1
|
Yang C, Zhang Y, Wang R, Cheng B, Wu Y, Fu X. IL-10 +CD19 + regulatory B cells induce CD4 +Foxp3 +regulatory T cells in serum of cervical cancer patients. Autoimmunity 2024; 57:2290909. [PMID: 38084896 DOI: 10.1080/08916934.2023.2290909] [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: 11/01/2022] [Accepted: 11/27/2023] [Indexed: 12/18/2023]
Abstract
Increase of regulatory T cells (Tregs) in the tumour microenvironment predicts worse survival of patients with various types of cancer. Recently, B cells play a significant role in the maintenance of Treg cells. However, the relevance of regulatory B cells (Bregs) to tumour immunity in humans remains elusive. Flow cytometry analysis was used to detect the Bregs and Tregs. Double staining results illustrated that the proportion of Bregs and Tregs were prominently higher in cervical cancer than normal tissues. Increase of Bregs and Tregs in cervical cancer microenvironment was associated with poor survival. Furthermore, Bregs cocultured with cervical cancer cell lines increased and induced Tregs. To sum up, the increased expression of Bregs contributes to the differentiation of CD4+ T cells into Tregs in the cervical cancer.
Collapse
Affiliation(s)
- Chunfeng Yang
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory for Major Obstetric Diseases; Guangdong Province Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macoa Greater Bay Area Higher Educaiton Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Obstetrics and Gynecology, Shenzhen Baoan Maternal and Child Health Hospital, Shenzhen, China
| | - Yuanyuan Zhang
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Rui Wang
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory for Major Obstetric Diseases; Guangdong Province Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macoa Greater Bay Area Higher Educaiton Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bing Cheng
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory for Major Obstetric Diseases; Guangdong Province Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macoa Greater Bay Area Higher Educaiton Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - You Wu
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Xi Fu
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory for Major Obstetric Diseases; Guangdong Province Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macoa Greater Bay Area Higher Educaiton Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
2
|
Wen YH, Lin YX, Zhou L, Lin C, Zhang L. The immune landscape in apical periodontitis: From mechanism to therapy. Int Endod J 2024; 57:1526-1545. [PMID: 39087849 DOI: 10.1111/iej.14125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/04/2024] [Accepted: 07/12/2024] [Indexed: 08/02/2024]
Abstract
Apical periodontitis (AP) is featured by a persistent inflammatory response and alveolar bone resorption initiated by microorganisms, posing risks to both dental and systemic health. Nonsurgical endodontic treatment is the recommended treatment plan for AP with a high success rate, but in some cases, periapical lesions may persist despite standard endodontic treatment. Better comprehension of the AP inflammatory microenvironment can help develop adjunct therapies to improve the outcome of endodontic treatment. This review presents an overview of the immune landscape in AP, elucidating how microbial invasion triggers host immune activation and shapes the inflammatory microenvironment, ultimately impacting bone homeostasis. The destructive effect of excessive immune activation on periapical tissues is emphasized. This review aimed to systematically discuss the immunological basis of AP, the inflammatory bone resorption and the immune cell network in AP, thereby providing insights into potential immunotherapeutic strategies such as targeted therapy, antioxidant therapy, adoptive cell therapy and cytokine therapy to mitigate AP-associated tissue destruction.
Collapse
Affiliation(s)
- Yuan-Hao Wen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yu-Xiu Lin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Cariology and Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lu Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Cariology and Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Chen Lin
- Department of Endodontics, Stomatological Hospital of Xiamen Medical College, Xiamen, China
| | - Lu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Cariology and Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
3
|
Liu JC, Zeng Q, Duan YG, Yeung WSB, Li RHW, Ng EHY, Cheung KW, Zhang Q, Chiu PCN. B cells: roles in physiology and pathology of pregnancy. Front Immunol 2024; 15:1456171. [PMID: 39434884 PMCID: PMC11491347 DOI: 10.3389/fimmu.2024.1456171] [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: 06/28/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
B cells constitute a diverse and adaptable immune cell population with functions that can vary according to the environment and circumstances. The involvement of B cells in pregnancy, as well as the associated molecular pathways, has yet to be investigated. This review consolidates current knowledge on B cell activities and regulation during pregnancy, with a particular focus on the roles of various B cell subsets and the effects of B cell-derived factors on pregnancy outcomes. Moreover, the review examines the significance of B cell-associated autoantibodies, cytokines, and signaling pathways in relation to pregnancy complications such as pregnancy loss, preeclampsia, and preterm birth.
Collapse
Affiliation(s)
- Jin-Chuan Liu
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Qunxiong Zeng
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yong-Gang Duan
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - William S. B. Yeung
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Raymond H. W. Li
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ernest H. Y. Ng
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ka-Wang Cheung
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Qingqing Zhang
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Philip C. N. Chiu
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
4
|
Iesari S, Nava FL, Zais IE, Coubeau L, Ferraresso M, Favi E, Lerut J. Advancing immunosuppression in liver transplantation: A narrative review. Hepatobiliary Pancreat Dis Int 2024; 23:441-448. [PMID: 38523030 DOI: 10.1016/j.hbpd.2024.03.001] [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: 05/08/2023] [Accepted: 03/14/2024] [Indexed: 03/26/2024]
Abstract
Immunosuppression is essential to ensure recipient and graft survivals after liver transplantation (LT). However, our understanding and management of the immune system remain suboptimal. Current immunosuppressive therapy cannot selectively inhibit the graft-specific immune response and entails a significant risk of serious side effects, i.e., among others, de novo cancers, infections, cardiovascular events, renal failure, metabolic syndrome, and late graft fibrosis, with progressive loss of graft function. Pharmacological research, aimed to develop alternative immunosuppressive agents in LT, is behind other solid-organ transplantation subspecialties, and, therefore, the development of new compounds and strategies should get priority in LT. The research trajectories cover mechanisms to induce T-cell exhaustion, to inhibit co-stimulation, to mitigate non-antigen-specific inflammatory response, and, lastly, to minimize the development and action of donor-specific antibodies. Moreover, while cellular modulation techniques are complex, active research is underway to foster the action of T-regulatory cells, to induce tolerogenic dendritic cells, and to promote the function of B-regulatory cells. We herein discuss current lines of research in clinical immunosuppression, particularly focusing on possible applications in the LT setting.
Collapse
Affiliation(s)
- Samuele Iesari
- General Surgery and Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 15 Via della Commenda, 20122 Milan, Italy
| | - Francesca Laura Nava
- General Surgery and Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 15 Via della Commenda, 20122 Milan, Italy
| | - Ilaria Elena Zais
- General Surgery and Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 15 Via della Commenda, 20122 Milan, Italy
| | - Laurent Coubeau
- Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, 10 Avenue Hippocrate, 1200 Brussels, Belgium; Service de Chirurgie et Transplantation Abdominale, Cliniques Universitaires Saint-Luc, 55 Avenue Hippocrate, 1200 Brussels, Belgium
| | - Mariano Ferraresso
- General Surgery and Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 15 Via della Commenda, 20122 Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 19 Via della Commenda, 20122 Milan, Italy
| | - Evaldo Favi
- General Surgery and Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 15 Via della Commenda, 20122 Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 19 Via della Commenda, 20122 Milan, Italy.
| | - Jan Lerut
- Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, 10 Avenue Hippocrate, 1200 Brussels, Belgium
| |
Collapse
|
5
|
Curioni AV, Borie R, Crestani B, Helou DG. Updates on the controversial roles of regulatory lymphoid cells in idiopathic pulmonary fibrosis. Front Immunol 2024; 15:1466901. [PMID: 39386201 PMCID: PMC11461235 DOI: 10.3389/fimmu.2024.1466901] [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: 07/19/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common and severe form of pulmonary fibrosis, characterized by scar formation in the lung interstitium. Transforming growth factor beta (TGF-β) is known as a key mediator in the fibrotic process, acting on fibroblasts and mediating their proliferation and differentiation into myofibroblasts. Although the immune system is not considered responsible for the initiation of IPF, markers of tolerogenic immunity define the pro-fibrotic microenvironment in the lungs. In homeostatic conditions, regulatory T cells (Tregs) constitute the main lymphoid population responsible for maintaining peripheral tolerance. Similar to Tregs, regulatory B cells (Bregs) represent a recently described subset of B lymphocytes with immunosuppressive functions. In the context of IPF, numerous studies have suggested a role for Tregs in enhancing fibrosis, mainly via the secretion of TGF-β. In humans, most studies show increased percentages of Tregs associated with the severity of IPF, although their exact role remains unclear. In mice, the most commonly used model involves triggering acute lung inflammation with bleomycin, leading to a subsequent fibrotic process. Consequently, data are still conflicting, as Tregs may play a protective role during the inflammatory phase and a deleterious role during the fibrotic phase. Bregs have been less studied in the context of IPF, but their role appears to be protective in experimental models of lung fibrosis. This review presents the latest updates on studies exploring the implication of regulatory lymphoid cells in IPF and compares the different approaches to better understand the origins of conflicting findings.
Collapse
Affiliation(s)
- Anna V. Curioni
- Université Paris Cité, Institut national de la santé et de la recherche médicale (INSERM), Physiopathologie et épidémiologie des maladies respiratoires (PHERE), Paris, France
| | - Raphaël Borie
- Université Paris Cité, Institut national de la santé et de la recherche médicale (INSERM), Physiopathologie et épidémiologie des maladies respiratoires (PHERE), Paris, France
- Service Pneumologie A, Assistance publique – Hôpitaux de Paris (AP-HP), Hôpital Bichat, Paris, France
| | - Bruno Crestani
- Université Paris Cité, Institut national de la santé et de la recherche médicale (INSERM), Physiopathologie et épidémiologie des maladies respiratoires (PHERE), Paris, France
- Service Pneumologie A, Assistance publique – Hôpitaux de Paris (AP-HP), Hôpital Bichat, Paris, France
| | - Doumet Georges Helou
- Université Paris Cité, Institut national de la santé et de la recherche médicale (INSERM), Physiopathologie et épidémiologie des maladies respiratoires (PHERE), Paris, France
| |
Collapse
|
6
|
Uvarova AN, Zheremyan EA, Ustiugova AS, Murashko MM, Bogomolova EA, Demin DE, Stasevich EM, Kuprash DV, Korneev KV. Autoimmunity-Associated SNP rs3024505 Disrupts STAT3 Binding in B Cells, Leading to IL10 Dysregulation. Int J Mol Sci 2024; 25:10196. [PMID: 39337678 PMCID: PMC11432243 DOI: 10.3390/ijms251810196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/11/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
Interleukin 10 (IL10) is a major anti-inflammatory cytokine that acts as a master regulator of the immune response. A single nucleotide polymorphism rs3024505(C/T), located downstream of the IL10 gene, is associated with several aggressive inflammatory diseases, including systemic lupus erythematosus, Sjögren's syndrome, Crohn's disease, and ulcerative colitis. In such autoimmune pathologies, IL10-producing B cells play a protective role by decreasing the level of inflammation and restoring immune homeostasis. This study demonstrates that rs3024505 is located within an enhancer that augments the activity of the IL10 promoter in a reporter system based on a human B cell line. The common rs3024505(C) variant creates a functional binding site for the transcription factor STAT3, whereas the risk allele rs3024505(T) disrupts STAT3 binding, thereby reducing the IL10 promoter activity. Our findings indicate that B cells from individuals carrying the minor rs3024505(T) allele may produce less IL10 due to the disrupted STAT3 binding site, contributing to the progression of inflammatory pathologies.
Collapse
Affiliation(s)
- Aksinya N. Uvarova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Elina A. Zheremyan
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Alina S. Ustiugova
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Matvey M. Murashko
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
| | - Elvina A. Bogomolova
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
| | - Denis E. Demin
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Ekaterina M. Stasevich
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
| | - Dmitry V. Kuprash
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Kirill V. Korneev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
7
|
Ahsan NF, Lourenço S, Psyllou D, Long A, Shankar S, Bashford-Rogers R. The current understanding of the phenotypic and functional properties of human regulatory B cells (Bregs). OXFORD OPEN IMMUNOLOGY 2024; 5:iqae012. [PMID: 39346706 PMCID: PMC11427547 DOI: 10.1093/oxfimm/iqae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/13/2024] [Accepted: 09/10/2024] [Indexed: 10/01/2024] Open
Abstract
B cells can have a wide range of pro- and anti- inflammatory functions. A subset of B cells called regulatory B cells (Bregs) can potently suppress immune responses. Bregs have been shown to maintain immune homeostasis and modulate inflammatory responses. Bregs are an exciting cellular target across a range of diseases, including Breg induction in autoimmunity, allergy and transplantation, and Breg suppression in cancers and infection. Bregs exhibit a remarkable phenotypic heterogeneity, rendering their unequivocal identification a challenging task. The lack of a universally accepted and exclusive surface marker set for Bregs across various studies contributes to inconsistencies in their categorization. This review paper presents a comprehensive overview of the current understanding of the phenotypic and functional properties of human Bregs while addressing the persisting ambiguities and discrepancies in their characterization. Finally, the paper examines the promising therapeutic opportunities presented by Bregs as their immunomodulatory capacities have gained attention in the context of autoimmune diseases, allergic conditions, and cancer. We explore the exciting potential in harnessing Bregs as potential therapeutic agents and the avenues that remain open for the development of Breg-based treatment strategies.
Collapse
Affiliation(s)
- Nawara Faiza Ahsan
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
- Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Stella Lourenço
- Keizo Asami Institute, Federal University of Pernambuco, Recife 50740-520, Brazil
| | - Dimitra Psyllou
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Alexander Long
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Sushma Shankar
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Rachael Bashford-Rogers
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
- Oxford Cancer Centre, University of Oxford, Oxford OX3 7LH, United Kingdom
| |
Collapse
|
8
|
Hovd AMK, Nayar S, Smith CG, Kanapathippillai P, Iannizzotto V, Barone F, Fenton KA, Pedersen HL. Podoplanin expressing macrophages and their involvement in tertiary lymphoid structures in mouse models of Sjögren's disease. Front Immunol 2024; 15:1455238. [PMID: 39355243 PMCID: PMC11442383 DOI: 10.3389/fimmu.2024.1455238] [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: 06/26/2024] [Accepted: 08/21/2024] [Indexed: 10/03/2024] Open
Abstract
Tertiary lymphoid structures (TLSs) are formed in tissues targeted by chronic inflammation processes, such as infection and autoimmunity. In Sjögren's disease, the organization of immune cells into TLS is an important part of disease progression. Here, we investigated the dynamics of tissue resident macrophages in the induction and expansion of salivary gland TLS. We induced Sjögren's disease by cannulation of the submandibular glands of C57BL/6J mice with LucAdV5. In salivary gland tissues from these mice, we analyzed the different macrophage populations prior to cannulation on day 0 and on day 2, 5, 8, 16 and 23 post-infection using multicolored flow cytometry, mRNA gene analysis, and histological evaluation of tissue specific macrophages. The histological localization of macrophages in the LucAdV5 induced inflamed salivary glands was compared to salivary glands of NZBW/F1 lupus prone mice, a spontaneous mouse model of Sjögren's disease. The evaluation of the dynamics and changes in macrophage phenotype revealed that the podoplanin (PDPN) expressing CX3CR1+ macrophage population was increased in the salivary gland tissue during LucAdV5 induced inflammation. This PDPN+ CX3CR1+ macrophage population was, together with PDPN+CD206+ macrophages, observed to be localized in the parenchyma during the acute inflammation phase as well as surrounding the TLS structure in the later stages of inflammation. This suggests a dual role of tissue resident macrophages, contributing to both proinflammatory and anti-inflammatory processes, as well as their possible interactions with other immune cells within the inflamed tissue. These macrophages may be involved with lymphoid neogenesis, which is associated with disease severity and progression. In conclusion, our study substantiates the involvement of proinflammatory and regulatory macrophages in autoimmune pathology and underlines the possible multifaceted functions of macrophages in lymphoid cell organization.
Collapse
Affiliation(s)
- Aud-Malin Karlsson Hovd
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Saba Nayar
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Charlotte G. Smith
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Premasany Kanapathippillai
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Valentina Iannizzotto
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Francesca Barone
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Kristin Andreassen Fenton
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Hege Lynum Pedersen
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
9
|
Park YH, Lee SW, Kim TC, Park HJ, Van Kaer L, Hong S. The iNKT cell ligand α-GalCer prevents murine septic shock by inducing IL10-producing iNKT and B cells. Front Immunol 2024; 15:1457690. [PMID: 39355237 PMCID: PMC11442275 DOI: 10.3389/fimmu.2024.1457690] [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: 07/01/2024] [Accepted: 08/26/2024] [Indexed: 10/03/2024] Open
Abstract
Introduction α-galactosylceramide (α-GalCer), a prototypical agonist of invariant natural killer T (iNKT) cells, stimulates iNKT cells to produce various cytokines such as IFNγ and IL4. Moreover, repeated α-GalCer treatment can cause protective or pathogenic outcomes in various immune-mediated diseases. However, the precise role of α-GalCer-activated iNKT cells in sepsis development remains unclear. To address this issue, we employed a lipopolysaccharide (LPS)/D-galactosamine (D-GalN)-induced murine sepsis model and two alternative models. Methods Sepsis was induced in wild-type (WT) C57BL/6 (B6) mice by three methods (LPS/D-GalN, α-GalCer/D-GalN, and cecal slurry), and these mice were monitored for survival rates. WT B6 mice were intraperitoneally injected with α-GalCer or OCH (an IL4-biased α-GalCer analog) one week prior to the induction of sepsis. To investigate the effects of α-GalCer-mediated iNKT cell activation on sepsis development, immune responses were analyzed by flow cytometry using splenocytes and liver-infiltrating leukocytes. In addition, a STAT6 inhibitor (AS1517499) and an IL10 inhibitor (AS101) were employed to evaluate the involvement of IL4 or IL10 signaling. Furthermore, we performed B cell adoptive transfers to examine the contribution of α-GalCer-induced regulatory B (Breg) cell populations in sepsis protection. Results In vivo α-GalCer pretreatment polarized iNKT cells towards IL4- and IL10-producing phenotypes, significantly attenuating LPS/D-GalN-induced septic lethality in WT B6 mice. Furthermore, α-GalCer pretreatment reduced the infiltration of immune cells to the liver and attenuated pro-inflammatory cytokine production. Treatment with a STAT6 inhibitor was unable to modulate disease progression, indicating that IL4 signaling did not significantly affect iNKT cell-mediated protection against sepsis. This finding was confirmed by pretreatment with OCH, which did not alter sepsis outcomes. However, interestingly, prophylactic effects of α-GalCer on sepsis were significantly suppressed by treatment with an IL10 antagonist, suggesting induction of IL10-dependent anti-inflammatory responses. In addition to IL10-producing iNKT cells, IL10-producing B cell populations were significantly increased after α-GalCer pretreatment. Conclusion Overall, our results identify α-GalCer-mediated induction of IL10 by iNKT and B cells as a promising option for controlling the pathogenesis of postoperative sepsis.
Collapse
Affiliation(s)
- Yun Hoo Park
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, Republic of Korea
| | - Sung Won Lee
- Department of Biomedical Laboratory Science, College of Health and Biomedical Services, Sangji University, Wonju, Republic of Korea
| | - Tae-Cheol Kim
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, Republic of Korea
| | - Hyun Jung Park
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, Republic of Korea
| | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Seokmann Hong
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, Republic of Korea
| |
Collapse
|
10
|
Luu M, Krause FF, Monning H, Wempe A, Leister H, Mainieri L, Staudt S, Ziegler-Martin K, Mangold K, Kappelhoff N, Shaul YD, Göttig S, Plaza-Sirvent C, Schulte LN, Bekeredjian-Ding I, Schmitz I, Steinhoff U, Visekruna A. Dissecting the metabolic signaling pathways by which microbial molecules drive the differentiation of regulatory B cells. Mucosal Immunol 2024:S1933-0219(24)00096-5. [PMID: 39265892 DOI: 10.1016/j.mucimm.2024.09.003] [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: 03/05/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
The host-microbiome axis has been implicated in promoting anti-inflammatory immune responses. Yet, the underlying molecular mechanisms of commensal-mediated IL-10 production by regulatory B cells (Bregs) are not fully elucidated. Here, we demonstrate that bacterial CpG motifs trigger the signaling downstream of TLR9 promoting IκBNS-mediated expression of Blimp-1, a transcription regulator of IL-10. Surprisingly, this effect was counteracted by the NF-κB transcription factor c-Rel. A functional screen for intestinal bacterial species identified the commensal Clostridium sporogenes, secreting high amounts of short-chain fatty acids (SCFAs) and branched-chain fatty acids (BCFAs), as an amplifier of IL-10 production by promoting sustained mTOR signaling in B cells. Consequently, enhanced Breg functionality was achieved by combining CpG with the SCFA butyrate or the BCFA isovalerate thereby synergizing TLR- and mTOR-mediated pathways. Collectively, Bregs required two bacterial signals (butyrate and CpG) to elicit their full suppressive capacity and ameliorate T cell-mediated intestinal inflammation. Our study has dissected the molecular pathways induced by bacterial factors, which might contribute not only to better understanding of host-microbiome interactions, but also to exploration of new strategies for improvement of anti-inflammatory cellular therapy.
Collapse
Affiliation(s)
- Maik Luu
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany; Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany.
| | - Felix F Krause
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Heide Monning
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Anne Wempe
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Hanna Leister
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Lisa Mainieri
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Sarah Staudt
- Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Kai Ziegler-Martin
- Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Kira Mangold
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany; Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Nora Kappelhoff
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Yoav D Shaul
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Stephan Göttig
- Institute for Medical Microbiology and Infection Control, University Hospital, Goethe University, Frankfurt am Main, Germany
| | | | - Leon N Schulte
- Institute for Lung Research, Philipps-University Marburg, Marburg, Germany
| | | | - Ingo Schmitz
- Department of Molecular Immunology, Ruhr-University Bochum, Bochum, Germany
| | - Ulrich Steinhoff
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Alexander Visekruna
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany.
| |
Collapse
|
11
|
Gao H, Zhao Y, Zhao S, Dai XQ, Qin XY, Zheng WL, He TT, Zhang N, Zhu C, Wang HM, Pan W, Zhu XM, Gao XM, Dai JF, Gong FY, Wang J. The ICF2 gene Zbtb24 specifically regulates the differentiation of B1 cells via promoting heme synthesis. Cell Mol Biol Lett 2024; 29:123. [PMID: 39277732 PMCID: PMC11401330 DOI: 10.1186/s11658-024-00641-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 08/29/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND Loss-of-function mutations of ZBTB24 cause immunodeficiency, centromeric instability, and facial anomalies syndrome 2 (ICF2). ICF2 is a rare autosomal recessive disorder with immunological defects in serum antibodies and circulating memory B cells, resulting in recurrent and sometimes fatal respiratory and gastrointestinal infections. The genotype-phenotype correlation in patients with ICF2 indicates an essential role of ZBTB24 in the terminal differentiation of B cells. METHODS We used the clustered regularly interspaced short palindromic repeats (CRISPER)/Cas9 technology to generate B cell specific Zbtb24-deficient mice and verified the deletion specificity and efficiency by quantitative polymerase chain reaction (Q-PCR) and western blotting analyses in fluorescence-activated cell sorting (FACS)-sorted cells. The development, phenotype of B cells and in vivo responses to T cell dependent or independent antigens post immunization were analyzed by flow cytometry and enzyme-linked immunosorbent assay (ELISA). Adoptive transfer experiment in combination with in vitro cultures of FACS-purified B cells and RNA-Seq analysis were utilized to specifically determine the impact of Zbtb24 on B cell biology as well as the underlying mechanisms. RESULTS Zbtb24 is dispensable for B cell development and maintenance in naive mice. Surprisingly, B cell specific deletion of Zbtb24 does not evidently compromise germinal center reactions and the resulting primary and secondary antibody responses induced by T cell dependent antigens (TD-Ags), but significantly inhibits T cell independent antigen-elicited antibody productions in vivo. At the cellular level, Zbtb24-deficiency specifically impedes the plasma cell differentiation of B1 cells without impairing their survival, activation and proliferation in vitro. Mechanistically, Zbtb24-ablation attenuates heme biosynthesis partially through mTORC1 in B1 cells, and addition of exogenous hemin abrogates the differentiation defects of Zbtb24-null B1 cells. CONCLUSIONS Zbtb24 seems to regulate antibody responses against TD-Ags B cell extrinsically, but it specifically promotes the plasma cell differentiation of B1 cells via heme synthesis in mice. Our study also suggests that defected B1 functions contribute to recurrent infections in patients with ICF2.
Collapse
Affiliation(s)
- He Gao
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Ying Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Sai Zhao
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Xiao-Qiu Dai
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Xiao-Yuan Qin
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Wei-Long Zheng
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Ting-Ting He
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Nan Zhang
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Can Zhu
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Hong-Min Wang
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Wen Pan
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Xue-Mei Zhu
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Xiao-Ming Gao
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Jian-Feng Dai
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China.
| | - Fang-Yuan Gong
- Department of Immunology, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China.
| | - Jun Wang
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
12
|
Wang C, Shu Q, Zeng N, Xie S, Zou J, Tang H, Zhou A. Immune response for acute Aeromonas hydrophila infection in two distinct color morphs of northern snakehead, Channa argus. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 52:101321. [PMID: 39260082 DOI: 10.1016/j.cbd.2024.101321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/13/2024]
Abstract
To compare and analyze the differences in immunological response between the two color morphs of Channa argus, a fish cohort was divided into four groups: black C argus + PBS (B-PBS), black C argus + Aeromonas hydrophila (B-Ah), white C. argus + PBS (W-PBS), and white C. argus + A hydrophila (W-Ah). The B-PBS and W-PBS groups received 100 μL PBS, while the B-Ah and W-Ah groups received 3.6 × 105 CFU/mL A. hydrophila in the same volume. The death rate in each group was noted, changes in plasma biochemical indicators and the expression of liver immune-related genes were examined, and transcriptome techniques were used to compare the differences between the two colors of C. argus following stress. No mortality occurred in the B-PBS and W-PBS groups. Mortality in the W-Ah and B-Ah groups showed an upward and then downward trend after A. hydrophila injection. The highest mortality occurred within 24 h and was higher in the W-Ah group than in the B-Ah group. MDA levels in the B-Ah and W-Ah groups increased and then decreased, while SOD and T-AOC showed the reverse tendency. The W-Ah and W-PBS groups differed significantly in MDA at 3, 12, and 24 h, SOD from 6 to 96 h, and T-AOC between 6 and 48 h. Plasma MDA and T-AOC levels at 12 h and SOD levels at 24 and 48 h differed significantly between the B-PBS and B-Ah groups. In both the W-Ah and B-Ah groups, the expression levels of IL-1β and IL-8 in the liver showed a temporal pattern with an initial increase followed by a decrease, reaching peak levels after 24 h, while IL-10 showed the reverse pattern. Transcriptome analysis of the liver revealed significant differences between the two C. argus colors. Differential genes in black C. argus were mainly enriched in steroid biosynthesis, glycolysis/gluconeogenesis, and glutathione and propanoate metabolism pathways 24 h after infection. In contrast, differential genes in white C. argus were mainly enriched in pathways such as oxidative phosphorylation, pancreatic secretion, and protein digestion and absorption 24 h after infection. After A. hydrophila infection, white C. argus had higher mortality, more severe oxidative stress and inflammatory responses, and lower antioxidant capacity than black C. argus.
Collapse
Affiliation(s)
- Chong Wang
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, Guangdong, China
| | - Qingsong Shu
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, Guangdong, China
| | - Nanyang Zeng
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, Guangdong, China
| | - Shaolin Xie
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, Guangdong, China
| | - Jixing Zou
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, Guangdong, China
| | - Huijuan Tang
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, Guangdong, China.
| | - Aiguo Zhou
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, Guangdong, China.
| |
Collapse
|
13
|
Poyia F, Neophytou CM, Christodoulou MI, Papageorgis P. The Role of Tumor Microenvironment in Pancreatic Cancer Immunotherapy: Current Status and Future Perspectives. Int J Mol Sci 2024; 25:9555. [PMID: 39273502 PMCID: PMC11395109 DOI: 10.3390/ijms25179555] [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: 07/26/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Pancreatic cancer comprises different subtypes, where most cases include ductal adenocarcinoma (PDAC). It is one of the deadliest tumor types, with a poor prognosis. In the majority of patients, the disease has already spread by the time of diagnosis, making full recovery unlikely and increasing mortality risk. Despite developments in its detection and management, including chemotherapy, radiotherapy, and targeted therapies as well as advances in immunotherapy, only in about 13% of PDAC patients does the overall survival exceed 5 years. This may be attributed, at least in part, to the highly desmoplastic tumor microenvironment (TME) that acts as a barrier limiting perfusion, drug delivery, and immune cell infiltration and contributes to the establishment of immunologically 'cold' conditions. Therefore, there is an urgent need to unravel the complexity of the TME that promotes PDAC progression and decipher the mechanisms of pancreatic tumors' resistance to immunotherapy. In this review, we provide an overview of the major cellular and non-cellular components of PDAC TME, as well as their biological interplays. We also discuss the current state of PDAC therapeutic treatments and focus on ongoing and future immunotherapy efforts and multimodal treatments aiming at remodeling the TME to improve therapeutic efficacy.
Collapse
Affiliation(s)
- Fotini Poyia
- Tumor Microenvironment, Metastasis and Experimental Therapeutics Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 2404, Cyprus
| | - Christiana M Neophytou
- Apoptosis and Cancer Chemoresistance Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 2404, Cyprus
| | - Maria-Ioanna Christodoulou
- Tumor Immunology and Biomarkers Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 2404, Cyprus
| | - Panagiotis Papageorgis
- Tumor Microenvironment, Metastasis and Experimental Therapeutics Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 2404, Cyprus
| |
Collapse
|
14
|
Qin M, Chen Y, Wang X, Zhang X, Pan X. Dexmedetomidine induces IL-10 secretion by B lymphocytes in the peripheral blood of patients with hepatocellular carcinoma. Immunobiology 2024; 229:152842. [PMID: 39154383 DOI: 10.1016/j.imbio.2024.152842] [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: 02/15/2024] [Revised: 07/26/2024] [Accepted: 08/13/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND/AIM To investigate the distribution of subpopulations of peripheral blood B lymphocytes in individuals with hepatocellular carcinoma (HCC), and to evaluate the effect of dexmedetomidine (DEX) on B lymphocyte differentiation in patients with HCC in vitro. METHODS Peripheral blood mononuclear cells (PBMCs) were collected from the HCC group and the healthy group, and the distribution of peripheral blood B-lymphocyte subpopulations in the two groups was examined by Flow Cytometry (FCM). B lymphocytes extracted from the peripheral blood of the HCC group were divided into D0, D1, D2 and D4 groups according to the different dose of DEX in the culture medium (0 μM, 1 μM, 2 μM and 4 μM). After 72 h of in vitro culture, FCM was used to detect differences in the percentage of apoptotic B lymphocytes and the percentage of B lymphocytes that can express interleukin 10(IL-10) and transforming growth factor-β (TGF-β) in each group. RESULTS In contrast to the healthy group, the HCC group exhibited a statistically significant increase in the proportion of CD19 + CD73 + B lymphocyte subpopulation (P<0.05). In the in vitro culture experiment, the differences in apoptosis of B lymphocytes and the percentage of TGF-β expression in each group were not statistically significant; When compared to the control group, there was a significant increase in the percentage of B lymphocytes expressing IL-10 across the D1, D2, and D4 groups (P<0.05). CONCLUSION The peripheral blood of HCC patients is characterized by an elevated presence of CD19 + CD73 + B lymphocyte subpopulations; DEX may have an immunosuppressive effect by promoting IL-10 secretion from peripheral blood B lymphocytes of HCC patients.
Collapse
Affiliation(s)
- Miaomiao Qin
- Department of Anesthesiology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, Jiangsu, China
| | - Yining Chen
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xinxin Wang
- Department of Anesthesiology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, Jiangsu, China
| | - Xiaobao Zhang
- Department of Anesthesiology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, Jiangsu, China.
| | - Xiongxiong Pan
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
15
|
Liu M. Effector and regulatory B-cell imbalance in systemic sclerosis: cooperation or competition? Clin Rheumatol 2024; 43:2783-2789. [PMID: 39080112 PMCID: PMC11330388 DOI: 10.1007/s10067-024-07086-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 08/18/2024]
Abstract
B cells play a central role in the pathogenesis of systemic sclerosis (SSc). Most B-cell studies have focused on their pathological role as antibody producers. However, in addition to immunoglobulin secretion, these cells have a wide range of functions in the immune response, including antigen presentation to T cells and cytokine production. Importantly, not all B-cell subsets promote the immune response. Regulatory B cells (Bregs) attenuate inflammation and contribute to the maintenance of immune tolerance. However, effector B cells (Beffs) positively modulate the immune response through the production of various cytokines. In SSc, Bregs are insufficient and/or dysfunctional. B-cell-targeting biologics have been trialled with promising results in the treatment of SSc. These therapies can affect Bregs or Beffs, which can potentially limit their long-term efficacy. Future strategies might involve the modulation of effector B cells in combination with the stimulation of regulatory subsets. Additionally, the monitoring of individual B-cell subsets in patients may lead to the discovery of novel biomarkers that could help predict disease relapse or progression. The purpose of this review is to summarize the relevant literatures and explain how Bregs and Beffs jointly participate in the pathogenesis of SSc.
Collapse
Affiliation(s)
- Mengguo Liu
- Department of Dermatology, Huashan Hospital, Fudan University, the 12Th Urumqi Road, Shanghai, 200040, China.
| |
Collapse
|
16
|
Petrara MR, Ruffoni E, Carmona F, Cavallari I, Zampieri S, Morello M, Del Bianco P, Rampon O, Cotugno N, Palma P, Rossi P, Giaquinto C, Giunco S, De Rossi A. HIV reservoir and premature aging: risk factors for aging-associated illnesses in adolescents and young adults with perinatally acquired HIV. PLoS Pathog 2024; 20:e1012547. [PMID: 39312589 PMCID: PMC11449303 DOI: 10.1371/journal.ppat.1012547] [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: 05/10/2024] [Revised: 10/03/2024] [Accepted: 08/30/2024] [Indexed: 09/25/2024] Open
Abstract
Despite receiving antiretroviral therapy (ART), an increasing number of adolescents and young adults with perinatally acquired HIV (PHIVAYA) are at risk of developing premature senescence and aging-associated illnesses, including cancer. Given this concern, it is crucial to assess aging biomarkers and their correlation with the HIV reservoir in order to comprehensively characterize and monitor these individuals. Fifty-five PHIVAYA (median age: 23, interquartile range [IQR]: 20-27 years, and 21 [18-23] years on ART at the time of study sampling) were studied along with 23 age-matched healthy controls. The PHIVAYA exhibited significantly higher percentages of activated, senescent, exhausted CD4 and CD8 T cells, shorter telomeres, reduced thymic output, and higher levels of circulating inflammatory markers (PAMPs, DAMPs, and pro-inflammatory cytokines IL-6, IL-8, and TNFα) as well as denervation biomarkers (neural cell adhesion molecule 1 [NCAM1] and C-terminal Agrin fragment [CAF]), compared to controls. HIV-DNA levels positively correlated with activated, senescent, exhausted CD4 and CD8 T cells, circulating biomarkers levels, and inversely with regulatory T and B cells and telomere length. According to their viremia over time, PHIVAYA were subgrouped into 14 Not Suppressed (NS)-PHIVAYA and 41 Suppressed (S)-PHIVAYA, of whom 6 who initiated ART within one year of age and maintained sustained viral suppression overtime were defined as Early Suppressed (ES)-PHIVAYA and the other 35 as Late Suppressed (LS)-PHIVAYA. ES-PHIVAYA exhibited significantly lower HIV-DNA reservoir, decreased percentages of senescent and exhausted CD4 and CD8 T cells, reduced levels of circulating inflammatory and denervation biomarkers, but longer telomere compared to LS- and NS-PHIVAYA. They differed significantly from healthy controls only in a few markers, including higher percentages of regulatory T and B cells, and higher levels of DAMPs. Overall, these results underscore the importance of initiating ART early and maintaining viral suppression to limit the establishment of the viral reservoir and to counteract immune and cellular premature aging. These findings also suggest new approaches for minimally invasive monitoring of individuals at high risk of developing premature aging and age-related illnesses.
Collapse
Affiliation(s)
- Maria Raffaella Petrara
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
- Oncology and Immunology Section, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Elena Ruffoni
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Francesco Carmona
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Ilaria Cavallari
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Sandra Zampieri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Marzia Morello
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Paola Del Bianco
- Clinical Research Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Osvalda Rampon
- Department of Women's and Children's Health, Division of Pediatric Infectious Diseases, University of Padova, Padova, Italy
| | - Nicola Cotugno
- Clinical and Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Roma, Italy
| | - Paolo Palma
- Clinical and Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Roma, Italy
| | - Paolo Rossi
- Clinical and Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Roma, Italy
| | - Carlo Giaquinto
- Department of Women's and Children's Health, Division of Pediatric Infectious Diseases, University of Padova, Padova, Italy
| | - Silvia Giunco
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
- Oncology and Immunology Section, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Anita De Rossi
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
- Oncology and Immunology Section, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| |
Collapse
|
17
|
Baridjavadi Z, Mahmoudi M, Abdollahi N, Ebadpour N, Mollazadeh S, Haghmorad D, Esmaeili SA. The humoral immune landscape in Parkinson's disease: Unraveling antibody and B cell changes. Cell Biochem Funct 2024; 42:e4109. [PMID: 39189398 DOI: 10.1002/cbf.4109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/28/2024]
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disorder characterized by the accumulation of α-synuclein (α-syn) in the brain and progressive loss of dopaminergic neurons in the substantia nigra (SN) region of the brain. Although the role of neuroinflammation and cellular immunity in PD has been extensively studied, the involvement of humoral immunity mediated by antibodies and B cells has received less attention. This article provides a comprehensive review of the current understanding of humoral immunity in PD. Here, we discuss alterations in B cells in PD, including changes in their number and phenotype. Evidence mostly indicates a decrease in the quantity of B cells in PD, accompanied by a shift in the population from naïve to memory cells. Furthermore, the existence of autoantibodies that target several antigens in PD has been investigated (i.e., anti-α-syn autoantibodies, anti-glial-derived antigen antibodies, anti-Tau antibodies, antineuromelanin antibodies, and antibodies against the renin-angiotensin system). Several autoantibodies are generated in PD, which may either provide protection or have harmful effects on disease progression. Furthermore, we have reviewed studies focusing on the utilization of antibodies as a potential treatment for PD, both in animal and clinical trials. This review sheds light on the intricate interplay between antibodies and the pathological processes in PD, including complement system activation.
Collapse
Affiliation(s)
- Zahra Baridjavadi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Narges Abdollahi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Ebadpour
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Mollazadeh
- Natural Products and Medicinal Plants Research Center, Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Dariush Haghmorad
- Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
18
|
Fadaghi S, Mahmoodi M, Derakhshani A, Sedghy F, Ranjkesh M, Behzadi A. Enhancement the antioxidative and immunomodulatory functions of mesenchymal stem cells by tetrandrine. Heliyon 2024; 10:e35667. [PMID: 39220890 PMCID: PMC11365297 DOI: 10.1016/j.heliyon.2024.e35667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
In this study, mesenchymal stem cells (MSCs) were primed with Tetrandrine (TET) having anti-inflammatory and immunomodulatory effects to examine the effects of this molecule on the antioxidative potential of MSCs as well as their modulatory effects on activated peripheral blood mononuclear cells (PBMCs). The viability of primed MSCs was detected using MTT assay and Trypan blue staining. Moreover, flow cytometry technique was applied to evaluate cell cycle distribution and immunophenotype of MSCs. The production of superoxide dismutase 3 (SOD3), malondialdehyde (MDA), kynurenine, TGF-β, and IFN-γ were also measured by spectrophotometry to assess the alteration of antioxidative and immunomodulatory potential of MSCs. Then, TET-primed MSCs were cocultured with PBMCs. The MTT assay was used to measure the proliferation of PBMCs. Cell cycle progression of PBMCs and frequency of regulatory T cells were evaluated using Flow cytometry. ELISA assay was also applied to determine the concentrations of TGF-β and IFN-γ after coculturing. According to our data, TET enhanced the secretion of SOD3 and kynurenine from MSCs, while the production of IFN-γ was reduced. No changes were observed in the viability, proliferation, and immunophenotype of MSCs after priming with TET. Moreover, the proliferation and frequency of PBMCs in the S and G2/M phases of cell cycle reduced after co-culturing with TET-primed MSCs. The concentration of TGF-β was increased in the supernatant of PBMCs, but the level of IFN-γ was reduced. Our data suggested this priming method as a novel strategy for increasing the antioxidative and immunomodulatory activity of MSCs.
Collapse
Affiliation(s)
- Shohreh Fadaghi
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Merat Mahmoodi
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Derakhshani
- Research Center for Hydatid Disease in Iran, Kerman University of Medical Sciences, Kerman, Iran
| | - Farnaz Sedghy
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahdi Ranjkesh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Ahmadreza Behzadi
- School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
19
|
Perezpayá I, Garcia SG, Clos-Sansalvador M, Sanroque-Muñoz M, Font-Morón M, Rodríguez-Martínez P, Vila-Santandreu A, Bover J, Borràs FE, Cañas L, Franquesa M. Molecular screening of transitional B cells as a prognostic marker of improved graft outcome and reduced rejection risk in kidney transplant. Front Immunol 2024; 15:1433832. [PMID: 39192987 PMCID: PMC11348389 DOI: 10.3389/fimmu.2024.1433832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/23/2024] [Indexed: 08/29/2024] Open
Abstract
Introduction Understanding immune cell dynamics in kidney transplantation may provide insight into the mechanisms of rejection and improve patient management. B cells have gained interest with a special relevance of the "regulatory" subsets and their graft outcome prognostic value. In this study, we aimed to prove that the direct immunophenotyping and target gene expression analysis of kidney transplant patients' fresh whole blood will help to identify graft rejection risk and assist in the monitoring of kidney transplanted patients. Methods We employed flow cytometry and qPCR techniques to characterize B and T cell subsets within fresh whole blood samples, with particular emphasis on transitional B cells (TrB) identified as CD19+CD24hiCD38hi. TrB are a relevant population in the context of kidney transplantation and are closely associated with regulatory B cells (Bregs) in humans. Patients were monitored, tracking pertinent clinical parameters and kidney-related events, including alterations in graft function and episodes of biopsy proven rejection. Results Higher percentages of TrB cells at 3 months after transplantation were positively associated with better graft outcomes and lower biopsy-proven acute rejection risk. Furthermore, a novel panel of B cell regulatory associated genes was validated at 3 months post-transplantation by qPCR analysis of peripheral blood mononuclear cell (PBMC) mRNA, showing high predictive power of graft events and prognostic value. Discussion These findings suggest that monitoring TrB may provide interesting patient management information, improve transplant outcomes, and allow for personalized drug regimens to minimize clinical complications.
Collapse
Affiliation(s)
- Inés Perezpayá
- REMAR-IGTP Group, Germans Trias i Pujol Research Institute (IGTP) & Nephrology Department, University Hospital Germans Trias i Pujol (HUGTiP), Barcelona, Catalonia, Spain
| | - Sergio G. Garcia
- REMAR-IGTP Group, Germans Trias i Pujol Research Institute (IGTP) & Nephrology Department, University Hospital Germans Trias i Pujol (HUGTiP), Barcelona, Catalonia, Spain
- Department of Cell Biology, Physiology and Immunology, Autonomous University of Barcelona, Bellaterra, Spain
| | - Marta Clos-Sansalvador
- REMAR-IGTP Group, Germans Trias i Pujol Research Institute (IGTP) & Nephrology Department, University Hospital Germans Trias i Pujol (HUGTiP), Barcelona, Catalonia, Spain
- Department of Cell Biology, Physiology and Immunology, Autonomous University of Barcelona, Bellaterra, Spain
| | - Marta Sanroque-Muñoz
- REMAR-IGTP Group, Germans Trias i Pujol Research Institute (IGTP) & Nephrology Department, University Hospital Germans Trias i Pujol (HUGTiP), Barcelona, Catalonia, Spain
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, Bellaterra, Spain
| | - Miriam Font-Morón
- REMAR-IGTP Group, Germans Trias i Pujol Research Institute (IGTP) & Nephrology Department, University Hospital Germans Trias i Pujol (HUGTiP), Barcelona, Catalonia, Spain
| | - Paula Rodríguez-Martínez
- Pathology Department, University Hospital Germans Trias i Pujol (HUGTiP), Barcelona, Catalonia, Spain
| | - Anna Vila-Santandreu
- REMAR-IGTP Group, Germans Trias i Pujol Research Institute (IGTP) & Nephrology Department, University Hospital Germans Trias i Pujol (HUGTiP), Barcelona, Catalonia, Spain
| | - Jordi Bover
- REMAR-IGTP Group, Germans Trias i Pujol Research Institute (IGTP) & Nephrology Department, University Hospital Germans Trias i Pujol (HUGTiP), Barcelona, Catalonia, Spain
| | - Francesc E. Borràs
- REMAR-IGTP Group, Germans Trias i Pujol Research Institute (IGTP) & Nephrology Department, University Hospital Germans Trias i Pujol (HUGTiP), Barcelona, Catalonia, Spain
- Department of Cell Biology, Physiology, and Immunology, Universitat de Barcelona (UB), Barcelona, Spain
| | - Laura Cañas
- REMAR-IGTP Group, Germans Trias i Pujol Research Institute (IGTP) & Nephrology Department, University Hospital Germans Trias i Pujol (HUGTiP), Barcelona, Catalonia, Spain
| | - Marcella Franquesa
- REMAR-IGTP Group, Germans Trias i Pujol Research Institute (IGTP) & Nephrology Department, University Hospital Germans Trias i Pujol (HUGTiP), Barcelona, Catalonia, Spain
| |
Collapse
|
20
|
Lee D, Jo MG, Min KY, Choi MY, Kim YM, Kim HS, Choi WS. IL-10 + regulatory B cells mitigate atopic dermatitis by suppressing eosinophil activation. Sci Rep 2024; 14:18164. [PMID: 39107352 PMCID: PMC11303538 DOI: 10.1038/s41598-024-68660-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
Atopic dermatitis (AD) presents significant therapeutic challenges due to its poorly understood etiology. Eosinophilia, a hallmark of allergic inflammation, is implicated in AD pathogenesis. Interleukin-10 (IL-10)-producing regulatory B (Breg) cells exhibit potent anti-inflammatory effects. However, their role in controlling AD-related eosinophilia is not well understood. To investigate the impact of eosinophils on AD, we employed IL-5Rα-deficient (Il5ra-/-) mice, which lack functional eosinophils. Induction of AD in these mice resulted in attenuated disease symptoms, underscoring the critical role of eosinophils in AD development. Additionally, the adoptive transfer of purified Breg cells into mice with AD significantly alleviated disease severity. Mechanistic studies revealed that IL-10 produced by Breg cells directly inhibits eosinophil activation and infiltration into the skin. In vitro experiments further confirmed that Breg cells inhibited eosinophil peroxidase secretion in an IL-10-dependent manner. Our collective findings demonstrate that IL-10 from Breg cells alleviates AD by suppressing eosinophil activation and tissue infiltration. This study elucidates a novel regulatory mechanism of Breg cells, providing a foundation for future Breg-mediated therapeutic strategies for AD.
Collapse
Affiliation(s)
- Dajeong Lee
- School of Medicine, Konkuk University, Chungju, 27478, Korea
| | - Min Geun Jo
- School of Medicine, Konkuk University, Chungju, 27478, Korea
| | - Keun Young Min
- School of Medicine, Konkuk University, Chungju, 27478, Korea
| | - Min Yeong Choi
- School of Medicine, Konkuk University, Chungju, 27478, Korea
| | - Young Mi Kim
- College of Pharmacy, Duksung Women's University, Seoul, 01369, Korea
| | - Hyuk Soon Kim
- Department of Biomedical Sciences, College of Natural Science and Department of Health Sciences, The Graduate School of Dong-A University, Busan, 49315, Korea.
| | - Wahn Soo Choi
- School of Medicine, Konkuk University, Chungju, 27478, Korea.
- Institute of Biomedical Sciences & Technology, Konkuk University, Seoul, 05029, Korea.
| |
Collapse
|
21
|
Wang Y, Wu L, Van Kaer L. Role of canonical and noncanonical autophagy pathways in shaping the life journey of B cells. Front Immunol 2024; 15:1426204. [PMID: 39139569 PMCID: PMC11319164 DOI: 10.3389/fimmu.2024.1426204] [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: 04/30/2024] [Accepted: 07/16/2024] [Indexed: 08/15/2024] Open
Abstract
Autophagy is a regulated intracellular catabolic process by which invading pathogens, damaged organelles, aggregated proteins, and other macromolecules are degraded in lysosomes. It has been widely appreciated that autophagic activity plays an important role in regulating the development, fate determination, and function of cells in the immune system, including B lymphocytes. Autophagy encompasses several distinct pathways that have been linked to B cell homeostasis and function. While B cell presentation of major histocompatibility complex (MHC) class II-restricted cytosolic antigens to T cells involves both macroautophagy and chaperone-mediated autophagy (CMA), plasma cells and memory B cells mainly rely on macroautophagy for their survival. Emerging evidence indicates that core autophagy factors also participate in processes related to yet clearly distinct from classical autophagy. These autophagy-related pathways, referred to as noncanonical autophagy or conjugation of ATG8 to single membranes (CASM), contribute to B cell homeostasis and functions, including MHC class II-restricted antigen presentation to T cells, germinal center formation, plasma cell differentiation, and recall responses. Dysregulation of B cell autophagy has been identified in several autoimmune and autoinflammatory diseases such as systemic lupus erythematosus, rheumatoid arthritis, and inflammatory bowel disease. In this review, we discuss recent advances in understanding the role of canonical and noncanonical autophagy in B cells, including B cell development and maturation, antigen processing and presentation, pathogen-specific antibody responses, cytokine secretion, and autoimmunity. Unraveling the molecular mechanisms of canonical and noncanonical autophagy in B cells will improve our understanding of B cell biology, with implications for the development of autophagy-based immunotherapies.
Collapse
Affiliation(s)
| | | | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States
| |
Collapse
|
22
|
Munteanu C, Schwartz B. Interactions between Dietary Antioxidants, Dietary Fiber and the Gut Microbiome: Their Putative Role in Inflammation and Cancer. Int J Mol Sci 2024; 25:8250. [PMID: 39125822 PMCID: PMC11311432 DOI: 10.3390/ijms25158250] [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: 06/20/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
The intricate relationship between the gastrointestinal (GI) microbiome and the progression of chronic non-communicable diseases underscores the significance of developing strategies to modulate the GI microbiota for promoting human health. The administration of probiotics and prebiotics represents a good strategy that enhances the population of beneficial bacteria in the intestinal lumen post-consumption, which has a positive impact on human health. In addition, dietary fibers serve as a significant energy source for bacteria inhabiting the cecum and colon. Research articles and reviews sourced from various global databases were systematically analyzed using specific phrases and keywords to investigate these relationships. There is a clear association between dietary fiber intake and improved colon function, gut motility, and reduced colorectal cancer (CRC) risk. Moreover, the state of health is reflected in the reciprocal and bidirectional relationships among food, dietary antioxidants, inflammation, and body composition. They are known for their antioxidant properties and their ability to inhibit angiogenesis, metastasis, and cell proliferation. Additionally, they promote cell survival, modulate immune and inflammatory responses, and inactivate pro-carcinogens. These actions collectively contribute to their role in cancer prevention. In different investigations, antioxidant supplements containing vitamins have been shown to lower the risk of specific cancer types. In contrast, some evidence suggests that taking antioxidant supplements can increase the risk of developing cancer. Ultimately, collaborative efforts among immunologists, clinicians, nutritionists, and dietitians are imperative for designing well-structured nutritional trials to corroborate the clinical efficacy of dietary therapy in managing inflammation and preventing carcinogenesis. This review seeks to explore the interrelationships among dietary antioxidants, dietary fiber, and the gut microbiome, with a particular focus on their potential implications in inflammation and cancer.
Collapse
Affiliation(s)
- Camelia Munteanu
- Department of Plant Culture, Faculty of Agriculture, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania
| | - Betty Schwartz
- The Institute of Biochemistry, Food Science and Nutrition, The School of Nutritional Sciences, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| |
Collapse
|
23
|
Donnenberg VS, Luketich JD, Popov B, Bartlett DL, Donnenberg AD. A common secretomic signature across epithelial cancers metastatic to the pleura supports IL-6 axis therapeutic targeting. Front Immunol 2024; 15:1404373. [PMID: 39114667 PMCID: PMC11303180 DOI: 10.3389/fimmu.2024.1404373] [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: 03/20/2024] [Accepted: 07/04/2024] [Indexed: 08/10/2024] Open
Abstract
Background Many cancers metastasize to the pleura, resulting in effusions that cause dyspnea and discomfort. Regardless of the tissue of origin, pleural malignancies are aggressive and uniformly fatal, with no treatment shown to prolong life. The pleural mesothelial monolayer is joined by tight junctions forming a contained bioreactor-like space, concentrating cytokines and chemokines secreted by the mesothelium, tumor, and infiltrating immune cells. This space represents a unique environment that profoundly influences tumor and immune cell behavior. Defining the pleural secretome is an important step in the rational development localized intrapleural immunotherapy. Method We measured cytokine/chemokine content of 252 malignant pleural effusion (MPE) samples across multiple cancers using a 40-analyte panel and Luminex multiplexing technology. Results Eleven analytes were consistently present in concentrations ≥ 10.0 pM: CXCL10/IP10 (geometric mean = 672.3 pM), CCL2/MCP1 (562.9 pM), sIL-6Rα (403.1 pM), IL-6 (137.6 pM), CXCL1/GRO (80.3 pM), TGFβ1 (76.8 pM), CCL22/MDC (54.8 pM), CXCL8/IL-8 (29.2 pM), CCL11/Eotaxin (12.6 pM), IL-10 (11.3 pM), and G-CSF (11.0 pM). All are capable of mediating chemotaxis, promotion of epithelial to mesenchymal transition, or immunosuppression, and many of are reportedly downstream of a pro-inflammatory cytokine cascade mediated by cytokine IL-6 and its soluble receptor. Conclusion The data indicate high concentrations of several cytokines and chemokines across epithelial cancers metastatic to the pleura and support the contention that the pleural environment is the major factor responsible for the clinical course of MPE across cancer types. A sIL-6Rα to IL-6 molar ratio of 2.7 ensures that virtually all epithelial, immune and vascular endothelial cells in the pleural environment are affected by IL-6 signaling. The central role likely played by IL-6 in the pathogenesis of MPE argues in favor of a therapeutic approach targeting the IL-6/IL-6R axis.
Collapse
Affiliation(s)
- Vera S. Donnenberg
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- UPMC Hillman Cancer Centers, Pittsburgh, PA, United States
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States
| | - James D. Luketich
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- UPMC Hillman Cancer Centers, Pittsburgh, PA, United States
| | - Bosko Popov
- UPMC Hillman Cancer Centers, Pittsburgh, PA, United States
| | - David L. Bartlett
- Allegheny Health Network Cancer Institute, Pittsburgh, PA, United States
- College of Medicine, Drexel University, Philadelphia, PA, United States
- Department of Surgery, College of Medicine, Drexel University, Philadelphia, PA, United States
| | - Albert D. Donnenberg
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States
- Allegheny Health Network Cancer Institute, Pittsburgh, PA, United States
- College of Medicine, Drexel University, Philadelphia, PA, United States
- Department of Medicine, College of Medicine, Drexel University, Philadelphia, PA, United States
| |
Collapse
|
24
|
Strandmoe AL, Bremer J, Diercks GFH, Gostyński A, Ammatuna E, Pas HH, Wouthuyzen-Bakker M, Huls GA, Heeringa P, Laman JD, Horváth B. Beyond the skin: B cells in pemphigus vulgaris, tolerance and treatment. Br J Dermatol 2024; 191:164-176. [PMID: 38504438 DOI: 10.1093/bjd/ljae107] [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: 12/05/2023] [Revised: 03/08/2024] [Accepted: 03/08/2024] [Indexed: 03/21/2024]
Abstract
Pemphigus vulgaris (PV) is a rare autoimmune bullous disease characterized by blistering of the skin and mucosa owing to the presence of autoantibodies against the desmosome proteins desmoglein 3 and occasionally in conjunction with desmoglein 1. Fundamental research into the pathogenesis of PV has revolutionized its treatment and outcome with rituximab, a B-cell-depleting therapy. The critical contribution of B cells to the pathogenesis of pemphigus is well accepted. However, the exact pathomechanism, mechanisms of onset, disease course and relapse remain unclear. In this narrative review, we provide an overview of the fundamental research progress that has unfolded over the past few centuries to give rise to current and emerging therapies. Furthermore, we summarize the multifaceted roles of B cells in PV, including their development, maturation and antibody activity. Finally, we explored how these various aspects of B-cell function contribute to disease pathogenesis and pave the way for innovative therapeutic interventions.
Collapse
Affiliation(s)
- Anne-Lise Strandmoe
- Departments of Medical Biology and Pathology
- Dermatology (Centre for Blistering Diseases)
| | | | - Gilles F H Diercks
- Departments of Medical Biology and Pathology
- Dermatology (Centre for Blistering Diseases)
| | - Antoni Gostyński
- Dermatology (Centre for Blistering Diseases)
- Department of Dermatology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | | | | | - Marjan Wouthuyzen-Bakker
- Medical Microbiology and Infection Prevention; University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | | | | | - Jon D Laman
- Departments of Medical Biology and Pathology
| | | |
Collapse
|
25
|
Daley AD, Bénézech C. Fat-associated lymphoid clusters: Supporting visceral adipose tissue B cell function in immunity and metabolism. Immunol Rev 2024; 324:78-94. [PMID: 38717136 DOI: 10.1111/imr.13339] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/09/2024] [Indexed: 07/23/2024]
Abstract
It is now widely understood that visceral adipose tissue (VAT) is a highly active and dynamic organ, with many functions beyond lipid accumulation and storage. In this review, we discuss the immunological role of this tissue, underpinned by the presence of fat-associated lymphoid clusters (FALCs). FALC's distinctive structure and stromal cell composition support a very different immune cell mix to that found in classical secondary lymphoid organs, which underlies their unique functions of filtration, surveillance, innate-like immune responses, and adaptive immunity within the serous cavities. FALCs are important B cell hubs providing B1 cell-mediated frontline protection against infection and supporting B2 cell-adaptative immune responses. Beyond these beneficial immune responses orchestrated by FALCs, immune cells within VAT play important homeostatic role. Dysregulation of immune cells during obesity and aging leads to chronic pathological "metabolic inflammation", which contributes to the development of cardiometabolic diseases. Here, we examine the emerging and complex functions of B cells in VAT homeostasis and the metabolic complications of obesity, highlighting the potential role that FALCs play and emphasize the areas where further research is needed.
Collapse
Affiliation(s)
- Alexander D Daley
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Cécile Bénézech
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
26
|
Giordano L, Cacciola R, Barone P, Vecchio V, Nasso ME, Alvaro ME, Gangemi S, Cacciola E, Allegra A. Autoimmune Diseases and Plasma Cells Dyscrasias: Pathogenetic, Molecular and Prognostic Correlations. Diagnostics (Basel) 2024; 14:1135. [PMID: 38893662 PMCID: PMC11171610 DOI: 10.3390/diagnostics14111135] [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: 04/13/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Multiple myeloma and monoclonal gammopathy of undetermined significance are plasma cell dyscrasias characterized by monoclonal proliferation of pathological plasma cells with uncontrolled production of immunoglobulins. Autoimmune pathologies are conditions in which T and B lymphocytes develop a tendency to activate towards self-antigens in the absence of exogenous triggers. The aim of our review is to show the possible correlations between the two pathological aspects. Molecular studies have shown how different cytokines that either cause inflammation or control the immune system play a part in the growth of immunotolerance conditions that make it easier for the development of neoplastic malignancies. Uncontrolled immune activation resulting in chronic inflammation is also known to be at the basis of the evolution toward neoplastic pathologies, as well as multiple myeloma. Another point is the impact that myeloma-specific therapies have on the course of concomitant autoimmune diseases. Indeed, cases have been observed of patients suffering from multiple myeloma treated with daratumumab and bortezomib who also benefited from their autoimmune condition or patients under treatment with immunomodulators in which there has been an arising or worsening of autoimmunity conditions. The role of bone marrow transplantation in the course of concomitant autoimmune diseases remains under analysis.
Collapse
Affiliation(s)
- Laura Giordano
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (L.G.); (P.B.); (V.V.); (M.E.N.); (M.E.A.)
| | - Rossella Cacciola
- Hemostasis/Hematology Unit, Department of Experimental and Clinical Medicine, University of Catania, 95123 Catania, Italy;
| | - Paola Barone
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (L.G.); (P.B.); (V.V.); (M.E.N.); (M.E.A.)
| | - Veronica Vecchio
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (L.G.); (P.B.); (V.V.); (M.E.N.); (M.E.A.)
| | - Maria Elisa Nasso
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (L.G.); (P.B.); (V.V.); (M.E.N.); (M.E.A.)
| | - Maria Eugenia Alvaro
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (L.G.); (P.B.); (V.V.); (M.E.N.); (M.E.A.)
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department and Experimental Medicine, University of Messina, 98125 Messina, Italy;
| | - Emma Cacciola
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy;
| | - Alessandro Allegra
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (L.G.); (P.B.); (V.V.); (M.E.N.); (M.E.A.)
| |
Collapse
|
27
|
Vilardi A, Przyborski S, Mobbs C, Rufini A, Tufarelli C. Current understanding of the interplay between extracellular matrix remodelling and gut permeability in health and disease. Cell Death Discov 2024; 10:258. [PMID: 38802341 PMCID: PMC11130177 DOI: 10.1038/s41420-024-02015-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/25/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024] Open
Abstract
The intestinal wall represents an interactive network regulated by the intestinal epithelium, extracellular matrix (ECM) and mesenchymal compartment. Under healthy physiological conditions, the epithelium undergoes constant renewal and forms an integral and selective barrier. Following damage, the healthy epithelium is restored via a series of signalling pathways that result in remodelling of the scaffolding tissue through finely-regulated proteolysis of the ECM by proteases such as matrix metalloproteinases (MMPs). However, chronic inflammation of the gastrointestinal tract, as occurs in Inflammatory Bowel Disease (IBD), is associated with prolonged disruption of the epithelial barrier and persistent damage to the intestinal mucosa. Increased barrier permeability exhibits distinctive signatures of inflammatory, immunological and ECM components, accompanied by increased ECM proteolytic activity. This narrative review aims to bring together the current knowledge of the interplay between gut barrier, immune and ECM features in health and disease, discussing the role of barrier permeability as a discriminant between homoeostasis and IBD.
Collapse
Affiliation(s)
- Aurora Vilardi
- Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, United Kingdom
| | - Stefan Przyborski
- Department of Biosciences, Durham University, Durham, DH1 3LE, United Kingdom
| | - Claire Mobbs
- Department of Biosciences, Durham University, Durham, DH1 3LE, United Kingdom
| | - Alessandro Rufini
- Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, United Kingdom.
- Department of Biosciences, University of Milan, Milan, 20133, Italy.
| | - Cristina Tufarelli
- Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, United Kingdom.
| |
Collapse
|
28
|
Pan T, Ding P, Huang A, Tang B, Song K, Sun G, Wu Y, Yang S, Chen X, Wang D, Zhu X. Reconstitution of double-negative T cells after cord blood transplantation and its predictive value for acute graft-versus-host disease. Chin Med J (Engl) 2024; 137:1207-1217. [PMID: 37620289 PMCID: PMC11101234 DOI: 10.1097/cm9.0000000000002807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND With an increasing number of patients with hematological malignancies being treated with umbilical cord blood transplantation (UCBT), the correlation between immune reconstitution (IR) after UCBT and graft-versus-host disease (GVHD) has been reported successively, but reports on double-negative T (DNT) cell reconstitution and its association with acute GVHD (aGVHD) after UCBT are lacking. METHODS A population-based observational study was conducted among 131 patients with hematological malignancies who underwent single-unit UCBT as their first transplant at the Department of Hematology, the First Affiliated Hospital of USTC, between August 2018 and June 2021. IR differences were compared between the patients with and without aGVHD. RESULTS The absolute number of DNT cells in the healthy Chinese population was 109 (70-157)/μL, accounting for 5.82 (3.98-8.19)% of lymphocytes. DNT cells showed delayed recovery and could not reach their normal levels even one year after transplantation. Importantly, the absolute number and percentage of DNT cells were significantly higher in UCBT patients without aGVHD than in those with aGVHD within one year ( F = 4.684, P = 0.039 and F = 5.583, P = 0.026, respectively). In addition, the number of DNT cells in the first month after transplantation decreased significantly with the degree of aGVHD increased, and faster DNT cell reconstitution in the first month after UCBT was an independent protective factor for aGVHD (HR = 0.46, 95% confidence interval [CI]: 0.23-0.93; P = 0.031). CONCLUSIONS Compared to the number of DNT cells in Chinese healthy people, the reconstitution of DNT cells in adults with hematological malignancies after UCBT was slow. In addition, the faster reconstitution of DNT cells in the early stage after transplantation was associated with a lower incidence of aGVHD.
Collapse
Affiliation(s)
- Tianzhong Pan
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
- Anhui Provincial Key Laboratory of Blood Research and Applications, Hefei, Anhui, 230001, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Peng Ding
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Aijie Huang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Baolin Tang
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
- Anhui Provincial Key Laboratory of Blood Research and Applications, Hefei, Anhui, 230001, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Kaidi Song
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Guangyu Sun
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
- Anhui Provincial Key Laboratory of Blood Research and Applications, Hefei, Anhui, 230001, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Yue Wu
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
- Anhui Provincial Key Laboratory of Blood Research and Applications, Hefei, Anhui, 230001, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Shiying Yang
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
- Anhui Provincial Key Laboratory of Blood Research and Applications, Hefei, Anhui, 230001, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Xingchi Chen
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
- Anhui Provincial Key Laboratory of Blood Research and Applications, Hefei, Anhui, 230001, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Dongyao Wang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Xiaoyu Zhu
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
- Anhui Provincial Key Laboratory of Blood Research and Applications, Hefei, Anhui, 230001, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| |
Collapse
|
29
|
Xu Z, Li R, Wang L, Wu Y, Tian Y, Su Y, Ma Y, Li R, Wei Y, Zhang C, Han S, Duan S, Peng H, Xue J. Pathogenic role of different phenotypes of immune cells in airway allergic diseases: a study based on Mendelian randomization. Front Immunol 2024; 15:1349470. [PMID: 38812518 PMCID: PMC11133742 DOI: 10.3389/fimmu.2024.1349470] [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: 12/04/2023] [Accepted: 04/25/2024] [Indexed: 05/31/2024] Open
Abstract
Background Airway allergic disease (AAD) is a class of autoimmune diseases with predominantly Th2-type inflammation, mainly including allergic rhinitis (AR), allergic asthma (AS), and chronic sinusitis (CRS). There are very complex regulatory mechanisms between immune cells and AAD; however, previous reports found that the functions of the same immune cells in AAD are not identical. Objective The aim of this study was to explore the causal relationship between different phenotypic immune cells and their association with AAD. Method Utilizing the publicly available Genome-Wide Association Studies (GWAS) database, this study conducted a bidirectional Mendelian randomization (MR) to assess the causal relationship between immune cells of 731 different immunophenotypes and AAD. The primary assessment methods included inverse variance weighting, weighted median, and MR Egger. Additionally, sensitivity analyses such as MR-PRESSO, leave-one-out, and scatter plots were employed to eliminate the interference of heterogeneity and pleiotropy, ensuring the stability of the causal inference. Result A total of 38 immune cells with different immunophenotypes were found to be positively and causally associated with AR, of which 26 were protective factors and 12 were risk factors. Positive associations were found between 33 immune cells and AS, of which 14 were protective factors and 19 were risk factors, as well as between 39 immune cells and CRS, of which 22 were protective factors and 17 were risk factors. Finally, the results of all relevant immune cells for the three diseases were taken and intersected, and it was found that CD3 on CD39+-activated Treg (IVWAR = 0.001, IVWCRS = 0.043, IVWAS = 0.027) may be the key immune cell that inhibits the development of AAD (ORAR = 0.940, ORAS = 0.967, ORCRS = 0.976). Conclusion This study reveals that different immune phenotypes of immune cells are closely related to AAD at the genetic level, which provides a theoretical basis for future clinical studies.
Collapse
Affiliation(s)
- Zhihan Xu
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ren Li
- Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Leigang Wang
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yisha Wu
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yuhe Tian
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yilin Su
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yuqiang Ma
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ruiying Li
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yao Wei
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Chen Zhang
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Shikai Han
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Siyu Duan
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Haiyi Peng
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jinmei Xue
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
30
|
Li Q, An N, Liu C, Ding Y, Yang C, Ma X, Yang W, Piao J, Zhu J, Liu J. Single-cell BCR and transcriptome analysis reveals peripheral immune signatures in patients with thyroid-associated ophthalmopathy. Aging (Albany NY) 2024; 16:8217-8245. [PMID: 38728262 PMCID: PMC11132005 DOI: 10.18632/aging.205814] [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: 11/17/2023] [Accepted: 04/09/2024] [Indexed: 05/12/2024]
Abstract
Thyroid-associated ophthalmopathy (TAO) is the most prevalent orbital disease in adults caused by an autoimmune disorder, which can lead to disfigurement and vision impairment. Developing effective treatments for this condition presents challenges due to our limited understanding of its underlying immune aberrations. In this study, we profiled the immune components in the peripheral blood of patients with TAO as well as healthy individuals, utilizing single-cell RNA sequencing and B-cell receptor repertoires (BCR) analysis. We observed a significant reduction in the proportions of regulatory B cells (Bregs) and type 2 conventional dendritic cells (DCs) in patients with TAO during the active phase. Conversely, there was a significant increase in the proportion of type 1 DCs. Further analysis of cell differentiation trajectory revealed potential impairment in the transition of B cells towards Breg phenotype during the active phase of TAO. Besides, the activation process of TAO appeared to involve inflammation and immune dysfunction, as indicated by the dynamic changes in the activities of key regulators. The abnormalities in the peripheral immune system, such as the reduced capacity of Bregs to suppress inflammation, were primarily driven by the enhanced interaction among Breg, DCs, and monocytes (i.e., CD22-PTPRC and BTLA-TNFRSF14). Collectively, our findings offer a comprehensive insight into the molecular regulation and cellular reconfiguration during the active phase of TAO at the single-cell level, in order to explore the pathogenesis of TAO and provide new ideas for the future treatment of TAO.
Collapse
Affiliation(s)
- Qian Li
- Department of Ophthalmology, Peoples’ Hospital of Ningxia Hui Autonomous Region, The Third Affiliated Clinical College of Ningxia Medical University, Yinchuan, Ningxia, P.R. China
| | - Ningyu An
- Department of Ophthalmology, Peoples’ Hospital of Ningxia Hui Autonomous Region, The Third Affiliated Clinical College of Ningxia Medical University, Yinchuan, Ningxia, P.R. China
| | - Cheng Liu
- Medical Science Research Institution of Ningxia Hui Autonomous Region, Medical Sci-Tech Research Center of Ningxia Medical University, Yinchuan, Ningxia, P.R. China
| | - Yungang Ding
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, P.R. China
| | - Cuixia Yang
- Department of Ophthalmology, Peoples’ Hospital of Ningxia Hui Autonomous Region, The Third Affiliated Clinical College of Ningxia Medical University, Yinchuan, Ningxia, P.R. China
| | - Xiumei Ma
- Department of Ophthalmology, Peoples’ Hospital of Ningxia Hui Autonomous Region, The Third Affiliated Clinical College of Ningxia Medical University, Yinchuan, Ningxia, P.R. China
| | - Wei Yang
- Department of Ophthalmology, Peoples’ Hospital of Ningxia Hui Autonomous Region, The Third Affiliated Clinical College of Ningxia Medical University, Yinchuan, Ningxia, P.R. China
| | - Junfeng Piao
- Department of Ophthalmology, Peoples’ Hospital of Ningxia Hui Autonomous Region, The Third Affiliated Clinical College of Ningxia Medical University, Yinchuan, Ningxia, P.R. China
- Department of Ophthalmology, Guro Hospital, Korea University College of Medicine, Guro-gu, Seoul 152–703, South Korea
| | - Jinyan Zhu
- Department of Ophthalmology, Peoples’ Hospital of Ningxia Hui Autonomous Region, The Third Affiliated Clinical College of Ningxia Medical University, Yinchuan, Ningxia, P.R. China
| | - Junxiu Liu
- Department of Ophthalmology, Peoples’ Hospital of Ningxia Hui Autonomous Region, The Third Affiliated Clinical College of Ningxia Medical University, Yinchuan, Ningxia, P.R. China
| |
Collapse
|
31
|
Chen W, Ning X, Liu Y, Shen T, Liu M, Yin H, Ding Y, Zhou J, Yin R, Cai L, Wu Y, Qian L. Myeloid-derived suppressor cells from tumour-bearing mice induce the population expansion of CD19 hiFcγRIIb hi regulatory B cells via PD-L1. Immunology 2024; 172:127-143. [PMID: 38332630 DOI: 10.1111/imm.13763] [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: 01/26/2023] [Accepted: 01/25/2024] [Indexed: 02/10/2024] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) increase in number and gain immunosuppressive functions in tumours and many other pathological conditions. MDSCs are characterized by their strong T-cell immunosuppressive capacity. The effects that MDSCs may have on B cells, especially within the tumour microenvironment, are less well understood. Here, we report that either monocytic MDSCs or polymorphonuclear MDSCs can promote increases in interleukin (IL)-10-expressing CD19hiFcγRIIbhi regulatory B cells in vitro and in vivo. Splenic transitional-1, -2, and -3 cells and marginal zone B cells, but not follicular B cells, differentiate into IL-10-expressing CD19hiFcγRIIbhi regulatory B cells. The adoptive transfer of CD19hiFcγRIIbhi regulatory B cells via tail vein injection can promote subcutaneous 3LL tumour growth in mice. The expression of programmed death-ligand 1 on MDSCs was found to be strongly associated with CD19hiFcγRIIbhi regulatory B cell population expansion. Furthermore, the frequency of circulating CD19+FcγRIIhi regulatory B cells was significantly increased in advanced-stage lung cancer patients. Our results unveil a critical role of MDSCs in regulatory B-cell differentiation and population expansion in lung cancer patients.
Collapse
Affiliation(s)
- Wenyan Chen
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Xiaomin Ning
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Yang Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Tingting Shen
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Mengru Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Hui Yin
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Yue Ding
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Jingwen Zhou
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Rui Yin
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Liangliang Cai
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Yuhan Wu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Li Qian
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, China
| |
Collapse
|
32
|
Bracamonte-Baran W, Kim ST. The Current and Future of Biomarkers of Immune Related Adverse Events. Rheum Dis Clin North Am 2024; 50:201-227. [PMID: 38670721 PMCID: PMC11232920 DOI: 10.1016/j.rdc.2024.01.004] [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] [Indexed: 04/28/2024]
Abstract
With their groundbreaking clinical responses, immune checkpoint inhibitors (ICIs) have ushered in a new chapter in cancer therapeutics. However, they are often associated with life-threatening or organ-threatening autoimmune/autoinflammatory phenomena, collectively termed immune-related adverse events (irAEs). In this review, we will first describe the mechanisms of action of ICIs as well as irAEs. Next, we will review biomarkers for predicting the development of irAEs or stratifying risks.
Collapse
Affiliation(s)
- William Bracamonte-Baran
- Department of Rheumatology, Allergy & Immunology, Yale University, 300 Cedar Street, TAC S541, New Haven, CT 06520, USA
| | - Sang T Kim
- Department of Rheumatology, Allergy & Immunology, Yale University, 300 Cedar Street, TAC S541, New Haven, CT 06520, USA.
| |
Collapse
|
33
|
Surendar J, Hackenberg RK, Schmitt-Sánchez F, Ossendorff R, Welle K, Stoffel-Wagner B, Sage PT, Burger C, Wirtz DC, Strauss AC, Schildberg FA. Osteomyelitis is associated with increased anti-inflammatory response and immune exhaustion. Front Immunol 2024; 15:1396592. [PMID: 38736874 PMCID: PMC11082283 DOI: 10.3389/fimmu.2024.1396592] [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: 03/06/2024] [Accepted: 04/12/2024] [Indexed: 05/14/2024] Open
Abstract
Introduction Osteomyelitis (OMS) is a bone infection causing bone pain and severe complications. A balanced immune response is critical to eradicate infection without harming the host, yet pathogens manipulate immunity to establish a chronic infection. Understanding OMS-driven inflammation is essential for disease management, but comprehensive data on immune profiles and immune cell activation during OMS are lacking. Methods Using high-dimensional flow cytometry, we investigated the detailed innate and adaptive systemic immune cell populations in OMS and age- and sex-matched controls. Results Our study revealed that OMS is associated with increased levels of immune regulatory cells, namely T regulatory cells, B regulatory cells, and T follicular regulatory cells. In addition, the expression of immune activation markers HLA-DR and CD86 was decreased in OMS, while the expression of immune exhaustion markers TIM-3, PD-1, PD-L1, and VISTA was increased. Members of the T follicular helper (Tfh) cell family as well as classical and typical memory B cells were significantly increased in OMS individuals. We also found a strong correlation between memory B cells and Tfh cells. Discussion We conclude that OMS skews the host immune system towards the immunomodulatory arm and that the Tfh memory B cell axis is evident in OMS. Therefore, immune-directed therapies may be a promising alternative for eradication and recurrence of infection in OMS, particularly in individuals and areas where antibiotic resistance is a major concern.
Collapse
Affiliation(s)
- Jayagopi Surendar
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Roslind K. Hackenberg
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Fabio Schmitt-Sánchez
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Robert Ossendorff
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Kristian Welle
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Birgit Stoffel-Wagner
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Peter T. Sage
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Christof Burger
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Dieter C. Wirtz
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Andreas C. Strauss
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Frank A. Schildberg
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
34
|
Liu Z, Zhang D, Chen S. Unveiling the gastric microbiota: implications for gastric carcinogenesis, immune responses, and clinical prospects. J Exp Clin Cancer Res 2024; 43:118. [PMID: 38641815 PMCID: PMC11027554 DOI: 10.1186/s13046-024-03034-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 03/29/2024] [Indexed: 04/21/2024] Open
Abstract
High-throughput sequencing has ushered in a paradigm shift in gastric microbiota, breaking the stereotype that the stomach is hostile to microorganisms beyond H. pylori. Recent attention directed toward the composition and functionality of this 'community' has shed light on its potential relevance in cancer. The microbial composition in the stomach of health displays host specificity which changes throughout a person's lifespan and is subject to both external and internal factors. Distinctive alterations in gastric microbiome signature are discernible at different stages of gastric precancerous lesions and malignancy. The robust microbes that dominate in gastric malignant tissue are intricately implicated in gastric cancer susceptibility, carcinogenesis, and the modulation of immunosurveillance and immune escape. These revelations offer fresh avenues for utilizing gastric microbiota as predictive biomarkers in clinical settings. Furthermore, inter-individual microbiota variations partially account for differential responses to cancer immunotherapy. In this review, we summarize current literature on the influence of the gastric microbiota on gastric carcinogenesis, anti-tumor immunity and immunotherapy, providing insights into potential clinical applications.
Collapse
Affiliation(s)
- Zhiyi Liu
- Department of Oncology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Dachuan Zhang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Siyu Chen
- Department of Oncology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China.
| |
Collapse
|
35
|
Wang Z, Li M, Bi L, Hu X, Wang Y. Traditional Chinese Medicine in Regulating Tumor Microenvironment. Onco Targets Ther 2024; 17:313-325. [PMID: 38617090 PMCID: PMC11016250 DOI: 10.2147/ott.s444214] [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: 10/31/2023] [Accepted: 03/15/2024] [Indexed: 04/16/2024] Open
Abstract
Tumor microenvironment (TME) is a complex and integrated system containing a variety of tumor-infiltrating immune cells and stromal cells. They are closely connected with cancer cells and influence the development and progression of cancer. Traditional Chinese medicine (TCM) is an important complementary therapy for cancer treatment in China. It mainly eliminates cancer cells by regulating TME. The aim of this review is to systematically summarize the crosstalk between tumor cells and TME, and to summarize the research progress of TCM in regulating TME. The review is of great significance in revealing the therapeutic mechanism of action of TCM, and provides an opportunity for the combined application of TCM and immunotherapy in cancer treatment.
Collapse
Affiliation(s)
- Ziwei Wang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Mengyao Li
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Ling Bi
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Xueqing Hu
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Yan Wang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| |
Collapse
|
36
|
Rodríguez-Zhurbenko N, Hernández AM. The role of B-1 cells in cancer progression and anti-tumor immunity. Front Immunol 2024; 15:1363176. [PMID: 38629061 PMCID: PMC11019000 DOI: 10.3389/fimmu.2024.1363176] [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: 12/29/2023] [Accepted: 03/14/2024] [Indexed: 04/19/2024] Open
Abstract
In recent years, in addition to the well-established role of T cells in controlling or promoting tumor growth, a new wave of research has demonstrated the active involvement of B cells in tumor immunity. B-cell subsets with distinct phenotypes and functions play various roles in tumor progression. Plasma cells and activated B cells have been linked to improved clinical outcomes in several types of cancer, whereas regulatory B cells have been associated with disease progression. However, we are only beginning to understand the role of a particular innate subset of B cells, referred to as B-1 cells, in cancer. Here, we summarize the characteristics of B-1 cells and review their ability to infiltrate tumors. We also describe the potential mechanisms through which B-1 cells suppress anti-tumor immune responses and promote tumor progression. Additionally, we highlight recent studies on the protective anti-tumor function of B-1 cells in both mouse models and humans. Understanding the functions of B-1 cells in tumor immunity could pave the way for designing more effective cancer immunotherapies.
Collapse
Affiliation(s)
- Nely Rodríguez-Zhurbenko
- Immunobiology Department, Immunology and Immunotherapy Division, Center of Molecular Immunology, Habana, Cuba
| | - Ana M. Hernández
- Applied Genetics Group, Department of Biochemistry, Faculty of Biology, University of Habana, Habana, Cuba
| |
Collapse
|
37
|
Zhou J, Zhou R, Zhu Y, Deng S, Muhuitijiang B, Li C, Shi X, Zhang L, Tan W. Investigating the impact of regulatory B cells and regulatory B cell-related genes on bladder cancer progression and immunotherapeutic sensitivity. J Exp Clin Cancer Res 2024; 43:101. [PMID: 38566204 PMCID: PMC10985985 DOI: 10.1186/s13046-024-03017-8] [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: 11/27/2023] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Regulatory B cells (Bregs), a specialized subset of B cells that modulate immune responses and maintain immune tolerance in malignant tumors, have not been extensively investigated in the context of bladder cancer (BLCA). This study aims to elucidate the roles of Bregs and Breg-related genes in BLCA. METHODS We assessed Breg infiltration levels in 34 pairs of BLCA and corresponding paracancerous tissues using immunohistochemical staining. We conducted transwell and wound healing assays to evaluate the impact of Bregs on the malignant phenotype of SW780 and T24 cells. Breg-related genes were identified through gene sets and transcriptional analysis. The TCGA-BLCA cohort served as the training set, while the IMvigor210 and 5 GEO cohorts were used as external validation sets. We employed LASSO regression and random forest for feature selection and developed a risk signature using Cox regression. Primary validation of the risk signature was performed through immunohistochemical staining and RT-qPCR experiments using the 34 local BLCA samples. Additionally, we employed transfection assays and flow cytometry to investigate Breg expansion ability and immunosuppressive functions. RESULTS Breg levels in BLCA tissues were significantly elevated compared to paracancerous tissues (P < 0.05) and positively correlated with tumor malignancy (P < 0.05). Co-incubation of SW780 and T24 cells with Bregs resulted in enhanced invasion and migration abilities (all P < 0.05). We identified 27 Breg-related genes, including CD96, OAS1, and CSH1, which were integrated into the risk signature. This signature demonstrated robust prognostic classification across the 6 cohorts (pooled HR = 2.25, 95% CI = 1.52-3.33). Moreover, the signature exhibited positive associations with advanced tumor stage (P < 0.001) and Breg infiltration ratios (P < 0.05) in the local samples. Furthermore, the signature successfully predicted immunotherapeutic sensitivity in three cohorts (all P < 0.05). Knockdown of CSH1 in B cells increased Breg phenotype and enhanced suppressive ability against CD8 + T cells (all P < 0.05). CONCLUSIONS Bregs play a pro-tumor role in the development of BLCA. The Breg-related gene signature established in this study holds great potential as a valuable tool for evaluating prognosis and predicting immunotherapeutic response in BLCA patients.
Collapse
Affiliation(s)
- Jiawei Zhou
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510080, China
- The First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Ranran Zhou
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510080, China
- The First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Yuanchao Zhu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510080, China
- The First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Shikai Deng
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, No. 1023-1063 Shatai South Road, Baiyun District, Guangzhou, Guangdong, 510080, China
| | - Bahaerguli Muhuitijiang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510080, China
- The First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Chengyao Li
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, No. 1023-1063 Shatai South Road, Baiyun District, Guangzhou, Guangdong, 510080, China
| | - Xiaojun Shi
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510080, China
- The First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Ling Zhang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, No. 1023-1063 Shatai South Road, Baiyun District, Guangzhou, Guangdong, 510080, China.
| | - Wanlong Tan
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510080, China.
- The First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
38
|
Gutiérrez-Vera C, García-Betancourt R, Palacios PA, Müller M, Montero DA, Verdugo C, Ortiz F, Simon F, Kalergis AM, González PA, Saavedra-Avila NA, Porcelli SA, Carreño LJ. Natural killer T cells in allergic asthma: implications for the development of novel immunotherapeutical strategies. Front Immunol 2024; 15:1364774. [PMID: 38629075 PMCID: PMC11018981 DOI: 10.3389/fimmu.2024.1364774] [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: 01/03/2024] [Accepted: 03/06/2024] [Indexed: 04/19/2024] Open
Abstract
Allergic asthma has emerged as a prevalent allergic disease worldwide, affecting most prominently both young individuals and lower-income populations in developing and developed countries. To devise effective and curative immunotherapy, it is crucial to comprehend the intricate nature of this condition, characterized by an immune response imbalance that favors a proinflammatory profile orchestrated by diverse subsets of immune cells. Although the involvement of Natural Killer T (NKT) cells in asthma pathology is frequently implied, their specific contributions to disease onset and progression remain incompletely understood. Given their remarkable ability to modulate the immune response through the rapid secretion of various cytokines, NKT cells represent a promising target for the development of effective immunotherapy against allergic asthma. This review provides a comprehensive summary of the current understanding of NKT cells in the context of allergic asthma, along with novel therapeutic approaches that leverage the functional response of these cells.
Collapse
Affiliation(s)
- Cristián Gutiérrez-Vera
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Richard García-Betancourt
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Pablo A. Palacios
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Marioly Müller
- Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - David A. Montero
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Carlos Verdugo
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Francisca Ortiz
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Felipe Simon
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Noemi A. Saavedra-Avila
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Steven A. Porcelli
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Leandro J. Carreño
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
39
|
Sanges S, Tian W, Dubucquoi S, Chang JL, Collet A, Launay D, Nicolls MR. B-cells in pulmonary arterial hypertension: friend, foe or bystander? Eur Respir J 2024; 63:2301949. [PMID: 38485150 PMCID: PMC11043614 DOI: 10.1183/13993003.01949-2023] [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: 11/02/2023] [Accepted: 03/01/2024] [Indexed: 04/22/2024]
Abstract
There is an unmet need for new therapeutic strategies that target alternative pathways to improve the prognosis of patients with pulmonary arterial hypertension (PAH). As immunity has been involved in the development and progression of vascular lesions in PAH, we review the potential contribution of B-cells in its pathogenesis and evaluate the relevance of B-cell-targeted therapies. Circulating B-cell homeostasis is altered in PAH patients, with total B-cell lymphopenia, abnormal subset distribution (expansion of naïve and antibody-secreting cells, reduction of memory B-cells) and chronic activation. B-cells are recruited to the lungs through local chemokine secretion, and activated by several mechanisms: 1) interaction with lung vascular autoantigens through cognate B-cell receptors; 2) costimulatory signals provided by T follicular helper cells (interleukin (IL)-21), type 2 T helper cells and mast cells (IL-4, IL-6 and IL-13); and 3) increased survival signals provided by B-cell activating factor pathways. This activity results in the formation of germinal centres within perivascular tertiary lymphoid organs and in the local production of pathogenic autoantibodies that target the pulmonary vasculature and vascular stabilisation factors (including angiotensin-II/endothelin-1 receptors and bone morphogenetic protein receptors). B-cells also mediate their effects through enhanced production of pro-inflammatory cytokines, reduced anti-inflammatory properties by regulatory B-cells, immunoglobulin (Ig)G-induced complement activation, and IgE-induced mast cell activation. Precision-medicine approaches targeting B-cell immunity are a promising direction for select PAH conditions, as suggested by the efficacy of anti-CD20 therapy in experimental models and a trial of rituximab in systemic sclerosis-associated PAH.
Collapse
Affiliation(s)
- Sébastien Sanges
- Univ. Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
- INSERM, F-59000 Lille, France
- CHU Lille, Département de Médecine Interne et Immunologie Clinique, F-59000 Lille, France
- Centre National de Référence Maladies Auto-immunes Systémiques Rares du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), F-59000 Lille, France
- Health Care Provider of the European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases Network (ReCONNET), F-59000 Lille, France
- Veteran Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, School of Medicine, Stanford, CA, USA
- Both authors contributed equally and share co-first authorship
| | - Wen Tian
- Veteran Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, School of Medicine, Stanford, CA, USA
- Both authors contributed equally and share co-first authorship
| | - Sylvain Dubucquoi
- Univ. Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
- INSERM, F-59000 Lille, France
- CHU Lille, Institut d'Immunologie, Pôle de Biologie Pathologie Génétique, F-59000 Lille, France
| | - Jason L Chang
- Veteran Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, School of Medicine, Stanford, CA, USA
| | - Aurore Collet
- Univ. Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
- INSERM, F-59000 Lille, France
- CHU Lille, Institut d'Immunologie, Pôle de Biologie Pathologie Génétique, F-59000 Lille, France
| | - David Launay
- Univ. Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
- INSERM, F-59000 Lille, France
- CHU Lille, Département de Médecine Interne et Immunologie Clinique, F-59000 Lille, France
- Centre National de Référence Maladies Auto-immunes Systémiques Rares du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), F-59000 Lille, France
- Health Care Provider of the European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases Network (ReCONNET), F-59000 Lille, France
- Veteran Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, School of Medicine, Stanford, CA, USA
- Both authors contributed equally and share co-last authorship
| | - Mark R Nicolls
- Veteran Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, School of Medicine, Stanford, CA, USA
- Both authors contributed equally and share co-last authorship
| |
Collapse
|
40
|
Deng Z, Fan T, Xiao C, Tian H, Zheng Y, Li C, He J. TGF-β signaling in health, disease, and therapeutics. Signal Transduct Target Ther 2024; 9:61. [PMID: 38514615 PMCID: PMC10958066 DOI: 10.1038/s41392-024-01764-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: 12/07/2022] [Revised: 08/31/2023] [Accepted: 01/31/2024] [Indexed: 03/23/2024] Open
Abstract
Transforming growth factor (TGF)-β is a multifunctional cytokine expressed by almost every tissue and cell type. The signal transduction of TGF-β can stimulate diverse cellular responses and is particularly critical to embryonic development, wound healing, tissue homeostasis, and immune homeostasis in health. The dysfunction of TGF-β can play key roles in many diseases, and numerous targeted therapies have been developed to rectify its pathogenic activity. In the past decades, a large number of studies on TGF-β signaling have been carried out, covering a broad spectrum of topics in health, disease, and therapeutics. Thus, a comprehensive overview of TGF-β signaling is required for a general picture of the studies in this field. In this review, we retrace the research history of TGF-β and introduce the molecular mechanisms regarding its biosynthesis, activation, and signal transduction. We also provide deep insights into the functions of TGF-β signaling in physiological conditions as well as in pathological processes. TGF-β-targeting therapies which have brought fresh hope to the treatment of relevant diseases are highlighted. Through the summary of previous knowledge and recent updates, this review aims to provide a systematic understanding of TGF-β signaling and to attract more attention and interest to this research area.
Collapse
Affiliation(s)
- Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - He Tian
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yujia Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
41
|
Mostkowska A, Rousseau G, Raynal NJM. Repurposing of rituximab biosimilars to treat B cell mediated autoimmune diseases. FASEB J 2024; 38:e23536. [PMID: 38470360 DOI: 10.1096/fj.202302259rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 03/13/2024]
Abstract
Rituximab, the first monoclonal antibody approved for the treatment of lymphoma, eventually became one of the most popular and versatile drugs ever in terms of clinical application and revenue. Since its patent expiration, and consequently, the loss of exclusivity of the original biologic, its repurposing as an off-label drug has increased dramatically, propelled by the development and commercialization of its many biosimilars. Currently, rituximab is prescribed worldwide to treat a vast range of autoimmune diseases mediated by B cells. Here, we present a comprehensive overview of rituximab repurposing in 115 autoimmune diseases across 17 medical specialties, sourced from over 1530 publications. Our work highlights the extent of its off-label use and clinical benefits, underlining the success of rituximab repurposing for both common and orphan immune-related diseases. We discuss the scientific mechanism associated with its clinical efficacy and provide additional indications for which rituximab could be investigated. Our study presents rituximab as a flagship example of drug repurposing owing to its central role in targeting cluster of differentiate 20 positive (CD20) B cells in 115 autoimmune diseases.
Collapse
Affiliation(s)
- Agata Mostkowska
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Guy Rousseau
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Noël J-M Raynal
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
- Centre de recherche du CHU Sainte-Justine, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
42
|
Wu P, Song Y, Chen Z, Xia J, Zhou Y. Changes of B cell subsets in different types of diabetes and its effect on the progression of latent autoimmune diabetes in adults. Endocrine 2024; 83:624-635. [PMID: 37755622 DOI: 10.1007/s12020-023-03539-9] [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: 06/25/2023] [Accepted: 09/13/2023] [Indexed: 09/28/2023]
Abstract
PURPOSE Developmental abnormalities in B cells is one of the key players in autoimmune diabetes, but little is known about its role in latent autoimmune diabetes in adults (LADA). This study aimed to investigate the distribution of B cell subsets in different types of diabetes and to analyze their correlations with other biochemical parameters. METHODS A total of 140 participants were prospectively enrolled from January 2021 to December 2022. Diabetes-related autoantibodies and laboratory indicators were tested. Flow cytometry was used to analyze the percentage of circulating B cell subsets and T follicular cells. The correlation of B cell subsets with different indicators was assessed by Spearman's correlation method. RESULTS We observed that the Naïve phenotype cells tended to be less frequent in patients with diabetes than in healthy controls. The frequency of plasmablasts (PB) and Breg cell-related phenotype (B10) were significantly higher in LADA. Notably, the percentage of PB was positively associated with levels of islet cell antibody (ICA) and insulin autoantibody (IAA), but inversely associated with fasting C-peptide (FCP), further indicating that PB may promote the destruction of β-cell in patients with diabetes. CONCLUSIONS This study showed that patients with LADA had significantly altered frequencies of B cell subsets, particularly in the naïve to memory B cell ratio. Our study provided valuable information on the distribution characteristics of B cell subsets in LADA and suggested the feasibility of B-cell targeted therapy in LADA patients.
Collapse
Affiliation(s)
- Peihao Wu
- Department of Clinical Laboratory, Women's Hospital, School of Medicine Zhejiang University, Hangzhou, Zhejiang, 310006, China
| | - Yingxiang Song
- Geriatric Medicine Center, Department of Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Zhuo Chen
- Department of Clinical Laboratory, Hangzhou Lin'an Third People's Hospital, Hangzhou, Zhejiang, 311311, China
| | - Jun Xia
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China.
| | - Yu Zhou
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China.
- Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang province, No.182 Tianmushan Road, Xihu District, Hangzhou, Zhejiang, 310063, China.
| |
Collapse
|
43
|
Woodruff MC, Faliti CE, Sanz I. Systems biology of B cells in COVID-19. Semin Immunol 2024; 72:101875. [PMID: 38489999 DOI: 10.1016/j.smim.2024.101875] [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: 01/29/2024] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 03/17/2024]
Abstract
The integration of multi-'omic datasets into complex systems-wide assessments has become a mainstay in immunologic investigation. This focus on high-dimensional data collection and analysis was on full display in the investigation of COVID-19, the respiratory illness resulting from infection by the novel coronavirus SARS-CoV-2. Particularly in the area of B cell biology, tremendous efforts in both cellular and serologic investigation have resulted in an increasingly detailed mapping of the coordinated effector, memory, and antibody secreting cell responses that underpin the development of humoral immunity in response to primary viral infection. Further, the rapid development and deployment of effective vaccines has allowed for the assessment of developing memory responses across a wide variety of immune contexts, including in patients with compromised immune function. The result has been a period of rapid gains in the understanding of B cell biology unrestricted to the study of COVID-19. Here, we outline the systems-level technologies that have been routinely implemented in these investigations throughout the pandemic, and discuss how their use has led to clear and applicable gains in pursuance of the amelioration of human infectious disease and beyond.
Collapse
Affiliation(s)
- Matthew C Woodruff
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA; Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA.
| | - Caterina E Faliti
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA; Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA.
| | - Ignacio Sanz
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA; Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| |
Collapse
|
44
|
Shyamsundar S, Pierson SK, Connolly CM, Teles M, Segev DL, Werbel WA, van Rhee F, Casper C, Brandstadter JD, Noy A, Fajgenbaum DC. Castleman disease patients report mild COVID-19 symptoms and mount a humoral response to SARS-CoV-2 vaccination. BLOOD NEOPLASIA 2024; 1:100002. [PMID: 39044861 PMCID: PMC11265787 DOI: 10.1016/j.bneo.2024.100002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
The coronavirus disease of 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has resulted in increased morbidity and mortality in patients with impaired immunity, hematologic malignancies, and immunosuppressive regimens. COVID-19 can cause a cytokine storm with some patients benefiting from blockade of the pro-inflammatory cytokine, interleukin 6 (IL6). As Castleman disease (CD) is an atypical lymphoproliferative disorder that can involve a cytokine storm and often requires immunosuppressive therapies, including IL6 inhibition, we sought to evaluate outcomes following COVID-19 and SARS-CoV-2 vaccination in CD patients. We administered a survey in April 2021 to characterize experiences with COVID-19 and SARS-CoV-2 vaccination among 300 CD patients enrolled in ACCELERATE, a natural history registry of CD patients. Among 128 respondents, the prevalence of SARS-CoV-2 infection (16/95, 17%), severe disease (1/16, 6%), vaccination rates (112/128, 88%), and vaccine adverse effects after dose one (62/112, 55%) were comparable to the general U.S. population. While there were two cases of CD flares occurring shortly after SARS-CoV-2 infection (N=1) and vaccination (N=1), over 100 patients in this study that were infected and/or vaccinated did not experience CD flares. The median anti-spike titer six months after the second dose among CD patients was comparable to individuals with other immune-related diseases and healthy populations. Data from this small cohort suggest that, despite being on immunosuppressive therapies, CD patients do not appear to be at increased risk of poor COVID-19 outcomes and can mount a humoral response to SARS-CoV-2 vaccination. This study was registered on clinicaltrials.gov (#NCT02817997).
Collapse
Affiliation(s)
- Saishravan Shyamsundar
- Center for Cytokine Storm Treatment & Laboratory, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sheila K. Pierson
- Center for Cytokine Storm Treatment & Laboratory, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Caoilfhionn M. Connolly
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Mayan Teles
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Dorry L. Segev
- Department of Surgery, NYU Grossman School of Medicine, New York, New York, 10016, USA
| | - William A. Werbel
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Frits van Rhee
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Corey Casper
- Access to Advanced Health Institute, Seattle, WA, 98102, USA
- Department of Global Health, University of Washington, Seattle, WA, 98105, USA
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Joshua D. Brandstadter
- Center for Cytokine Storm Treatment & Laboratory, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ariela Noy
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Weill Cornell Medical College, New York, NY, 10065, USA
| | - David C. Fajgenbaum
- Center for Cytokine Storm Treatment & Laboratory, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
45
|
Huang F, Zhang J, Zhou H, Qu T, Wang Y, Jiang K, Liu Y, Xu Y, Chen M, Chen L. B cell subsets contribute to myocardial protection by inducing neutrophil apoptosis after ischemia and reperfusion. JCI Insight 2024; 9:e167201. [PMID: 38290007 DOI: 10.1172/jci.insight.167201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/17/2024] [Indexed: 02/01/2024] Open
Abstract
A robust, sterile inflammation underlies myocardial ischemia and reperfusion injury (MIRI). Several subsets of B cells possess the immunoregulatory capacity that limits tissue damage, yet the role of B cells in MIRI remains elusive. Here, we sought to elucidate the contribution of B cells to MIRI by transient ligation of the left anterior descending coronary artery in B cell-depleted or -deficient mice. Following ischemia and reperfusion (I/R), regulatory B cells are rapidly recruited to the heart. B cell-depleted or -deficient mice exhibited exacerbated tissue damage, adverse cardiac remodeling, and an augmented inflammatory response after I/R. Rescue and chimeric experiments indicated that the cardioprotective effect of B cells was not solely dependent on IL-10. Coculture experiments demonstrated that B cells induced neutrophil apoptosis through contact-dependent interactions, subsequently promoting reparative macrophage polarization by facilitating the phagocytosis of neutrophils by macrophages. The in vivo cardioprotective effect of B cells was undetectable in the absence of neutrophils after I/R. Mechanistically, ligand-receptor imputation identified FCER2A as a potential mediator of interactions between B cells and neutrophils. Blocking FCER2A on B cells resulted in a reduction in the percentage of apoptotic neutrophils, contributing to the deterioration of cardiac remodeling. Our findings unveil a potential cardioprotective role of B cells in MIRI through mechanisms involving FCER2A, neutrophils, and macrophages.
Collapse
Affiliation(s)
- Fangyang Huang
- Department of Cardiology
- State Key Laboratory of Biotherapy and Cancer Center
- Laboratory of Heart Valve Disease
| | - Jialiang Zhang
- Department of Cardiology
- Laboratory of Heart Valve Disease
| | - Hao Zhou
- Laboratory of Heart Valve Disease
| | | | - Yan Wang
- Department of Cardiology
- Laboratory of Heart Valve Disease
| | - Kexin Jiang
- Department of Cardiology
- West China School of Medicine, and
| | | | | | - Mao Chen
- Department of Cardiology
- Laboratory of Heart Valve Disease
| | - Li Chen
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
46
|
Liu J, Zhang B, Zhang G, Shang D. Reprogramming of regulatory T cells in inflammatory tumor microenvironment: can it become immunotherapy turning point? Front Immunol 2024; 15:1345838. [PMID: 38449875 PMCID: PMC10915070 DOI: 10.3389/fimmu.2024.1345838] [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: 11/28/2023] [Accepted: 01/29/2024] [Indexed: 03/08/2024] Open
Abstract
Overcoming the immunosuppressive tumor microenvironment and identifying widely used immunosuppressants with minimal side effects are two major challenges currently hampering cancer immunotherapy. Regulatory T cells (Tregs) are present in almost all cancer tissues and play an important role in preserving autoimmune tolerance and tissue homeostasis. The tumor inflammatory microenvironment causes the reprogramming of Tregs, resulting in the conversion of Tregs to immunosuppressive phenotypes. This process ultimately facilitates tumor immune escape or tumor progression. However, current systemic Treg depletion therapies may lead to severe autoimmune toxicity. Therefore, it is crucial to understand the mechanism of Treg reprogramming and develop immunotherapies that selectively target Tregs within tumors. This article provides a comprehensive review of the potential mechanisms involved in Treg cell reprogramming and explores the application of Treg cell immunotherapy. The interference with reprogramming pathways has shown promise in reducing the number of tumor-associated Tregs or impairing their function during immunotherapy, thereby improving anti-tumor immune responses. Furthermore, a deeper understanding of the mechanisms that drive Treg cell reprogramming could reveal new molecular targets for future treatments.
Collapse
Affiliation(s)
- Jinming Liu
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Biao Zhang
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guolin Zhang
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Dong Shang
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| |
Collapse
|
47
|
Veh J, Ludwig C, Schrezenmeier H, Jahrsdörfer B. Regulatory B Cells-Immunopathological and Prognostic Potential in Humans. Cells 2024; 13:357. [PMID: 38391970 PMCID: PMC10886933 DOI: 10.3390/cells13040357] [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: 12/27/2023] [Revised: 02/05/2024] [Accepted: 02/10/2024] [Indexed: 02/24/2024] Open
Abstract
The aim of the following review is to shed light on the putative role of regulatory B cells (Bregs) in various human diseases and highlight their potential prognostic and therapeutic relevance in humans. Regulatory B cells are a heterogeneous group of B lymphocytes capable of suppressing inflammatory immune reactions. In this way, Bregs contribute to the maintenance of tolerance and immune homeostasis by limiting ongoing immune reactions temporally and spatially. Bregs play an important role in attenuating pathological inflammatory reactions that can be associated with transplant rejection, graft-versus-host disease, autoimmune diseases and allergies but also with infectious, neoplastic and metabolic diseases. Early studies of Bregs identified IL-10 as an important functional molecule, so the IL-10-secreting murine B10 cell is still considered a prototype Breg, and IL-10 has long been central to the search for human Breg equivalents. However, over the past two decades, other molecules that may contribute to the immunosuppressive function of Bregs have been discovered, some of which are only present in human Bregs. This expanded arsenal includes several anti-inflammatory cytokines, such as IL-35 and TGF-β, but also enzymes such as CD39/CD73, granzyme B and IDO as well as cell surface proteins including PD-L1, CD1d and CD25. In summary, the present review illustrates in a concise and comprehensive manner that although human Bregs share common functional immunosuppressive features leading to a prominent role in various human immunpathologies, they are composed of a pool of different B cell types with rather heterogeneous phenotypic and transcriptional properties.
Collapse
Affiliation(s)
- Johanna Veh
- Institute for Transfusion Medicine, Ulm University Hospitals and Clinics, 89081 Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Donation Service Baden-Württemberg-Hessen, 89081 Ulm, Germany
| | - Carolin Ludwig
- Institute for Transfusion Medicine, Ulm University Hospitals and Clinics, 89081 Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Donation Service Baden-Württemberg-Hessen, 89081 Ulm, Germany
| | - Hubert Schrezenmeier
- Institute for Transfusion Medicine, Ulm University Hospitals and Clinics, 89081 Ulm, Germany
| | - Bernd Jahrsdörfer
- Institute for Transfusion Medicine, Ulm University Hospitals and Clinics, 89081 Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Donation Service Baden-Württemberg-Hessen, 89081 Ulm, Germany
| |
Collapse
|
48
|
Wang Y, Chen S, Bao S, Yao L, Wen Z, Xu L, Chen X, Guo S, Pang H, Zhou Y, Zhou P. Deciphering the fibrotic process: mechanism of chronic radiation skin injury fibrosis. Front Immunol 2024; 15:1338922. [PMID: 38426100 PMCID: PMC10902513 DOI: 10.3389/fimmu.2024.1338922] [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: 11/16/2023] [Accepted: 02/02/2024] [Indexed: 03/02/2024] Open
Abstract
This review explores the mechanisms of chronic radiation-induced skin injury fibrosis, focusing on the transition from acute radiation damage to a chronic fibrotic state. It reviewed the cellular and molecular responses of the skin to radiation, highlighting the role of myofibroblasts and the significant impact of Transforming Growth Factor-beta (TGF-β) in promoting fibroblast-to-myofibroblast transformation. The review delves into the epigenetic regulation of fibrotic gene expression, the contribution of extracellular matrix proteins to the fibrotic microenvironment, and the regulation of the immune system in the context of fibrosis. Additionally, it discusses the potential of biomaterials and artificial intelligence in medical research to advance the understanding and treatment of radiation-induced skin fibrosis, suggesting future directions involving bioinformatics and personalized therapeutic strategies to enhance patient quality of life.
Collapse
Affiliation(s)
- Yiren Wang
- School of Nursing, Southwest Medical University, Luzhou, China
- Wound Healing Basic Research and Clinical Application Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Shouying Chen
- School of Nursing, Southwest Medical University, Luzhou, China
- Wound Healing Basic Research and Clinical Application Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Shuilan Bao
- School of Nursing, Southwest Medical University, Luzhou, China
- Wound Healing Basic Research and Clinical Application Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Li Yao
- School of Nursing, Southwest Medical University, Luzhou, China
- Wound Healing Basic Research and Clinical Application Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Zhongjian Wen
- School of Nursing, Southwest Medical University, Luzhou, China
- Wound Healing Basic Research and Clinical Application Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Lixia Xu
- School of Nursing, Southwest Medical University, Luzhou, China
| | - Xiaoman Chen
- School of Nursing, Southwest Medical University, Luzhou, China
| | - Shengmin Guo
- Department of Nursing, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Haowen Pang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yun Zhou
- School of Medical Information and Engineering, Southwest Medical University, Luzhou, China
| | - Ping Zhou
- Wound Healing Basic Research and Clinical Application Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
49
|
Mititelu A, Onisâi MC, Roșca A, Vlădăreanu AM. Current Understanding of Immune Thrombocytopenia: A Review of Pathogenesis and Treatment Options. Int J Mol Sci 2024; 25:2163. [PMID: 38396839 PMCID: PMC10889445 DOI: 10.3390/ijms25042163] [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: 12/21/2023] [Revised: 01/24/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
The management of immune thrombocytopenia (ITP) and the prediction of patient response to therapy still represent a significant and constant challenge in hematology. ITP is a heterogeneous disease with an unpredictable evolution. Although the pathogenesis of ITP is currently better known and its etiology has been extensively studied, up to 75% of adult patients with ITP may develop chronicity, which represents a significant burden on patients' quality of life. A major risk of ITP is bleeding, but knowledge on the exact relationship between the degree of thrombocytopenia and bleeding symptoms, especially at a lower platelet count, is lacking. The actual management of ITP is based on immune suppression (corticosteroids and intravenous immunoglobulins), or the use of thrombopoietin receptor agonists (TPO-RAs), rituximab, or spleen tyrosine kinase (Syk) inhibitors. A better understanding of the underlying pathology has facilitated the development of a number of new targeted therapies (Bruton's tyrosine kinase inhibitors, neonatal Fc receptors, strategies targeting B and plasma cells, strategies targeting T cells, complement inhibitors, and newer TPO-RAs for improving megakaryopoiesis), which seem to be highly effective and well tolerated and result in a significant improvement in patients' quality of life. The disadvantage is that there is a lack of knowledge of the predictive factors of response to treatments, which would help in the development of an optimized treatment algorithm for selected patients.
Collapse
Affiliation(s)
- Alina Mititelu
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, Emergency University Hospital of Bucharest, 050098 Bucharest, Romania; (M.-C.O.); (A.M.V.)
| | - Minodora-Cezarina Onisâi
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, Emergency University Hospital of Bucharest, 050098 Bucharest, Romania; (M.-C.O.); (A.M.V.)
| | - Adrian Roșca
- Department of Physiology, Carol Davila University of Medicine and Pharmacy, 050471 Bucharest, Romania;
| | - Ana Maria Vlădăreanu
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, Emergency University Hospital of Bucharest, 050098 Bucharest, Romania; (M.-C.O.); (A.M.V.)
| |
Collapse
|
50
|
Tieck MP, Vasilenko N, Ruschil C, Kowarik MC. Peripheral memory B cells in multiple sclerosis vs. double negative B cells in neuromyelitis optica spectrum disorder: disease driving B cell subsets during CNS inflammation. Front Cell Neurosci 2024; 18:1337339. [PMID: 38385147 PMCID: PMC10879280 DOI: 10.3389/fncel.2024.1337339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 01/16/2024] [Indexed: 02/23/2024] Open
Abstract
B cells are fundamental players in the pathophysiology of autoimmune diseases of the central nervous system, such as multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD). A deeper understanding of disease-specific B cell functions has led to the differentiation of both diseases and the development of different treatment strategies. While NMOSD is strongly associated with pathogenic anti-AQP4 IgG antibodies and proinflammatory cytokine pathways, no valid autoantibodies have been identified in MS yet, apart from certain antigen targets that require further evaluation. Although both diseases can be effectively treated with B cell depleting therapies, there are distinct differences in the peripheral B cell subsets that influence CNS inflammation. An increased peripheral blood double negative B cells (DN B cells) and plasmablast populations has been demonstrated in NMOSD, but not consistently in MS patients. Furthermore, DN B cells are also elevated in rheumatic diseases and other autoimmune entities such as myasthenia gravis and Guillain-Barré syndrome, providing indirect evidence for a possible involvement of DN B cells in other autoantibody-mediated diseases. In MS, the peripheral memory B cell pool is affected by many treatments, providing indirect evidence for the involvement of memory B cells in MS pathophysiology. Moreover, it must be considered that an important effector function of B cells in MS may be the presentation of antigens to peripheral immune cells, including T cells, since B cells have been shown to be able to recirculate in the periphery after encountering CNS antigens. In conclusion, there are clear differences in the composition of B cell populations in MS and NMOSD and treatment strategies differ, with the exception of broad B cell depletion. This review provides a detailed overview of the role of different B cell subsets in MS and NMOSD and their implications for treatment options. Specifically targeting DN B cells and plasmablasts in NMOSD as opposed to memory B cells in MS may result in more precise B cell therapies for both diseases.
Collapse
Affiliation(s)
| | | | | | - M. C. Kowarik
- Department of Neurology and Stroke, Center for Neurology, and Hertie-Institute for Clinical Brain Research Eberhard-Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|