151
|
Wei Y, Huang CX, Xiao X, Chen DP, Shan H, He H, Kuang DM. B cell heterogeneity, plasticity, and functional diversity in cancer microenvironments. Oncogene 2021; 40:4737-4745. [PMID: 34188249 DOI: 10.1038/s41388-021-01918-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/14/2021] [Accepted: 06/17/2021] [Indexed: 02/08/2023]
Abstract
B cells constitute a major component of tumor-infiltrating leukocytes. However, the influence of these cells on malignancy is currently under debate, reflecting the heterogeneity of B cell subsets in tumors. With recent advances, it becomes apparent that this debate includes not only the evaluation of B cells themselves, but also the underlying immune microenvironment network, which scripts the highly heterogeneous B cell populations in tumors and directs the roles of those sub-populations in disease progression and clinical treatment. In this review, we summarize recent findings on the heterogeneous subset composition of B cells in both human and mouse tumor models and their different impacts on disease progression. We further describe the multidimensional interplays between B cells and other immune cells in the tumor microenvironment, which account for the regulation of B cell differentiation and function in situ. We also assess the potential influences of distinct sub-tumor locations on B cell function in primary tumors during development and those under immunotherapy treatment. Illuminating the heterogeneous nature of B cell subset composition, generation, localization, and related immune network in tumor is of immense significance for comprehensively understanding B cell response in tumor and designing more efficacious cancer immunotherapies.
Collapse
Affiliation(s)
- Yuan Wei
- The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Chun-Xiang Huang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiao Xiao
- Cancer Program, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Dong-Ping Chen
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Hong Shan
- The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.
| | - Huanhuan He
- The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.
| | - Dong-Ming Kuang
- The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
152
|
Eslami M, Schneider P. Function, occurrence and inhibition of different forms of BAFF. Curr Opin Immunol 2021; 71:75-80. [PMID: 34182216 DOI: 10.1016/j.coi.2021.06.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 05/26/2021] [Accepted: 06/06/2021] [Indexed: 01/27/2023]
Abstract
B cell activating factor (BAFF or BLyS), an important cytokine for B cell survival and humoral immune responses, is targeted in the clinic for the treatment of systemic lupus erythematosus. This review focuses on the structure, function and inhibition profiles of membrane-bound BAFF, soluble BAFF 3-mer and soluble BAFF 60-mer, all of which have distinct properties. BAFF contains a loop region not required for receptor binding but essential for receptor activation via promotion of BAFF-to-BAFF contacts. This loop region additionally allows formation of BAFF 60-mer, in which epitopes of the BAFF inhibitor belimumab are inaccessible. If 60-mer forms in humans, it is predicted to be short-lived and to act locally because adult serum contains a BAFF 60-mer dissociating activity. Cord blood contains elevated levels of BAFF, part of which displays attributes of 60-mer, suggesting a role for this form of BAFF in the development of foetal or neonate B cells.
Collapse
Affiliation(s)
- Mahya Eslami
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland.
| |
Collapse
|
153
|
McAllister E, Jellusova J. BAFF signaling in B cell metabolism. Curr Opin Immunol 2021; 71:69-74. [PMID: 34174517 DOI: 10.1016/j.coi.2021.05.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/30/2022]
Abstract
BAFF is an essential cytokine primarily known for its role in maintaining B cell homeostasis via induction of a pro-survival gene expression profile. Additionally, recent evidence suggests that BAFF induced signaling also drives a metabolic program that is needed for homeostatic cell mass maintenance in resting B cells and which increases the cells' capacity to divide. Many components of the signaling cascades initiated by BAFF, the alternative NFκB pathway and the PI3K/AKT/mTOR pathway, are active in roles beyond their classically assigned function. These components can directly or indirectly impact metabolic reprogramming. Further exploration of the role BAFF signaling plays in B cell metabolism could help to identify metabolic vulnerabilities of hyperactive B cells in the context of autoimmunity.
Collapse
Affiliation(s)
- Ellen McAllister
- Institute of Biology III at the Faculty of Biology, Albert-Ludwigs-University of Freiburg, Schänzlestr. 1, 79104 Freiburg, Germany
| | - Julia Jellusova
- Institute for Clinical Chemistry and Pathobiochemistry, Klinikum Rechts der Isar, School of Medicine, Technical University Munich, Ismaningerstr. 22, 81675 Munich, Germany; TranslaTUM, Center for Translational Cancer Research, Technical University Munich, Einsteinstr.25, 81675 Munich, Germany.
| |
Collapse
|
154
|
Oniszczuk J, Beldi-Ferchiou A, Audureau E, Azzaoui I, Molinier-Frenkel V, Frontera V, Karras A, Moktefi A, Pillebout E, Zaidan M, El Karoui K, Delfau-Larue MH, Hénique C, Ollero M, Sahali D, Mahévas M, Audard V. Circulating plasmablasts and high level of BAFF are hallmarks of minimal change nephrotic syndrome in adults. Nephrol Dial Transplant 2021; 36:609-617. [PMID: 33241414 DOI: 10.1093/ndt/gfaa279] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The recent success achieved with the use of B cell-depleting agents in some patients with minimal change nephrotic syndrome (MCNS) suggests an unexpected role for B lymphocytes in the pathogenesis of this immune-mediated glomerular disease. Nevertheless, no extensive B-cell phenotyping analysis has ever been performed in untreated adult patients soon after MCNS diagnosis. METHODS We investigated the distribution of the different B-cell subpopulations in 22 untreated adult patients with biopsy-proven MCNS [MCNS relapse (MCNS-Rel)]. We compared these data with those for 24 healthy controls, 13 MCNS patients in remission (with no specific treatment) and 19 patients with idiopathic membranous nephropathy (IMN). RESULTS Patients with MCNS-Rel or IMN had higher proteinuria and lower serum albumin and gammaglobulin levels (P < 0.0001 for all comparisons) than MCNS patients in remission. Plasmablasts were the only B-cell subsets present at significantly higher levels in MCNS-Rel patients than in the patients of the other three groups (P < 0.05 for all comparisons). The lower albumin levels and higher proteinuria levels were positively correlated with the percentage of circulating plasmablasts (Spearman test's ρ = -0.54, P = 0.01 and ρ = 0.65, P = 0.002, respectively). Similarly, the increase of immunoglobulin M (IgM) and the decrease of IgG levels were significantly associated with the percentage of plasmablasts in MCNS-Rel patients (Spearman's ρ = 0.36, P = 0.01 and Spearman's ρ = -0.60, P = 0.01, respectively). Increased production of interleukin (IL)-21, IL-6 and B-cell activating factor (BAFF) in the serum of MCNS-Rel patients was found significantly correlated with the percentage of plasmablasts (ρ = 0.72, P = 0.0002, ρ = 0.49, P = 0.04 and ρ = 0.62, P = 0.009, respectively). CONCLUSIONS An increase in the proportion of circulating plasmablasts seems to be a hallmark of untreated MCNS in adult patients. Further studies are required to more precisely determine the phenotype and functions of these cells.
Collapse
Affiliation(s)
- Julie Oniszczuk
- Assistance Publique des Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Service de Néphrologie et Transplantation, Centre de Référence Maladie Rare « Syndrome Néphrotique Idiopathique », Fédération Hospitalo-Universitaire « Innovative Therapy for Immune Disorders », Créteil, France.,Univ Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale (INSERM) U955, Institut Mondor de Recherche Biomédicale (IMRB), Equipe 21, Créteil, France
| | - Asma Beldi-Ferchiou
- AP-HP, Hôpitaux Universitaires Henri Mondor, Département d'Hématologie et Immunologie Biologiques, Créteil, France.,Univ Paris Est Créteil, INSERM U955, IMRB, Equipe Immunorégulation et Biothérapie, Créteil, France
| | - Etienne Audureau
- AP-HP, Hôpitaux Universitaires Henri Mondor, Service de Santé Publique, Unité de Recherche Clinique, Université Paris Est Créteil, INSERM U955, IMRB, Equipe CEPIA, Créteil, France
| | - Imane Azzaoui
- Univ Paris Est Créteil, INSERM U955, IMRB, Equipe 2, Créteil, France
| | - Valérie Molinier-Frenkel
- AP-HP, Hôpitaux Universitaires Henri Mondor, Département d'Hématologie et Immunologie Biologiques, Créteil, France.,Univ Paris Est Créteil, INSERM U955, IMRB, Equipe Immunorégulation et Biothérapie, Créteil, France
| | - Vincent Frontera
- Univ Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale (INSERM) U955, Institut Mondor de Recherche Biomédicale (IMRB), Equipe 21, Créteil, France
| | - Alexandre Karras
- AP-HP, Hôpital Européen Georges Pompidou, Service de Néphrologie, Paris, France.,Faculté de Médecine Paris-Descartes, Université de Paris, Paris, France
| | - Anissa Moktefi
- Univ Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale (INSERM) U955, Institut Mondor de Recherche Biomédicale (IMRB), Equipe 21, Créteil, France.,AP-HP, Hôpitaux Universitaires Henri Mondor, Département de Pathologie, Créteil, France
| | - Evangeline Pillebout
- Service de Néphrologie et Transplantation, AP-HP, Hôpital Saint Louis, Paris, France
| | - Mohamad Zaidan
- AP-HP, Hôpitaux Universitaires du Kremlin Bicêtre, Service de Néphrologie-Dialyse-Transplantation, Le Kremlin-Bicêtre, France
| | - Khalil El Karoui
- Assistance Publique des Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Service de Néphrologie et Transplantation, Centre de Référence Maladie Rare « Syndrome Néphrotique Idiopathique », Fédération Hospitalo-Universitaire « Innovative Therapy for Immune Disorders », Créteil, France.,Univ Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale (INSERM) U955, Institut Mondor de Recherche Biomédicale (IMRB), Equipe 21, Créteil, France
| | - Marie-Hélène Delfau-Larue
- AP-HP, Hôpitaux Universitaires Henri Mondor, Département d'Hématologie et Immunologie Biologiques, Créteil, France.,Univ Paris Est Créteil, INSERM U955, IMRB, Equipe 9, Créteil, France
| | - Carole Hénique
- Univ Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale (INSERM) U955, Institut Mondor de Recherche Biomédicale (IMRB), Equipe 21, Créteil, France
| | - Mario Ollero
- Univ Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale (INSERM) U955, Institut Mondor de Recherche Biomédicale (IMRB), Equipe 21, Créteil, France
| | - Dil Sahali
- Assistance Publique des Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Service de Néphrologie et Transplantation, Centre de Référence Maladie Rare « Syndrome Néphrotique Idiopathique », Fédération Hospitalo-Universitaire « Innovative Therapy for Immune Disorders », Créteil, France.,Univ Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale (INSERM) U955, Institut Mondor de Recherche Biomédicale (IMRB), Equipe 21, Créteil, France
| | - Matthieu Mahévas
- Univ Paris Est Créteil, INSERM U955, IMRB, Equipe 2, Créteil, France.,AP-HP, Hôpitaux Universitaires Henri Mondor, Service de Médecine interne, Centre de Référence Maladie Rare « Cytopénies Auto-immunes », Créteil, France
| | - Vincent Audard
- Assistance Publique des Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Service de Néphrologie et Transplantation, Centre de Référence Maladie Rare « Syndrome Néphrotique Idiopathique », Fédération Hospitalo-Universitaire « Innovative Therapy for Immune Disorders », Créteil, France.,Univ Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale (INSERM) U955, Institut Mondor de Recherche Biomédicale (IMRB), Equipe 21, Créteil, France
| |
Collapse
|
155
|
Dugan HL, Stamper CT, Li L, Changrob S, Asby NW, Halfmann PJ, Zheng NY, Huang M, Shaw DG, Cobb MS, Erickson SA, Guthmiller JJ, Stovicek O, Wang J, Winkler ES, Madariaga ML, Shanmugarajah K, Jansen MO, Amanat F, Stewart I, Utset HA, Huang J, Nelson CA, Dai YN, Hall PD, Jedrzejczak RP, Joachimiak A, Krammer F, Diamond MS, Fremont DH, Kawaoka Y, Wilson PC. Profiling B cell immunodominance after SARS-CoV-2 infection reveals antibody evolution to non-neutralizing viral targets. Immunity 2021; 54:1290-1303.e7. [PMID: 34022127 PMCID: PMC8101792 DOI: 10.1016/j.immuni.2021.05.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/06/2021] [Accepted: 04/29/2021] [Indexed: 12/16/2022]
Abstract
Dissecting the evolution of memory B cells (MBCs) against SARS-CoV-2 is critical for understanding antibody recall upon secondary exposure. Here, we used single-cell sequencing to profile SARS-CoV-2-reactive B cells in 38 COVID-19 patients. Using oligo-tagged antigen baits, we isolated B cells specific to the SARS-CoV-2 spike, nucleoprotein (NP), open reading frame 8 (ORF8), and endemic human coronavirus (HCoV) spike proteins. SARS-CoV-2 spike-specific cells were enriched in the memory compartment of acutely infected and convalescent patients several months post symptom onset. With severe acute infection, substantial populations of endemic HCoV-reactive antibody-secreting cells were identified and possessed highly mutated variable genes, signifying preexisting immunity. Finally, MBCs exhibited pronounced maturation to NP and ORF8 over time, especially in older patients. Monoclonal antibodies against these targets were non-neutralizing and non-protective in vivo. These findings reveal antibody adaptation to non-neutralizing intracellular antigens during infection, emphasizing the importance of vaccination for inducing neutralizing spike-specific MBCs.
Collapse
Affiliation(s)
- Haley L Dugan
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| | | | - Lei Li
- University of Chicago Department of Medicine, Section of Rheumatology, Chicago, IL 60637, USA
| | - Siriruk Changrob
- University of Chicago Department of Medicine, Section of Rheumatology, Chicago, IL 60637, USA
| | - Nicholas W Asby
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Peter J Halfmann
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53711
| | - Nai-Ying Zheng
- University of Chicago Department of Medicine, Section of Rheumatology, Chicago, IL 60637, USA
| | - Min Huang
- University of Chicago Department of Medicine, Section of Rheumatology, Chicago, IL 60637, USA
| | - Dustin G Shaw
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| | - Mari S Cobb
- Section of Genetic Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Steven A Erickson
- University of Chicago Department of Medicine, Section of Rheumatology, Chicago, IL 60637, USA
| | - Jenna J Guthmiller
- University of Chicago Department of Medicine, Section of Rheumatology, Chicago, IL 60637, USA
| | - Olivia Stovicek
- University of Chicago Department of Medicine, Section of Rheumatology, Chicago, IL 60637, USA
| | - Jiaolong Wang
- University of Chicago Department of Medicine, Section of Rheumatology, Chicago, IL 60637, USA
| | - Emma S Winkler
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63130, USA; Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63130, USA
| | | | | | - Maud O Jansen
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Fatima Amanat
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Isabelle Stewart
- University of Chicago Department of Medicine, Section of Rheumatology, Chicago, IL 60637, USA
| | - Henry A Utset
- University of Chicago Department of Medicine, Section of Rheumatology, Chicago, IL 60637, USA
| | - Jun Huang
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA; Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Christopher A Nelson
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63130, USA
| | - Ya-Nan Dai
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63130, USA
| | - Paige D Hall
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63130, USA
| | - Robert P Jedrzejczak
- Center for Structural Genomics of Infectious Diseases, Consortium for Advanced Science and Engineering, University of Chicago, Chicago, IL 60637, USA; Structural Biology Center, X-ray Science Division, Argonne National Laboratory, Lemont, IL 60439, USA
| | - Andrzej Joachimiak
- Center for Structural Genomics of Infectious Diseases, Consortium for Advanced Science and Engineering, University of Chicago, Chicago, IL 60637, USA; Structural Biology Center, X-ray Science Division, Argonne National Laboratory, Lemont, IL 60439, USA; Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63130, USA; Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63130, USA; Department of Molecular Immunology, Washington University School of Medicine, St Louis, MO 63130, USA
| | - Daved H Fremont
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63130, USA
| | - Yoshihiro Kawaoka
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53711; Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, 108-8639 Tokyo, Japan
| | - Patrick C Wilson
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA; University of Chicago Department of Medicine, Section of Rheumatology, Chicago, IL 60637, USA.
| |
Collapse
|
156
|
Neys SFH, Hendriks RW, Corneth OBJ. Targeting Bruton's Tyrosine Kinase in Inflammatory and Autoimmune Pathologies. Front Cell Dev Biol 2021; 9:668131. [PMID: 34150760 PMCID: PMC8213343 DOI: 10.3389/fcell.2021.668131] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/07/2021] [Indexed: 12/15/2022] Open
Abstract
Bruton's tyrosine kinase (BTK) was discovered due to its importance in B cell development, and it has a critical role in signal transduction downstream of the B cell receptor (BCR). Targeting of BTK with small molecule inhibitors has proven to be efficacious in several B cell malignancies. Interestingly, recent studies reveal increased BTK protein expression in circulating resting B cells of patients with systemic autoimmune disease (AID) compared with healthy controls. Moreover, BTK phosphorylation following BCR stimulation in vitro was enhanced. In addition to its role in BCR signaling, BTK is involved in many other pathways, including pattern recognition, Fc, and chemokine receptor signaling in B cells and myeloid cells. This broad involvement in several immunological pathways provides a rationale for the targeting of BTK in the context of inflammatory and systemic AID. Accordingly, numerous in vitro and in vivo preclinical studies support the potential of BTK targeting in these conditions. Efficacy of BTK inhibitors in various inflammatory and AID has been demonstrated or is currently evaluated in clinical trials. In addition, very recent reports suggest that BTK inhibition may be effective as immunosuppressive therapy to diminish pulmonary hyperinflammation in coronavirus disease 2019 (COVID-19). Here, we review BTK's function in key signaling pathways in B cells and myeloid cells. Further, we discuss recent advances in targeting BTK in inflammatory and autoimmune pathologies.
Collapse
|
157
|
Prieto-Peña D, Genre F, Remuzgo-Martínez S, Pulito-Cueto V, Atienza-Mateo B, Llorca J, Sevilla-Pérez B, Ortego-Centeno N, Lera-Gómez L, Leonardo MT, Peñalba A, Narváez J, Martín-Penagos L, Rodrigo E, Miranda-Filloy JA, Caminal-Montero L, Collado P, Pérez JS, de Argila D, Rubio E, Luque ML, Blanco-Madrigal JM, Galíndez-Agirregoikoa E, Gualillo O, Martín J, Castañeda S, Blanco R, González-Gay MA, López-Mejías R. BAFF, APRIL and BAFFR on the pathogenesis of Immunoglobulin-A vasculitis. Sci Rep 2021; 11:11510. [PMID: 34075170 PMCID: PMC8169776 DOI: 10.1038/s41598-021-91055-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/21/2021] [Indexed: 12/15/2022] Open
Abstract
BAFF, APRIL and BAFF-R are key proteins involved in the development of B-lymphocytes and autoimmunity. Additionally, BAFF, APRIL and BAFFR polymorphisms were associated with immune-mediated conditions, being BAFF GCTGT>A a shared insertion-deletion genetic variant for several autoimmune diseases. Accordingly, we assessed whether BAFF, APRIL and BAFFR represent novel genetic risk factors for Immunoglobulin-A vasculitis (IgAV), a predominantly B-lymphocyte inflammatory condition. BAFF rs374039502, which colocalizes with BAFF GCTGT>A, and two tag variants within APRIL (rs11552708 and rs6608) and BAFFR (rs7290134 and rs77874543) were genotyped in 386 Caucasian IgAV patients and 806 matched healthy controls. No genotypes or alleles differences were observed between IgAV patients and controls when BAFF, APRIL and BAFFR variants were analysed independently. Likewise, no statistically significant differences were found in the genotype and allele frequencies of BAFF, APRIL or BAFFR when IgAV patients were stratified according to the age at disease onset or to the presence/absence of gastrointestinal (GI) or renal manifestations. Similar results were disclosed when APRIL and BAFFR haplotypes were compared between IgAV patients and controls and between IgAV patients stratified according to the clinical characteristics mentioned above. Our results suggest that BAFF, APRIL and BAFFR do not contribute to the genetic network underlying IgAV.
Collapse
Affiliation(s)
- Diana Prieto-Peña
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Division of Rheumatology, Hospital Universitario Marqués de Valdecilla, Avenida Cardenal Herrera Oria s/n, 39011, Santander, Spain
| | - Fernanda Genre
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Division of Rheumatology, Hospital Universitario Marqués de Valdecilla, Avenida Cardenal Herrera Oria s/n, 39011, Santander, Spain
| | - Sara Remuzgo-Martínez
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Division of Rheumatology, Hospital Universitario Marqués de Valdecilla, Avenida Cardenal Herrera Oria s/n, 39011, Santander, Spain
| | - Verónica Pulito-Cueto
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Division of Rheumatology, Hospital Universitario Marqués de Valdecilla, Avenida Cardenal Herrera Oria s/n, 39011, Santander, Spain
| | - Belén Atienza-Mateo
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Division of Rheumatology, Hospital Universitario Marqués de Valdecilla, Avenida Cardenal Herrera Oria s/n, 39011, Santander, Spain.,`López Albo´ Post-Residency Programme, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Javier Llorca
- Epidemiology and Computational Biology Department, School of Medicine, Universidad de Cantabria, and CIBER Epidemiología y Salud Pública (CIBERESP), Santander, Spain
| | | | | | - Leticia Lera-Gómez
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Division of Rheumatology, Hospital Universitario Marqués de Valdecilla, Avenida Cardenal Herrera Oria s/n, 39011, Santander, Spain
| | - María Teresa Leonardo
- Division of Paediatrics, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Ana Peñalba
- Division of Paediatrics, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Javier Narváez
- Division of Rheumatology, Hospital Universitario de Bellvitge, Barcelona, Spain
| | - Luis Martín-Penagos
- Division of Nephrology, Hospital Universitario Marqués de Valdecilla, IDIVAL-REDINREN, Santander, Spain
| | - Emilio Rodrigo
- Division of Nephrology, Hospital Universitario Marqués de Valdecilla, IDIVAL-REDINREN, Santander, Spain
| | | | - Luis Caminal-Montero
- Internal Medicine Dept, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Paz Collado
- Division of Rheumatology, Hospital Universitario Severo Ochoa, Madrid, Spain
| | | | - Diego de Argila
- Division of Dermatology, Hospital Universitario de La Princesa, Madrid, Spain
| | - Esteban Rubio
- Division of Rheumatology, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Manuel León Luque
- Division of Rheumatology, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | | | | | - Oreste Gualillo
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain
| | - Javier Martín
- Instituto de Parasitología y Biomedicina 'López-Neyra', CSIC, PTS Granada, Granada, Spain
| | - Santos Castañeda
- Rheumatology Department, Hospital Universitario de La Princesa, IIS-Princesa, Madrid, Spain
| | - Ricardo Blanco
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Division of Rheumatology, Hospital Universitario Marqués de Valdecilla, Avenida Cardenal Herrera Oria s/n, 39011, Santander, Spain
| | - Miguel A González-Gay
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Division of Rheumatology, Hospital Universitario Marqués de Valdecilla, Avenida Cardenal Herrera Oria s/n, 39011, Santander, Spain.,School of Medicine, Universidad de Cantabria, Santander, Spain.,Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Raquel López-Mejías
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Division of Rheumatology, Hospital Universitario Marqués de Valdecilla, Avenida Cardenal Herrera Oria s/n, 39011, Santander, Spain.
| |
Collapse
|
158
|
Smets I, Prezzemolo T, Imbrechts M, Mallants K, Mitera T, Humblet-Baron S, Dubois B, Matthys P, Liston A, Goris A. Treatment-Induced BAFF Expression and B Cell Biology in Multiple Sclerosis. Front Immunol 2021; 12:676619. [PMID: 34122439 PMCID: PMC8187869 DOI: 10.3389/fimmu.2021.676619] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/07/2021] [Indexed: 01/12/2023] Open
Abstract
Although fingolimod and interferon-β are two mechanistically different multiple sclerosis (MS) treatments, they both induce B cell activating factor (BAFF) and shift the B cell pool towards a regulatory phenotype. However, whether there is a shared mechanism between both treatments in how they influence the B cell compartment remains elusive. In this study, we collected a cross-sectional study population of 112 MS patients (41 untreated, 42 interferon-β, 29 fingolimod) and determined B cell subsets, cell-surface and RNA expression of BAFF-receptor (BAFF-R) and transmembrane activator and cyclophilin ligand interactor (TACI) as well as plasma and/or RNA levels of BAFF, BAFF splice forms and interleukin-10 (IL-10) and -35 (IL-35). We added an in vitro B cell culture with four stimulus conditions (Medium, CpG, BAFF and CpG+BAFF) for untreated and interferon-β treated patients including measurement of intracellular IL-10 levels. Our flow experiments showed that interferon-β and fingolimod induced BAFF protein and mRNA expression (P ≤ 3.15 x 10-4) without disproportional change in the antagonizing splice form. Protein BAFF correlated with an increase in transitional B cells (P = 5.70 x 10-6), decrease in switched B cells (P = 3.29 x 10-4), and reduction in B cell-surface BAFF-R expression (P = 2.70 x 10-10), both on TACI-positive and -negative cells. TACI and BAFF-R RNA levels remained unaltered. RNA, plasma and in vitro experiments demonstrated that BAFF was not associated with increased IL-10 and IL-35 levels. In conclusion, treatment-induced BAFF correlates with a shift towards transitional B cells which are enriched for cells with an immunoregulatory function. However, BAFF does not directly influence the expression of the immunoregulatory cytokines IL-10 and IL-35. Furthermore, the post-translational mechanism of BAFF-induced BAFF-R cell surface loss was TACI-independent. These observations put the failure of pharmaceutical anti-BAFF strategies in perspective and provide insights for targeted B cell therapies.
Collapse
Affiliation(s)
- Ide Smets
- Department of Neurosciences, Laboratory for Neuroimmunology, KU Leuven, Leuven, Belgium.,Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Teresa Prezzemolo
- Department of Microbiology, Immunology and Transplantation, Laboratory for Adaptive Immunology, KU Leuven, Belgium.,VIB Center for Brain & Disease Research, Leuven, Belgium
| | - Maya Imbrechts
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Immunobiology, KU Leuven, Leuven, Belgium
| | - Klara Mallants
- Department of Neurosciences, Laboratory for Neuroimmunology, KU Leuven, Leuven, Belgium.,Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Tania Mitera
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Immunobiology, KU Leuven, Leuven, Belgium
| | - Stéphanie Humblet-Baron
- Department of Microbiology, Immunology and Transplantation, Laboratory for Adaptive Immunology, KU Leuven, Belgium
| | - Bénédicte Dubois
- Department of Neurosciences, Laboratory for Neuroimmunology, KU Leuven, Leuven, Belgium.,Leuven Brain Institute, KU Leuven, Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Patrick Matthys
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Immunobiology, KU Leuven, Leuven, Belgium
| | - Adrian Liston
- Department of Microbiology, Immunology and Transplantation, Laboratory for Adaptive Immunology, KU Leuven, Belgium.,VIB Center for Brain & Disease Research, Leuven, Belgium.,Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, Cambridge, United Kingdom
| | - An Goris
- Department of Neurosciences, Laboratory for Neuroimmunology, KU Leuven, Leuven, Belgium.,Leuven Brain Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
159
|
Takeshita M, Suzuki K, Nakazawa M, Kamata H, Ishii M, Oyamada Y, Oshima H, Takeuchi T. Antigen-driven autoantibody production in lungs of interstitial lung disease with autoimmune disease. J Autoimmun 2021; 121:102661. [PMID: 34034155 DOI: 10.1016/j.jaut.2021.102661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 11/26/2022]
Abstract
Interstitial lung disease (ILD) sometimes becomes a life-threatening complication of systemic autoimmune diseases; however, little is known about the immune response in lung lesions. We aimed to investigate humoural immunity in ILD associated with rheumatoid arthritis (RA), sjögren's syndrome (SjS), and mixed connective tissue disease (MCTD), using bronchoalveolar fluid (BALF) and serum samples from 15 patients with autoimmune disease associated-ILD. We first showed that BALF contained higher titers of disease-related autoantibodies than serum, suggesting the possibility of autoantibody production in lungs. Next, we produced 326 monoclonal antibodies from antibody-secreting cells in BALF, and the reactivity and their revertants, in which somatic hypermutations were reverted to germline, were analyzed. Among 123 antibodies from RA-ILD, 16 disease-related antibodies (anti-modified protein antibodies and rheumatoid factors) were identified, of which one antibody had both properties. The revertant antibodies changed their target modification in a complicated manner, suggesting that the antibodies were selected against various modifications in lungs. Among 146 antibodies from SjS-ILD and/or MCTD-ILD, seven anti-SSA/Ro60 antibodies and 15 anti-RNP antibodies were identified. Some of the anti-RNP antibodies recognized multiple RNP constituent proteins simultaneously, indicating that epitope spreading may progress in lungs. Our results revealed the existence of an active autoimmunity in the lungs of autoimmune disease associated-ILD.
Collapse
Affiliation(s)
- Masaru Takeshita
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.
| | - Katsuya Suzuki
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Maho Nakazawa
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hirofumi Kamata
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Makoto Ishii
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yoshitaka Oyamada
- Department of Respiratory Medicine, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Hisaji Oshima
- Department of Connective Tissue Diseases, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Tsutomu Takeuchi
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
160
|
The role of BAFF and G-CSF for rituximab-induced late-onset neutropenia (LON) in lymphomas. Med Oncol 2021; 38:70. [PMID: 34003398 PMCID: PMC8131291 DOI: 10.1007/s12032-021-01516-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 04/24/2021] [Indexed: 11/22/2022]
Abstract
Mechanisms for late-onset neutropenia (LON) after rituximab treatment are poorly defined both for non-Hodgkin lymphoma (NHL) and for autoimmune disorders. We performed a case–control analysis of a prospective cohort of 169 evaluable consecutive rituximab-treated NHL patients to assess cytokines involved in neutro- and lymphopoiesis (G-CSF, SDF1, BAFF, APRIL) and inflammation (CRP) as possible LON mechanisms. Fifteen patients (9%) developed LON (peripheral blood /PB/ absolute neutrophil counts /ANC/ < 0.5 G/L, all with marked depletion of CD20+ B-lymphocytes in bone marrows); they were compared with 20 matched NHL controls without LON. At start of LON, significantly higher PB G-CSF and BAFF levels (P = 0.0004 and 0.006, respectively), as well as CRP rises were noted compared to controls; these G-CSF and BAFF and most CRP values returned to levels of the controls in post-LON samples. G-CSF (but not BAFF) changes correlated to CRP rises (but not to ANC levels). BAFF levels correlated significantly to absolute monocyte counts and PB large granular lymphocyte counts (but not to ANC, C-CSF or CRP values). No changes of SDF1 or APRIL levels were noted. Neither LON cases nor controls displayed anti-neutrophil autoantibodies. Collectively, LON in NHL patients was timewise related to transient bursts of blood G-CSF and BAFF concentrations, suggesting that these neutro- and lymphopoiesis growth factors play a role in emergence of rituximab-induced LON, and that inflammation may be a trigger for G-CSF production during LON.
Collapse
|
161
|
Mosquera Orgueira A, Ferreiro Ferro R, Díaz Arias JÁ, Aliste Santos C, Antelo Rodríguez B, Bao Pérez L, Alonso Vence N, Bendaña López Á, Abuin Blanco A, Melero Valentín P, Peleteiro Raindo A, Cid López M, Pérez Encinas MM, González Pérez MS, Fraga Rodríguez MF, Bello López JL. Detection of new drivers of frequent B-cell lymphoid neoplasms using an integrated analysis of whole genomes. PLoS One 2021; 16:e0248886. [PMID: 33945543 PMCID: PMC8096002 DOI: 10.1371/journal.pone.0248886] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 01/19/2021] [Indexed: 12/21/2022] Open
Abstract
B-cell lymphoproliferative disorders exhibit a diverse spectrum of diagnostic entities with heterogeneous behaviour. Multiple efforts have focused on the determination of the genomic drivers of B-cell lymphoma subtypes. In the meantime, the aggregation of diverse tumors in pan-cancer genomic studies has become a useful tool to detect new driver genes, while enabling the comparison of mutational patterns across tumors. Here we present an integrated analysis of 354 B-cell lymphoid disorders. 112 recurrently mutated genes were discovered, of which KMT2D, CREBBP, IGLL5 and BCL2 were the most frequent, and 31 genes were putative new drivers. Mutations in CREBBP, TNFRSF14 and KMT2D predominated in follicular lymphoma, whereas those in BTG2, HTA-A and PIM1 were more frequent in diffuse large B-cell lymphoma. Additionally, we discovered 31 significantly mutated protein networks, reinforcing the role of genes such as CREBBP, EEF1A1, STAT6, GNA13 and TP53, but also pointing towards a myriad of infrequent players in lymphomagenesis. Finally, we report aberrant expression of oncogenes and tumor suppressors associated with novel noncoding mutations (DTX1 and S1PR2), and new recurrent copy number aberrations affecting immune check-point regulators (CD83, PVR) and B-cell specific genes (TNFRSF13C). Our analysis expands the number of mutational drivers of B-cell lymphoid neoplasms, and identifies several differential somatic events between disease subtypes.
Collapse
Affiliation(s)
- Adrián Mosquera Orgueira
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Galicia, Spain
- Department of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Galicia, Spain
- University of Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Roi Ferreiro Ferro
- Department of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Galicia, Spain
| | - José Ángel Díaz Arias
- Department of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Galicia, Spain
| | - Carlos Aliste Santos
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Galicia, Spain
- Department of Pathology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Galicia, Spain
| | - Beatriz Antelo Rodríguez
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Galicia, Spain
- Department of Pathology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Galicia, Spain
| | - Laura Bao Pérez
- Department of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Galicia, Spain
| | - Natalia Alonso Vence
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Galicia, Spain
- Department of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Galicia, Spain
| | - Ággeles Bendaña López
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Galicia, Spain
- Department of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Galicia, Spain
- University of Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Aitor Abuin Blanco
- Department of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Galicia, Spain
| | - Paula Melero Valentín
- Department of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Galicia, Spain
| | - And´res Peleteiro Raindo
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Galicia, Spain
- Department of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Galicia, Spain
- University of Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Miguel Cid López
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Galicia, Spain
- Department of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Galicia, Spain
- University of Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Manuel Mateo Pérez Encinas
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Galicia, Spain
- Department of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Galicia, Spain
- University of Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Marta Sonia González Pérez
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Galicia, Spain
- Department of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Galicia, Spain
| | - Máximo Francisco Fraga Rodríguez
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Galicia, Spain
- University of Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Department of Pathology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Galicia, Spain
| | - José Luis Bello López
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Galicia, Spain
- Department of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Galicia, Spain
- University of Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| |
Collapse
|
162
|
Meghnem D, Oldford SA, Haidl ID, Barrett L, Marshall JS. Histamine receptor 2 blockade selectively impacts B and T cells in healthy subjects. Sci Rep 2021; 11:9405. [PMID: 33931709 PMCID: PMC8087813 DOI: 10.1038/s41598-021-88829-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 04/12/2021] [Indexed: 11/18/2022] Open
Abstract
Histamine receptor 2 (H2R) blockade is commonly used in patients with gastric, duodenal ulcers or gastroesophageal reflux disease. Beyond the gastrointestinal tract, H2R is expressed by multiple immune cells, yet little is known about the immunomodulatory effects of such treatment. Clinical reports have associated H2R blockade with leukopenia, neutropenia, and myelosuppression, and has been shown to provide clinical benefit in certain cancer settings. To systematically assess effects of H2R blockade on key immune parameters, a single-center, single-arm clinical study was conducted in 29 healthy subjects. Subjects received daily high dose ranitidine for 6 weeks. Peripheral blood immunophenotyping and mediator analysis were performed at baseline, 3 and 6 weeks into treatment, and 12 weeks after treatment cessation. Ranitidine was well-tolerated, and no drug related adverse events were observed. Ranitidine had no effect on number of neutrophils, basophils or eosinophils. However, ranitidine decreased numbers of B cells and IL-2Rα (CD25) expressing T cells that remained lower even after treatment cessation. Reduced serum levels of IL-2 were also observed and remained low after treatment. These observations highlight a previously unrecognised immunomodulatory sustained impact of H2R blockade. Therefore, the immune impacts of H2R blockade may require greater consideration in the context of vaccination and immunotherapy.
Collapse
Affiliation(s)
- Dihia Meghnem
- Dalhousie Human Immunology and Inflammation Group, Department of Microbiology and Immunology, Dalhousie University, Sir Charles Tupper Medical Building, Room 7-C2, 5850 College Street, PO Box 15000, Halifax, NS, B3H 4R2, Canada
| | - Sharon A Oldford
- Dalhousie Human Immunology and Inflammation Group, Department of Microbiology and Immunology, Dalhousie University, Sir Charles Tupper Medical Building, Room 7-C2, 5850 College Street, PO Box 15000, Halifax, NS, B3H 4R2, Canada.,Senescence, Aging, Infection and Immunity Laboratory, Department of Medicine, Dalhousie University, Halifax, NS, Canada.,Division of Infectious Diseases, Nova Scotia Health Authority, Halifax, NS, Canada
| | - Ian D Haidl
- Dalhousie Human Immunology and Inflammation Group, Department of Microbiology and Immunology, Dalhousie University, Sir Charles Tupper Medical Building, Room 7-C2, 5850 College Street, PO Box 15000, Halifax, NS, B3H 4R2, Canada
| | - Lisa Barrett
- Dalhousie Human Immunology and Inflammation Group, Department of Microbiology and Immunology, Dalhousie University, Sir Charles Tupper Medical Building, Room 7-C2, 5850 College Street, PO Box 15000, Halifax, NS, B3H 4R2, Canada.,Senescence, Aging, Infection and Immunity Laboratory, Department of Medicine, Dalhousie University, Halifax, NS, Canada.,Division of Infectious Diseases, Nova Scotia Health Authority, Halifax, NS, Canada
| | - Jean S Marshall
- Dalhousie Human Immunology and Inflammation Group, Department of Microbiology and Immunology, Dalhousie University, Sir Charles Tupper Medical Building, Room 7-C2, 5850 College Street, PO Box 15000, Halifax, NS, B3H 4R2, Canada. .,Division of Infectious Diseases, Nova Scotia Health Authority, Halifax, NS, Canada.
| |
Collapse
|
163
|
Li G, Zhang Q, Liu Z, Shen H, Zhu Y, Zhou Z, Ding W, Han S, Zhou J, Ou R, Luo M, Liu S. TriBAFF-CAR-T cells eliminate B-cell malignancies with BAFFR-expression and CD19 antigen loss. Cancer Cell Int 2021; 21:223. [PMID: 33865370 PMCID: PMC8052726 DOI: 10.1186/s12935-021-01923-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/07/2021] [Indexed: 02/08/2023] Open
Abstract
Background To investigate the effect of TriBAFF-CAR-T cells on hematological tumor cells. Methods TriBAFF-CAR-T and CD19-CAR-T cells were co-cultured with BAFFR-bearing B-cell malignancies at different effector/target ratios to evaluate the anti-tumor effects. In vivo, TriBAFF-CAR-T and CD19-CAR-T cells were intravenously injected into Raji-luciferase xenograft mice. CD19 antigens losing lymphoblasts was simulated by Raji knocking out CD19 (CD19KO) to investigate the effect of TriBAFF-CAR-T cells on CD19KO Raji. Results Both TriBAFF-CAR-T and CD19-CAR-T cells significantly induced the lysis of Raji, BALL-1, and Jeko-1. Moreover, when CD19-CAR-T cells specifically caused the lysis of K562 with overexpressed CD19, the lethal effect of TriBAFF-CAR-T cells was also specific for BAFFR-bearing K562 with increasing levels of interleukin-2 and INF-γ. The TriBAFF-CAR-T have the same effect with CD19-CAR-T cells in treating Raji xenofraft mice. TriBAFF-CAR-T cells also have great effect in CD19KO Raji cells. Conclusions In this study, we successfully constructed novel TriBAFF-CAR-T cells to eliminate BAFFR-bearing and CD19 antigen loss in hematological tumor cells.
Collapse
Affiliation(s)
- Guangchao Li
- Department of Hematology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong Province, 510317, China.,Guangzhou Bio-gene Technology Co., Ltd, Guangzhou, Guangdong Province, 510530, China
| | - Qing Zhang
- Department of Hematology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong Province, 510317, China
| | - Zhi Liu
- Department of Hematology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong Province, 510317, China
| | - Huijuan Shen
- Department of Hematology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong Province, 510317, China
| | - Yangmin Zhu
- Department of Hematology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong Province, 510317, China
| | - Zhao Zhou
- Guangzhou Bio-gene Technology Co., Ltd, Guangzhou, Guangdong Province, 510530, China
| | - Wen Ding
- Guangzhou Bio-gene Technology Co., Ltd, Guangzhou, Guangdong Province, 510530, China
| | - Siqi Han
- Department of Medical Oncology, Jinling Hospital, Nanjing Clinical School of Southern Medical University, Nanjing, Jiangsu Province, 210002, China
| | - Jie Zhou
- Department of Hematology, People's Hospital of Deyang City, Deyang, Sichuan Province, 618000, China
| | - Ruiming Ou
- Department of Hematology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong Province, 510317, China. .,Department of Hematology, Guangdong Second Provincial General Hospital, Xin Gang Zhong Road 466#, Haizhu Distict, Guangzhou, Guangdong Province, 510317, China.
| | - Min Luo
- Department of Hematology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong Province, 510317, China. .,Guangzhou Bio-gene Technology Co., Ltd, Guangzhou, Guangdong Province, 510530, China. .,Department of Hematology, Guangdong Second Provincial General Hospital, Xin Gang Zhong Road 466#, Haizhu Distict, Guangzhou, Guangdong Province, 510317, China.
| | - Shuang Liu
- Department of Hematology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong Province, 510317, China. .,Department of Hematology, Guangdong Second Provincial General Hospital, Xin Gang Zhong Road 466#, Haizhu Distict, Guangzhou, Guangdong Province, 510317, China.
| |
Collapse
|
164
|
The impact of rare and low-frequency genetic variants in common variable immunodeficiency (CVID). Sci Rep 2021; 11:8308. [PMID: 33859323 PMCID: PMC8050305 DOI: 10.1038/s41598-021-87898-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 04/01/2021] [Indexed: 02/07/2023] Open
Abstract
Next Generation Sequencing (NGS) has uncovered hundreds of common and rare genetic variants involved in complex and rare diseases including immune deficiencies in both an autosomal recessive and autosomal dominant pattern. These rare variants however, cannot be classified clinically, and common variants only marginally contribute to disease susceptibility. In this study, we evaluated the multi-gene panel results of Common Variable Immunodeficiency (CVID) patients and argue that rare variants located in different genes play a more prominent role in disease susceptibility and/or etiology. We performed NGS on DNA extracted from the peripheral blood leukocytes from 103 patients using a panel of 19 CVID-related genes: CARD11, CD19, CD81, ICOS, CTLA4, CXCR4, GATA2, CR2, IRF2BP2, MOGS, MS4A1, NFKB1, NFKB2, PLCG2, TNFRSF13B, TNFRSF13C, TNFSF12, TRNT1 and TTC37. Detected variants were evaluated and classified based on their impact, pathogenicity classification and population frequency as well as the frequency within our study group. NGS revealed 112 different (a total of 227) variants with under 10% population frequency in 103 patients of which 22(19.6%) were classified as benign, 29(25.9%) were classified as likely benign, 4(3.6%) were classified as likely pathogenic and 2(1.8%) were classified as pathogenic. Moreover, 55(49.1%) of the variants were classified as variants of uncertain significance. We also observed different variant frequencies when compared to population frequency databases. Case-control data is not sufficient to unravel the genetic etiology of immune deficiencies. Thus, it is important to understand the incidence of co-occurrence of two or more rare variants to aid in illuminating their potential roles in the pathogenesis of immune deficiencies.
Collapse
|
165
|
Lin YH, Liang Y, Wang H, Tung LT, Förster M, Subramani PG, Di Noia JM, Clare S, Langlais D, Nijnik A. Regulation of B Lymphocyte Development by Histone H2A Deubiquitinase BAP1. Front Immunol 2021; 12:626418. [PMID: 33912157 PMCID: PMC8072452 DOI: 10.3389/fimmu.2021.626418] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/12/2021] [Indexed: 01/08/2023] Open
Abstract
BAP1 is a deubiquitinase (DUB) of the Ubiquitin C-terminal Hydrolase (UCH) family that regulates gene expression and other cellular processes, via deubiquitination of histone H2AK119ub and other substrates. BAP1 is an important tumor suppressor in human, expressed and functional across many cell-types and tissues, including those of the immune system. B lymphocytes are the mediators of humoral immune response, however the role of BAP1 in B cell development and physiology remains poorly understood. Here we characterize a mouse line with a selective deletion of BAP1 within the B cell lineage (Bap1fl/fl mb1-Cre) and establish a cell intrinsic role of BAP1 in the regulation of B cell development. We demonstrate a depletion of large pre-B cells, transitional B cells, and mature B cells in Bap1fl/fl mb1-Cre mice. We characterize broad transcriptional changes in BAP1-deficient pre-B cells, map BAP1 binding across the genome, and analyze the effects of BAP1-loss on histone H2AK119ub levels and distribution. Overall, our work establishes a cell intrinsic role of BAP1 in B lymphocyte development, and suggests its contribution to the regulation of the transcriptional programs of cell cycle progression, via the deubiquitination of histone H2AK119ub.
Collapse
Affiliation(s)
- Yun Hsiao Lin
- Department of Physiology, McGill University, Montreal, QC, Canada
- McGill Research Centre on Complex Traits, McGill University, Montreal, QC, Canada
| | - Yue Liang
- Department of Physiology, McGill University, Montreal, QC, Canada
- McGill Research Centre on Complex Traits, McGill University, Montreal, QC, Canada
| | - HanChen Wang
- Department of Physiology, McGill University, Montreal, QC, Canada
- McGill Research Centre on Complex Traits, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- McGill University Genome Centre, Montreal, QC, Canada
| | - Lin Tze Tung
- Department of Physiology, McGill University, Montreal, QC, Canada
- McGill Research Centre on Complex Traits, McGill University, Montreal, QC, Canada
| | - Michael Förster
- Department of Physiology, McGill University, Montreal, QC, Canada
- McGill Research Centre on Complex Traits, McGill University, Montreal, QC, Canada
| | - Poorani Ganesh Subramani
- Institut de Recherches Cliniques de Montréal, Montreal, QC, Canada
- Department of Medicine, McGill University, Montreal, QC, Canada
| | - Javier M. Di Noia
- Institut de Recherches Cliniques de Montréal, Montreal, QC, Canada
- Department of Medicine, McGill University, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Biochemistry & Molecular Medicine, Université de Montréal, Montreal, QC, Canada
| | - Simon Clare
- Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - David Langlais
- McGill Research Centre on Complex Traits, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- McGill University Genome Centre, Montreal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Anastasia Nijnik
- Department of Physiology, McGill University, Montreal, QC, Canada
- McGill Research Centre on Complex Traits, McGill University, Montreal, QC, Canada
| |
Collapse
|
166
|
Yao X, Ren Y, Zhao Q, Chen X, Jiang J, Liu D, Hu P. Pharmacokinetics analysis based on target-mediated drug distribution for RC18, a novel BLyS/APRIL fusion protein to treat systemic lupus erythematosus and rheumatoid arthritis. Eur J Pharm Sci 2021; 159:105704. [PMID: 33440243 DOI: 10.1016/j.ejps.2021.105704] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 01/03/2021] [Accepted: 01/05/2021] [Indexed: 01/27/2023]
Abstract
BACKGROUND AND PURPOSE RC18 is a novel recombinant fusion protein targeting on B lymphocyte stimulator (BLyS). We aimed to develop and qualify a population pharmacokinetics (PopPK) model for RC18 in systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) patients, taking into account the mechanistic target-mediated drug disposition (TMDD) process. METHODS A TMDD model of RC18 was developed using data from two phase I clinical trial (n = 23). The TMDD structural model was developed by simultaneous fitting of the serum free RC18 and serum RC18-BLyS complex. Potential covariates were screened using stepwise method, and predictive performance was qualified using a prediction-corrected visual predictive check (pcVPC) and bootstrap. RESULTS A two compartment TMDD model with first order absorption for subcutaneous administration was built. The final model included a significant relationship between distribution volume of the central compartment and body weight. And the baseline of immunoglobulin IgG had significant effect on the baseline of target BLyS. The plots from goodness-of-fit and pcVPC confirmed good predictive performance of this TMDDmodel. CONCLUSIONS This mechanistic TMDD model integrated the interaction of RC18 with its target BLyS and accurately predicts both RC18 and RC18-BLyS complex profiles in RA and SLE patients. Simulated target change profiles can be used to help guide rational dose regimen selection and used as a biomarker for efficacy evaluation.
Collapse
Affiliation(s)
- Xueting Yao
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100032, China; Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Pking Union Medical College Hospital, Beijing 100032, China; Drug Clinical Trial Center, Peking University Third Hospital, Beijing 100191, China
| | - Yupeng Ren
- Johnson & Johnson Pharmaceuticals (Shanghai) Ltd., Shanghai 200240, China
| | - Qian Zhao
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100032, China; Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Pking Union Medical College Hospital, Beijing 100032, China
| | - Xia Chen
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Ji Jiang
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100032, China; Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Pking Union Medical College Hospital, Beijing 100032, China
| | - Dongyang Liu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100032, China; Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Pking Union Medical College Hospital, Beijing 100032, China; Drug Clinical Trial Center, Peking University Third Hospital, Beijing 100191, China
| | - Pei Hu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100032, China; Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Pking Union Medical College Hospital, Beijing 100032, China.
| |
Collapse
|
167
|
Lázaro SF, Tonhati H, Oliveira HR, Silva AA, Nascimento AV, Santos DJA, Stefani G, Brito LF. Genomic studies of milk-related traits in water buffalo (Bubalus bubalis) based on single-step genomic best linear unbiased prediction and random regression models. J Dairy Sci 2021; 104:5768-5793. [PMID: 33685677 DOI: 10.3168/jds.2020-19534] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 01/02/2021] [Indexed: 01/14/2023]
Abstract
Genomic selection has been widely implemented in many livestock breeding programs, but it remains incipient in buffalo. Therefore, this study aimed to (1) estimate variance components incorporating genomic information in Murrah buffalo; (2) evaluate the performance of genomic prediction for milk-related traits using single- and multitrait random regression models (RRM) and the single-step genomic best linear unbiased prediction approach; and (3) estimate longitudinal SNP effects and candidate genes potentially associated with time-dependent variation in milk, fat, and protein yields, as well as somatic cell score (SCS) in multiple parities. The data used to estimate the genetic parameters consisted of a total of 323,140 test-day records. The average daily heritability estimates were moderate (0.35 ± 0.02 for milk yield, 0.22 ± 0.03 for fat yield, 0.42 ± 0.03 for protein yield, and 0.16 ± 0.03 for SCS). The highest heritability estimates, considering all traits studied, were observed between 20 and 280 d in milk (DIM). The genetic correlation estimates at different DIM among the evaluated traits ranged from -0.10 (156 to 185 DIM for SCS) to 0.61 (36 to 65 DIM for fat yield). In general, direct selection for any of the traits evaluated is expected to result in indirect genetic gains for milk yield, fat yield, and protein yield but also increase SCS at certain lactation stages, which is undesirable. The predicted RRM coefficients were used to derive the genomic estimated breeding values (GEBV) for each time point (from 5 to 305 DIM). In general, the tuning parameters evaluated when constructing the hybrid genomic relationship matrices had a small effect on the GEBV accuracy and a greater effect on the bias estimates. The SNP solutions were back-solved from the GEBV predicted from the Legendre random regression coefficients, which were then used to estimate the longitudinal SNP effects (from 5 to 305 DIM). The daily SNP effect for 3 different lactation stages were performed considering 3 different lactation stages for each trait and parity: from 5 to 70, from 71 to 150, and from 151 to 305 DIM. Important genomic regions related to the analyzed traits and parities that explain more than 0.50% of the total additive genetic variance were selected for further analyses of candidate genes. In general, similar potential candidate genes were found between traits, but our results suggest evidence of differential sets of candidate genes underlying the phenotypic expression of the traits across parities. These results contribute to a better understanding of the genetic architecture of milk production traits in dairy buffalo and reinforce the relevance of incorporating genomic information to genetically evaluate longitudinal traits in dairy buffalo. Furthermore, the candidate genes identified can be used as target genes in future functional genomics studies.
Collapse
Affiliation(s)
- Sirlene F Lázaro
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907; Department of Animal Science, College of Agricultural and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal, 14884-900, SP, Brazil
| | - Humberto Tonhati
- Department of Animal Science, College of Agricultural and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal, 14884-900, SP, Brazil
| | - Hinayah R Oliveira
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907; Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, N1G 2W1, ON, Canada
| | - Alessandra A Silva
- Department of Animal Science, College of Agricultural and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal, 14884-900, SP, Brazil
| | - André V Nascimento
- Department of Animal Science, College of Agricultural and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal, 14884-900, SP, Brazil
| | - Daniel J A Santos
- Department of Animal and Avian Science, University of Maryland, College Park 20742
| | - Gabriela Stefani
- Department of Animal Science, College of Agricultural and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal, 14884-900, SP, Brazil
| | - Luiz F Brito
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907.
| |
Collapse
|
168
|
Zhang K, Roy NK, Vicioso Y, Woo J, Beck R, de Lima M, Caimi P, Feinberg D, Parameswaran R. BAFF receptor antibody for mantle cell lymphoma therapy. Oncoimmunology 2021; 10:1893501. [PMID: 33747637 PMCID: PMC7939563 DOI: 10.1080/2162402x.2021.1893501] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Mantle cell lymphoma (MCL) is an aggressive form of B cell non-Hodgkin’s lymphoma and remains incurable under current treatment modalities. One of the main reasons for treatment failure is the development of drug resistance. Accumulating evidence suggests that B cell activating factor (BAFF) and BAFF receptor (BAFF-R) play an important role in the proliferation and survival of malignant B cells. High serum BAFF levels are often correlated with poor drug response and relapse in MCL patients. Our study shows that BAFF-R is expressed on both MCL patient cells and cell lines. BAFF-R knockdown leads to MCL cell death showing the importance of BAFF-R signaling in MCL survival. Moderate knockdown of BAFF-R in MCL cells did not affect its viability, but sensitized them to cytarabine treatment in vitro and in vivo, with prolonged mice survival. Anti-BAFF-R antibody treatment promoted drug-induced MCL cell death. Conversely, the addition of recombinant BAFF (rhBAFF) to MCL cells protected them from cytarabine-induced apoptosis. We tested the efficacy of a humanized defucosylated ADCC optimized anti-BAFF-R antibody in killing MCL. Our data show both in vitro and in vivo efficacy of this antibody for MCL therapy. To conclude, our data indicate that BAFF/BAFF-R signaling is crucial for survival and involved in drug resistance of MCL. Targeting BAFF-R using BAFF-R antibody might be a promising therapeutical strategy to treat MCL patients resistant to chemotherapy.
Collapse
Affiliation(s)
- Keman Zhang
- Division of Hematology/Oncology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Nand K Roy
- Division of Hematology/Oncology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Yorleny Vicioso
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Janghee Woo
- Novartis Institute for BioMedical Research, Translational Clinical Oncology, NJ, USA
| | - Rose Beck
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Marcos de Lima
- Hematology and Oncology, University Hospitals, Cleveland, Ohio, USA.,The Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Paolo Caimi
- Hematology and Oncology, University Hospitals, Cleveland, Ohio, USA.,The Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Daniel Feinberg
- Division of Hematology/Oncology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Reshmi Parameswaran
- Division of Hematology/Oncology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,The Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
169
|
Lin Z, Süsskind D. Exploring the role of BAFF as biomarker in the detection of uveal melanoma metastases. J Cancer Res Clin Oncol 2021; 147:1389-1405. [PMID: 33665679 DOI: 10.1007/s00432-021-03555-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/04/2021] [Indexed: 12/29/2022]
Abstract
PURPOSE While B-cell activating factor (BAFF) was identified to promote the invasion in other malignancies, its role in the progression of uveal melanoma (UM) still remains unexplored. Here, we analysed the serum level of BAFF in UM patients with regard to its significance as biomarker for the metastases. METHODS In this retrospective study, serum BAFF levels in 173 UM patients (36 with metastases and 137 without), and 23 healthy controls were measured with a multiplexed sandwich ELISA system and then correlated with clinicopathological characteristics such as primary tumor size, tumor location, histological cell type, sex, cancer stage, cytogenetic alterations of chromosome 3, and the metastatic burden. Immunohistochemical staining of 50 UM tissue specimens was also performed to evaluate the expression of BAFF and its receptors BAFF-R and TACI. RESULTS The metastatic patients were identified to have significantly higher serum BAFF levels (mean ± SD, 1520.8 ± 1182.1 pg/ml) than those without metastases (950.4 ± 494.6 pg/ml) and controls (810.3 ± 140.5 pg/ml). While no distinctions were detected with regard to tumor location, histological cell type, gender, and monosomy 3, the patients in cancer stages II, III, and IV displayed higher serum BAFF levels than those in stage I. The serum BAFF level was significantly correlated with the metastatic burden. The serum BAFF level of 1120 pg/ml was identified to have the best performance for distinguishing the metastatic patients from non-metastatic patients. In the kinetic study, we noticed that 20.8% of the analysed patients already demonstrated elevated serum BAFF concentrations before the clinical diagnosis of metastases. Positive BAFF staining was detected in the cytoplasm of single tumor cells (in 13 specimens), macrophages (in 12 specimens), and tumor-infiltrating lymphocytes (TILs) (in 13 specimens). The expressions of BAFF-R and TACI were also observed in 17 and 36 of the 50 tested UM specimens, respectively. CONCLUSIONS Our study first suggests that BAFF might be a promising serum marker for the detection of UM metastases.
Collapse
Affiliation(s)
- Zenan Lin
- Center for Ophthalmology, University Eye Hospital, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany.
| | - Daniela Süsskind
- Center for Ophthalmology, University Eye Hospital, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| |
Collapse
|
170
|
Armitage LH, Wallet MA, Mathews CE. Influence of PTPN22 Allotypes on Innate and Adaptive Immune Function in Health and Disease. Front Immunol 2021; 12:636618. [PMID: 33717184 PMCID: PMC7946861 DOI: 10.3389/fimmu.2021.636618] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/18/2021] [Indexed: 01/18/2023] Open
Abstract
Protein tyrosine phosphatase, non-receptor type 22 (PTPN22) regulates a panoply of leukocyte signaling pathways. A single nucleotide polymorphism (SNP) in PTPN22, rs2476601, is associated with increased risk of Type 1 Diabetes (T1D) and other autoimmune diseases. Over the past decade PTPN22 has been studied intensely in T cell receptor (TCR) and B cell receptor (BCR) signaling. However, the effect of the minor allele on PTPN22 function in TCR signaling is controversial with some reports concluding it has enhanced function and blunts TCR signaling and others reporting it has reduced function and increases TCR signaling. More recently, the core function of PTPN22 as well as functional derangements imparted by the autoimmunity-associated variant allele of PTPN22 have been examined in monocytes, macrophages, dendritic cells, and neutrophils. In this review we will discuss the known functions of PTPN22 in human cells, and we will elaborate on how autoimmunity-associated variants influence these functions across the panoply of immune cells that express PTPN22. Further, we consider currently unresolved questions that require clarification on the role of PTPN22 in immune cell function.
Collapse
Affiliation(s)
- Lucas H. Armitage
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| | - Mark A. Wallet
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, United States
- Immuno-Oncology at Century Therapeutics, LLC, Philadelphia, PA, United States
| | - Clayton E. Mathews
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
171
|
Johansson D, Rauld C, Roux J, Regairaz C, Galli E, Callegari I, Raad L, Waldt A, Cuttat R, Roma G, Diebold M, Becher B, Kuhle J, Derfuss T, Carballido JM, Sanderson NSR. Mass Cytometry of CSF Identifies an MS-Associated B-cell Population. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/2/e943. [PMID: 33589541 PMCID: PMC8057060 DOI: 10.1212/nxi.0000000000000943] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 10/28/2020] [Indexed: 11/15/2022]
Abstract
Objective To identify an MS-specific immune cell population by deep immune phenotyping and relate it to soluble signaling molecules in CSF. Methods We analyzed surface expression of 22 markers in paired blood/CSF samples from 39 patients using mass cytometry (cytometry by time of flight). We also measured the concentrations of 296 signaling molecules in CSF using proximity extension assay. Results were analyzed using highly automated unsupervised algorithmic informatics. Results Mass cytometry objectively identified a B-cell population characterized by the expression of CD49d, CD69, CD27, CXCR3, and human leukocyte antigen (HLA)-DR as clearly associated with MS. Concentrations of the B cell–related factors, notably FCRL2, were increased in MS CSF, especially in early stages of the disease. The B-cell trophic factor B cell activating factor (BAFF) was decreased in MS. Proteins involved in neural plasticity were also reduced in MS. Conclusion When analyzed without a priori assumptions, both the soluble and the cellular compartments of the CSF in MS were characterized by markers related to B cells, and the strongest candidate for an MS-specific cell type has a B-cell phenotype.
Collapse
Affiliation(s)
- David Johansson
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Céline Rauld
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Julien Roux
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Camille Regairaz
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Edoardo Galli
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Ilaria Callegari
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Layla Raad
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Annick Waldt
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Rachel Cuttat
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Guglielmo Roma
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Martin Diebold
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Burkhard Becher
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Jens Kuhle
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Tobias Derfuss
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - José M Carballido
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Nicholas S R Sanderson
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland.
| |
Collapse
|
172
|
Rozmus J. Monogenic Immune Diseases Provide Insights Into the Mechanisms and Treatment of Chronic Graft-Versus-Host Disease. Front Immunol 2021; 11:574569. [PMID: 33613511 PMCID: PMC7889949 DOI: 10.3389/fimmu.2020.574569] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 12/07/2020] [Indexed: 12/22/2022] Open
Abstract
Chronic graft-versus-host disease (GvHD) has become a leading cause of morbidity and mortality following allogeneic hematopoietic stem cell transplantation (HSCT) and can burden patients with devastating and lifelong health effects. Our understanding of the pathogenic mechanisms underlying chronic GvHD remains incomplete and this lack of understanding is reflected by lack of clear therapeutic approaches to steroid refractory disease. Observations predominantly from mouse models and human correlative studies currently support a three phase model for the initiation and development of chronic GvHD: 1) early inflammation and tissue damage triggers the innate immune system. This leads to inflammatory cytokine/chemokine patterns that recruit effector immune cell populations; 2) chronic inflammation causes the loss of central and peripheral tolerance mechanisms leading to emergence of pathogenic B and T cell populations that promote autoimmune and alloimmune reactions; 3) the dysregulated immunity causes altered macrophage polarization, aberrant tissue repair leading to scarring and end organ fibrosis. This model has led to the evaluation of many new therapies aimed at limiting inflammation, targeting dysregulated signaling pathways and restoring tolerance mechanisms. However, chronic GvHD is a multisystem disease with complex clinical phenotypes and it remains unclear as to which cluster of patients will respond best to specific therapeutic strategies. However, it is possible to gain novel insights from immune-related monogenic diseases. These diseases either share common clinical manifestations, replicate steps from the three phase chronic GvHD model or serve as surrogates for perfectly targeted drugs being investigated in chronic GvHD therapy. In this review, we will summarize the evidence from these monogenic immune related diseases that provide insight into pathogenic pathways in chronic GvHD, rationales for current therapies and novel directions for future drug discovery.
Collapse
Affiliation(s)
- Jacob Rozmus
- Division of Pediatric Hematology, Oncology & BMT, Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada.,Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, Vancouver, BC, Canada
| |
Collapse
|
173
|
Increased let-7b-5p is associated with enhanced BAFF-R expression and B cell survival in immune thrombocytopenia. Int Immunopharmacol 2021; 93:107393. [PMID: 33529914 DOI: 10.1016/j.intimp.2021.107393] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/04/2021] [Accepted: 01/11/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND B cells play a key role in the pathogenesis of immune thrombocytopenia (ITP) by producing platelet autoantibodies. Accumulating evidence suggest that microRNA (miRNA) is a critical regulator in B cells. The contribution of miRNA to B cell dysfunction in ITP has not been described. The aim of this study was to examine the expression of miRNA let-7b-5p in B cells of ITP patients and investigate its possible association with B cell function in ITP. METHODS The CD19+ cells were isolated from peripheral mononuclear cells of ITP patients and healthy controls using immunomagnetic microbeads. B cell survival in vitro was evaluated by cell counting. The level of let-7b-5p was quantified by quantitative PCR. The surface expression of B cell activating factor receptor (BAFF-R) was detected by flow cytometry. The role of let-7b-5p was examined in isolated B cells by transfecting miRNA mimics or inhibitors. RESULTS The results showed that let-7b-5p in B cells was elevated, and B cell survival was enhanced in ITP patients compared with healthy controls. BAFF and B cell receptor stimulation can induce the expression of let-7b-5p in vitro. Overexpression of let-7b-5p in B cells enhanced the expression of surface BAFF-R and promoted B cell survival. Moreover, let-7b-5p enhanced the phosphorylation of NF-κB2 p100 and upregulated the expression of survival factor Bcl-xL after BAFF induction. CONCLUSION Let-7b-5p is a pro-survival miRNA in B cells and increased let-7b-5p is associated with enhanced surface BAFF-R in ITP.
Collapse
|
174
|
BAFF Inhibition Effectively Suppresses the Development of Anti-HLA.A2 Antibody in the Highly Sensitized Mouse Model. Int J Mol Sci 2021; 22:ijms22020861. [PMID: 33467096 PMCID: PMC7830620 DOI: 10.3390/ijms22020861] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/13/2021] [Accepted: 01/13/2021] [Indexed: 11/23/2022] Open
Abstract
B cell activating factor (BAFF) is a cytokine that plays a role in the survival, proliferation and differentiation of B cells. We proposed to observe the effects of BAFF inhibition on the humoral immune responses of an allosensitized mouse model using HLA.A2 transgenic mice. Wild-type C57BL/6 mice were sensitized with skin allografts from C57BL/6-Tg (HLA-A2.1)1Enge/J mice and were treated with anti-BAFF monoclonal antibody (mAb) (named Sandy-2) or control IgG1 antibody. HLA.A2-specific IgG was reduced in BAFF-inhibited mice compared to the control group (Δ-13.62 vs. Δ27.07, p < 0.05). BAFF inhibition also resulted in increased pre-pro and immature B cell proportions and decreased mature B cells in the bone marrow (p < 0.05 vs. control). In the spleen, an increase in transitional B cells was observed with a significant decrease in marginal and follicular B cells (p < 0.05 vs. control). There was no significant difference in the proportions of long-lived plasma and memory B cells. Microarray analysis showed that 19 gene probes were significantly up- (>2-fold, p < 0.05) or down-regulated (≤2-fold, p < 0.05) in the BAFF-inhibited group. BAFF inhibition successfully reduced alloimmune responses through the reduction in alloantibody production and suppression of B cell differentiation and maturation. Our data suggest that BAFF suppression may serve as a useful target in desensitization therapy.
Collapse
|
175
|
BST-2/Tetherin is involved in BAFF-enhanced proliferation and survival via canonical NF-κB signaling in neoplastic B-lymphoid cells. Exp Cell Res 2020; 398:112399. [PMID: 33245890 DOI: 10.1016/j.yexcr.2020.112399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 11/19/2020] [Accepted: 11/22/2020] [Indexed: 01/08/2023]
Abstract
The development of Sjögren's syndrome (SS) is accompanied by B cell hyperproliferation and mutation. Our previous study identified aberrant expression of BST-2 (also known as Tetherin/CD317) in B cells from either the peripheral blood or infiltrated salivary glands. However, the roles of BST-2 in the regulation of B cell activation remain unknown. In this study, we identified that BST-2 can respond to BAFF simulation but not to other B cell simulators in neoplastic B cell lines. A CCK-8 assay, an EdU assay and Annexin V/PI staining indicated that BST-2 inhibition attenuated BAFF-enhanced proliferation and survival in both Raji cells and Daudi cells. Screening of BAFF-related signaling in neoplastic B-lymphoid cells indicated that BST-2 was involved in the regulation of NF-κB signaling upon BAFF simulation. However, inhibition of NF-κB by JSH-23 significantly reduced the proliferation and survival of Raji and Daudi cells under both normal and BAFF-simulated conditions. Collectively, our results indicate that BST-2/Tetherin is a BAFF-responsive membrane factor involved in the regulation of NF-κB signaling, thereby assisting in the proliferation and survival of neoplastic B-lymphoid cells. Our study provides a potential molecular mechanism underlying aberrant overactivation of B cells upon SS development.
Collapse
|
176
|
Eslami M, Meinl E, Eibel H, Willen L, Donzé O, Distl O, Schneider H, Speiser DE, Tsiantoulas D, Yalkinoglu Ö, Samy E, Schneider P. BAFF 60-mer, and Differential BAFF 60-mer Dissociating Activities in Human Serum, Cord Blood and Cerebrospinal Fluid. Front Cell Dev Biol 2020; 8:577662. [PMID: 33240880 PMCID: PMC7677505 DOI: 10.3389/fcell.2020.577662] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022] Open
Abstract
B cell activation factor of the TNF family (BAFF/BLyS), an essential B cell survival factor of which circulating levels are elevated in several autoimmune disorders, is targeted in the clinic for the treatment of systemic lupus erythematosus (SLE). The soluble form of BAFF can exist as 3-mer, or as 60-mer that results from the ordered assembly of twenty 3-mers and that can be obtained from naturally cleaved membrane-bound BAFF or made as a recombinant protein. However, which forms of soluble BAFF exist and act in humans is unclear. In this study, BAFF 3-mer and 60-mer in biological fluids were characterized for size, activity and response to specific stimulators or inhibitors of BAFF. Human cerebrospinal fluids (CSF) from patients with multiple sclerosis and adult human sera contained exclusively BAFF 3-mer in these assays, also when BAFF concentrations were moderately SLE or highly (BAFFR-deficient individual) increased. Human sera, but not CSF, contained a high molecular weight, saturable activity that dissociated preformed recombinant BAFF 60-mer into 3-mer. This activity was lower in cord blood. Cord blood displayed BAFF levels 10-fold higher than in adults and consistently contained a fair proportion of active high molecular weight BAFF able to dissociate into 3-mer but not endowed with all properties of recombinant BAFF 60-mer. If BAFF 60-mer is produced in humans, it is dissociated, or at least attenuated in the circulation.
Collapse
Affiliation(s)
- Mahya Eslami
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, University Hospital of the Ludwig-Maximilians-Universität München, Munich, Germany
| | - Hermann Eibel
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Freiburg, Germany
| | - Laure Willen
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | | | - Ottmar Distl
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Holm Schneider
- Department of Pediatrics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Daniel E Speiser
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | | | - Özkan Yalkinoglu
- Clinical Pharmacology, Quantitative Pharmacology, Translational Medicine, Merck KGaA, Darmstadt, Germany
| | - Eileen Samy
- Business of Merck KGaA, EMD Serono Research & Development Institute, Inc., Billerica, MA, United States
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
177
|
Im J, Baik JE, Lee D, Park OJ, Park DH, Yun CH, Han SH. Bacterial Lipoproteins Induce BAFF Production via TLR2/MyD88/JNK Signaling Pathways in Dendritic Cells. Front Immunol 2020; 11:564699. [PMID: 33123136 PMCID: PMC7566273 DOI: 10.3389/fimmu.2020.564699] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/17/2020] [Indexed: 11/13/2022] Open
Abstract
B-cell activating factor (BAFF) plays a crucial role in survival, differentiation, and antibody secretion of B cells. Microbial products with B-cell mitogenic properties can indirectly promote expansion and activation of B cells by stimulating accessory cells, such as dendritic cells (DCs), to induce BAFF. Although bacterial lipoproteins are potent B-cell mitogen like lipopolysaccharides (LPSs), it is uncertain whether they can stimulate DCs to induce BAFF expression. Here, we evaluated the effect of bacterial lipoproteins on BAFF expression in mouse bone marrow-derived DCs. Lipoprotein-deficient Staphylococcus aureus mutant induced relatively low expression level of membrane-bound BAFF (mBAFF) and the mRNA compared with its wild-type strain, implying that bacterial lipoproteins can positively regulate BAFF induction. The synthetic lipopeptides Pam2CSK4 and Pam3CSK4, which mimic bacterial lipoproteins, dose-dependently induced BAFF expression, and their BAFF-inducing capacities were comparable to those of LPS in DCs. Induction of BAFF by the lipopeptide was higher than the induction by other microbe-associated molecular patterns, including peptidoglycan, flagellin, zymosan, lipoteichoic acid, and poly(I:C). Pam3CSK4 induced both mBAFF and soluble BAFF expression in a dose- and time-dependent manner. BAFF expression by Pam3CSK4 was completely absent in DCs from TLR2- or MyD88-deficient mice. Among various MAP kinase inhibitors, only JNK inhibitors blocked Pam3CSK4-induced BAFF mRNA expression, while inhibitors blocking ERK or p38 kinase had no such effect. Furthermore, Pam3CSK4 increased the DNA-binding activities of NF-κB and Sp1, but not that of C/EBP. Pam3CSK4-induced BAFF promoter activity via TLR2/1 was blocked by NF-κB or Sp1 inhibitor. Collectively, these results suggest that bacterial lipoproteins induce expression of BAFF through TLR2/MyD88/JNK signaling pathways leading to NF-κB and Sp1 activation in DCs, and BAFF derived from bacterial lipoprotein-stimulated DCs induces B-cell proliferation.
Collapse
Affiliation(s)
- Jintaek Im
- Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Jung Eun Baik
- Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Dongwook Lee
- Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Ok-Jin Park
- Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Dong Hyun Park
- Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul, South Korea
| |
Collapse
|
178
|
Wang A, Rojas O, Lee D, Gommerman JL. Regulation of neuroinflammation by B cells and plasma cells. Immunol Rev 2020; 299:45-60. [PMID: 33107072 DOI: 10.1111/imr.12929] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 09/07/2020] [Indexed: 02/06/2023]
Abstract
The remarkable success of anti-CD20 B cell depletion therapies in reducing the burden of multiple sclerosis (MS) disease has prompted significant interest in how B cells contribute to neuroinflammation. Most focus has been on identifying pathogenic CD20+ B cells. However, an increasing number of studies have also identified regulatory functions of B lineage cells, particularly the production of IL-10, as being associated with disease remission in anti-CD20-treated MS patients. Moreover, IL-10-producing B cells have been linked to the attenuation of inflammation in experimental autoimmune encephalomyelitis (EAE), the animal model of MS. In addition to IL-10-producing B cells, antibody-producing plasma cells (PCs) have also been implicated in suppressing neuroinflammation. This review will examine regulatory roles for B cells and PCs in MS and EAE. In addition, we speculate on the involvement of regulatory PCs and the cytokine BAFF in the context of anti-CD20 treatment. Lastly, we explore how the microbiota could influence anti-inflammatory B cell behavior. A better understanding of the contributions of different B cell subsets to the regulation of neuroinflammation, and factors that impact the development, maintenance, and migration of such subsets, will be important for rationalizing next-generation B cell-directed therapies for the treatment of MS.
Collapse
Affiliation(s)
- Angela Wang
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Olga Rojas
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Dennis Lee
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
179
|
Abdelhamid L, Cabana-Puig X, Mu Q, Moarefian M, Swartwout B, Eden K, Das P, Seguin RP, Xu L, Lowen S, Lavani M, Hrubec TC, Jones CN, Luo XM. Quaternary Ammonium Compound Disinfectants Reduce Lupus-Associated Splenomegaly by Targeting Neutrophil Migration and T-Cell Fate. Front Immunol 2020; 11:575179. [PMID: 33193366 PMCID: PMC7609861 DOI: 10.3389/fimmu.2020.575179] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/30/2020] [Indexed: 12/13/2022] Open
Abstract
Hypersensitivity reactions and immune dysregulation have been reported with the use of quaternary ammonium compound disinfectants (QACs). We hypothesized that QAC exposure would exacerbate autoimmunity associated with systemic lupus erythematosus (lupus). Surprisingly, however, we found that compared to QAC-free mice, ambient exposure of lupus-prone mice to QACs led to smaller spleens with no change in circulating autoantibodies or the severity of glomerulonephritis. This suggests that QACs may have immunosuppressive effects on lupus. Using a microfluidic device, we showed that ambient exposure to QACs reduced directional migration of bone marrow-derived neutrophils toward an inflammatory chemoattractant ex vivo. Consistent with this, we found decreased infiltration of neutrophils into the spleen. While bone marrow-derived neutrophils appeared to exhibit a pro-inflammatory profile, upregulated expression of PD-L1 was observed on neutrophils that infiltrated the spleen, which in turn interacted with PD-1 on T cells and modulated their fate. Specifically, QAC exposure hindered activation of splenic T cells and increased apoptosis of effector T-cell populations. Collectively, these results suggest that ambient QAC exposure decreases lupus-associated splenomegaly likely through neutrophil-mediated toning of T-cell activation and/or apoptosis. However, our findings also indicate that even ambient exposure could alter immune cell phenotypes, functions, and their fate. Further investigations on how QACs affect immunity under steady-state conditions are warranted.
Collapse
Affiliation(s)
- Leila Abdelhamid
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Xavier Cabana-Puig
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Qinghui Mu
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
- School of Medicine, Stanford University, Stanford, CA, United States
| | - Maryam Moarefian
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA, United States
| | - Brianna Swartwout
- Translational Biology, Medicine and Health Graduate Program, Virginia Tech, Roanoke, VA, United States
| | - Kristin Eden
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Prerna Das
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Ryan P. Seguin
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA, United States
| | - Libin Xu
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA, United States
| | - Sarah Lowen
- Department of Anatomical Sciences, Edward Via College of Osteopathic Medicine-Virginia Campus, Blacksburg, VA, United States
| | - Mital Lavani
- Department of Anatomical Sciences, Edward Via College of Osteopathic Medicine-Virginia Campus, Blacksburg, VA, United States
| | - Terry C. Hrubec
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
- Department of Anatomical Sciences, Edward Via College of Osteopathic Medicine-Virginia Campus, Blacksburg, VA, United States
| | - Caroline N. Jones
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
- Department of Bioengineering, University of Texas, Dallas, TX, United States
| | - Xin M. Luo
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
180
|
Liang ZQ, Tu PC, Ji JJ, Xing QQ, Zhao X. Gu-Ben-Fang-Xiao attenuates allergic airway inflammation by inhibiting BAFF-mediated B cell activation. Biomed Pharmacother 2020; 132:110801. [PMID: 33049582 DOI: 10.1016/j.biopha.2020.110801] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/20/2020] [Accepted: 09/25/2020] [Indexed: 12/15/2022] Open
Abstract
Allergic airway inflammation is one of the major pathological events involved in the development of asthma. The B cell-activating factor (BAFF)-mediated abnormal activation of B cells plays a key role in developing allergic airway inflammation. Here, we investigated the effects of Gu-Ben-Fang-Xiao decoction (GBFXD), a TCM decoction used in the prevention and treatment of allergic asthma, on allergic airway inflammation and BAFF-mediated B cell activation. A mouse model of OVA-Severe respiratory syncytial virus (RSV) induced asthma in the remission stage was administrated with GBFXD by gavage for four weeks, after which, the pulmonary function was evaluated. Pathological changes of the lung were observed by hematoxylin and eosin (HE) staining, and serum levels of IgE, BAFF, and inflammatory factors were detected by ELISA. The expression of BAFF, APRIL, and their related receptors in the lung and spleen was detected by Western blotting and RT-qPCR. Flow cytometry detected B cell subsets in the spleen, PBC, and monocyte subsets in bronchoalveolar lavage fluid (BALF). The results showed that GBFXD improved the lung function, alleviated the inflammatory changes of the lung tissue in OVA-RSV sensitized mice, and reduced levels of IL-6, TNF-α, IL1-β, INOS, IL13 as well as IL-15, IgE, BAFF in the serum of OVA-RAV mice. Additionally, GBFXD significantly reduced the proportion of CD19+CD27+ B cell subpopulation and IgE + B cell subpopulation in the PBC and spleen cells of mice. Furthermore, the expression of BAFF, APRIL, BAFFR, TACI, and AID decreased in the lung and spleen of GBFXD-treated mice, as well as the proportion of CD11b + BAFF + cell subsets in BALF. In conclusion, GBFXD has an inhibitory effect on the secretion of BAFF by pulmonary macrophages and the expression of BAFF-related receptors, thereby reducing B cell activation and the release of IgE. This proposed mechanism contributes to the improvement of allergic airway inflammation and respiratory function in an asthmatic mouse model.
Collapse
Affiliation(s)
- Zhong-Qing Liang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China; Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China; Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, 210023, China
| | - Peng-Cheng Tu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China; Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
| | - Jian-Jian Ji
- Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, 210023, China
| | - Qiong-Qiong Xing
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China; Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China; Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, 210023, China
| | - Xia Zhao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China; Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China; Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, 210023, China.
| |
Collapse
|
181
|
Mathias LM, Stohl W. Systemic lupus erythematosus (SLE): emerging therapeutic targets. Expert Opin Ther Targets 2020; 24:1283-1302. [PMID: 33034541 DOI: 10.1080/14728222.2020.1832464] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with a heterogeneous clinical presentation whose etiologies are multifactorial. A myriad of genetic, hormonal, immunologic, and environmental factors contribute to its pathogenesis, and its diverse biological basis and phenotypic presentations make development of therapeutics difficult. In the past decade, tens of therapeutic targets with hundreds of individual candidate therapeutics have been investigated. AREAS COVERED We used a PUBMED database search through April 2020 to review the relevant literature. This review discusses therapeutic targets in the adaptive and innate immune systems, specifically: B cell surface antigens, B cell survival factors, Bruton's tyrosine kinase, costimulators, IL-12/IL-23, the calcineurin pathway, the JAK/STAT pathway, and interferons. EXPERT OPINION Our ever-improving understanding of SLE pathophysiology in the past decade has allowed us to identify new therapeutic targets. Multiple new drugs are on the horizon that target different elements of the adaptive and innate immune systems. SLE research remains challenging due to the heterogenous clinical presentation of SLE, confounding from background immunosuppressives being taken by SLE patients, animal models that inadequately recapitulate human disease, and imperfect and complicated outcome measures. Despite these limitations, research is promising and ongoing. The search for new therapies that target specific elements of SLE pathophysiology are discussed as well as key findings, pitfalls, and questions surrounding these targets.
Collapse
Affiliation(s)
- Lauren M Mathias
- Division of Rheumatology, Department of Medicine, University of Southern California Keck School of Medicine , Los Angeles, CA, USA
| | - William Stohl
- Division of Rheumatology, Department of Medicine, University of Southern California Keck School of Medicine , Los Angeles, CA, USA
| |
Collapse
|
182
|
Yang B, Zhao M, Wu H, Lu Q. A Comprehensive Review of Biological Agents for Lupus: Beyond Single Target. Front Immunol 2020; 11:539797. [PMID: 33123125 PMCID: PMC7573553 DOI: 10.3389/fimmu.2020.539797] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 09/01/2020] [Indexed: 12/29/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that involves multiple immune cells. Due to its complex pathogenesis, the effectiveness of traditional treatment methods is limited. Many patients have developed resistance to conventional treatment or are not sensitive to steroid and immunosuppressant therapy, and so emerging therapeutic antibodies have become an alternative and have been shown to work well in many patients with moderate and severe SLE. This review summarizes the biological agents that are in the preclinical and clinical trial study of SLE. In addition to the various monoclonal antibodies that have been studied for a long time, such as belimumab and rituximab, we focused on another treatment for SLE, bispecific antibodies (BsAbs) such as tibulizumab, which simultaneously targets multiple pathogenic cytokines or pathways. Although the application of BsAbs in cancer has been intensively studied, their application in autoimmune diseases is still in the infant stage. This unique combined mechanism of action may provide a novel therapeutic strategy for SLE.
Collapse
Affiliation(s)
- Bingyi Yang
- Department of Dermatology, The Second Xiangya Hospital of Central South University; Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ming Zhao
- Department of Dermatology, The Second Xiangya Hospital of Central South University; Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Haijing Wu
- Department of Dermatology, The Second Xiangya Hospital of Central South University; Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qianjin Lu
- Department of Dermatology, The Second Xiangya Hospital of Central South University; Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
183
|
NF-κB signaling induces inductive expression of the downstream molecules and IgD gene in the freshwater carp, Catla catla. 3 Biotech 2020; 10:445. [PMID: 33014688 DOI: 10.1007/s13205-020-02435-7] [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: 05/15/2020] [Accepted: 09/08/2020] [Indexed: 10/23/2022] Open
Abstract
Toll-like receptors (TLRs) in innate immune system act as primary sensors in detecting the microbial components and activate their signaling cascades to induce NF-κB (nuclear factor NF-κB) towards the augmentation of immunoglobulin (Ig) synthesis. To gain insights into the efficacy of NF-κB pathway in immunoglobulin D (IgD) synthesis in the Indian Major Carp Catla catla, cloning and sequencing of TLR-signaling downstream molecules [TRAF3 (TNF receptor-associated factor 3), NEMO (nuclear factor-kappa B essential modulator), NF-κB and BAFF (B cell activating factor)] were performed by infecting the fish with pathogens. mRNA expression analysis of the downstream molecules and IgD showed significant up-regulation of these genes in kidney (P ≤ 0.001) as compared to spleen (P ≤ 0.05). To ascertain the role of NF-κB pathway in IgD synthesis, the primary cell culture of kidney and spleen in monolayer cell suspension was treated with NF-κB inhibitor (BAY 11-7082) and down-regulation of BAFF, NEMO, NF-κB, and IgD gene was observed. These results highlight the importance of NF-κB signaling pathway in augmenting the IgD gene expression in the freshwater carp, Catla catla.
Collapse
|
184
|
Tandler C, Schmidt M, Heitmann JS, Hierold J, Schmidt J, Schneider P, Dörfel D, Walz J, Salih HR. Neutralization of B-Cell Activating Factor (BAFF) by Belimumab Reinforces Small Molecule Inhibitor Treatment in Chronic Lymphocytic Leukemia. Cancers (Basel) 2020; 12:cancers12102725. [PMID: 32977449 PMCID: PMC7598196 DOI: 10.3390/cancers12102725] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/16/2020] [Accepted: 09/21/2020] [Indexed: 01/29/2023] Open
Abstract
Simple Summary Chronic lymphocytic leukemia (CLL) is the most common form of leukemia in Western countries. Despite the substantial progress achieved by the recent introduction of the novel small molecule inhibitors idelalisib, ibrutinib and venetoclax in CLL treatment, therapy resistance occurs frequently and the disease so far remains incurable. In the present study we report that BAFF, a member of the TNF protein family, protects CLL cells from treatment-induced cell death. In turn, the therapeutic effects of idelalisib, ibrutinib and venetoclax can be reinforced by neutralizing BAFF with belimumab, an antibody which presently is clinically approved for treatment of systemic lupus erythematosus. Based on the data presented in this study, a clinical study to evaluate whether drug repurposing of belimumab for BAFF neutralization can serve to improve response to small molecule inhibitor treatment in CLL is in preparation. Abstract The introduction of idelalisib, ibrutinib and venetoclax for treatment of chronic lymphocytic leukemia (CLL) has greatly improved long term survival of patients. However, many patients do not achieve complete remission and suffer from development of resistance upon treatment with these small molecule inhibitors. Here we report that the TNF family member B-cell activating factor (BAFF) mediates resistance of CLL cells to idelalisib, ibrutinib and venetoclax by sustaining survival and preventing apoptosis of the malignant B cells as revealed by analysis of cellular ATP levels and mitochondrial membrane integrity as well as caspase activation, respectively. As BAFF also plays a prominent role in autoimmune diseases, the BAFF-neutralizing antibody belimumab was developed and approved for treatment of systemic lupus erythematosus (SLE). When we employed belimumab in the context of CLL treatment with idelalisib, ibrutinib and venetoclax, BAFF neutralization was found to significantly increase the sensitivity of the leukemic cells to all three small molecule inhibitors. Notably, BAFF neutralization proved to be beneficial independently of clinical stage according to Binet and Rai or IgVH mutational status. Our results identify drug repurposing of belimumab for neutralization of BAFF to complement small molecule inhibitor treatment as a promising therapeutic approach in CLL that is presently undergoing clinical evaluation.
Collapse
Affiliation(s)
- Claudia Tandler
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (C.T.); (M.S.); (J.S.H.); (J.H.); (J.S.); (D.D.); (J.W.)
| | - Moritz Schmidt
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (C.T.); (M.S.); (J.S.H.); (J.H.); (J.S.); (D.D.); (J.W.)
| | - Jonas S. Heitmann
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (C.T.); (M.S.); (J.S.H.); (J.H.); (J.S.); (D.D.); (J.W.)
| | - Julia Hierold
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (C.T.); (M.S.); (J.S.H.); (J.H.); (J.S.); (D.D.); (J.W.)
| | - Jonas Schmidt
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (C.T.); (M.S.); (J.S.H.); (J.H.); (J.S.); (D.D.); (J.W.)
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland;
| | - Daniela Dörfel
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (C.T.); (M.S.); (J.S.H.); (J.H.); (J.S.); (D.D.); (J.W.)
| | - Juliane Walz
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (C.T.); (M.S.); (J.S.H.); (J.H.); (J.S.); (D.D.); (J.W.)
- DFG Cluster of Excellence 2180 ‘Image-Guided and Functional Instructed Tumor Therapy’ (iFIT), Eberhard Karls University, 72076 Tuebingen, Germany
| | - Helmut R. Salih
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (C.T.); (M.S.); (J.S.H.); (J.H.); (J.S.); (D.D.); (J.W.)
- DFG Cluster of Excellence 2180 ‘Image-Guided and Functional Instructed Tumor Therapy’ (iFIT), Eberhard Karls University, 72076 Tuebingen, Germany
- Correspondence: ; Tel.: +49-7071/29-83275
| |
Collapse
|
185
|
Mendez ME, Murugesh DK, Sebastian A, Hum NR, McCloy SA, Kuhn EA, Christiansen BA, Loots GG. Antibiotic Treatment Prior to Injury Improves Post-Traumatic Osteoarthritis Outcomes in Mice. Int J Mol Sci 2020; 21:E6424. [PMID: 32899361 PMCID: PMC7503363 DOI: 10.3390/ijms21176424] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/28/2020] [Accepted: 08/29/2020] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis (OA) is a painful and debilitating disease characterized by the chronic and progressive degradation of articular cartilage. Post-traumatic OA (PTOA) is a secondary form of OA that develops in ~50% of cases of severe articular injury. Inflammation and re-occurring injury have been implicated as contributing to the progression of PTOA after the initial injury. However, there is very little known about external factors prior to injury that could affect the risk of PTOA development. To examine how the gut microbiome affects PTOA development we used a chronic antibiotic treatment regimen starting at weaning for six weeks prior to ACL rupture, in mice. A six-weeks post-injury histological examination showed more robust cartilage staining on the antibiotic (AB)-treated mice than the untreated controls (VEH), suggesting slower disease progression in AB cohorts. Injured joints also showed an increase in the presence of anti-inflammatory M2 macrophages in the AB group. Molecularly, the phenotype correlated with a significantly lower expression of inflammatory genes Tlr5, Ccl8, Cxcl13, and Foxo6 in the injured joints of AB-treated animals. Our results indicate that a reduced state of inflammation at the time of injury and a lower expression of Wnt signaling modulatory protein, Rspo1, caused by AB treatment can slow down or improve PTOA outcomes.
Collapse
Affiliation(s)
- Melanie E. Mendez
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Deepa K. Murugesh
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Aimy Sebastian
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Nicholas R. Hum
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
- UC Merced, School of Natural Sciences, Merced, CA 95343, USA
| | - Summer A. McCloy
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Edward A. Kuhn
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | | | - Gabriela G. Loots
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
- UC Merced, School of Natural Sciences, Merced, CA 95343, USA
| |
Collapse
|
186
|
Siu JH, Motallebzadeh R, Pettigrew GJ. Humoral autoimmunity after solid organ transplantation: Germinal ideas may not be natural. Cell Immunol 2020; 354:104131. [DOI: 10.1016/j.cellimm.2020.104131] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/11/2020] [Accepted: 05/11/2020] [Indexed: 12/22/2022]
|
187
|
Nascimento M, Huot-Marchand S, Gombault A, Panek C, Bourinet M, Fanny M, Savigny F, Schneider P, Le Bert M, Ryffel B, Riteau N, Quesniaux VFJ, Couillin I. B-Cell Activating Factor Secreted by Neutrophils Is a Critical Player in Lung Inflammation to Cigarette Smoke Exposure. Front Immunol 2020; 11:1622. [PMID: 32849550 PMCID: PMC7405926 DOI: 10.3389/fimmu.2020.01622] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/17/2020] [Indexed: 12/18/2022] Open
Abstract
Cigarette smoke (CS) is the major cause of chronic lung injuries, such as chronic obstructive pulmonary disease (COPD). In patients with severe COPD, tertiary lymphoid follicles containing B lymphocytes and B cell-activating factor (BAFF) overexpression are associated with disease severity. In addition, BAFF promotes adaptive immunity in smokers and mice chronically exposed to CS. However, the role of BAFF in the early phase of innate immunity has never been investigated. We acutely exposed C57BL/6J mice to CS and show early BAFF expression in the bronchoalveolar space and lung tissue that correlates to airway neutrophil and macrophage influx. Immunostaining analysis revealed that neutrophils are the major source of BAFF. We confirmed in vitro that neutrophils secrete BAFF in response to cigarette smoke extract (CSE) stimulation. Antibody-mediated neutrophil depletion significantly dampens lung inflammation to CS exposure but only partially decreases BAFF expression in lung tissue and bronchoalveolar space suggesting additional sources of BAFF. Importantly, BAFF deficient mice displayed decreased airway neutrophil recruiting chemokines and neutrophil influx while the addition of exogenous BAFF significantly enhanced this CS-induced neutrophilic inflammation. This demonstrates that BAFF is a key proinflammatory cytokine and that innate immune cells in particular neutrophils, are an unconsidered source of BAFF in early stages of CS-induced innate immunity.
Collapse
Affiliation(s)
| | | | | | - Corinne Panek
- University of Orleans and CNRS, INEM-UMR7355, Orléans, France
| | - Manon Bourinet
- University of Orleans and CNRS, INEM-UMR7355, Orléans, France
| | - Manoussa Fanny
- University of Orleans and CNRS, INEM-UMR7355, Orléans, France
| | | | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Épalinges, Switzerland
| | - Marc Le Bert
- University of Orleans and CNRS, INEM-UMR7355, Orléans, France
| | - Bernhard Ryffel
- University of Orleans and CNRS, INEM-UMR7355, Orléans, France
| | - Nicolas Riteau
- University of Orleans and CNRS, INEM-UMR7355, Orléans, France
| | | | | |
Collapse
|
188
|
Sehnert B, Burkhardt H, Dübel S, Voll RE. Cell-Type Targeted NF-kappaB Inhibition for the Treatment of Inflammatory Diseases. Cells 2020; 9:E1627. [PMID: 32640727 PMCID: PMC7407293 DOI: 10.3390/cells9071627] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/29/2022] Open
Abstract
Deregulated NF-k activation is not only involved in cancer but also contributes to the pathogenesis of chronic inflammatory diseases like rheumatoid arthritis (RA) and multiple sclerosis (MS). Ideally, therapeutic NF-KappaB inhibition should only take place in those cell types that are involved in disease pathogenesis to maintain physiological cell functions in all other cells. In contrast, unselective NF-kappaB inhibition in all cells results in multiple adverse effects, a major hindrance in drug development. Hitherto, various substances exist to inhibit different steps of NF-kappaB signaling. However, powerful tools for cell-type specific NF-kappaB inhibition are not yet established. Here, we review the role of NF-kappaB in inflammatory diseases, current strategies for drug delivery and NF-kappaB inhibition and point out the "sneaking ligand" approach. Sneaking ligand fusion proteins (SLFPs) are recombinant proteins with modular architecture consisting of three domains. The prototype SLC1 binds specifically to the activated endothelium and blocks canonical NF-kappaB activation. In vivo, SLC1 attenuated clinical and histological signs of experimental arthritides. The SLFP architecture allows an easy exchange of binding and effector domains and represents an attractive approach to study disease-relevant biological targets in a broad range of diseases. In vivo, SLFP treatment might increase therapeutic efficacy while minimizing adverse effects.
Collapse
Affiliation(s)
- Bettina Sehnert
- Department of Rheumatology and Clinical Immunology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| | - Harald Burkhardt
- Division of Rheumatology, University Hospital Frankfurt, Goethe University, and Branch for Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, 60590 Frankfurt am Main, Germany;
| | - Stefan Dübel
- Institute of Biochemistry and Biotechnology, Technical University Braunschweig, 38106 Braunschweig, Germany;
| | - Reinhard E. Voll
- Department of Rheumatology and Clinical Immunology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| |
Collapse
|
189
|
Abstract
PURPOSE OF REVIEW The advent of enhanced genetic testing has allowed for the discovery of gene defects underlying two broad categories of antibody deficiency in children: agammaglobulinemia and common variable immunodeficiency (CVID). This review describes the underlying gene defects and the clinical manifestations. RECENT FINDINGS Because novel monogenetic defects have been discovered in both categories, a strict dichotomous classification of B cell disorders as either X-linked agammaglobulinemia or common variable immunodeficiency is no longer appropriate. Advances in genetic testing technology and the decreasing cost of such testing permit more precise diagnosis of B cell disorders, more helpful information for genetic counselors, and a better understanding of the complex process of B cell development and function. More disorders await discovery.
Collapse
Affiliation(s)
- Bailee Gilchrist
- Department of Pediatrics, Allergy-Immunology and Pediatric Rheumatology Division, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - William K Dolen
- Department of Pediatrics, Allergy-Immunology and Pediatric Rheumatology Division, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, USA.
| |
Collapse
|
190
|
Ghezzi A, Banwell B, Bar-Or A, Chitnis T, Dale RC, Gorman M, Kornek B, Krupp L, Krysko KM, Nosadini M, Rostasy K, Salzer J, Schreiner T, Tenembaum S, Waubant E. Rituximab in patients with pediatric multiple sclerosis and other demyelinating disorders of the CNS: Practical considerations. Mult Scler 2020; 27:1814-1822. [PMID: 32552353 DOI: 10.1177/1352458520932798] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Anti-CD20 therapies have established efficacy in the treatment of immune-mediated neurological and non-neurological diseases. Rituximab, one of the first B-cell-directed therapies, is relatively inexpensive compared to newer anti-CD20 molecules, is available in many countries, and has been used off-label in pediatric patients with neuroimmune conditions. The objective of this paper is to describe the experience with rituximab in pediatric multiple sclerosis and other inflammatory immune-mediated disorders of the central nervous system (CNS), and to define a protocol for its use in clinical practice, in particular addressing doses, interval of administration, duration of treatment, and tests to perform at baseline and during follow-up.
Collapse
Affiliation(s)
- Angelo Ghezzi
- Centro Studi Sclerosi Multipla, Ospedale di Gallarate, ASST Valleolona, Gallarate, Italy
| | - Brenda Banwell
- Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amit Bar-Or
- Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA/Center for Neuroinflammation and Experimental Therapeutics and the Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tanuja Chitnis
- Partners Pediatric MS Center, Massachusetts General Hospital, Boston, MA, USA
| | - Russell C Dale
- Kids Neuroscience Centre and Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Mark Gorman
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Barbara Kornek
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Lauren Krupp
- Multiple Sclerosis Comprehensive Care Center, Department of Neurology, NYU Langone Health, New York, NY, USA
| | - Kristen M Krysko
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Margherita Nosadini
- Pediatric Neurology and Neurophysiology Unit, Department of Women's and Children's Health, University Hospital of Padua, Padua, Italy
| | - Kevin Rostasy
- Department of Pediatric Neurology, Children's Hospital Datteln, University Witten/Herdecke, Witten, Germany
| | - Jonatan Salzer
- Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - Teri Schreiner
- Children's Hospital Colorado, University of Colorado, Aurora, CO, USA
| | - Silvia Tenembaum
- Pediatric Neuroimmunology Program, Department of Neurology, National Pediatric Hospital Dr. Juan P. Garrahan, Buenos Aires, Argentina
| | - Emmanuelle Waubant
- UCSF Pediatric MS Clinic and UCSF Adult MS Clinic, Department of Neurology, University of California at San Francisco, San Francisco CA, USA
| |
Collapse
|
191
|
Barroso R, Morrison WI, Morrison LJ. Molecular Dissection of the Antibody Response: Opportunities and Needs for Application in Cattle. Front Immunol 2020; 11:1175. [PMID: 32595642 PMCID: PMC7304342 DOI: 10.3389/fimmu.2020.01175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/13/2020] [Indexed: 12/11/2022] Open
Abstract
Improving understanding of the bovine adaptive immune response would equip researchers to more efficiently design interventions against pathogens that impact upon food security and animal welfare. There are features of the bovine antibody response that differ substantially from other mammalian species, including the best understood models in the human and mouse. These include the ability to generate a functionally diverse immunoglobulin response despite having a fraction of the germline gene diversity that underpins this process in humans and mice, and the unique structure of a subset of immunoglobulins with "ultralong" HCDR3 domains, which are of significant interest with respect to potential therapeutics, including against human pathogens. However, a more detailed understanding of the B cell response and the production of an effective antibody response in the bovine is currently hampered by the lack of reagents for the B cell lineage. In this article we outline the current state of knowledge and capabilities with regard to B cell and antibody responses in cattle, highlight resource gaps, and summarize recent advances that have the potential to fundamentally advance our understanding of this process in the bovine host.
Collapse
Affiliation(s)
- Ruben Barroso
- Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, United Kingdom
| | - W Ivan Morrison
- Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, United Kingdom
| | - Liam J Morrison
- Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, United Kingdom
| |
Collapse
|
192
|
Sanoja-Flores L, Flores-Montero J, Pérez-Andrés M, Puig N, Orfao A. Detection of Circulating Tumor Plasma Cells in Monoclonal Gammopathies: Methods, Pathogenic Role, and Clinical Implications. Cancers (Basel) 2020; 12:E1499. [PMID: 32521788 PMCID: PMC7352573 DOI: 10.3390/cancers12061499] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 12/28/2022] Open
Abstract
Cancer dissemination and distant metastasis most frequently require the release of tumor cells into the blood circulation, both in solid tumors and most hematological malignancies, including plasma cell neoplasms. However, detection of blood circulating tumor cells in solid tumors and some hematological malignancies, such as the majority of mature/peripheral B-cell lymphomas and monoclonal gammopathies, has long been a challenge due to their very low frequency. In recent years, the availability of highly-sensitive and standardized methods for the detection of circulating tumor plasma cells (CTPC) in monoclonal gammopathies, e.g., next-generation flow cytometry (NGF), demonstrated the systematic presence of CTPC in blood in virtually every smoldering (SMM) and symptomatic multiple myeloma (MM) patient studied at diagnosis, and in the majority of patients with newly-diagnosed monoclonal gammopathies of undetermined significance (MGUS). These methods set the basis for further detailed characterization of CTPC vs. their bone marrow counterpart in monoclonal gammopathies, to investigate their role in the biology of the disease, and to confirm their strong impact on patient outcome when measured both at diagnosis and after initiating therapy. Here, we review the currently available techniques for the detection of CTPC, and determine their biological features, physiopathological role and clinical significance in patients diagnosed with distinct diagnostic categories of plasma cell neoplasms.
Collapse
Affiliation(s)
- Luzalba Sanoja-Flores
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)- University of Salamanca, 37007 Salamanca, Spain; (L.S.-F.); (J.F.-M.); (M.P.-A.)
- Centro de Investigación Biomédica en Red de Cáncer, CIBER-ONC number CB16/12/00400, Instituto Carlos III, 28029 Madrid, Spain
| | - Juan Flores-Montero
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)- University of Salamanca, 37007 Salamanca, Spain; (L.S.-F.); (J.F.-M.); (M.P.-A.)
- Centro de Investigación Biomédica en Red de Cáncer, CIBER-ONC number CB16/12/00400, Instituto Carlos III, 28029 Madrid, Spain
| | - Martín Pérez-Andrés
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)- University of Salamanca, 37007 Salamanca, Spain; (L.S.-F.); (J.F.-M.); (M.P.-A.)
- Centro de Investigación Biomédica en Red de Cáncer, CIBER-ONC number CB16/12/00400, Instituto Carlos III, 28029 Madrid, Spain
| | - Noemí Puig
- Department of Hematology, University Hospital of Salamanca, IBSAL, IBMCC (USAL-CSIC), 37007 Salamanca, Spain;
- Centro de Investigación Biomédica en Red de Cáncer, CIBER-ONC number CB16/12/00233, Instituto Carlos III, 28029 Madrid, Spain
| | - Alberto Orfao
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)- University of Salamanca, 37007 Salamanca, Spain; (L.S.-F.); (J.F.-M.); (M.P.-A.)
- Centro de Investigación Biomédica en Red de Cáncer, CIBER-ONC number CB16/12/00400, Instituto Carlos III, 28029 Madrid, Spain
| |
Collapse
|
193
|
Alturaiki W. The roles of B cell activation factor (BAFF) and a proliferation-inducing ligand (APRIL) in allergic asthma. Immunol Lett 2020; 225:25-30. [PMID: 32522667 DOI: 10.1016/j.imlet.2020.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/20/2020] [Accepted: 06/01/2020] [Indexed: 12/11/2022]
Abstract
Allergic asthma, which is the most common type of asthma, is mediated by the IgE response, and B cells are key drivers of allergic inflammation in the lungs. B cell activation factor (BAFF) and proliferation inducing ligand (APRIL) are members of the TNF superfamily. BAFF and APRIL interact with three receptors, namely the B cell activation factor receptor (BAFF-r), B cell maturation antigen (BCMA), and transmembrane activator; calcium modulator; and cyclophilin ligand interactor (TACI). The interaction of BAFF and APRIL with their receptors induces B cell activation, differentiation, and antibody production. BAFF and APRIL are produced by airway epithelial cells during the response to allergens or infectious agents, and have shown to induce local IgE production, thus establishing allergic inflammation in the airways. BAFF can maintain in inflamed airways during infection and can inhibit regulatory T cells (Tregs), thereby promoting allergic inflammation in the airways. This review aims to outline current knowledge about BAFF/APRIL systems in humans as well as in murine models of allergic asthma. The precise role of BAFF and APRIL and their receptors in allergic asthma remains unclear. Therefore, further studies are required to identify and elucidate their roles in enhancing IgE production and activating immune cells that drive the Th2 effector response and initiate allergic inflammation in asthma. Targeting BAFF/APRIL or their cognate receptors may offer a novel therapeutic approach in asthma treatment.
Collapse
Affiliation(s)
- Wael Alturaiki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia.
| |
Collapse
|
194
|
Giltiay NV, Giordano D, Clark EA. The Plasticity of Newly Formed B Cells. THE JOURNAL OF IMMUNOLOGY 2020; 203:3095-3104. [PMID: 31818922 DOI: 10.4049/jimmunol.1900928] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 09/26/2019] [Indexed: 12/21/2022]
Abstract
Newly formed B cells (NF-B cells) that emerge from the bone marrow to the periphery have often been referred to as immature or transitional B cells. However, NF-B cells have several striking characteristics, including a distinct BCR repertoire, high expression of AID, high sensitivity to PAMPs, and the ability to produce cytokines. A number of findings do not support their designation as immature because NF-B cells have the potential to become Ab-producing cells and to undergo class-switch recombination. In this review, we provide a fresh perspective on NF-B cell functions and describe some of the signals driving their activation. We summarize growing evidence supporting a role for NF-B cells in protection against infections and as a potential source of autoantibody-producing cells in autoimmune diseases such as systemic lupus erythematosus.
Collapse
Affiliation(s)
- Natalia V Giltiay
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, WA 98109; and
| | - Daniela Giordano
- Department of Immunology, University of Washington, Seattle, WA 98109
| | - Edward A Clark
- Department of Immunology, University of Washington, Seattle, WA 98109
| |
Collapse
|
195
|
Khlaiphuengsin A, Chuaypen N, Sodsai P, Buranapraditkun S, Boonpiyathad T, Hirankarn N, Tangkijvanich P. Decreased of BAFF-R expression and B cells maturation in patients with hepatitis B virus-related hepatocellular carcinoma. World J Gastroenterol 2020; 26:2645-2656. [PMID: 32523317 PMCID: PMC7265148 DOI: 10.3748/wjg.v26.i20.2645] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/27/2020] [Accepted: 05/12/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Recent evidence has indicated the role of B cells and B cell-activating factor (BAFF) in the development of hepatocellular carcinoma (HCC).
AIM To characterize circulating BAFF receptor expression and B cell subpopulations in patients with hepatitis B virus (HBV)-related HCC.
METHODS Peripheral blood samples collected from 41 patients with chronic HBV infection (25 patients without HCC and 16 patients with HCC) and 9 healthy controls were assessed for BAFF receptors [BAFF-R(B cell-activating factor receptor), transmembrane activator and cyclophilin ligand interactor, B-cell maturation antigen] and B cell subpopulations by multicolor flow cytometry.
RESULTS The frequency of BAFF-R expressing B cells to total B cells was significantly lower in patients with HCC (3.39% ± 2.12%) compared with the non-HCC group (5.37% ± 1.90%) and healthy controls (6.23% ± 2.32%), whereas there was no difference in transmembrane activator and cyclophilin ligand interactor and B-cell maturation antigen. The frequencies of CD27+IgD+ memory B cells, CD27+IgD- class-switched memory B cells and plasmablasts were significantly lower in the patients with HCC compared to patients without HCC (1.23 ± 1.17 vs 3.09 ± 1.55, P = 0.001, 0.60 ± 0.44 vs 1.69 ± 0.86, P < 0.0001 and 0.16 ± 0.12 vs 0.37 ± 0.30, P = 0.014, respectively). However, the ratio of naïve and transitional B cell did not differ significantly between the three groups. In addition, decreased BAFF-R expression on B cells was significantly correlated with large tumor size and advanced tumor stage.
CONCLUSION Our data demonstrated BAFF-R expression was reduced in B cells that involved with the frequencies of B cells maturation in patients with HCC. The depletion of BAFF-R might play an important role in the development of HCC in patients with chronic HBV infection.
Collapse
Affiliation(s)
- Apichaya Khlaiphuengsin
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Natthaya Chuaypen
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pimpayao Sodsai
- Center of Excellence in Immunology and Immune-mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Supranee Buranapraditkun
- Division of Allergy and Clinical Immunology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10330, Thailand
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center), Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | | | - Nattiya Hirankarn
- Center of Excellence in Immunology and Immune-mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pisit Tangkijvanich
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
196
|
Deenick EK, Lau A, Bier J, Kane A. Molecular and cellular mechanisms underlying defective antibody responses. Immunol Cell Biol 2020; 98:467-479. [PMID: 32348596 DOI: 10.1111/imcb.12345] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/25/2020] [Accepted: 04/27/2020] [Indexed: 12/18/2022]
Abstract
Primary immune deficiency is caused by genetic mutations that result in immune dysfunction and subsequent susceptibility to infection. Over the last decade there has been a dramatic increase in the number of genetically defined causes of immune deficiency including those which affect B-cell function. This has not only identified critical nonredundant pathways that control the generation of protective antibody responses but also revealed that immunodeficiency and autoimmunity are often closely linked. Here we explore the molecular and cellular mechanisms of these rare monogenic conditions that disrupt antibody production, which also have implications for understanding the causes of more common polygenic immune dysfunction.
Collapse
Affiliation(s)
- Elissa K Deenick
- Immunity and Inflammatory Diseases, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Anthony Lau
- Immunity and Inflammatory Diseases, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Julia Bier
- Immunity and Inflammatory Diseases, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Alisa Kane
- Immunity and Inflammatory Diseases, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,South Western Sydney Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia.,Department of Immunology and HIV, St Vincent's Hospital, Darlinghurst, NSW, Australia.,Department of Immunology, Allergy and HIV, Liverpool Hospital, Liverpool, NSW, Australia
| |
Collapse
|
197
|
Zakzuk J, Acevedo N, Harb H, Eick L, Renz H, Potaczek DP, Caraballo L. IgE Levels to Ascaris and House Dust Mite Allergens Are Associated With Increased Histone Acetylation at Key Type-2 Immune Genes. Front Immunol 2020; 11:756. [PMID: 32425942 PMCID: PMC7204827 DOI: 10.3389/fimmu.2020.00756] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/03/2020] [Indexed: 12/18/2022] Open
Abstract
Background Epigenetic changes in response to allergen exposure are still not well understood. The aim of this study was to evaluate histone acetylation levels in peripheral blood leukocytes from humans naturally infected by intestinal parasites and perennially exposed to house dust mites (HDM). Methods Peripheral blood mononuclear cells (PBMCs) were isolated by gradient centrifugation from 20 infected and 21 non-infected individuals living in a rural/village in Colombia. Histone 3 acetylation (H3Ac) and histone 4 acetylation (H4Ac) levels were measured in six immune genes previously associated with helminth immunity by chromatin immunoprecipitation (ChIP)-quantitative PCR. Then we analyzed the association between histone acetylation levels with total parasite egg burden and IgE levels. Results We found an inverse correlation between H4Ac levels in the IL13 gene and egg worm burden that remained significant after adjustment by age [−0.20 (−0.32 to −0.09), p < 0.0001]. Moreover, we found significant associations between H4Ac levels in IL4 [0.32 (0.05–0.60), p = 0.02] and CHI3L1 [0.29 (0.08–0.51), p = 0.008] with the IgE levels to Ascaris lumbricoides. In addition, the levels of specific IgE antibodies to HDM were associated with H4Ac levels in the gene TNFSF13B encoding the B cell activating factor (BAFF) [0.51 (0.26–0.76), p < 0.001]. All values are presented as beta (95% CI). Conclusion Histone acetylation levels at key type-2 immune genes in humans were modified by nematode infection and HDM allergens and are associated with the intensity of the IgE response.
Collapse
Affiliation(s)
- Josefina Zakzuk
- Institute for Immunological Research, University of Cartagena, Cartagena, Colombia
| | - Nathalie Acevedo
- Institute for Immunological Research, University of Cartagena, Cartagena, Colombia
| | - Hani Harb
- Institute of Laboratory Medicine, Member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University Marburg, Marburg, Germany
| | - Lisa Eick
- Institute of Laboratory Medicine, Member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University Marburg, Marburg, Germany
| | - Harald Renz
- Institute of Laboratory Medicine, Member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University Marburg, Marburg, Germany
| | - Daniel P Potaczek
- Institute of Laboratory Medicine, Member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University Marburg, Marburg, Germany.,John Paul II Hospital, Krakow, Poland
| | - Luis Caraballo
- Institute for Immunological Research, University of Cartagena, Cartagena, Colombia
| |
Collapse
|
198
|
Gupta S, Agrawal S. In vitro Effects of CD8+ Regulatory T Cells on Human B Cell Subpopulations. Int Arch Allergy Immunol 2020; 181:476-480. [PMID: 32248193 DOI: 10.1159/000506806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 02/26/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND CD8+ regulatory T cells (CD8+ Tregs) are relatively recently described T cell subsets that have been shown to regulate various T cell responses and appear to play a role in autoimmunity. However, their effects on B cells have not been explored. OBJECTIVES In this investigation we examine the effect of CD8+ Tregs on various subsets of peripheral B cells include naïve B cells, transitional B cells, marginal zone B cells, IgM memory B cells, class switched memory B cells, and plasmablasts, and on the expression of B cell-activating factor receptor (BAFF-R). METHODS CD8+ T cells were first purified and then activated with anti-CD3/CD28 beads to generate CD8+ Tregs. Purified CD19+ B cells were cultured alone or with sorted CD8+ Tregs (CD8+CD183+CCR7+CD45RA-) and activated with anti-CD40 monoclonal antibody and CpG. B cell subsets and the expression of BAFF-R on naïve and memory B cells were analyzed using various monoclonal antibodies and corresponding control isotypes. Ten thousand cells were acquired and analyzed by FACSCalibur using the FlowJo software. RESULTS CD8+ Tregs selectively and significantly suppressed plasmablasts without any significant effect on other B cell subsets or on the expression of BAFF-R. CONCLUSION CD8+ Tregs may play a role in autoimmunity by regulating antibody production via suppression of plasmablasts.
Collapse
Affiliation(s)
- Sudhir Gupta
- Division of Basic and Clinical Immunology, Department of Medicine, School of Medicine, University of California at Irvine, Irvine, California, USA,
| | - Sudhanshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, School of Medicine, University of California at Irvine, Irvine, California, USA
| |
Collapse
|
199
|
Peripheral B Cell Subsets in Autoimmune Diseases: Clinical Implications and Effects of B Cell-Targeted Therapies. J Immunol Res 2020; 2020:9518137. [PMID: 32280720 PMCID: PMC7125470 DOI: 10.1155/2020/9518137] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 03/01/2020] [Accepted: 03/10/2020] [Indexed: 12/14/2022] Open
Abstract
Antibody-secreting cells (ASCs) play a fundamental role in humoral immunity. The aberrant function of ASCs is related to a number of disease states, including autoimmune diseases and cancer. Recent insights into activated B cell subsets, including naïve B cell to ASC stages and their resultant cellular disturbances, suggest that aberrant ASC differentiation occurs during autoimmune diseases and is closely related to disease severity. However, the mechanisms underlying highly active ASC differentiation and the B cell subsets in autoimmune patients remain undefined. Here, we first review the processes of ASC generation. From the perspective of novel therapeutic target discovery, prediction of disease progression, and current clinical challenges, we further summarize the aberrant activity of B cell subsets including specialized memory CD11chiT-bet+ B cells that participate in the maintenance of autoreactive ASC populations. An improved understanding of subgroups may also enhance the knowledge of antigen-specific B cell differentiation. We further discuss the influence of current B cell therapies on B cell subsets, specifically focusing on systemic lupus erythematosus, rheumatoid arthritis, and myasthenia gravis.
Collapse
|
200
|
Rosales C. Neutrophils at the crossroads of innate and adaptive immunity. J Leukoc Biol 2020; 108:377-396. [DOI: 10.1002/jlb.4mir0220-574rr] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 02/17/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022] Open
Affiliation(s)
- Carlos Rosales
- Departamento de Inmunología Instituto de Investigaciones Biomédicas Universidad Nacional Autónoma de México Mexico City Mexico
| |
Collapse
|