1
|
Crescioli S, Jatiani S, Moise L. With great power, comes great responsibility: the importance of broadly measuring Fc-mediated effector function early in the antibody development process. MAbs 2025; 17:2453515. [PMID: 39819511 PMCID: PMC11810086 DOI: 10.1080/19420862.2025.2453515] [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: 10/23/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/19/2025] Open
Abstract
The field of antibody therapeutics is rapidly growing, with over 210 antibodies currently approved or in regulatory review and ~ 1,250 antibodies in clinical development. Antibodies are highly versatile molecules that, with strategic design of their antigen-binding domain (Fab) and the domain responsible for mediating effector functions (Fc), can be used in a wide range of therapeutic indications. Building on many years of progress, the biopharmaceutical industry is now advancing innovative research and development by exploring new targets and new formats and using antibody engineering to fine-tune functions tailored to specific disease requirements. In addition to considering the target and the disease context, however, the unique features of each therapeutic antibody trigger a diverse set of Fc-mediated effector functions. To avoid unexpected results on safety and efficacy outcomes during the later stages of the development process, it is crucial to measure the impact of antibody design on Fc-mediated effector function early in the antibody development process. Given the breadth of effector functions antibodies can deploy and the close interplay between the antibody Fab and Fc functional domains, it is important to conduct a comprehensive evaluation of Fc-mediated functions using an array of antigen-specific biophysical and cell-mediated functional assays. Here, we review antibody and Fc receptor properties that influence Fc effector functions and discuss their implications on development of safe and efficacious antibody therapeutics.
Collapse
|
2
|
Malherbe DC, Kimble JB, Atyeo C, Fischinger S, Meyer M, Cody SG, Hyde M, Alter G, Bukreyev A. A Single-Dose Intranasal Combination Panebolavirus Vaccine. J Infect Dis 2023; 228:S648-S659. [PMID: 37469133 PMCID: PMC10651208 DOI: 10.1093/infdis/jiad266] [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: 02/01/2023] [Revised: 06/23/2023] [Accepted: 07/10/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND Ebolaviruses Ebola (EBOV), Sudan (SUDV), and Bundibugyo (BDBV) cause severe human disease, which may be accompanied by hemorrhagic syndrome, with high case fatality rates. Monovalent vaccines do not offer cross-protection against these viruses whose endemic areas overlap. Therefore, development of a panebolavirus vaccine is a priority. As a vaccine vector, human parainfluenza virus type 3 (HPIV3) has the advantages of needle-free administration and induction of both systemic and local mucosal antibody responses in the respiratory tract. METHODS To minimize the antivector immunity, genes encoding the HPIV3 envelope proteins F and HN were removed from the vaccine constructs, resulting in expression of only the ebolavirus envelope protein-glycoprotein. These second-generation vaccine constructs were used to develop a combination vaccine against EBOV, SUDV, and BDBV. RESULTS A single intranasal vaccination of guinea pigs or ferrets with the trivalent combination vaccine elicited humoral responses to each of the targeted ebolaviruses, including binding and neutralizing antibodies, as well as Fc-mediated effector functions. This vaccine protected animals from death and disease caused by lethal challenges with EBOV, SUDV, or BDBV. CONCLUSIONS The combination vaccine elicited protection that was comparable to that induced by the monovalent vaccines, thus demonstrating the value of this combination trivalent vaccine.
Collapse
Affiliation(s)
- Delphine C Malherbe
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, Galveston, Texas, USA
| | - J Brian Kimble
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, Galveston, Texas, USA
| | - Caroline Atyeo
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts, USA
| | - Stephanie Fischinger
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts, USA
| | - Michelle Meyer
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, Galveston, Texas, USA
| | - S Gabrielle Cody
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Matthew Hyde
- Galveston National Laboratory, Galveston, Texas, USA
| | - Galit Alter
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts, USA
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, Galveston, Texas, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
3
|
Wu J, Ma Y, Nie Y, Wang J, Feng G, Hao L, Huang W, Li Y, Liu Z. Functional Characterization of Largemouth Bass ( Micropterus salmoides) Soluble FcγR Homolog in Response to Bacterial Infection. Int J Mol Sci 2022; 23:ijms232213788. [PMID: 36430268 PMCID: PMC9699129 DOI: 10.3390/ijms232213788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/30/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
Fc receptors (FcRs) are key players in antibody-dependent cellular phagocytosis (ADCP) with their specific recognition of the Fc portion of an immunoglobulin. Despite reports of FcγR-mediated phagocytosis in mammals, little is known about the effects of soluble FcγRs on the immune response. In this study, FcγRIα was cloned from the largemouth bass (Micropterus salmoides) (MsFcγRIα). Without a transmembrane segment or a cytoplasmic tail, MsFcγRIα was identified as a soluble form protein and widely distributed in the spleen, head kidney, and intestine. The native MsFcγRIα was detected in the serum of Nocardia seriolae-infected largemouth bass and the supernatants of transfected HEK293 cells. Additionally, it was verified that the transfected cells' surface secreted MsFcRIα could bind to largemouth bass IgM. Moreover, the expression changes of MsFcγRIα, Syk, and Lyn indicated that MsFcγRIα was engaged in the acute phase response to bacteria, and the FcγR-mediated phagocytosis pathway was activated by Nocardia seriolae stimulation. Furthermore, recombinant MsFcγRIα could enhance both reactive oxygen species (ROS) and phagocytosis to Nocardia seriolae of leukocytes, presumably through the interaction of MsFcγRIα with a complement receptor. In conclusion, these findings provided a better understanding of the function of soluble FcγRs in the immune response and further shed light on the mechanism of phagocytosis in teleosts.
Collapse
Affiliation(s)
- Jing Wu
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yanping Ma
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Guangzhou 510640, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Collaborative Innovation Center of Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Yifan Nie
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Guangzhou 510640, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Collaborative Innovation Center of Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Jingya Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Guoqing Feng
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Guangzhou 510640, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Collaborative Innovation Center of Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Le Hao
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Guangzhou 510640, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Collaborative Innovation Center of Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Wen Huang
- Collaborative Innovation Center of Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Yugu Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Correspondence: (Y.L.); (Z.L.)
| | - Zhenxing Liu
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Guangzhou 510640, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Collaborative Innovation Center of Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- Correspondence: (Y.L.); (Z.L.)
| |
Collapse
|
4
|
Haist M, Ries F, Gunzer M, Bednarczyk M, Siegel E, Kuske M, Grabbe S, Radsak M, Bros M, Teschner D. Neutrophil-Specific Knockdown of β2 Integrins Impairs Antifungal Effector Functions and Aggravates the Course of Invasive Pulmonal Aspergillosis. Front Immunol 2022; 13:823121. [PMID: 35734179 PMCID: PMC9207500 DOI: 10.3389/fimmu.2022.823121] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 04/28/2022] [Indexed: 12/12/2022] Open
Abstract
β2-integrins are heterodimeric surface receptors that are expressed specifically by leukocytes and consist of a variable α (CD11a-d) and a common β-subunit (CD18). Functional impairment of CD18, which causes leukocyte adhesion deficiency type-1 results in an immunocompromised state characterized by severe infections, such as invasive pulmonary aspergillosis (IPA). The underlying immune defects have largely been attributed to an impaired migratory and phagocytic activity of polymorphonuclear granulocytes (PMN). However, the exact contribution of β2-integrins for PMN functions in-vivo has not been elucidated yet, since the mouse models available so far display a constitutive CD18 knockout (CD18-/- or CD18hypo). To determine the PMN-specific role of β2-integrins for innate effector functions and pathogen control, we generated a mouse line with a Ly6G-specific knockdown of the common β-subunit (CD18Ly6G cKO). We characterized CD18Ly6G cKO mice in-vitro to confirm the PMN-specific knockdown of β2-integrins. Next, we investigated the clinical course of IPA in A. fumigatus infected CD18Ly6G cKO mice with regard to the fungal burden, pulmonary inflammation and PMN response towards A. fumigatus. Our results revealed that the β2-integrin knockdown was restricted to PMN and that CD18Ly6G cKO mice showed an aggravated course of IPA. In accordance, we observed a higher fungal burden and lower levels of proinflammatory innate cytokines, such as TNF-α, in lungs of IPA-infected CD18Ly6G cKO mice. Bronchoalveolar lavage revealed higher levels of CXCL1, a stronger PMN-infiltration, but concomitantly elevated apoptosis of PMN in lungs of CD18Ly6G cKO mice. Ex-vivo analysis further unveiled a strong impairment of PMN effector function, as reflected by an attenuated phagocytic activity, and a diminished generation of reactive oxygen species (ROS) and neutrophil-extracellular traps (NET) in CD18-deficient PMN. Overall, our study demonstrates that β2-integrins are required specifically for PMN effector functions and contribute to the clearance of A. fumigatus by infiltrating PMN, and the establishment of an inflammatory microenvironment in infected lungs.
Collapse
Affiliation(s)
- Maximilian Haist
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
- *Correspondence: Maximilian Haist,
| | - Frederic Ries
- Department of Hematology, Medical Oncology and Pneumology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital, University Duisburg-Essen, Essen, Germany
- Leibniz-Institut für Analytische Wissenschaften ISAS -e.V, Dortmund, Germany
| | - Monika Bednarczyk
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Ekkehard Siegel
- Institute for Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Michael Kuske
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Markus Radsak
- Department of Hematology, Medical Oncology and Pneumology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Daniel Teschner
- Department of Hematology, Medical Oncology and Pneumology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
5
|
Gunn BM, Bai S. Building a better antibody through the Fc: advances and challenges in harnessing antibody Fc effector functions for antiviral protection. Hum Vaccin Immunother 2021; 17:4328-4344. [PMID: 34613865 PMCID: PMC8827636 DOI: 10.1080/21645515.2021.1976580] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 12/15/2022] Open
Abstract
Antibodies can provide antiviral protection through neutralization and recruitment of innate effector functions through the Fc domain. While neutralization has long been appreciated for its role in antibody-mediated protection, a growing body of work indicates that the antibody Fc domain also significantly contributes to antiviral protection. Recruitment of innate immune cells such as natural killer cells, neutrophils, monocytes, macrophages, dendritic cells and the complement system by antibodies can lead to direct restriction of viral infection as well as promoting long-term antiviral immunity. Monoclonal antibody therapeutics against viruses are increasingly incorporating Fc-enhancing features to take advantage of the Fc domain, uncovering a surprising breadth of mechanisms through which antibodies can control viral infection. Here, we review the recent advances in our understanding of antibody-mediated innate immune effector functions in protection from viral infection and review the current approaches and challenges to effectively leverage innate immune cells via antibodies.
Collapse
Affiliation(s)
- Bronwyn M. Gunn
- Paul G. Allen School of Global Health, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Shuangyi Bai
- Paul G. Allen School of Global Health, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| |
Collapse
|
6
|
Takagi K, Somiya M, Jung J, Iijima M, Kuroda S. Polymerized Albumin Receptor of Hepatitis B Virus for Evading the Reticuloendothelial System. Pharmaceuticals (Basel) 2021; 14:ph14050408. [PMID: 33923102 PMCID: PMC8145202 DOI: 10.3390/ph14050408] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 01/05/2023] Open
Abstract
Various strategies, such as optimization of surface chemistry, size, shape, and charge, have been undertaken to develop nanoparticles (NPs) as DDS (drug delivery system) nanocarriers for evading the reticuloendothelial system (RES) in vivo. We previously developed a hollow NP composed of hepatitis B virus (HBV) surface antigen L proteins and lipid bilayers, hereinafter referred to as bio-nanocapsule (BNC), as a nonviral DDS nanocarrier. Such a BNC harbors the HBV-derived human hepatic cell-specific infection mechanism, and intravenously injected BNCs by themselves were shown to avoid clearance by RES-rich organs and accumulate in target tissues. In this study, since the surface modification with albumins is known to prolong the circulation time of nanomedicines, we examined whether the polymerized albumin receptor (PAR) of BNCs contributes to RES evasion in mouse liver. Our results show that NPs conjugated with peptides possessing sufficient PAR activity were captured by Kupffer cells less efficiently in vitro and were able to circulate for a longer period of time in vivo. Comparing with polyethylene glycol, PAR peptides were shown to reduce the recognition by RES to equal content. Taken together, our results strongly suggest that the PAR domain of BNCs, as well as HBV, harbors an innate RES evasion mechanism. Therefore, the surface modification with PAR peptides could be an alternative strategy for improving the pharmacodynamics and pharmacokinetics of forthcoming nanomedicines.
Collapse
Affiliation(s)
- Kurumi Takagi
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan; (K.T.); (M.I.)
| | - Masaharu Somiya
- Department of Biomolecular Science and Reaction, The Institute of Scientific and Industrial Research, Osaka University, Osaka 567-0047, Japan;
| | - Joohee Jung
- College of Pharmacy, Duksung Women’s University, Seoul 132-714, Korea;
| | - Masumi Iijima
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan; (K.T.); (M.I.)
- Department of Nutritional Science and Food Safety, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo 156-8502, Japan
| | - Shun’ichi Kuroda
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan; (K.T.); (M.I.)
- Department of Biomolecular Science and Reaction, The Institute of Scientific and Industrial Research, Osaka University, Osaka 567-0047, Japan;
- Correspondence:
| |
Collapse
|
7
|
Zent CS, Pinney JJ, Chu CC, Elliott MR. Complement Activation in the Treatment of B-Cell Malignancies. Antibodies (Basel) 2020; 9:E68. [PMID: 33271825 PMCID: PMC7709106 DOI: 10.3390/antib9040068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 09/30/2020] [Accepted: 11/22/2020] [Indexed: 12/13/2022] Open
Abstract
Unconjugated monoclonal antibodies (mAb) have revolutionized the treatment of B-cell malignancies. These targeted drugs can activate innate immune cytotoxicity for therapeutic benefit. mAb activation of the complement cascade results in complement-dependent cytotoxicity (CDC) and complement receptor-mediated antibody-dependent cellular phagocytosis (cADCP). Clinical and laboratory studies have showed that CDC is therapeutically important. In contrast, the biological role and clinical effects of cADCP are less well understood. This review summarizes the available data on the role of complement activation in the treatment of mature B-cell malignancies and proposes future research directions that could be useful in optimizing the efficacy of this important class of drugs.
Collapse
Affiliation(s)
- Clive S. Zent
- Wilmot Cancer Institute and Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Jonathan J. Pinney
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA; (J.J.P.); (M.R.E.)
- Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908, USA
| | - Charles C. Chu
- Wilmot Cancer Institute and Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Michael R. Elliott
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA; (J.J.P.); (M.R.E.)
- Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
8
|
Saleh R, Sasidharan Nair V, Al-Dhaheri M, Khawar M, Abu Nada M, Alajez NM, Elkord E. RNA-Seq Analysis of Colorectal Tumor-Infiltrating Myeloid-Derived Suppressor Cell Subsets Revealed Gene Signatures of Poor Prognosis. Front Oncol 2020; 10:604906. [PMID: 33312958 PMCID: PMC7703275 DOI: 10.3389/fonc.2020.604906] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 10/15/2020] [Indexed: 12/11/2022] Open
Abstract
Elevated levels of myeloid-derived suppressor cells (MDSCs), including polymorphonuclear MDSCs (PMN-MDSCs) and immature MDSCs (I-MDSCs), are usually associated with disease progression in cancer patients, including colorectal cancer (CRC). However, biological mechanisms and molecular pathways regulated by MDSC subpopulations in the CRC tumor microenvironment (TME) have not been fully investigated. In this study, we performed transcriptomic analysis of tumor-infiltrating I-MDSCs and PMN-MDSCs isolated from tumor tissues of six CRC patients, compared to antigen-presenting cells (APCs). We also compared the transcriptomic profiles of tumor-infiltrating PMN-MDSCs to I-MDSCs. Our results showed different molecular pathways regulated by each MDSC subset, potentially reflecting their phenotypical/molecular/functional characteristics in the CRC TME. Moreover, we identified gene signatures in PMN-MDSC and I-MDSC of poor overall survival (OS) and disease-free survival (DFS) using the Cancer Genome Atlas (TCGA) dataset from patients with colon adenocarcinoma (COAD). However, functional studies are required to validate these findings.
Collapse
Affiliation(s)
- Reem Saleh
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Varun Sasidharan Nair
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | | | - Mahwish Khawar
- Department of Surgery, Hamad Medical Corporation, Doha, Qatar
| | | | - Nehad M Alajez
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Eyad Elkord
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar.,Biomedical Research Center, School of Science, Engineering and Environment, University of Salford, Manchester, United Kingdom
| |
Collapse
|
9
|
Saleh R, Taha RZ, Sasidharan Nair V, Toor SM, Alajez NM, Elkord E. Transcriptomic Profiling of Circulating HLA-DR - Myeloid Cells, Compared with HLA-DR + Myeloid Antigen-presenting Cells. Immunol Invest 2020; 50:952-963. [PMID: 32727251 DOI: 10.1080/08820139.2020.1795875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells with potent immunosuppressive functions, which can inhibit the activation of immune responses under a steady-state condition and pathological conditions. We performed transcriptomic profiling of circulating CD33+HLA-DR+ myeloid antigen-presenting cells (APCs) and CD33+HLA-DR- myeloid cells (potentially MDSCs) in healthy individuals. We sorted both subpopulations from peripheral blood mononuclear cells (PBMCs) of 10 healthy donors and performed RNA sequencing (RNA-Seq). We found that several signaling pathways associated with the positive regulation of immune responses, such as antigen presentation/processing, FcγR-mediated phagocytosis and immune cell trafficking, phosphoinositide 3-kinase (PI3K)/Akt signaling, DC maturation, triggering receptor expressed on myeloid cells 1 (TREM1) signaling, nuclear factor of activated T cells (NFAT) and IL-8 signaling were downregulated in CD33+HLA-DR- myeloid cells. In contrast, pathways implicated in tumor suppression and anti-inflammation, including peroxisome proliferator-activated receptor (PPAR) and phosphatase and tensin homolog (PTEN), were upregulated in CD33+HLA-DR- myeloid cells. These data indicate that PPAR/PTEN axis could be upregulated in myeloid cells to keep the immune system in check in normal physiological conditions. Our data reveal some of the molecular and functional differences between CD33+HLA-DR+ APCs and CD33+HLA-DR- myeloid cells in a steady-state condition, reflecting the potential suppressive function of CD33+HLA-DR- myeloid cells to maintain immune tolerance. For future studies, the same methodological approach could be applied to perform transcriptomic profiling of myeloid subsets in pathological conditions.
Collapse
Affiliation(s)
- Reem Saleh
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Rowaida Z Taha
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Varun Sasidharan Nair
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Salman M Toor
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Nehad M Alajez
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Eyad Elkord
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| |
Collapse
|
10
|
Heterogeneity in neutrophil responses to immune complexes. Blood Adv 2019; 3:2778-2789. [PMID: 31554616 DOI: 10.1182/bloodadvances.2019000235] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 08/13/2019] [Indexed: 11/20/2022] Open
Abstract
Immune complexes (ICs) can trigger inflammation and thrombosis, in part, by activating neutrophils. Much attention has focused on the serologic characteristics of ICs and Fc receptors associated with cellular activation, but few studies have examined host susceptibility to neutrophil activation by ICs. Here, we use a novel whole blood system to investigate the ability of ICs to cause neutrophil activation and degranulation. Using monoclonal anti-platelet factor 4/heparin (PF4/heparin), anti-protamine/heparin antibodies, patient-derived anti-PF4/heparin antibodies, and heat-aggregated immunoglobulin G as model ICs, we demonstrate that heparin-containing ICs cause robust, heparin-dependent neutrophil activation and degranulation which is mediated by both FcγRIIa and complement. Longitudinal testing over a 1-year period shows that an individual's neutrophil response to ICs represents a fixed phenotype resulting in high, intermediate, or low reactivity. Examination of individuals at the extremes of reactivity (high vs low) shows that phenotypic variation resides in the cellular compartment and is correlated with host white blood cell count and absolute neutrophil count, but not age, sex, race, polymorphisms in neutrophil Fcγ receptors, or CR1, CR3, and Fcγ receptor expression on neutrophils. Together, these studies demonstrate that susceptibility to neutrophil activation by ICs is intrinsic to the host and is likely genetic in origin. These findings may be relevant to the heterogeneous clinical outcomes seen in patients with heparin-induced thrombocytopenia and other IC-mediated disorders and could potentially identify patients at high risk for thrombotic and inflammatory complications.
Collapse
|
11
|
Naik U, Nguyen QPH, Harrison RE. Binding and uptake of single and dual-opsonized targets by macrophages. J Cell Biochem 2019; 121:183-199. [PMID: 31172552 DOI: 10.1002/jcb.29043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 05/03/2019] [Indexed: 12/15/2022]
Abstract
Our current understanding of phagocytosis is largely derived from studies of individual receptor-ligand interactions and their downstream signaling pathways. Because phagocytes are exposed to a variety of ligands on heterogeneous target particles in vivo, it is important to observe the engagement of multiple receptors simultaneously and the triggered involvement of downstream signaling pathways. Potential crosstalk between the two well-characterized opsonic receptors, FcγR and CR3, was briefly explored in the early 1970s, where macrophages were challenged with dual-opsonized targets. However, subsequent studies on receptor crosstalk were primarily restricted to using single opsonins on different targets, typically at saturating opsonin conditions. Beyond validating these initial explorations on receptor crosstalk, we identify the early signaling mechanisms that underlie the binding and phagocytosis during the simultaneous activation of both opsonic receptors, through the presence of a dual-opsonized target (immunoglobulin G [IgG] and C3bi), compared with single receptor activation. For this purpose, we used signaling protein inhibitor studies as well as live cell brightfield and fluorescent imaging to fully understand the role of tyrosine kinases, F-actin dynamics and internalization kinetics for FcγR and CR3. Importantly, opsonic receptors were studied together and in isolation, in the context of sparsely opsonized targets. We observed enhanced particle binding and a synergistic effect on particle internalization during the simultaneous activation of FcγR and CR3 engaged with sparsely opsonized targets. Inhibition of early signaling and cytoskeletal molecules revealed a differential involvement of Src kinase for FcγR- vs CR3- and dual receptor-mediated phagocytosis. Src activity recruits Syk kinase and we observed intermediate levels of Syk phosphorylation in dual-opsonized particles compared with those opsonized with IgG or C3bi alone. These results likely explain the intermediate levels of F-actin that is recruited to sites of dual-opsonized particle uptake and the notoriously delayed internalization of C3bi-opsonized targets by macrophages.
Collapse
Affiliation(s)
- Urja Naik
- Department of Cell & Systems Biology, University of Toronto Scarborough, Toronto, Ontario, Canada
| | - Quynh Phuong Hai Nguyen
- Department of Cell & Systems Biology, University of Toronto Scarborough, Toronto, Ontario, Canada
| | - Rene E Harrison
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
| |
Collapse
|
12
|
Teschner D, Cholaszczyńska A, Ries F, Beckert H, Theobald M, Grabbe S, Radsak M, Bros M. CD11b Regulates Fungal Outgrowth but Not Neutrophil Recruitment in a Mouse Model of Invasive Pulmonary Aspergillosis. Front Immunol 2019; 10:123. [PMID: 30778357 PMCID: PMC6369709 DOI: 10.3389/fimmu.2019.00123] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 01/15/2019] [Indexed: 12/28/2022] Open
Abstract
ß2 integrin receptors consist of an alpha subunit (CD11a-CD11d) and CD18 as the common beta subunit, and are differentially expressed by leukocytes. ß2 integrins are required for cell-cell interaction, transendothelial migration, uptake of opsonized pathogens, and cell signaling processes. Functional loss of CD18—termed leukocyte-adhesion deficiency type 1 (LAD1)—results in an immunocompromised state characterized by frequent occurrence of severe infections. In immunosuppressed individuals Aspergillus fumigatus is a frequent cause of invasive pulmonary fungal infection, and often occurs in patients suffering from LAD1. Here, we asked for the importance of CD11b/CD18 also termed MAC-1 which is required for phagocytosis of opsonized A. fumigatus conidia by polymorphonuclear neutrophils (PMN) for control of pulmonary A. fumigatus infection. We show that CD11b−/− mice infected with A. fumigatus were unaffected in long term survival, similar to wild type (WT) mice. However, bronchoalveolar lavage (BAL) performed 1 day after infection revealed a higher lung infiltration of PMN in case of infected CD11b−/− mice than observed for WT mice. BAL derived from infected CD11b−/− mice also contained a higher amount of leukocyte-attracting CCL5 chemokine, but lower amounts of proinflammatory innate cytokines. In accordance, lung tissue of A. fumigatus infected CD11b−/− mice was characterized by lower cellular inflammation, and a higher fungal burden. In agreement, CD11b−/−PMN exerted lower phagocytic activity on serum-opsonized A. fumigatus conidia than WT PMN in vitro. Our study shows that MAC-1 is required for effective clearance of A. fumigatus by infiltrating PMN, and the establishment of an inflammatory microenvironment in infected lung. Enhanced infiltration of CD11b−/− PMN may serve to compensate impaired PMN function.
Collapse
Affiliation(s)
- Daniel Teschner
- Department of Hematology, Medical Oncology and Pneumology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Anna Cholaszczyńska
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Frederic Ries
- Department of Hematology, Medical Oncology and Pneumology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Hendrik Beckert
- Department of Pulmonary Medicine, University Medical Center Essen-Ruhrlandklinik, Essen, Germany
| | - Matthias Theobald
- Department of Hematology, Medical Oncology and Pneumology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Markus Radsak
- Department of Hematology, Medical Oncology and Pneumology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
13
|
Syk-dependent tyrosine phosphorylation of 3BP2 is required for optimal FcRγ-mediated phagocytosis and chemokine expression in U937 cells. Sci Rep 2017; 7:11480. [PMID: 28904407 PMCID: PMC5597638 DOI: 10.1038/s41598-017-11915-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 08/31/2017] [Indexed: 11/24/2022] Open
Abstract
The adaptor protein c-Abl SH3 domain binding protein-2 (3BP2) is tyrosine phosphorylated by Syk in response to cross-linking of antigen receptors, which in turn activates various immune responses. Recently, a study using the mouse model of cherubism, a dominant inherited disorder caused by mutations in the gene encoding 3BP2, showed that 3BP2 is involved in the regulation of phagocytosis mediated by Fc receptor for IgG (FcγR) in macrophages. However, the molecular mechanisms underlying 3BP2-mediated regulation of phagocytosis and the physiological relevance of 3BP2 tyrosine phosphorylation remains elusive. In this study, we established various gene knockout U937 cell lines using the CRISPR/Cas9 system and found that 3BP2 is rapidly tyrosine phosphorylated by Syk in response to cross-linking of FcγRI. Depletion of 3BP2 caused significant reduction in the Fc receptor γ chain (FcRγ)-mediated phagocytosis in addition to the FcγRI-mediated induction of chemokine mRNA for IL-8, CCL3L3 and CCL4L2. Syk-dependent tyrosine phosphorylation of 3BP2 was required for overcoming these defects. Finally, we found that the PH and SH2 domains play important roles on FcγRI-mediated tyrosine phosphorylation of 3BP2 in HL-60 cells. Taken together, these results indicate that Syk-dependent tyrosine phosphorylation of 3BP2 is required for optimal FcRγ-mediated phagocytosis and chemokine expression.
Collapse
|
14
|
Abstract
Phagocytosis refers to the active process that allows cells to take up large particulate material upon binding to surface receptors. The discovery of phagocytosis in 1883 by Elie Metchnikoff, leading to the concept that specialized cells are implicated in the defense against microbes, was one of the starting points of the field of immunology. After more than a century of research, phagocytosis is now appreciated to be a widely used process that enables the cellular uptake of a remarkable variety of particles, including bacteria, fungi, parasites, viruses, dead cells, and assorted debris and solid materials. Uptake of foreign particles is performed almost exclusively by specialized myeloid cells, commonly termed "professional phagocytes": neutrophils, monocytes, macrophages, and dendritic cells. Phagocytosis of microbes not only stops or at least restricts the spread of infection but also plays an important role in regulating the innate and adaptive immune responses. Activation of the myeloid cells upon phagocytosis leads to the secretion of cytokines and chemokines that convey signals to a variety of immune cells. Moreover, foreign antigens generated by the degradation of microbes following phagocytosis are loaded onto the major histocompatibility complex for presentation to specific T lymphocytes. However, phagocytosis is not restricted to professional myeloid phagocytes; an expanding diversity of cell types appear capable of engulfing apoptotic bodies and debris, playing a critical role in tissue remodeling and in the clearance of billions of effete cells every day.
Collapse
|
15
|
Imprime PGG-Mediated Anti-Cancer Immune Activation Requires Immune Complex Formation. PLoS One 2016; 11:e0165909. [PMID: 27812183 PMCID: PMC5094785 DOI: 10.1371/journal.pone.0165909] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/19/2016] [Indexed: 11/19/2022] Open
Abstract
Imprime PGG (Imprime), an intravenously-administered, soluble β-glucan, has shown compelling efficacy in multiple phase 2 clinical trials with tumor targeting or anti-angiogenic antibodies. Mechanistically, Imprime acts as pathogen-associated molecular pattern (PAMP) directly activating innate immune effector cells, triggering a coordinated anti-cancer immune response. Herein, using whole blood from healthy human subjects, we show that Imprime-induced anti-cancer functionality is dependent on immune complex formation with naturally-occurring, anti-β glucan antibodies (ABA). The formation of Imprime-ABA complexes activates complement, primarily via the classical complement pathway, and is opsonized by iC3b. Immune complex binding depends upon Complement Receptor 3 and Fcg Receptor IIa, eliciting phenotypic activation of, and enhanced chemokine production by, neutrophils and monocytes, enabling these effector cells to kill antibody-opsonized tumor cells via the generation of reactive oxygen species and antibody-dependent cellular phagocytosis. Importantly, these innate immune cell changes were not evident in subjects with low ABA levels but could be rescued with exogenous ABA supplementation. Together, these data indicate that pre-existing ABA are essential for Imprime-mediated anti-cancer immune activation and suggest that pre-treatment ABA levels may provide a plausible patient selection biomarker to delineate patients most likely to benefit from Imprime-based therapy.
Collapse
|
16
|
Ricklin D, Reis ES, Lambris JD. Complement in disease: a defence system turning offensive. Nat Rev Nephrol 2016; 12:383-401. [PMID: 27211870 DOI: 10.1038/nrneph.2016.70] [Citation(s) in RCA: 405] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Although the complement system is primarily perceived as a host defence system, a more versatile, yet potentially more harmful side of this innate immune pathway as an inflammatory mediator also exists. The activities that define the ability of the complement system to control microbial threats and eliminate cellular debris - such as sensing molecular danger patterns, generating immediate effectors, and extensively coordinating with other defence pathways - can quickly turn complement from a defence system to an aggressor that drives immune and inflammatory diseases. These host-offensive actions become more pronounced with age and are exacerbated by a variety of genetic factors and autoimmune responses. Complement can also be activated inappropriately, for example in response to biomaterials or transplants. A wealth of research over the past two decades has led to an increasingly finely tuned understanding of complement activation, identified tipping points between physiological and pathological behaviour, and revealed avenues for therapeutic intervention. This Review summarizes our current view of the key activating, regulatory, and effector mechanisms of the complement system, highlighting important crosstalk connections, and, with an emphasis on kidney disease and transplantation, discusses the involvement of complement in clinical conditions and promising therapeutic approaches.
Collapse
Affiliation(s)
- Daniel Ricklin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, 401 Stellar Chance, 422 Curie Boulevard, Philadelphia, Pennsylvania 19104, USA
| | - Edimara S Reis
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, 401 Stellar Chance, 422 Curie Boulevard, Philadelphia, Pennsylvania 19104, USA
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, 401 Stellar Chance, 422 Curie Boulevard, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
17
|
Amash A, Wang L, Wang Y, Bhakta V, Fairn GD, Hou M, Peng J, Sheffield WP, Lazarus AH. CD44 Antibody Inhibition of Macrophage Phagocytosis Targets Fcγ Receptor– and Complement Receptor 3–Dependent Mechanisms. THE JOURNAL OF IMMUNOLOGY 2016; 196:3331-40. [DOI: 10.4049/jimmunol.1502198] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 02/08/2016] [Indexed: 11/19/2022]
|
18
|
Fcγ and Complement Receptors and Complement Proteins in Neutrophil Activation in Rheumatoid Arthritis: Contribution to Pathogenesis and Progression and Modulation by Natural Products. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:429878. [PMID: 26346244 PMCID: PMC4540990 DOI: 10.1155/2015/429878] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 06/30/2015] [Accepted: 07/05/2015] [Indexed: 12/29/2022]
Abstract
Rheumatoid arthritis (RA) is a highly disabling disease that affects all structures of the joint and significantly impacts on morbidity and mortality in RA patients. RA is characterized by persistent inflammation of the synovial membrane lining the joint associated with infiltration of immune cells. Eighty to 90% of the leukocytes infiltrating the synovia are neutrophils. The specific role that neutrophils play in the onset of RA is not clear, but recent studies have evidenced that they have an important participation in joint damage and disease progression through the release of proteolytic enzymes, reactive oxygen species (ROS), cytokines, and neutrophil extracellular traps, in particular during frustrated phagocytosis of immune complexes (ICs). In addition, the local and systemic activation of the complement system contributes to the pathogenesis of RA and other IC-mediated diseases. This review discusses (i) the participation of Fcγ and complement receptors in mediating the effector functions of neutrophils in RA; (ii) the contribution of the complement system and ROS-dependent and ROS-independent mechanisms to joint damage in RA; and (iii) the use of plant extracts, dietary compounds, and isolated natural compounds in the treatment of RA, focusing on modulation of the effector functions of neutrophils and the complement system activity and/or activation.
Collapse
|
19
|
Wu X, Lahiri A, Sarin R, Abraham C. T cell-extrinsic CD18 attenuates antigen-dependent CD4+ T cell activation in vivo. THE JOURNAL OF IMMUNOLOGY 2015; 194:4122-9. [PMID: 25801431 DOI: 10.4049/jimmunol.1401328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 03/04/2015] [Indexed: 12/13/2022]
Abstract
The β2 integrins (CD11/CD18) are heterodimeric leukocyte adhesion molecules expressed on hematopoietic cells. The role of T cell-intrinsic CD18 in trafficking of naive T cells to secondary lymphoid organs and in Ag-dependent T cell activation in vitro and in vivo has been well defined. However, the T cell-extrinsic role for CD18, including on APC, in contributing to T cell activation in vivo is less well understood. We examined the role for T cell-extrinsic CD18 in the activation of wild-type CD4(+) T cells in vivo through the adoptive transfer of DO11.10 Ag-specific CD4(+) T cells into CD18(-/-) mice. We found that T cell-extrinsic CD18 was required for attenuating OVA-induced T cell proliferation in peripheral lymph nodes (PLN). The increased proliferation of wild-type DO11.10 CD4(+) T cells in CD18(-/-) PLN was associated with a higher percentage of APC, and these APC demonstrated an increased activation profile and increased Ag uptake, in particular in F4/80(+) APC. Depletion of F4/80(+) cells both reduced and equalized Ag-dependent T cell proliferation in CD18(-/-) relative to littermate control PLN, demonstrating that these cells play a critical role in the enhanced T cell proliferation in CD18(-/-) mice. Consistently, CD11b blockade, which is expressed on F4/80(+) macrophages, enhanced the proliferation of DO11.10 CD4(+) T cells in CD18(+/-) PLN. Thus, in contrast to the T cell-intrinsic essential role for CD18 in T cell activation, T cell-extrinsic expression of CD18 attenuates Ag-dependent CD4(+) T cell activation in PLN in vivo.
Collapse
Affiliation(s)
- Xingxin Wu
- Department of Internal Medicine, Yale University, New Haven, CT 06510
| | - Amit Lahiri
- Department of Internal Medicine, Yale University, New Haven, CT 06510
| | - Ritu Sarin
- Department of Internal Medicine, Yale University, New Haven, CT 06510
| | - Clara Abraham
- Department of Internal Medicine, Yale University, New Haven, CT 06510
| |
Collapse
|
20
|
Esser-von Bieren J, Volpe B, Kulagin M, Sutherland DB, Guiet R, Seitz A, Marsland BJ, Verbeek JS, Harris NL. Antibody-mediated trapping of helminth larvae requires CD11b and Fcγ receptor I. THE JOURNAL OF IMMUNOLOGY 2014; 194:1154-63. [PMID: 25548226 DOI: 10.4049/jimmunol.1401645] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Infections with intestinal helminths severely impact on human and veterinary health, particularly through the damage that these large parasites inflict when migrating through host tissues. Host immunity often targets the motility of tissue-migrating helminth larvae, which ideally should be mimicked by anti-helminth vaccines. However, the mechanisms of larval trapping are still poorly defined. We have recently reported an important role for Abs in the rapid trapping of tissue-migrating larvae of the murine parasite Heligmosomoides polygyrus bakeri. Trapping was mediated by macrophages (MΦ) and involved complement, activating FcRs, and Arginase-1 (Arg1) activity. However, the receptors and Ab isotypes responsible for MΦ adherence and Arg1 induction remained unclear. Using an in vitro coculture assay of H. polygyrus bakeri larvae and bone marrow-derived MΦ, we now identify CD11b as the major complement receptor mediating MΦ adherence to the larval surface. However, larval immobilization was largely independent of CD11b and instead required the activating IgG receptor FcγRI (CD64) both in vitro and during challenge H. polygyrus bakeri infection in vivo. FcγRI signaling also contributed to the upregulation of MΦ Arg1 expression in vitro and in vivo. Finally, IgG2a/c was the major IgG subtype from early immune serum bound by FcγRI on the MΦ surface, and purified IgG2c could trigger larval immobilization and Arg1 expression in MΦ in vitro. Our findings reveal a novel role for IgG2a/c-FcγRI-driven MΦ activation in the efficient trapping of tissue-migrating helminth larvae and thus provide important mechanistic insights vital for anti-helminth vaccine development.
Collapse
Affiliation(s)
- Julia Esser-von Bieren
- Swiss Vaccine Research Institute, Global Health Institute, Swiss Federal Institute of Technology, Lausanne, 1015 Lausanne, Switzerland
| | - Beatrice Volpe
- Swiss Vaccine Research Institute, Global Health Institute, Swiss Federal Institute of Technology, Lausanne, 1015 Lausanne, Switzerland
| | - Manuel Kulagin
- Swiss Vaccine Research Institute, Global Health Institute, Swiss Federal Institute of Technology, Lausanne, 1015 Lausanne, Switzerland
| | - Duncan B Sutherland
- Swiss Vaccine Research Institute, Global Health Institute, Swiss Federal Institute of Technology, Lausanne, 1015 Lausanne, Switzerland
| | - Romain Guiet
- Bioimaging and Optics Core Facility, Swiss Federal Institute of Technology, Lausanne, 1015 Lausanne, Switzerland
| | - Arne Seitz
- Bioimaging and Optics Core Facility, Swiss Federal Institute of Technology, Lausanne, 1015 Lausanne, Switzerland
| | - Benjamin J Marsland
- Faculty of Biology and Medicine, Respiratory Division, University Hospital, Vaud, University of Lausanne, 1011 Lausanne, Switzerland; and
| | - J Sjef Verbeek
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Nicola L Harris
- Swiss Vaccine Research Institute, Global Health Institute, Swiss Federal Institute of Technology, Lausanne, 1015 Lausanne, Switzerland;
| |
Collapse
|
21
|
Smolny M, Rogers ML, Shafton A, Rush RA, Stebbing MJ. Development of non-viral vehicles for targeted gene transfer into microglia via the integrin receptor CD11b. Front Mol Neurosci 2014; 7:79. [PMID: 25346658 PMCID: PMC4191133 DOI: 10.3389/fnmol.2014.00079] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 09/16/2014] [Indexed: 12/12/2022] Open
Abstract
Microglial activation is a central event in neurodegeneration. Novel technologies are sought for that specifically manipulate microglial function in order to delineate their role in onset and progression of neuropathologies. We investigated for the first time whether non-viral gene delivery based on polyethyleneglycol-polyethyleneimine conjugated to the monoclonal anti-CD11b antibody OX42 ("OX42-immunogene") could be used to specifically target microglia. We first conducted immunofluorescence studies with the OX42 antibody and identified its microglial integrin receptor CD11b as a potential target for receptor-mediated gene transfer based on its cellular specificity in mixed glia culture and in vivo and found that the OX42 antibody is rapidly internalized and trafficked to acidic organelles in absence of activation of the respiratory burst. We then performed transfection experiments with the OX42-immunogene in vitro and in rat brain showing that the OX42-immunogene although internalized was degraded intracellularly and did not cause substantial gene expression in microglia. Investigation of specific barriers to microglial gene transfer revealed that aggregated OX42-immunogene polyplexes stimulated the respiratory burst that likely involved Fcγ-receptors. Transfections in the presence of the endosomolytic agent chloroquine improved transfection efficiency indicating that endosomal escape may be limited. This study identifies CD11b as an entry point for antibody-mediated gene transfer into microglia and takes important steps toward the further development of OX42-immunogenes.
Collapse
Affiliation(s)
- Markus Smolny
- School of Medical Sciences and Health Innovations Research Institute, Royal Melbourne Institute of Technology UniversityBundoora, VIC, Australia
| | - Mary-Louise Rogers
- Department of Human Physiology, Centre for Neuroscience, Flinders UniversityAdelaide, SA, Australia
| | - Anthony Shafton
- The Florey Institute of Neuroscience and Mental Health, The University of MelbourneParkville, VIC, Australia
| | - Robert A. Rush
- Department of Human Physiology, Centre for Neuroscience, Flinders UniversityAdelaide, SA, Australia
| | - Martin J. Stebbing
- School of Medical Sciences and Health Innovations Research Institute, Royal Melbourne Institute of Technology UniversityBundoora, VIC, Australia
| |
Collapse
|
22
|
Jones TB. Lymphocytes and autoimmunity after spinal cord injury. Exp Neurol 2014; 258:78-90. [PMID: 25017889 DOI: 10.1016/j.expneurol.2014.03.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 03/05/2014] [Accepted: 03/06/2014] [Indexed: 12/23/2022]
Abstract
Over the past 15 years an immense amount of data has accumulated regarding the infiltration and activation of lymphocytes in the traumatized spinal cord. Although the impact of the intraspinal accumulation of lymphocytes is still unclear, modulation of the adaptive immune response via active and passive vaccination is being evaluated for its preclinical efficacy in improving the outcome for spinal-injured individuals. The complexity of the interaction between the nervous and the immune systems is highlighted in the contradictions that appear in response to these modulations. Current evidence regarding augmentation and inhibition of the adaptive immune response to spinal cord injury is reviewed with an aim toward reconciling conflicting data and providing consensus issues that may be exploited in future therapies. Opportunities such an approach may provide are highlighted as well as the obstacles that must be overcome before such approaches can be translated into clinical trials.
Collapse
Affiliation(s)
- T Bucky Jones
- Department of Anatomy, Arizona College of Medicine, Midwestern University, Glendale, AZ, USA.
| |
Collapse
|
23
|
van der Heijden J, Nagelkerke S, Zhao X, Geissler J, Rispens T, van den Berg TK, Kuijpers TW. Haplotypes of FcγRIIa and FcγRIIIb polymorphic variants influence IgG-mediated responses in neutrophils. THE JOURNAL OF IMMUNOLOGY 2014; 192:2715-21. [PMID: 24554771 DOI: 10.4049/jimmunol.1203570] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Human blood neutrophils normally express two FcγRs (FcγRIIa and FcγRIIIb) that, upon multivalent binding of IgG in immune complexes or on opsonized targets, mediate responses such as phagocytosis, Ab-dependent cellular cytotoxicity, and respiratory burst. Allelic variants have been described for both FcγRIIa (131H/R) and FcγRIIIb (NA1/NA2/SH), with different binding affinity for IgG subclasses. Because neither of these variants acts alone, we have set out to systematically analyze in a large cohort of healthy FCGR2/3-genotyped volunteers how the different haplotypes of neutrophil FcγRs functionally interact. Maximal IgG-induced H2O2 production by neutrophils from individuals with different (homozygous) haplotypes was observed in the following order: 131HH-NA2NA2 > 131RR-NA1NA1 > 131HH-NA1NA1 > 131RR-NA2NA2. Although FcγRIIa 131H is known to bind IgG1 and IgG2 more avidly, no such differences in affinity are known for FcγRIIIb variants. Nonetheless, a remarkable impact of the FcγRIIIb variants on IgG-mediated neutrophil activity was thus demonstrated, which was not explained by differences in FcγR surface expression. The FcγR expression profile was changed by overnight G-CSF/IFN-γ activation of the neutrophils and eliminated any haplotypic impact on the respiratory burst. To our knowledge, our results are the first to provide an integrated functional analysis of neutrophil FcγR haplotypes and suggest that particularly the early phase of IgG-mediated neutrophil reactivity is influenced by FCGR2/3 genotypic variation.
Collapse
Affiliation(s)
- Joris van der Heijden
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
24
|
Ke Y, Chen Z, Yang R. Yersinia pestis: mechanisms of entry into and resistance to the host cell. Front Cell Infect Microbiol 2013; 3:106. [PMID: 24400226 PMCID: PMC3871965 DOI: 10.3389/fcimb.2013.00106] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 12/10/2013] [Indexed: 12/28/2022] Open
Abstract
During infection, Yersinia, a facultative intracellular bacterial species, exhibits the ability to first invade host cells and then counteract phagocytosis by the host cells. During these two distinct stages, invasion or antiphagocytic factors assist bacteria in manipulating host cells to accomplish each of these functions; however, the mechanism through which Yersinia regulates these functions during each step remains unclear. Here, we discuss those factors that seem to function reversely and give some hypothesis about how bacteria switch between the two distinct status.
Collapse
Affiliation(s)
- Yuehua Ke
- Institute of Disease Control and Prevention, Academy of Military Medical Sciences Beijing, China ; Laboratory of Analytical Microbiology, State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences Beijing, China
| | - Zeliang Chen
- Institute of Disease Control and Prevention, Academy of Military Medical Sciences Beijing, China
| | - Ruifu Yang
- Laboratory of Analytical Microbiology, State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences Beijing, China
| |
Collapse
|
25
|
Taylor RP, Lindorfer MA. The role of complement in mAb-based therapies of cancer. Methods 2013; 65:18-27. [PMID: 23886909 DOI: 10.1016/j.ymeth.2013.07.027] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 07/16/2013] [Indexed: 11/26/2022] Open
Abstract
The ability of complement to promote lysis of antibody-opsonized cells is well-established. Virtually all of the molecular details of this reaction have been elucidated and numerous points of regulation have also been delineated. Use of this information, along with the techniques that were first applied in the fundamental studies of complement, has allowed for investigations of the role of complement in mAb-based immunotherapies of cancer. These studies, which have often combined in vitro investigations with parallel correlative clinical measurements, have revealed that several FDA-approved mAbs make use of complement as an effector function in promoting opsonization and killing of targeted malignant cells. We describe the key methods used in this work, and discuss how the results of these studies provide rational approaches for making more effective use of complement in mAb-based cancer immunotherapy.
Collapse
Affiliation(s)
- Ronald P Taylor
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Margaret A Lindorfer
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
26
|
Vaughn CN, Iafrate JL, Henley JB, Stevenson EK, Shlifer IG, Jones TB. Cellular Neuroinflammation in a Lateral Forceps Compression Model of Spinal Cord Injury. Anat Rec (Hoboken) 2013; 296:1229-46. [DOI: 10.1002/ar.22730] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/31/2013] [Accepted: 05/17/2013] [Indexed: 12/17/2022]
Affiliation(s)
- Chloe N. Vaughn
- Biomedical Sciences Program; Midwestern University; Glendale Arizona
| | - Julia L. Iafrate
- College of Osteopathic Medicine; Midwestern University; Glendale Arizona
| | | | | | - Igor G. Shlifer
- College of Osteopathic Medicine; Midwestern University; Glendale Arizona
| | - T. Bucky Jones
- College of Osteopathic Medicine; Midwestern University; Glendale Arizona
- Department of Anatomy; Midwestern University; Glendale Arizona
| |
Collapse
|
27
|
|
28
|
Bréchard S, Plançon S, Tschirhart EJ. New insights into the regulation of neutrophil NADPH oxidase activity in the phagosome: a focus on the role of lipid and Ca(2+) signaling. Antioxid Redox Signal 2013; 18:661-76. [PMID: 22867131 PMCID: PMC3549206 DOI: 10.1089/ars.2012.4773] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
SIGNIFICANCE Reactive oxygen species, produced by the phagosomal NADPH oxidase of neutrophils, play a significant physiological role during normal defense. Their role is not only to kill invading pathogens, but also to act as modulators of global physiological functions of phagosomes. Given the importance of NADPH oxidase in the immune system, its activity has to be decisively controlled by distinctive mechanisms to ensure appropriate regulation at the phagosome. RECENT ADVANCES Here, we describe the signal transduction pathways that regulate phagosomal NADPH oxidase in neutrophils, with an emphasis on the role of lipid metabolism and intracellular Ca(2+) mobilization. CRITICAL ISSUES The potential involvement of Ca(2+)-binding S100A8 and S100A9 proteins, known to interact with the plasma membrane NADPH oxidase, is also considered. FUTURE DIRECTIONS Recent technical progress in advanced live imaging microscopy will permit to focus more accurately on phagosomal rather than plasma membrane NADPH oxidase regulation during neutrophil phagocytosis.
Collapse
Affiliation(s)
- Sabrina Bréchard
- Calcium Signaling and Inflammation Group, Life Sciences Research Unit, University of Luxembourg, Luxembourg, Luxembourg
| | | | | |
Collapse
|
29
|
Andrade MF, Kabeya LM, Azzolini AECS, Santos EOL, Figueiredo-Rinhel ASG, Paris MRP, Emery FS, Pupo MT, Lucisano-Valim YM. 3-Phenylcoumarin derivatives selectively modulate different steps of reactive oxygen species production by immune complex-stimulated human neutrophils. Int Immunopharmacol 2013; 15:387-94. [PMID: 23333455 DOI: 10.1016/j.intimp.2013.01.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 01/02/2013] [Indexed: 01/19/2023]
Abstract
Immune complex (IC) deposition in tissues triggers the release of harmful oxidant and lytic compounds by neutrophils. We examined how ten 3-phenylcoumarin derivatives affect the reactive oxygen species (ROS) production by IC-stimulated human neutrophils. Most of the 3-phenylcoumarins inhibited the luminol-enhanced chemiluminescence (CL-lum) more strongly than they inhibited the lucigenin-enhanced chemiluminescence (CL-luc), without clear signs of toxicity. The most effective CL-lum inhibitors, 6,7-dihydroxy-3-[3',4'-methylenedioxyphenyl]-coumarin (5) and 6,7-dihydroxy-3-[3',4'-dihydroxyphenyl]-coumarin (19), also inhibited myeloperoxidase activity more potently and had higher hypochlorous acid scavenging ability, but did not affect the NADPH-oxidase activity. The type, number, and position of the substituent influenced the pharmacological effects of 3-phenylcoumarins; however, the structural requirements for CL-lum and CL-luc inhibition were a little different. Compounds 5 and 19 are promising prototypes of therapeutic molecules to modulate ROS production by neutrophils in IC-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Micássio F Andrade
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, Avenida Bandeirantes n. 3900, CEP 14049-900, Ribeirão Preto, SP, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Carter C. Alzheimer's Disease: APP, Gamma Secretase, APOE, CLU, CR1, PICALM, ABCA7, BIN1, CD2AP, CD33, EPHA1, and MS4A2, and Their Relationships with Herpes Simplex, C. Pneumoniae, Other Suspect Pathogens, and the Immune System. Int J Alzheimers Dis 2011; 2011:501862. [PMID: 22254144 PMCID: PMC3255168 DOI: 10.4061/2011/501862] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 09/02/2011] [Indexed: 12/26/2022] Open
Abstract
Alzheimer's disease susceptibility genes, APP and gamma-secretase, are involved in the herpes simplex life cycle, and that of other suspect pathogens (C. pneumoniae, H. pylori, C. neoformans, B. burgdorferri, P. gingivalis) or immune defence. Such pathogens promote beta-amyloid deposition and tau phosphorylation and may thus be causative agents, whose effects are conditioned by genes. The antimicrobial effects of beta-amyloid, the localisation of APP/gamma-secretase in immunocompetent dendritic cells, and gamma secretase cleavage of numerous pathogen receptors suggest that this network is concerned with pathogen disposal, effects which may be abrogated by the presence of beta-amyloid autoantibodies in the elderly. These autoantibodies, as well as those to nerve growth factor and tau, also observed in Alzheimer's disease, may well be antibodies to pathogens, due to homology between human autoantigens and pathogen proteins. NGF or tau antibodies promote beta-amyloid deposition, neurofibrillary tangles, or cholinergic neuronal loss, and, with other autoantibodies, such as anti-ATPase, are potential agents of destruction, whose formation is dictated by sequence homology between pathogen and human proteins, and thus by pathogen strain and human genes. Pathogen elimination in the ageing population and removal of culpable autoantibodies might reduce the incidence and offer hope for a cure in this affliction.
Collapse
Affiliation(s)
- Chris Carter
- PolygenicPathways, Flat 2, 40 Baldslow Road, Hastings, East Sussex TN34 2EY, UK
| |
Collapse
|
31
|
Alvarez Y, Municio C, Hugo E, Zhu J, Alonso S, Hu X, Fernández N, Sánchez Crespo M. Notch- and transducin-like enhancer of split (TLE)-dependent histone deacetylation explain interleukin 12 (IL-12) p70 inhibition by zymosan. J Biol Chem 2011; 286:16583-95. [PMID: 21402701 DOI: 10.1074/jbc.m111.222158] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The fungal analog zymosan induces IL-23 and low amounts of IL-12 p70. This study addresses the molecular mechanisms underlying this cytokine pattern in human monocyte-derived dendritic cells. The transcriptional regulation of il23a, one of the chains of IL-23, depended on the activation of c-Rel and histone H3 phosphorylation, as judged from the association of c-Rel with the il23a promoter and the correlation between IL-23 production and Ser-10-histone H3 phosphorylation. Consistent with its reduced ability to produce IL-12 p70, zymosan induced a transient occupancy of the il12a promoter by c-Rel, blocked the production of IL-12 p70 and the transcription of il12a induced by other stimuli, and triggered the expression and nuclear translocation of the transcriptional repressors of the Notch family hairy and enhancer of split (Hes)-1, Hes5, hairy/enhancer-of-split related with YRPW motif protein (Hey)-1, and transducin-like enhancer of split (TLE). Zymosan also induced the interaction of Hes1 and TLE with histone H3 phosphorylated on Ser-10 and deacetylated on Lys-14. Inhibition of class III histone deacetylases increased the production of IL-12 p70 and partially blunted the inhibitory effect of zymosan on the production of IL-12 p70 elicited by LPS and IFN-γ. These results indicate that the selective induction of IL-23 by β-glucans is explained by the activation of c-Rel associated with Ser-10-histone H3 phosphorylation in the il23a promoter mediated by mitogen- and stress-activated kinase and/or protein kinase A and inhibition of il12a transcription by a mechanism involving activation of several corepressors with the ability to bind TLE and to promote histone deacetylation.
Collapse
Affiliation(s)
- Yolanda Alvarez
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, 47003-Valladolid, Spain
| | | | | | | | | | | | | | | |
Collapse
|