1
|
Bulut O, Kilic G, Debisarun PA, Röring RJ, Sun S, Kolkman M, van Rijssen E, Ten Oever J, Koenen H, Barreiro L, Domínguez-Andrés J, Netea MG. Alendronate modulates cytokine responses in healthy young individuals after BCG vaccination. Immunol Lett 2024; 267:106851. [PMID: 38479480 DOI: 10.1016/j.imlet.2024.106851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/28/2024] [Accepted: 03/10/2024] [Indexed: 05/31/2024]
Abstract
Bacillus Calmette-Guérin (BCG) vaccination induces memory characteristics in innate immune cells and their progenitors, a process called trained immunity mediated by epigenetic and metabolic reprogramming. Cholesterol synthesis plays an amplifying role in trained immunity through mevalonate release. Nitrogen-containing bisphosphonates (N-BPs), such as alendronate, can inhibit cholesterol synthesis. We explored their effects on trained immunity induced by BCG in a placebo-controlled clinical study (NL74082.091.20) in young, healthy individuals. Participants receiving single-dose oral alendronate on the day of BCG vaccination had more neutrophils and plasma cells one month after treatment. Alendronate led to reduced proinflammatory cytokine production by PBMCs stimulated with heterologous bacterial and viral stimuli one month later. Furthermore, the addition of alendronate transcriptionally suppressed multiple immune response pathways in PBMCs upon stimulation. Our findings indicate that N-BPs modulate the long-lasting effects of BCG vaccination on the cytokine production capacity of innate immune cells.
Collapse
Affiliation(s)
- Ozlem Bulut
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gizem Kilic
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Priya A Debisarun
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rutger Jan Röring
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sarah Sun
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Medical Scientist Training Program, University of Chicago, Chicago, IL, USA
| | - Manon Kolkman
- Department of Laboratory Medicine, Laboratory for Medical Immunology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Esther van Rijssen
- Department of Laboratory Medicine, Laboratory for Medical Immunology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jaap Ten Oever
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hans Koenen
- Department of Laboratory Medicine, Laboratory for Medical Immunology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Luis Barreiro
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Jorge Domínguez-Andrés
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands; Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany.
| |
Collapse
|
2
|
Mooney AH, Draper SL, Burn OK, Anderson RJ, Compton BJ, Tang C, Farrand KJ, Di Lucia P, Ravà M, Fumagalli V, Giustini L, Bono E, Godfrey DI, Heath WR, Yuan W, Chisari FV, Guidotti LG, Iannacone M, Sidney J, Sette A, Gulab SA, Painter GF, Hermans IF. Preclinical evaluation of therapeutic vaccines for chronic hepatitis B that stimulate antiviral activities of T cells and NKT cells. JHEP Rep 2024; 6:101038. [PMID: 38694959 PMCID: PMC11061331 DOI: 10.1016/j.jhepr.2024.101038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 05/04/2024] Open
Abstract
Background & Aims Liver diseases resulting from chronic HBV infection are a significant cause of morbidity and mortality. Vaccines that elicit T-cell responses capable of controlling the virus represent a treatment strategy with potential for long-term effects. Here, we evaluated vaccines that induce the activity of type I natural killer T (NKT) cells to limit viral replication and license stimulation of conventional antiviral T-cells. Methods Vaccines were prepared by conjugating peptide epitopes to an NKT-cell agonist to promote co-delivery to antigen-presenting cells, encouraging NKT-cell licensing and stimulation of T cells. Activity of the conjugate vaccines was assessed in transgenic mice expressing the complete HBV genome, administered intravenously to maximise access to NKT cell-rich tissues. Results The vaccines induced only limited antiviral activity in unmanipulated transgenic hosts, likely attributable to NKT-cell activation as T-cell tolerance to viral antigens is strong. However, in a model of chronic hepatitis B involving transfer of naive HBcAg-specific CD8+ T cells into the transgenic mice, which typically results in specific T-cell dysfunction without virus control, vaccines containing the targeted HBcAg epitope induced prolonged antiviral activity because of qualitatively improved T-cell stimulation. In a step towards a clinical product, vaccines were prepared using synthetic long peptides covering clusters of known HLA-binding epitopes and shown to be immunogenic in HLA transgenic mice. Predictions based on HLA distribution suggest a product containing three selected SLP-based vaccines could give >90 % worldwide coverage, with an average of 3.38 epitopes targeted per individual. Conclusions The novel vaccines described show promise for further clinical development as a treatment for chronic hepatitis B. Impact and Implications Although there are effective prophylactic vaccines for HBV infection, it is estimated that 350-400 million people worldwide have chronic hepatitis B, putting these individuals at significant risk of life-threatening liver diseases. Therapeutic vaccination aimed at activating or boosting HBV-specific T-cell responses holds potential as a strategy for treating chronic infection, but has so far met with limited success. Here, we show that a glycolipid-peptide conjugate vaccine designed to coordinate activity of type I NKT cells alongside conventional antiviral T cells has antiviral activity in a mouse model of chronic infection. It is anticipated that a product based on a combination of three such conjugates, each prepared using long peptides covering clusters of known HLA-binding epitopes, could be developed further as a treatment for chronic hepatitis B with broad global HLA coverage.
Collapse
Affiliation(s)
- Anna H. Mooney
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Sarah L. Draper
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
| | - Olivia K. Burn
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Regan J. Anderson
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
| | - Benjamin J. Compton
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
| | - Chingwen Tang
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | | | - Pietro Di Lucia
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Micol Ravà
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Valeria Fumagalli
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Leonardo Giustini
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisa Bono
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Dale I. Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Australia
| | - William R. Heath
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia
| | - Weiming Yuan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Francis V. Chisari
- Department of Immunology & Microbial Sciences, The Scripps Research Institute, La Jolla, CA, USA
| | - Luca G. Guidotti
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - John Sidney
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Shivali A. Gulab
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
- Avalia Immunotherapies Limited, Wellington, New Zealand
| | - Gavin F. Painter
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
| | - Ian F. Hermans
- Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
3
|
Zareein A, Mahmoudi M, Jadhav SS, Wilmore J, Wu Y. Biomaterial engineering strategies for B cell immunity modulations. Biomater Sci 2024; 12:1981-2006. [PMID: 38456305 PMCID: PMC11019864 DOI: 10.1039/d3bm01841e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 02/23/2024] [Indexed: 03/09/2024]
Abstract
B cell immunity has a penetrating effect on human health and diseases. Therapeutics aiming to modulate B cell immunity have achieved remarkable success in combating infections, autoimmunity, and malignancies. However, current treatments still face significant limitations in generating effective long-lasting therapeutic B cell responses for many conditions. As the understanding of B cell biology has deepened in recent years, clearer regulation networks for B cell differentiation and antibody production have emerged, presenting opportunities to overcome current difficulties and realize the full therapeutic potential of B cell immunity. Biomaterial platforms have been developed to leverage these emerging concepts to augment therapeutic humoral immunity by facilitating immunogenic reagent trafficking, regulating T cell responses, and modulating the immune microenvironment. Moreover, biomaterial engineering tools have also advanced our understanding of B cell biology, further expediting the development of novel therapeutics. In this review, we will introduce the general concept of B cell immunobiology and highlight key biomaterial engineering strategies in the areas including B cell targeted antigen delivery, sustained B cell antigen delivery, antigen engineering, T cell help optimization, and B cell suppression. We will also discuss our perspective on future biomaterial engineering opportunities to leverage humoral immunity for therapeutics.
Collapse
Affiliation(s)
- Ali Zareein
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Mina Mahmoudi
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Shruti Sunil Jadhav
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
| | - Joel Wilmore
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Yaoying Wu
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
4
|
Aguilar CC, Kalia A, Brisse ME, Dowd KA, Wise-Dent O, Burgomaster KE, Droppo J, Pierson TC, Hickman HD. Subcapsular sinus macrophages maximize germinal center development in non-draining lymph nodes during blood-borne viral infection. Sci Immunol 2024; 9:eadi4926. [PMID: 38457515 DOI: 10.1126/sciimmunol.adi4926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 01/29/2024] [Indexed: 03/10/2024]
Abstract
Lymph node (LN) germinal centers (GCs) are critical sites for B cell activation and differentiation. GCs develop after specialized CD169+ macrophages residing in LN sinuses filter antigens (Ags) from the lymph and relay these Ags into proximal B cell follicles. Many viruses, however, first reach LNs through the blood during viremia (virus in the blood), rather than through lymph drainage from infected tissue. How LNs capture viral Ag from the blood to allow GC development is not known. Here, we followed Zika virus (ZIKV) dissemination in mice and subsequent GC formation in both infected tissue-draining and non-draining LNs. From the footpad, ZIKV initially disseminated through two LN chains, infecting LN macrophages and leading to GC formation. Despite rapid ZIKV viremia, non-draining LNs were not infected for several days. Non-draining LN infection correlated with virus-induced vascular leakage and neutralization of permeability reduced LN macrophage attrition. Depletion of non-draining LN macrophages significantly decreased GC B cells in these nodes. Thus, although LNs inefficiently captured viral Ag directly from the blood, GC formation in non-draining LNs proceeded similarly to draining LNs through LN sinus CD169+ macrophages. Together, our findings reveal a conserved pathway allowing LN macrophages to activate antiviral B cells in LNs distal from infected tissue after blood-borne viral infection.
Collapse
Affiliation(s)
- Cynthia C Aguilar
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Anurag Kalia
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Morgan E Brisse
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kimberly A Dowd
- Arbovirus Immunity Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Olivia Wise-Dent
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Katherine E Burgomaster
- Arbovirus Immunity Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Joanna Droppo
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Theodore C Pierson
- Arbovirus Immunity Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Heather D Hickman
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
5
|
Zhang X, Wei M, Zhang Z, Zeng Y, Zou F, Zhang S, Wang Z, Chen F, Xiong H, Li Y, Zhou L, Li T, Zheng Q, Yu H, Zhang J, Gu Y, Zhao Q, Li S, Xia N. Risedronate-functionalized manganese-hydroxyapatite amorphous particles: A potent adjuvant for subunit vaccines and cancer immunotherapy. J Control Release 2024; 367:13-26. [PMID: 38244843 DOI: 10.1016/j.jconrel.2024.01.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 01/22/2024]
Abstract
The cGAS-STING pathway and the Mevalonate Pathway are druggable targets for vaccine adjuvant discovery. Manganese (Mn) and bisphosphonates are known to exert adjuvant effects by targeting these two pathways, respectively. This study found the synergistic potential of the two pathways in enhancing immune response. Risedronate (Ris) significantly amplified the Mn adjuvant early antibody response by 166-fold and fortified its cellular immunity. However, direct combination of Mn2+ and Ris resulted in increased adjuvant toxicity (40% mouse mortality). By the combination of doping property of hydroxyapatite (HA) and its high affinity for Ris, we designed Ris-functionalized Mn-HA micro-nanoparticles as an organic-inorganic hybrid adjuvant, named MnHARis. MnHARis alleviated adjuvant toxicity (100% vs. 60% survival rate) and exhibited good long-term stability. When formulated with the varicella-zoster virus glycoprotein E (gE) antigen, MnHARis triggered a 274.3-fold increase in IgG titers and a 61.3-fold surge in neutralization titers while maintaining a better long-term humoral immunity compared to the aluminum adjuvant. Its efficacy spanned other antigens, including ovalbumin, HPV18 VLP, and SARS-CoV-2 spike protein. Notably, the cellular immunity elicited by the group of gE + MnHARis was comparable to the renowned Shingrix®. Moreover, intratumoral co-administration with an anti-trophoblast cell surface antigen 2 nanobody revealed synergistic antitumor capabilities. These findings underscore the potential of MnHARis as a potent adjuvant for augmenting vaccine immune responses and improving cancer immunotherapy outcomes.
Collapse
Affiliation(s)
- Xiuli Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Mingjing Wei
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Zhigang Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Yarong Zeng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Feihong Zou
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Sibo Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Zhiping Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Fentian Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Hualong Xiong
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Yufang Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Lizhi Zhou
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Tingting Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Qingbing Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Hai Yu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Jun Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Ying Gu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Qinjian Zhao
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China.
| | - Shaowei Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China.
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
6
|
Fumagalli V, Iannacone M. The interplay of drug therapeutics and immune responses to SARS-CoV-2. Cell Mol Immunol 2024; 21:197-200. [PMID: 37964122 PMCID: PMC10805708 DOI: 10.1038/s41423-023-01098-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/22/2023] [Indexed: 11/16/2023] Open
Abstract
The SARS-CoV-2 pandemic has necessitated rapid therapeutic and preventative responses. While vaccines form the frontline of defense, antiviral treatments such as nirmatrelvir have emerged as vital adjunctive measures, particularly for those unable or unwilling to be vaccinated. This review delves into the potential influence of nirmatrelvir on enduring immunity. In parallel, the potential of drug repurposing is explored, with bisphosphonates being examined for their possible effects against COVID-19 due to their immunomodulatory properties. The importance of rigorous clinical trials and careful interpretation of preliminary data is emphasized.
Collapse
Affiliation(s)
- Valeria Fumagalli
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Matteo Iannacone
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
- Experimental Imaging Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
7
|
Mazzarello AN, Gugiatti E, Cossu V, Bertola N, Bagnara D, Carta S, Ravera S, Salvetti C, Ibatici A, Ghiotto F, Colombo M, Cutrona G, Marini C, Sambuceti G, Fais F, Bruno S. Unexpected chronic lymphocytic leukemia B cell activation by bisphosphonates. Cancer Immunol Immunother 2024; 73:27. [PMID: 38280019 PMCID: PMC10821833 DOI: 10.1007/s00262-023-03588-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/25/2023] [Indexed: 01/29/2024]
Abstract
Chronic lymphocytic leukemia (CLL) is a disease of the elderly, often presenting comorbidities like osteoporosis and requiring, in a relevant proportion of cases, treatment with bisphosphonates (BPs). This class of drugs was shown in preclinical investigations to also possess anticancer properties. We started an in vitro study of the effects of BPs on CLL B cells activated by microenvironment-mimicking stimuli and observed that, depending on drug concentration, hormetic effects were induced on the leukemic cells. Higher doses induced cytotoxicity whereas at lower concentrations, more likely occurring in vivo, the drugs generated a protective effect from spontaneous and chemotherapy-induced apoptosis, and augmented CLL B cell activation/proliferation. This CLL-activation effect promoted by the BPs was associated with markers of poor CLL prognosis and required the presence of bystander stromal cells. Functional experiments suggested that this phenomenon involves the release of soluble factors and is increased by cellular contact between stroma and CLL B cells. Since CLL patients often present comorbidities such as osteoporosis and considering the diverse outcomes in both CLL disease progression and CLL response to treatment among patients, illustrating this phenomenon holds potential significance in driving additional investigations.
Collapse
Affiliation(s)
- Andrea N Mazzarello
- Department of Experimental Medicine (DIMES), University of Genoa, Via De Toni 14, 16132, Genoa, Italy
| | - Elena Gugiatti
- Department of Experimental Medicine (DIMES), University of Genoa, Via De Toni 14, 16132, Genoa, Italy
| | - Vanessa Cossu
- Department of Experimental Medicine (DIMES), University of Genoa, Via De Toni 14, 16132, Genoa, Italy
| | - Nadia Bertola
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Davide Bagnara
- Department of Experimental Medicine (DIMES), University of Genoa, Via De Toni 14, 16132, Genoa, Italy
| | - Sonia Carta
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Silvia Ravera
- Department of Experimental Medicine (DIMES), University of Genoa, Via De Toni 14, 16132, Genoa, Italy
| | - Chiara Salvetti
- Clinic of Hematology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Adalberto Ibatici
- Division of Hematology and Bone Marrow Transplant, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Fabio Ghiotto
- Department of Experimental Medicine (DIMES), University of Genoa, Via De Toni 14, 16132, Genoa, Italy
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Monica Colombo
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Giovanna Cutrona
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Cecilia Marini
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Milan, Italy
| | - Gianmario Sambuceti
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Franco Fais
- Department of Experimental Medicine (DIMES), University of Genoa, Via De Toni 14, 16132, Genoa, Italy
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Silvia Bruno
- Department of Experimental Medicine (DIMES), University of Genoa, Via De Toni 14, 16132, Genoa, Italy.
| |
Collapse
|
8
|
Nie M, Wu S, Chen Y, Wu Y, Chen R, Liu Y, Yue M, Jiang Y, Qiu D, Yang M, Wang Z, Gao J, Xiong H, Qi R, He J, Zhang J, Zhang L, Wang Y, Fang M, Que Y, Yao Y, Li S, Zhang J, Zhao Q, Yuan Q, Zhang T, Xia N. Micronanoparticled risedronate exhibits potent vaccine adjuvant effects. J Control Release 2024; 365:369-383. [PMID: 37972764 DOI: 10.1016/j.jconrel.2023.11.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/26/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
Micro/Nano-scale particles are widely used as vaccine adjuvants to enhance immune response and improve antigen stability. While aluminum salt is one of the most common adjuvants approved for human use, its immunostimulatory capacity is suboptimal. In this study, we modified risedronate, an immunostimulant and anti-osteoporotic drug, to create zinc salt particle-based risedronate (Zn-RS), also termed particulate risedronate. Compared to soluble risedronate, micronanoparticled Zn-RS adjuvant demonstrated increased recruitment of innate cells, enhanced antigen uptake locally, and a similar antigen depot effect as aluminum salt. Furthermore, Zn-RS adjuvant directly and quickly stimulated immune cells, accelerated the formulation of germinal centers in lymph nodes, and facilitated the rapid production of antibodies. Importantly, Zn-RS adjuvant exhibited superior performance in both young and aged mice, effectively protecting against respiratory diseases such as SARS-CoV-2 challenge. Consequently, particulate risedronate showed great potential as an immune-enhancing vaccine adjuvant, particularly beneficial for vaccines targeting the susceptible elderly.
Collapse
Affiliation(s)
- Meifeng Nie
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Shuyu Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Yiyi Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Yangtao Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Ruitong Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Yue Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Mingxi Yue
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Yao Jiang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Dekui Qiu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Man Yang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Zikang Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Jiahua Gao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Hualong Xiong
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Ruoyao Qi
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Jinhang He
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Jinlei Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Liang Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Yingbin Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Mujin Fang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Yuqiong Que
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Youliang Yao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Shaowei Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China.
| | - Jun Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China.
| | - Qinjian Zhao
- College of Pharmacy, Chongqing Medical University, Chongqing, Chongqing 400016, China.
| | - Quan Yuan
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China.
| | - Tianying Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China.
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
9
|
Thompson J, Wang Y, Dreischulte T, Barreiro O, Gonzalez RJ, Hanč P, Matysiak C, Neely HR, Rottenkolber M, Haskell T, Endres S, von Andrian UH. Association between bisphosphonate use and COVID-19 related outcomes. eLife 2023; 12:e79548. [PMID: 37534876 PMCID: PMC10691801 DOI: 10.7554/elife.79548] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 06/28/2023] [Indexed: 08/04/2023] Open
Abstract
Background Although there are several efficacious vaccines against COVID-19, vaccination rates in many regions around the world remain insufficient to prevent continued high disease burden and emergence of viral variants. Repurposing of existing therapeutics that prevent or mitigate severe COVID-19 could help to address these challenges. The objective of this study was to determine whether prior use of bisphosphonates is associated with reduced incidence and/or severity of COVID-19. Methods A retrospective cohort study utilizing payer-complete health insurance claims data from 8,239,790 patients with continuous medical and prescription insurance January 1, 2019 to June 30, 2020 was performed. The primary exposure of interest was use of any bisphosphonate from January 1, 2019 to February 29, 2020. Bisphosphonate users were identified as patients having at least one bisphosphonate claim during this period, who were then 1:1 propensity score-matched to bisphosphonate non-users by age, gender, insurance type, primary-care-provider visit in 2019, and comorbidity burden. Main outcomes of interest included: (a) any testing for SARS-CoV-2 infection; (b) COVID-19 diagnosis; and (c) hospitalization with a COVID-19 diagnosis between March 1, 2020 and June 30, 2020. Multiple sensitivity analyses were also performed to assess core study outcomes amongst more restrictive matches between BP users/non-users, as well as assessing the relationship between BP-use and other respiratory infections (pneumonia, acute bronchitis) both during the same study period as well as before the COVID outbreak. Results A total of 7,906,603 patients for whom continuous medical and prescription insurance information was available were selected. A total of 450,366 bisphosphonate users were identified and 1:1 propensity score-matched to bisphosphonate non-users. Bisphosphonate users had lower odds ratios (OR) of testing for SARS-CoV-2 infection (OR = 0.22; 95%CI:0.21-0.23; p<0.001), COVID-19 diagnosis (OR = 0.23; 95%CI:0.22-0.24; p<0.001), and COVID-19-related hospitalization (OR = 0.26; 95%CI:0.24-0.29; p<0.001). Sensitivity analyses yielded results consistent with the primary analysis. Bisphosphonate-use was also associated with decreased odds of acute bronchitis (OR = 0.23; 95%CI:0.22-0.23; p<0.001) or pneumonia (OR = 0.32; 95%CI:0.31-0.34; p<0.001) in 2019, suggesting that bisphosphonates may protect against respiratory infections by a variety of pathogens, including but not limited to SARS-CoV-2. Conclusions Prior bisphosphonate-use was associated with dramatically reduced odds of SARS-CoV-2 testing, COVID-19 diagnosis, and COVID-19-related hospitalizations. Prospective clinical trials will be required to establish a causal role for bisphosphonate-use in COVID-19-related outcomes. Funding This study was supported by NIH grants, AR068383 and AI155865, a grant from MassCPR (to UHvA) and a CRI Irvington postdoctoral fellowship, CRI2453 (to PH).
Collapse
Affiliation(s)
| | - Yidi Wang
- Dept. of Immunology, Harvard Medical SchoolBostonUnited States
| | - Tobias Dreischulte
- Institute of General Practice and Family Medicine, University Hospital of Ludwig Maximilians-University MunichMunichGermany
| | - Olga Barreiro
- Dept. of Immunology, Harvard Medical SchoolBostonUnited States
| | | | - Pavel Hanč
- Dept. of Immunology, Harvard Medical SchoolBostonUnited States
| | | | - Harold R Neely
- Dept. of Immunology, Harvard Medical SchoolBostonUnited States
| | - Marietta Rottenkolber
- Institute of General Practice and Family Medicine, University Hospital of Ludwig Maximilians-University MunichMunichGermany
| | | | - Stefan Endres
- Center of Integrated Protein Science Munich and Division of Clinical Pharmacology, University Hospital, LMU Munich, GermanyMunichGermany
| | | |
Collapse
|
10
|
Petruk N, Siddiqui A, Tadayon S, Määttä J, Mattila PK, Jukkola A, Sandholm J, Selander KS. CD73 regulates zoledronate-induced lymphocyte infiltration in triple-negative breast cancer tumors and lung metastases. Front Immunol 2023; 14:1179022. [PMID: 37533856 PMCID: PMC10390692 DOI: 10.3389/fimmu.2023.1179022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/23/2023] [Indexed: 08/04/2023] Open
Abstract
Introduction Bisphosphonates (BPs) are bone-protecting osteoclast inhibitors, typically used in the treatment of osteoporosis and skeletal complications of malignancies. When given in the adjuvant setting, these drugs may also prevent relapses and prolong overall survival in early breast cancer (EBC), specifically among postmenopausal patients. Because of these findings, adjuvant nitrogen-containing BPs (N-BPs), such as zoledronate (ZOL), are now the standard of care for high-risk EBC patients, but there are no benefit-associated biomarkers, and the efficacy remains low. BPs have been demonstrated to possess anti-tumor activities, but the mechanisms by which they provide the beneficial effects in EBC are not known. Methods We used stably transfected 4T1 breast cancer cells together with suppression of CD73 (sh-CD73) or control cells (sh-NT). We compared ZOL effects on tumor growth and infiltrating lymphocytes (TILs) into tumors and lung metastases using two mouse models. B cell depletion was performed using anti-CD20 antibody. Results Sh-CD73 4T1 cells were significantly more sensitive to the growth inhibitory effects of n-BPs in vitro. However, while ZOL-induced growth inhibition was similar between the tumor groups in vivo, ZOL enhanced B and T lymphocyte infiltration into the orthotopic tumors with down-regulated CD73. A similar trend was detected in lung metastases. ZOL-induced tumor growth inhibition was found to be augmented with B cell depletion in sh-NT tumors, but not in sh-CD73 tumors. As an internal control, ZOL effects on bone were similar in mice bearing both tumor groups. Discussion Taken together, these results indicate that ZOL modifies TILs in breast cancer, both in primary tumors and metastases. Our results further demonstrate that B cells may counteract the growth inhibitory effects of ZOL. However, all ZOL-induced TIL effects may be influenced by immunomodulatory characteristics of the tumor.
Collapse
Affiliation(s)
- Nataliia Petruk
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Arafat Siddiqui
- Institute of Biomedicine, University of Turku, Turku, Finland
- Western Cancer Centre FICAN West, Turku, Finland
| | - Sina Tadayon
- MediCity Research Laboratory, University of Turku, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Jorma Määttä
- Institute of Biomedicine, University of Turku, Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Pieta K. Mattila
- Institute of Biomedicine, University of Turku, Turku, Finland
- MediCity Research Laboratory, University of Turku, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Arja Jukkola
- Department of Oncology, Tampere University Hospital, Tays Cancer Center, Tampere, Finland
| | - Jouko Sandholm
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Katri S. Selander
- Department of Oncology and Radiation Therapy, Oulu University Hospital, Oulu, Finland
- Cancer Research and Translational Medicine Research Unit, University of Oulu, Oulu, Finland
| |
Collapse
|
11
|
Efficacy of mucosal vaccination using a protozoan parasite as a vehicle for antigen delivery: IgG and neutralizing response after rectal administration of LeCoVax-2, a candidate vaccine against COVID-19. Pharmacol Res 2022; 186:106546. [PMCID: PMC9633108 DOI: 10.1016/j.phrs.2022.106546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/02/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
|
12
|
Zhou F, Zhang G, Wu Y, Xiong Y. Inflammasome Complexes: Crucial mediators in osteoimmunology and bone diseases. Int Immunopharmacol 2022; 110:109072. [DOI: 10.1016/j.intimp.2022.109072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/15/2022] [Accepted: 07/14/2022] [Indexed: 11/05/2022]
|
13
|
Zhang YN, Paynter J, Sou C, Fourfouris T, Wang Y, Abraham C, Ngo T, Zhang Y, He L, Zhu J. Mechanism of a COVID-19 nanoparticle vaccine candidate that elicits a broadly neutralizing antibody response to SARS-CoV-2 variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.03.26.437274. [PMID: 33791704 PMCID: PMC8010731 DOI: 10.1101/2021.03.26.437274] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Vaccines that induce potent neutralizing antibody (NAb) responses against emerging variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are essential for combating the coronavirus disease 2019 (COVID-19) pandemic. We demonstrated that mouse plasma induced by self-assembling protein nanoparticles (SApNPs) that present 20 rationally designed S2GΔHR2 spikes of the ancestral Wuhan-Hu-1 strain can neutralize the B.1.1.7, B.1.351, P.1, and B.1.617 variants with the same potency. The adjuvant effect on vaccine-induced immunity was investigated by testing 16 formulations for the multilayered I3-01v9 SApNP. Using single-cell sorting, monoclonal antibodies (mAbs) with diverse neutralization breadth and potency were isolated from mice immunized with the receptor binding domain (RBD), S2GΔHR2 spike, and SApNP vaccines. The mechanism of vaccine-induced immunity was examined in mice. Compared with the soluble spike, the I3-01v9 SApNP showed 6-fold longer retention, 4-fold greater presentation on follicular dendritic cell dendrites, and 5-fold stronger germinal center reactions in lymph node follicles.
Collapse
Affiliation(s)
- Yi-Nan Zhang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Jennifer Paynter
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Cindy Sou
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Tatiana Fourfouris
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Ying Wang
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, Pennsylvania 19140, USA
- Department of Microbiology and Immunology, Temple University, Philadelphia, Pennsylvania 19140, USA
| | - Ciril Abraham
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, Pennsylvania 19140, USA
| | - Timothy Ngo
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Yi Zhang
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, Pennsylvania 19140, USA
- Department of Microbiology and Immunology, Temple University, Philadelphia, Pennsylvania 19140, USA
| | - Linling He
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Jiang Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California 92037, USA
| |
Collapse
|
14
|
Wu D, Cline-Smith A, Shashkova E, Perla A, Katyal A, Aurora R. T-Cell Mediated Inflammation in Postmenopausal Osteoporosis. Front Immunol 2021; 12:687551. [PMID: 34276675 PMCID: PMC8278518 DOI: 10.3389/fimmu.2021.687551] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/04/2021] [Indexed: 12/11/2022] Open
Abstract
Osteoporosis is the most prevalent metabolic bone disease that affects half the women in the sixth and seventh decade of life. Osteoporosis is characterized by uncoupled bone resorption that leads to low bone mass, compromised microarchitecture and structural deterioration that increases the likelihood of fracture with minimal trauma, known as fragility fractures. Several factors contribute to osteoporosis in men and women. In women, menopause - the cessation of ovarian function, is one of the leading causes of primary osteoporosis. Over the past three decades there has been growing appreciation that the adaptive immune system plays a fundamental role in the development of postmenopausal osteoporosis, both in humans and in mouse models. In this review, we highlight recent data on the interactions between T cells and the skeletal system in the context of postmenopausal osteoporosis. Finally, we review recent studies on the interventions to ameliorate osteoporosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Rajeev Aurora
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
15
|
Zarubova J, Zhang X, Hoffman T, Hasani-Sadrabadi MM, Li S. Biomaterial-based immunoengineering to fight COVID-19 and infectious diseases. MATTER 2021; 4:1528-1554. [PMID: 33723531 PMCID: PMC7942141 DOI: 10.1016/j.matt.2021.02.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Infection by SARS-CoV-2 virus often induces the dysregulation of immune responses, tissue damage, and blood clotting. Engineered biomaterials from the nano- to the macroscale can provide targeted drug delivery, controlled drug release, local immunomodulation, enhanced immunity, and other desirable functions to coordinate appropriate immune responses and to repair tissues. Based on the understanding of COVID-19 disease progression and immune responses to SARS-CoV-2, we discuss possible immunotherapeutic strategies and highlight biomaterial approaches from the perspectives of preventive immunization, therapeutic immunomodulation, and tissue healing and regeneration. Successful development of biomaterial platforms for immunization and immunomodulation will not only benefit COVID-19 patients, but also have broad applications for a variety of infectious diseases.
Collapse
Affiliation(s)
- Jana Zarubova
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, CA 90095, USA
| | - Xuexiang Zhang
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, CA 90095, USA
| | - Tyler Hoffman
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, CA 90095, USA
| | - Mohammad Mahdi Hasani-Sadrabadi
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, CA 90095, USA
| | - Song Li
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, CA 90095, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
16
|
Proto MC, Fiore D, Piscopo C, Pagano C, Galgani M, Bruzzaniti S, Laezza C, Gazzerro P, Bifulco M. Lipid homeostasis and mevalonate pathway in COVID-19: Basic concepts and potential therapeutic targets. Prog Lipid Res 2021; 82:101099. [PMID: 33915202 PMCID: PMC8074527 DOI: 10.1016/j.plipres.2021.101099] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/21/2022]
Abstract
Despite encouraging progresses achieved in the management of viral diseases, efficient strategies to counteract infections are still required. The current global challenge highlighted the need to develop a rapid and cost-effective strategy to counteract the SARS-CoV-2 pandemic. Lipid metabolism plays a crucial role in viral infections. Viruses can use the host lipid machinery to support their life cycle and to impair the host immune response. The altered expression of mevalonate pathway-related genes, induced by several viruses, assures survival and spread in host tissue. In some infections, statins, HMG-CoA-reductase inhibitors, reduce cholesterol in the plasma membrane of permissive cells resulting in lower viral titers and failure to internalize the virus. Statins can also counteract viral infections through their immunomodulatory, anti-inflammatory and anti-thrombotic effects. Beyond statins, interfering with the mevalonate pathway could have an adjuvant effect in therapies aimed at mitigating endothelial dysfunction and deregulated inflammation in viral infection. In this review we depicted the historical and current evidence highlighting how lipid homeostasis and mevalonate pathway targeting represents a valid approach to rapidly neutralize viruses, focusing our attention to their potential use as effective targets to hinder SARS-CoV-2 morbidity and mortality. Pros and cons of statins and Mevalonate-pathway inhibitors have been also dissected.
Collapse
Affiliation(s)
- Maria Chiara Proto
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy
| | - Donatella Fiore
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy
| | - Chiara Piscopo
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy
| | - Cristina Pagano
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", 80131 Naples, Italy
| | - Mario Galgani
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", 80131 Naples, Italy; Institute of Endocrinology and Experimental Oncology, IEOS CNR, 80131 Naples, Italy
| | - Sara Bruzzaniti
- Institute of Endocrinology and Experimental Oncology, IEOS CNR, 80131 Naples, Italy; Department of Biology, University of Naples "Federico II", 80126 Naples, Italy
| | - Chiara Laezza
- Institute of Endocrinology and Experimental Oncology, IEOS CNR, 80131 Naples, Italy
| | - Patrizia Gazzerro
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy.
| | - Maurizio Bifulco
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", 80131 Naples, Italy.
| |
Collapse
|
17
|
Fumagalli V, Di Lucia P, Venzin V, Bono EB, Jordan R, Frey CR, Delaney W, Chisari FV, Guidotti LG, Iannacone M. Serum HBsAg clearance has minimal impact on CD8+ T cell responses in mouse models of HBV infection. J Exp Med 2021; 217:152002. [PMID: 32761167 PMCID: PMC7596822 DOI: 10.1084/jem.20200298] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/20/2020] [Accepted: 06/18/2020] [Indexed: 12/23/2022] Open
Abstract
Antibody-mediated clearance of hepatitis B surface antigen (HBsAg) from the circulation of chronically infected patients (i.e., seroconversion) is usually associated with increased HBV-specific T cell responsiveness. However, a causative link between serum HBsAg levels and impairment of intrahepatic CD8+ T cells has not been established. Here we addressed this issue by using HBV replication-competent transgenic mice that are depleted of circulating HBsAg, via either spontaneous seroconversion or therapeutic monoclonal antibodies, as recipients of HBV-specific CD8+ T cells. Surprisingly, we found that serum HBsAg clearance has only a minimal effect on the expansion of HBV-specific naive CD8+ T cells undergoing intrahepatic priming. It does not alter their propensity to become dysfunctional, nor does it enhance the capacity of IL-2–based immunotherapeutic strategies to increase their antiviral function. In summary, our results reveal that circulating HBsAg clearance does not improve HBV-specific CD8+ T cell responses in vivo and may have important implications for the treatment of chronic HBV infection.
Collapse
Affiliation(s)
- Valeria Fumagalli
- Division of Immunology, Transplantation and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Pietro Di Lucia
- Division of Immunology, Transplantation and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Valentina Venzin
- Division of Immunology, Transplantation and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Elisa B Bono
- Division of Immunology, Transplantation and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | | | | | | | - Francis V Chisari
- Deparment of Immunology and Microbial Sciences, The Scripps Research Institute, La Jolla, CA
| | - Luca G Guidotti
- Division of Immunology, Transplantation and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy.,Experimental Imaging Center, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
18
|
Zhang YN, Poon W, Sefton E, Chan WCW. Suppressing Subcapsular Sinus Macrophages Enhances Transport of Nanovaccines to Lymph Node Follicles for Robust Humoral Immunity. ACS NANO 2020; 14:9478-9490. [PMID: 32479046 DOI: 10.1021/acsnano.0c02240] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Nanovaccines need to be transported to lymph node follicles to induce humoral immunity and generate neutralizing antibodies. Here, we discovered that subcapsular sinus macrophages play a barrier role to prevent nanovaccines from accessing lymph node follicles. This is illustrated by measuring the humoral immune responses after removing or functionally altering these cells in the nanovaccine transport process. We achieved up to 60 times more antigen-specific antibody production after suppressing subcapsular sinus macrophages. The degree of the enhanced antibody production is dependent on the nanovaccine dose and size, formulation, and administration time. We further found that pharmacological agents that disrupt the macrophage uptake function can be considered as adjuvants in vaccine development. Immunizing mice using nanovaccines formulated with these agents can induce more than 30 times higher antigen-specific antibody production compared to nanovaccines alone. These findings suggest that altering transport barriers to enable more of the nanovaccine to be delivered to the lymph node follicles for neutralizing antibody production is an effective strategy to boost vaccination.
Collapse
Affiliation(s)
- Yi-Nan Zhang
- Institute of Biomaterials & Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Wilson Poon
- Institute of Biomaterials & Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Elana Sefton
- Institute of Biomaterials & Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Warren C W Chan
- Institute of Biomaterials & Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
- Department of Materials Science & Engineering, University of Toronto, Toronto, Ontario M5S 1A1, Canada
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada
| |
Collapse
|
19
|
Kim EH, Woodruff MC, Grigoryan L, Maier B, Lee SH, Mandal P, Cortese M, Natrajan MS, Ravindran R, Ma H, Merad M, Gitlin AD, Mocarski ES, Jacob J, Pulendran B. Squalene emulsion-based vaccine adjuvants stimulate CD8 T cell, but not antibody responses, through a RIPK3-dependent pathway. eLife 2020; 9:52687. [PMID: 32515732 PMCID: PMC7314549 DOI: 10.7554/elife.52687] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 06/08/2020] [Indexed: 12/26/2022] Open
Abstract
The squalene-based oil-in-water emulsion (SE) vaccine adjuvant MF59 has been administered to more than 100 million people in more than 30 countries, in both seasonal and pandemic influenza vaccines. Despite its wide use and efficacy, its mechanisms of action remain unclear. In this study we demonstrate that immunization of mice with MF59 or its mimetic AddaVax (AV) plus soluble antigen results in robust antigen-specific antibody and CD8 T cell responses in lymph nodes and non-lymphoid tissues. Immunization triggered rapid RIPK3-kinase dependent necroptosis in the lymph node which peaked at 6 hr, followed by a sequential wave of apoptosis. Immunization with alum plus antigen did not induce RIPK3-dependent signaling. RIPK3-dependent signaling induced by MF59 or AV was essential for cross-presentation of antigen to CD8 T cells by Batf3-dependent CD8+ DCs. Consistent with this, RIPK3 deficient or Batf3 deficient mice were impaired in their ability to mount adjuvant-enhanced CD8 T cell responses. However, CD8 T cell responses were unaffected in mice deficient in MLKL, a downstream mediator of necroptosis. Surprisingly, antibody responses were unaffected in RIPK3-kinase or Batf3 deficient mice. In contrast, antibody responses were impaired by in vivo administration of the pan-caspase inhibitor Z-VAD-FMK, but normal in caspase-1 deficient mice, suggesting a contribution from apoptotic caspases, in the induction of antibody responses. These results demonstrate that squalene emulsion-based vaccine adjuvants induce antigen-specific CD8 T cell and antibody responses, through RIPK3-dependent and-independent pathways, respectively.
Collapse
Affiliation(s)
- Eui Ho Kim
- Emory Vaccine Center, Emory University, Atlanta, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, United States.,Viral Immunology Laboratory, Institut Pasteur Korea, Seongnam, Republic of Korea
| | - Matthew C Woodruff
- Emory Vaccine Center, Emory University, Atlanta, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, United States
| | - Lilit Grigoryan
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, United States
| | - Barbara Maier
- Department of Oncological Sciences, Tisch Cancer Institute and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York City, United States
| | - Song Hee Lee
- Emory Vaccine Center, Emory University, Atlanta, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, United States
| | - Pratyusha Mandal
- Emory Vaccine Center, Emory University, Atlanta, United States.,Department of Microbiology and Immunology, Emory Vaccine Center, School of Medicine, Emory University, Atlanta, United States
| | - Mario Cortese
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, United States
| | | | - Rajesh Ravindran
- Emory Vaccine Center, Emory University, Atlanta, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, United States
| | - Huailiang Ma
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, United States
| | - Miriam Merad
- Department of Oncological Sciences, Tisch Cancer Institute and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York City, United States
| | - Alexander D Gitlin
- Department of Physiological Chemistry, Genentech, South San Francisco, United States.,Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, United States
| | - Edward S Mocarski
- Emory Vaccine Center, Emory University, Atlanta, United States.,Department of Microbiology and Immunology, Emory Vaccine Center, School of Medicine, Emory University, Atlanta, United States
| | - Joshy Jacob
- Emory Vaccine Center, Emory University, Atlanta, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, United States.,Department of Microbiology and Immunology, Emory Vaccine Center, School of Medicine, Emory University, Atlanta, United States
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, United States.,Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, United States.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, United States
| |
Collapse
|
20
|
Sala E, Kuka M. The Suppressive Attitude of Inflammatory Monocytes in Antiviral Antibody Responses. Viral Immunol 2020; 33:327-333. [PMID: 32027238 PMCID: PMC7247028 DOI: 10.1089/vim.2019.0132] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Inflammatory monocytes play important functions in antiviral immune responses, including release of inflammatory cytokines and antigen presentation to T lymphocytes. Depending on the pathological context, these functions might translate into beneficial or detrimental effects in the resolution of the disease. Recent literature has highlighted a role for inflammatory monocytes also in direct suppression of B cell responses. In this review, we will briefly discuss research showing the relationship between inflammatory monocytes and B lymphocytes, its functional consequences on antiviral antibody responses, and possible implications in the design of future vaccination strategies.
Collapse
Affiliation(s)
- Eleonora Sala
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Mirela Kuka
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
21
|
Spatiotemporal regulation of type I interferon expression determines the antiviral polarization of CD4 + T cells. Nat Immunol 2020; 21:321-330. [PMID: 32066949 PMCID: PMC7043938 DOI: 10.1038/s41590-020-0596-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 01/10/2020] [Indexed: 01/01/2023]
Abstract
Differentiation of CD4+ T cells into either follicular helper T (TFH) or type 1 helper T (TH1) cells influences the balance between humoral and cellular adaptive immunity, but the mechanisms whereby pathogens elicit distinct effector cells are incompletely understood. Here, we analyzed the spatiotemporal dynamics of CD4+ T cells during infection with recombinant vesicular stomatitis virus (VSV), which induces early, potent neutralizing antibodies or recombinant lymphocytic choriomeningitis virus (LCMV), which induces a vigorous cellular response, but inefficient neutralizing antibodies, expressing the same T cell epitope. Early exposure of dendritic cells to type I interferon (IFN), which occurred during infection with VSV, induced the production of the cytokine IL-6 and drove TFH cell polarization, while late exposure to type I IFN, which occurred during infection with LCMV, did not induce IL-6 and allowed differentiation into TH1 cells. Thus, tight spatiotemporal regulation of type I IFN shapes antiviral CD4+ T cell differentiation, and might instruct vaccine design strategies.
Collapse
|
22
|
Dynamics and genomic landscape of CD8 + T cells undergoing hepatic priming. Nature 2019; 574:200-205. [PMID: 31582858 PMCID: PMC6858885 DOI: 10.1038/s41586-019-1620-6] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 08/21/2019] [Indexed: 12/15/2022]
Abstract
The responses of CD8+ T cells to hepatotropic viruses such as hepatitis B range from dysfunction to differentiation into effector cells, but the mechanisms that underlie these distinct outcomes remain poorly understood. Here we show that priming by Kupffer cells, which are not natural targets of hepatitis B, leads to differentiation of CD8+ T cells into effector cells that form dense, extravascular clusters of immotile cells scattered throughout the liver. By contrast, priming by hepatocytes, which are natural targets of hepatitis B, leads to local activation and proliferation of CD8+ T cells but not to differentiation into effector cells; these cells form loose, intravascular clusters of motile cells that coalesce around portal tracts. Transcriptomic and chromatin accessibility analyses reveal unique features of these dysfunctional CD8+ T cells, with limited overlap with those of exhausted or tolerant T cells; accordingly, CD8+ T cells primed by hepatocytes cannot be rescued by treatment with anti-PD-L1, but instead respond to IL-2. These findings suggest immunotherapeutic strategies against chronic hepatitis B infection.
Collapse
|
23
|
Wang PW, Li YZ, Zhuang HF, Yu HM, Cai SQ, Xu H, Chen ZH, Lin JK, Yao XD. Anti-Osteoporosis Medications Associated with Decreased Mortality after Hip Fracture. Orthop Surg 2019; 11:777-783. [PMID: 31429532 PMCID: PMC6819189 DOI: 10.1111/os.12517] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 06/25/2019] [Accepted: 07/21/2019] [Indexed: 12/31/2022] Open
Abstract
Objective To study the effect of anti‐osteoporosis therapies on mortality after hip fracture. Methods This retrospective study was carried out in the Second Affiliated Hospital of Fujian Medical University and enrolled 690 patients 50 years of age and older who were admitted with hip fractures between 2010 and 2015. The patients were followed in 2017: 690 patients aged was from 50 to 103 years. There were 456 women and 234 men. There were 335 patients with fractures of the femoral neck and 355 patients with intertrochanteric fractures of the femur. There were 444 (64.35%) patients who also had internal diseases. The Charlson comorbidity index was 0–6. The anti‐osteoporosis medications were classified into no anti‐osteoporosis medication, calcium + vitamin D supplementations, non‐bisphosphonate medication, and bisphosphonate medication. The physicians followed the patients or family members by personal visit and telephone. Multivariable Cox regression analyses were done with known risk factors for mortality of hip fracture, such as gender, age, number of combined internal diseases, fracture type, place of residence, and Charlson comorbidity index, to show which anti‐osteoporosis medications had significant effects on mortality after adjustment for these variables. Results Out of 690 patients with hip fractures, 149 patients received no anti‐osteoporosis medication, 63 patients received calcium +vitamin D supplementations, 398 patients received non‐bisphosphonate medication, and 80 patients received bisphosphonate medication. The patients were followed between 7 months and 52 months, with the average of 28.53 ± 9.75 months. A total of 166 patients died during the follow‐up period. Of 166 deaths, 43 occurred within 3 months, 65 within 6 months, and 99 within 1 year after the hip fracture. In this study, fracture type, place of residence, and Charlson comorbidity index were not associated with the mortality, and the male gender, age > 75 years, and ≥ 2 combined internal diseases were the independent factors for deaths post‐hip fracture. The cumulative mortality was 36.24% in the patients receiving no anti‐osteoporosis medication. The hazard ratio for mortality after hip fracture with bisphosphonate medication, non‐bisphosphonate medication, and calcium/vitamin D supplementation was 0.355 (95% CI, 0.194–0.648), 0.492 (95% CI, 0.347–0.699) and 0.616 (95% CI, 0.341–1.114), respectively, as compared with no anti‐osteoporosis group. Bisphosphonate and non‐bisphosphonate medications for osteoporosis were significantly associated with the reduction of cumulative mortality post‐hip fracture (P < 0.01). Conclusions Bisphosphonate and non‐bisphosphonate medications for osteoporosis were significantly associated with decreased mortality after fragility hip fracture.
Collapse
Affiliation(s)
- Pei-Wen Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yi-Zhong Li
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Hua-Feng Zhuang
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Hai-Ming Yu
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Si-Qing Cai
- Department of Radiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Hao Xu
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Zhen-Hui Chen
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Jin-Kuang Lin
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xue-Dong Yao
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
24
|
Miyachi Y, Kaido T, Yao S, Shirai H, Kobayashi A, Hamaguchi Y, Kamo N, Yagi S, Uemoto S. Bone Mineral Density as a Risk Factor for Patients Undergoing Surgery for Hepatocellular Carcinoma. World J Surg 2019; 43:920-928. [PMID: 30465085 DOI: 10.1007/s00268-018-4861-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND We have reported the impact of sarcopenia and body composition on patients undergoing hepatectomy for hepatocellular carcinoma (HCC). However, the impact of bone mineral density (BMD) on outcomes after hepatectomy for HCC and correlation with other parameters including sarcopenia are unclear. METHODS We retrospectively analyzed 465 patients who underwent primary hepatectomy for HCC between April 2005 and March 2015. We analyzed the plain CT images at the level of the eleventh thoracic vertebra with the region of interest and defined as preoperative BMD. RESULTS In this cohort, male (n = 367) and female (n = 98) patients showed significant heterogeneity in age, body composition markers, tumor factors, peri-operative parameters and so on. The median preoperative BMD in male and female patients was 155 and 139 HU, respectively (P = 0.005). BMD was negatively correlated with age in female (r = -0.590, P < 0.001) and intramuscular adipose tissue content in both male and female (r = -0.332 and -0.359, respectively, P < 0.001). For males, BMD < 160 HU was associated with worse cancer-specific survival post-hepatectomy (P = 0.015). In contrast, females were not (P = 0.135). For male patients, multivariate analysis identified low BMD as an independent risk factor for death (hazard ratio 1.720, 95% confidence interval 1.038-2.922, P = 0.035) after hepatectomy for HCC. CONCLUSION Preoperative low BMD was an independent risk factor for cancer-specific mortality after hepatectomy for HCC.
Collapse
Affiliation(s)
- Yosuke Miyachi
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Toshimi Kaido
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Siuan Yao
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hisaya Shirai
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Atsushi Kobayashi
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yuhei Hamaguchi
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Naoko Kamo
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Shintaro Yagi
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Shinji Uemoto
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
25
|
Abstract
Antibodies play a crucial role in virus control. The production of antibodies requires virus-specific B cells to encounter viral antigens in lymph nodes, become activated, interact with different immune cells, proliferate and enter specific differentiation programmes. Each step occurs in distinct lymph node niches, requiring a coordinated migration of B cells between different subcompartments. The development of multiphoton intravital microscopy has enabled researchers to begin to elucidate the precise cellular and molecular events by which lymph nodes coordinate humoral responses. This Review discusses recent studies that clarify how viruses interfere with antibody responses, highlighting how these mechanisms relate to our topological and temporal understanding of B cell activation within secondary lymphoid organs.
Collapse
Affiliation(s)
- Mirela Kuka
- Division of Immunology, Transplantation and Infectious Diseases and Experimental Imaging Center, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Via Olgettina 58, Milan 20132, Italy
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases and Experimental Imaging Center, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Via Olgettina 58, Milan 20132, Italy
| |
Collapse
|
26
|
Medaglia C, Giladi A, Stoler-Barak L, De Giovanni M, Salame TM, Biram A, David E, Li H, Iannacone M, Shulman Z, Amit I. Spatial reconstruction of immune niches by combining photoactivatable reporters and scRNA-seq. Science 2017; 358:1622-1626. [PMID: 29217582 DOI: 10.1126/science.aao4277] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 11/27/2017] [Indexed: 12/20/2022]
Abstract
Cellular functions are strongly dependent on surrounding cells and environmental factors. Current technologies are limited in their ability to characterize the spatial location and gene programs of cells in poorly structured and dynamic niches. We developed a method, NICHE-seq, that combines photoactivatable fluorescent reporters, two-photon microscopy, and single-cell RNA sequencing (scRNA-seq) to infer the cellular and molecular composition of niches. We applied NICHE-seq to examine the high-order assembly of immune cell networks. NICHE-seq is highly reproducible in spatial tissue reconstruction, enabling identification of rare niche-specific immune subpopulations and gene programs, including natural killer cells within infected B cell follicles and distinct myeloid states in the spleen and tumor. This study establishes NICHE-seq as a broadly applicable method for elucidating high-order spatial organization of cell types and their molecular pathways.
Collapse
Affiliation(s)
- Chiara Medaglia
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Amir Giladi
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Liat Stoler-Barak
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Marco De Giovanni
- Division of Immunology, Transplantation and Infectious Diseases and Experimental Imaging Center, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milan 20132, Italy
| | - Tomer Meir Salame
- Flow Cytometry Unit, Department of Biological Services, Weizmann Institute of Science, Rehovot, Israel
| | - Adi Biram
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Hanjie Li
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases and Experimental Imaging Center, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milan 20132, Italy.
| | - Ziv Shulman
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel.
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
27
|
Jiménez de Oya N, De Giovanni M, Fioravanti J, Übelhart R, Di Lucia P, Fiocchi A, Iacovelli S, Efremov DG, Caligaris-Cappio F, Jumaa H, Ghia P, Guidotti LG, Iannacone M. Pathogen-specific B-cell receptors drive chronic lymphocytic leukemia by light-chain-dependent cross-reaction with autoantigens. EMBO Mol Med 2017; 9:1482-1490. [PMID: 28899929 PMCID: PMC5666309 DOI: 10.15252/emmm.201707732] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 08/11/2017] [Accepted: 08/15/2017] [Indexed: 12/27/2022] Open
Abstract
Several lines of evidence indirectly suggest that antigenic stimulation through the B-cell receptor (BCR) supports chronic lymphocytic leukemia (CLL) development. In addition to self-antigens, a number of microbial antigens have been proposed to contribute to the selection of the immunoglobulins expressed in CLL. How pathogen-specific BCRs drive CLL development remains, however, largely unexplored. Here, we utilized mouse models of CLL pathogenesis to equip B cells with virus-specific BCRs and study the effect of antigen recognition on leukemia growth. Our results show that BCR engagement is absolutely required for CLL development. Unexpectedly, however, neither acute nor chronic exposure to virus-derived antigens influenced leukemia progression. Rather, CLL clones preferentially selected light chains that, when paired with virus-specific heavy chains, conferred B cells the ability to recognize a broad range of autoantigens. Taken together, our results suggest that pathogens may drive CLL pathogenesis by selecting and expanding pathogen-specific B cells that cross-react with one or more self-antigens.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Autoantigens/immunology
- Disease Models, Animal
- Immunoglobulin Light Chains/metabolism
- Immunoglobulins/metabolism
- Intercellular Adhesion Molecule-3/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Protein Array Analysis
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Receptors, Antigen, B-Cell/metabolism
- Spleen/cytology
- Spleen/metabolism
- Vesicular stomatitis Indiana virus/genetics
- Vesicular stomatitis Indiana virus/metabolism
Collapse
Affiliation(s)
- Nereida Jiménez de Oya
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco De Giovanni
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Jessica Fioravanti
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rudolf Übelhart
- Institute of Immunology, University Hospital Ulm, Ulm, Germany
| | - Pietro Di Lucia
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Amleto Fiocchi
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Iacovelli
- Molecular Hematology Unit, International Centre for Genetic Engineering & Biotechnology, Trieste, Italy
| | - Dimitar G Efremov
- Molecular Hematology Unit, International Centre for Genetic Engineering & Biotechnology, Trieste, Italy
| | | | - Hassan Jumaa
- Institute of Immunology, University Hospital Ulm, Ulm, Germany
- Department of Molecular Immunology, Faculty of Biology, Albert-Ludwigs University of Freiburg, Freiburg, Germany
| | - Paolo Ghia
- Vita-Salute San Raffaele University, Milan, Italy
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca G Guidotti
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
28
|
Xue F, Goli V, Petraro P, McMullan T, Sprafka JM, Tchetgen Tchetgen EJ. Marginal structural model to evaluate the association between cumulative osteoporosis medication and infection using claims data. Osteoporos Int 2017; 28:2893-2901. [PMID: 28685279 PMCID: PMC5624978 DOI: 10.1007/s00198-017-4129-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 06/14/2017] [Indexed: 11/05/2022]
Abstract
UNLABELLED Due to the suboptimal persistence to osteoporosis (OP) treatment, factors triggering treatment discontinuation/switching may be causing time-varying confounding. BP treatment was associated with the risk of overall infection in opposite directions in the unweighted Cox model versus the weighted MSM. The discrepancy of effect estimates for overall infection in the MSM suggested there may be time-varying confounding. INTRODUCTION Due to the suboptimal persistence to osteoporosis (OP) treatment, factors triggering treatment discontinuation/switching may be affected by prior treatment and confound the subsequent treatment effect, causing time-varying confounding. METHODS In a US insurance database, the association between joint treatment of bisphosphonates (BP) and other OP medication and the incidence of infections among postmenopausal women was assessed using a marginal structural model (MSM). Stabilized weights were estimated by modeling treatment and censoring processes conditioning on past treatment, and baseline and time-varying covariates. RESULTS BP treatment was associated with the risk of overall infection in opposite directions in the unweighted Cox model {incidence rate ratio [IRR] [95% confidence interval (CI)] = 1.15 [1.14-1.17]} versus the weighted MSM [IRR (95% CI) = 0.79 (0.77-0.81)], but was consistently associated with a lower risk of serious infection in both the unweighted Cox model [IRR (95% CI] = 0.79 (0.78-0.81)) and the weighted MSM [IRR (95% CI) = 0.71 (0.68-0.75)]. Similar results were found when current and past treatments were simultaneously assessed. CONCLUSIONS The discrepancy of effect estimates for overall but not serious infection comparing unweighted models and MSM suggested analyses of composite outcomes with a wide range of disease severity may be more susceptible to time-varying confounding.
Collapse
Affiliation(s)
- F. Xue
- Center for Observational Research, Amgen Inc., Thousand Oaks, CA USA
| | - V. Goli
- Center for Observational Research, Amgen Inc., Thousand Oaks, CA USA
| | - P. Petraro
- Center for Observational Research, Amgen Inc., Thousand Oaks, CA USA
| | - T. McMullan
- Global Biostatistics, Amgen Inc., Thousand Oaks, CA USA
| | - J. M. Sprafka
- Center for Observational Research, Amgen Inc., Thousand Oaks, CA USA
| | - E. J. Tchetgen Tchetgen
- Department of Biostatistics and Epidemiology, Harvard School of Public Health, Boston, MA USA
| |
Collapse
|
29
|
Inhaled Fine Particles Induce Alveolar Macrophage Death and Interleukin-1α Release to Promote Inducible Bronchus-Associated Lymphoid Tissue Formation. Immunity 2017; 45:1299-1310. [PMID: 28002730 DOI: 10.1016/j.immuni.2016.11.010] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 09/12/2016] [Accepted: 10/17/2016] [Indexed: 12/11/2022]
Abstract
Particulate pollution is thought to function as an adjuvant that can induce allergic responses. However, the exact cell types and immunological factors that initiate the lung-specific immune responses are unclear. We found that upon intratracheal instillation, particulates such as aluminum salts and silica killed alveolar macrophages (AMs), which then released interleukin-1α (IL-1α) and caused inducible bronchus-associated lymphoid tissue (iBALT) formation in the lung. IL-1α release continued for up to 2 weeks after particulate exposure, and type-2 allergic immune responses were induced by the inhalation of antigen during IL-1α release and iBALT formation, even long after particulate instillation. Recombinant IL-1α was sufficient to induce iBALTs, which coincided with subsequent immunoglobulin E responses, and IL-1-receptor-deficient mice failed to induce iBALT formation. Therefore, the AM-IL-1α-iBALT axis might be a therapeutic target for particulate-induced allergic inflammation.
Collapse
|
30
|
Effector CD8 + T cell-derived interleukin-10 enhances acute liver immunopathology. J Hepatol 2017; 67:543-548. [PMID: 28483675 PMCID: PMC7127652 DOI: 10.1016/j.jhep.2017.04.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 03/27/2017] [Accepted: 04/25/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Besides secreting pro-inflammatory cytokines, chemokines and effector molecules, effector CD8+ T cells that arise upon acute infection with certain viruses have been shown to produce the regulatory cytokine interleukin (IL)-10 and, therefore, contain immunopathology. Whether the same occurs during acute hepatitis B virus (HBV) infection and role that IL-10 might play in liver disease is currently unknown. METHODS Mouse models of acute HBV pathogenesis, as well as chimpanzees and patients acutely infected with HBV, were used to analyse the role of CD8+ T cell-derived IL-10 in liver immunopathology. RESULTS Mouse HBV-specific effector CD8+ T cells produce significant amounts of IL-10 upon in vivo antigen encounter. This is corroborated by longitudinal data in a chimpanzee acutely infected with HBV, where serum IL-10 was readily detectable and correlated with intrahepatic CD8+ T cell infiltration and liver disease severity. Unexpectedly, mouse and human CD8+ T cell-derived IL-10 was found to act in an autocrine/paracrine fashion to enhance IL-2 responsiveness, thus preventing antigen-induced HBV-specific effector CD8+ T cell apoptosis. Accordingly, the use of mouse models of HBV pathogenesis revealed that the IL-10 produced by effector CD8+ T cells promoted their own intrahepatic survival and, thus supported, rather than suppressed liver immunopathology. CONCLUSION Effector CD8+ T cell-derived IL-10 enhances acute liver immunopathology. Altogether, these results extend our understanding of the cell- and tissue-specific role that IL-10 exerts in immune regulation. Lay summary: Interleukin-10 is mostly regarded as an immunosuppressive cytokine. We show here that HBV-specific CD8+ T cells produce IL-10 upon antigen recognition and that this cytokine enhances CD8+ T cell survival. As such, IL-10 paradoxically promotes rather than suppresses liver disease.
Collapse
|
31
|
Immune-mediated syndromes following intravenous bisphosphonate therapy. Inflammopharmacology 2017; 25:665-671. [PMID: 28567535 DOI: 10.1007/s10787-017-0365-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 05/24/2017] [Indexed: 01/28/2023]
Abstract
OBJECTIVES Intravenous (IV) infusion of aminobisphosphonates (ABP) induces cytokine release by peripheral blood Vγ9δ2 T cells, resulting in an immediate short-term inflammatory response in up to 50% of patients. We evaluated possible long-term pro-inflammatory effects of IV ABP. METHODS Retrospective case-series study from one rheumatology specialist's clinic. 2261 electronic charts were reviewed for administration of 'zoledronate' or different brand names of zoledronic acid, and relevant clinical data was retrieved for patients who had received the infusion. RESULTS Thirteen patients had recieved zoledronate. In six, new-onset or exacerbation of a previous inflammatory/autoimmune disorder was diagnosed within 3 months following infusion. Of these, one patient developed new-onset rheumatoid arthritis (RA), two polymyalgia rheumatica (PMR), two suffered a flare of Crohn's disease-related and aromatase inhibitor-induced arthralgias, and one patient acquired autoimmune hemophilia. Pre-existing malignancy and immediate inflammatory response following zoledronate were more frequent in patients experiencing new or worsening immunologic manifestations (3/6 vs. 0/7, and 5/6 vs. 2/7, respectively). CONCLUSIONS Intravenous ABP may trigger induction of persistent autoimmune syndromes, especially when accompanied by an immediate adverse reaction or pre-existing malignancy.
Collapse
|
32
|
Abstract
The field of vaccination is moving from the use of attenuated or inactivated pathogens to safer but less immunogenic protein and peptide antigens, which require stronger adjuvant compositions. Antigen delivery carriers appear to play an important role in vaccine development, providing not only antigen protection and controlled release but also an intrinsic adjuvant potential. Among them, carriers based on polymers and lipids are the most representative ones. Patent applications in this area have disclosed, either the design and preparation methods for new biocompatible antigen delivery systems or the application of the previously developed systems for the delivery of novel antigens. Some of them have also reported the use of these technologies for modern therapeutic vaccination approaches.
Collapse
|
33
|
Guidotti LG, Inverso D, Sironi L, Di Lucia P, Fioravanti J, Ganzer L, Fiocchi A, Vacca M, Aiolfi R, Sammicheli S, Mainetti M, Cataudella T, Raimondi A, Gonzalez-Aseguinolaza G, Protzer U, Ruggeri ZM, Chisari FV, Isogawa M, Sitia G, Iannacone M. Immunosurveillance of the liver by intravascular effector CD8(+) T cells. Cell 2015; 161:486-500. [PMID: 25892224 DOI: 10.1016/j.cell.2015.03.005] [Citation(s) in RCA: 217] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 12/18/2014] [Accepted: 02/24/2015] [Indexed: 02/06/2023]
Abstract
Effector CD8(+) T cells (CD8 TE) play a key role during hepatotropic viral infections. Here, we used advanced imaging in mouse models of hepatitis B virus (HBV) pathogenesis to understand the mechanisms whereby these cells home to the liver, recognize antigens, and deploy effector functions. We show that circulating CD8 TE arrest within liver sinusoids by docking onto platelets previously adhered to sinusoidal hyaluronan via CD44. After the initial arrest, CD8 TE actively crawl along liver sinusoids and probe sub-sinusoidal hepatocytes for the presence of antigens by extending cytoplasmic protrusions through endothelial fenestrae. Hepatocellular antigen recognition triggers effector functions in a diapedesis-independent manner and is inhibited by the processes of sinusoidal defenestration and capillarization that characterize liver fibrosis. These findings reveal the dynamic behavior whereby CD8 TE control hepatotropic pathogens and suggest how liver fibrosis might reduce CD8 TE immune surveillance toward infected or transformed hepatocytes.
Collapse
Affiliation(s)
- Luca G Guidotti
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Department of Immunology and Microbial Sciences, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Donato Inverso
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Laura Sironi
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Department of Physics, University of Milano Bicocca, 20126 Milan, Italy
| | - Pietro Di Lucia
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Jessica Fioravanti
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Lucia Ganzer
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Department of Physics, University of Milano Bicocca, 20126 Milan, Italy
| | - Amleto Fiocchi
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Maurizio Vacca
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Roberto Aiolfi
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Stefano Sammicheli
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Marta Mainetti
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Tiziana Cataudella
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Andrea Raimondi
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | - Ulrike Protzer
- Institute of Virology, Technical University of Munich, 81675 Munich, Germany
| | - Zaverio M Ruggeri
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Francis V Chisari
- Department of Immunology and Microbial Sciences, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Masanori Isogawa
- Department of Immunology and Microbial Sciences, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Giovanni Sitia
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Vita-Salute San Raffaele University, 20132 Milan, Italy; Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.
| |
Collapse
|
34
|
Marcu-Malina V, Balbir-Gurman A, Dardik R, Braun-Moscovici Y, Segel MJ, Bank I. A Novel Prothrombotic Pathway in Systemic Sclerosis Patients: Possible Role of Bisphosphonate-Activated γδ T Cells. Front Immunol 2014; 5:414. [PMID: 25250025 PMCID: PMC4157565 DOI: 10.3389/fimmu.2014.00414] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 08/15/2014] [Indexed: 11/17/2022] Open
Abstract
Objectives: Infusions of aminobisphonates (ABP) activate Vγ9δ2T cells in vivo and induce an acute inflammatory response in 30% of patients treated for osteoporosis. Following the observation of digital thrombosis in a systemic sclerosis (SSc) patient after treatment with an intravenous ABP, zoledronate (Zol), we evaluated whether patient and control peripheral blood (PB) mononuclear cell (MC, PBMC) acquire a prothrombotic phenotype in response to Zol. Results: Vγ9δ2T cells of both patients and healthy donors (HD) upregulated the CD69 activation antigen and secreted tumor necrosis factor (TNF)α in response to Zol in vitro. In addition, exposure to either Zol or lipopolysaccharide (LPS), or to both additively, induced expression of the highly procoagulant, tissue factor (TF)-1 on CD14+ monocytes. Importantly, only Zol-induced TF-1 was blocked by a monoclonal antibody to TNFα. Interestingly, we found that SSc, but not HD, Vδ1+ T cells were concurrently activated by Zol to produce interleukin (IL)-4. Addition of plasma from the blood of the SSc patient who developed critical digital ischemia after infusion of Zol, but neither plasma from a second patient with no adverse clinical response to Zol infusion nor of a HD, strongly enhanced Zol-induced monocyte TF-1, which could still be blocked by anti-TNFα. Conclusion: Aminobisphonates induced secretion of TNFα by Vγ9δ2+ T cells may lead to TNFα-dependent induction of procoagulant TF-1 induction on monocytes. In certain clinical settings, e.g., SSc, TF-1+ monocytes could play a role in triggering clinically relevant thrombosis.
Collapse
Affiliation(s)
| | - Alexandra Balbir-Gurman
- B Shine Rheumatology Unit, Rambam Health Care Campus, Rambam Medical Center , Haifa , Israel
| | - Rima Dardik
- Institutes of Thrombosis and Hemostasis, Sheba Medical Center , Ramat Gan , Israel
| | - Yolanda Braun-Moscovici
- B Shine Rheumatology Unit, Rambam Health Care Campus, Rambam Medical Center , Haifa , Israel
| | - Michael J Segel
- Institute of Pulmonary Diseases, Sheba Medical Center , Ramat Gan , Israel
| | - Ilan Bank
- Laboratory of Immunoregulation, Sheba Medical Center , Ramat Gan , Israel ; Department of Medicine F, Sheba Medical Center , Ramat Gan , Israel ; Department of Medicine, Sackler School of Medicine, Tel Aviv University , Tel Aviv , Israel
| |
Collapse
|
35
|
Kuka M, Iannacone M. The role of lymph node sinus macrophages in host defense. Ann N Y Acad Sci 2014; 1319:38-46. [DOI: 10.1111/nyas.12387] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Mirela Kuka
- Division of Immunology; Transplantation and Infectious Diseases; San Raffaele Scientific Institute and Vita-Salute San Raffaele University; Milan Italy
| | - Matteo Iannacone
- Division of Immunology; Transplantation and Infectious Diseases; San Raffaele Scientific Institute and Vita-Salute San Raffaele University; Milan Italy
| |
Collapse
|
36
|
The adjuvant action of bisphosphonates. Nat Rev Immunol 2013. [DOI: 10.1038/nri3578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|