1
|
Dong H, Liao Y, Shang M, Fu Y, Zhang H, Luo M, Hu B. Effects of co-infection with Clonorchis sinensis on T cell exhaustion levels in patients with chronic hepatitis B. J Helminthol 2024; 98:e13. [PMID: 38263743 DOI: 10.1017/s0022149x23000871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
To investigate the effects of co-infection with Clonorchis sinensis (C. sinensis) on T cell exhaustion levels in patients with chronic hepatitis B, we enrolled clinical cases in this study, including the patients with concomitant C. sinensis and HBV infection. In this study, we detected inhibitory receptors and cytokine expression in circulating CD4+ and CD8+ T cells by flow cytometry. PD-1 and TIM-3 expression levels were significantly higher on CD4+ T and CD8+ T cells from co-infected patients than on those from the HBV patients. In addition, CD4+ T cells and CD8+ T cells function were significantly inhibited by C. sinensis and HBV co-infection compared with HBV single infection, secreting lower levels of Interferon gamma (IFN-γ), Interleukin-2 (IL-2), and TNF-α. Our current results suggested that C. sinensis co-infection could exacerbate T cell exhaustion in patients with chronic hepatitis B. PD-1 and TIM-3 could be novel biomarkers for T cell exhaustion in patients with Clonorchis sinensis and chronic hepatitis B co-infection. Furthermore, it may be one possible reason for the weaker response to antiviral therapies and the chronicity of HBV infection in co-infected patients. We must realize the importance of C. sinensis treatment for HBV-infected patients. It might provide useful information for clinical doctors to choose the right treatment plans.
Collapse
Affiliation(s)
- Huimin Dong
- Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yuan Liao
- Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Mei Shang
- Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yuechun Fu
- Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hongbin Zhang
- Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Minqi Luo
- Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Bo Hu
- Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
2
|
Guo M, Yang K, Lin S, Tang J, Liu M, Zhou H, Lin H, Fan H. Coinfection with porcine circovirus type 2 and Glaesserella parasuis serotype 4 enhances pathogenicity in piglets. Vet Microbiol 2023; 278:109663. [PMID: 36680971 DOI: 10.1016/j.vetmic.2023.109663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023]
Abstract
Coinfection of Porcine circovirus type 2 (PCV2) and Glaesserella parasuis type 4 (GPS4) is widespread clinically, resulting in high morbidity and mortality, however, interactions between the two pathogens during coinfection and the coinfection pathogenesis are poorly understood. In this study, a piglet model coinfected with PCV2 and GPS4 was established; coinfection of the piglets' group showed more obvious symptoms, such as high fever and emaciation, and more severe histological lesions appeared in various organs. Importantly, piglets in the coinfection group produced lower levels of PCV2 and GPS4 antibodies, and showed high levels of inflammatory cytokines, TLR2, and TLR4, while the levels of CD4, CD8, MHC II, costimulatory molecules, and IL-12p40 were decreased. In addition, a model of macrophage 3D4/21 cells coinfection with PCV2 and GPS4 was established, coinfected cells exhibited increased expression of the cytokines IL-6, IL-8, TNF-α, IL-1β, and the receptors TLR2, TLR4, while decreased MHC II. We further demonstrate that cytokine production is associated with the activation of NF-κB and NLRP3 inflammasome signaling pathways, and TLR4 is also involved. Altogether, our findings suggest that coinfection with PCV2 and GPS4 exacerbates the inflammatory response, resulting in severe tissue damage, and probably impaired macrophage antigen presentation and T cell activation, resulting in immune dysregulation, aggravating host infection.
Collapse
Affiliation(s)
- Mengru Guo
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Kaiyue Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Shaojie Lin
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Jinsheng Tang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Mingxing Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Hong Zhou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Huixing Lin
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Hongjie Fan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
3
|
Abbadessa G, Miele G, Maida E, Vanore E, Cipriano L, Coppola C, Lavorgna L, Bonavita S. Immunomodulatory effects of ocrelizumab and candidate biomarkers for monitoring treatment response in multiple sclerosis. Mult Scler 2023:13524585221147635. [PMID: 36683286 DOI: 10.1177/13524585221147635] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Ocrelizumab is a humanized monoclonal antibody designed to bind to the CD20 molecule, resulting in a rapid depletion of B-cells; however, it has been shown that lymphocyte subpopulations other than B-cells are affected by the drug. To review the effects of ocrelizumab on circulating lymphocytes and identify candidate biomarkers to predict and monitor treatment response. A literature search for the most relevant articles from 2006 to 2022 was conducted in PubMed and Scopus. The effect of ocrelizumab on the peripheral immune system goes beyond B-cells; it also depletes T CD20 + lymphocytes. Further, ocrelizumab reshapes the T-cell response toward a low inflammatory profile and induces an increase in T CD8 + regulatory cell percentage. A higher Body Mass Index and higher B-cell count at baseline have been associated with early B-cell reappearance. Serum neurofilament light chain reduction has been associated with treatment response. Ocrelizumab treatment exerts a broad immunomodulatory effect and may be tailored based on patients' clinical and biological profiles.
Collapse
Affiliation(s)
- Gianmarco Abbadessa
- Second Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Giuseppina Miele
- Second Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Elisabetta Maida
- Second Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Emanuele Vanore
- Second Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Lorenzo Cipriano
- Second Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Cinzia Coppola
- Second Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Luigi Lavorgna
- Second Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Simona Bonavita
- Second Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
4
|
Bevers S, Kooijmans SAA, Van de Velde E, Evers MJW, Seghers S, Gitz-Francois JJJM, van Kronenburg NCH, Fens MHAM, Mastrobattista E, Hassler L, Sork H, Lehto T, Ahmed KE, El Andaloussi S, Fiedler K, Breckpot K, Maes M, Van Hoorick D, Bastogne T, Schiffelers RM, De Koker S. mRNA-LNP vaccines tuned for systemic immunization induce strong antitumor immunity by engaging splenic immune cells. Mol Ther 2022; 30:3078-3094. [PMID: 35821637 PMCID: PMC9273295 DOI: 10.1016/j.ymthe.2022.07.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 06/23/2022] [Accepted: 07/09/2022] [Indexed: 12/19/2022] Open
Abstract
mRNA vaccines have recently proven to be highly effective against SARS-CoV-2. Key to their success is the lipid-based nanoparticle (LNP), which enables efficient mRNA expression and endows the vaccine with adjuvant properties that drive potent antibody responses. Effective cancer vaccines require long-lived, qualitative CD8 T cell responses instead of antibody responses. Systemic vaccination appears to be the most effective route, but necessitates adaptation of LNP composition to deliver mRNA to antigen presenting cells. Using a design-of-experiments methodology, we tailored mRNA-LNP compositions to achieve high magnitude tumor-specific CD8 T cell responses within a single round of optimization. Optimized LNP compositions resulted in enhanced mRNA uptake by multiple splenic immune cell populations. Type I interferon and phagocytes were found essential for the T cell response. Surprisingly, we also discovered a yet unidentified role of B cells in stimulating the vaccine-elicited CD8 T cell response. Optimized LNPs displayed a similar, spleen-centered biodistribution profile in non-human primates and did not trigger histopathological changes in liver and spleen, warranting their further assessment in clinical studies. Taken together, our study clarifies the relationship between nanoparticle composition and their T cell stimulatory capacity and provides novel insights into the underlying mechanisms of effective mRNA-LNP based antitumor immunotherapy.
Collapse
Affiliation(s)
- Sanne Bevers
- eTheRNA Immunotherapies, 2845 Niel, Belgium; Laboratory for Molecular and Cellular Therapy (LMCT), Free University of Brussels, 1090 Jette, Belgium
| | - Sander A A Kooijmans
- CDL Research, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| | | | - Martijn J W Evers
- CDL Research, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| | | | | | - Nicky C H van Kronenburg
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Marcel H A M Fens
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Enrico Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, the Netherlands
| | | | - Helena Sork
- Institute of Technology, University of Tartu, 50411 Tartu, Estonia
| | - Taavi Lehto
- Institute of Technology, University of Tartu, 50411 Tartu, Estonia; Department of Laboratory Medicine, Karolinksa Institutet, 141 52 Huddinge, Sweden
| | - Kariem E Ahmed
- Department of Laboratory Medicine, Karolinksa Institutet, 141 52 Huddinge, Sweden
| | - Samir El Andaloussi
- Department of Laboratory Medicine, Karolinksa Institutet, 141 52 Huddinge, Sweden
| | | | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy (LMCT), Free University of Brussels, 1090 Jette, Belgium
| | | | | | - Thierry Bastogne
- CYBERnano, 54000 Nancy, France; CRAN, Université de Lorraine, CNRS, INRIA BIGS, 54506 Vandœuvre-lès-Nancy, France
| | | | | |
Collapse
|
5
|
Lu J, Cen Z, Tang Q, Dong J, Qin L, Wu W. The absence of B cells disrupts splenic and myocardial Treg homeostasis in coxsackievirus B3-induced myocarditis. Clin Exp Immunol 2022; 208:1-11. [PMID: 35262174 PMCID: PMC9113299 DOI: 10.1093/cei/uxac015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 01/27/2022] [Accepted: 02/05/2022] [Indexed: 01/12/2023] Open
Abstract
Although B cells are essential for humoral immunity and show noteworthy immunomodulatory activity through antibody-independent functions, the role of B cells in regulating Treg cell responses remains controversial. Tregs (CD4+CD25+Foxp3+) are considered to play an immunoprotective role in viral myocarditis (VMC) by controlling autoimmune effector T cells. Here, we proved that B-cell knockout can not only lead to significant reductions in Tregs in the spleen, blood, and heart of VMC mice but also decrease the activation and immune function of splenic Tregs, which was reversed by adoptive transfer of B cells; the transcription levels of TGF-β and Foxp3 in the myocardium were also significantly reduced. B-cell depletion by anti-CD20 impaired the anti-inflammatory function of splenic Tregs and the homeostasis of myocardial Tregs population. Moreover, B cells can convert CD4+CD25- T cells into Foxp3+ and Foxp3-, two functionally suppressive Treg subgroups. Although the reduction in myocardial inflammation in BKO mice indicates that B cells may play a proinflammatory role, the beneficial side of B cells cannot be ignored, that is, to control autoimmunity by maintaining Treg numbers. The results observed in the animal model of VMC highlight the potential harm of rituximab in the nonselective depletion of B cells in clinical applications.
Collapse
Affiliation(s)
- Jing Lu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road 6, Nanning, Guangxi Zhuang Autonomous Region 530021, Peoples Republic of China
| | - Zhihong Cen
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road 6, Nanning, Guangxi Zhuang Autonomous Region 530021, Peoples Republic of China
| | - Quan Tang
- Coronary Care Unit, Nanning First People”s Hospital. Qixing Road 89, Nanning, Guangxi Zhuang Autonomous Region 530021, Peoples Republic of China
| | - Jingwei Dong
- Department of nuclear medicine, Liuzhou People’s Hospital, Wenchang Road 8, Liuzhou, Guangxi Zhuang Autonomous Region 530021, Peoples Republic of China
| | - Lin Qin
- Coronary Care Unit, Nanning First People”s Hospital. Qixing Road 89, Nanning, Guangxi Zhuang Autonomous Region 530021, Peoples Republic of China
| | - Weifeng Wu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road 6, Nanning, Guangxi Zhuang Autonomous Region 530021, Peoples Republic of China
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine, Guangxi Medical University, Shuangyong Road 22, Nanning, Guangxi Zhuang Autonomous Region 530021, Peoples Republic of China
| |
Collapse
|
6
|
Carpenter SM, Lu LL. Leveraging Antibody, B Cell and Fc Receptor Interactions to Understand Heterogeneous Immune Responses in Tuberculosis. Front Immunol 2022; 13:830482. [PMID: 35371092 PMCID: PMC8968866 DOI: 10.3389/fimmu.2022.830482] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/07/2022] [Indexed: 12/25/2022] Open
Abstract
Despite over a century of research, Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), continues to kill 1.5 million people annually. Though less than 10% of infected individuals develop active disease, the specific host immune responses that lead to Mtb transmission and death, as well as those that are protective, are not yet fully defined. Recent immune correlative studies demonstrate that the spectrum of infection and disease is more heterogenous than has been classically defined. Moreover, emerging translational and animal model data attribute a diverse immune repertoire to TB outcomes. Thus, protective and detrimental immune responses to Mtb likely encompass a framework that is broader than T helper type 1 (Th1) immunity. Antibodies, Fc receptor interactions and B cells are underexplored host responses to Mtb. Poised at the interface of initial bacterial host interactions and in granulomatous lesions, antibodies and Fc receptors expressed on macrophages, neutrophils, dendritic cells, natural killer cells, T and B cells have the potential to influence local and systemic adaptive immune responses. Broadening the paradigm of protective immunity will offer new paths to improve diagnostics and vaccines to reduce the morbidity and mortality of TB.
Collapse
Affiliation(s)
- Stephen M. Carpenter
- Division of Infectious Disease and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Cleveland Medical Center, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Lenette L. Lu
- Division of Geographic Medicine and Infectious Diseases, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, United States
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX, United States
- Parkland Health and Hospital System, Dallas, TX, United States
| |
Collapse
|
7
|
Lee CY, Shah MK, Hoyos D, Solovyov A, Douglas M, Taur Y, Maslak P, Babady NE, Greenbaum B, Kamboj M, Vardhana SA. Prolonged SARS-CoV-2 infection in patients with lymphoid malignancies. Cancer Discov 2021; 12:62-73. [PMID: 34753749 DOI: 10.1158/2159-8290.cd-21-1033] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/18/2021] [Accepted: 10/27/2021] [Indexed: 11/16/2022]
Abstract
Coronavirus disease 2019 (COVID-19) infection results in both acute mortality and persistent and/or recurrent disease in patients with hematologic malignancies, but the drivers of persistent infection in this population are unknown. We found that B-cell lymphomas were at particularly high risk for persistent SARS-CoV-2 positivity. Further analysis of these patients identified discrete risk factors for initial disease severity as compared to disease chronicity. Active therapy and diminished T-cell counts were drivers of acute mortality in COVID-19 infected lymphoma patients. Conversely, B-cell-depleting therapy was the primary driver of re-hospitalization for COVID-19. In patients with persistent SARS-CoV-2 positivity, we observed high levels of viral entropy consistent with intrahost viral evolution, particularly in patients with impaired CD8+ T-cell immunity. These results suggest that persistent COVID-19 infection is likely to remain a risk in patients with impaired adaptive immunity and that additional therapeutic strategies are needed to enable viral clearance in this high-risk population.
Collapse
Affiliation(s)
- Christina Y Lee
- Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, USA
| | - Monika K Shah
- Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, USA
| | - David Hoyos
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center
| | | | - Melanie Douglas
- Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, USA
| | - Ying Taur
- Infectious Diseases, Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Peter Maslak
- Laboratory Medicine, Memorial Sloan Kettering Cancer Center
| | | | | | | | - Santosha A Vardhana
- Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, USA
| |
Collapse
|
8
|
B cell depletion changes the immune cell profile in multiple sclerosis patients: One-year report. J Neuroimmunol 2021; 359:577676. [PMID: 34364105 DOI: 10.1016/j.jneuroim.2021.577676] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/27/2021] [Accepted: 07/27/2021] [Indexed: 01/02/2023]
Abstract
B cell depletion therapy has been shown to be beneficial in multiple sclerosis (MS). However, the mechanism by which B cell depletion mediates its beneficial effects in MS is still unclear. To better understand how B cell depletion may benefit patients with a disease previously thought to be primarily mediated by CD4 T cells, immune profiles were monitored in 48 patients in a phase II trial of ublituximab, a glycoengineered CD20 monoclonal antibody, at 18 time points over a year. As we previously described there was a significant shift in the percentages of T cells, NK cells, and myeloid cells following the initial dose of ublituximab, but this shift normalized within a week and these populations remained stable for the duration of the study. However, T cell subsets changed with an increase in the percentage of naïve CD4 and CD8 T cells and a decline in memory T cells. Importantly, the percentage of Th1 and CD4+GM-CSF+ T cells decreased, while the percentage of Tregs continued to increase over the year. Ublituximab not only depleted CD20+ B cells, but also CD20+ T cells. The favorable changes in the T cell subsets may contribute to the beneficial effects of B cell depletion therapy.
Collapse
|
9
|
Márquez AC, Shanina I, Horwitz MS. Multiple Sclerosis-Like Symptoms in Mice Are Driven by Latent γHerpesvirus-68 Infected B Cells. Front Immunol 2020; 11:584297. [PMID: 33329556 PMCID: PMC7711133 DOI: 10.3389/fimmu.2020.584297] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/21/2020] [Indexed: 12/16/2022] Open
Abstract
Multiple sclerosis (MS) is caused by a combination of genetic and environmental factors. It is believed that previous infection with Epstein Barr Virus (EBV) plays an important role in the development of MS. Previously, we developed a murine model where latent infection with gamma herpesvirus 68 (γHV-68), a murine homolog to EBV, enhanced the symptoms of experimental autoimmune encephalomyelitis (EAE), resulting in disease that more closely resembles MS in humans. Here, we explored the conditions that were necessary for EAE enhancement. We showed that latently infected CD19+IgD− B cells were capable of enhancing EAE symptoms when transferred from mice previously infected with γHV-68 into uninfected mice. We also observed a prevention of enhancement when B cells were depleted before infection. However, depletion after the establishment of latency only partially reduced EAE. This indicated the existence of a mechanism where B cells play an important role as antigen presenting cells (APCs) prior to EAE induction for the priming of Th1 cells. It is possible that these signals persist even after B cell depletion, strongly suggesting a paracrine signaling modulation of non-B cell APCs. These results strongly support the concept that EBV contributes to the development of autoimmunity and highlights the need for a vaccine against EBV that could limit or prevent multiple sclerosis development.
Collapse
Affiliation(s)
- Ana Citlali Márquez
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC, Canada
| | - Iryna Shanina
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC, Canada
| | - Marc Steven Horwitz
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
10
|
Zrzavy T, Wimmer I, Rommer PS, Berger T. Immunology of COVID-19 and disease-modifying therapies: The good, the bad and the unknown. Eur J Neurol 2020; 28:3503-3516. [PMID: 33090599 PMCID: PMC7675490 DOI: 10.1111/ene.14578] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/04/2020] [Indexed: 01/08/2023]
Abstract
Objective The outbreak of the SARS‐CoV‐2 pandemic, caused by a previously unknown infectious agent, posed unprecedented challenges to healthcare systems and unmasked their vulnerability and limitations worldwide. Patients with long‐term immunomodulatory/suppressive therapies, as well as their physicians, were and are concerned about balancing the risk of infection and effects of disease‐modifying therapy. Over the last few months, knowledge regarding SARS‐CoV‐2 has been growing tremendously, and the first experiences of infections in patients with multiple sclerosis (MS) have been reported. Methods This review summarizes the currently still limited knowledge about SARS‐CoV‐2 immunology and the commonly agreed modes of action of approved drugs in immune‐mediated diseases of the central nervous system (MS and neuromyelitis optica spectrum disorder). Specifically, we discuss whether immunosuppressive/immunomodulatory drugs may increase the risk of SARS‐CoV‐2 infection and, conversely, may decrease the severity of a COVID‐19 disease course. Results At present, it can be recommended in general that none of those therapies with a definite indication needs to be stopped per se. A possibly increased risk of infection for most medications is accompanied by the possibility to reduce the severity of COVID‐19. Conclusions Despite the knowledge gain over the last few months, current evidence remains limited, and, thus, further clinical vigilance and systematic documentation is essential.
Collapse
Affiliation(s)
- Tobias Zrzavy
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Isabella Wimmer
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Paulus S Rommer
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
11
|
Davis JS, Ferreira D, Paige E, Gedye C, Boyle M. Infectious Complications of Biological and Small Molecule Targeted Immunomodulatory Therapies. Clin Microbiol Rev 2020; 33:e00035-19. [PMID: 32522746 PMCID: PMC7289788 DOI: 10.1128/cmr.00035-19] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The past 2 decades have seen a revolution in our approach to therapeutic immunosuppression. We have moved from relying on broadly active traditional medications, such as prednisolone or methotrexate, toward more specific agents that often target a single receptor, cytokine, or cell type, using monoclonal antibodies, fusion proteins, or targeted small molecules. This change has transformed the treatment of many conditions, including rheumatoid arthritis, cancers, asthma, and inflammatory bowel disease, but along with the benefits have come risks. Contrary to the hope that these more specific agents would have minimal and predictable infectious sequelae, infectious complications have emerged as a major stumbling block for many of these agents. Furthermore, the growing number and complexity of available biologic agents makes it difficult for clinicians to maintain current knowledge, and most review articles focus on a particular target disease or class of agent. In this article, we review the current state of knowledge about infectious complications of biologic and small molecule immunomodulatory agents, aiming to create a single resource relevant to a broad range of clinicians and researchers. For each of 19 classes of agent, we discuss the mechanism of action, the risk and types of infectious complications, and recommendations for prevention of infection.
Collapse
Affiliation(s)
- Joshua S Davis
- Department of Infectious Diseases and Immunology, John Hunter Hospital, Newcastle, NSW, Australia
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia
| | - David Ferreira
- School of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Emma Paige
- Department of Infectious Diseases, Alfred Hospital, Melbourne, VIC, Australia
| | - Craig Gedye
- School of Medicine, University of New South Wales, Sydney, NSW, Australia
- Department of Oncology, Calvary Mater Hospital, Newcastle, NSW, Australia
| | - Michael Boyle
- Department of Infectious Diseases and Immunology, John Hunter Hospital, Newcastle, NSW, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
12
|
Abstract
Monoclonal antibody targeting the CD20 antigen on B cells is used to treat the majority of non-Hodgkin lymphoma patients and some autoimmune disorders. This therapy generates adverse effects, notably opportunistic infections and activation of viruses from latency. Here, using the infection murine model with the intracellular parasite Trypanosoma cruzi, we report that anti-CD20 treatment affects not only B cell responses but also CD8+ T cell responses, representing the most important immune effectors involved in control of intracellular pathogens. Anti-CD20 treatment, directly or indirectly, affects cytotoxic T cell number and function, and this deficient response was rescued by the cytokine IL-17A. The identification of IL-17A as the cytokine capable of reversing the poor response of CD8+ T cells provides information about a potential therapeutic treatment aimed at enhancing defective immunity induced by B cell depletion. Treatment with anti-CD20, used in many diseases in which B cells play a pathogenic role, has been associated with susceptibility to intracellular infections. Here, we studied the effect of anti-CD20 injection on CD8+ T cell immunity using an experimental model of Trypanosoma cruzi infection, in which CD8+ T cells play a pivotal role. C57BL/6 mice were treated with anti-CD20 for B cell depletion prior to T. cruzi infection. Infected anti-CD20-treated mice exhibited a CD8+ T cell response with a conserved expansion phase followed by an early contraction, resulting in a strong reduction in total and parasite-specific CD8+ T cell numbers at 20 days postinfection. Anti-CD20 injection increased the frequency of apoptotic CD8+ T cells, decreased the number of effector and memory CD8+ T cells, and reduced the frequency of proliferating and cytokine-producing CD8+ T cells. Accordingly, infected anti-CD20-treated mice presented lower cytotoxicity of T. cruzi peptide-pulsed target cells in vivo. All of these alterations in CD8+ T cell immunity were associated with increased tissue parasitism. Anti-CD20 injection also dampened the CD8+ T cell response, when this had already been generated, indicating that B cells were involved in the maintenance rather than the induction of CD8+ T cell immunity. Anti-CD20 injection also resulted in a marked reduction in the frequency of interleukin-6 (IL-6)- and IL-17A-producing cells, and recombinant IL-17A (rIL-17A) injection partially restored the CD8+ T cell response in infected anti-CD20-treated mice. Thus, anti-CD20 reduced CD8+ T cell immunity, and IL-17A is a candidate for rescuing deficient responses either directly or indirectly.
Collapse
|
13
|
Porcine Circovirus Type 2 Rep Enhances IL-10 Production in Macrophages via Activation of p38-MAPK Pathway. Viruses 2019; 11:v11121141. [PMID: 31835539 PMCID: PMC6950681 DOI: 10.3390/v11121141] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/06/2019] [Accepted: 12/08/2019] [Indexed: 12/21/2022] Open
Abstract
Porcine circovirus type 2 (PCV2) is one of the major threats to pig farms worldwide. Although PCV2 has been identified to promote IL-10 production, the detailed regulatory roles of PCV2 Rep for IL-10 production remain unclear. Herein, we first found that PCV2 Rep, rather than PCV1 Rep, enhanced IL-10 expression at the later phase of PCV2 infection in porcine alveolar macrophages (PAMs). Furthermore, we found that PCV2 Rep directly activated the p38-MAPK pathway to promote transcription factors NF-κB p50 and Sp1 binding to the il10 promoter, but PCV1 Rep did not. During PCV2 infection, however, PCV2 Rep promoted the binding activities of NF-κB p50 and Sp1 with the il10 promoter only at the later phase of PCV2 infection, since Rep proteins only expressed at the later phase of the infection. Moreover, silence of the thymine DNA glycosylase (TDG), a Rep-binding protein, significantly reduced the binding activities of NF-κB p50 and Sp1 with il10 promoter, resulting in the reduction of IL-10 production in PCV2-inoculated PAMs at the later phase of infection. Taken together, our results demonstrate that Rep proteins enhance IL-10 production during PCV2 infection of PAMs via activation of p38-MAPK pathways, in which host TDG is a critical mediator.
Collapse
|
14
|
Milcent B, Josseaume N, Petitprez F, Riller Q, Amorim S, Loiseau P, Toubert A, Brice P, Thieblemont C, Teillaud JL, Sibéril S. Recovery of central memory and naive peripheral T cells in Follicular Lymphoma patients receiving rituximab-chemotherapy based regimen. Sci Rep 2019; 9:13471. [PMID: 31530876 PMCID: PMC6748924 DOI: 10.1038/s41598-019-50029-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 09/04/2019] [Indexed: 02/08/2023] Open
Abstract
Preclinical models and clinical studies have shown that anti-CD20-based treatment has multifaceted consequences on T-cell immunity. We have performed a prospective study of peripheral T-cell compartment in FL patients, all exhibiting high tumor burden and receiving rituximab-chemotherapy-based regimen (R-CHOP). Before treatment, FL patients harbor low amounts of peripheral naive T cells, but high levels of CD4+ TEM, CD4+ Treg and CD8+ TEMRA subsets and significant amounts of CD38+ HLA-DR+ activated T cells. A portion of these activated/differentiated T cells also expressed PD-1 and/or TIGIT immune checkpoints. Hierarchical clustering of phenotyping data revealed that 5/8 patients with only a partial response to R-CHOP induction therapy or with disease progression segregate into a group exhibiting a highly activated/differentiated T cell profile and a markedly low proportion of naive T cells before treatment. Rituximab-based therapy induced a shift of CD4+ and CD8+ T cells toward a central memory phenotype and of CD8+ T cells to a naive phenotype. In parallel, a decrease in the number of peripheral T cells expressing both PD-1 and TIGIT was detected. These observations suggest that the standard rituximab-based therapy partially reverts the profound alterations observed in T-cell subsets in FL patients, and that blood T-cell phenotyping could provide a better understanding of the mechanisms of rituximab-based treatment.
Collapse
Affiliation(s)
- B Milcent
- Cordeliers Research Center-Inserm UMR-S 1138, "Cancer, Immune Control and Escape" Laboratory, Paris, 75006, France.,Sorbonne Université, UMR-S 1138, Paris, 75006, France.,Paris Descartes-Paris 5 University, UMR-S 1138, Paris, 75006, France
| | - N Josseaume
- Cordeliers Research Center-Inserm UMR-S 1138, "Cancer, Immune Control and Escape" Laboratory, Paris, 75006, France.,Sorbonne Université, UMR-S 1138, Paris, 75006, France.,Paris Descartes-Paris 5 University, UMR-S 1138, Paris, 75006, France
| | - F Petitprez
- Cordeliers Research Center-Inserm UMR-S 1138, "Cancer, Immune Control and Escape" Laboratory, Paris, 75006, France.,Sorbonne Université, UMR-S 1138, Paris, 75006, France.,Paris Descartes-Paris 5 University, UMR-S 1138, Paris, 75006, France.,Ligue Nationale Contre le Cancer, Programme Cartes d'Identité des Tumeurs, Paris, 75014, France
| | - Q Riller
- Cordeliers Research Center-Inserm UMR-S 1138, "Cancer, Immune Control and Escape" Laboratory, Paris, 75006, France.,Sorbonne Université, UMR-S 1138, Paris, 75006, France.,Paris Descartes-Paris 5 University, UMR-S 1138, Paris, 75006, France
| | - S Amorim
- APHP, Saint-Louis Hospital, Hemato-oncology - Diderot University, Sorbonne Paris Cité, Paris, France
| | - P Loiseau
- Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint-Louis, Paris, France.,Inserm UMR-S 1160, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, 7, France
| | - A Toubert
- Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint-Louis, Paris, France.,Inserm UMR-S 1160, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, 7, France
| | - P Brice
- APHP, Saint-Louis Hospital, Hemato-oncology - Diderot University, Sorbonne Paris Cité, Paris, France
| | - C Thieblemont
- APHP, Saint-Louis Hospital, Hemato-oncology - Diderot University, Sorbonne Paris Cité, Paris, France.,EA7324 Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - J-L Teillaud
- Cordeliers Research Center-Inserm UMR-S 1138, "Cancer, Immune Control and Escape" Laboratory, Paris, 75006, France.,Sorbonne Université, UMR-S 1138, Paris, 75006, France.,Paris Descartes-Paris 5 University, UMR-S 1138, Paris, 75006, France.,Laboratory "Immune Microenvironment and Biotherapy", Sorbonne University UMRS1135, INSERM U.1135, Centre d'Immunologie et des Maladies Infectieuses (CIMI), Paris, France
| | - S Sibéril
- Cordeliers Research Center-Inserm UMR-S 1138, "Cancer, Immune Control and Escape" Laboratory, Paris, 75006, France. .,Sorbonne Université, UMR-S 1138, Paris, 75006, France. .,Paris Descartes-Paris 5 University, UMR-S 1138, Paris, 75006, France.
| |
Collapse
|
15
|
Dong Y, Li X, Zhang L, Zhu Q, Chen C, Bao J, Chen Y. CD4 + T cell exhaustion revealed by high PD-1 and LAG-3 expression and the loss of helper T cell function in chronic hepatitis B. BMC Immunol 2019; 20:27. [PMID: 31390978 PMCID: PMC6686459 DOI: 10.1186/s12865-019-0309-9] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 07/29/2019] [Indexed: 12/17/2022] Open
Abstract
Background Immune inhibitory receptors play an important role in chronic infections. However, little is known about their role in hepatitis B virus (HBV) infection. Here, we analyzed the relationship between programmed death-1 (PD-1) and lymphocyte activation gene-3 (LAG-3) expression on CD4+ T cells and HBV disease progression. Results PD-1 and LAG-3 expression was significantly higher on CD4+ T cells from HBV patients than on those from the HCs. In addition, a significant positive correlation was found between the PD-1 and LAG-3 expression levels and the ALT(alanine aminotransferase) level. CD4+ T cell function was inhibited by high PD-1 and LAG-3 levels, and CD4+ T cells with high PD-1 and LAG-3 expression lost the ability to secrete IFN-γ, IL-2 and TNF-α. Furthermore, blockade of the PD-1 and LAG-3 pathways reversed the damage to CD4+ T cell proliferation and cytokine secretion. Conclusions CD4+ T cell exhaustion during chronic HBV had high PD-1 and LAG-3 expression and the absence of helper T cell cytokines, including IFN-γ, IL-2 and TNF-α. After blocking PD-L1 and LAG-3, CD4+ T cell function in chronic hepatitis B patients was partially restored.
Collapse
Affiliation(s)
- Yuejiao Dong
- Department of Laboratory Medicine, Key Laboratory of Clinical In Vitro Diagnostic Techniques of Zhejiang Province, First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, China
| | - Xuefen Li
- Department of Laboratory Medicine, Key Laboratory of Clinical In Vitro Diagnostic Techniques of Zhejiang Province, First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, China
| | - Lu Zhang
- Department of Laboratory Medicine, Key Laboratory of Clinical In Vitro Diagnostic Techniques of Zhejiang Province, First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, China
| | - Qiaoyun Zhu
- Department of Laboratory Medicine, Key Laboratory of Clinical In Vitro Diagnostic Techniques of Zhejiang Province, First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, China
| | - Chunlei Chen
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, China
| | - Jiaqi Bao
- Department of Laboratory Medicine, Key Laboratory of Clinical In Vitro Diagnostic Techniques of Zhejiang Province, First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, China
| | - Yu Chen
- Department of Laboratory Medicine, Key Laboratory of Clinical In Vitro Diagnostic Techniques of Zhejiang Province, First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, China. .,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, China.
| |
Collapse
|
16
|
Platt JL, Cascalho M. Non-canonical B cell functions in transplantation. Hum Immunol 2019; 80:363-377. [PMID: 30980861 PMCID: PMC6544480 DOI: 10.1016/j.humimm.2019.04.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/04/2019] [Accepted: 04/09/2019] [Indexed: 12/18/2022]
Abstract
B cells are differentiated to recognize antigen and respond by producing antibodies. These activities, governed by recognition of ancillary signals, defend the individual against microorganisms and the products of microorganisms and constitute the canonical function of B cells. Despite the unique differentiation (e.g. recombination and mutation of immunoglobulin gene segments) toward this canonical function, B cells can provide other, "non-canonical" functions, such as facilitating of lymphoid organogenesis and remodeling and fashioning T cell repertoires and modifying T cell responses. Some non-canonical functions are exerted by antibodies, but most are mediated by other products and/or direct actions of B cells. The diverse set of non-canonical functions makes the B cell as much as any cell a central organizer of innate and adaptive immunity. However, the diverse products and actions also confound efforts to weigh the importance of individual non-canonical B cell functions. Here we shall describe the non-canonical functions of B cells and offer our perspective on how those functions converge in the development and governance of immunity, particularly immunity to transplants, and hurdles to advancing understanding of B cell functions in transplantation.
Collapse
Affiliation(s)
- Jeffrey L Platt
- Departments of Surgery and of Microbiology & Immunology, University of Michigan, Ann Arbor, MI, United States.
| | - Marilia Cascalho
- Departments of Surgery and of Microbiology & Immunology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
17
|
Mature IgD low/- B cells maintain tolerance by promoting regulatory T cell homeostasis. Nat Commun 2019; 10:190. [PMID: 30643147 PMCID: PMC6331566 DOI: 10.1038/s41467-018-08122-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 12/19/2018] [Indexed: 12/31/2022] Open
Abstract
A number of different B cell subsets have been shown to exhibit regulatory activity using a variety of mechanisms to attenuate inflammatory diseases. Here we show, using anti-CD20-mediated partial B cell depletion in mice, that a population of mature B cells distinguishable by IgDlow/- expression maintains tolerance by, at least in part, promoting CD4+Foxp3+ regulatory T cell homeostatic expansion via glucocorticoid-induced tumor necrosis factor receptor ligand, or GITRL. Cell surface phenotyping, transcriptome analysis and developmental study data show that B cells expressing IgD at a low level (BDL) are a novel population of mature B cells that emerge in the spleen from the transitional-2 stage paralleling the differentiation of follicular B cells. The cell surface phenotype and regulatory function of BDL are highly suggestive that they are a new B cell subset. Human splenic and peripheral blood IgDlow/- B cells also exhibit BDL regulatory activity, rendering them of therapeutic interest.
Collapse
|
18
|
Sanjo N, Nose Y, Shishido-Hara Y, Mizutani S, Sekijima Y, Aizawa H, Tanizawa T, Yokota T. A controlled inflammation and a regulatory immune system are associated with more favorable prognosis of progressive multifocal leukoencephalopathy. J Neurol 2018; 266:369-377. [PMID: 30511098 DOI: 10.1007/s00415-018-9140-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/20/2018] [Accepted: 11/24/2018] [Indexed: 01/09/2023]
Abstract
OBJECTIVE In the present study, we analyzed the inflammatory profiles of brain tissues obtained from patients with progressive multifocal leukoencephalopathy (PML) due to John Cunningham (JC) virus infection to identify potential prognostic factors. METHODS The study included seven patients (two men, five women) who had been pathologically diagnosed with PML, and all of whom were HIV negative. Fixed brain samples were analyzed via hematoxylin and eosin (HE) staining and Klüver-Barrera (KB) staining. We then performed immunohistochemistry (IHC) specific to JC virus capsid proteins (VP1 and VP2/3) and lymphocyte surface markers (CD4, CD8, CD138, and PD-1). RESULTS The mean age at onset was 53.4, while the mean duration until biopsy/autopsy was 4.7 months. Four patients were included in the good prognosis (GP) group, while three were included in the poor prognosis (PP) group. Pathological analysis revealed a significantly larger number of CD4-positive T-cell infiltrations (P = .029) in the GP group, along with a preserved CD4:CD8 ratio. Larger numbers of CD138-positive plasma cells were also observed in the GP group (P = .029) than in the PP group. Linear regression analyses revealed a significant association between the numbers of CD138-positive plasma cells and PD-1-positive cells (R2 = 0.80). CONCLUSIONS Viral loads in the cerebrospinal fluid, a controlled inflammatory response mediated by CD4- and CD8-positive T cells, and plasma cells are associated with PML prognosis. Our findings further indicate that regulatory plasma cells may regulate inflammatory T-cell activity via a PD-1/PD-L1 immuno-checkpoint pathway, thereby protecting the uninfected brain from excessive immune-mediated damage during an active JC virus infection.
Collapse
Affiliation(s)
- Nobuo Sanjo
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University Graduate School of Medical and Dental Sciences, 1-5-45 Yushima Bunkyo-ku, Tokyo, 113-8510, Japan.
| | - Yurie Nose
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University Graduate School of Medical and Dental Sciences, 1-5-45 Yushima Bunkyo-ku, Tokyo, 113-8510, Japan
| | | | - Saneyuki Mizutani
- Department of Internal Medicine (Neurology), Tokyo Metropolitan Bokutoh Hospital, Tokyo, Japan
| | - Yoshiki Sekijima
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Nagano, Japan
| | - Hitoshi Aizawa
- Department of Neurology, Tokyo Medical University, Tokyo, Japan
| | - Toru Tanizawa
- Department of Pathology, Tokyo Metropolitan Bokutoh Hospital, Tokyo, Japan
| | - Takanori Yokota
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University Graduate School of Medical and Dental Sciences, 1-5-45 Yushima Bunkyo-ku, Tokyo, 113-8510, Japan
| |
Collapse
|
19
|
Nguyen TG. Immune-modulation via IgD B-cell receptor suppresses allergic skin inflammation in experimental contact hypersensitivity models despite of a Th2-favoured humoral response. Immunol Lett 2018; 203:29-39. [PMID: 30218740 DOI: 10.1016/j.imlet.2018.09.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/30/2018] [Accepted: 09/11/2018] [Indexed: 12/11/2022]
Abstract
Atopic dermatitis (AD) and allergic contact dermatitis (ACD) are common skin inflammatory conditions. B and T cells are strongly implicated in allergic contact hypersensitivity (CHS) conditions. Activation of IgD B-cell receptor (BCR) by anti-IgD stimulation depletes mature B cells and modulates T-helper cell type 1/2 (Th1/2) responses in vivo. It is not known whether these effects by anti-IgD exacerbates or ameliorates chronic skin inflammations. This study investigated the effects of anti-IgD and B-cell depleting anti-CD20 antibody on skin inflammation in CHS murine models. Chronic CHS were induced by challenges with allergens trimellitic anhydride (TMA) or 2,4 dinitrochlorobenzene (DNCB). Mice were treated with an anti-IgD or anti-CD20 at various time-points following allergen challenges. This study revealed that early therapeutic treatments with anti-IgD at 4 h after allergen challenge significantly reduced skin inflammation in both TMA- and DNCB-induced CHS models (P < 0.05). In contrast, anti-CD20 treatment exacerbated skin inflammation in DNCB-induced CHS despite of an extensive B cell depletion (P < 0.05). Anti-IgD treatment depleted mature CD19+IgD+ B cells but enhanced allergen-specific IgM and total IgE productions, suggesting a Th2-favoured humoral response. Anti-IgD reduced neutrophilic infiltrations but increases accumulation of mast cells in dermal tissues. The anti-inflammatory effects of anti-IgD were supported by evidence of an increase in the percentage of regulatory B cells and T cells. Collectively, this study demonstrates that immune-modulation by anti-IgD treatment suppresses Th2-mediated allergic skin inflammation in murine models despite a skew toward a Th2-favvoured humoral response and therefore may present a novel treatment for chronic human AD and ACD.
Collapse
Affiliation(s)
- Tue G Nguyen
- Autoimmunity and Immunotherapy Research, Kolling Institute, Australia; Perinatal Research, Kolling Institute at Royal North Shore Hospital, St Leonards, NSW, 2065, Australia; ImmunoTherapeutic Mab Group, Macquarie Park, Sydney, NSW, 2113, Australia.
| |
Collapse
|
20
|
T cell deficiencies as a common risk factor for drug associated progressive multifocal leukoencephalopathy. Immunobiology 2018; 223:508-517. [PMID: 29472141 DOI: 10.1016/j.imbio.2018.01.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/19/2018] [Accepted: 01/21/2018] [Indexed: 01/07/2023]
Abstract
Progressive multifocal leukoencephalopathy (PML) is a disease of the central nervous system caused by neuropathogenic prototypes of ubiquitous community-acquired JC virus (JCV). The disease became of particular concern following its association with certain therapies that modulate immune system function without heavy immunosuppression. Due to lack of prophylactic/treatment options and poor outcomes, which often include severe disability or death, PML is a considerable concern for development of new drugs that interfere with immune system functions. In this review of clinical and research findings, we discuss the evidence that deficiencies in CD4+ T helper cells, cytotoxic CD8+ T cells, and interferon gamma are of crucial importance for development of PML under a variety of circumstances, including those associated with use of various drugs, regardless of differences in their mechanisms of action. These deficiencies apparently enable transformation of the harmless JCV archetype into neuropathogenic prototypes, but the site(s), and the mechanisms, of this transformation are yet to be elucidated. Here we discuss the evidence for brain as one of the sites of this transformation, and propose a model of PML pathogenesis that emphasizes the central role of T cell deficiencies in the two life cycles of the JCV, one non-pathogenic and one neuropathogenic. Finally, we conclude that the development of clinical grade T cell functional tests and more consistent use of already available laboratory tests for T cell subset analysis would greatly aid the effort to more accurately predict and assess the magnitude of PML risk for concerned therapeutic interventions.
Collapse
|
21
|
Grebenciucova E, Pruitt A. Infections in Patients Receiving Multiple Sclerosis Disease-Modifying Therapies. Curr Neurol Neurosci Rep 2017; 17:88. [PMID: 28940162 DOI: 10.1007/s11910-017-0800-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW This paper will systemically review the risk of infections associated with current disease-modifying treatments and will discuss pre-treatment testing recommendations, infection monitoring strategies, and patient education. RECENT FINDINGS Aside from glatiramer acetate and interferon-beta therapies, all other multiple sclerosis treatments to various degrees impair immune surveillance and may predispose patients to the development of both community-acquired and opportunistic infections. Some of these infections are rarely seen in neurologic practice, and neurologists should be aware of how to monitor for these infections and how to educate patients about medication-specific risks. Of particular interest in this discussion is the risk of PML in association with the recently approved B cell depleting therapy, ocrelizumab, particularly when switching from natalizumab. The risk of infection in association with MS treatments has become one of the most important factors in the choice of therapy. Balance of the overall risk versus benefit should be continuously re-evaluated during treatment.
Collapse
Affiliation(s)
- Elena Grebenciucova
- Multiple Sclerosis Division, Davee Department of Neurology and Clinical Neurosciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| | - Amy Pruitt
- Multiple Sclerosis Division, the Department of Neurology, Perelman School of Medicine, The University of Pennsylvania, 3400 Convention Avenue, Philadelphia, PA, 19104, USA
| |
Collapse
|
22
|
Fortner KA, Bond JP, Austin JW, Boss JM, Budd RC. The molecular signature of murine T cell homeostatic proliferation reveals both inflammatory and immune inhibition patterns. J Autoimmun 2017; 82:47-61. [PMID: 28551033 PMCID: PMC5902411 DOI: 10.1016/j.jaut.2017.05.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 05/04/2017] [Accepted: 05/09/2017] [Indexed: 01/21/2023]
Abstract
T lymphocyte homeostatic proliferation, driven by the engagement of T cell antigen receptor with self-peptide/major histocompatibility complexes, and signaling through the common γ-chain-containing cytokine receptors, is critical for the maintenance of the T cell compartment and is regulated by the Fas death receptor (Fas, CD95). In the absence of Fas, Fas-deficient lymphoproliferation spontaneous mutation (lpr) mice accumulate homeostatically expanded T cells. The functional consequences of sequential rounds of homeostatic expansion are not well defined. We thus examined the gene expression profiles of murine wild-type and Fas-deficient lpr CD8+ T cell subsets that have undergone different amounts of homeostatic proliferation as defined by their level of CD44 expression, and the CD4-CD8-TCRαβ+ T cell subset that results from extensive homeostatic expansion of CD8+ T cells. Our studies show that recurrent T cell homeostatic proliferation results in global gene expression changes, including the progressive upregulation of both cytolytic proteins such as Fas-Ligand and granzyme B as well as inhibitory proteins such as programmed cell death protein 1 (PD-1) and lymphocyte activating 3 (Lag3). These findings provide an explanation for how augmented T cell homeostatic expansion could lead to the frequently observed clinical paradox of simultaneous autoinflammatory and immunodeficiency syndromes and provide further insight into the regulatory programs that control chronically stimulated T cells.
Collapse
Affiliation(s)
- Karen A Fortner
- Department of Medicine, The University of Vermont College of Medicine, Burlington, VT 05405-0068, USA.
| | - Jeffrey P Bond
- Department of Microbiology and Molecular Genetics, The University of Vermont College of Medicine, Burlington, VT 05405-0068, USA
| | - James W Austin
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jeremy M Boss
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ralph C Budd
- Department of Medicine, The University of Vermont College of Medicine, Burlington, VT 05405-0068, USA
| |
Collapse
|
23
|
Sage AP, Nus M, Bagchi Chakraborty J, Tsiantoulas D, Newland SA, Finigan AJ, Masters L, Binder CJ, Mallat Z. X-Box Binding Protein-1 Dependent Plasma Cell Responses Limit the Development of Atherosclerosis. Circ Res 2017; 121:270-281. [DOI: 10.1161/circresaha.117.310884] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 06/14/2017] [Accepted: 06/15/2017] [Indexed: 02/06/2023]
Abstract
Rationale:
Diverse B cell responses and functions may be involved in atherosclerosis. Protective antibody responses, such as those against oxidized lipid epitopes, are thought to mainly derive from T cell-independent innate B cell subsets. In contrast, both pathogenic and protective roles have been associated with T cell-dependent antibodies, and their importance in both humans and mouse models is still unclear.
Objective:
To specifically target antibody production by plasma cells and determine the impact on atherosclerotic plaque development in mice with and without CD4+ T cells.
Methods and Results:
We combined a model of specific antibody deficiency, B cell-specific CD79a-
Cre
x XBP1 (X-box binding protein-1) floxed mice (XBP1-conditional knockout), with antibody-mediated depletion of CD4+ T cells. Ldlr knockout mice transplanted with XBP1-conditional knockout (or wild-type control littermate) bone marrow were fed western diet for 8 weeks with or without anti-CD4 depletion. All groups had similar levels of serum cholesterol. In Ldlr/XBP1-conditional knockout mice, serum levels of IgG, IgE, and IgM were significantly attenuated, and local antibody deposition in atherosclerotic plaque was absent. Antibody deficiency significantly accelerated atherosclerosis at both the aortic root and aortic arch. T cell and monocyte responses were not modulated, but necrotic core size was greater, even when adjusting for plaque size, and collagen deposition significantly lower. Anti-CD4 depletion in Ldlr/wild-type mice led to a decrease of serum IgG1 and IgG2c but not IgG3, as well as decreased IgM, associated with increased atherosclerosis and necrotic cores, and a decrease in plaque collagen. The combination of antibody deficiency and anti-CD4 depletion has no additive effects on aortic root atherosclerosis.
Conclusions:
The endogenous T cell-dependent humoral response can be protective. This has important implications for novel vaccine strategies for atherosclerosis and in understanding the impacts of immunotherapies used in patients at high risk for cardiovascular disease.
Collapse
Affiliation(s)
- Andrew P. Sage
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Meritxell Nus
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Jayashree Bagchi Chakraborty
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Dimitrios Tsiantoulas
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Stephen A. Newland
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Alison J. Finigan
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Leanne Masters
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Christoph J. Binder
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Ziad Mallat
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| |
Collapse
|
24
|
Sage AP, Mallat Z. Readapting the adaptive immune response - therapeutic strategies for atherosclerosis. Br J Pharmacol 2017; 174:3926-3939. [PMID: 28052311 DOI: 10.1111/bph.13700] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/13/2016] [Accepted: 12/20/2016] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases remain a major global health issue, with the development of atherosclerosis as a major underlying cause. Our treatment of cardiovascular disease has improved greatly over the past three decades, but much remains to be done reduce disease burden. Current priorities include reducing atherosclerosis advancement to clinically significant stages and preventing plaque rupture or erosion. Inflammation and involvement of the adaptive immune system influences all these aspects and therefore is one focus for future therapeutic development. The atherosclerotic vascular wall is now recognized to be invaded from both sides (arterial lumen and adventitia), for better or worse, by the adaptive immune system. Atherosclerosis is also affected at several stages by adaptive immune responses, overall providing many opportunities to target these responses and to reduce disease progression. Protective influences that may be defective in diseased individuals include humoral responses to modified LDL and regulatory T cell responses. There are many strategies in development to boost these pathways in humans, including vaccine-based therapies. The effects of various existing adaptive immune targeting therapies, such as blocking critical co-stimulatory pathways or B cell depletion, on cardiovascular disease are beginning to emerge with important consequences for both autoimmune disease patients and the potential for wider use of such therapies. Entering the translation phase for adaptive immune targeting therapies is an exciting and promising prospect. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- Andrew P Sage
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK.,INSERM U970, Paris Cardiovascular Research Center, Paris, France, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
25
|
Biswas M, Rogers GL, Sherman A, Byrne BJ, Markusic DM, Jiang H, Herzog RW. Combination therapy for inhibitor reversal in haemophilia A using monoclonal anti-CD20 and rapamycin. Thromb Haemost 2016; 117:33-43. [PMID: 27683758 DOI: 10.1160/th16-05-0404] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 09/12/2016] [Indexed: 01/19/2023]
Abstract
Development of antibodies (inhibitors) against coagulation factor VIII (FVIII) is a major complication of intravenous replacement therapy in haemophilia A (HA). Current immune tolerance induction (ITI) regimens are not universally effective. Rituximab, a B cell-depleting antibody against CD20, has shown mixed results for inhibitor reversal in patients. This study aims to develop a combinatorial therapy for inhibitor reversal in HA, using anti-murine CD20 (anti-mCD20) antibody and rapamycin, which targets both B and T cell responses. Additionally, it extensively characterises the role of the IgG backbone in B cell depletion by anti-CD20 antibodies. For this, inhibitors were generated in BALB/c-HA mice by weekly IV injection of FVIII. Subsequently, anti-mCD20 (18B12) with IgG2a or IgG1 backbone was injected IV in two doses three weeks apart and B cell depletion and recovery was characterised. Rapamycin was administered orally 3x/week (for 1 month) while continuing FVIII injections. Altering the IgG backbone of anti-mCD20 from IgG2a to IgG1 reduced overall depletion of B cells (including memory B cells), and marginal zone, B-10, and B-1b cells were specifically unaffected. While neither antibody was effective alone, in combination with rapamycin, anti-mCD20 IgG2a but not IgG1 was able to reverse inhibitors in HA mice. This regimen was particularly effective for starting titres of ~10 BU. Although IgG1 anti-mCD20 spared potentially tolerogenic B cell subsets, IgG2a directed sustained hyporesponsiveness when administered in conjunction with rapamycin. This regimen represents a promising treatment for inhibitor reversal in HA, as both of these compounds have been extensively used in human patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Roland W Herzog
- Roland W. Herzog, PhD, University of Florida, Cancer and Genetics Research Complex, 2033 Mowry Road, Gainesville, FL 32610, USA, Tel.: +1 352 273 8113, Fax: +1 352 273 8342, E-mail:
| |
Collapse
|
26
|
Schaheen B, Downs EA, Serbulea V, Almenara CCP, Spinosa M, Su G, Zhao Y, Srikakulapu P, Butts C, McNamara CA, Leitinger N, Upchurch GR, Meher AK, Ailawadi G. B-Cell Depletion Promotes Aortic Infiltration of Immunosuppressive Cells and Is Protective of Experimental Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2016; 36:2191-2202. [PMID: 27634836 DOI: 10.1161/atvbaha.116.307559] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 09/02/2016] [Indexed: 01/09/2023]
Abstract
OBJECTIVE B-cell depletion therapy is widely used for treatment of cancers and autoimmune diseases. B cells are abundant in abdominal aortic aneurysms (AAA); however, it is unknown whether B-cell depletion therapy affects AAA growth. Using experimental models of murine AAA, we aim to examine the effect of B-cell depletion on AAA formation. APPROACH AND RESULTS Wild-type or apolipoprotein E-knockout mice were treated with mouse monoclonal anti-CD20 or control antibodies and subjected to an elastase perfusion or angiotensin II infusion model to induce AAA, respectively. Anti-CD20 antibody treatment significantly depleted B1 and B2 cells, and strikingly suppressed AAA growth in both models. B-cell depletion resulted in lower circulating IgM levels, but did not affect the levels of IgG or cytokine/chemokine levels. Although the total number of leukocyte remained unchanged in elastase-perfused aortas after anti-CD20 antibody treatment, the number of B-cell subtypes was significantly lower. Interestingly, plasmacytoid dendritic cells expressing the immunomodulatory enzyme indole 2,3-dioxygenase were detected in the aortas of B-cell-depleted mice. In accordance with an increase in indole 2,3-dioxygenase+ plasmacytoid dendritic cells, the number of regulatory T cells was higher, whereas the expression of proinflammatory genes was lower in aortas of B-cell-depleted mice. In a coculture model, the presence of B cells significantly lowered the number of indole 2,3-dioxygenase+ plasmacytoid dendritic cells without affecting total plasmacytoid dendritic cell number. CONCLUSIONS The present results demonstrate that B-cell depletion protects mice from experimental AAA formation and promotes emergence of an immunosuppressive environment in aorta.
Collapse
Affiliation(s)
- Basil Schaheen
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Emily A Downs
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Vlad Serbulea
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Camila C P Almenara
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Michael Spinosa
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Gang Su
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Yunge Zhao
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Prasad Srikakulapu
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Cherié Butts
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Coleen A McNamara
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Norbert Leitinger
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Gilbert R Upchurch
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Akshaya K Meher
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville.
| | - Gorav Ailawadi
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| |
Collapse
|
27
|
Durali D, de Goër de Herve MG, Gasnault J, Taoufik Y. B cells and progressive multifocal leukoencephalopathy: search for the missing link. Front Immunol 2015; 6:241. [PMID: 26042124 PMCID: PMC4437032 DOI: 10.3389/fimmu.2015.00241] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 05/05/2015] [Indexed: 12/23/2022] Open
Abstract
Progressive multifocal leukoencephalopathy (PML) is a deadly demyelinating disease due to JC virus (JCV) replication in the brain. PML classically occurs in patients with severe immunodepression, and cases have recently been linked to therapeutic monoclonal antibodies such as natalizumab and also rituximab, which depletes B cells. B cells appear to play a complex role in the pathogenesis of PML. They may act as a viral reservoir and as a vector for viral dissemination in the central nervous system. Anti-JCV antibody responses appear to have a limited effect on JCV replication in the brain. However, accumulating evidence suggests that B cells may considerably influence T cell responses through their cytokine secretion. This immunomodulatory function of B cells may play an important role in the control of JCV infection and in the pathogenesis of PML, including rituximab-induced PML.
Collapse
Affiliation(s)
- Deniz Durali
- Immunology Research Laboratory, Department of Medical Microbiology, School of Medicine, Istanbul Medipol University , Istanbul , Turkey
| | | | - Jacques Gasnault
- IMVA-INSERM U1184, Department of Immunology, Bicetre Hospital, University Paris-sud , Le Kremlin-Bicêtre , France
| | - Yassine Taoufik
- IMVA-INSERM U1184, Department of Immunology, Bicetre Hospital, University Paris-sud , Le Kremlin-Bicêtre , France
| |
Collapse
|