1
|
Cáceres E, Olivella JC, Di Napoli M, Raihane AS, Divani AA. Immune Response in Traumatic Brain Injury. Curr Neurol Neurosci Rep 2024; 24:593-609. [PMID: 39467990 PMCID: PMC11538248 DOI: 10.1007/s11910-024-01382-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2024] [Indexed: 10/30/2024]
Abstract
PURPOSE OF REVIEW This review aims to comprehensively examine the immune response following traumatic brain injury (TBI) and how its disruption can impact healing and recovery. RECENT FINDINGS The immune response is now considered a key element in the pathophysiology of TBI, with consequences far beyond the acute phase after injury. A delicate equilibrium is crucial for a healthy recovery. When this equilibrium is disrupted, chronic inflammation and immune imbalance can lead to detrimental effects on survival and disability. Globally, traumatic brain injury (TBI) imposes a substantial burden in terms of both years of life lost and years lived with disability. Although its epidemiology exhibits dynamic trends over time and across regions, TBI disproportionally affects the younger populations, posing psychosocial and financial challenge for communities and families. Following the initial trauma, the primary injury is succeeded by an inflammatory response, primarily orchestrated by the innate immune system. The inflammasome plays a pivotal role during this stage, catalyzing both programmed cell death pathways and the up-regulation of inflammatory cytokines and transcription factors. These events trigger the activation and differentiation of microglia, thereby intensifying the inflammatory response to a systemic level and facilitating the migration of immune cells and edema. This inflammatory response, initially originated in the brain, is monitored by our autonomic nervous system. Through the vagus nerve and adrenergic and cholinergic receptors in various peripheral lymphoid organs and immune cells, bidirectional communication and regulation between the immune and nervous systems is established.
Collapse
Affiliation(s)
- Eder Cáceres
- Unisabana Center for Translational Science, Universidad de La Sabana, Chía, Colombia.
- School of Medicine, Universidad de La Sabana, Chía, Colombia.
- Bioscience PhD. School of Engineering, Universidad de La Sabana, Chía, Colombia.
| | | | - Mario Di Napoli
- Neurological Service, SS Annunziata Hospital, Sulmona, L'Aquila, Italy
| | - Ahmed S Raihane
- School of Medicine, University of New Mexico, Albuquerque, NM, USA
- Department of Neurology, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Afshin A Divani
- Department of Neurology, University of New Mexico Health Science Center, Albuquerque, NM, USA
| |
Collapse
|
2
|
Du Y, Xie J, Liu D, Zhao J, Chen P, He X, Hong P, Fu Y, Hong Y, Liu WH, Xiao C. Critical and differential roles of eIF4A1 and eIF4A2 in B-cell development and function. Cell Mol Immunol 2024:10.1038/s41423-024-01234-x. [PMID: 39516355 DOI: 10.1038/s41423-024-01234-x] [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: 07/16/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
Eukaryotic initiation factor 4 A (eIF4A) plays critical roles during translation initiation of cellular mRNAs by forming the cap-binding eIF4F complex, recruiting the 40S small ribosome subunit, and scanning the 5' untranslated region (5' UTR) for the start codon. eIF4A1 and eIF4A2, two isoforms of eIF4A, are highly conserved and exchange freely within eIF4F complexes. The understanding of their biological and molecular functions remains incomplete if not fragmentary. In this study, we showed that eIF4A1 and eIF4A2 exhibit different expression patterns during B-cell development and activation. Mouse genetic analyses showed that they play critical but differential roles during B-cell development and humoral immune responses. While eIF4A1 controls global protein synthesis, eIF4A2 regulates the biogenesis of 18S ribosomal RNA and the 40S ribosome subunit. This study demonstrates the distinct cellular and molecular functions of eIF4A1 and eIF4A2 and reveals a new role of eIF4A2 in controlling 40S ribosome biogenesis.
Collapse
Affiliation(s)
- Ying Du
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Jun Xie
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Dewang Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Jiayi Zhao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Pengda Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Xiaoyu He
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Peicheng Hong
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yubing Fu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yazhen Hong
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Wen-Hsien Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Changchun Xiao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China.
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA.
- Sanofi Institute for Biomedical Research, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
3
|
Zhong Z, Quiñones-Pérez M, Dai Z, Juarez VM, Bhatia E, Carlson CR, Shah SB, Patel A, Fang Z, Hu T, Allam M, Hicks SL, Gupta M, Gupta SL, Weeks E, Vagelos SD, Molina A, Mulero-Russe A, Mora-Boza A, Joshi DJ, Sekaly RP, Sulchek T, Goudy SL, Wrammert J, Roy K, Boss JM, Coskun AF, Scharer CD, García AJ, Koff JL, Singh A. Human immune organoids to decode B cell response in healthy donors and patients with lymphoma. NATURE MATERIALS 2024:10.1038/s41563-024-02037-1. [PMID: 39506098 DOI: 10.1038/s41563-024-02037-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/30/2024] [Indexed: 11/08/2024]
Abstract
Antibodies are produced when naive B cells differentiate into plasma cells within germinal centres (GCs) of lymphoid tissues. Patients with B cell lymphoma on effective immunotherapies exhibit diminished antibody production, leading to higher infection rates and reduced vaccine efficacy, even after B cell recovery. Current ex vivo models fail to sustain long-term GC reactions and effectively test B cell responses. Here we developed synthetic hydrogels mimicking the lymphoid tissue microenvironment, enabling human GCs from tonsils and peripheral blood mononuclear cell-derived B cells. Immune organoids derived from peripheral blood mononuclear cells maintain GC B cells and plasma cells longer than tonsil-derived ones and exhibit unique B cell programming, including GC compartments, somatic hypermutation, immunoglobulin class switching and B cell clones. Chemical inhibition of transcriptional and epigenetic processes enhances plasma cell formation. While integrating polarized CXCL12 protein in a lymphoid organ-on-chip modulates GC responses in healthy donor B cells, it fails with B cells derived from patients with lymphoma. Our system allows rapid, controlled modelling of immune responses and B cell disorders.
Collapse
Affiliation(s)
- Zhe Zhong
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Manuel Quiñones-Pérez
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Zhonghao Dai
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Valeria M Juarez
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Eshant Bhatia
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Christopher R Carlson
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Shivem B Shah
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Anjali Patel
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Zhou Fang
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Thomas Hu
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Mayar Allam
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Sakeenah L Hicks
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Mansi Gupta
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Sneh Lata Gupta
- Emory Vaccine Center, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Ethan Weeks
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Stephanie D Vagelos
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Alejandro Molina
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Adriana Mulero-Russe
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Ana Mora-Boza
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Devyani J Joshi
- Emory Vaccine Center, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Rafick P Sekaly
- Emory Vaccine Center, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Todd Sulchek
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Steven L Goudy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Department of Otolaryngology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Jens Wrammert
- Emory Vaccine Center, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Krishnendu Roy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jeremy M Boss
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Ahmet F Coskun
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Andrés J García
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Jean L Koff
- Winship Cancer Center, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Ankur Singh
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA.
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
4
|
Zhang X, Ma C, Lu Y, Wang J, Yun H, Jiang H, Wu M, Feng X, Gai W, Xu G, Deng H, Feng J, Liu W, Shi T, Cheng Q, Zhang J. Rack1 regulates B-cell development and function by binding to and stabilizing the transcription factor Pax5. Cell Mol Immunol 2024; 21:1282-1295. [PMID: 39256480 PMCID: PMC11528059 DOI: 10.1038/s41423-024-01213-2] [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: 12/24/2023] [Accepted: 08/26/2024] [Indexed: 09/12/2024] Open
Abstract
The transcription factor Pax5 activates genes essential for B-cell development and function. However, the regulation of Pax5 expression remains elusive. The adaptor Rack1 can interact with multiple transcription factors and modulate their activation and/or stability. However, its role in the transcriptional control of B-cell fates is largely unknown. Here, we show that CD19-driven Rack1 deficiency leads to pro-B accumulation and a simultaneous reduction in B cells at later developmental stages. The generation of bone marrow chimeras indicates a cell-intrinsic role of Rack1 in B-cell homeostasis. Moreover, Rack1 augments BCR and TLR signaling in mature B cells. On the basis of the aberrant expression of Pax5-regulated genes, including CD19, upon Rack1 deficiency, further exploration revealed that Rack1 maintains the protein level of Pax5 through direct interaction and consequently prevents Pax5 ubiquitination. Accordingly, Mb1-driven Rack1 deficiency almost completely blocks B-cell development at the pro-B-cell stage. Ectopic expression of Pax5 in Rack1-deficient pro-B cells partially rescues B-cell development. Thus, Rack1 regulates B-cell development and function through, at least partially, binding to and stabilizing Pax5.
Collapse
Affiliation(s)
- Xueting Zhang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Chenke Ma
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yuchen Lu
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jing Wang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Hongfang Yun
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Hui Jiang
- Beijing Institute of Basic Medical Sciences, Beijing, China
- University of South China, Hengyang Medical School, Hengyang, China
| | - Mengyao Wu
- Beijing Institute of Basic Medical Sciences, Beijing, China
- Anhui Medical University, Hefei, China
| | - Xiaoyao Feng
- Beijing Institute of Basic Medical Sciences, Beijing, China
- Henan University Joint National Laboratory for Antibody Drug Engineering, Kaifeng, China
| | - Wenbin Gai
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Guanglei Xu
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Hongbin Deng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiannan Feng
- Henan University Joint National Laboratory for Antibody Drug Engineering, Kaifeng, China
- National Engineering Research Center for Emergence Drugs, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Wanli Liu
- Institute of Life Sciences, Tsinghua University, Beijing, China
| | - Taoxing Shi
- Beijing Institute of Basic Medical Sciences, Beijing, China.
| | - Qianqian Cheng
- Beijing Institute of Basic Medical Sciences, Beijing, China.
| | - Jiyan Zhang
- Beijing Institute of Basic Medical Sciences, Beijing, China.
- University of South China, Hengyang Medical School, Hengyang, China.
- Anhui Medical University, Hefei, China.
- Henan University Joint National Laboratory for Antibody Drug Engineering, Kaifeng, China.
- Chinese Institute for Brain Research, Beijing, China.
| |
Collapse
|
5
|
Pyuza JJ, van Dorst MM, Stam K, Wammes L, König M, Kullaya VI, Kruize Y, Huisman W, Andongolile N, Ngowi A, Shao ER, Mremi A, Hogendoorn PC, Msuya SE, Jochems SP, de Steenhuijsen Piters WA, Yazdanbakhsh M. Lifestyle score is associated with cellular immune profiles in healthy Tanzanian adults. Brain Behav Immun Health 2024; 41:100863. [PMID: 39398291 PMCID: PMC11470418 DOI: 10.1016/j.bbih.2024.100863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/31/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024] Open
Abstract
Immune system and vaccine responses vary across geographical locations worldwide, not only between high and low-middle income countries (LMICs), but also between rural and urban populations within the same country. Lifestyle factors such as housing conditions, exposure to microorganisms and parasites and diet are associated with rural-and urban-living. However, the relationships between these lifestyle factors and immune profiles have not been mapped in detail. Here, we profiled the immune system of 100 healthy Tanzanians living across four rural/urban areas using mass cytometry. We developed a lifestyle score based on an individual's household assets, housing condition and recent dietary history and studied the association with cellular immune profiles. Seventeen out of 80 immune cell clusters were associated with living location or lifestyle score, with eight identifiable only using lifestyle score. Individuals with low lifestyle score, most of whom live in rural settings, showed higher frequencies of NK cells, plasmablasts, atypical memory B cells, T helper 2 cells, regulatory T cells and activated CD4+ T effector memory cells expressing CD38, HLA-DR and CTLA-4. In contrast, those with high lifestyle score, most of whom live in urban areas, showed a less activated state of the immune system illustrated by higher frequencies of naïve CD8+ T cells. Using an elastic net machine learning model, we identified cellular immune signatures most associated with lifestyle score. Assuming a link between these immune profiles and vaccine responses, these signatures may inform us on the cellular mechanisms underlying poor responses to vaccines, but also reduced autoimmunity and allergies in low- and middle-income countries.
Collapse
Affiliation(s)
- Jeremia J. Pyuza
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
- Department of Pathology, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
- Institute of Public Health, Kilimanjaro Christian University Medical College (KCMUCo), Moshi, Tanzania
- Kilimanjaro Clinical Research Institute (KCRI), Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Marloes M.A.R. van Dorst
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Koen Stam
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Linda Wammes
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Marion König
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Vesla I. Kullaya
- Kilimanjaro Clinical Research Institute (KCRI), Kilimanjaro Christian Medical Centre, Moshi, Tanzania
- Department of Medical Biochemistry and Molecular Biology, Kilimanjaro Christian Medical College (KCMUCo), Moshi, Tanzania
| | - Yvonne Kruize
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Wesley Huisman
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Nikuntufya Andongolile
- Department of Community Medicine, Kilimanjaro Christian Medical Centre (KCMC), Moshi, Tanzania
| | - Anastazia Ngowi
- Department of Community Medicine, Kilimanjaro Christian Medical Centre (KCMC), Moshi, Tanzania
| | - Elichilia R. Shao
- Department of Internal Medicine, Kilimanjaro Christian Medical University College (KCMUCo), Moshi, Tanzania
- Department of Internal Medicine, Kilimanjaro Christian Medical Centre (KCMC), Moshi, Tanzania
| | - Alex Mremi
- Department of Pathology, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | | | - Sia E. Msuya
- Institute of Public Health, Kilimanjaro Christian University Medical College (KCMUCo), Moshi, Tanzania
- Department of Community Medicine, Kilimanjaro Christian Medical Centre (KCMC), Moshi, Tanzania
| | - Simon P. Jochems
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | | | - Maria Yazdanbakhsh
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| |
Collapse
|
6
|
Bazarbachi AH, Avet-Loiseau H, Harousseau JL, Bazarbachi A, Mohty M. Precision medicine for multiple myeloma: The case for translocation (11;14). Cancer Treat Rev 2024; 130:102823. [PMID: 39255732 DOI: 10.1016/j.ctrv.2024.102823] [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: 06/09/2024] [Revised: 08/11/2024] [Accepted: 09/02/2024] [Indexed: 09/12/2024]
Abstract
The t(11;14) translocation is among the most prevalent cytogenetic abnormalities in multiple myeloma (MM), distinguished by its unique features and biology that have been thoroughly explored for decades. What further sets this MM subtype apart is its oscillating prognostic significance, from initially being considered a favorable alteration to intermediate risk and potential future reclassification as favorable risk. Despite not being inherently a high-risk alteration indicative of an aggressive phenotype, it appears that t(11;14)-MM is less responsive to novel agents like proteasome inhibitors and immunomodulatory drugs which have otherwise transformed the disease's treatment landscape, perhaps partially explained by its reduced propensity for immunoglobulin production and oligosecretory nature. However, its distinct reliance on Bcl-2 has heightened its sensitivity to venetoclax. Further subclassification based on morphological and genomic characteristics could enhance our prediction models of treatment responses and enable more tailored therapeutic strategies for patients. This review aims to encapsulate the existing research evidence in this area.
Collapse
Affiliation(s)
- Abdul-Hamid Bazarbachi
- Division of Hematology/Oncology, Columbia University Irving Medical Center/New York-Presbyterian Hospital, New York, NY, USA.
| | - Hervé Avet-Loiseau
- Cancer Research Center of Toulouse, INSERM, Myeloma Genomics Laboratory, University Cancer Institute Toulouse Oncopole, Université Paul Sabatier, Toulouse, France
| | - Jean-Luc Harousseau
- Institut de Cancerologie de l'Ouest, Centre René Gauducheau, Nantes-St Herblain, France
| | - Ali Bazarbachi
- Department of Internal Medicine, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Mohamad Mohty
- Sorbonne University, Service d'Hematologie Clinique et Thérapie Cellulaire, Hôpital Saint Antoine, and INSERM UMR 938, Paris, France.
| |
Collapse
|
7
|
Lam N, Lee Y, Farber DL. A guide to adaptive immune memory. Nat Rev Immunol 2024; 24:810-829. [PMID: 38831162 DOI: 10.1038/s41577-024-01040-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2024] [Indexed: 06/05/2024]
Abstract
Immune memory - comprising T cells, B cells and plasma cells and their secreted antibodies - is crucial for human survival. It enables the rapid and effective clearance of a pathogen after re-exposure, to minimize damage to the host. When antigen-experienced, memory T cells become activated, they proliferate and produce effector molecules at faster rates and in greater magnitudes than antigen-inexperienced, naive cells. Similarly, memory B cells become activated and differentiate into antibody-secreting cells more rapidly than naive B cells, and they undergo processes that increase their affinity for antigen. The ability of T cells and B cells to form memory cells after antigen exposure is the rationale behind vaccination. Understanding immune memory not only is crucial for the design of more-efficacious vaccines but also has important implications for immunotherapies in infectious disease and cancer. This 'guide to' article provides an overview of the current understanding of the phenotype, function, location, and pathways for the generation, maintenance and protective capacity of memory T cells and memory B cells.
Collapse
Affiliation(s)
- Nora Lam
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - YoonSeung Lee
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Donna L Farber
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
8
|
Su X, Yu H, Lei Q, Chen X, Tong Y, Zhang Z, Yang W, Guo Y, Lin L. Systemic lupus erythematosus: pathogenesis and targeted therapy. MOLECULAR BIOMEDICINE 2024; 5:54. [PMID: 39472388 PMCID: PMC11522254 DOI: 10.1186/s43556-024-00217-8] [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: 07/14/2024] [Accepted: 10/16/2024] [Indexed: 11/02/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a multifaceted autoimmune disorder characterized by dysregulated immune responses and autoantibody production, which affects multiple organs and varies in clinical presentation and disease severity. The development of SLE is intricate, encompassing dysregulation within the immune system, a collapse of immunological tolerance, genetic susceptibilities to the disease, and a variety of environmental factors that can act as triggers. This review provides a comprehensive discussion of the pathogenesis and treatment strategies of SLE and focuses on the progress and status of traditional and emerging treatment strategies for SLE. Traditional treatment strategies for SLE have mainly employed non-specific approaches, including cytotoxic and immunosuppressive drugs, antimalarials, glucocorticoids, and NSAIDs. These strategies are effective in mitigating the effects of the disease, but they are not a complete cure and are often accompanied by adverse reactions. Emerging targeted therapeutic drugs, on the other hand, aim to control and treat SLE by targeting B and T cells, inhibiting their activation and function, as well as the abnormal activation of the immune system. A deeper understanding of the pathogenesis of SLE and the exploration of new targeted treatment strategies are essential to advance the treatment of this complex autoimmune disease.
Collapse
Affiliation(s)
- Xu Su
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Hui Yu
- Department of Urology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610014, China
| | - Qingqiang Lei
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400000, China
| | - Xuerui Chen
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Yanli Tong
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades, Paris, F-75015, France
| | - Zhongyang Zhang
- Department of Health Technology, The Danish National Research Foundation and Villum Foundation's Center IDUN, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Wenyong Yang
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
- Department of Neurosurgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610014, China.
| | - Yuanbiao Guo
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
| | - Liangbin Lin
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
- Obesity and Metabolism Medicine-Engineering Integration Laboratory, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China.
- The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
9
|
Yu-Hong Cheng R, Helmers AE, Kreuser S, Dahl N, Honaker Y, Lopez C, Rawlings DJ, James RG. Generation, expansion, gene delivery, and single-cell profiling in rhesus macaque plasma B cells. CELL REPORTS METHODS 2024; 4:100878. [PMID: 39406231 DOI: 10.1016/j.crmeth.2024.100878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 07/18/2024] [Accepted: 09/20/2024] [Indexed: 10/25/2024]
Abstract
A key step in developing engineered B cells for therapeutic purposes is evaluation in immunocompetent, large-animal models. Therefore, we developed methods to purify, expand, and differentiate non-human primate (NHP; rhesus macaque) B cells. After 7 days in culture, B cells expanded 10-fold, differentiated into a plasma cell phenotype (CD38, CD138), and secreted immunoglobulin G. Using single-cell sequencing and flow cytometry, we verified the presence of plasma cell genes in differentiated NHP B cells and unearthed less-recognized markers, such as CD59 and CD79A. In contrast with human cells, we found that the immune checkpoint molecule CD274 (PD-L1) and major histocompatibility complex (MHC) class I molecules were upregulated in NHP plasma cells in the transcriptional data. Lastly, we established the conditions for efficient transduction of NHP B cells with adeno-associated virus (AAV) vectors, achieving a delivery rate of approximately 60%. We envision that this work will accelerate proof-of-concept studies using engineered B cells in NHPs.
Collapse
Affiliation(s)
- Rene Yu-Hong Cheng
- Center of Immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA 98101, USA; Molecular Engineering and Science Institute, University of Washington, Seattle, WA 98195, USA
| | - Anna E Helmers
- Center of Immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA 98101, USA
| | - Shannon Kreuser
- Center of Immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA 98101, USA
| | - Noelle Dahl
- Center of Immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA 98101, USA
| | - Yuchi Honaker
- Center of Immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA 98101, USA
| | - Christina Lopez
- Center of Immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA 98101, USA
| | - David J Rawlings
- Center of Immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Richard G James
- Center of Immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA 98101, USA; Molecular Engineering and Science Institute, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA; Brotman-Baty Institute for Precision Medicine, Seattle, WA 98195, USA.
| |
Collapse
|
10
|
Cui X, Xiang Q, Huang Y, Ji Q, Hu Z, Shi T, Bao G, Liu Y. Mixed Th1/Th2/Th17 Responses Induced by Plant Oil Adjuvant-Based B. bronchiseptica Vaccine in Mice, with Mechanisms Unraveled by RNA-Seq, 16S rRNA and Metabolomics. Vaccines (Basel) 2024; 12:1182. [PMID: 39460348 PMCID: PMC11512391 DOI: 10.3390/vaccines12101182] [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: 09/05/2024] [Revised: 10/05/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES The current Bordetella bronchiseptica (Bb) vaccine, when adjuvanted with alum, does not elicit adequate robust cellular immunity or effective antibody defense against Bb attacks. Unfortunately, antibiotic treatment generally represents an ineffective strategy due to the development of resistance against a broad range of antibiotics. METHODS The present study was designed to investigate the immune response, protective capabilities and underlying mechanisms of a plant oil-based adjuvant E515 formulated with inactivated Bb antigen as a potential vaccine candidate against Bordetella bronchiseptica. RESULTS Immunization studies revealed that a combination of SO, VE and GS (E515) exhibited a good synergistic adjuvant effect. The E515 adjuvanted Bb vaccine was proven to be highly efficacious and induced a mixed Th1/Th2/Th17 immune response in mice, leading to a significant increase in Bb-specific IgG, IgG1 and IgG2a antibodies, proliferative lymphocyte responses and cytokine levels (by lymphocytes and serum) and effectively induced responses by CD4+ TE, TM cells and B cells. The E515 adjuvant significantly enhanced the immune protection provided by the Bb vaccine in a mice model, as indicated by a reduced bacterial burden in the lungs. Multi-omics sequencing analysis revealed that E515 functions as an adjuvant by modulating critical pathways, including cytokine-cytokine receptor interaction, the IL-17 signaling pathway and the chemokine signaling pathway. This modulation also included interactions with beneficial species of bacteria including Alistipes, Odoribacter and Colidextribacter, as well as energy and lipid-related metabolites, thus highlighting its role as an immunomodulatory agent. CONCLUSION Collectively, our results demonstrate the huge potential of E515-Bb vaccine candidates, thus highlighting the vegetable oil original adjuvant E515 as a promising agent for the development of new veterinary vaccines.
Collapse
Affiliation(s)
- Xuemei Cui
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (X.C.); (Q.X.); (Y.H.); (Q.J.); (Z.H.); (T.S.)
| | - Qiuju Xiang
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (X.C.); (Q.X.); (Y.H.); (Q.J.); (Z.H.); (T.S.)
- College of Animal Science and Technology·College of Veterinary Medicine, Zhejiang A&F University, Hangzhou 311300, China
| | - Yee Huang
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (X.C.); (Q.X.); (Y.H.); (Q.J.); (Z.H.); (T.S.)
| | - Quanan Ji
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (X.C.); (Q.X.); (Y.H.); (Q.J.); (Z.H.); (T.S.)
| | - Zizhe Hu
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (X.C.); (Q.X.); (Y.H.); (Q.J.); (Z.H.); (T.S.)
| | - Tuanyuan Shi
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (X.C.); (Q.X.); (Y.H.); (Q.J.); (Z.H.); (T.S.)
| | - Guolian Bao
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (X.C.); (Q.X.); (Y.H.); (Q.J.); (Z.H.); (T.S.)
| | - Yan Liu
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (X.C.); (Q.X.); (Y.H.); (Q.J.); (Z.H.); (T.S.)
| |
Collapse
|
11
|
Cordas Dos Santos DM, Toenges R, Bertamini L, Alberge JB, Ghobrial IM. New horizons in our understanding of precursor multiple myeloma and early interception. Nat Rev Cancer 2024:10.1038/s41568-024-00755-x. [PMID: 39414947 DOI: 10.1038/s41568-024-00755-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/06/2024] [Indexed: 10/18/2024]
Abstract
Multiple myeloma is an incurable plasma cell malignancy that evolves over decades through the selection and malignant transformation of monoclonal plasma cells. The evolution from precursor states to symptomatic disease is characterized by an increasing complexity of genomic alterations within the plasma cells and a remodelling of the microenvironment towards an immunosuppressive state. Notably, in patients with advanced disease, similar mechanisms of tumour escape and immune dysfunction mediate resistance to modern T cell-based therapies, such as T cell-engaging bispecific antibodies and chimeric antigen receptor (CAR)-T cells. Thus, an increasing number of clinical trials are assessing the efficiency and safety of these therapies in individuals with newly diagnosed multiple myeloma and high-risk smoldering multiple myeloma. In this Review, we summarize the current knowledge about tumour intrinsic and extrinsic processes underlying progression from precursor states to symptomatic myeloma and discuss the rationale for early interception including the use of T cell-redirecting therapies.
Collapse
Affiliation(s)
- David M Cordas Dos Santos
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Rosa Toenges
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Luca Bertamini
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Hematology, Erasmus MC Cancer Institute Rotterdam, Rotterdam, The Netherlands
| | - Jean-Baptiste Alberge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Irene M Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
12
|
Schapfl MA, LoMastro GM, Braun VZ, Hirai M, Levine MS, Kiermaier E, Labi V, Holland AJ, Villunger A. Centrioles are frequently amplified in early B cell development but dispensable for humoral immunity. Nat Commun 2024; 15:8890. [PMID: 39406735 PMCID: PMC11480410 DOI: 10.1038/s41467-024-53222-4] [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: 10/20/2023] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Centrioles define centrosome structure and function. Deregulation of centriole numbers can cause developmental defects and cancer. The p53 tumor suppressor limits the growth of cells lacking or harboring additional centrosomes and can be engaged by the "mitotic surveillance" or the "PIDDosome pathway", respectively. Here, we show that early B cell progenitors frequently present extra centrioles, ensuing their high proliferative activity and related DNA damage. Extra centrioles are efficiently cleared during B cell maturation. In contrast, centriole loss upon Polo-like kinase 4 (Plk4) deletion causes apoptosis and arrests B cell development. This defect can be rescued by co-deletion of Usp28, a critical component of the mitotic surveillance pathway, that restores cell survival and maturation. Centriole-deficient mature B cells are proliferation competent and mount a humoral immune response. Our findings imply that progenitor B cells are intolerant to centriole loss but permissive to centriole amplification, a feature potentially facilitating their malignant transformation.
Collapse
Affiliation(s)
- Marina A Schapfl
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Gina M LoMastro
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vincent Z Braun
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Maretoshi Hirai
- Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Michelle S Levine
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eva Kiermaier
- Life and Medical Sciences Institute, Immune and Tumor Biology, University of Bonn, Bonn, Germany
| | - Verena Labi
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Andrew J Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andreas Villunger
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria.
- The Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria.
| |
Collapse
|
13
|
Moysidou E, Christodoulou M, Lioulios G, Stai S, Karamitsos T, Dimitroulas T, Fylaktou A, Stangou M. Lymphocytes Change Their Phenotype and Function in Systemic Lupus Erythematosus and Lupus Nephritis. Int J Mol Sci 2024; 25:10905. [PMID: 39456692 PMCID: PMC11508046 DOI: 10.3390/ijms252010905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease, characterized by considerable changes in peripheral lymphocyte structure and function, that plays a critical role in commencing and reviving the inflammatory and immune signaling pathways. In healthy individuals, B lymphocytes have a major role in guiding and directing defense mechanisms against pathogens. Certain changes in B lymphocyte phenotype, including alterations in surface and endosomal receptors, occur in the presence of SLE and lead to dysregulation of peripheral B lymphocyte subpopulations. Functional changes are characterized by loss of self-tolerance, intra- and extrafollicular activation, and increased cytokine and autoantibody production. T lymphocytes seem to have a supporting, rather than a leading, role in the disease pathogenesis. Substantial aberrations in peripheral T lymphocyte subsets are evident, and include a reduction of cytotoxic, regulatory, and advanced differentiated subtypes, together with an increase of activated and autoreactive forms and abnormalities in follicular T cells. Up-regulated subpopulations, such as central and effector memory T cells, produce pre-inflammatory cytokines, activate B lymphocytes, and stimulate cell signaling pathways. This review explores the pivotal roles of B and T lymphocytes in the pathogenesis of SLE and Lupus Nephritis, emphasizing the multifaceted mechanisms and interactions and their phenotypic and functional dysregulations.
Collapse
Affiliation(s)
- Eleni Moysidou
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Michalis Christodoulou
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Georgios Lioulios
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Stamatia Stai
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Theodoros Karamitsos
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Cardiology, AHEPA University Hospital, 54636 Thessaloniki, Greece
| | - Theodoros Dimitroulas
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 4th Department of Medicine, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Asimina Fylaktou
- Department of Immunology, National Histocompatibility Center, Hippokration General Hospital, 54642 Thessaloniki, Greece;
| | - Maria Stangou
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| |
Collapse
|
14
|
Xiao R, Han J, Deng Y, Zhang L, Qian Y, Tian N, Yang Z, Zhang L. AGTR1: a potential biomarker associated with the occurrence and prognosis of lung adenocarcinoma. Front Oncol 2024; 14:1441235. [PMID: 39450258 PMCID: PMC11499140 DOI: 10.3389/fonc.2024.1441235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Introduction Lung adenocarcinoma, a disease with complex pathogenesis, high mortality and poor prognosis, is one of the subtypes of lung cancer. Hence, it is very crucial to find novel biomarkers as diagnostic and therapeutic targets for LUAD. Methods GSE10072 was used for DEGs and WGCNA, and the intersection genes were subjected to enrichment analysis through Metascape and GSEA. Key genes were screened by three machine learning methods. Further, the reliability of key genes was identified by ROC, COX regression analysis and qRT-PCR. CIBERSORT and Spearman analysis were used for understanding the relationships of LUAD, immunity and key genes. In addition, ceRNA networks and potential drugs of key genes were constructed and predicted. Results After overlapping 631 DEGs and key module genes, 623 intersection genes were obtained. Subsequently, DUOX1, CD36, AGTR1, FHL5 and SSR4 were further selected using three machine learning methods. Reliability analysis demonstrated that AGTR1 possesses important predictive value for the occurrence and prognosis of LUAD. The enrichment analysis showed that AGTR1 was significantly enriched in the GPCR-related pathways. Immune infiltration analysis showed that the development of LUAD was related to the changes of immune cells such as M2 macrophages and neutrophils, which were regulated by AGTR1. Further, AGTR1 is also involved in regulating immune chemokines, checkpoints and immune regulatory factors such as PECAM1, ADARB1, SPP1 and ENO1, all of them playing important roles in immune cell regulation, tumor cell proliferation and migration. Further, the drug-gene interaction network screened out 13 potential drugs such as Benazepril, Valsartan, Eprosartan, and so on. Discussion AGTR1 is a potential biomarker for the occurrence and progression of LUAD, closely related to tumor immunity, proliferation and migration. It can serve as a new target for the diagnosis and treatment of LUAD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lin Zhang
- College of Life Science/Institute of Molecular Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
15
|
Komura K. CD19: a promising target for systemic sclerosis. Front Immunol 2024; 15:1454913. [PMID: 39421745 PMCID: PMC11484411 DOI: 10.3389/fimmu.2024.1454913] [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: 06/25/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune disease characterized by immune dysregulation, vascular damage, and fibrosis. B cells play a significant role in SSc through autoantibody production, cytokine secretion, and T cell regulation. Autoantibodies like anti-topoisomerase I and anti-RNA polymerase III are specific to SSc and linked to clinical features such as skin and lung involvement. B cell depletion therapies, particularly anti-CD20 antibodies like rituximab, have shown benefits in treating SSc, improving skin and lung disease symptoms. However, CD19, another B cell marker, is more widely expressed and has emerged as a promising target in autoimmune diseases. CD19-targeted therapies, such as CAR T cells and Uplizna® (inebilizumab), have demonstrated potential in treating refractory autoimmune diseases, including SSc. Uplizna® offers advantages over rituximab by targeting a broader range of B cells and showing higher efficacy in specific patient subsets. Clinical trials currently investigate Uplizna®'s effectiveness in SSc, particularly in severe cases. While these therapies offer hope, long-term safety and efficacy remain unknown. SSc is still a complex disease, but advancing B cell-targeted treatments could significantly improve patient outcomes and knowledge about the pathogenesis.
Collapse
Affiliation(s)
- Kazuhiro Komura
- Department of Dermatology, Kanazawa Red Cross Hospital, Japanese Red Cross Society, Kanazawa, Ishikawa, Japan
| |
Collapse
|
16
|
Rubino V, Carriero F, Palatucci AT, Giovazzino A, Salemi F, Carrano R, Sabbatini M, Ruggiero G, Terrazzano G. T R3-56 and Treg Regulatory T Cell Subsets as Potential Indicators of Graft Tolerance Control in Kidney Transplant Recipients. Int J Mol Sci 2024; 25:10610. [PMID: 39408939 PMCID: PMC11477056 DOI: 10.3390/ijms251910610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Identification of early signatures of immune rejection represents a key challenge in the clinical management of kidney transplant. To address such an issue, we enrolled 53 kidney transplant recipients without signs of graft rejection, no infectious episodes and no change in the immunosuppressive regimen in the last 6 months. An extensive immune profile revealed increased activation of the T cells, a decreased amount and growth ability of the Treg and a higher level of the TR3-56 regulatory T cell subset, described by us as involved in the preferential control of cytotoxic T lymphocytes. In renal transplant recipients, the high level of the TR3-56 cells associates with a reduction in both the amount and the growth ability of the Treg. Moreover, when the transplanted subjects were categorised according to their stable or unstable disease status, as defined by changes in serum creatinine ≥0.2 mg/dL in two consecutive detections, a higher TR3-56 level and defective Treg growth ability were observed to characterise patients with unstable graft control. Further studies are required to substantiate the hypothesis that immune profiling, including TR3-56 evaluation, might represent a valuable diagnostic tool to identify patients at risk of developing significant anti-donor allo-immune responses.
Collapse
Affiliation(s)
- Valentina Rubino
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli “Federico II”, 80131 Napoli, Italy; (V.R.); (A.G.)
| | - Flavia Carriero
- Dipartimento di Scienze Della Salute, Università Della Basilicata, 85100 Potenza, Italy; (F.C.); (A.T.P.); (G.T.)
| | - Anna Teresa Palatucci
- Dipartimento di Scienze Della Salute, Università Della Basilicata, 85100 Potenza, Italy; (F.C.); (A.T.P.); (G.T.)
| | - Angela Giovazzino
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli “Federico II”, 80131 Napoli, Italy; (V.R.); (A.G.)
| | - Fabrizio Salemi
- Percorso Clinico Assistenziale in Nefrologia e Trapianto Renale, Azienda Ospedaliera Universitaria “Federico II”, 80131 Napoli, Italy; (F.S.); (R.C.)
| | - Rosa Carrano
- Percorso Clinico Assistenziale in Nefrologia e Trapianto Renale, Azienda Ospedaliera Universitaria “Federico II”, 80131 Napoli, Italy; (F.S.); (R.C.)
| | - Massimo Sabbatini
- Dipartimento di Sanità Pubblica, Sezione di Nefrologia, Università di Napoli “Federico II”, 80131 Napoli, Italy;
| | - Giuseppina Ruggiero
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli “Federico II”, 80131 Napoli, Italy; (V.R.); (A.G.)
| | - Giuseppe Terrazzano
- Dipartimento di Scienze Della Salute, Università Della Basilicata, 85100 Potenza, Italy; (F.C.); (A.T.P.); (G.T.)
| |
Collapse
|
17
|
Yan M, Man S, Ma L, Guo L, Huang L, Gao W. Immunological mechanisms in steatotic liver diseases: An overview and clinical perspectives. Clin Mol Hepatol 2024; 30:620-648. [PMID: 38988278 PMCID: PMC11540396 DOI: 10.3350/cmh.2024.0315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 07/12/2024] Open
Abstract
Steatotic liver diseases (SLD) are the principal worldwide cause of cirrhosis and end-stage liver cancer, affecting nearly a quarter of the global population. SLD includes metabolic dysfunction-associated alcoholic liver disease (MetALD) and metabolic dysfunction-associated steatotic liver disease (MASLD), resulting in asymptomatic liver steatosis, fibrosis, cirrhosis and associated complications. The immune processes include gut dysbiosis, adiposeliver organ crosstalk, hepatocyte death and immune cell-mediated inflammatory processes. Notably, various immune cells such as B cells, plasma cells, dendritic cells, conventional CD4+ and CD8+ T cells, innate-like T cells, platelets, neutrophils and macrophages play vital roles in the development of MetALD and MASLD. Immunological modulations targeting hepatocyte death, inflammatory reactions and gut microbiome include N-acetylcysteine, selonsertib, F-652, prednisone, pentoxifylline, anakinra, JKB-121, HA35, obeticholic acid, probiotics, prebiotics, antibiotics and fecal microbiota transplantation. Understanding the immunological mechanisms underlying SLD is crucial for advancing clinical therapeutic strategies.
Collapse
Affiliation(s)
- Mengyao Yan
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Shuli Man
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Long Ma
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Lanping Guo
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Luqi Huang
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenyuan Gao
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin, China
| |
Collapse
|
18
|
Wang R, Lan C, Benlagha K, Camara NOS, Miller H, Kubo M, Heegaard S, Lee P, Yang L, Forsman H, Li X, Zhai Z, Liu C. The interaction of innate immune and adaptive immune system. MedComm (Beijing) 2024; 5:e714. [PMID: 39286776 PMCID: PMC11401974 DOI: 10.1002/mco2.714] [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: 10/09/2023] [Revised: 08/11/2024] [Accepted: 08/11/2024] [Indexed: 09/19/2024] Open
Abstract
The innate immune system serves as the body's first line of defense, utilizing pattern recognition receptors like Toll-like receptors to detect pathogens and initiate rapid response mechanisms. Following this initial response, adaptive immunity provides highly specific and sustained killing of pathogens via B cells, T cells, and antibodies. Traditionally, it has been assumed that innate immunity activates adaptive immunity; however, recent studies have revealed more complex interactions. This review provides a detailed dissection of the composition and function of the innate and adaptive immune systems, emphasizing their synergistic roles in physiological and pathological contexts, providing new insights into the link between these two forms of immunity. Precise regulation of both immune systems at the same time is more beneficial in the fight against immune-related diseases, for example, the cGAS-STING pathway has been found to play an important role in infections and cancers. In addition, this paper summarizes the challenges and future directions in the field of immunity, including the latest single-cell sequencing technologies, CAR-T cell therapy, and immune checkpoint inhibitors. By summarizing these developments, this review aims to enhance our understanding of the complexity interactions between innate and adaptive immunity and provides new perspectives in understanding the immune system.
Collapse
Affiliation(s)
- Ruyuan Wang
- Department of Thyroid and Breast Surgery Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Caini Lan
- Cancer Center Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Kamel Benlagha
- Alloimmunity, Autoimmunity and Transplantation Université de Paris, Institut de Recherche Saint-Louis, EMiLy, INSERM U1160 Paris France
| | - Niels Olsen Saraiva Camara
- Department of Immunology Institute of Biomedical Sciences University of São Paulo (USP) São Paulo São Paulo Brazil
| | - Heather Miller
- Coxiella Pathogenesis Section, Laboratory of Bacteriology Rocky Mountain Laboratories National Institute of Allergy and Infectious Diseases, National Institutes of Health Hamilton Montana USA
| | - Masato Kubo
- Division of Molecular Pathology Research Institute for Biomedical Sciences (RIBS) Tokyo University of Science Noda Chiba Japan
| | - Steffen Heegaard
- Department of Ophthalmology Rigshospitalet Hospital Copenhagen University Copenhagen Denmark
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong China
| | - Lu Yang
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology Wuhan Hubei China
| | - Huamei Forsman
- Department of Laboratory Medicine Institute of Biomedicine, University of Gothenburg Gothenburg Sweden
| | - Xingrui Li
- Department of Thyroid and Breast Surgery Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Zhimin Zhai
- Department of Hematology The Second Hospital of Anhui Medical University Hefei China
| | - Chaohong Liu
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology Wuhan Hubei China
| |
Collapse
|
19
|
Wang W, Dong G, Yang Z, Li S, Li J, Wang L, Zhu Q, Wang Y. Single-cell analysis of tumor microenvironment and cell adhesion reveals that interleukin-1 beta promotes cancer cell proliferation in breast cancer. Animal Model Exp Med 2024; 7:617-625. [PMID: 38860503 PMCID: PMC11528385 DOI: 10.1002/ame2.12445] [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: 01/29/2024] [Accepted: 05/20/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC), which is so called because of the lack of estrogen receptors (ER), progesterone receptors (PR), and human epidermal growth factor receptor 2 (HER2) receptors on the cancer cells, accounts for 10%-15% of all breast cancers. The heterogeneity of the tumor microenvironment is high. However, the role of plasma cells controlling the tumor migration progression in TNBC is still not fully understood. METHODS We analyzed single-cell RNA sequencing data from five HER2 positive, 12 ER positive/PR positive, and nine TNBC samples. The potential targets were validated by immunohistochemistry. RESULTS Plasma cells were enriched in TNBC samples, which was consistent with validation using data from The Cancer Genome Atlas. Cell communication analysis revealed that plasma cells interact with T cells through the intercellular adhesion molecule 2-integrin-aLb2 complex, and then release interleukin 1 beta (IL1B), as verified by immunohistochemistry, ultimately promoting tumor growth. CONCLUSION Our results revealed the role of plasma cells in TNBC and identified IL1B as a new prognostic marker for TNBC.
Collapse
Affiliation(s)
- Wenyan Wang
- Department of General Surgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Gehong Dong
- Department of Pathology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Ziguo Yang
- Department of General Surgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Shaoxiang Li
- Department of Pathology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Jia Li
- Department of Pathology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Lin Wang
- Department of General Surgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Qiang Zhu
- Department of General Surgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Yuchen Wang
- Department of PharmacologyInstitute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
20
|
Song HS, Kim YM. Efficient In Vitro Plasma Cell Differentiation by B Cell Receptor Activation and Cytokine Stimulation. Immune Netw 2024; 24:e35. [PMID: 39513030 PMCID: PMC11538606 DOI: 10.4110/in.2024.24.e35] [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: 08/21/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 11/15/2024] Open
Abstract
Plasma cells (PCs) constitute a small proportion of B cells, limiting their biochemical characterization. An in vitro culture system that reliably generates PCs could provide an alternative method to obtain PCs for further analysis and manipulation. To date, most in vitro PC differentiation methods rely on B cell receptor (BCR)-independent stimulants, including TLR ligands, CD40L, and cytokines. However, these methods do not fully recapitulate the natural T cell-dependent PC differentiation process, in which BCR activation is the initial events. In this study, we established an efficient in vitro PC differentiation method incorporating BCR stimulation. Naïve B cells were first stimulated with anti-IgM and anti-CD40 Abs, followed by stimulation with various cytokines. By screening cytokines known to participate in PC differentiation in vivo, we identified that the combination of IL-4 and IL-5 induced the most efficient PC differentiation. The in vitro generated PCs highly expressed PC-associated surface markers and regulatory genes. Additionally, they secreted high amounts of IgM and IgG Abs. Moreover, retroviral transduction of B cells resulted in efficient target gene expression in PCs. Our new method closely mimics natural PC differentiation and effectively generates a large quantity of PCs for various applications, including elucidating the molecular mechanisms underlying PC differentiation.
Collapse
Affiliation(s)
- Hyun-Sup Song
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - You-Me Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| |
Collapse
|
21
|
Ma C, Luo C, Deng F, Yu C, Chen Y, Zhong G, Zhan Y, Nie L, Huang Y, Xia Y, Cai Z, Xu K, Cai H, Wang F, Lu Z, Zeng X, Zhu Y, Liu S. Major vault protein directly enhances adaptive immunity induced by Influenza A virus or indirectly through innate immunity. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167441. [PMID: 39069011 DOI: 10.1016/j.bbadis.2024.167441] [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: 01/08/2024] [Revised: 07/05/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
As we previously revealed, major vault protein (MVP) is a virus-induced host factor, and its expression is crucial for innate immune responses. Nevertheless, the function of MVP in adaptive immunity is poorly known. Here, we demonstrate that Mvp knockout mice had attenuated antibody responses and reduced survival after rechallenge with homologous influenza A virus (IAV) relative to wild-type mice. Analysis of B cell populations showed that MVP promoted germinal center (GC) responses to develop optimal antiviral humoral immunity. Although MVP-deficient T cells and dendritic cells (DCs) were not intrinsically damaged, MVP promoted activating effector T cells and T follicular helper responses and regulated specific DC subsets. These findings suggest that MVP directs an effective adaptive immune response against IAV by directly engaging in GC reactions or indirectly augmenting cellular immunity via innate immune pathways.
Collapse
Affiliation(s)
- Caijiao Ma
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Chuanjin Luo
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Feiyan Deng
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Chen Yu
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yumeng Chen
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Gechang Zhong
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yuxin Zhan
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Longyu Nie
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yu Huang
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yongfang Xia
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Zeng Cai
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Ke Xu
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Huanhuan Cai
- Institute of Myocardial Injury and Repair, Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
| | - Fubing Wang
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan 430072, China
| | - Zhibing Lu
- Institute of Myocardial Injury and Repair, Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
| | - Xiangtai Zeng
- Department of General Surgery, Ganzhou Key Laboratory of Thyroid Cancer, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, China
| | - Ying Zhu
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Shi Liu
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China; Institute of Myocardial Injury and Repair, Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430072, China; Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan 430072, China; Department of General Surgery, Ganzhou Key Laboratory of Thyroid Cancer, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, China.
| |
Collapse
|
22
|
Atef Y, Ito T, Masuda A, Kato Y, Nishimura A, Kanda Y, Kunisawa J, Kusakabe T, Nishida M. Diabetic Mice Spleen Vulnerability Contributes to Decreased Persistence of Antibody Production after SARS-CoV-2 Vaccine. Int J Mol Sci 2024; 25:10379. [PMID: 39408710 PMCID: PMC11476529 DOI: 10.3390/ijms251910379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 09/14/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
During the COVID-19 pandemic, diabetic and obese patients experienced higher rates of hospital admissions, severe illness, and mortality. However, vaccinations failed to provide those vulnerable populations the same level of protection against COVID-19 severity as those without diabetic and obese phenotypes. Our study aimed to investigate how diabetes mellitus (DM) impacts the immune response following vaccination including the artificially designed trimeric SARS-CoV-2 spike (S)-protein. By using two diabetic mouse models, ob/ob mice (obese, hyperglycemic, and insulin-resistant) and STZ-treated mice (insulin-deficient and hyperglycemic), we observed a significant reduction in S-protein-specific IgG antibody titer post-vaccination in both diabetic models compared to wild-type (WT) mice. Both diabetic mouse models exhibited significant abnormalities in spleen tissue, including marked reductions in splenic weight and the size of the white pulp regions. Furthermore, the splenic T-cell and B-cell zones were notably diminished, suggesting an underlying immune dysfunction that could contribute to impaired antibody production. Notably, vaccination with the S-protein, when paired with an optimal adjuvant, did not exacerbate diabetic cardiomyopathy, blood glucose levels, or liver function, providing reassurance about the vaccine's safety. These findings offer valuable insights into potential mechanisms responsible for the decreased persistence of antibody production in diabetic patients.
Collapse
Affiliation(s)
- Yara Atef
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Tomoya Ito
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Akitsu Masuda
- Laboratory of Creative Science for Insect Industries, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 819-0395, Japan;
| | - Yuri Kato
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Akiyuki Nishimura
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences (NINS), Okazaki 444-8787, Japan;
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences (NINS), Okazaki 444-8787, Japan
- Department of Physiological Sciences, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki 210-9501, Japan;
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
- Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Takahiro Kusakabe
- Laboratory of Insect Genome Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Fukuoka 819-0395, Japan;
| | - Motohiro Nishida
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences (NINS), Okazaki 444-8787, Japan;
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences (NINS), Okazaki 444-8787, Japan
- Department of Physiological Sciences, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan
| |
Collapse
|
23
|
Li H, Xia N. The multifaceted roles of B lymphocytes in metabolic dysfunction-associated steatotic liver disease. Front Immunol 2024; 15:1447391. [PMID: 39372417 PMCID: PMC11449700 DOI: 10.3389/fimmu.2024.1447391] [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: 06/11/2024] [Accepted: 09/05/2024] [Indexed: 10/08/2024] Open
Abstract
Recent evidence suggests that adaptive immune cells are important contributors to metabolic dysfunction-associated steatotic liver disease (MASLD, formerly non-alcoholic fatty liver disease, NAFLD). In liver biopsies from MASLD patients, the accumulation of intrahepatic B cells is positively correlated with the MASLD activity score. Hepatic B-cell infiltration is observed in experimental models of metabolic dysfunction-associated steatohepatitis (MASH, formerly non-alcoholic steatohepatitis, NASH). Intrahepatic B2 cells have been shown to contribute to MASLD/MASH by activating T cells, macrophages and hepatic stellate cells, and by producing pathogenic IgG antibodies. In mice fed a MASH diet, selective depletion of B2 cells reduces steatohepatitis and fibrosis. Intestinal B cells are metabolically activated in MASH and promote T-cell activation independently of TCR signaling. In addition, B cells have been shown to contribute to liver fibrosis by activating monocyte-derived macrophages through the secretion of IgA immunoglobulins. Furthermore, our recent study indicates that certain B cell subsets, very likely regulatory B cells, may play a protective role in MASLD. This review summarizes the molecular mechanisms of B cell functions and discusses future research directions on the different roles of B cells in MASLD and MASH.
Collapse
Affiliation(s)
- Huige Li
- Department of Pharmacology, University Medical Center, Johannes Gutenberg
University, Mainz, Germany
| | - Ning Xia
- Department of Pharmacology, University Medical Center, Johannes Gutenberg
University, Mainz, Germany
| |
Collapse
|
24
|
Carreto-Binaghi LE, Sztein MB, Booth JS. Role of cellular effectors in the induction and maintenance of IgA responses leading to protective immunity against enteric bacterial pathogens. Front Immunol 2024; 15:1446072. [PMID: 39324143 PMCID: PMC11422102 DOI: 10.3389/fimmu.2024.1446072] [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: 06/08/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
The mucosal immune system is a critical first line of defense to infectious diseases, as many pathogens enter the body through mucosal surfaces, disrupting the balanced interactions between mucosal cells, secretory molecules, and microbiota in this challenging microenvironment. The mucosal immune system comprises of a complex and integrated network that includes the gut-associated lymphoid tissues (GALT). One of its primary responses to microbes is the secretion of IgA, whose role in the mucosa is vital for preventing pathogen colonization, invasion and spread. The mechanisms involved in these key responses include neutralization of pathogens, immune exclusion, immune modulation, and cross-protection. The generation and maintenance of high affinity IgA responses require a delicate balance of multiple components, including B and T cell interactions, innate cells, the cytokine milieu (e.g., IL-21, IL-10, TGF-β), and other factors essential for intestinal homeostasis, including the gut microbiota. In this review, we will discuss the main cellular components (e.g., T cells, innate lymphoid cells, dendritic cells) in the gut microenvironment as mediators of important effector responses and as critical players in supporting B cells in eliciting and maintaining IgA production, particularly in the context of enteric infections and vaccination in humans. Understanding the mechanisms of humoral and cellular components in protection could guide and accelerate the development of more effective mucosal vaccines and therapeutic interventions to efficiently combat mucosal infections.
Collapse
Affiliation(s)
- Laura E Carreto-Binaghi
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Laboratorio de Inmunobiologia de la Tuberculosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Marcelo B Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Tumor Immunology and Immunotherapy Program, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Jayaum S Booth
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
25
|
Deguine J, Xavier RJ. B cell tolerance and autoimmunity: Lessons from repertoires. J Exp Med 2024; 221:e20231314. [PMID: 39093312 PMCID: PMC11296956 DOI: 10.1084/jem.20231314] [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: 04/25/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
Adaptive immune cell function is regulated by a highly diverse receptor recombined from variable germline-encoded segments that can recognize an almost unlimited array of epitopes. While this diversity enables the recognition of any pathogen, it also poses a risk of self-recognition, leading to autoimmunity. Many layers of regulation are present during both the generation and activation of B cells to prevent this phenomenon, although they are evidently imperfect. In recent years, our ability to analyze immune repertoires at scale has drastically increased, both through advances in sequencing and single-cell analyses. Here, we review the current knowledge on B cell repertoire analyses, focusing on their implication for autoimmunity. These studies demonstrate that a failure of tolerance occurs at multiple independent checkpoints in different autoimmune contexts, particularly during B cell maturation, plasmablast differentiation, and within germinal centers. These failures are marked by distinct repertoire features that may be used to identify disease- or patient-specific therapeutic approaches.
Collapse
Affiliation(s)
- Jacques Deguine
- Immunology Program, Broad Institute of Massachusetts Institute of Technology and Harvard , Cambridge, MA, USA
| | - Ramnik J Xavier
- Immunology Program, Broad Institute of Massachusetts Institute of Technology and Harvard , Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School , Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
26
|
Han XQ, Cui ZW, Ma ZY, Wang J, Hu YZ, Li J, Ye JM, Tafalla C, Zhang YA, Zhang XJ. Phagocytic Plasma Cells in Teleost Fish Provide Insights into the Origin and Evolution of B Cells in Vertebrates. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:730-742. [PMID: 38984862 DOI: 10.4049/jimmunol.2400182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 06/17/2024] [Indexed: 07/11/2024]
Abstract
Teleost IgM+ B cells can phagocytose, like mammalian B1 cells, and secrete Ag-specific IgM, like mammalian B2 cells. Therefore, teleost IgM+ B cells may have the functions of both mammalian B1 and B2 cells. To support this view, we initially found that grass carp (Ctenopharyngodon idella) IgM+ plasma cells (PCs) exhibit robust phagocytic ability, akin to IgM+ naive B cells. Subsequently, we sorted grass carp IgM+ PCs into two subpopulations: nonphagocytic (Pha-IgM+ PCs) and phagocytic IgM+ PCs (Pha+IgM+ PCs), both of which demonstrated the capacity to secrete natural IgM with LPS and peptidoglycan binding capacity. Remarkably, following immunization of grass carp with an Ag, we observed that both Pha-IgM+ PCs and Pha+IgM+ PCs could secrete Ag-specific IgM. Furthermore, in vitro concatenated phagocytosis experiments in which Pha-IgM+ PCs from an initial phagocytosis experiment were sorted and exposed again to beads confirmed that these cells also have phagocytic capabilities, thereby suggesting that all teleost IgM+ B cells have phagocytic potential. Additionally, we found that grass carp IgM+ PCs display classical phenotypic features of macrophages, providing support for the hypothesis that vertebrate B cells evolved from ancient phagocytes. These findings together reveal that teleost B cells are a primitive B cell type with functions reminiscent of both mammalian B1 and B2 cells, providing insights into the origin and evolution of B cells in vertebrates.
Collapse
Affiliation(s)
- Xue-Qing Han
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Zheng-Wei Cui
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Zi-You Ma
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Jie Wang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Ya-Zhen Hu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Jun Li
- School of Science and Medicine, Lake Superior State University, Sault Sainte Marie, MI
| | - Jian-Min Ye
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Carolina Tafalla
- Animal Health Research Center (CISA), National Institute for Agricultural and Food Research and Technology (INIA), National Research Council (CSIC), Madrid, Spain
| | - Yong-An Zhang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Xu-Jie Zhang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| |
Collapse
|
27
|
Nguyen AA, Platt CD. Flow Cytometry-based Immune Phenotyping of T and B Lymphocytes in the Evaluation of Immunodeficiency and Immune Dysregulation. Clin Lab Med 2024; 44:479-493. [PMID: 39089753 DOI: 10.1016/j.cll.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
There are approximately 500 congenital disorders that impair immune cell development and/or function. Patients with these disorders may present with a wide range of symptoms, including increased susceptibility to infection, autoimmunity, autoinflammation, lymphoproliferation, and/or atopy. Flow cytometry-based immune phenotyping of T and B lymphocytes plays an essential role in the evaluation of patients with these presentations. In this review, we describe the clinical utility of flow cytometry as part of a comprehensive evaluation of immune function and how this testing may be used as a diagnostic tool to identify underlying aberrant immune pathways, monitor disease activity, and assess infection risk.
Collapse
Affiliation(s)
- Alan A Nguyen
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Fegan Building 6th Floor, Boston, MA 02115, USA
| | - Craig D Platt
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, 1 Blackfan Circle, Karp Building 10th Floor, Boston, MA 02115, USA.
| |
Collapse
|
28
|
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
|
29
|
Rohrbacher S, Seefried S, Hartmannsberger B, Annabelle R, Appeltshauser L, Arlt FA, Brämer D, Dresel C, Dorst J, Elmas Z, Franke C, Geis C, Högen T, Krause S, Marziniak M, Mäurer M, Prüss H, Schoeberl F, Schrank B, Steen C, Teichtinger H, Thieme A, Wessely L, Zernecke A, Sommer C, Doppler K. Different Patterns of Autoantibody Secretion by Peripheral Blood Mononuclear Cells in Autoimmune Nodopathies. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200295. [PMID: 39173087 PMCID: PMC11379437 DOI: 10.1212/nxi.0000000000200295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
BACKGROUND AND OBJECTIVES Autoimmune nodopathies with antibodies against the paranodal proteins show a distinct phenotype of a severe sensorimotor neuropathy. In some patients, complete remission can be achieved after treatment with rituximab whereas others show a chronic course. For optimal planning of treatment, predicting the course of disease and therapeutic response is crucial. METHODS We stimulated peripheral blood mononuclear cells in vitro to find out whether secretion of specific autoantibodies may be a predictor of the course of disease and response to rituximab. RESULTS Three patterns could be identified: In most patients with anti-Neurofascin-155-, anti-Contactin-1-, and anti-Caspr1-IgG4 autoantibodies, in vitro production of autoantibodies was detected, indicating autoantigen-specific memory B cells and short-lived plasma cells/plasmablasts as the major source of autoantibodies. These patients generally showed a good response to rituximab. In a subgroup of patients with anti-Neurofascin-155-IgG4 autoantibodies and insufficient response to rituximab, no in vitro autoantibody production was found despite high serum titers, indicating autoantibody secretion by long-lived plasma cells outside the peripheral blood. In the patients with anti-pan-Neurofascin autoantibodies-all with a monophasic course of disease-no in vitro autoantibody production could be measured, suggesting a lack of autoantigen-specific memory B cells. In some of them, autoantibody production by unstimulated cells was detectable, presumably corresponding to high amounts of autoantigen-specific plasmablasts-well in line with a severe but monophasic course of disease. DISCUSSION Our data suggest that different B-cell responses may occur in autoimmune nodopathies and may serve as markers of courses of disease and response to rituximab.
Collapse
Affiliation(s)
- Sophia Rohrbacher
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Sabine Seefried
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Beate Hartmannsberger
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Rosa Annabelle
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Luise Appeltshauser
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Friederike A Arlt
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Dirk Brämer
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Christian Dresel
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Johannes Dorst
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Zeynep Elmas
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Christiana Franke
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Christian Geis
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Tobias Högen
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Sabine Krause
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Martin Marziniak
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Mathias Mäurer
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Harald Prüss
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Florian Schoeberl
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Bertold Schrank
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Claudia Steen
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Helena Teichtinger
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Andrea Thieme
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Lena Wessely
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Alma Zernecke
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Claudia Sommer
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Kathrin Doppler
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| |
Collapse
|
30
|
Bahabayi A, Guan Z, Zheng M, Yu H, Hasimu A, Nie Y, Zhao M, Zhu Y, Ren J, Zhao Y, Ma X, Li Q, Zhang Z, Zeng X, Liu C. Expression of GPR56 reflects a hypoactivated state of circulating B cells and is downregulated in B cell subsets in patients with early-stage lung adenocarcinoma. Immunology 2024; 173:172-184. [PMID: 38840413 DOI: 10.1111/imm.13819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 05/17/2024] [Indexed: 06/07/2024] Open
Abstract
Lung adenocarcinoma (LUAD) is the most prevalent subtype of lung cancer, and the early detection and diagnosis of this disease are crucial in reducing mortality rates. The timely diagnosis of LUAD is essential for controlling tumour development and enabling early surgical treatment. GPR56 is a vital G protein-coupled receptor and its role in T lymphocytes has received considerable attention. However, its function in B cells remains unclear. This study aimed to investigate the significance of GPR56 in LUAD. We found that GPR56 exhibited a significant increase in circulating plasmablasts and a decrease in new memory B cells. GPR56 expression in B cells was significantly reduced after LPS stimulation and the proportion of HLA-DR+ and CD40+ proportions were also decreased in GPR56+ B cells after stimulation. Additionally, GPR56 exhibited significant down-regulation in circulating B cell subsets of early-stage LUAD patients, and there were significant correlations between GPR56+ B cell subsets and tumour markers. In conclusion, GPR56 could reflect the hypoactivation state of B cells and the decreased proportion of GPR56+ B cell subset in LUAD patients can signify the active humoral immunity in vivo. The expression of GPR56 in B cells could potentially hold value in the early diagnosis of LUAD.
Collapse
Affiliation(s)
- Ayibaota Bahabayi
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Zhao Guan
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Mohan Zheng
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - He Yu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Ainizati Hasimu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Yuying Nie
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Ming Zhao
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Yaoyi Zhu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Jiaxin Ren
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Yiming Zhao
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Xiancan Ma
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Qi Li
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Zhonghui Zhang
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Xingyue Zeng
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Chen Liu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| |
Collapse
|
31
|
Anderson J, Quah L, Mangano K, Pellicci DG, Mazarakis N, Licciardi PV. A 38-colour high dimensional immunophenotyping panel for human peripheral blood mononuclear cells. J Immunol Methods 2024; 532:113726. [PMID: 38992764 DOI: 10.1016/j.jim.2024.113726] [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: 03/27/2024] [Revised: 05/28/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024]
Abstract
High dimensional immunophenotyping panels are invaluable resources for performing extensive phenotyping on peripheral blood mononuclear cells (PBMCs). We designed a 38-colour high dimensional phenotyping panel to measure innate (monocytes, dendritic cells, NK cells, basophils, innate like lymphoid cells), T cell (γδ T cells, MAIT cells, CD4 and CD8 memory, Th1, Th2, Th17, Tfh, Treg) and B cell (memory, plasma cells, transitional B cells, plasmablasts, IgG, IgM) subsets in addition to their activation status using the 5-laser Cytek Aurora. We optimised optimal fluorochrome combinations and titres to minimise spread and autofluorescence of rare immune cell populations and tested this panel on PBMCs from 15 healthy adults. This high dimensional panel will be invaluable for direct ex vivo studies to evaluate immune cells in the context of human health and disease, especially when samples are limited.
Collapse
Affiliation(s)
- Jeremy Anderson
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia.
| | - Leanne Quah
- Murdoch Children's Research Institute, Melbourne, Australia
| | - Kiara Mangano
- Murdoch Children's Research Institute, Melbourne, Australia
| | - Daniel G Pellicci
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Microbiology and Immunology, University of Melbourne, Melbourne, Australia
| | - Nadia Mazarakis
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Paul V Licciardi
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| |
Collapse
|
32
|
Yang Y, Chen X, Pan J, Ning H, Zhang Y, Bo Y, Ren X, Li J, Qin S, Wang D, Chen MM, Zhang Z. Pan-cancer single-cell dissection reveals phenotypically distinct B cell subtypes. Cell 2024; 187:4790-4811.e22. [PMID: 39047727 DOI: 10.1016/j.cell.2024.06.038] [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: 06/09/2023] [Revised: 04/25/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024]
Abstract
Characterizing the compositional and phenotypic characteristics of tumor-infiltrating B cells (TIBs) is important for advancing our understanding of their role in cancer development. Here, we establish a comprehensive resource of human B cells by integrating single-cell RNA sequencing data of B cells from 649 patients across 19 major cancer types. We demonstrate substantial heterogeneity in their total abundance and subtype composition and observe immunoglobulin G (IgG)-skewness of antibody-secreting cell isotypes. Moreover, we identify stress-response memory B cells and tumor-associated atypical B cells (TAABs), two tumor-enriched subpopulations with prognostic potential, shared in a pan-cancer manner. In particular, TAABs, characterized by a high clonal expansion level and proliferative capacity as well as by close interactions with activated CD4 T cells in tumors, are predictive of immunotherapy response. Our integrative resource depicts distinct clinically relevant TIB subsets, laying a foundation for further exploration of functional commonality and diversity of B cells in cancer.
Collapse
Affiliation(s)
- Yu Yang
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Xueyan Chen
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Jieying Pan
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Huiheng Ning
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Yaojun Zhang
- State Key Laboratory of Oncology in South China, Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yufei Bo
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Xianwen Ren
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Jiesheng Li
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Shishang Qin
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Dongfang Wang
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China.
| | - Min-Min Chen
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China.
| |
Collapse
|
33
|
Fischer K, Lulla A, So TY, Pereyra-Gerber P, Raybould MIJ, Kohler TN, Yam-Puc JC, Kaminski TS, Hughes R, Pyeatt GL, Leiss-Maier F, Brear P, Matheson NJ, Deane CM, Hyvönen M, Thaventhiran JED, Hollfelder F. Rapid discovery of monoclonal antibodies by microfluidics-enabled FACS of single pathogen-specific antibody-secreting cells. Nat Biotechnol 2024:10.1038/s41587-024-02346-5. [PMID: 39143416 DOI: 10.1038/s41587-024-02346-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/27/2024] [Indexed: 08/16/2024]
Abstract
Monoclonal antibodies are increasingly used to prevent and treat viral infections and are pivotal in pandemic response efforts. Antibody-secreting cells (ASCs; plasma cells and plasmablasts) are an excellent source of high-affinity antibodies with therapeutic potential. Current methods to study antigen-specific ASCs either have low throughput, require expensive and labor-intensive screening or are technically demanding and therefore not widely accessible. Here we present a straightforward technology for the rapid discovery of monoclonal antibodies from ASCs. Our approach combines microfluidic encapsulation of single cells into an antibody capture hydrogel with antigen bait sorting by conventional flow cytometry. With our technology, we screened millions of mouse and human ASCs and obtained monoclonal antibodies against severe acute respiratory syndrome coronavirus 2 with high affinity (<1 pM) and neutralizing capacity (<100 ng ml-1) in 2 weeks with a high hit rate (>85% of characterized antibodies bound the target). By facilitating access to the underexplored ASC compartment, the approach enables efficient antibody discovery and immunological studies into the generation of protective antibodies.
Collapse
Affiliation(s)
- Katrin Fischer
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Aleksei Lulla
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Tsz Y So
- MRC Toxicology Unit, Gleeson Building, Cambridge, UK
| | - Pehuén Pereyra-Gerber
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK
| | - Matthew I J Raybould
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, Oxford, UK
| | - Timo N Kohler
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | - Tomasz S Kaminski
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Department of Molecular Biology, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Robert Hughes
- MRC Toxicology Unit, Gleeson Building, Cambridge, UK
| | | | | | - Paul Brear
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Nicholas J Matheson
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK
- NHS Blood and Transplant, Cambridge, UK
| | - Charlotte M Deane
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, Oxford, UK
| | - Marko Hyvönen
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | | |
Collapse
|
34
|
Pyles GM, Huckaby AB, Gutierrez MDLP, Witt WT, Mateu-Borrás M, Dublin SR, Rocuskie-Marker C, Sesti BN, Peasak K, Bitzer GJ, Rader N, Weaver KL, Boehm DT, Fitzgerald N, Chapman J, Ulicny S, Damron FH, Barbier M. Virus-like particles displaying the mature C-terminal domain of filamentous hemagglutinin are immunogenic and protective against Bordetella pertussis respiratory infection in mice. Infect Immun 2024; 92:e0027024. [PMID: 39023271 PMCID: PMC11320929 DOI: 10.1128/iai.00270-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/20/2024] Open
Abstract
Bordetella pertussis, the bacterium responsible for whooping cough, remains a significant public health challenge despite the existing licensed pertussis vaccines. Current acellular pertussis vaccines, though having favorable reactogenicity and efficacy profiles, involve complex and costly production processes. In addition, acellular vaccines have functional challenges such as short-lasting duration of immunity and limited antigen coverage. Filamentous hemagglutinin (FHA) is an adhesin of B. pertussis that is included in all multivalent pertussis vaccine formulations. Antibodies to FHA have been shown to prevent bacterial attachment to respiratory epithelial cells, and T cell responses to FHA facilitate cell-mediated immunity. In this study, FHA's mature C-terminal domain (MCD) was evaluated as a novel vaccine antigen. MCD was conjugated to virus-like particles via SpyTag-SpyCatcher technology. Prime-boost vaccine studies were performed in mice to characterize immunogenicity and protection against the intranasal B. pertussis challenge. MCD-SpyVLP was more immunogenic than SpyTag-MCD antigen alone, and in Tohama I strain challenge studies, improved protection against challenge was observed in the lungs at day 3 and in the trachea and nasal wash at day 7 post-challenge. Furthermore, a B. pertussis strain encoding genetically inactivated pertussis toxin was used to evaluate MCD-SpyVLP vaccine immunity. Mice vaccinated with MCD-SpyVLP had significantly lower respiratory bacterial burden at both days 3 and 7 post-challenge compared to mock-vaccinated animals. Overall, these data support the use of SpyTag-SpyCatcher VLPs as a platform for use in vaccine development against B. pertussis and other pathogens.
Collapse
Affiliation(s)
- Gage M. Pyles
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Annalisa B. Huckaby
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Maria de la Paz Gutierrez
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - William T. Witt
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Margalida Mateu-Borrás
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Spencer R. Dublin
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Carleena Rocuskie-Marker
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Bethany N. Sesti
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Kerrington Peasak
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Graham J. Bitzer
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Nathaniel Rader
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Kelly L. Weaver
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Dylan T. Boehm
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Nicholas Fitzgerald
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Joshua Chapman
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Samuel Ulicny
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - F. Heath Damron
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Mariette Barbier
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| |
Collapse
|
35
|
Went M, Duran-Lozano L, Halldorsson GH, Gunnell A, Ugidos-Damboriena N, Law P, Ekdahl L, Sud A, Thorleifsson G, Thodberg M, Olafsdottir T, Lamarca-Arrizabalaga A, Cafaro C, Niroula A, Ajore R, Lopez de Lapuente Portilla A, Ali Z, Pertesi M, Goldschmidt H, Stefansdottir L, Kristinsson SY, Stacey SN, Love TJ, Rognvaldsson S, Hajek R, Vodicka P, Pettersson-Kymmer U, Späth F, Schinke C, Van Rhee F, Sulem P, Ferkingstad E, Hjorleifsson Eldjarn G, Mellqvist UH, Jonsdottir I, Morgan G, Sonneveld P, Waage A, Weinhold N, Thomsen H, Försti A, Hansson M, Juul-Vangsted A, Thorsteinsdottir U, Hemminki K, Kaiser M, Rafnar T, Stefansson K, Houlston R, Nilsson B. Deciphering the genetics and mechanisms of predisposition to multiple myeloma. Nat Commun 2024; 15:6644. [PMID: 39103364 DOI: 10.1038/s41467-024-50932-7] [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: 01/03/2024] [Accepted: 07/24/2024] [Indexed: 08/07/2024] Open
Abstract
Multiple myeloma (MM) is an incurable malignancy of plasma cells. Epidemiological studies indicate a substantial heritable component, but the underlying mechanisms remain unclear. Here, in a genome-wide association study totaling 10,906 cases and 366,221 controls, we identify 35 MM risk loci, 12 of which are novel. Through functional fine-mapping and Mendelian randomization, we uncover two causal mechanisms for inherited MM risk: longer telomeres; and elevated levels of B-cell maturation antigen (BCMA) and interleukin-5 receptor alpha (IL5RA) in plasma. The largest increase in BCMA and IL5RA levels is mediated by the risk variant rs34562254-A at TNFRSF13B. While individuals with loss-of-function variants in TNFRSF13B develop B-cell immunodeficiency, rs34562254-A exerts a gain-of-function effect, increasing MM risk through amplified B-cell responses. Our results represent an analysis of genetic MM predisposition, highlighting causal mechanisms contributing to MM development.
Collapse
Affiliation(s)
- Molly Went
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Laura Duran-Lozano
- Department of Laboratory Medicine, Lund University, SE-221 84, Lund, Sweden
- Lund Stem Cell Center, Lund University, SE-221 84, Lund, Sweden
| | | | - Andrea Gunnell
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Nerea Ugidos-Damboriena
- Department of Laboratory Medicine, Lund University, SE-221 84, Lund, Sweden
- Lund Stem Cell Center, Lund University, SE-221 84, Lund, Sweden
| | - Philip Law
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Ludvig Ekdahl
- Department of Laboratory Medicine, Lund University, SE-221 84, Lund, Sweden
- Lund Stem Cell Center, Lund University, SE-221 84, Lund, Sweden
| | - Amit Sud
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| | | | - Malte Thodberg
- Department of Laboratory Medicine, Lund University, SE-221 84, Lund, Sweden
- Lund Stem Cell Center, Lund University, SE-221 84, Lund, Sweden
| | | | - Antton Lamarca-Arrizabalaga
- Department of Laboratory Medicine, Lund University, SE-221 84, Lund, Sweden
- Lund Stem Cell Center, Lund University, SE-221 84, Lund, Sweden
| | - Caterina Cafaro
- Department of Laboratory Medicine, Lund University, SE-221 84, Lund, Sweden
- Lund Stem Cell Center, Lund University, SE-221 84, Lund, Sweden
| | - Abhishek Niroula
- Department of Laboratory Medicine, Lund University, SE-221 84, Lund, Sweden
- Lund Stem Cell Center, Lund University, SE-221 84, Lund, Sweden
| | - Ram Ajore
- Department of Laboratory Medicine, Lund University, SE-221 84, Lund, Sweden
- Lund Stem Cell Center, Lund University, SE-221 84, Lund, Sweden
| | - Aitzkoa Lopez de Lapuente Portilla
- Department of Laboratory Medicine, Lund University, SE-221 84, Lund, Sweden
- Lund Stem Cell Center, Lund University, SE-221 84, Lund, Sweden
| | - Zain Ali
- Department of Laboratory Medicine, Lund University, SE-221 84, Lund, Sweden
- Lund Stem Cell Center, Lund University, SE-221 84, Lund, Sweden
| | - Maroulio Pertesi
- Department of Laboratory Medicine, Lund University, SE-221 84, Lund, Sweden
- Lund Stem Cell Center, Lund University, SE-221 84, Lund, Sweden
| | - Hartmut Goldschmidt
- Department of Internal Medicine V, University of Heidelberg, 69120, Heidelberg, Germany
| | | | - Sigurdur Y Kristinsson
- Landspitali, National University Hospital of Iceland, IS-101, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, IS-101, Reykjavik, Iceland
| | - Simon N Stacey
- deCODE Genetics/Amgen, Sturlugata 8, IS-101, Reykjavik, Iceland
| | - Thorvardur J Love
- Landspitali, National University Hospital of Iceland, IS-101, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, IS-101, Reykjavik, Iceland
| | - Saemundur Rognvaldsson
- Landspitali, National University Hospital of Iceland, IS-101, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, IS-101, Reykjavik, Iceland
| | - Roman Hajek
- University Hospital Ostrava and University of Ostrava, Ostrava, Czech Republic
| | - Pavel Vodicka
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | - Florentin Späth
- Department of Radiation Sciences, Umeå University, SE-901 87, Umeå, Sweden
| | - Carolina Schinke
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Frits Van Rhee
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Patrick Sulem
- deCODE Genetics/Amgen, Sturlugata 8, IS-101, Reykjavik, Iceland
| | | | | | | | | | - Gareth Morgan
- Perlmutter Cancer Center, Langone Health, New York University, New York, NY, USA
| | - Pieter Sonneveld
- Department of Hematology, Erasmus MC Cancer Institute, 3075 EA, Rotterdam, The Netherlands
| | - Anders Waage
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Box 8905, N-7491, Trondheim, Norway
| | - Niels Weinhold
- Department of Internal Medicine V, University of Heidelberg, 69120, Heidelberg, Germany
- German Cancer Research Center (DKFZ), D-69120, Heidelberg, Germany
| | | | - Asta Försti
- German Cancer Research Center (DKFZ), D-69120, Heidelberg, Germany
- Hopp Children's Cancer Center, Heidelberg, Germany
| | - Markus Hansson
- Department of Laboratory Medicine, Lund University, SE-221 84, Lund, Sweden
- Section of Hematology, Sahlgrenska University Hospital, Gothenburg, SE-413 45, Sweden
- Skåne University Hospital, SE-221 85, Lund, Sweden
| | - Annette Juul-Vangsted
- Department of Haematology, University Hospital of Copenhagen at Rigshospitalet, Blegdamsvej 9, DK-2100, Copenhagen, Denmark
| | - Unnur Thorsteinsdottir
- deCODE Genetics/Amgen, Sturlugata 8, IS-101, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, IS-101, Reykjavik, Iceland
| | - Kari Hemminki
- German Cancer Research Center (DKFZ), D-69120, Heidelberg, Germany
- Faculty of Medicine in Pilsen, Charles University, 30605, Pilsen, Czech Republic
| | - Martin Kaiser
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Thorunn Rafnar
- deCODE Genetics/Amgen, Sturlugata 8, IS-101, Reykjavik, Iceland
| | - Kari Stefansson
- deCODE Genetics/Amgen, Sturlugata 8, IS-101, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, IS-101, Reykjavik, Iceland
| | - Richard Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK.
| | - Björn Nilsson
- Department of Laboratory Medicine, Lund University, SE-221 84, Lund, Sweden.
- Lund Stem Cell Center, Lund University, SE-221 84, Lund, Sweden.
- Broad Institute, 415 Main Street, Cambridge, MA, 02142, USA.
| |
Collapse
|
36
|
Zhou Y, Gong J, Deng X, Shen L, Liu L. Novel insights: crosstalk with non-puerperal mastitis and immunity. Front Immunol 2024; 15:1431681. [PMID: 39148739 PMCID: PMC11324573 DOI: 10.3389/fimmu.2024.1431681] [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: 05/12/2024] [Accepted: 07/16/2024] [Indexed: 08/17/2024] Open
Abstract
The two primary types of non-puerperal mastitis (NPM) are granulomatous lobular mastitis (GLM) and plasma cell mastitis (PCM). Existing research indicates that immune inflammatory response is considered to be the core of the pathogenesis of GLM and PCM, and both innate and adaptive immune responses play an important role in the pathophysiology of PCM and GLM. However, the regulatory balance between various immune cells in these diseases is still unclear. Consequently, we present a comprehensive summary of the immune-related variables and recent advances in GLM and PCM.
Collapse
Affiliation(s)
- Yao Zhou
- Department of Galactophore, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jie Gong
- Department of Galactophore, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xianguang Deng
- Department of Galactophore, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lele Shen
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lifang Liu
- Department of Galactophore, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
37
|
Lanahan SM, Yang L, Jones KM, Qi Z, Cabrera EC, Cominsky LY, Ramaswamy A, Barmada A, Gabernet G, Uthaya Kumar DB, Xu L, Shan P, Wymann MP, Kleinstein SH, Rao VK, Mustillo P, Romberg N, Abraham RS, Lucas CL. PI3Kγ in B cells promotes antibody responses and generation of antibody-secreting cells. Nat Immunol 2024; 25:1422-1431. [PMID: 38961274 DOI: 10.1038/s41590-024-01890-1] [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: 07/06/2023] [Accepted: 06/07/2024] [Indexed: 07/05/2024]
Abstract
The differentiation of naive and memory B cells into antibody-secreting cells (ASCs) is a key feature of adaptive immunity. The requirement for phosphoinositide 3-kinase-delta (PI3Kδ) to support B cell biology has been investigated intensively; however, specific functions of the related phosphoinositide 3-kinase-gamma (PI3Kγ) complex in B lineage cells have not. In the present study, we report that PI3Kγ promotes robust antibody responses induced by T cell-dependent antigens. The inborn error of immunity caused by human deficiency in PI3Kγ results in broad humoral defects, prompting our investigation of roles for this kinase in antibody responses. Using mouse immunization models, we found that PI3Kγ functions cell intrinsically within activated B cells in a kinase activity-dependent manner to transduce signals required for the transcriptional program supporting differentiation of ASCs. Furthermore, ASC fate choice coincides with upregulation of PIK3CG expression and is impaired in the context of PI3Kγ disruption in naive B cells on in vitro CD40-/cytokine-driven activation, in memory B cells on toll-like receptor activation, or in human tonsillar organoids. Taken together, our study uncovers a fundamental role for PI3Kγ in supporting humoral immunity by integrating signals instructing commitment to the ASC fate.
Collapse
Affiliation(s)
- Stephen M Lanahan
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Lucas Yang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Kate M Jones
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Zhihong Qi
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Emylette Cruz Cabrera
- Division of Immunology and Allergy, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lauren Y Cominsky
- Immunology Graduate Group, Perelman School of Medicine, Philadelphia, PA, USA
| | - Anjali Ramaswamy
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Anis Barmada
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Gisela Gabernet
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Lan Xu
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Peiying Shan
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Steven H Kleinstein
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - V Koneti Rao
- Primary Immunodeficiency Clinic, Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
| | - Peter Mustillo
- Division of Allergy and Immunology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, OH, USA
| | - Neil Romberg
- Division of Immunology and Allergy, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, Philadelphia, PA, USA
| | - Roshini S Abraham
- Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - Carrie L Lucas
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
38
|
Shi X, Xiao B, Feng R. Identification of a glycolysis-related miRNA Signature for Predicting Breast cancer Survival. Mol Biotechnol 2024; 66:1988-2006. [PMID: 37535159 DOI: 10.1007/s12033-023-00837-5] [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: 02/26/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023]
Abstract
Breast cancer (BC) is a common type of cancer and has a poor prognosis. In this study, we collected the mRNA and miRNA expression profiles of BC patients were obtained from The Cancer Genome Atlas (TCGA) to explore a novel prognostic strategy for BC patients using bioinformatics tools. We found that six glycolysis-related miRNAs (GRmiRs, including hsa-mir-1247, hsa-mir148b, hsa-mir-133a-2, has-mir-1307, hsa-mir-195 and hsa-mir-1258) were correlated with prognosis of BC samples. The risk score model was established based on 6 prognosis-associated GRmiRs. The outcome of high risk group was significantly poorer. Cox regression analysis showed that risk score was an independent prognostic factor. Differentially expressed genes identified between high and low risk groups were mainly enriched in inflammation and immune-related signaling pathways. The proportion of infiltration of 12 kinds of immune cells in high and low risk groups were significantly different. Risk score was closely associated with many immune indexes. Multiple DEGRGs and miRNAs were associated with drugs. In conclusion, glycolysis-related miRNA signature effectively predicts BC prognosis.
Collapse
Affiliation(s)
- Xuejing Shi
- Department of Galactophore, Tianjin Central Hospital of Gynecology and Obstetrics, No. 156 Nankai Sanma Road, Tianjin, Nankai District, 300100, P.R. China
| | - Baoqiang Xiao
- Department of General Surgery, Tianjin Hospital, Tianjin, Hexi District, 300211, P.R. China
| | - Rui Feng
- Department of Galactophore, Tianjin Central Hospital of Gynecology and Obstetrics, No. 156 Nankai Sanma Road, Tianjin, Nankai District, 300100, P.R. China.
| |
Collapse
|
39
|
Alaterre E, Ovejero S, Bret C, Dutrieux L, Sika D, Fernandez Perez R, Espéli M, Fest T, Cogné M, Martin-Subero JI, Milpied P, Cavalli G, Moreaux J. Integrative single-cell chromatin and transcriptome analysis of human plasma cell differentiation. Blood 2024; 144:496-509. [PMID: 38643512 PMCID: PMC11406183 DOI: 10.1182/blood.2023023237] [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: 11/13/2023] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/23/2024] Open
Abstract
ABSTRACT Plasma cells (PCs) are highly specialized cells representing the end stage of B-cell differentiation. We have shown that PC differentiation can be reproduced in vitro using elaborate culture systems. The molecular changes occurring during PC differentiation are recapitulated in this in vitro differentiation model. However, a major challenge exists to decipher the spatiotemporal epigenetic and transcriptional programs that drive the early stages of PC differentiation. We combined single cell (sc) RNA sequencing (RNA-seq) and assay for transposase-accessible chromatin with high throughput sequencing (scATAC-seq) to decipher the trajectories involved in PC differentiation. ScRNA-seq experiments revealed a strong heterogeneity of the preplasmablastic and plasmablastic stages. Among genes that were commonly identified using scATAC-seq and scRNA-seq, we identified several transcription factors with significant stage specific potential importance in PC differentiation. Interestingly, differentially accessible peaks characterizing the preplasmablastic stage were enriched in motifs of BATF3, FOS and BATF, belonging to activating protein 1 (AP-1) transcription factor family that may represent key transcriptional nodes involved in PC differentiation. Integration of transcriptomic and epigenetic data at the single cell level revealed that a population of preplasmablasts had already undergone epigenetic remodeling related to PC profile together with unfolded protein response activation and are committed to differentiate in PC. These results and the supporting data generated with our in vitro PC differentiation model provide a unique resource for the identification of molecular circuits that are crucial for early and mature PC maturation and biological functions. These data thus provide critical insights into epigenetic- and transcription-mediated reprogramming events that sustain PC differentiation.
Collapse
Affiliation(s)
- Elina Alaterre
- Institute of Human Genetics, Unité Mixte de Recherche, Centre National de la Recherche Scientifique, Université Montpellier, Montpellier, France
| | - Sara Ovejero
- Institute of Human Genetics, Unité Mixte de Recherche, Centre National de la Recherche Scientifique, Université Montpellier, Montpellier, France
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
| | - Caroline Bret
- Institute of Human Genetics, Unité Mixte de Recherche, Centre National de la Recherche Scientifique, Université Montpellier, Montpellier, France
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
| | - Laure Dutrieux
- Institute of Human Genetics, Unité Mixte de Recherche, Centre National de la Recherche Scientifique, Université Montpellier, Montpellier, France
| | - Dassou Sika
- Institute of Human Genetics, Unité Mixte de Recherche, Centre National de la Recherche Scientifique, Université Montpellier, Montpellier, France
| | | | - Marion Espéli
- INSERM U1160 EMiLy, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris, France
| | - Thierry Fest
- Université de Rennes 1, INSERM, Établissement Français du Sang de Bretagne, Team B_DEVIL, UMR_S1236, Rennes, France
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire, Rennes, France
| | - Michel Cogné
- Institut National de La Santé et de La Recherche Médicale, Unité Mixte de Recherche U1236, Université de Rennes, Etablissement Français Du Sang Bretagne, Rennes, France
- Centre Hospitalier Universitaire de Rennes, Suivi Immunologique des Thérapies Innovantes, Pôle Biologie, Rennes, France
| | - José Ignacio Martin-Subero
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Departament de Fonaments Clínics, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Pierre Milpied
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Giacomo Cavalli
- Institute of Human Genetics, Unité Mixte de Recherche, Centre National de la Recherche Scientifique, Université Montpellier, Montpellier, France
| | - Jérôme Moreaux
- Institute of Human Genetics, Unité Mixte de Recherche, Centre National de la Recherche Scientifique, Université Montpellier, Montpellier, France
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
- University of Montpellier, UFR Medicine, Montpellier, France
- Institut Universitaire de France, Paris, France
| |
Collapse
|
40
|
Qiu GH, Yu B, Ma M. G protein-coupled receptor-mediated signaling of immunomodulation in tumor progression. FASEB J 2024; 38:e23829. [PMID: 39017658 DOI: 10.1096/fj.202400458r] [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: 03/01/2024] [Revised: 06/07/2024] [Accepted: 07/08/2024] [Indexed: 07/18/2024]
Abstract
G protein-coupled receptors (GPCRs) are essential contributors to tumor growth and metastasis due to their roles in immune cell regulation. Therefore, GPCRs are potential targets for cancer immunotherapy. Here, we discuss the current understanding of the roles of GPCRs and their signaling pathways in tumor progression from an immunocellular perspective. Additionally, we focus on the roles of GPCRs in regulating immune checkpoint proteins involved in immune evasion. Finally, we review the progress of clinical trials of GPCR-targeted drugs for cancer treatment, which may be combined with immunotherapy to improve treatment efficacy. This expanded understanding of the role of GPCRs may shed light on the mechanisms underlying tumor progression and provide a novel perspective on cancer immunotherapy.
Collapse
Affiliation(s)
- Guang-Hong Qiu
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, PR China
| | - Bin Yu
- Department of General Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, PR China
| | - Mei Ma
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, PR China
| |
Collapse
|
41
|
Kim J, Maharjan R, Park J. Current Trends and Innovative Approaches in Cancer Immunotherapy. AAPS PharmSciTech 2024; 25:168. [PMID: 39044047 DOI: 10.1208/s12249-024-02883-x] [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: 01/30/2024] [Accepted: 07/05/2024] [Indexed: 07/25/2024] Open
Abstract
Immunotherapy is one of the most promising therapeutic approaches in the field of cancer treatment. As a tumor progresses, tumor cells employ an array of immune-regulatory mechanisms to suppress immune responses within the tumor microenvironment. Using our understanding of these mechanisms, cancer immunotherapy has been developed to enhance the immune system's effectiveness in treating cancer. Numerous cancer immunotherapies are currently in clinical use, yet many others are either in different stages of development or undergoing clinical studies. In this paper, we briefly discuss the features and current status of cancer immunotherapies. This includes the application of monoclonal antibodies, immune checkpoint inhibitors, adoptive cell therapy, cytokine therapy, cancer vaccines, and gene therapy, all of which have gained significant recognition in clinical practice. Additionally, we discuss limitations that may hinder successful clinical utilization and promising strategies, such as combining immunotherapy with nanotechnology.
Collapse
Affiliation(s)
- Jaechang Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40506, USA
| | - Ruby Maharjan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40506, USA
| | - Jonghyuck Park
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40506, USA.
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY, USA.
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
42
|
Eslami M, Schuepbach-Mallepell S, Diana D, Willen L, Kowalczyk-Quintas C, Desponds C, Peter B, Vigolo M, Renevey F, Donzé O, Luther SA, Yalkinoglu Ö, Alouche N, Schneider P. Unique and redundant roles of mouse BCMA, TACI, BAFF, APRIL, and IL-6 in supporting antibody-producing cells in different tissues. Proc Natl Acad Sci U S A 2024; 121:e2404309121. [PMID: 38990948 PMCID: PMC11260164 DOI: 10.1073/pnas.2404309121] [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: 03/05/2024] [Accepted: 06/03/2024] [Indexed: 07/13/2024] Open
Abstract
Antibody-producing plasma cells fuel humoral immune responses. They also contribute to autoimmune diseases such as systemic lupus erythematosus or IgA nephropathy. Interleukin-6 and the tumor necrosis factor (TNF) family ligands BAFF (B cell-activating factor) and APRIL (a proliferation-inducing ligand) participate in plasma cell survival. BAFF binds to three receptors, BAFFR (BAFF receptor), TACI (transmembrane activator and CAML interactor), and BCMA (B cell maturation antigen), while APRIL binds to TACI, BCMA, and proteoglycans. However, which ligand-receptor pair(s) are required to maintain plasma cells in different body locations remains unknown. Here, by combining mouse genetic and pharmacological approaches, we found that plasma cells required BCMA and/or TACI but not BAFFR. BCMA responded exclusively to APRIL, while TACI responded to both BAFF and APRIL, identifying three self-sufficient ligand-receptor pairs for plasma cell maintenance: BAFF-TACI, APRIL-TACI, and APRIL-BCMA. Together, these actors accounted for 90% of circulating antibodies. In BAFF-ko mice, the reduction of plasma cells upon APRIL inhibition indicated that APRIL could function in the absence of BAFF-APRIL heteromers. No evidence was found that in the absence of BCMA and TACI, binding of APRIL to proteoglycans would help maintain plasma cells. IL-6, alone or together with BAFF and APRIL, supported mainly splenic plasmablasts and plasma cells and contributed to circulating IgG but not IgA levels. In conclusion, survival factors for plasma cells can vary with body location and with the antibody isotype that plasma cells produce. To efficiently target plasma cells, in particular IgA-producing ones, dual inhibition of BAFF and APRIL is required.
Collapse
Affiliation(s)
- Mahya Eslami
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | | | - Daniela Diana
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | - Laure Willen
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | | | - Chantal Desponds
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | - Benjamin Peter
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | - Michele Vigolo
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | - François Renevey
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | | | - Sanjiv A. Luther
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | - Özkan Yalkinoglu
- Clinical Pharmacology, Global Early Development, The Healthcare Business of Merck KGaA, Darmstadt64293, Germany
| | - Nagham Alouche
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | - Pascal Schneider
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| |
Collapse
|
43
|
Armstrong D, Chang CY, Hong MJ, Green L, Hudson W, Shen Y, Song LZ, Jammi S, Casal B, Creighton CJ, Carisey A, Zhang XHF, McKenna NJ, Kang SW, Lee HS, Corry DB, Kheradmand F. MAGE-A4-Responsive Plasma Cells Promote Non-Small Cell Lung Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.602985. [PMID: 39071307 PMCID: PMC11275715 DOI: 10.1101/2024.07.10.602985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Adaptive immunity is critical to eliminate malignant cells, while multiple tumor-intrinsic factors can alter this protective function. Melanoma antigen-A4 (MAGE-A4), a cancer-testis antigen, is expressed in several solid tumors and correlates with poor survival in non-small cell lung cancer (NSCLC), but its role in altering antitumor immunity remains unclear. We found that expression of MAGE-A4 was highly associated with the loss of PTEN , a tumor suppressor, in human NSCLC. Here we show that constitutive expression of human MAGE-A4 combined with the loss of Pten in mouse airway epithelial cells results in metastatic adenocarcinoma enriched in CD138 + CXCR4 + plasma cells, predominantly expressing IgA. Consistently, human NSCLC expressing MAGE-A4 showed increased CD138 + IgA + plasma cell density surrounding tumors. The abrogation of MAGE-A4-responsive plasma cells (MARPs) decreased tumor burden, increased T cell infiltration and activation, and reduced CD163 + CD206 + macrophages in mouse lungs. These findings suggest MAGE-A4 promotes NSCLC tumorigenesis, in part, through the recruitment and retention of IgA + MARPs in the lungs.
Collapse
|
44
|
Martini S, Drzeniek NM, Stark R, Kollert MR, Du W, Reinke S, Ort M, Hardt S, Kotko I, Kath J, Schlickeiser S, Geißler S, Wagner DL, Krebs AC, Volk HD. Long-term in vitromaintenance of plasma cells in a hydrogel-enclosed human bone marrow microphysiological 3D model system. Biofabrication 2024; 16:045005. [PMID: 38955197 DOI: 10.1088/1758-5090/ad5dfe] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 07/01/2024] [Indexed: 07/04/2024]
Abstract
Plasma cells (PCs) in bone marrow (BM) play an important role in both protective and pathogenic humoral immune responses, e.g. in various malignant and non-malignant diseases such as multiple myeloma, primary and secondary immunodeficiencies and autoimmune diseases. Dedicated microenvironmental niches in the BM provide PCs with biomechanical and soluble factors that support their long-term survival. There is a high need for appropriate and robust model systems to better understand PCs biology, to develop new therapeutic strategies for PCs-related diseases and perform targeted preclinical studies with high predictive value. Most preclinical data have been derived fromin vivostudies in mice, asin vitrostudies of human PCs are limited due to restricted survival and functionality in conventional 2D cultures that do not reflect the unique niche architecture of the BM. We have developed a microphysiological, dynamic 3D BM culture system (BM-MPS) based on human primary tissue (femoral biopsies), mechanically supported by a hydrogel scaffold casing. While a bioinert agarose casing did not support PCs survival, a photo-crosslinked collagen-hyaluronic acid (Col-HA) hydrogel preserved the native BM niche architecture and allowed PCs survivalin vitrofor up to 2 weeks. Further, the Col-HA hydrogel was permissive to lymphocyte migration into the microphysiological system´s circulation. Long-term PCs survival was related to the stable presence in the culture of soluble factors, as APRIL, BAFF, and IL-6. Increasing immunoglobulins concentrations in the medium confirm their functionality over culture time. To the best of our knowledge, this study is the first report of successful long-term maintenance of primary-derived non-malignant PCsin vitro. Our innovative model system is suitable for in-depthin vitrostudies of human PCs regulation and exploration of targeted therapeutic approaches such as CAR-T cell therapy or biologics.
Collapse
Affiliation(s)
- Stefania Martini
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Norman Michael Drzeniek
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Regina Stark
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Matthias Reiner Kollert
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Julius Wolff Institute, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Weijie Du
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Simon Reinke
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Melanie Ort
- Julius Wolff Institute, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Sebastian Hardt
- Center for Musculoskeletal Surgery Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Iuliia Kotko
- Institute of Medical Immunology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Jonas Kath
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Stephan Schlickeiser
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- CheckImmune GmbH, Berlin, Germany
| | - Sven Geißler
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Julius Wolff Institute, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Dimitrios Laurin Wagner
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Institute of Transfusion Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Anna-Catharina Krebs
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Hans-Dieter Volk
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
45
|
Gu D, Lim J, Han KY, Seo IH, Jee JH, Cho SJ, Choi YH, Choi SC, Koh JH, Lee JY, Kang M, Jung DH, Park WY. Single-cell analysis of human PBMCs in healthy and type 2 diabetes populations: dysregulated immune networks in type 2 diabetes unveiled through single-cell profiling. Front Endocrinol (Lausanne) 2024; 15:1397661. [PMID: 39072276 PMCID: PMC11272961 DOI: 10.3389/fendo.2024.1397661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/27/2024] [Indexed: 07/30/2024] Open
Abstract
Abnormalities in glucose metabolism that precede the onset of type 2 diabetes (T2D) activate immune cells, leading to elevated inflammatory factors and chronic inflammation. However, no single-cell RNA sequencing (scRNA-seq) studies have characterized the properties and networks of individual immune cells in T2D. Here, we analyzed peripheral blood mononuclear cells (PBMCs) from non-diabetes and T2D patients by scRNA-seq. We found that CD14 monocytes in T2D patients were in a pro-inflammatory state and intermediate monocytes expressed more MHC class II genes. In T2D patients, cytotoxic CD4 T cells, effector memory CD8 T cells, and γδ T cells have increased cytotoxicity and clonal expansion. B cells were characterized by increased differentiation into intermediate B cells, plasma cells, and isotype class switching with increased expression of soluble antibody genes. These results suggest that monocytes, T cells, and B cells could interact to induce chronic inflammation in T2D patients with pro-inflammatory characteristics.
Collapse
Affiliation(s)
- Doeon Gu
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea
- Samsung Genome Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Jinyeong Lim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea
- Samsung Genome Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Kyung Yeon Han
- Samsung Genome Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - In-Ho Seo
- Department of Biomedical Science, College of Life Science CHA University, Gyeonggi-do, Republic of Korea
| | - Jae Hwan Jee
- Department of Health Promotion Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Soo Jin Cho
- Department of Health Promotion Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yoon Ho Choi
- Department of Health Promotion Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sung Chul Choi
- Department of Health Promotion Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jang Hyun Koh
- Department of Health Promotion Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jin-Young Lee
- Department of Health Promotion Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Mira Kang
- Department of Health Promotion Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Digital Health, Samsung Advanced Institute of Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
- Digital Transformation Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Dong-Hyuk Jung
- Department of Family Medicine, Yongin Severance Hospital, Gyeonggi-do, Republic of Korea
| | - Woong-Yang Park
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea
- Samsung Genome Institute, Samsung Medical Center, Seoul, Republic of Korea
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
46
|
Chen Y, Jiang B, Qu C, Jiang C, Zhang C, Wang Y, Chen F, Sun X, Su L, Luo Y. Genetically predicted metabolites mediate the causal associations between autoimmune thyroiditis and immune cells. Front Endocrinol (Lausanne) 2024; 15:1424957. [PMID: 39045270 PMCID: PMC11263034 DOI: 10.3389/fendo.2024.1424957] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
Introduction We aimed to comprehensively investigate the causal relationship between 731 immune cell traits and autoimmune thyroiditis (AIT) and to identify and quantify the role of 1400 metabolic traits as potential mediators in between. Methods Using summary-level data from genome-wide association studies (GWAS) we performed a two-sample bidirectional Mendelian randomization (MR) analysis of genetically predicted AIT and 731 immune cell traits. Furthermore, we used a two-step MR analysis to quantify the proportion of the total effects (that the immune cells exerted on the risk of AIT) mediated by potential metabolites. Results We identified 24 immune cell traits (with odds ratio (OR) ranging from 1.3166 6 to 0.6323) and 10 metabolic traits (with OR ranging from 1.7954 to 0.6158) to be causally associated with AIT, respectively. Five immune cell traits (including CD38 on IgD+ CD24-, CD28 on CD28+ CD45RA+ CD8br, HLA DR+ CD4+ AC, TD CD4+ %CD4+, and CD8 on EM CD8br) were found to be associated with the risk of AIT, which were partially mediated by metabolites (including glycolithocholate sulfate, 5alpha-androstan-3alpha,17beta-diol disulfate, arachidonoylcholine, X-15486, and kynurenine). The proportion of genetically predicted AIT mediated by the identified metabolites could range from 5.58% to 17.7%. Discussion Our study identified causal associations between AIT and immune cells which were partially mediated by metabolites, thus providing guidance for future clinical and basic research.
Collapse
Affiliation(s)
- Yongzhao Chen
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Bo Jiang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Cheng Qu
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Chaoyu Jiang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Chen Zhang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yanxue Wang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Fei Chen
- General Surgery Center, Department of Thyroid Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xitai Sun
- Division of Pancreas and Metabolism Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Lei Su
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yuqian Luo
- Clinical Medicine Research Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
47
|
Noble J, Cabezas L, Truffot A, Dumolard L, Jouve T, Malvezzi P, Rostaing L, Dard C, Saas P, Cravedi P, Macek-Jilkova Z. Glycolysis Changes in Alloreactive Memory B Cells in Highly Sensitized Kidney Transplant Recipients Undergonig Desensitization Therapy. Transpl Int 2024; 37:13029. [PMID: 39081904 PMCID: PMC11287219 DOI: 10.3389/ti.2024.13029] [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: 03/23/2024] [Accepted: 06/25/2024] [Indexed: 08/02/2024]
Abstract
Despite the growing use of desensitization strategies, hyperimmune patients remain at high risk of antibody-mediated rejection suggesting that, even when donor-specific antibodies (DSA) are effectively depleted, anti-donor specific B cells persist. We included 10 highly sensitized recipients that underwent desensitization with plasmapheresis and B cell depletion prior to kidney transplantation. We quantified changes in DSA (luminex), total B-cell subsets (flow cytometry), anti-donor HLA B cells (fluorospot), and single-cell metabolism in serially collected samples before desensitization, at the time of transplant, and at 6 and 12 months thereafter. Desensitization was associated with a decrease in DSA and total memory B cell and naive B cell percentage, while plasma cells and memory anti-donor HLA circulating B cells persisted up to 12 months after transplant. At 12-month post-transplantation, memory B cells increased their glycolytic capacity, while proliferative KI67+ plasma cells modified their metabolism by increasing fatty acid and amino acid oxidation capacity and decreasing their glucose dependence. Despite effective DSA depletion, anti-donor B cells persist in kidney transplant recipients. Due to the reliance of these cells on glycolysis, glycolysis-targeting therapies might represent a valuable treatment strategy.
Collapse
Affiliation(s)
- Johan Noble
- Nephrology, Hemodialysis Apheresis and Kidney Transplantation, Department, CHU Grenoble Alpes, Grenoble, France
- University Grenoble Alpes, CNRS, Inserm, CHU Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
- Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Lara Cabezas
- Nephrology, Hemodialysis Apheresis and Kidney Transplantation, Department, CHU Grenoble Alpes, Grenoble, France
- University Grenoble Alpes, CNRS, Inserm, CHU Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
| | - Aurelie Truffot
- Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Virology Department, University Hospital Grenoble, Grenoble, France
| | - Lucile Dumolard
- University Grenoble Alpes, CNRS, Inserm, CHU Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
| | - Thomas Jouve
- Nephrology, Hemodialysis Apheresis and Kidney Transplantation, Department, CHU Grenoble Alpes, Grenoble, France
- University Grenoble Alpes, CNRS, Inserm, CHU Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
| | - Paolo Malvezzi
- Nephrology, Hemodialysis Apheresis and Kidney Transplantation, Department, CHU Grenoble Alpes, Grenoble, France
| | - Lionel Rostaing
- Nephrology, Hemodialysis Apheresis and Kidney Transplantation, Department, CHU Grenoble Alpes, Grenoble, France
- University Grenoble Alpes, CNRS, Inserm, CHU Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
| | - Céline Dard
- EFS, Recherche et Développement, Grenoble, France
| | - Philippe Saas
- University Grenoble Alpes, CNRS, Inserm, CHU Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
- EFS, Recherche et Développement, Grenoble, France
| | - Paolo Cravedi
- Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Zuzana Macek-Jilkova
- University Grenoble Alpes, CNRS, Inserm, CHU Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
- Hepato-Gastroenterology and Digestive Oncology Department, CHU Grenoble Alpes, Grenoble, France
| |
Collapse
|
48
|
Ziaka M, Exadaktylos A. Gut-derived immune cells and the gut-lung axis in ARDS. Crit Care 2024; 28:220. [PMID: 38965622 PMCID: PMC11225303 DOI: 10.1186/s13054-024-05006-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 06/26/2024] [Indexed: 07/06/2024] Open
Abstract
The gut serves as a vital immunological organ orchestrating immune responses and influencing distant mucosal sites, notably the respiratory mucosa. It is increasingly recognized as a central driver of critical illnesses, with intestinal hyperpermeability facilitating bacterial translocation, systemic inflammation, and organ damage. The "gut-lung" axis emerges as a pivotal pathway, where gut-derived injurious factors trigger acute lung injury (ALI) through the systemic circulation. Direct and indirect effects of gut microbiota significantly impact immune responses. Dysbiosis, particularly intestinal dysbiosis, termed as an imbalance of microbial species and a reduction in microbial diversity within certain bodily microbiomes, influences adaptive immune responses, including differentiating T regulatory cells (Tregs) and T helper 17 (Th17) cells, which are critical in various lung inflammatory conditions. Additionally, gut and bone marrow immune cells impact pulmonary immune activity, underscoring the complex gut-lung interplay. Moreover, lung microbiota alterations are implicated in diverse gut pathologies, affecting local and systemic immune landscapes. Notably, lung dysbiosis can reciprocally influence gut microbiota composition, indicating bidirectional gut-lung communication. In this review, we investigate the pathophysiology of ALI/acute respiratory distress syndrome (ARDS), elucidating the role of immune cells in the gut-lung axis based on recent experimental and clinical research. This exploration aims to enhance understanding of ALI/ARDS pathogenesis and to underscore the significance of gut-lung interactions in respiratory diseases.
Collapse
Affiliation(s)
- Mairi Ziaka
- Clinic of Geriatric Medicine, Center of Geriatric Medicine and Rehabilitation, Kantonsspital Baselland, Bruderholz, Switzerland.
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland.
| | - Aristomenis Exadaktylos
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
49
|
Li Y, Li YJ, Fang X, Chen DQ, Yu WQ, Zhu ZQ. Peripheral inflammation as a potential mechanism and preventive strategy for perioperative neurocognitive disorder under general anesthesia and surgery. Front Cell Neurosci 2024; 18:1365448. [PMID: 39022312 PMCID: PMC11252726 DOI: 10.3389/fncel.2024.1365448] [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: 01/04/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
General anesthesia, as a commonly used medical intervention, has been widely applied during surgical procedures to ensure rapid loss of consciousness and pain relief for patients. However, recent research suggests that general anesthesia may be associated with the occurrence of perioperative neurocognitive disorder (PND). PND is characterized by a decline in cognitive function after surgery, including impairments in attention, memory, learning, and executive functions. With the increasing trend of population aging, the burden of PND on patients and society's health and economy is becoming more evident. Currently, the clinical consensus tends to believe that peripheral inflammation is involved in the pathogenesis of PND, providing strong support for further investigating the mechanisms and prevention of PND.
Collapse
Affiliation(s)
- Yuan Li
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Anesthesiology, Mianyang Hospital of Traditional Chinese Medicine, Mianyang, China
| | - Ying-Jie Li
- Department of General Surgery, Mianyang Hospital of Traditional Chinese Medicine, Mianyang, China
| | - Xu Fang
- Department of Anesthesiology, Nanchong Central Hospital, The Second Clinical Medical School of North Sichuan Medical College, Zunyi, China
| | - Dong-Qin Chen
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Wan-Qiu Yu
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhao-Qiong Zhu
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Early Clinical Research Ward of Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
50
|
Labeur-Iurman L, Harker JA. Mechanisms of antibody mediated immunity - Distinct in early life. Int J Biochem Cell Biol 2024; 172:106588. [PMID: 38768890 DOI: 10.1016/j.biocel.2024.106588] [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] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/22/2024]
Abstract
Immune responses in early life are characterized by a failure to robustly generate long-lasting protective responses against many common pathogens or upon vaccination. This is associated with a reduced ability to generate T-cell dependent high affinity antibodies. This review highlights the differences in T-cell dependent antibody responses observed between infants and adults, in particular focussing on the alterations in immune cell function that lead to reduced T follicular helper cell-B cell crosstalk within germinal centres in early life. Understanding the distinct functional characteristics of early life humoral immunity, and how these are regulated, will be critical in guiding age-appropriate immunological interventions in the very young.
Collapse
Affiliation(s)
- Lucia Labeur-Iurman
- National Heart & Lung Institute, Imperial College London, London, United Kingdom.
| | - James A Harker
- National Heart & Lung Institute, Imperial College London, London, United Kingdom; Centre for Paediatrics and Child Health, Imperial College London, London, United Kingdom.
| |
Collapse
|