1
|
Qi X, Fang Z, Meng L, Xiang X, Ju Y, Cai X, An T, Sun M, Wang H. The C3d-fused Porcine circovirus type 2d virus-like particle induced early and enhanced immune response and protected pigs against challenge. Vet Microbiol 2024; 299:110305. [PMID: 39536690 DOI: 10.1016/j.vetmic.2024.110305] [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: 07/04/2024] [Revised: 11/05/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Porcine circovirus type 2 (PCV2) is an economically significant pathogen affecting the global swine industry. Vaccination is considered the most effective and best way to prevent PCV2-associated disease. The PCV2d genotype has become predominant by replacing the previous PCV2b genotype. The potential increase in the virulence of PCV2d has drawn attention, spurring the development of PCV2d vaccines. Virus-like particle (VLP) is an ideal vaccine candidate for its safety and potent immunogenicity. C3d is a molecular adjuvant that can be used to promote the protective efficacy of the PCV2 vaccine. In this study, we expressed PCV2d Cap protein fused with C3d epitope using E. coli expression system. The purified recombinant Cap protein assembled into VLP, which was designated as PCV2d-C3d-VLP. Through assessments in mice and piglets, we demonstrated that the PCV2d-C3d-VLP elicited robust humoral responses, notably accelerating antibody production one week earlier compared to a commercial PCV2d subunit vaccine. Furthermore, vaccination substantially reduced PCV2d viral load in piglets. These results present an innovative strategy for developing a more efficacious and cost-effective PCV2d VLP vaccine.
Collapse
Affiliation(s)
- Xinyu Qi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Zheng Fang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Liang Meng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Xuyan Xiang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Yan Ju
- Department of research and development, Harbin Weike Biotechnology Co., LTD, Harbin 150069, China
| | - Xuehui Cai
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China; Harbin Veterinary Research Institute, Heilongjiang Provincial Research Center for Veterinary Biomedicine, Harbin, China
| | - Tongqing An
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China; Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin 150069, China
| | - Mingxia Sun
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Haiwei Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China; Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin 150069, China.
| |
Collapse
|
2
|
Saxena R, Gottlin EB, Campa MJ, Bushey RT, Guo J, Patz EF, He YW. Complement factor H: a novel innate immune checkpoint in cancer immunotherapy. Front Cell Dev Biol 2024; 12:1302490. [PMID: 38389705 PMCID: PMC10883309 DOI: 10.3389/fcell.2024.1302490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/08/2024] [Indexed: 02/24/2024] Open
Abstract
The elimination of cancer cells critically depends on the immune system. However, cancers have evolved a variety of defense mechanisms to evade immune monitoring, leading to tumor progression. Complement factor H (CFH), predominately known for its function in inhibiting the alternative pathway of the complement system, has recently been identified as an important innate immunological checkpoint in cancer. CFH-mediated immunosuppression enhances tumor cells' ability to avoid immune recognition and produce an immunosuppressive tumor microenvironment. This review explores the molecular underpinnings, interactions with immune cells, clinical consequences, and therapeutic possibilities of CFH as an innate immune checkpoint in cancer control. The difficulties and opportunities of using CFH as a target in cancer immunotherapy are also explored.
Collapse
Affiliation(s)
- Ruchi Saxena
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, United States
| | - Elizabeth B Gottlin
- Department of Radiology, Duke University School of Medicine, Durham, NC, United States
| | - Michael J Campa
- Department of Radiology, Duke University School of Medicine, Durham, NC, United States
| | - Ryan T Bushey
- Department of Radiology, Duke University School of Medicine, Durham, NC, United States
| | - Jian Guo
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, United States
| | - Edward F Patz
- Department of Radiology, Duke University School of Medicine, Durham, NC, United States
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, United States
| | - You-Wen He
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
3
|
Loh JM, Aghababa H, Proft T. Eluding the immune system's frontline defense: Secreted complement evasion factors of pathogenic Gram-positive cocci. Microbiol Res 2023; 277:127512. [PMID: 37826985 DOI: 10.1016/j.micres.2023.127512] [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: 08/23/2023] [Revised: 10/01/2023] [Accepted: 10/04/2023] [Indexed: 10/14/2023]
Abstract
The human complement system is an important part of the innate immune response in the fight against invasive bacteria. Complement responses can be activated independently by the classical pathway, the lectin pathway, or the alternative pathway, each resulting in the formation of a C3 convertase that produces the anaphylatoxin C3a and the opsonin C3b by specifically cutting C3. Other important features of complement are the production of the chemotactic C5a peptide and the generation of the membrane attack complex to lyse intruding pathogens. Invasive pathogens like Staphylococcus aureus and several species of the genus Streptococcus have developed a variety of complement evasion strategies to resist complement activity thereby increasing their virulence and potential to cause disease. In this review, we focus on secreted complement evasion factors that assist the bacteria to avoid opsonization and terminal pathway lysis. We also briefly discuss the potential role of complement evasion factors for the development of vaccines and therapeutic interventions.
Collapse
Affiliation(s)
- Jacelyn Ms Loh
- Department of Molecular Medicine & Pathology, School of Medical Sciences, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Haniyeh Aghababa
- Department of Molecular Medicine & Pathology, School of Medical Sciences, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Thomas Proft
- Department of Molecular Medicine & Pathology, School of Medical Sciences, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand.
| |
Collapse
|
4
|
Kim HW, Ko MK, Park SH, Shin S, Kim SM, Park JH, Lee MJ. Bestatin, A Pluripotent Immunomodulatory Small Molecule, Drives Robust and Long-Lasting Immune Responses as an Adjuvant in Viral Vaccines. Vaccines (Basel) 2023; 11:1690. [PMID: 38006022 PMCID: PMC10675184 DOI: 10.3390/vaccines11111690] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/24/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
An inactivated whole-virus vaccine is currently used to prevent foot-and-mouth disease (FMD). Although this vaccine is effective, it offers short-term immunity that requires regular booster immunizations and has several side effects, including local reactions at the vaccination site. To address these limitations, herein, we evaluated the efficacy of bestatin as a novel small molecule adjuvant for inactivated FMD vaccines. Our findings showed that the FMD vaccine formulated with bestatin enhanced early, intermediate-, and particularly long-term immunity in experimental animals (mice) and target animals (pigs). Furthermore, cytokines (interferon (IFN)α, IFNβ, IFNγ, and interleukin (IL)-29), retinoic acid-inducible gene (RIG)-I, and T-cell and B-cell core receptors (cluster of differentiation (CD)28, CD19, CD21, and CD81) markedly increased in the group that received the FMD vaccine adjuvanted with bestatin in pigs compared with the control. These results indicate the significant potential of bestatin to improve the efficacy of inactivated FMD vaccines in terms of immunomodulatory function for the simultaneous induction of potent cellular and humoral immune response and a long-lasting memory response.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Min Ja Lee
- Center for Foot-and-Mouth Disease Vaccine Research, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon-si 39660, Gyeongsangbuk-do, Republic of Korea; (H.W.K.); (M.-K.K.); (S.H.P.); (S.S.); (S.-M.K.); (J.-H.P.)
| |
Collapse
|
5
|
C3d(g), iron nanoparticles, hemin and cytochrome c may induce oxidative cytotoxicity in tumors and reduce tumor-associated myeloid cells-mediated immunosuppression. Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
6
|
Rohrer B, Parsons N, Annamalai B, Nicholson C, Obert E, Jones BW, Dick AD. Peptide-based immunotherapy against oxidized elastin ameliorates pathology in mouse model of smoke-induced ocular injury. Exp Eye Res 2021; 212:108755. [PMID: 34487725 PMCID: PMC9753162 DOI: 10.1016/j.exer.2021.108755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/24/2021] [Accepted: 08/30/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Age-related macular degeneration (AMD), the leading cause of blindness in western populations, is associated with an overactive complement system, and an increase in circulating antibodies against certain epitopes, including elastin. As loss of the elastin layer of Bruch's membrane (BrM) has been reported in aging and AMD, we previously showed that immunization with elastin peptide oxidatively modified by cigarette smoke (ox-elastin), exacerbated ocular pathology in the smoke-induced ocular pathology (SIOP) model. Here we asked whether ox-elastin peptide-based immunotherapy (PIT) ameliorates damage. METHODS C57BL/6J mice were injected with ox-elastin peptide at two doses via weekly subcutaneous administration, while exposed to cigarette smoke for 6 months. FcγR-/- and uninjected C57BL/6J mice served as controls. Retinal morphology was assessed by electron microscopy, and complement activation, antibody deposition and mechanisms of immunological tolerance were assessed by Western blotting and ELISA. RESULTS Elimination of Fcγ receptors, preventing antigen/antibody-dependent cytotoxicity, protected against SIOP. Mice receiving PIT with low dose ox-elastin (LD-PIT) exhibited reduced humoral immunity, reduced complement activation and IgG/IgM deposition in the RPE/choroid, and largely a preserved BrM. While there is no direct evidence of ox-elastin pathogenicity, LD-PIT reduced IFNγ and increased IL-4 within RPE/choroid. High dose PIT was not protective. CONCLUSIONS These data further support ox-elastin role in ocular damage in part via elastin-specific antibodies, and support the corollary that PIT with ox-elastin attenuates ocular pathology. Overall, damage is associated with complement activation, antibody-dependent cell-mediated cytotoxicity, and altered cytokine signature.
Collapse
Affiliation(s)
- Bärbel Rohrer
- Departments of Ophthalmology and Neurosciences Division of Research, Medical University of South Carolina, Charleston, SC, USA; Departments of Neurosciences Division of Research, Medical University of South Carolina, Charleston, SC, USA; Departments of Ralph H. Johnson VA Medical Center, Division of Research, Charleston, SC, 29401, USA.
| | - Nathaniel Parsons
- Departments of Ophthalmology and Neurosciences Division of Research, Medical University of South Carolina, Charleston, SC, USA
| | - Balasubramaniam Annamalai
- Departments of Ophthalmology and Neurosciences Division of Research, Medical University of South Carolina, Charleston, SC, USA
| | - Crystal Nicholson
- Departments of Ophthalmology and Neurosciences Division of Research, Medical University of South Carolina, Charleston, SC, USA
| | - Elisabeth Obert
- Departments of Ophthalmology and Neurosciences Division of Research, Medical University of South Carolina, Charleston, SC, USA
| | - Bryan W Jones
- Department of Ophthalmology, University of Utah, Salt Lake City, UT, 84132, USA
| | - Andrew D Dick
- University of Bristol, Bristol BS8 1TD, UK and University College London-Institute of Ophthalmology and the National Institute for Health Research Biomedical Research Centre, Moorfields Eye Hospital, London, EC1V 9EL, UK.
| |
Collapse
|
7
|
Rosser C, Sage D. Approaches for the characterization of clinically relevant pre-transplant human leucocyte antigen (HLA) antibodies in solid organ transplant patients. Int J Immunogenet 2021; 48:385-402. [PMID: 34346180 DOI: 10.1111/iji.12552] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/30/2021] [Accepted: 07/22/2021] [Indexed: 11/27/2022]
Abstract
The avoidance of antibody-mediated rejection (AMR) attributed to human leucocyte antigen (HLA) antibody incompatibility remains an essential function of clinical Histocompatibility and Immunogenetics (H&I) laboratories who are supporting solid organ transplantation. Developments in HLA antibody identification assays over the past thirty years have greatly reduced unexpected positive cellular crossmatches and improved solid organ transplant outcomes. For sensitized patients, the decision to register unacceptable HLA antigen mismatches is often heavily influenced by results from solid phase antibody assays, particularly the Luminex® Single Antigen Bead (SAB) assays, although the clinical relevance of antibodies identified solely by these assays remains unclear. As such, the identification of non-clinically relevant antibodies may proportionally increase the number of unacceptable transplant mismatches registered, with an associated increase in waiting time for a compatible organ. We reflect on the clinical relevance of antibodies identified solely by the Luminex SAB® assays and consider whether the application of additional assays and/or tools could further develop our ability to define the clinical relevance of antibodies identified in patient sera. Improvements in this area would assist equity of access to a compatible transplant for highly sensitized patients awaiting a solid organ transplant.
Collapse
Affiliation(s)
- Carla Rosser
- NHS Blood and Transplant (Tooting), Histocompatibility and Immunogenetics, London, UK
| | - Deborah Sage
- NHS Blood and Transplant (Tooting), Histocompatibility and Immunogenetics, London, UK
| |
Collapse
|
8
|
Abstract
Complement protein C3dg, a key linkage between innate and adaptive immunity, is capable of stimulating both humoral and cell-mediated immune responses, leading to considerable interest in its use as a molecular adjuvant. However, the potential of C3dg as an adjuvant is limited without ways of controllably assembling multiple copies of it into vaccine platforms. Here, we report a strategy to assemble C3dg into supramolecular nanofibers with excellent compositional control, using β-tail fusion tags. These assemblies were investigated as therapeutic active immunotherapies, which may offer advantages over existing biologics, particularly toward chronic inflammatory diseases. Supramolecular assemblies based on the Q11 peptide system containing β-tail-tagged C3dg, B cell epitopes from TNF, and the universal T cell epitope PADRE raised strong antibody responses against both TNF and C3dg, and prophylactic immunization with these materials significantly improved protection in a lethal TNF-mediated inflammation model. Additionally, in a murine model of psoriasis induced by imiquimod, the C3dg-adjuvanted nanofiber vaccine performed as well as anti-TNF monoclonal antibodies. Nanofibers containing only β-tail-C3dg and lacking the TNF B cell epitope also showed improvements in both models, suggesting that supramolecular C3dg, by itself, played an important therapeutic role. We observed that immunization with β-tail-C3dg caused the expansion of an autoreactive C3dg-specific T cell population, which may act to dampen the immune response, preventing excessive inflammation. These findings indicate that molecular assemblies displaying C3dg warrant further development as active immunotherapies.
Collapse
|
9
|
Mellors J, Tipton T, Longet S, Carroll M. Viral Evasion of the Complement System and Its Importance for Vaccines and Therapeutics. Front Immunol 2020; 11:1450. [PMID: 32733480 PMCID: PMC7363932 DOI: 10.3389/fimmu.2020.01450] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/04/2020] [Indexed: 12/17/2022] Open
Abstract
The complement system is a key component of innate immunity which readily responds to invading microorganisms. Activation of the complement system typically occurs via three main pathways and can induce various antimicrobial effects, including: neutralization of pathogens, regulation of inflammatory responses, promotion of chemotaxis, and enhancement of the adaptive immune response. These can be vital host responses to protect against acute, chronic, and recurrent viral infections. Consequently, many viruses (including dengue virus, West Nile virus and Nipah virus) have evolved mechanisms for evasion or dysregulation of the complement system to enhance viral infectivity and even exacerbate disease symptoms. The complement system has multifaceted roles in both innate and adaptive immunity, with both intracellular and extracellular functions, that can be relevant to all stages of viral infection. A better understanding of this virus-host interplay and its contribution to pathogenesis has previously led to: the identification of genetic factors which influence viral infection and disease outcome, the development of novel antivirals, and the production of safer, more effective vaccines. This review will discuss the antiviral effects of the complement system against numerous viruses, the mechanisms employed by these viruses to then evade or manipulate this system, and how these interactions have informed vaccine/therapeutic development. Where relevant, conflicting findings and current research gaps are highlighted to aid future developments in virology and immunology, with potential applications to the current COVID-19 pandemic.
Collapse
Affiliation(s)
- Jack Mellors
- Public Health England, National Infection Service, Salisbury, United Kingdom.,Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Tom Tipton
- Public Health England, National Infection Service, Salisbury, United Kingdom
| | - Stephanie Longet
- Public Health England, National Infection Service, Salisbury, United Kingdom
| | - Miles Carroll
- Public Health England, National Infection Service, Salisbury, United Kingdom
| |
Collapse
|
10
|
Nguyen H, Alawieh A, Bastian D, Kuril S, Dai M, Daenthanasanmak A, Zhang M, Iamsawat S, Schutt SD, Wu Y, Sleiman MM, Shetty A, Atkinson C, Sun S, Varela JC, Tomlinson S, Yu XZ. Targeting the Complement Alternative Pathway Permits Graft Versus Leukemia Activity while Preventing Graft Versus Host Disease. Clin Cancer Res 2020; 26:3481-3490. [PMID: 31919135 PMCID: PMC7334060 DOI: 10.1158/1078-0432.ccr-19-1717] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 10/03/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022]
Abstract
PURPOSE Application of allogeneic hematopoietic cell transplantation (allo-HCT) for patients with hematologic disorders is limited by the development of GVHD. Separation of GVHD and graft-versus-leukemia (GVL) remains a great challenge in the field. We investigated the contribution of individual pathways involved in the complement cascade in GVH and GVL responses to identify specific targets by which to separate these two processes. EXPERIMENTAL DESIGN We used multiple preclinical murine and human-to-mouse xenograft models involving allo-HCT recipients lacking components of the alternative pathway (AP) or classical pathway (CP)/lectin pathway (LP) to dissect the role of each individual pathway in GVHD pathogenesis and the GVL effect. For translational purposes, we used the AP-specific complement inhibitor, CR2-fH, which localizes in injured target organs to allow specific blockade of complement activation at sites of inflammation. RESULTS Complement deposition was evident in intestines of mice and patients with GVHD. In a preclinical setting, ablation of the AP, but not the CP/LP, significantly improved GVHD outcomes. Complement activation through the AP in host hematopoietic cells, and specifically dendritic cells (DC), was required for GVHD progression. AP deficiency in recipients decreased donor T-cell migration and Th1/Th2 differentiation, while increasing the generation of regulatory T cells. This was because of decreased activation and stimulatory activity of recipient DCs in GVHD target organs. Treatment with CR2-fH effectively prevented GVHD while preserving GVL activity. CONCLUSIONS This study highlights the AP as a new therapeutic target to prevent GVHD and tumor relapse after allo-HCT. Targeting the AP by CR2-fH represents a promising therapeutic approach for GVHD treatment.
Collapse
Affiliation(s)
- Hung Nguyen
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina.
| | - Ali Alawieh
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
- Medical Scientist Training Program, College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - David Bastian
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Sandeepkumar Kuril
- Department of Pediatric, Medical University of South Carolina, Charleston, South Carolina
| | - Min Dai
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Anusara Daenthanasanmak
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Mengmeng Zhang
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Supinya Iamsawat
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Steven D Schutt
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Yongxia Wu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - M Mahdi Sleiman
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Akshay Shetty
- Department of Pathology, Medical University of South Carolina, Charleston, South Carolina
| | - Carl Atkinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Shaoli Sun
- Department of Pathology, Medical University of South Carolina, Charleston, South Carolina
| | - Juan Carlos Varela
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
- Ralph H. Johnson Veterans Affairs Medical Center, Medical University of South Carolina, Charleston, South Carolina
| | - Xue-Zhong Yu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina.
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
11
|
Holland-Tummillo KM, Shoudy LE, Steiner D, Kumar S, Rosa SJ, Namjoshi P, Singh A, Sellati TJ, Gosselin EJ, Hazlett KRO. Autotransporter-Mediated Display of Complement Receptor Ligands by Gram-Negative Bacteria Increases Antibody Responses and Limits Disease Severity. Pathogens 2020; 9:E375. [PMID: 32422907 PMCID: PMC7281241 DOI: 10.3390/pathogens9050375] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/06/2020] [Accepted: 05/11/2020] [Indexed: 12/13/2022] Open
Abstract
The targeting of immunogens/vaccines to specific immune cells is a promising approach for amplifying immune responses in the absence of exogenous adjuvants. However, the targeting approaches reported thus far require novel, labor-intensive reagents for each vaccine and have primarily been shown as proof-of-concept with isolated proteins and/or inactivated bacteria. We have engineered a plasmid-based, complement receptor-targeting platform that is readily applicable to live forms of multiple gram-negative bacteria, including, but not limited to, Escherichia coli, Klebsiella pneumoniae, and Francisella tularensis. Using F. tularensis as a model, we find that targeted bacteria show increased binding and uptake by macrophages, which coincides with increased p38 and p65 phosphorylation. Mice vaccinated with targeted bacteria produce higher titers of specific antibody that recognizes a greater diversity of bacterial antigens. Following challenge with homologous or heterologous isolates, these mice exhibited less weight loss and/or accelerated weight recovery as compared to counterparts vaccinated with non-targeted immunogens. Collectively, these findings provide proof-of-concept for plasmid-based, complement receptor-targeting of live gram-negative bacteria.
Collapse
Affiliation(s)
- Kristen M Holland-Tummillo
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (K.M.H.-T.); (D.S.); (S.K.); (S.J.R.); (P.N.); (E.J.G.)
| | - Lauren E Shoudy
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208, USA;
| | - Donald Steiner
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (K.M.H.-T.); (D.S.); (S.K.); (S.J.R.); (P.N.); (E.J.G.)
| | - Sudeep Kumar
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (K.M.H.-T.); (D.S.); (S.K.); (S.J.R.); (P.N.); (E.J.G.)
| | - Sarah J Rosa
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (K.M.H.-T.); (D.S.); (S.K.); (S.J.R.); (P.N.); (E.J.G.)
| | - Prachi Namjoshi
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (K.M.H.-T.); (D.S.); (S.K.); (S.J.R.); (P.N.); (E.J.G.)
| | - Anju Singh
- Department of Infectious Disease, Southern Research Institute, Birmingham, AL 35211, USA; (A.S.); (T.J.S.)
| | - Timothy J Sellati
- Department of Infectious Disease, Southern Research Institute, Birmingham, AL 35211, USA; (A.S.); (T.J.S.)
| | - Edmund J Gosselin
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (K.M.H.-T.); (D.S.); (S.K.); (S.J.R.); (P.N.); (E.J.G.)
| | - Karsten RO Hazlett
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (K.M.H.-T.); (D.S.); (S.K.); (S.J.R.); (P.N.); (E.J.G.)
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208, USA;
| |
Collapse
|
12
|
De Groot AS, Moise L, Terry F, Gutierrez AH, Hindocha P, Richard G, Hoft DF, Ross TM, Noe AR, Takahashi Y, Kotraiah V, Silk SE, Nielsen CM, Minassian AM, Ashfield R, Ardito M, Draper SJ, Martin WD. Better Epitope Discovery, Precision Immune Engineering, and Accelerated Vaccine Design Using Immunoinformatics Tools. Front Immunol 2020; 11:442. [PMID: 32318055 PMCID: PMC7154102 DOI: 10.3389/fimmu.2020.00442] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 02/26/2020] [Indexed: 12/19/2022] Open
Abstract
Computational vaccinology includes epitope mapping, antigen selection, and immunogen design using computational tools. Tools that facilitate the in silico prediction of immune response to biothreats, emerging infectious diseases, and cancers can accelerate the design of novel and next generation vaccines and their delivery to the clinic. Over the past 20 years, vaccinologists, bioinformatics experts, and advanced programmers based in Providence, Rhode Island, USA have advanced the development of an integrated toolkit for vaccine design called iVAX, that is secure and user-accessible by internet. This integrated set of immunoinformatic tools comprises algorithms for scoring and triaging candidate antigens, selecting immunogenic and conserved T cell epitopes, re-engineering or eliminating regulatory T cell epitopes, and re-designing antigens to induce immunogenicity and protection against disease for humans and livestock. Commercial and academic applications of iVAX have included identifying immunogenic T cell epitopes in the development of a T-cell based human multi-epitope Q fever vaccine, designing novel influenza vaccines, identifying cross-conserved T cell epitopes for a malaria vaccine, and analyzing immune responses in clinical vaccine studies. Animal vaccine applications to date have included viral infections of pigs such as swine influenza A, PCV2, and African Swine Fever. “Rapid-Fire” applications for biodefense have included a demonstration project for Lassa Fever and Q fever. As recent infectious disease outbreaks underscore the significance of vaccine-driven preparedness, the integrated set of tools available on the iVAX toolkit stand ready to help vaccine developers deliver genome-derived, epitope-driven vaccines.
Collapse
Affiliation(s)
- Anne S De Groot
- EpiVax, Inc., Providence, RI, United States.,Institute for Immunology and Informatics, Providence, RI, United States
| | - Leonard Moise
- EpiVax, Inc., Providence, RI, United States.,Institute for Immunology and Informatics, Providence, RI, United States
| | | | - Andres H Gutierrez
- EpiVax, Inc., Providence, RI, United States.,Institute for Immunology and Informatics, Providence, RI, United States
| | | | | | - Daniel Fredric Hoft
- Departments of Molecular Microbiology & Immunology and Internal Medicine, Division of Infectious Diseases, Allergy & Immunology, Saint Louis University, St. Louis, MO, United States
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States
| | - Amy R Noe
- Leidos Life Sciences, Frederick, MD, United States
| | | | | | - Sarah E Silk
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | | | | | | - Simon J Draper
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
13
|
Yang Y, Back CR, Gräwert MA, Wahid AA, Denton H, Kildani R, Paulin J, Wörner K, Kaiser W, Svergun DI, Sartbaeva A, Watts AG, Marchbank KJ, van den Elsen JMH. Utilization of Staphylococcal Immune Evasion Protein Sbi as a Novel Vaccine Adjuvant. Front Immunol 2019; 9:3139. [PMID: 30687332 PMCID: PMC6336717 DOI: 10.3389/fimmu.2018.03139] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 12/19/2018] [Indexed: 02/04/2023] Open
Abstract
Co-ligation of the B cell antigen receptor with complement receptor 2 on B-cells via a C3d-opsonised antigen complex significantly lowers the threshold required for B cell activation. Consequently, fusions of antigens with C3d polymers have shown great potential in vaccine design. However, these linear arrays of C3d multimers do not mimic the natural opsonisation of antigens with C3d. Here we investigate the potential of using the unique complement activating characteristics of Staphylococcal immune-evasion protein Sbi to develop a pro-vaccine approach that spontaneously coats antigens with C3 degradation products in a natural way. We show that Sbi rapidly triggers the alternative complement pathway through recruitment of complement regulators, forming tripartite complexes that act as competitive antagonists of factor H, resulting in enhanced complement consumption. These functional results are corroborated by the structure of the complement activating Sbi-III-IV:C3d:FHR-1 complex. Finally, we demonstrate that Sbi, fused with Mycobacterium tuberculosis antigen Ag85b, causes efficient opsonisation with C3 fragments, thereby enhancing the immune response significantly beyond that of Ag85b alone, providing proof of concept for our pro-vaccine approach.
Collapse
Affiliation(s)
- Yi Yang
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Catherine R Back
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Melissa A Gräwert
- Hamburg Unit, European Molecular Biology Laboratory, Deutsches Elektronen-Synchrotron, Hamburg, Germany
| | - Ayla A Wahid
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Harriet Denton
- Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Rebecca Kildani
- Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Joshua Paulin
- Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | | | | | - Dmitri I Svergun
- Hamburg Unit, European Molecular Biology Laboratory, Deutsches Elektronen-Synchrotron, Hamburg, Germany
| | - Asel Sartbaeva
- Department of Chemistry, University of Bath, Bath, United Kingdom
| | - Andrew G Watts
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | - Kevin J Marchbank
- Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | | |
Collapse
|
14
|
Zhang Y, Guo J, Ning L, Tian J, Yao X, Liu H. The molecular mechanism of pH-regulating C3d-CR2 interactions: Insights from molecular dynamics simulation. Chem Biol Drug Des 2018; 93:628-637. [PMID: 30566277 DOI: 10.1111/cbdd.13460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/09/2018] [Accepted: 12/07/2018] [Indexed: 11/27/2022]
Abstract
The interactions of complement receptor 2 (CR2) and the degradation fragment C3d of complement component C3 mediate the innate and adaptive immune systems. Due to the importance of C3d-CR2 interaction in the design of vaccines, many studies have indicated the interactions are pH-dependent. Moreover, C3d-CR2 interactions at pH 5.0 are unknown. To investigate the molecular mechanism of pH-regulating C3d-CR2 interaction, molecular dynamics simulations for C3d-CR2 complex in different pH are performed. Our results revealed that the protonation of His9 in C3d at pH 6.0 slightly weakens C3d-CR2 association as reducing pH from 7.4 to 6.0, initiated from a key hydrogen bond formed between Gly270 and His9 in C3d at pH 6.0. When reducing pH from 6.0 to 5.0, the protonation of His33 in C3d weakens C3d-SCR1 association by changing the hydrogen-bond network of Asp36, Glu37, and Glu39 in C3d with Arg13 in CR2. In addition, the protonation of His90 significantly enhances C3d-SCR2 association. This is because the enhanced hydrogen-bond interactions of His90 with Glu63 and Ser69 of the linker change the conformations of the linker, Cys112-Asn116 and Pro87-Gly91 regions. This study uncovers the molecular mechanism of the mediation of pH on C3d-CR2 interaction, which is valuable for vaccine design.
Collapse
Affiliation(s)
- Yan Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, China.,Guiyang College of Traditional Chinese Medicine, Guiyang, China.,State Key Laboratory of Applied Organic Chemistry, Department of Chemistry, Lanzhou University, Lanzhou, China
| | - Jingjing Guo
- Henan Engineering Research Center of Chiral Hydroxyl Pharmaceutical, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, China
| | - Lulu Ning
- State Key Laboratory of Applied Organic Chemistry, Department of Chemistry, Lanzhou University, Lanzhou, China
| | - Jiaqi Tian
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Xiaojun Yao
- State Key Laboratory of Applied Organic Chemistry, Department of Chemistry, Lanzhou University, Lanzhou, China
| | - Huanxiang Liu
- School of Pharmacy, Lanzhou University, Lanzhou, China.,State Key Laboratory of Applied Organic Chemistry, Department of Chemistry, Lanzhou University, Lanzhou, China
| |
Collapse
|
15
|
He YG, Pappworth IY, Rossbach A, Paulin J, Mavimba T, Hayes C, Kulik L, Holers VM, Knight AM, Marchbank KJ. A novel C3d-containing oligomeric vaccine provides insight into the viability of testing human C3d-based vaccines in mice. Immunobiology 2018; 223:125-134. [PMID: 29017821 PMCID: PMC5849677 DOI: 10.1016/j.imbio.2017.10.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 10/03/2017] [Indexed: 11/30/2022]
Abstract
The use of C3d, the final degradation product of complement protein C3, as a "natural" adjuvant has been widely examined since the initial documentation of its immunogenicity-enhancing properties as a consequence of binding to complement receptor 2. Subsequently it was demonstrated that these effects are most evident when oligomeric, rather than when monomeric forms of C3d, are linked to various test protein antigens. In this study, we examined the feasibility of enhancing the adjuvant properties of human C3d further by utilizing C4b-binding protein (C4BP) to provide an oligomeric arrayed scaffold fused to the model antigen, tetanus toxin C fragment (TTCF). High molecular weight, C3d-containing oligomeric vaccines were successfully expressed, purified from mammalian cells and used to immunize groups of mice. Surprisingly, anti-TTCF antibody responses measured in these mice were poor. Subsequently we established by in vitro and in vivo analysis that, in the presence of mouse C3, human C3d does not interact with either mouse or even human complement receptor 2. These data confirm the requirement to develop murine versions of C3d based adjuvant compounds to test in mice or that mice would need to be developed that express both human C3 and human CR2 to allow the testing of human C3d based adjuvants in mouse in any capacity.
Collapse
Affiliation(s)
- Yong-Gang He
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle-upon-Tyne, NE2 4HH, UK
| | - Isabel Y Pappworth
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle-upon-Tyne, NE2 4HH, UK
| | | | - Joshua Paulin
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle-upon-Tyne, NE2 4HH, UK.
| | - Tarirai Mavimba
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle-upon-Tyne, NE2 4HH, UK
| | - Christine Hayes
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle-upon-Tyne, NE2 4HH, UK
| | - Liudmila Kulik
- Departments of Medicine and Immunology, University of Colorado, SOM, Denver, CO, USA
| | - V Michael Holers
- Departments of Medicine and Immunology, University of Colorado, SOM, Denver, CO, USA
| | - Andrew M Knight
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle-upon-Tyne, NE2 4HH, UK; School of Biomedical Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle-upon-Tyne, NE2 4HH, UK
| | - Kevin J Marchbank
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle-upon-Tyne, NE2 4HH, UK.
| |
Collapse
|
16
|
Platt JL, Silva I, Balin SJ, Lefferts AR, Farkash E, Ross TM, Carroll MC, Cascalho M. C3d regulates immune checkpoint blockade and enhances antitumor immunity. JCI Insight 2017; 2:90201. [PMID: 28469081 PMCID: PMC5414554 DOI: 10.1172/jci.insight.90201] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 04/04/2017] [Indexed: 01/27/2023] Open
Abstract
Despite expression of immunogenic polypeptides, tumors escape immune surveillance by engaging T cell checkpoint regulators and expanding Tregs, among other mechanisms. What orchestrates these controls is unknown. We report that free C3d, a fragment of the third component of complement, inside tumor cells - or associated with irradiated tumor cells and unattached to antigen - recruits, accelerates, and amplifies antitumor T cell responses, allowing immunity to reverse or even to prevent tumor growth. C3d enhances antitumor immunity independently of B cells, NK cells, or antibodies, but it does so by increasing tumor infiltrating CD8+ lymphocytes, by depleting Tregs, and by suppressing expression of programmed cell death protein 1 (PD-1) by T cells. These properties of C3d appear specific for the tumor and dependent on complement receptor 2, and they incur no obvious systemic toxicity. The heretofore unrecognized properties of free C3d suggest that protein might determine the effectiveness of immune surveillance and that increasing availability of the protein might prove advantageous in the treatment or prevention of cancer and premalignant conditions.
Collapse
Affiliation(s)
| | - Inês Silva
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Samuel J. Balin
- Department of Dermatology, UCLA, Los Angeles, California, USA
| | - Adam R. Lefferts
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Evan Farkash
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Ted M. Ross
- Center for Vaccines and Immunology, Department of Infectious Diseases, The University of Georgia, Athens, Georgia, USA
| | - Michael C. Carroll
- Department of Pediatrics, Graduate Program in Immunology, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
17
|
Parente R, Clark SJ, Inforzato A, Day AJ. Complement factor H in host defense and immune evasion. Cell Mol Life Sci 2016; 74:1605-1624. [PMID: 27942748 PMCID: PMC5378756 DOI: 10.1007/s00018-016-2418-4] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 11/08/2016] [Accepted: 11/09/2016] [Indexed: 12/30/2022]
Abstract
Complement is the major humoral component of the innate immune system. It recognizes pathogen- and damage-associated molecular patterns, and initiates the immune response in coordination with innate and adaptive immunity. When activated, the complement system unleashes powerful cytotoxic and inflammatory mechanisms, and thus its tight control is crucial to prevent damage to host tissues and allow restoration of immune homeostasis. Factor H is the major soluble inhibitor of complement, where its binding to self markers (i.e., particular glycan structures) prevents complement activation and amplification on host surfaces. Not surprisingly, mutations and polymorphisms that affect recognition of self by factor H are associated with diseases of complement dysregulation, such as age-related macular degeneration and atypical haemolytic uremic syndrome. In addition, pathogens (i.e., non-self) and cancer cells (i.e., altered-self) can hijack factor H to evade the immune response. Here we review recent (and not so recent) literature on the structure and function of factor H, including the emerging roles of this protein in the pathophysiology of infectious diseases and cancer.
Collapse
Affiliation(s)
- Raffaella Parente
- Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Simon J Clark
- Division of Evolution and Genomic Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Antonio Inforzato
- Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089, Milan, Italy. .,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Via Vanvitelli 32, 20129, Milan, Italy.
| | - Anthony J Day
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
18
|
Clement CC, Becerra A, Yin L, Zolla V, Huang L, Merlin S, Follenzi A, Shaffer SA, Stern LJ, Santambrogio L. The Dendritic Cell Major Histocompatibility Complex II (MHC II) Peptidome Derives from a Variety of Processing Pathways and Includes Peptides with a Broad Spectrum of HLA-DM Sensitivity. J Biol Chem 2016; 291:5576-5595. [PMID: 26740625 DOI: 10.1074/jbc.m115.655738] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Indexed: 12/26/2022] Open
Abstract
The repertoire of peptides displayed in vivo by MHC II molecules derives from a wide spectrum of proteins produced by different cell types. Although intracellular endosomal processing in dendritic cells and B cells has been characterized for a few antigens, the overall range of processing pathways responsible for generating the MHC II peptidome are currently unclear. To determine the contribution of non-endosomal processing pathways, we eluted and sequenced over 3000 HLA-DR1-bound peptides presented in vivo by dendritic cells. The processing enzymes were identified by reference to a database of experimentally determined cleavage sites and experimentally validated for four epitopes derived from complement 3, collagen II, thymosin β4, and gelsolin. We determined that self-antigens processed by tissue-specific proteases, including complement, matrix metalloproteases, caspases, and granzymes, and carried by lymph, contribute significantly to the MHC II self-peptidome presented by conventional dendritic cells in vivo. Additionally, the presented peptides exhibited a wide spectrum of binding affinity and HLA-DM susceptibility. The results indicate that the HLA-DR1-restricted self-peptidome presented under physiological conditions derives from a variety of processing pathways. Non-endosomal processing enzymes add to the number of epitopes cleaved by cathepsins, altogether generating a wider peptide repertoire. Taken together with HLA-DM-dependent and-independent loading pathways, this ensures that a broad self-peptidome is presented by dendritic cells. This work brings attention to the role of "self-recognition" as a dynamic interaction between dendritic cells and the metabolic/catabolic activities ongoing in every parenchymal organ as part of tissue growth, remodeling, and physiological apoptosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Simone Merlin
- the School of Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Antonia Follenzi
- From the Departments of Pathology and; the School of Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Scott A Shaffer
- Biochemistry and Molecular Pharmacology and; the Proteomics and Mass Spectrometry Facility, University of Massachusetts Medical School, Worcester, Massachusetts 01655, and
| | - Lawrence J Stern
- the Departments of Pathology and; Biochemistry and Molecular Pharmacology and
| | - Laura Santambrogio
- From the Departments of Pathology and; Microbiology and Immunology, Albert Einstein College of Medicine, New York, New York 10461,.
| |
Collapse
|