 4001, Australia
| |
Collapse
|
33
|
Lee BH, Bang YJ, Lim SH, Kang SJ, Kim SH, Kim-Schulze S, Park CG, Kim HJ, Kim TG. High-dimensional profiling of regulatory T cells in psoriasis reveals an impaired skin-trafficking property. EBioMedicine 2024; 100:104985. [PMID: 38306895 PMCID: PMC10847473 DOI: 10.1016/j.ebiom.2024.104985] [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: 11/14/2022] [Revised: 12/21/2023] [Accepted: 01/13/2024] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Psoriasis is a chronic inflammatory skin disease with a Th17-skewed immune phenotype. Although it has been generally accepted that regulatory T cells (Tregs) in lesional psoriatic skin have functional impairment due to the local inflammatory microenvironment, the molecular properties of skin-homing psoriatic Tregs have not been well explored. METHODS We designed an extensive 39 marker mass cytometry (CyTOF) panel to deeply profile the immune landscape of skin-homing Tregs from 31 people with psoriasis stratified by psoriasis area severity index score as mild (n = 15) to moderate-severe (n = 16) and 32 healthy controls. We further validated the findings with an in-vitro chemokine-mediated Treg migration assay, immunofluorescent imaging of normal and psoriatic lesional skin and analysed public single-cell RNA-sequencing datasets to expand upon our findings into the local tissue microenvironments. FINDINGS We discovered an overall decrease in CLAhi Tregs and specifically, CLAhiCCR5+ Tregs in psoriasis. Functional markers CD39 and FoxP3 were elevated in psoriatic Tregs. However, CCR7 expression was significantly increased while CCR4 and CLA expression was reduced in psoriatic Tregs and CLAhi Tregs, which was associated with disease severity. Moreover, psoriatic Tregs revealed increased migratory capacity towards CCR7's ligands, CCL19/CCL21. Interrogation of public single-cell RNA sequencing data confirmed reduced expression of skin-trafficking markers in lesional-skin Tregs compared to non-lesioned skin, further substantiated by immunofluorescent staining. INTERPRETATION Psoriatic circulating Tregs showed an impaired skin-trafficking phenotype thus leading to insufficient suppression of ongoing inflammation in the lesional skin, expanding upon our current understanding of the impairment of Treg-mediated immunosuppression in psoriasis. FUNDING This research was supported by the Basic Science Research Program through the National Research Foundation of Korea funded by the Ministry of Science and Information and Communications Technology (2020R1C1C1014513, 2021R1A4A5032185, 2020R1F1A1073692); and the new faculty research seed money grant of Yonsei University College of Medicine for 2021 (2021-32-0033).
Collapse
Affiliation(s)
- Brian Hyohyoung Lee
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yoon Ji Bang
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea
| | - Sung Ha Lim
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Seong-Jun Kang
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea; Cancer Research Institute, Seoul National University, Seoul, South Korea
| | - Sung Hee Kim
- Department of Dermatology, Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Seunghee Kim-Schulze
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chung-Gyu Park
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea; Cancer Research Institute, Seoul National University, Seoul, South Korea; Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea; Institute of Endemic Diseases, Seoul National University Medical Research Center, Seoul, South Korea; Transplantation Research Institute, Seoul National University Medical Research Center, Seoul, South Korea; Seoul National University Hospital, Seoul, South Korea.
| | - Hyun Je Kim
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea; Cancer Research Institute, Seoul National University, Seoul, South Korea; Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea; Institute of Endemic Diseases, Seoul National University Medical Research Center, Seoul, South Korea; Seoul National University Hospital, Seoul, South Korea; Genome Medicine Institute, Seoul National University Medical Research Center, Seoul, South Korea.
| | - Tae-Gyun Kim
- Department of Dermatology, Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, South Korea; Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
34
|
Santosh Nirmala S, Kayani K, Gliwiński M, Hu Y, Iwaszkiewicz-Grześ D, Piotrowska-Mieczkowska M, Sakowska J, Tomaszewicz M, Marín Morales JM, Lakshmi K, Marek-Trzonkowska NM, Trzonkowski P, Oo YH, Fuchs A. Beyond FOXP3: a 20-year journey unravelling human regulatory T-cell heterogeneity. Front Immunol 2024; 14:1321228. [PMID: 38283365 PMCID: PMC10811018 DOI: 10.3389/fimmu.2023.1321228] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/19/2023] [Indexed: 01/30/2024] Open
Abstract
The initial idea of a distinct group of T-cells responsible for suppressing immune responses was first postulated half a century ago. However, it is only in the last three decades that we have identified what we now term regulatory T-cells (Tregs), and subsequently elucidated and crystallized our understanding of them. Human Tregs have emerged as essential to immune tolerance and the prevention of autoimmune diseases and are typically contemporaneously characterized by their CD3+CD4+CD25high CD127lowFOXP3+ phenotype. It is important to note that FOXP3+ Tregs exhibit substantial diversity in their origin, phenotypic characteristics, and function. Identifying reliable markers is crucial to the accurate identification, quantification, and assessment of Tregs in health and disease, as well as the enrichment and expansion of viable cells for adoptive cell therapy. In our comprehensive review, we address the contributions of various markers identified in the last two decades since the master transcriptional factor FOXP3 was identified in establishing and enriching purity, lineage stability, tissue homing and suppressive proficiency in CD4+ Tregs. Additionally, our review delves into recent breakthroughs in innovative Treg-based therapies, underscoring the significance of distinct markers in their therapeutic utilization. Understanding Treg subsets holds the key to effectively harnessing human Tregs for immunotherapeutic approaches.
Collapse
Affiliation(s)
| | - Kayani Kayani
- Centre for Liver and Gastrointestinal Research and National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Department of Academic Surgery, Queen Elizabeth Hospital, University of Birmingham, Birmingham, United Kingdom
- Department of Renal Surgery, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Mateusz Gliwiński
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | - Yueyuan Hu
- Center for Regenerative Therapies Dresden, Technical University Dresden, Dresden, Germany
| | | | | | - Justyna Sakowska
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | - Martyna Tomaszewicz
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Kavitha Lakshmi
- Center for Regenerative Therapies Dresden, Technical University Dresden, Dresden, Germany
| | | | - Piotr Trzonkowski
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | - Ye Htun Oo
- Centre for Liver and Gastrointestinal Research and National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Liver Transplant and Hepatobiliary Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
- Birmingham Advanced Cellular Therapy Facility, University of Birmingham, Birmingham, United Kingdom
- Centre for Rare Diseases, European Reference Network - Rare Liver Centre, Birmingham, United Kingdom
| | - Anke Fuchs
- Center for Regenerative Therapies Dresden, Technical University Dresden, Dresden, Germany
| |
Collapse
|
35
|
Wang B, Zhou A, Pan Q, Li Y, Xi Z, He K, Li D, Li B, Liu Y, Liu Y, Xia Q. Adenosinergic metabolism pathway: an emerging target for improving outcomes of solid organ transplantation. Transl Res 2024; 263:93-101. [PMID: 37678756 DOI: 10.1016/j.trsl.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/25/2023] [Accepted: 09/01/2023] [Indexed: 09/09/2023]
Abstract
Extracellular nucleotides are widely recognized as crucial modulators of immune responses in peripheral tissues. Adenosine triphosphate (ATP) and adenosine are key components of extracellular nucleotides, the balance of which contributes to immune homeostasis. Under tissue injury, ATP exerts its pro-inflammatory function, while the adenosinergic pathway rapidly degrades ATP to immunosuppressive adenosine, thus inhibiting excessive and uncontrolled inflammatory responses. Previous reviews have explored the immunoregulatory role of extracellular adenosine in various pathological conditions, especially inflammation and malignancy. However, current knowledge regarding adenosine and adenosinergic metabolism in the context of solid organ transplantation remains fragmented. In this review, we summarize the latest information on adenosine metabolism and the mechanisms by which it suppresses the effector function of immune cells, as well as highlight the protective role of adenosine in all stages of solid organ transplantation, including reducing ischemia reperfusion injury during organ procurement, alleviating rejection, and promoting graft regeneration after transplantation. Finally, we discuss the potential for future clinical translation of adenosinergic pathway in solid organ transplantation.
Collapse
Affiliation(s)
- Bingran Wang
- Department of liver surgery, Renji Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China; Shanghai Institute of Transplantation, Shanghai, China
| | - Aiwei Zhou
- Department of liver surgery, Renji Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China; Shanghai Institute of Transplantation, Shanghai, China
| | - Qi Pan
- Department of liver surgery, Renji Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China; Shanghai Institute of Transplantation, Shanghai, China
| | - Yanran Li
- Department of liver surgery, Renji Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China; Shanghai Institute of Transplantation, Shanghai, China
| | - Zhifeng Xi
- Department of liver surgery, Renji Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China; Shanghai Institute of Transplantation, Shanghai, China
| | - Kang He
- Department of liver surgery, Renji Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China; Shanghai Institute of Transplantation, Shanghai, China
| | - Dan Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongbo Liu
- Department of liver surgery, Renji Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China; Shanghai Institute of Transplantation, Shanghai, China
| | - Yuan Liu
- Department of liver surgery, Renji Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China; Shanghai Institute of Transplantation, Shanghai, China.
| | - Qiang Xia
- Department of liver surgery, Renji Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China; Shanghai Institute of Transplantation, Shanghai, China
| |
Collapse
|
36
|
Elsaghir A, El-Sabaa EMW, Zahran AM, Mandour SA, Salama EH, Aboulfotuh S, El-Morshedy RM, Tocci S, Mandour AM, Ali WE, Abdel-Wahid L, Sayed IM, El-Mokhtar MA. Elevated CD39+T-Regulatory Cells and Reduced Levels of Adenosine Indicate a Role for Tolerogenic Signals in the Progression from Moderate to Severe COVID-19. Int J Mol Sci 2023; 24:17614. [PMID: 38139439 PMCID: PMC10744088 DOI: 10.3390/ijms242417614] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Viral infections trigger inflammation by controlling ATP release. CD39 ectoenzymes hydrolyze ATP/ADP to AMP, which is converted by CD73 into anti-inflammatory adenosine (ADO). ADO is an anti-inflammatory and immunosuppressant molecule which can enhance viral persistence and severity. The CD39-CD73-adenosine axis contributes to the immunosuppressive T-reg microenvironment and may affect COVID-19 disease progression. Here, we investigated the link between CD39 expression, mostly on T-regs, and levels of CD73, adenosine, and adenosine receptors with COVID-19 severity and progression. Our study included 73 hospitalized COVID-19 patients, of which 33 were moderately affected and 40 suffered from severe infection. A flow cytometric analysis was used to analyze the frequency of T-regulatory cells (T-regs), CD39+ T-regs, and CD39+CD4+ T-cells. Plasma concentrations of adenosine, IL-10, and TGF-β were quantified via an ELISA. An RT-qPCR was used to analyze the gene expression of CD73 and adenosine receptors (A1, A2A, A2B, and A3). T-reg cells were higher in COVID-19 patients compared to healthy controls (7.4 ± 0.79 vs. 2.4 ± 0.28; p < 0.0001). Patients also had a higher frequency of the CD39+ T-reg subset. In addition, patients who suffered from a severe form of the disease had higher CD39+ T-regs compared with moderately infected patients. CD39+CD4+ T cells were increased in patients compared to the control group. An analysis of serum adenosine levels showed a marked decrease in their levels in patients, particularly those suffering from severe illness. However, this was paralleled with a marked decline in the expression levels of CD73. IL-10 and TGF-β levels were higher in COVID-19; in addition, their values were also higher in the severe group. In conclusion, there are distinct immunological alterations in CD39+ lymphocyte subsets and a dysregulation in the adenosine signaling pathway in COVID-19 patients which may contribute to immune dysfunction and disease progression. Understanding these immunological alterations in the different immune cell subsets and adenosine signaling provides valuable insights into the pathogenesis of the disease and may contribute to the development of novel therapeutic approaches targeting specific immune mechanisms.
Collapse
Affiliation(s)
- Alaa Elsaghir
- Department of Microbiology & Immunology, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt
| | - Ehsan M. W. El-Sabaa
- Department of Microbiology & Immunology, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt
| | - Asmaa M. Zahran
- Department of Clinical Pathology, South Egypt Cancer Institute, Assiut University, Assiut 71515, Egypt
| | - Sahar A. Mandour
- Department of Microbiology and Immunology, Faculty of Pharmacy, Deraya University, Minia 11566, Egypt
| | - Eman H. Salama
- Department of Clinical Pathology, Faculty of Medicine, Sohag University, Sohag 82524, Egypt
| | - Sahar Aboulfotuh
- Department of Clinical Pathology, Faculty of Medicine, Sohag University, Sohag 82524, Egypt
| | - Reham M. El-Morshedy
- Department of Chest Diseases and Tuberculosis, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| | - Stefania Tocci
- Department of Biomedical & Nutritional Sciences, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - Ahmed Mohamed Mandour
- Department of Anesthesia and ICU, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| | - Wael Esmat Ali
- Department of Clinical Pathology, Faculty of Medicine, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| | - Lobna Abdel-Wahid
- Gastroenterology and Hepatology Unit, Internal Medicine Department, Assiut University, Assiut 71515, Egypt
| | - Ibrahim M. Sayed
- Department of Biomedical & Nutritional Sciences, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - Mohamed A. El-Mokhtar
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos P.O. Box 36, Lebanon
- Department of Medical Microbiology & Immunology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| |
Collapse
|
37
|
Vafadar A, Vosough P, Jahromi HK, Tajbakhsh A, Savardshtaki A, Butler AE, Sahebkar A. The role of efferocytosis and transplant rejection: Strategies in promoting transplantation tolerance using apoptotic cell therapy and/or synthetic particles. Cell Biochem Funct 2023; 41:959-977. [PMID: 37787641 DOI: 10.1002/cbf.3852] [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: 06/05/2023] [Revised: 07/26/2023] [Accepted: 08/24/2023] [Indexed: 10/04/2023]
Abstract
Recently, efforts have been made to recognize the precise reason(s) for transplant failure and the process of rejection utilizing the molecular signature. Most transplant recipients do not appreciate the unknown length of survival of allogeneic grafts with the existing standard of care. Two noteworthy immunological pathways occur during allogeneic transplant rejection. A nonspecific innate immune response predominates in the early stages of the immune reaction, and allogeneic antigens initiate a donor-specific adaptive reaction. Though the adaptive response is the major cause of allograft rejection, earlier pro-inflammatory responses that are part of the innate immune response are also regarded as significant in graft loss. The onset of the innate and adaptive immune response causes chronic and acute transplant rejection. Currently employed immunosuppressive medications have shown little or no influence on chronic rejection and, as a result, on overall long-term transplant survival. Furthermore, long-term pharmaceutical immunosuppression is associated with side effects, toxicity, and an increased risk of developing diseases, both infectious and metabolic. As a result, there is a need for the development of innovative donor-specific immunosuppressive medications to regulate the allorecognition pathways that induce graft loss and to reduce the side effects of immunosuppression. Efferocytosis is an immunomodulatory mechanism with fast and efficient clearance of apoptotic cells (ACs). As such, AC therapy strategies have been suggested to limit transplant-related sequelae. Efferocytosis-based medicines/treatments can also decrease the use of immunosuppressive drugs and have no detrimental side effects. Thus, this review aims to investigate the impact of efferocytosis on transplant rejection/tolerance and identify approaches using AC clearance to increase transplant viability.
Collapse
Affiliation(s)
- Asma Vafadar
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parisa Vosough
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Kargar Jahromi
- Research Center for Non-Communicable Disease, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Amir Tajbakhsh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardshtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland - Bahrain, Adliya, Bahrain
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
38
|
Bei KF, Moshkelgosha S, Liu BJ, Juvet S. Intragraft regulatory T cells in the modern era: what can high-dimensional methods tell us about pathways to allograft acceptance? Front Immunol 2023; 14:1291649. [PMID: 38077395 PMCID: PMC10701590 DOI: 10.3389/fimmu.2023.1291649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 10/31/2023] [Indexed: 12/18/2023] Open
Abstract
Replacement of diseased organs with transplanted healthy donor ones remains the best and often only treatment option for end-stage organ disease. Immunosuppressants have decreased the incidence of acute rejection, but long-term survival remains limited. The broad action of current immunosuppressive drugs results in global immune impairment, increasing the risk of cancer and infections. Hence, achievement of allograft tolerance, in which graft function is maintained in the absence of global immunosuppression, has long been the aim of transplant clinicians and scientists. Regulatory T cells (Treg) are a specialized subset of immune cells that control a diverse array of immune responses, can prevent allograft rejection in animals, and have recently been explored in early phase clinical trials as an adoptive cellular therapy in transplant recipients. It has been established that allograft residency by Tregs can promote graft acceptance, but whether intragraft Treg functional diversification and spatial organization contribute to this process is largely unknown. In this review, we will explore what is known regarding the properties of intragraft Tregs during allograft acceptance and rejection. We will summarize recent advances in understanding Treg tissue residency through spatial, transcriptomic and high-dimensional cytometric methods in both animal and human studies. Our discussion will explore properties of intragraft Tregs in mediating operational tolerance to commonly transplanted solid organs. Finally, given recent developments in Treg cellular therapy, we will review emerging knowledge of whether and how these adoptively transferred cells enter allografts in humans. An understanding of the properties of intragraft Tregs will help lay the foundation for future therapies that will promote immune tolerance.
Collapse
Affiliation(s)
- Ke Fan Bei
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Sajad Moshkelgosha
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Bo Jie Liu
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Stephen Juvet
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
39
|
Kott KA, Chan AS, Vernon ST, Hansen T, Kim T, de Dreu M, Gunasegaran B, Murphy AJ, Patrick E, Psaltis PJ, Grieve SM, Yang JY, Fazekas de St Groth B, McGuire HM, Figtree GA. Mass cytometry analysis reveals altered immune profiles in patients with coronary artery disease. Clin Transl Immunology 2023; 12:e1462. [PMID: 37927302 PMCID: PMC10621005 DOI: 10.1002/cti2.1462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 05/09/2023] [Accepted: 08/09/2023] [Indexed: 11/07/2023] Open
Abstract
Objective The importance of inflammation in atherosclerosis is well accepted, but the role of the adaptive immune system is not yet fully understood. To further explore this, we assessed the circulating immune cell profile of patients with coronary artery disease (CAD) to identify discriminatory features by mass cytometry. Methods Mass cytometry was performed on patient samples from the BioHEART-CT study, gated to detect 82 distinct cell subsets. CT coronary angiograms were analysed to categorise patients as having CAD (CAD+) or having normal coronary arteries (CAD-). Results The discovery cohort included 117 patients (mean age 61 ± 12 years, 49% female); 79 patients (68%) were CAD+. Mass cytometry identified changes in 15 T-cell subsets, with higher numbers of proliferating, highly differentiated and cytotoxic cells and decreases in naïve T cells. Five T-regulatory subsets were related to an age and gender-independent increase in the odds of CAD incidence when expressing CCR2 (OR 1.12), CCR4 (OR 1.08), CD38 and CD45RO (OR 1.13), HLA-DR (OR 1.06) and Ki67 (OR 1.22). Markers of proliferation and differentiation were also increased within B cells, while plasmacytoid dendritic cells were decreased. This combination of changes was assessed using SVM models in discovery and validation cohorts (area under the curve = 0.74 for both), confirming the robust nature of the immune signature detected. Conclusion We identified differences within immune subpopulations of CAD+ patients which are indicative of a systemic immune response to coronary atherosclerosis. This immune signature needs further study via incorporation into risk scoring tools for the precision diagnosis of CAD.
Collapse
Affiliation(s)
- Katharine A Kott
- Cardiothoracic and Vascular HealthKolling Institute of Medical ResearchSydneyNSWAustralia
- Department of Cardiology, Royal North Shore HospitalNorthern Sydney Local Health DistrictSydneyNSWAustralia
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
| | - Adam S Chan
- School of Mathematics and StatisticsUniversity of SydneySydneyNSWAustralia
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
| | - Stephen T Vernon
- Cardiothoracic and Vascular HealthKolling Institute of Medical ResearchSydneyNSWAustralia
- Department of Cardiology, Royal North Shore HospitalNorthern Sydney Local Health DistrictSydneyNSWAustralia
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
| | - Thomas Hansen
- Cardiothoracic and Vascular HealthKolling Institute of Medical ResearchSydneyNSWAustralia
| | - Taiyun Kim
- School of Mathematics and StatisticsUniversity of SydneySydneyNSWAustralia
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
| | - Macha de Dreu
- School of Medical Sciences, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
| | - Bavani Gunasegaran
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
- School of Medical Sciences, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
| | | | - Ellis Patrick
- School of Mathematics and StatisticsUniversity of SydneySydneyNSWAustralia
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
| | | | - Stuart M Grieve
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
- School of Medical Sciences, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
- Department of RadiologyRoyal Prince Alfred HospitalSydneyNSWAustralia
- Imaging and Phenotyping Laboratory, Charles Perkins Centre, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
| | - Jean Y Yang
- School of Mathematics and StatisticsUniversity of SydneySydneyNSWAustralia
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
| | - Barbara Fazekas de St Groth
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
- School of Medical Sciences, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
- Ramaciotti Facility for Human Systems BiologyUniversity of SydneySydneyNSWAustralia
| | - Helen M McGuire
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
- School of Medical Sciences, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
- Ramaciotti Facility for Human Systems BiologyUniversity of SydneySydneyNSWAustralia
| | - Gemma A Figtree
- Cardiothoracic and Vascular HealthKolling Institute of Medical ResearchSydneyNSWAustralia
- Department of Cardiology, Royal North Shore HospitalNorthern Sydney Local Health DistrictSydneyNSWAustralia
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
| |
Collapse
|
40
|
Pichichero ME. Variability of vaccine responsiveness in early life. Cell Immunol 2023; 393-394:104777. [PMID: 37866234 DOI: 10.1016/j.cellimm.2023.104777] [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: 06/15/2023] [Revised: 09/18/2023] [Accepted: 10/14/2023] [Indexed: 10/24/2023]
Abstract
Vaccinations in early life elicit variable antibody and cellular immune responses, sometimes leaving fully vaccinated children unprotected against life-threatening infectious diseases. Specific immune cell populations and immune networks may have a critical period of development and calibration in a window of opportunity occurring during the first 100 days of early life. Among the early life determinants of vaccine responses, this review will focus on modifiable factors involving development of the infant microbiota and metabolome: antibiotic exposure, breast versus formula feeding, and Caesarian section versus vaginal delivery of newborns. How microbiota may serve as natural adjuvants for vaccine responses and how microbiota-derived metabolites influence vaccine responses are also reviewed. Early life poor vaccine responsiveness can be linked to increased infection susceptibility because both phenotypes share similar immunity dysregulation profiles. An early life pre-vaccination endotype, when interventions have the highest potential for success, should be sought that predicts vaccine response trajectories.
Collapse
Affiliation(s)
- Michael E Pichichero
- Center for Infectious Diseases and Immunology, Research Institute, Rochester General Hospital, 1425 Portland Ave, Rochester, NY 14621, USA.
| |
Collapse
|
41
|
O'Neil TR, Harman AN, Cunningham AL, Nasr N, Bertram KM. OMIP-096: A 24-color flow cytometry panel to identify and characterize CD4+ and CD8+ tissue-resident T cells in human skin, intestinal, and type II mucosal tissue. Cytometry A 2023; 103:851-856. [PMID: 37772977 PMCID: PMC10953338 DOI: 10.1002/cyto.a.24782] [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: 09/03/2022] [Revised: 04/06/2023] [Accepted: 07/24/2023] [Indexed: 09/30/2023]
Abstract
There is a great need to understand human immune cells within tissue, where disease manifests and infection occurs. Tissue-resident memory T cells (TRMs) were discovered over a decade ago, there is a great need to understand their role in human disease. We developed a 24-color flow cytometry panel to comprehensively interrogate CD4+ and CD8+ TRMs isolated from human tissues. When interrogating cells within human tissue, enzymatic methods used to liberate cells from within the tissue can cause cleavage of cell surface markers needed to phenotype these cells. Here we carefully select antibody clones and evaluate the effect of enzymatic digestion on the expression of markers relevant to the identification of T cell residency, as well as markers relevant to the activation and immunoregulation status of these cells. We have designed this panel to be applicable across a range of human tissues including skin, intestine, and type II mucosae such as the vagina.
Collapse
Affiliation(s)
- Thomas R. O'Neil
- Centre for Virus Research, The Westmead Institute for Medical ResearchWestmeadAustralia
- The Westmead Clinical School, Faculty of Medicine and HealthThe University of SydneySydneyAustralia
| | - Andrew N. Harman
- Centre for Virus Research, The Westmead Institute for Medical ResearchWestmeadAustralia
- The Westmead Clinical School, Faculty of Medicine and HealthThe University of SydneySydneyAustralia
| | - Anthony L. Cunningham
- Centre for Virus Research, The Westmead Institute for Medical ResearchWestmeadAustralia
- The Westmead Clinical School, Faculty of Medicine and HealthThe University of SydneySydneyAustralia
| | - Najla Nasr
- Centre for Virus Research, The Westmead Institute for Medical ResearchWestmeadAustralia
- The Westmead Clinical School, Faculty of Medicine and HealthThe University of SydneySydneyAustralia
| | - Kirstie M. Bertram
- Centre for Virus Research, The Westmead Institute for Medical ResearchWestmeadAustralia
- The Westmead Clinical School, Faculty of Medicine and HealthThe University of SydneySydneyAustralia
| |
Collapse
|
42
|
Vollmer C, Dias A, Sales M, Sacramento PM, Silva JC, Oyamada HAA, Linhares UC, Gupta S, Kasahara TM, Bento CAM. Leptin favors imbalance of antigen-specific CD4 + T-cells associated with severity of cat allergy. Front Immunol 2023; 14:1290740. [PMID: 37954580 PMCID: PMC10639137 DOI: 10.3389/fimmu.2023.1290740] [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: 09/08/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023] Open
Abstract
Introduction Obesity can complicate IgE-mediated allergic diseases. In the present study, we aimed to investigate the ability of obesity-related concentrations of leptin to modulate the in vitro effector and regulatory Fel d1-specific CD4+ T-cell subsets in patients allergic to cat, considered the third most common cause of respiratory allergy in humans. Methods For this study, plasma and peripheral blood mononuclear cells (PBMC) from 30 cat-allergic patients with mild, moderate and severe respiratory symptoms were obtained. The PBMC cultures were stimulated with Fel d1 antigen (10 µg/mL) in the presence or absence of obesity-related leptin dose (50 ηg/mL). After 6 days, the levels of cytokines and IgE in the supernatants were evaluated by multiplex and ELISA, respectively. The frequency of different non-follicular (CXCR5-) and follicular (CXCR5+) Fel d1-specific CD4+ T cell subsets was determined by flow cytometry. The plasma levels of leptin and IgE anti-cat titers were evaluated by ELISA and ImmunoCAP, respectively. Results and conclusions Fel d1 induced both IgE production and release of cytokines related to Th2, Th9 and Th17 cell phenotypes. Feld1 was more efficient in increasing the frequency of TFHIL-21- cells positive for IL-4, IL-5 and IL-13 than TFHIL-21+ cell subsets. Leptin favored the expansion Th2-like and Th9-like cells and TFHIL-21- cells positive for IL-4, IL-5 and IL-13, but reduced the proportion of conventional (Treg/Tr-1) and follicular (TFR) regulatory CD4+ T-cell subsets expressing or not CD39 marker. Finally, many of the imbalances between Fel d1-specific CD4+ T-cells were also correlated with plasma leptin and anti-Fel d1 IgE titers. In summary, hyperleptinemia should negatively impact on the severity of cat allergies by favoring the expansion of pathogenic Fel d1-specific CD4+ T-cell phenotypes and damaging the functional status of regulatory CD4+ T-cell subsets.
Collapse
Affiliation(s)
- Carolina Vollmer
- Post-graduate Program in Cellular and Molecular Biology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Aleida Dias
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Post-graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marisa Sales
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Post-graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Priscila M. Sacramento
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Júlio Cesar Silva
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Hugo A. A. Oyamada
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Post-graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ulisses C. Linhares
- Department of Morphological Sciences, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sudhir Gupta
- Department of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Taissa M. Kasahara
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cleonice A. M. Bento
- Post-graduate Program in Cellular and Molecular Biology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Post-graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
43
|
Happel AU, Rametse L, Perumaul B, Diener C, Gibbons SM, Nyangahu DD, Donald KA, Gray C, Jaspan HB. Bifidobacterium infantis supplementation versus placebo in early life to improve immunity in infants exposed to HIV: a protocol for a randomized trial. BMC Complement Med Ther 2023; 23:367. [PMID: 37853370 PMCID: PMC10583347 DOI: 10.1186/s12906-023-04208-0] [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: 10/02/2023] [Accepted: 10/08/2023] [Indexed: 10/20/2023] Open
Abstract
INTRODUCTION Infants who are born from mothers with HIV (infants who are HIV exposed but uninfected; iHEU) are at higher risk of morbidity and display multiple immune alterations compared to infants who are HIV-unexposed (iHU). Easily implementable strategies to improve immunity of iHEU, and possibly subsequent clinical health outcomes, are needed. iHEU have altered gut microbiome composition and bifidobacterial depletion, and relative abundance of Bifidobacterium infantis has been associated with immune ontogeny, including humoral and cellular vaccine responses. Therefore, we will assess microbiological and immunological phenotypes and clinical outcomes in a randomized, double-blinded trial of B. infantis Rosell®-33 versus placebo given during the first month of life in South African iHEU. METHODS This is a parallel, randomised, controlled trial. Two-hundred breastfed iHEU will be enrolled from the Khayelitsha Site B Midwife Obstetric Unit in Cape Town, South Africa and 1:1 randomised to receive 8 × 109 CFU B. infantis Rosell®-33 daily or placebo for the first 4 weeks of life, starting on day 1-3 of life. Infants will be followed over 36 weeks with extensive collection of meta-data and samples. Primary outcomes include gut microbiome composition and diversity, intestinal inflammation and microbial translocation and cellular vaccine responses. Additional outcomes include biological (e.g. gut metabolome and T cell phenotypes) and clinical (e.g. growth and morbidity) outcome measures. DISCUSSION The results of this trial will provide evidence whether B. infantis supplementation during early life could improve health outcomes for iHEU. ETHICS AND DISSEMINATION Approval for this study has been obtained from the ethics committees at the University of Cape Town (HREC Ref 697/2022) and Seattle Children's Research Institute (STUDY00003679). TRIAL REGISTRATION Pan African Clinical Trials Registry Identifier: PACTR202301748714019. CLINICAL TRIALS gov: NCT05923333. PROTOCOL VERSION Version 1.8, dated 18 July 2023.
Collapse
Affiliation(s)
- Anna-Ursula Happel
- Department of Pathology, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa.
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa.
| | - Lerato Rametse
- Department of Pathology, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa
| | - Brandon Perumaul
- Department of Pathology, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa
| | | | - Sean M Gibbons
- Institute for Systems Biology, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
- eScience Institute, University of Washington, Seattle, WA, 98195, USA
| | - Donald D Nyangahu
- Seattle Children's Research Institute, 307 Westlake Ave. N, Seattle, WA, 98109, USA
| | - Kirsten A Donald
- Division of Developmental Paediatrics, Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Klipfontein Road Rondebosch, Cape Town, 7700, South Africa
- The Neuroscience Institute, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa
| | - Clive Gray
- Division of Molecular Biology and Human Genetics, Stellenbosch University, Francie Van Zijl Drive, Tygerberg, 7505, South Africa
| | - Heather B Jaspan
- Department of Pathology, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa
- Seattle Children's Research Institute, 307 Westlake Ave. N, Seattle, WA, 98109, USA
- Department of Pediatrics, University of Washington, 1959 NE Pacific St, Seattle, WA, 98195, USA
- Department of Global Health, University of Washington, 1510 San Juan Road NE, Seattle, WA, 98195, USA
| |
Collapse
|
44
|
de Silva TA, Apte S, Voisey J, Spann K, Tan M, Divithotawela C, Chambers D, O’Sullivan B. Single-Cell Profiling of Cells in the Lung of a Patient with Chronic Hypersensitivity Pneumonitis Reveals Inflammatory Niche with Abundant CD39+ T Cells with Functional ATPase Phenotype: A Case Study. Int J Mol Sci 2023; 24:14442. [PMID: 37833889 PMCID: PMC10572861 DOI: 10.3390/ijms241914442] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/13/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
This study investigated immune cell characteristics in chronic hypersensitivity pneumonitis (HP), focusing on CD39-expressing cells' impact on inflammation and tissue remodelling. Lung tissue from an HP patient was analysed using single-cell transcriptomics, flow cytometry, and gene expression profiling. The tissue revealed diverse cell types like macrophages, T cells, fibroblasts, and regulatory T cells (Tregs). CD39-expressing Tregs exhibited heightened ATP hydrolysis capacity and regulatory gene expression. CD39hi cells displayed markers of both Tregs and proinflammatory Th17 cells, suggesting transitional properties. Communication networks involving molecules like SPP1, collagen, CSF1, and IL-1β were identified, hinting at interactions between cell types in HP pathogenesis. This research provides insights into the immune response and cell interactions in chronic HP. CD39-expressing cells dual nature as Tregs and Th17 cells suggests a role in modulating lung inflammation, potentially affecting disease progression. These findings lay the groundwork for further research, underscoring CD39-expressing cells as potential therapeutic targets in HP.
Collapse
Affiliation(s)
- Tharushi Ayanthika de Silva
- Centre for Genomics and Personalised Health, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Chermside, Brisbane, QLD 4000, Australia
| | - Simon Apte
- Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Chermside, Brisbane, QLD 4000, Australia
- Facility of Clinical Medicine, The University of Queensland, Brisbane, QLD 4000, Australia
| | - Joanne Voisey
- Centre for Genomics and Personalised Health, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| | - Kirsten Spann
- Centre for Immunology and Infection Control, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| | - Maxine Tan
- Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Chermside, Brisbane, QLD 4000, Australia
- Facility of Clinical Medicine, The University of Queensland, Brisbane, QLD 4000, Australia
| | - Chandima Divithotawela
- Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Chermside, Brisbane, QLD 4000, Australia
| | - Daniel Chambers
- Centre for Genomics and Personalised Health, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Chermside, Brisbane, QLD 4000, Australia
- Facility of Clinical Medicine, The University of Queensland, Brisbane, QLD 4000, Australia
| | - Brendan O’Sullivan
- Centre for Genomics and Personalised Health, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Chermside, Brisbane, QLD 4000, Australia
- Facility of Clinical Medicine, The University of Queensland, Brisbane, QLD 4000, Australia
| |
Collapse
|
45
|
Mallardo D, Fordellone M, White A, Ottaviano M, Sparano F, Bailey M, Facchini AB, Ong S, Maiolino P, Caracò C, Church S, Cavalcanti E, Warren S, Budillon A, Cesano A, Simeone E, Chiodini P, Ascierto PA. CD39 and LDHA affects the prognostic role of NLR in metastatic melanoma patients treated with immunotherapy. J Transl Med 2023; 21:610. [PMID: 37684649 PMCID: PMC10492378 DOI: 10.1186/s12967-023-04419-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/05/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND Identifying response markers is highly needed to guide the treatment strategy in patients with metastatic melanoma. METHODS A retrospective study was carried out in patients with unresectable/metastatic melanoma (stage IIIb-IV), treated with anti-PD-1 in the first line setting, to better explore the role and the timing of neutrophil/lymphocyte ratio (NLR) as potential biomarker of response. The relationship of NLR with inflammation-immune mediators and the underlying negative effect of raising NLR during immunotherapy, have been investigated with transcriptomic gene analysis. RESULTS The results confirmed previous findings that a high baseline NLR is associated with a poorer prognosis and with higher serum level of lactate dehydrogenase (LDH), regardless of the presence of brain metastases. The transcriptomic analysis showed that high baseline NLR is associated with a characteristic gene signature CCNA1, LDHA and IL18R1, which correlates with inflammation and tumorigenesis. Conversely, low baseline NLR is associated with the signature CD3, SH2D1A, ZAP70 and CD45RA, linked to the immune-activation. The genes positively associated with NLR (CD39 (ENTPD1), PTEN, MYD88, MMP9 and LDH) are involved in processes of immunosuppression, inflammation and tumor-promoting activity. Increased expression of CD39 correlated with TGFβ2, a marker of the N2 neutrophils with immunosuppressive activity. CONCLUSIONS These results suggest that increasing NLR is associated with an increased neutrophil population, with polarization to the N2 phenotype, and this process may be the basis for the negatively prognostic role of NLR.
Collapse
Affiliation(s)
- Domenico Mallardo
- Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - Mario Fordellone
- Universitiy of Campania "Luigi Vanvitelli", 81100, Naples, Italy
| | | | | | - Francesca Sparano
- Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | | | | | - Sufey Ong
- NanoString Technologies Inc, Seattle, WA, USA
| | - Piera Maiolino
- Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - Corrado Caracò
- Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | | | | | | | - Alfredo Budillon
- Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | | | - Ester Simeone
- Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - Paolo Chiodini
- Universitiy of Campania "Luigi Vanvitelli", 81100, Naples, Italy
| | | |
Collapse
|
46
|
Koppensteiner L, Mathieson L, Pattle S, Dorward DA, O'Connor R, Akram AR. Location of CD39 + T cell subpopulations within tumors predict differential outcomes in non-small cell lung cancer. J Immunother Cancer 2023; 11:e006770. [PMID: 37648263 PMCID: PMC10471883 DOI: 10.1136/jitc-2023-006770] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2023] [Indexed: 09/01/2023] Open
Abstract
PURPOSE An improved mechanistic understanding of immunosuppressive pathways in non-small cell lung cancer (NSCLC) is important to develop novel diagnostic and therapeutic approaches. Here, we investigate the prognostic significance of the ectonucleotidases CD39 and CD73 in NSCLC. EXPERIMENTAL DESIGN The expression and localization of CD39, CD73 and CD103 was digitally quantified in a cohort of 162 early treatment naïve NSCLC patients using multiplex-immunofluorescence and related to patient outcome. Expression among different cell-populations was assessed via flow cytometry. Targeted RNA-Seq was performed on CD4+ and CD8+ T cells from digested NSCLC tumor tissue and single-cell RNA-Seq data was analyzed to investigate the functional significance of CD39+ T cell populations. RESULTS We demonstrate that flow cytometry of early untreated NSCLC patients shows an upregulation of CD39 expression in the tumor tissue among natural killer (NK) cells, fibroblasts and T cells. CD73 expression is mainly found among fibroblasts and Epcam+cells in the tumor tissue. Multiplex Immunofluorescence in a cohort of 162 early untreated NSCLC patients demonstrates that CD39 expression is mainly localized in the tumor stroma while CD73 expression is equally distributed between tumor nest and stroma, and high expression of CD39 and CD73 in the tumor stroma is associated with poor recurrence-free survival (RFS) at 5 years. Additionally, we find that CD8+T cells located in the tumor nest express CD103 and the density of CD39+CD103+CD8+ T cells in the tumor nest predicts improved RFS at 5 years. Targeted RNA-Seq shows that the tumor microenvironment of NSCLC upregulates regulatory pathways in CD4+ T cells and exhaustion in CD8+ T cells, and analysis of a single cell RNA sequencing dataset shows that CD39+CD4+ cells are enriched in Treg signature gene-sets, and CD39+CD103+ cytotoxic T lymphocyte show gene signatures indicative of an exhausted cytotoxic phenotype with upregulated expression of CXCL13. CONCLUSIONS Knowledge of patterns of distribution and location are required to understand the prognostic impact of CD39+ T cell populations in NSCLC. This study provides an improved understanding of spatial and functional characteristics of CD39+ T cells and their significance to patient outcome.
Collapse
Affiliation(s)
| | - Layla Mathieson
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, UK
| | - Samuel Pattle
- Department of Pathology, Royal Infirmary, Edinburgh, UK
| | | | - Richard O'Connor
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, UK
| | - Ahsan R Akram
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, UK
- Cancer Research UK Edinburgh Centre, Institute of Genetics & Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
47
|
O’Connor RA, Martinez BR, Koppensteiner L, Mathieson L, Akram AR. Cancer-associated fibroblasts drive CXCL13 production in activated T cells via TGF-beta. Front Immunol 2023; 14:1221532. [PMID: 37520560 PMCID: PMC10373066 DOI: 10.3389/fimmu.2023.1221532] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Tumour-reactive T cells producing the B-cell attractant chemokine CXCL13, in solid tumours, promote development of tertiary lymphoid structures (TLS) and are associated with improved prognosis and responsiveness to checkpoint immunotherapy. Cancer associated fibroblasts are the dominant stromal cell type in non-small cell lung cancer (NSCLC) where they co-localise with T cells and can influence T cell activation and exhaustion. We questioned whether CAF directly promote CXCL13-production during T cell activation. Methods We characterised surface markers, cytokine production and transcription factor expression in CXCL13-producing T cells in NSCLC tumours and paired non-cancerous lung samples using flow cytometry. We then assessed the influence of human NSCLC-derived primary CAF lines on T cells from healthy donors and NSCLC patients during activation in vitro measuring CXCL13 production and expression of cell-surface markers and transcription factors by flow cytometry. Results CAFs significantly increased the production of CXCL13 by both CD4+ and CD8+ T cells. CAF-induced CXCL13-producing cells lacked expression of CXCR5 and BCL6 and displayed a T peripheral helper cell phenotype. Furthermore, we demonstrate CXCL13 production by T cells is induced by TGF-β and limited by IL-2. CAF provide TGF-β during T cell activation and reduce availability of IL-2 both directly (by reducing the capacity for IL-2 production) and indirectly, by expanding a population of activated Treg. Inhibition of TGF-β signalling prevented both CAF-driven upregulation of CXCL13 and Treg expansion. Discussion Promoting CXCL13 production represents a newly described immune-regulatory function of CAF with the potential to shape the immune infiltrate of the tumour microenvironment both by altering the effector-function of tumour infiltrating T-cells and their capacity to attract B cells and promote TLS formation.
Collapse
Affiliation(s)
- Richard A. O’Connor
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Begoña Roman Martinez
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Lilian Koppensteiner
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Layla Mathieson
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Ahsan R. Akram
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
48
|
Abhishek K, Nidhi M, Chandran S, Shevkoplyas SS, Mohan C. Manufacturing regulatory T cells for adoptive cell therapy in immune diseases: A critical appraisal. Clin Immunol 2023; 251:109328. [PMID: 37086957 PMCID: PMC11003444 DOI: 10.1016/j.clim.2023.109328] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/04/2023] [Accepted: 04/07/2023] [Indexed: 04/24/2023]
Abstract
Regulatory T cells (Tregs) are a unique subset of lymphocytes that play a vital role in regulating the immune system by suppressing unwanted immune responses and thus preventing autoimmune diseases and inappropriate inflammatory reactions. In preclinical and clinical trials, these cells have demonstrated the ability to prevent and treat graft vs. host disease, alleviate autoimmune symptoms, and promote transplant tolerance. In this review, we provide a background on Treg cells with a focus on important Treg cell markers and Treg subsets, and outline the methodology currently used for manufacturing adoptive regulatory T cell therapies (TRACT). Finally, we discuss the approaches and outcomes of several clinical trials in which Tregs have been adoptively transferred to patients.
Collapse
Affiliation(s)
- Kumar Abhishek
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204-5060, United States of America
| | - Malavika Nidhi
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204-5060, United States of America
| | - Srinandhini Chandran
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204-5060, United States of America
| | - Sergey S Shevkoplyas
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204-5060, United States of America.
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204-5060, United States of America.
| |
Collapse
|
49
|
Sligar C, Cuthbertson P, Miles NA, Adhikary SR, Elhage A, Zhang G, Alexander SI, Sluyter R, Watson D. Tocilizumab increases regulatory T cells, reduces natural killer cells and delays graft-versus-host disease development in humanized mice treated with post-transplant cyclophosphamide. Immunol Cell Biol 2023. [PMID: 37191045 DOI: 10.1111/imcb.12652] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/02/2023] [Accepted: 04/19/2023] [Indexed: 05/17/2023]
Abstract
Graft-versus-host disease (GVHD) is a life-threatening complication following donor hematopoietic stem cell transplantation, where donor T cells damage host tissues. This study investigated the effect of tocilizumab (TOC) combined with post-transplant cyclophosphamide (PTCy) on immune cell engraftment and GVHD development in a humanized mouse model. NOD-scid-IL2Rγnull (NSG) mice were injected intraperitoneally with 2 × 107 human (h) peripheral blood mononuclear cells and cyclophosphamide (33 mg kg-1 ) or saline on days 3 and 4, then TOC or control antibody (0.5 mg mouse-1 ) twice weekly for 28 days. Mice were monitored for clinical signs of GVHD for either 28 or 70 days. Spleens and livers were assessed for human leukocyte subsets, and serum cytokines and tissue histology were analyzed. In the short-term model (day 28), liver and lung damage were reduced in PTCy + TOC compared with control mice. All groups showed similar splenic hCD45+ leukocyte engraftment (55-60%); however, PTCy + TOC mice demonstrated significantly increased (1.5-2-fold) splenic regulatory T cells. Serum human interferon gamma was significantly reduced in PTCy + TOC compared with control mice. Long-term (day 70), prolonged survival was similar in PTCy + TOC (median survival time, > 70 days) and PTCy mice (median survival time, 56 days). GVHD onset was significantly delayed in PTCy + TOC, compared with TOC or control mice. Notably, natural killer cells were reduced (77.5%) in TOC and PTCy + TOC mice. Overall, combining PTCy with TOC increases regulatory T cells and reduces clinical signs of early GVHD, but does not improve long-term survival compared with PTCy alone.
Collapse
Affiliation(s)
- Chloe Sligar
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Peter Cuthbertson
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Nicole A Miles
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Sam R Adhikary
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Amal Elhage
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Geoff Zhang
- The Centre for Kidney Research, The Children's Hospital at Westmead, NSW, Westmead, Australia
| | - Stephen I Alexander
- The Centre for Kidney Research, The Children's Hospital at Westmead, NSW, Westmead, Australia
| | - Ronald Sluyter
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Debbie Watson
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| |
Collapse
|
50
|
Rahim MK, Okholm TLH, Jones KB, McCarthy EE, Liu CC, Yee JL, Tamaki SJ, Marquez DM, Tenvooren I, Wai K, Cheung A, Davidson BR, Johri V, Samad B, O'Gorman WE, Krummel MF, van Zante A, Combes AJ, Angelo M, Fong L, Algazi AP, Ha P, Spitzer MH. Dynamic CD8 + T cell responses to cancer immunotherapy in human regional lymph nodes are disrupted in metastatic lymph nodes. Cell 2023; 186:1127-1143.e18. [PMID: 36931243 PMCID: PMC10348701 DOI: 10.1016/j.cell.2023.02.021] [Citation(s) in RCA: 209] [Impact Index Per Article: 104.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/28/2022] [Accepted: 02/16/2023] [Indexed: 03/18/2023]
Abstract
CD8+ T cell responses are critical for anti-tumor immunity. While extensively profiled in the tumor microenvironment, recent studies in mice identified responses in lymph nodes (LNs) as essential; however, the role of LNs in human cancer patients remains unknown. We examined CD8+ T cells in human head and neck squamous cell carcinomas, regional LNs, and blood using mass cytometry, single-cell genomics, and multiplexed ion beam imaging. We identified progenitor exhausted CD8+ T cells (Tpex) that were abundant in uninvolved LN and clonally related to terminally exhausted cells in the tumor. After anti-PD-L1 immunotherapy, Tpex in uninvolved LNs reduced in frequency but localized near dendritic cells and proliferating intermediate-exhausted CD8+ T cells (Tex-int), consistent with activation and differentiation. LN responses coincided with increased circulating Tex-int. In metastatic LNs, these response hallmarks were impaired, with immunosuppressive cellular niches. Our results identify important roles for LNs in anti-tumor immune responses in humans.
Collapse
Affiliation(s)
- Maha K Rahim
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Trine Line H Okholm
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kyle B Jones
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA 94143, USA; Pharma Technical Cell and Gene Therapy, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Elizabeth E McCarthy
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Candace C Liu
- Department of Pathology, Stanford University, Stanford, CA 94304, USA
| | - Jacqueline L Yee
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Stanley J Tamaki
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Diana M Marquez
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Iliana Tenvooren
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Katherine Wai
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alexander Cheung
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Brittany R Davidson
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Vrinda Johri
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Bushra Samad
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - William E O'Gorman
- Department of Translational Medicine, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Matthew F Krummel
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| | - Annemieke van Zante
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alexis J Combes
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael Angelo
- Department of Pathology, Stanford University, Stanford, CA 94304, USA
| | - Lawrence Fong
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| | - Alain P Algazi
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Patrick Ha
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthew H Spitzer
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|