1
|
Rafeek RAM, Ketheesan N, Good MF, Pandey M, Lepletier A. Low-dose interleukin 2 therapy halts the progression of post-streptococcal autoimmune complications in a rat model of rheumatic heart disease. mBio 2025; 16:e0382324. [PMID: 39998162 PMCID: PMC11980396 DOI: 10.1128/mbio.03823-24] [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: 12/12/2024] [Accepted: 01/10/2025] [Indexed: 02/26/2025] Open
Abstract
Acute rheumatic fever (ARF) is an autoimmune disease triggered by antibodies and T cells targeting the group A Streptococcus (GAS, Strep A) bacterium, often leading to rheumatic heart disease (RHD) and Sydenham's chorea. Long-term monthly penicillin injections are recognized as a cornerstone of public health programs to prevent Strep A reinfection and progression of ARF. However, compliance is poor, and better tools are required to slow disease progression. Preclinical evidence suggests that this can be achieved. Using a rat model that replicates post-streptococcal autoimmune complications, we explored the potential of low-dose interleukin-2 (LD-IL-2) as an immunotherapeutic intervention for ARF/RHD. In this model, injections of recombinant M protein from Strep A type 5 (rM5) to Lewis rats induce cardiac tissue inflammation, conduction abnormalities, and cross-reactive antibodies against cardiac and brain proteins central to disease pathogenesis. In animals injected with rM5 and treated with LD-IL-2, no cardiac functional or histological changes was observed. LD-IL-2 therapy effectively reduced the production of cross-reactive antibodies raised against host proteins and significantly increased regulatory T cells in the mediastinal lymph nodes. These novel findings suggest that LD-IL-2 will be an effective immunotherapeutic agent for treating ARF and has the potential to replace the standard monthly penicillin injections. IMPORTANCE Post-streptococcal autoimmune syndromes, including acute rheumatic fever, rheumatic heart disease, and Sydenham's chorea, represent a significant yet often under-recognized health and economic burden. This is especially true in low-income countries and among Indigenous populations in high-income nations, where the disease burden is most severe. These conditions arise from an autoimmune response to group A Streptococcus infections, leading to long-term health complications, disability, and premature death. Despite their widespread impact, no vaccine is currently available to prevent reinfections, and no specific therapy exists to treat the resulting autoimmune process. This study uses a rat model of rheumatic heart disease to evaluate the potential of low-dose interleukin 2 therapy in improving clinical outcomes and reducing the incidence of autoimmune diseases triggered by streptococcal infections.
Collapse
Affiliation(s)
| | - Natkunam Ketheesan
- School of Science and Technology, University of New England, New South Wales, Australia
| | - Michael F. Good
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Manisha Pandey
- School of Science and Technology, University of New England, New South Wales, Australia
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Ailin Lepletier
- School of Science and Technology, University of New England, New South Wales, Australia
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
2
|
Xian S, Zeng Z. Methods for Establishing a Rat Model of Rheumatic Heart Disease. Rev Cardiovasc Med 2024; 25:346. [PMID: 39355577 PMCID: PMC11440394 DOI: 10.31083/j.rcm2509346] [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: 01/04/2024] [Revised: 04/23/2024] [Accepted: 05/07/2024] [Indexed: 10/03/2024] Open
Abstract
Rheumatic heart disease (RHD) is responsible for nearly 250,000 deaths annually and poses a significant health threat in developing areas. The unclear pathogenesis of RHD makes the development of cost-effective treatments challenging, particularly as current surgical options are expensive and technologically demanding, exacerbating the economic and quality-of-life burdens for patients. Given the risks associated with direct human experimentation due to the uncertain pathogenesis, using a rat model infected with Group A Streptococcus (GAS) has become a crucial experimental strategy for RHD research. The development of an RHD rat model, refined over 23 years, now stands as a pivotal approach in studies aiming to understand the disease's pathogenesis. This review summarizes the evolution, characteristics, advantages, and limitations of the RHD rat model, offering insights into potential areas for improvement. It aims to provide researchers with a comprehensive understanding of the model, supporting the advancement of research methodologies and the discovery of innovative treatments for RHD.
Collapse
Affiliation(s)
- Shenglin Xian
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, 530021 Nanning, Guangxi, China
- Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, 530021 Nanning, Guangxi, China
- Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, 530021 Nanning, Guangxi, China
| | - Zhiyu Zeng
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, 530021 Nanning, Guangxi, China
- Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, 530021 Nanning, Guangxi, China
- Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, 530021 Nanning, Guangxi, China
| |
Collapse
|
3
|
Alharbi N, Shalash AO, Koirala P, Boer JC, Hussein WM, Khalil ZG, Capon RJ, Plebanski M, Toth I, Skwarczynski M. Cholesterol as an inbuilt immunoadjuvant for a lipopeptide vaccine against group A Streptococcus infection. J Colloid Interface Sci 2024; 663:43-52. [PMID: 38387185 DOI: 10.1016/j.jcis.2024.02.134] [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: 11/05/2023] [Revised: 01/21/2024] [Accepted: 02/17/2024] [Indexed: 02/24/2024]
Abstract
Peptide-based vaccines can trigger highly specific immune responses, although peptides alone are usually unable to confer strong humoral or cellular immunity. Consequently, peptide antigens are administered with immunostimulatory adjuvants, but only a few are safe and effective for human use. To overcome this obstacle, herein a peptide antigen was lipidated to effectively anchor it to liposomes and emulsion. A peptide antigen B cell epitope from Group A Streptococcus M protein was conjugated to a universal T helper epitope, the pan DR-biding epitope (PADRE), alongside a lipidic moiety cholesterol. Compared to a free peptide antigen, the lipidated version (LP1) adopted a helical conformation and self-assembled into small nanoparticles. Surprisingly, LP1 alone induced the same or higher antibody titers than liposomes or emulsion-based formulations. In addition, antibodies produced by mice immunized with LP1 were more opsonic than those induced by administering the antigen with incomplete Freund's adjuvant. No side effects were observed in the immunized mice and no excessive inflammatory immune responses were detected. Overall, this study demonstrated how simple conjugation of cholesterol to a peptide antigen can produce a safe and efficacious vaccine against Group A Streptococcus - the leading cause of superficial infections and the bacteria responsible for deadly post-infection autoimmune disorders.
Collapse
Affiliation(s)
- Nedaa Alharbi
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; University of Jeddah, College of Science, Department of Chemistry, Jeddah, Saudi Arabia
| | - Ahmed O Shalash
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Prashamsa Koirala
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jennifer C Boer
- School of Health and Biomedical Sciences, RMIT University, VIC 3083, Australia
| | - Waleed M Hussein
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Zeinab G Khalil
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Robert J Capon
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, VIC 3083, Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia; School of Pharmacy, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
4
|
Kirvan CA, Canini H, Swedo SE, Hill H, Veasy G, Jankelow D, Kosanke S, Ward K, Zhao YD, Alvarez K, Hedrick A, Cunningham MW. IgG2 rules: N-acetyl-β-D-glucosamine-specific IgG2 and Th17/Th1 cooperation may promote the pathogenesis of acute rheumatic heart disease and be a biomarker of the autoimmune sequelae of Streptococcus pyogenes. Front Cardiovasc Med 2023; 9:919700. [PMID: 36815140 PMCID: PMC9939767 DOI: 10.3389/fcvm.2022.919700] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 12/29/2022] [Indexed: 02/09/2023] Open
Abstract
Antecedent group A streptococcal pharyngitis is a well-established cause of acute rheumatic fever (ARF) where rheumatic valvular heart disease (RHD) and Sydenham chorea (SC) are major manifestations. In ARF, crossreactive antibodies and T cells respond to streptococcal antigens, group A carbohydrate, N-acetyl-β-D-glucosamine (GlcNAc), and M protein, respectively, and through molecular mimicry target heart and brain tissues. In this translational human study, we further address our hypothesis regarding specific pathogenic humoral and cellular immune mechanisms leading to streptococcal sequelae in a small pilot study. The aims of the study were to (1) better understand specific mechanisms of pathogenesis in ARF, (2) identify a potential early biomarker of ARF, (3) determine immunoglobulin G (IgG) subclasses directed against GlcNAc, the immunodominant epitope of the group A carbohydrate, by reaction of ARF serum IgG with GlcNAc, M protein, and human neuronal cells (SK-N-SH), and (4) determine IgG subclasses deposited on heart tissues from RHD. In 10 pediatric patients with RHD and 6 pediatric patients with SC, the serum IgG2 subclass reacted significantly with GlcNAc, and distinguished ARF from 7 pediatric patients with uncomplicated pharyngitis. Three pediatric patients who demonstrated only polymigrating arthritis, a major manifestation of ARF and part of the Jones criteria for diagnosis, lacked the elevated IgG2 subclass GlcNAc-specific reactivity. In SC, the GlcNAc-specific IgG2 subclass in cerebrospinal fluid (CSF) selectively targeted human neuronal cells as well as GlcNAc in the ELISA. In rheumatic carditis, the IgG2 subclass preferentially and strongly deposited in valve tissues (n = 4) despite elevated concentrations of IgG1 and IgG3 in RHD sera as detected by ELISA to group A streptococcal M protein. Although our human study of ARF includes a very small limited sample set, our novel research findings suggest a strong IgG2 autoantibody response against GlcNAc in RHD and SC, which targeted heart valves and neuronal cells. Cardiac IgG2 deposition was identified with an associated IL-17A/IFN-γ cooperative signature in RHD tissue which displayed both IgG2 deposition and cellular infiltrates demonstrating these cytokines simultaneously. GlcNAc-specific IgG2 may be an important autoantibody in initial stages of the pathogenesis of group A streptococcal sequelae, and future studies will determine if it can serve as a biomarker for risk of RHD and SC or early diagnosis of ARF.
Collapse
Affiliation(s)
- Christine A. Kirvan
- Department of Biological Sciences, California State University, Sacramento, CA, United States
| | - Heather Canini
- Department of Biological Sciences, California State University, Sacramento, CA, United States
| | - Susan E. Swedo
- Pediatrics and Developmental Neuropsychiatry Branch, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, United States
| | - Harry Hill
- Departments of Pediatrics, Infectious Diseases, Cardiology, and Pathology, University of Utah College of Medicine, Salt Lake City, UT, United States
| | - George Veasy
- Departments of Pediatrics, Infectious Diseases, Cardiology, and Pathology, University of Utah College of Medicine, Salt Lake City, UT, United States
| | - David Jankelow
- Division of Cardiology, University of Witwatersrand, Johannesburg, South Africa
| | - Stanley Kosanke
- Department of Comparative Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Kent Ward
- Department of Pediatrics, Division of Cardiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Yan D. Zhao
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Kathy Alvarez
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Andria Hedrick
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Madeleine W. Cunningham
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
5
|
Reynolds S, Rafeek RAM, Hamlin A, Lepletier A, Pandey M, Ketheesan N, Good MF. Streptococcus pyogenes vaccine candidates do not induce autoimmune responses in a rheumatic heart disease model. NPJ Vaccines 2023; 8:9. [PMID: 36739443 PMCID: PMC9899064 DOI: 10.1038/s41541-023-00604-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 01/23/2023] [Indexed: 02/06/2023] Open
Abstract
We have developed a candidate vaccine to protect against multiple strains of Streptococcus pyogenes infections. The candidate vaccine contains two synthetic peptides derived from S. pyogenes proteins: the M-protein epitope, p*17 and the IL-8 degrading S. pyogenes Cell-Envelope Proteinase (SpyCEP) epitope, K4S2. In this study we utilise a rat autoimmune valvulitis model that displays both the cardiac and neurobehavioural pathology associated with post-streptococcal sequelae, to assess if the vaccine candidate antigens induce autoimmune complications and inflammatory pathology. Each antigen was conjugated to carrier protein diphtheria toxoid (DT) and independently assessed for potential to induce autoimmune pathology in female Lewis rats. Rats were administered three subcutaneous doses, and one intranasal dose over a four-week study with a two-week recovery period. A positive control group received recombinant S. pyogenes M5 (rM5) protein, and the negative control group received PBS. Rats that received rM5 developed significant cardiac and neurological pathologies. There was no evidence of these pathologies in the PBS control group, or the rats administered either P*17-DT or K4S2-DT. This study provides further preclinical evidence of the safety of the vaccine candidates p*17 and K4S2 and their appropriateness as candidates in human clinical trials.
Collapse
Affiliation(s)
- Simone Reynolds
- grid.1022.10000 0004 0437 5432Institute for Glycomics, Griffith University, Southport, Queensland Australia
| | - Rukshan Ahamed Mohamed Rafeek
- grid.1020.30000 0004 1936 7371School of Science & Technology, University of New England, Armidale, New South Wales Australia
| | - Adam Hamlin
- grid.1020.30000 0004 1936 7371School of Science & Technology, University of New England, Armidale, New South Wales Australia
| | - Ailin Lepletier
- grid.1022.10000 0004 0437 5432Institute for Glycomics, Griffith University, Southport, Queensland Australia
| | - Manisha Pandey
- grid.1022.10000 0004 0437 5432Institute for Glycomics, Griffith University, Southport, Queensland Australia
| | - Natkunam Ketheesan
- grid.1022.10000 0004 0437 5432Institute for Glycomics, Griffith University, Southport, Queensland Australia ,grid.1020.30000 0004 1936 7371School of Science & Technology, University of New England, Armidale, New South Wales Australia
| | - Michael F. Good
- grid.1022.10000 0004 0437 5432Institute for Glycomics, Griffith University, Southport, Queensland Australia
| |
Collapse
|
6
|
Rafeek RAM, Hamlin AS, Andronicos NM, Lawlor CS, McMillan DJ, Sriprakash KS, Ketheesan N. Characterization of an experimental model to determine streptococcal M protein–induced autoimmune cardiac and neurobehavioral abnormalities. Immunol Cell Biol 2022; 100:653-666. [PMID: 35792671 PMCID: PMC9545610 DOI: 10.1111/imcb.12571] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/28/2022] [Accepted: 07/05/2022] [Indexed: 11/29/2022]
Abstract
Group A streptococcal (GAS) infection is associated with a spectrum of autoimmune diseases including acute rheumatic fever/rheumatic heart disease (ARF/RHD) and neurobehavioral abnormalities. Antibodies against GAS M proteins cross‐react with host tissue proteins in the heart and brain leading to the symptomatology observed in ARF/RHD. As throat carriage of Streptococcus dysgalactiae subspecies equisimilis (SDSE) has been reported to be relatively high in some ARF/RHD endemic regions compared with GAS, and both SDSE and GAS express coiled‐coil surface protein called M protein, we hypothesized that streptococci other than GAS can also associated with ARF/RHD and neurobehavioral abnormalities. Neurobehavioral assessments and electrocardiography were performed on Lewis rats before and after exposure to recombinant GAS and SDSE M proteins. Histological assessments were performed to confirm inflammatory changes in cardiac and neuronal tissues. ELISA and Western blot analysis were performed to determine the cross‐reactivity of antibodies with host connective, cardiac and neuronal tissue proteins. Lewis rats injected with M proteins either from GAS or SDSE developed significant cardiac functional and neurobehavioral abnormalities in comparison to control rats injected with phosphate‐buffered saline. Antibodies against GAS and SDSE M proteins cross‐reacted with cardiac, connective and neuronal proteins. Serum from rats injected with streptococcal antigens showed higher immunoglobulin G binding to the striatum and cortex of the brain. Cardiac and neurobehavioral abnormalities observed in our experimental model were comparable to the cardinal symptoms observed in patients with ARF/RHD. Here for the first time, we demonstrate in an experimental model that M proteins from different streptococcal species could initiate and drive the autoimmune‐mediated cardiac tissue damage and neurobehavioral abnormalities.
Collapse
Affiliation(s)
- Rukshan AM Rafeek
- School of Science & Technology University of New England Armidale NSW Australia
| | - Adam S Hamlin
- School of Science & Technology University of New England Armidale NSW Australia
| | | | - Craig S Lawlor
- School of Science & Technology University of New England Armidale NSW Australia
| | - David J McMillan
- School of Science & Technology University of New England Armidale NSW Australia
- School of Science, Technology, Engineering and Genecology Research Centre University of the Sunshine Coast Sippy Downs QLDAustralia
| | - Kadaba S Sriprakash
- School of Science & Technology University of New England Armidale NSW Australia
- Infection and Inflammation Laboratory QIMR Berghofer Medical Research Institute Herston QLDAustralia
| | - Natkunam Ketheesan
- School of Science & Technology University of New England Armidale NSW Australia
- School of Science, Technology, Engineering and Genecology Research Centre University of the Sunshine Coast Sippy Downs QLDAustralia
| |
Collapse
|
7
|
Distinct serotypes of streptococcal M proteins mediate fibrinogen-dependent platelet activation and pro-inflammatory effects. Infect Immun 2021; 90:e0046221. [PMID: 34898252 PMCID: PMC8852700 DOI: 10.1128/iai.00462-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Sepsis is a life-threatening complication of infection that is characterized by a dysregulated inflammatory state and disturbed hemostasis. Platelets are the main regulators of hemostasis, and they also respond to inflammation. The human pathogen Streptococcus pyogenes can cause local infection that may progress to sepsis. There are more than 200 serotypes of S. pyogenes defined according to sequence variations in the M protein. The M1 serotype is among 10 serotypes that are predominant in invasive infection. M1 protein can be released from the surface and has previously been shown to generate platelet, neutrophil, and monocyte activation. The platelet-dependent proinflammatory effects of other serotypes of M protein associated with invasive infection (M3, M5, M28, M49, and M89) are now investigated using a combination of multiparameter flow cytometry, enzyme-linked immunosorbent assay (ELISA), aggregometry, and quantitative mass spectrometry. We demonstrate that only M1, M3, and M5 protein serotypes can bind fibrinogen in plasma and mediate fibrinogen- and IgG-dependent platelet activation and aggregation, release of granule proteins, upregulation of CD62P to the platelet surface, and complex formation with neutrophils and monocytes. Neutrophil and monocyte activation, determined as upregulation of surface CD11b, is also mediated by M1, M3, and M5 protein serotypes, while M28, M49, and M89 proteins failed to mediate activation of platelets or leukocytes. Collectively, our findings reveal novel aspects of the immunomodulatory role of fibrinogen acquisition and platelet activation during streptococcal infections.
Collapse
|
8
|
Rafeek RAM, Lobbe CM, Wilkinson EC, Hamlin AS, Andronicos NM, McMillan DJ, Sriprakash KS, Ketheesan N. Group A streptococcal antigen exposed rat model to investigate neurobehavioral and cardiac complications associated with post-streptococcal autoimmune sequelae. Animal Model Exp Med 2021; 4:151-161. [PMID: 34179722 PMCID: PMC8212825 DOI: 10.1002/ame2.12164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/03/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The neuropsychiatric disorders due to post-streptococcal autoimmune complications such as Sydenham's chorea (SC) are associated with acute rheumatic fever and rheumatic heart disease (ARF/RHD). An animal model that exhibits characteristics of both cardiac and neurobehavioral defects in ARF/RHD would be an important adjunct for future studies. Since age, gender, strain differences, and genotypes impact on the development of autoimmunity, we investigated the behavior of male and female Wistar and Lewis rat strains in two age cohorts (<6 weeks and >12 weeks) under normal husbandry conditions and following exposure to group A streptococcus (GAS). METHODS Standard behavioral assessments were performed to determine the impairments in fine motor control (food manipulation test), gait and balance (beam walking test), and obsessive-compulsive behavior (grooming and marble burying tests). Furthermore, electrocardiography, histology, and behavioral assessments were performed on male and female Lewis rats injected with GAS antigens. RESULTS For control Lewis rats there were no significant age and gender dependent differences in marble burying, food manipulation, beam walking and grooming behaviors. In contrast significant age-dependent differences were observed in Wistar rats in all the behavioral tests except for food manipulation. Therefore, Lewis rats were selected for further experiments to determine the effect of GAS. After exposure to GAS, Lewis rats demonstrated neurobehavioral abnormalities and cardiac pathology akin to SC and ARF/RHD, respectively. CONCLUSION We have characterised a new model that provides longitudinal stability of age-dependent behavior, to simultaneously investigate both neurobehavioral and cardiac abnormalities associated with post-streptococcal complications.
Collapse
Affiliation(s)
| | - Catherine M. Lobbe
- School of Science & TechnologyUniversity of New EnglandArmidaleNSWAustralia
| | - Ethan C. Wilkinson
- School of Science & TechnologyUniversity of New EnglandArmidaleNSWAustralia
| | - Adam S. Hamlin
- School of Science & TechnologyUniversity of New EnglandArmidaleNSWAustralia
| | | | - David J. McMillan
- School of Science & TechnologyUniversity of New EnglandArmidaleNSWAustralia
- School of Science, Technology, Engineering and Genecology Research CentreUniversity of the Sunshine CoastMaroochydore DCQLDAustralia
| | - Kadaba S. Sriprakash
- School of Science & TechnologyUniversity of New EnglandArmidaleNSWAustralia
- QIMR Berghofer Medical Research InstituteHerstonQLDAustralia
| | - Natkunam Ketheesan
- School of Science & TechnologyUniversity of New EnglandArmidaleNSWAustralia
| |
Collapse
|
9
|
McMillan DJ, Rafeek RAM, Norton RE, Good MF, Sriprakash KS, Ketheesan N. In Search of the Holy Grail: A Specific Diagnostic Test for Rheumatic Fever. Front Cardiovasc Med 2021; 8:674805. [PMID: 34055941 PMCID: PMC8160110 DOI: 10.3389/fcvm.2021.674805] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/22/2021] [Indexed: 12/13/2022] Open
Abstract
Current diagnosis of Acute Rheumatic Fever and Rheumatic Heart Disease (ARF/RHD) relies on a battery of clinical observations aided by technologically advanced diagnostic tools and non-specific laboratory tests. The laboratory-based assays fall into two categories: those that (1) detect "evidence of preceding streptococcal infections" (ASOT, anti-DNAse B, isolation of the Group A Streptococcus from a throat swab) and (2) those that detect an ongoing inflammatory process (ESR and CRP). These laboratory tests are positive during any streptococcal infection and are non-specific for the diagnosis of ARF/RHD. Over the last few decades, we have accumulated considerable knowledge about streptococcal biology and the immunopathological mechanisms that contribute to the development, progression and exacerbation of ARF/RHD. Although our knowledge is incomplete and many more years will be devoted to understanding the exact molecular and cellular mechanisms involved in the spectrum of clinical manifestations of ARF/RHD, in this commentary we contend that there is sufficient understanding of the disease process that using currently available technologies it is possible to identify pathogen associated peptides and develop a specific test for ARF/RHD. It is our view that with collaboration and sharing of well-characterised serial blood samples from patients with ARF/RHD from different regions, antibody array technology and/or T-cell tetramers could be used to identify streptococcal peptides specific to ARF/RHD. The availability of an appropriate animal model for this uniquely human disease can further facilitate the determination as to whether these peptides are pathognomonic. Identification of such peptides will also facilitate testing of potential anti-streptococcal vaccines for safety and avoid potential candidates that may pre-dispose potential vaccine recipients to adverse outcomes. Such peptides can also be readily incorporated into a universally affordable point of care device for both primary and tertiary care.
Collapse
Affiliation(s)
- David J. McMillan
- School of Science and Technology, Engineering and Genecology Research Centre, University of the Sunshine Coast, Maroochydore, QLD, Australia
- School of Science and Technology, University of New England, Armidale, NSW, Australia
| | - Rukshan A. M. Rafeek
- School of Science and Technology, University of New England, Armidale, NSW, Australia
| | - Robert E. Norton
- School of Science and Technology, University of New England, Armidale, NSW, Australia
- Pathology Queensland, Townsville University Hospital, Douglas, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Michael F. Good
- Laboratory of Vaccines for the Developing World, Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Kadaba S. Sriprakash
- School of Science and Technology, University of New England, Armidale, NSW, Australia
- Queensland Institute of Medical Research Berghofer (QIMR) Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Natkunam Ketheesan
- School of Science and Technology, University of New England, Armidale, NSW, Australia
| |
Collapse
|
10
|
Rafeek RAM, Sikder S, Hamlin AS, Andronicos NM, McMillan DJ, Sriprakash KS, Ketheesan N. Requirements for a Robust Animal Model to Investigate the Disease Mechanism of Autoimmune Complications Associated With ARF/RHD. Front Cardiovasc Med 2021; 8:675339. [PMID: 34026876 PMCID: PMC8131511 DOI: 10.3389/fcvm.2021.675339] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/09/2021] [Indexed: 01/03/2023] Open
Abstract
The pathogenesis of Acute Rheumatic Fever/Rheumatic Heart Disease (ARF/RHD) and associated neurobehavioral complications including Sydenham's chorea (SC) is complex. Disease complications triggered by Group A streptococcal (GAS) infection are confined to human and determining the early events leading to pathology requires a robust animal model that reflects the hallmark features of the disease. However, modeling these conditions in a laboratory animal, of a uniquely human disease is challenging. Animal models including cattle, sheep, pig, dog, cat, guinea pigs rats and mice have been used extensively to dissect molecular mechanisms of the autoimmune inflammatory responses in ARF/RHD. Despite the characteristic limitations of some animal models, several rodent models have significantly contributed to better understanding of the fundamental mechanisms underpinning features of ARF/RHD. In the Lewis rat autoimmune valvulitis model the development of myocarditis and valvulitis with the infiltration of mononuclear cells along with generation of antibodies that cross-react with cardiac tissue proteins following exposure to GAS antigens were found to be similar to ARF/RHD. We have recently shown that Lewis rats injected with recombinant GAS antigens simultaneously developed cardiac and neurobehavioral changes. Since ARF/RHD is multifactorial in origin, an animal model which exhibit the characteristics of several of the cardinal diagnostic criteria observed in ARF/RHD, would be advantageous to determine the early immune responses to facilitate biomarker discovery as well as provide a suitable model to evaluate treatment options, safety and efficacy of vaccine candidates. This review focuses on some of the common small animals and their advantages and limitations.
Collapse
Affiliation(s)
- Rukshan A. M. Rafeek
- School of Science and Technology, University of New England, Armidale, NSW, Australia
| | - Suchandan Sikder
- School of Science and Technology, University of New England, Armidale, NSW, Australia
- Department of Medicine and Surgery, Chattogram Veterinary and Animal Sciences University, Chattogram, Bangladesh
| | - Adam S. Hamlin
- School of Science and Technology, University of New England, Armidale, NSW, Australia
| | | | - David J. McMillan
- School of Science and Technology, University of New England, Armidale, NSW, Australia
- School of Science, Technology, Engineering and Genecology Research Centre, University of the Sunshine Coast, Maroochydore, QLD, Australia
| | - Kadaba S. Sriprakash
- School of Science and Technology, University of New England, Armidale, NSW, Australia
- Queensland Institute of Medical Research Berghofer, Brisbane, QLD, Australia
| | - Natkunam Ketheesan
- School of Science and Technology, University of New England, Armidale, NSW, Australia
| |
Collapse
|
11
|
Dooley LM, Ahmad TB, Pandey M, Good MF, Kotiw M. Rheumatic heart disease: A review of the current status of global research activity. Autoimmun Rev 2020; 20:102740. [PMID: 33333234 DOI: 10.1016/j.autrev.2020.102740] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 09/04/2020] [Indexed: 01/17/2023]
Abstract
Rheumatic heart disease (RHD) is a serious and long-term consequence of acute rheumatic fever (ARF), an autoimmune sequela of a mucosal infection by Streptococcus pyogenes (Group A Streptococcus, Strep A). The pathogenesis of ARF and RHD is complex and not fully understood but involves host and bacterial factors, molecular mimicry, and aberrant host innate and adaptive immune responses that result in loss of self-tolerance and subsequent cross-reactivity with host tissues. RHD is entirely preventable yet claims an estimated 320 000 lives annually. The major burden of disease is carried by developing nations and Indigenous populations within developed nations, including Australia. This review will focus on the epidemiology, pathogenesis and treatment of ARF and RHD in Australia, where: streptococcal pyoderma, rather than streptococcal pharyngitis, and Group C and Group G Streptococcus, have been implicated as antecedents to ARF; the rates of RHD in remote Indigenous communities are persistently among the highest in the world; government register-based programs coordinate disease screening and delivery of prophylaxis with variable success; and researchers are making significant progress in the development of a broad-spectrum vaccine against Strep A.
Collapse
Affiliation(s)
- Leanne M Dooley
- School of Health and Wellbeing, University of Southern Queensland, Toowoomba, Queensland, Australia; Institute for Life Sciences and the Environment, University of Southern Queensland, Toowoomba, Queensland, Australia.
| | - Tarek B Ahmad
- School of Health and Wellbeing, University of Southern Queensland, Toowoomba, Queensland, Australia; Institute for Life Sciences and the Environment, University of Southern Queensland, Toowoomba, Queensland, Australia.
| | - Manisha Pandey
- The Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia.
| | - Michael F Good
- The Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia.
| | - Michael Kotiw
- School of Health and Wellbeing, University of Southern Queensland, Toowoomba, Queensland, Australia; Institute for Life Sciences and the Environment, University of Southern Queensland, Toowoomba, Queensland, Australia.
| |
Collapse
|
12
|
Jia T, Wang C, Han Z, Wang X, Ding M, Wang Q. Experimental Rodent Models of Cardiovascular Diseases. Front Cardiovasc Med 2020; 7:588075. [PMID: 33365329 PMCID: PMC7750387 DOI: 10.3389/fcvm.2020.588075] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 11/09/2020] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular diseases, as the most common non-communicable disease in the world, cause a high mortality rate today and bring a serious medical burden to countries worldwide, especially in low- and middle-income countries. Experimental rodent models are widely used for cardiovascular diseases researches due to the effective simulation of human cardiovascular diseases, strong reproductive ability, and easy detection. Herein, we will summarize the pathological manifestations of common cardiovascular diseases and illustrate the establishment of corresponding experimental rodent models in detail.
Collapse
Affiliation(s)
- Tian Jia
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Chen Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zhengxi Han
- School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaozhi Wang
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Ming Ding
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Quanyi Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
13
|
Xian S, Chen A, Wu X, Lu C, Wu Y, Huang F, Zeng Z. Activation of activin/Smad2 and 3 signaling pathway and the potential involvement of endothelial‑mesenchymal transition in the valvular damage due to rheumatic heart disease. Mol Med Rep 2020; 23:10. [PMID: 33179113 PMCID: PMC7673319 DOI: 10.3892/mmr.2020.11648] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
Rheumatic heart disease (RHD) is an autoimmune disease caused by rheumatic fever following group A hemolytic streptococcal infection and primarily affects the mitral valve. RHD is currently a major global health problem. However, the exact pathological mechanisms associated with RHD-induced cardiac valve damage remain to be elucidated. The endothelial-mesenchymal transition (EndMT) serves a key role in a number of diseases with an important role in cardiac fibrosis and the activin/Smad2 and 3 signaling pathway is involved in regulating the EndMT. Nevertheless, there are no studies to date, to the best of the authors' knowledge, investigating the association between RHD and EndMT. Thus, the aim of the current study was to investigate the potential role of EndMT in cardiac valve damage and assess whether activin/Smad2 and 3 signaling was activated during RHD-induced valvular injury in a rat model of RHD induced by inactivated Group A streptococci and complete Freund's adjuvant. Inflammation and fibrosis were assessed by hematoxylin and eosin and Sirius red staining. Serum cytokine and rheumatoid factor levels were measured using ELISA kits. Expression levels of activin/Smad2 and 3 signaling pathway-related factors [activin A, Smad2, Smad3, phosphorylated (p-)Smad2 and p-Smad3], EndMT-related factors [lymphoid enhancer factor-1 (LEF-1), Snail1, TWIST, zinc finger E-box-binding homeobox (ZEB)1, ZEB2, α smooth muscle actin (α-SMA) and type I collagen α 1 (COL1A1)], apoptosis-related markers (BAX and cleaved caspase-3) and valvular inflammation markers (NF-κB and p-NF-κB) were detected using reverse transcription-quantitative PCR and western blot analyses. Compared with the control group, the degree of valvular inflammation and fibrosis, serum levels of IL-6, IL-17, TNF-α and expression of apoptosis-related markers (BAX and cleaved caspase-3) and valvular inflammation marker (p-NF-κB), activin/Smad2 and 3 signaling pathway-related factors (activin A, p-Smad2 and p-Smad3), EndMT-related factors (LEF-1, Snail1, TWIST, ZEB 1, ZEB2, α-SMA and COL1A1) were significantly increased in the RHD group. These results suggested that the activin/Smad2 and 3 signaling pathway was activated during the development of valvular damage caused by RHD and that the EndMT is involved in RHD-induced cardiac valve damage.
Collapse
Affiliation(s)
- Shenglin Xian
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Ang Chen
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiaodan Wu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Chuanghong Lu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yunjiao Wu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Feng Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zhiyu Zeng
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
14
|
Skwarczynski M, Zhao G, Boer JC, Ozberk V, Azuar A, Cruz JG, Giddam AK, Khalil ZG, Pandey M, Shibu MA, Hussein WM, Nevagi RJ, Batzloff MR, Wells JW, Capon RJ, Plebanski M, Good MF, Toth I. Poly(amino acids) as a potent self-adjuvanting delivery system for peptide-based nanovaccines. SCIENCE ADVANCES 2020; 6:eaax2285. [PMID: 32064333 PMCID: PMC6989150 DOI: 10.1126/sciadv.aax2285] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 11/21/2019] [Indexed: 05/05/2023]
Abstract
To be optimally effective, peptide-based vaccines need to be administered with adjuvants. Many currently available adjuvants are toxic, not biodegradable; they invariably invoke adverse reactions, including allergic responses and excessive inflammation. A nontoxic, biodegradable, biocompatible, self-adjuvanting vaccine delivery system is urgently needed. Herein, we report a potent vaccine delivery system fulfilling the above requirements. A peptide antigen was coupled with poly-hydrophobic amino acid sequences serving as self-adjuvanting moieties using solid-phase synthesis, to produce fully defined single molecular entities. Under aqueous conditions, these molecules self-assembled into distinct nanoparticles and chain-like aggregates. Following subcutaneous immunization in mice, these particles successfully induced opsonic epitope-specific antibodies without the need of external adjuvant. Mice immunized with entities bearing 15 leucine residues were able to clear bacterial load from target organs without triggering the release of soluble inflammatory mediators. Thus, we have developed a well-defined and effective self-adjuvanting delivery system for peptide antigens.
Collapse
Affiliation(s)
- Mariusz Skwarczynski
- The University of Queensland, School of Chemistry & Molecular Biosciences, Lucia, QLD 4072, Australia
| | - Guangzu Zhao
- The University of Queensland, School of Chemistry & Molecular Biosciences, Lucia, QLD 4072, Australia
| | - Jennifer C. Boer
- School of Health and Biomedical Sciences, RMIT University, Victoria 3083, Australia
| | - Victoria Ozberk
- Griffith University, Institute for Glycomics, Gold Coast, QLD 4222, Australia
| | - Armira Azuar
- The University of Queensland, School of Chemistry & Molecular Biosciences, Lucia, QLD 4072, Australia
| | - Jazmina Gonzalez Cruz
- The University of Queensland, Diamantina Institute, Translational Research Institute, Brisbane, QLD 4102, Australia
| | | | - Zeinab G. Khalil
- The University of Queensland, Diamantina Institute, Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Manisha Pandey
- Griffith University, Institute for Glycomics, Gold Coast, QLD 4222, Australia
| | - Mohini A. Shibu
- The University of Queensland, School of Chemistry & Molecular Biosciences, Lucia, QLD 4072, Australia
| | - Waleed M. Hussein
- The University of Queensland, School of Chemistry & Molecular Biosciences, Lucia, QLD 4072, Australia
| | - Reshma J. Nevagi
- The University of Queensland, School of Chemistry & Molecular Biosciences, Lucia, QLD 4072, Australia
| | - Michael R. Batzloff
- Griffith University, Institute for Glycomics, Gold Coast, QLD 4222, Australia
| | - James W. Wells
- The University of Queensland, Diamantina Institute, Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Robert J. Capon
- The University of Queensland, Institute for Molecular Bioscience, St Lucia, QLD 4072, Australia
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Victoria 3083, Australia
| | - Michael F. Good
- Griffith University, Institute for Glycomics, Gold Coast, QLD 4222, Australia
| | - Istvan Toth
- The University of Queensland, School of Chemistry & Molecular Biosciences, Lucia, QLD 4072, Australia
- The University of Queensland, Institute for Molecular Bioscience, St Lucia, QLD 4072, Australia
- The University of Queensland, School of Pharmacy, Woolloongabba, QLD 4102, Australia
| |
Collapse
|
15
|
Chen A, Wen J, Lu C, Lin B, Xian S, Huang F, Wu Y, Zeng Z. Inhibition of miR‑155‑5p attenuates the valvular damage induced by rheumatic heart disease. Int J Mol Med 2019; 45:429-440. [PMID: 31894293 PMCID: PMC6984794 DOI: 10.3892/ijmm.2019.4420] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 11/20/2019] [Indexed: 12/26/2022] Open
Abstract
Autoimmunity is involved in the valvular damage caused by rheumatic heart disease (RHD). Increased evidence has linked microRNAs (miRNAs/miRs) to autoimmune disease. Signal transducer and activator of transcription 3 (STAT3) and sphingosine-1-phosphate receptor 1 (S1PR1) and suppressor of cytokine signaling 1 (SOCS1) have been widely studied for their roles in autoimmunity and inflammation. Thus, the current study aims to investigate the role played by miR-155-5p in RHD-induced valvular damage via the S1PR1, SOCS1/STAT3 and interleukin (IL)-6/STAT3 signaling pathways. An RHD rat model was induced by inactivated Group A streptococci and complete Freund's adjuvant. A recombinant adeno-associated virus (AAV-miR155-inhibitor) was used to inhibit the expression of miR-155-5p in the heart. Inflammation and fibrosis were assessed by hematoxylin and eosin staining and Sirius red staining. The expression of miR-155-5p in valvular tissues and serum exosomes was detected by reverse transcription-quantitative PCR. S1PR1, SOCS1, STAT3, phosphorylated STAT3, IL-6 and IL-17 protein expression was detected by western blotting and immunohistochemistry. The relationships between miR-155-5p and S1PR1 and SOCS1 were detected by dual luciferase assays. Cytokine concentrations were measured by ELISA. The expression of miR-155-5p in valve tissues and serum exosomes was increased along with decreased S1PR1 and activated SOCS1/STAT3 signaling in the RHD model. The expression of IL-6 and IL-17 was increased in the valves and the serum. Dual luciferase assays showed that miR-155-5p directly targeted S1PR1 and SOCS1. Inhibition of valvular miR-155-5p through AAV pretreatment increased S1PR1 expression and inhibited activation of the SOCS1/STAT3 signal pathway as a result of attenuated valvular inflammation and fibrosis as well as a decrease in IL-6 and IL-17 in the valves and serum. These results suggest that inhibition of miR-155-5p can reduce RHD-induced valvular damage via the S1PR1, SOCS1/STAT3 and IL-6/STAT3 signaling pathways.
Collapse
Affiliation(s)
- Ang Chen
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jianlin Wen
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Chuanghong Lu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Beiyou Lin
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Shenglin Xian
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Feng Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yunjiao Wu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zhiyu Zeng
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
16
|
Sikder S, Williams NL, Sorenson AE, Alim MA, Vidgen ME, Moreland NJ, Rush CM, Simpson RS, Govan BL, Norton RE, Cunningham MW, McMillan DJ, Sriprakash KS, Ketheesan N. Group G Streptococcus Induces an Autoimmune Carditis Mediated by Interleukin 17A and Interferon γ in the Lewis Rat Model of Rheumatic Heart Disease. J Infect Dis 2019; 218:324-335. [PMID: 29236994 DOI: 10.1093/infdis/jix637] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 12/08/2017] [Indexed: 12/17/2022] Open
Abstract
Acute rheumatic fever and rheumatic heart disease (ARF/RHD) have long been described as autoimmune sequelae of Streptococcus pyogenes or group A streptococcal (GAS) infection. Both antibody and T-cell responses against immunodominant GAS virulence factors, including M protein, cross-react with host tissue proteins, triggering an inflammatory response leading to permanent heart damage. However, in some ARF/RHD-endemic regions, throat carriage of GAS is low. Because Streptococcus dysgalactiae subspecies equisimilis organisms, also known as β-hemolytic group C streptococci and group G streptococci (GGS), also express M protein, we postulated that streptococci other than GAS may have the potential to initiate or exacerbate ARF/RHD. Using a model initially developed to investigate the uniquely human disease of ARF/RHD, we have discovered that GGS causes interleukin 17A/interferon γ-induced myocarditis and valvulitis, hallmarks of ARF/RHD. Remarkably the histological, immunological, and functional changes in the hearts of rats exposed to GGS are identical to those exposed to GAS. Furthermore, antibody cross-reactivity to cardiac myosin was comparable in both GGS- and GAS-exposed animals, providing additional evidence that GGS can induce and/or exacerbate ARF/RHD.
Collapse
Affiliation(s)
- Suchandan Sikder
- College of Public Health, Medical and Veterinary Sciences, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville
| | - Natasha L Williams
- College of Public Health, Medical and Veterinary Sciences, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville
| | - Alanna E Sorenson
- College of Public Health, Medical and Veterinary Sciences, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville
| | - Md A Alim
- College of Public Health, Medical and Veterinary Sciences, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville
| | - Miranda E Vidgen
- INFLAME Biomedical Research Cluster, School of Health and Sport Sciences, University of the Sunshine Coast, Maroochydore
| | | | - Catherine M Rush
- College of Public Health, Medical and Veterinary Sciences, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville
| | | | - Brenda L Govan
- College of Public Health, Medical and Veterinary Sciences, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville
| | | | - Madeleine W Cunningham
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City
| | - David J McMillan
- INFLAME Biomedical Research Cluster, School of Health and Sport Sciences, University of the Sunshine Coast, Maroochydore
| | - Kadaba S Sriprakash
- Bacterial Pathogenesis Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia
| | - Natkunam Ketheesan
- College of Public Health, Medical and Veterinary Sciences, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville
| |
Collapse
|
17
|
Sikder S, Price G, Alim MA, Gautam A, Scott Simpson R, Margaret Rush C, Lee Govan B, Ketheesan N. Group A streptococcal M-protein specific antibodies and T-cells drive the pathology observed in the rat autoimmune valvulitis model. Autoimmunity 2019; 52:78-87. [PMID: 31062619 DOI: 10.1080/08916934.2019.1605356] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Acute rheumatic fever (ARF) and rheumatic heart disease (RHD) are autoimmune mediated diseases triggered by group A streptococcal (GAS) infections. Molecular mimicry between GAS M-proteins and host tissue proteins has been proposed as the mechanism that initiates autoreactive immune responses in ARF/RHD. However, the individual role of antibodies and T-cells specific for GAS M-proteins in the pathogenesis of autoimmune carditis remains under-explored. The current study investigated the role of antibodies and T-cells in the development of carditis in the Lewis rat autoimmune valvultis (RAV) model by transferring serum and/or splenic T-cells from rats previously injected with GAS recombinant M5 protein. Here we report that serum antibodies alone and serum plus in vitro expanded rM5-specific T-cells from hyperimmune rats were capable of transferring carditis to naïve syngeneic animals. Moreover, the rats that received combined serum and T-cells developed more severe carditis. Recipient rats developed mitral valvulitis and myocarditis and showed prolongation of P-R intervals in electrocardiography. GAS M5 protein-specific IgG reactivity and T-cell recall response were also demonstrated in recipient rats indicating long-term persistence of antibodies and T-cells following transfer. The results suggest that both anti-GAS M5 antibodies and T-cells have differential propensity to induce autoimmune mediated carditis in syngeneic rats following transfer. The results highlight that antibodies and effector T-cells generated by GAS M protein injection can also independently home into cardiac tissue to cross-react with tissue proteins causing autoimmune mediated immunopathology.
Collapse
Affiliation(s)
- Suchandan Sikder
- a College of Public Health Medical and Veterinary Sciences , Australian Institute of Tropical Health and Medicine, James Cook University , Townsville , Australia.,b Department of Medicine and Surgery , Chittagong Veterinary and Animal Sciences University , Chittagong , Bangladesh
| | - Georgina Price
- a College of Public Health Medical and Veterinary Sciences , Australian Institute of Tropical Health and Medicine, James Cook University , Townsville , Australia
| | - Md Abdul Alim
- a College of Public Health Medical and Veterinary Sciences , Australian Institute of Tropical Health and Medicine, James Cook University , Townsville , Australia.,c Department of Pathology and Parasitology , Chittagong Veterinary and Animal Sciences University , Chittagong , Bangladesh
| | - Anil Gautam
- d The Townsville Hospital , Townsville , Australia
| | | | - Catherine Margaret Rush
- a College of Public Health Medical and Veterinary Sciences , Australian Institute of Tropical Health and Medicine, James Cook University , Townsville , Australia
| | - Brenda Lee Govan
- a College of Public Health Medical and Veterinary Sciences , Australian Institute of Tropical Health and Medicine, James Cook University , Townsville , Australia
| | - Natkunam Ketheesan
- f School of Science & Technology , University of New England , Armidale , Australia
| |
Collapse
|
18
|
Sikder S, Rush CM, Govan BL, Norton RE, Cunningham MW, McMillan DJ, Sriprakash KS, Ketheesan N. Reply to Dale and Shulman. J Infect Dis 2019; 219:675-676. [PMID: 30247617 DOI: 10.1093/infdis/jiy561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 09/20/2018] [Indexed: 11/12/2022] Open
Affiliation(s)
- Suchandan Sikder
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia
| | - Catherine M Rush
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia
| | - Brenda L Govan
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia
| | | | - Madeleine W Cunningham
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City
| | - David J McMillan
- Inflammation and Healing Biomedical Research Cluster, School of Health and Sport Sciences, University of the Sunshine Coast, Maroochydore, Australia
| | | | | |
Collapse
|
19
|
Meier LA, Auger JL, Engelson BJ, Cowan HM, Breed ER, Gonzalez-Torres MI, Boyer JD, Verma M, Marath A, Binstadt BA. CD301b/MGL2 + Mononuclear Phagocytes Orchestrate Autoimmune Cardiac Valve Inflammation and Fibrosis. Circulation 2018; 137:2478-2493. [PMID: 29386201 PMCID: PMC5988921 DOI: 10.1161/circulationaha.117.033144] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/08/2018] [Indexed: 11/16/2022]
Abstract
BACKGROUND Valvular heart disease is common and affects the mitral valve (MV) most frequently. Despite the prevalence of MV disease (MVD), the cellular and molecular pathways that initiate and perpetuate it are not well understood. METHODS K/B.g7 T-cell receptor transgenic mice spontaneously develop systemic autoantibody-associated autoimmunity, leading to fully penetrant fibroinflammatory MVD and arthritis. We used multiparameter flow cytometry, intracellular cytokine staining, and immunofluorescent staining to characterize the cells in inflamed K/B.g7 MVs. We used genetic approaches to study the contribution of mononuclear phagocytes (MNPs) to MVD in this model. Specifically, we generated K/B.g7 mice in which either CX3CR1 or CD301b/macrophage galactose N-acetylgalactosamine-specific lectin 2 (MGL2)-expressing MNPs were ablated. Using K/B.g7 mice expressing Cx3Cr1-Cre, we conditionally deleted critical inflammatory molecules from MNPs, including the Fc-receptor signal-transducing tyrosine kinase Syk and the cell adhesion molecule very late antigen-4. We performed complementary studies using monoclonal antibodies to block key inflammatory molecules. We generated bone marrow chimeric mice to define the origin of the inflammatory cells present in the MV and to determine which valve cells respond to the proinflammatory cytokine tumor necrosis factor (TNF). Finally, we examined specimens from patients with rheumatic heart disease to correlate our findings to human pathology. RESULTS MNPs comprised the vast majority of MV-infiltrating cells; these MNPs expressed CX3CR1 and CD301b/MGL2. Analogous cells were present in human rheumatic heart disease valves. K/B.g7 mice lacking CX3CR1 or in which CD301b/MGL2-expressing MNPs were ablated were protected from MVD. The valve-infiltrating CD301b/MGL2+ MNPs expressed tissue-reparative molecules including arginase-1 and resistin-like molecule α. These MNPs also expressed the proinflammatory cytokines TNF and interleukin-6, and antibody blockade of these cytokines prevented MVD. Deleting Syk from CX3CR1-expressing MNPs reduced their TNF and interleukin-6 production and also prevented MVD. TNF acted through TNF receptor-1 expressed on valve-resident cells to increase the expression of vascular cell adhesion molecule-1. Conditionally deleting the vascular cell adhesion molecule-1 ligand very late antigen-4 from CX3CR1-expressing MNPs prevented MVD. CONCLUSIONS CD301b/MGL2+ MNPs are key drivers of autoimmune MVD in K/B.g7 mice and are also present in human rheumatic heart disease. We define key inflammatory molecules that drive MVD in this model, including Syk, TNF, interleukin-6, very late antigen-4, and vascular cell adhesion molecule-1.
Collapse
Affiliation(s)
- Lee A Meier
- Center for Immunology (L.A.M., J.L.A., B.J.E., H.M.C., M.I.G.-T., B.A.B.)
- Department of Pediatrics (L.A.M., J.L.A., M.I.G.-T., B.A.B.)
- University of Minnesota Medical School (L.A.M., B.J.E., E.R.B., M.I.G.-T., M.V., B.A.B.), Minneapolis
| | - Jennifer L Auger
- Center for Immunology (L.A.M., J.L.A., B.J.E., H.M.C., M.I.G.-T., B.A.B.)
- Department of Pediatrics (L.A.M., J.L.A., M.I.G.-T., B.A.B.)
| | - Brianna J Engelson
- Center for Immunology (L.A.M., J.L.A., B.J.E., H.M.C., M.I.G.-T., B.A.B.)
- University of Minnesota Medical School (L.A.M., B.J.E., E.R.B., M.I.G.-T., M.V., B.A.B.), Minneapolis
| | - Hannah M Cowan
- Center for Immunology (L.A.M., J.L.A., B.J.E., H.M.C., M.I.G.-T., B.A.B.)
| | - Elise R Breed
- Department of Laboratory Medicine and Pathology (E.R.B.)
- University of Minnesota Medical School (L.A.M., B.J.E., E.R.B., M.I.G.-T., M.V., B.A.B.), Minneapolis
| | - Mayra I Gonzalez-Torres
- Center for Immunology (L.A.M., J.L.A., B.J.E., H.M.C., M.I.G.-T., B.A.B.)
- Department of Pediatrics (L.A.M., J.L.A., M.I.G.-T., B.A.B.)
- University of Minnesota Medical School (L.A.M., B.J.E., E.R.B., M.I.G.-T., M.V., B.A.B.), Minneapolis
| | - Joshua D Boyer
- Department of Medicine, University of California, San Diego (J.D.B.)
| | - Mayank Verma
- Department of Integrative Biology and Physiology (M.V.)
- University of Minnesota Medical School (L.A.M., B.J.E., E.R.B., M.I.G.-T., M.V., B.A.B.), Minneapolis
| | | | - Bryce A Binstadt
- Center for Immunology (L.A.M., J.L.A., B.J.E., H.M.C., M.I.G.-T., B.A.B.)
- Department of Pediatrics (L.A.M., J.L.A., M.I.G.-T., B.A.B.)
- University of Minnesota Medical School (L.A.M., B.J.E., E.R.B., M.I.G.-T., M.V., B.A.B.), Minneapolis
| |
Collapse
|
20
|
Streptococcal pharyngitis and rheumatic heart disease: the superantigen hypothesis revisited. INFECTION GENETICS AND EVOLUTION 2018. [PMID: 29530660 DOI: 10.1016/j.meegid.2018.03.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Streptococcus pyogenes is a human-specific and globally prominent bacterial pathogen that despite causing numerous human infections, this bacterium is normally found in an asymptomatic carrier state. This review provides an overview of both bacterial and human factors that likely play an important role in nasopharyngeal colonization and pharyngitis, as well as the development of acute rheumatic fever and rheumatic heart disease. Here we highlight a recently described role for bacterial superantigens in promoting acute nasopharyngeal infection, and discuss how these immune system activating toxins could be crucial to initiate the autoimmune process in rheumatic heart disease.
Collapse
|
21
|
Xiao P, Zhang K, Tao Z, Liu N, Ge B, Xu M, Lu X. Bmi1 and BRG1 drive myocardial repair by regulating cardiac stem cell function in acute rheumatic heart disease. Exp Ther Med 2017; 14:3812-3816. [PMID: 29042984 DOI: 10.3892/etm.2017.4936] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 04/21/2017] [Indexed: 01/14/2023] Open
Abstract
Rheumatic heart disease (RHD) occurs due to the accumulation of complications associated with rheumatic fever, and it results in high morbidity and mortality. The majority of cases of RHD are diagnosed in the chronic stages, when treatment options are limited. A small reservoir of cardiac stem cells is responsible for maintaining cardiac homeostasis and repairing tissue damage. Understanding the role of cardiac stem cells and the various proteins responsible for their functions in different pathological stages of RHD is an important area of investigation. Polycomb complex protein BMI-1 (Bmi1) and transcription activator BRG1 (BRG1) are associated with the maintenance of stemness in various types of stem cells. The present study investigated the role served by Bmi1 and BRG1 in cardiac stem cells during various pathological stages of RHD through immunohistochemistry and western blotting. A rat model of RHD was established via immunization with the Group A Streptococcus M5 protein. The rat was demonstrated to develop acute RHD 2 months after the final immunization, characterized by cardiac inflammation and tissue damage. Chronic RHD was identified 4 months after the final immunization, revealed by cardiac tissue compression and shrinkage. Expression of the cardiac stem cell marker mast/stem cell growth factor receptor kit was identified to be elevated during acute RHD, but downregulated in the chronic stages of RHD. A similar pattern of expression was revealed for Bmi1 and BRG1, indicating that they serve a role in regulating cardiac stem cell proliferation during acute RHD. These results suggest that cardiac stem cells serve a supportive role in the acute, but not chronic, stages of RHD via expression of Bmi1 and BRG1.
Collapse
Affiliation(s)
- Pingxi Xiao
- Department of Cardiology, Sir Run Run Shaw Hospital, Nanjing Medical University, Nanjing, Jiangsu 211100, P.R. China
| | - Kai Zhang
- Department of Cardiology, Sir Run Run Shaw Hospital, Nanjing Medical University, Nanjing, Jiangsu 211100, P.R. China
| | - Zhiwen Tao
- Department of Cardiology, Sir Run Run Shaw Hospital, Nanjing Medical University, Nanjing, Jiangsu 211100, P.R. China
| | - Niannian Liu
- Department of Cardiology, Sir Run Run Shaw Hospital, Nanjing Medical University, Nanjing, Jiangsu 211100, P.R. China
| | - Bangshun Ge
- Central Laboratory, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211100, P.R. China
| | - Min Xu
- Department of Cardiac Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211100, P.R. China
| | - Xinzheng Lu
- Department of Cardiology, Sir Run Run Shaw Hospital, Nanjing Medical University, Nanjing, Jiangsu 211100, P.R. China
| |
Collapse
|
22
|
Oner T, Ozdemir R, Genc DB, Kucuk M, Karadeniz C, Demirpence S, Yilmazer MM, Mese T, Tavli V, Genel F. Parameters indicative of persistence of valvular pathology at initial diagnosis in acute rheumatic carditis: the role of albumin and CD19 expression. J Pediatr (Rio J) 2016; 92:581-587. [PMID: 27553592 DOI: 10.1016/j.jped.2016.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 01/04/2016] [Accepted: 01/15/2016] [Indexed: 10/21/2022] Open
Abstract
OBJECTIVE The aim of this study is to define the predictors of chronic carditis in patients with acute rheumatic carditis (ARC). METHODS Patients diagnosed with ARC between May 2010 and May 2011 were included in the study. Echocardiography, electrocardiography, lymphocyte subset analysis, acute phase reactants, plasma albumin levels, and antistreptolysin-O (ASO) tests were performed at initial presentation. The echocardiographic assessments were repeated at the sixth month of follow-up. The patients were divided into two groups according to persistence of valvular pathology at 6th month as Group 1 and Group 2, and all clinical and laboratory parameters at admission were compared between two groups of valvular involvement. RESULTS During the one-year study period, 22 patients had valvular disease. Seventeen (77.2%) patients showed regression in valvular pathology. An initial mild regurgitation disappeared in eight patients (36.3%). Among seven (31.8%) patients with moderate regurgitation initially, the regurgitation disappeared in three, and four patients improved to mild regurgitation. Two patients with a severe regurgitation initially improved to moderate regurgitation (9.1%). In five (22.8%) patients, the grade of regurgitation [moderate regurgitation in one (4.6%), and severe regurgitation in 4 (18.2%)] remained unchanged. The albumin level was significantly lower at diagnosis in Group 2 (2.6±0.48g/dL). Lymphocyte subset analysis showed a significant decrease in the CD8 percentage and a significant increase in CD19 percentage at diagnosis in Group 2 compared to Group 1. CONCLUSION The blood albumin level and the percentage of CD8 and CD19 (+) lymphocytes at diagnosis may help to predict chronic valvular disease risk in patients with acute rheumatic carditis.
Collapse
Affiliation(s)
- Taliha Oner
- Izmir Dr. Behcet Uz Children's Hospital, Pediatric Cardiology, Izmir, Turkey
| | - Rahmi Ozdemir
- Izmir Dr. Behcet Uz Children's Hospital, Pediatric Cardiology, Izmir, Turkey.
| | - Dildar Bahar Genc
- Sisli Etfal Training and Research Hospital, Pediatric Oncology, Istanbul, Turkey
| | - Mehmet Kucuk
- Izmir Dr. Behcet Uz Children's Hospital, Pediatric Cardiology, Izmir, Turkey
| | - Cem Karadeniz
- Izmir Dr. Behcet Uz Children's Hospital, Pediatric Cardiology, Izmir, Turkey
| | - Savas Demirpence
- Izmir Dr. Behcet Uz Children's Hospital, Pediatric Cardiology, Izmir, Turkey
| | | | - Timur Mese
- Izmir Dr. Behcet Uz Children's Hospital, Pediatric Cardiology, Izmir, Turkey
| | - Vedide Tavli
- Izmir Dr. Behcet Uz Children's Hospital, Pediatric Cardiology, Izmir, Turkey
| | - Ferah Genel
- Izmir Dr. Behçet Uz Children's Hospital, Pediatric Immunology, Izmir, Turkey
| |
Collapse
|
23
|
Oner T, Ozdemir R, Genc DB, Kucuk M, Karadeniz C, Demirpence S, Yilmazer MM, Mese T, Tavli V, Genel F. Parameters indicative of persistence of valvular pathology at initial diagnosis in acute rheumatic carditis: the role of albumin and CD19 expression. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2016. [DOI: 10.1016/j.jpedp.2016.08.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
24
|
Gorton D, Sikder S, Williams NL, Chilton L, Rush CM, Govan BL, Cunningham MW, Ketheesan N. Repeat exposure to group A streptococcal M protein exacerbates cardiac damage in a rat model of rheumatic heart disease. Autoimmunity 2016; 49:563-570. [PMID: 27562362 PMCID: PMC5177596 DOI: 10.1080/08916934.2016.1217999] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Rheumatic fever and rheumatic heart disease (RF/RHD) develop following repeated infection with group A streptococci (GAS). We used the Rat Autoimmune Valvulitis (RAV) model of RF/RHD to demonstrate that repetitive booster immunization with GAS-derived recombinant M protein (rM5) resulted in an enhanced anti-cardiac myosin antibody response that may contribute to the breaking of immune tolerance leading to RF/RHD and increased infiltration of heart valves by mononuclear cells. With each boost, more inflammatory cells were observed infiltrating heart tissue which could lead to severe cardiac damage. We also found evidence that both complement and anti-M protein antibodies in serum from rM5-immunized rats have the potential to contribute to inflammation in heart valves by activating cardiac endothelium. More importantly, we have demonstrated by electrocardiography for the first time in the RAV model that elongation of P-R interval follows repetitive boost with rM5. Our observations provide experimental evidence for cardiac alterations following repeated exposure to GAS M protein with immunological and electrophysiological features resembling that seen in humans following recurrent GAS infection.
Collapse
Affiliation(s)
- Davina Gorton
- a Biomedicine, College of Public Health, Medical & Veterinary Sciences, and Australian Institute of Tropical Health and Medicine, James Cook University , Townsville , Australia and
| | - Suchandan Sikder
- a Biomedicine, College of Public Health, Medical & Veterinary Sciences, and Australian Institute of Tropical Health and Medicine, James Cook University , Townsville , Australia and
| | - Natasha L Williams
- a Biomedicine, College of Public Health, Medical & Veterinary Sciences, and Australian Institute of Tropical Health and Medicine, James Cook University , Townsville , Australia and
| | - Lisa Chilton
- a Biomedicine, College of Public Health, Medical & Veterinary Sciences, and Australian Institute of Tropical Health and Medicine, James Cook University , Townsville , Australia and
| | - Catherine M Rush
- a Biomedicine, College of Public Health, Medical & Veterinary Sciences, and Australian Institute of Tropical Health and Medicine, James Cook University , Townsville , Australia and
| | - Brenda L Govan
- a Biomedicine, College of Public Health, Medical & Veterinary Sciences, and Australian Institute of Tropical Health and Medicine, James Cook University , Townsville , Australia and
| | - Madeleine W Cunningham
- b Department of Microbiology and Immunology , University of Oklahoma Health Sciences Center , Oklahoma City , OK , USA
| | - Natkunam Ketheesan
- a Biomedicine, College of Public Health, Medical & Veterinary Sciences, and Australian Institute of Tropical Health and Medicine, James Cook University , Townsville , Australia and
| |
Collapse
|
25
|
Batzloff MR, Fane A, Gorton D, Pandey M, Rivera-Hernandez T, Calcutt A, Yeung G, Hartas J, Johnson L, Rush CM, McCarthy J, Ketheesan N, Good MF. Preclinical immunogenicity and safety of a Group A streptococcal M protein-based vaccine candidate. Hum Vaccin Immunother 2016; 12:3089-3096. [PMID: 27541593 DOI: 10.1080/21645515.2016.1222999] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Streptococcus pyogenes (group A streptococcus, GAS) causes a wide range of clinical manifestations ranging from mild self-limiting pyoderma to invasive diseases such as sepsis. Also of concern are the post-infectious immune-mediated diseases including rheumatic heart disease. The development of a vaccine against GAS would have a large health impact on populations at risk of these diseases. However, there is a lack of suitable models for the safety evaluation of vaccines with respect to post-infectious complications. We have utilized the Lewis Rat model for cardiac valvulitis to evaluate the safety of the J8-DT vaccine formulation in parallel with a rabbit toxicology study. These studies demonstrated that the vaccine did not induce abnormal pathology. We also show that in mice the vaccine is highly immunogenic but that 3 doses are required to induce protection from a GAS skin challenge even though 2 doses are sufficient to induce a high antibody titer.
Collapse
Affiliation(s)
- Michael R Batzloff
- a Institute for Glycomics, Gold Coast Campus, Griffith University , Queensland , Australia
| | - Anne Fane
- b Infectious Diseases and Immunopathogenesis Research Group , Australian Institute of Tropical Health and Medicine, James Cook University , Queensland , Australia
| | - Davina Gorton
- b Infectious Diseases and Immunopathogenesis Research Group , Australian Institute of Tropical Health and Medicine, James Cook University , Queensland , Australia
| | - Manisha Pandey
- a Institute for Glycomics, Gold Coast Campus, Griffith University , Queensland , Australia
| | - Tania Rivera-Hernandez
- a Institute for Glycomics, Gold Coast Campus, Griffith University , Queensland , Australia
| | - Ainslie Calcutt
- a Institute for Glycomics, Gold Coast Campus, Griffith University , Queensland , Australia
| | - Grace Yeung
- a Institute for Glycomics, Gold Coast Campus, Griffith University , Queensland , Australia
| | - Jon Hartas
- a Institute for Glycomics, Gold Coast Campus, Griffith University , Queensland , Australia
| | - Linda Johnson
- b Infectious Diseases and Immunopathogenesis Research Group , Australian Institute of Tropical Health and Medicine, James Cook University , Queensland , Australia
| | - Catherine M Rush
- b Infectious Diseases and Immunopathogenesis Research Group , Australian Institute of Tropical Health and Medicine, James Cook University , Queensland , Australia
| | - James McCarthy
- c QIMR Berghofer Medical Research Institute , Herston , Queensland , Australia
| | - Natkunam Ketheesan
- b Infectious Diseases and Immunopathogenesis Research Group , Australian Institute of Tropical Health and Medicine, James Cook University , Queensland , Australia
| | - Michael F Good
- a Institute for Glycomics, Gold Coast Campus, Griffith University , Queensland , Australia
| |
Collapse
|
26
|
McNeilly C, Cosh S, Vu T, Nichols J, Henningham A, Hofmann A, Fane A, Smeesters PR, Rush CM, Hafner LM, Ketheesan N, Sriprakash KS, McMillan DJ. Predicted Coverage and Immuno-Safety of a Recombinant C-Repeat Region Based Streptococcus pyogenes Vaccine Candidate. PLoS One 2016; 11:e0156639. [PMID: 27310707 PMCID: PMC4911098 DOI: 10.1371/journal.pone.0156639] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 05/17/2016] [Indexed: 12/14/2022] Open
Abstract
The C-terminal region of the M-protein of Streptococcus pyogenes is a major target for vaccine development. The major feature is the C-repeat region, consisting of 35–42 amino acid repeat units that display high but not perfect identity. SV1 is a S. pyogenes vaccine candidate that incorporates five 14mer amino acid sequences (called J14i variants) from differing C-repeat units in a single recombinant construct. Here we show that the J14i variants chosen for inclusion in SV1 are the most common variants in a dataset of 176 unique M-proteins. Murine antibodies raised against SV1 were shown to bind to each of the J14i variants present in SV1, as well as variants not present in the vaccine. Antibodies raised to the individual J14i variants were also shown to bind to multiple but different combinations of J14i variants, supporting the underlying rationale for the design of SV1. A Lewis Rat Model of valvulitis was then used to assess the capacity of SV1 to induce deleterious immune response associated with rheumatic heart disease. In this model, both SV1 and the M5 positive control protein were immunogenic. Neither of these antibodies were cross-reactive with cardiac myosin or collagen. Splenic T cells from SV1/CFA and SV1/alum immunized rats did not proliferate in response to cardiac myosin or collagen. Subsequent histological examination of heart tissue showed that 4 of 5 mice from the M5/CFA group had valvulitis and inflammatory cell infiltration into valvular tissue, whereas mice immunised with SV1/CFA, SV1/alum showed no sign of valvulitis. These results suggest that SV1 is a safe vaccine candidate that will elicit antibodies that recognise the vast majority of circulating GAS M-types.
Collapse
Affiliation(s)
- Celia McNeilly
- Bacterial Pathogenesis Laboratory, QIMR Berghofer Medical Research Institute, 300 Herston Rd, Herston, QLD, 4006, Australia
| | - Samantha Cosh
- Bacterial Pathogenesis Laboratory, QIMR Berghofer Medical Research Institute, 300 Herston Rd, Herston, QLD, 4006, Australia
| | - Therese Vu
- Bacterial Pathogenesis Laboratory, QIMR Berghofer Medical Research Institute, 300 Herston Rd, Herston, QLD, 4006, Australia
| | - Jemma Nichols
- Inflammation and Healing Research Cluster, School of Health and Sport Sciences, University of the Sunshine Coast, Maroochydore, QLD, 4558, Australia
| | - Anna Henningham
- Australian Infectious Disease Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Andreas Hofmann
- Structural Chemistry Program, Eskitis Institute for Cell and Molecular Therapies, Griffith University, Nathan, and Queensland Tropical Health Alliance, Smithfield, QLD, Australia
| | - Anne Fane
- Australian Institute of Tropical Medicine, James Cook University, Townsville, QLD, 4811, Australia
| | - Pierre R Smeesters
- Laboratoire de Génétique et Physiologie Bactérienne, Institut de Biologie et de Médecine Moléculaires, Faculté des Sciences, Université Libre de Bruxelles, Gosselies, Belgium, and Murdoch Children Research Institute, Melbourne, VIC, 3052, Australia
| | - Catherine M Rush
- Australian Institute of Tropical Medicine, James Cook University, Townsville, QLD, 4811, Australia
| | - Louise M Hafner
- School of Biomedical Sciences, Faculty of Health & Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology, Brisbane, QLD, 4001, Australia
| | - Natkuman Ketheesan
- Australian Institute of Tropical Medicine, James Cook University, Townsville, QLD, 4811, Australia
| | - Kadaba S Sriprakash
- Bacterial Pathogenesis Laboratory, QIMR Berghofer Medical Research Institute, 300 Herston Rd, Herston, QLD, 4006, Australia
| | - David J McMillan
- Bacterial Pathogenesis Laboratory, QIMR Berghofer Medical Research Institute, 300 Herston Rd, Herston, QLD, 4006, Australia.,Inflammation and Healing Research Cluster, School of Health and Sport Sciences, University of the Sunshine Coast, Maroochydore, QLD, 4558, Australia
| |
Collapse
|
27
|
Pandey M, Mortensen R, Calcutt A, Powell J, Batzloff MR, Dietrich J, Good MF. Combinatorial Synthetic Peptide Vaccine Strategy Protects against Hypervirulent CovR/S Mutant Streptococci. THE JOURNAL OF IMMUNOLOGY 2016; 196:3364-74. [PMID: 26969753 DOI: 10.4049/jimmunol.1501994] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 02/16/2016] [Indexed: 11/19/2022]
Abstract
Cluster of virulence responder/sensor (CovR/S) mutant group A streptococci (GAS) are serious human pathogens of multiple M protein strains that upregulate expression of virulence factors, including the IL-8 proteaseStreptococcus pyogenescell envelope proteinase (SpyCEP), thus blunting neutrophil-mediated killing and enabling ingress of bacteria from a superficial wound to deep tissue. We previously showed that a combination vaccine incorporating J8-DT (conserved peptide vaccine from the M protein) and a recombinant SpyCEP fragment protects against CovR/S mutants. To enhance the vaccine's safety profile, we identified a minimal epitope (S2) that was the target for anti-SpyCEP Abs that could protect IL-8 from SpyCEP-mediated proteolysis. Abs from healthy humans and from mice experimentally infected with GAS also recognized S2, albeit at low titers. Native SpyCEP may be poorly immunogenic (cryptic or subdominant), and it would be to the organism's advantage if the host did not induce a strong Ab response against it. However, S2 conjugated to diphtheria toxoid is highly immunogenic and induces Abs that recognize and neutralize SpyCEP. Hence, we describe a two-component peptide vaccine that induces Abs (anti-S2) that protect IL-8 from proteolysis and other Abs (anti-J8) that cause strain-independent killing in the presence of neutrophils. We show that either component alone is ineffectual in preventing skin infection and bacteremia due to CovR/S mutants but that the combination induces complete protection. This protection correlated with a significant influx of neutrophils to the infection site. The data strongly suggest that the lack of natural immunity to hypervirulent GAS strains in humans could be rectified by this combination vaccine.
Collapse
Affiliation(s)
- Manisha Pandey
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland 4222, Australia;
| | - Rasmus Mortensen
- Department of Infectious Disease Immunology, Statens Serum Institut, 2300 Copenhagen, Denmark; and Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Ainslie Calcutt
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland 4222, Australia
| | - Jessica Powell
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland 4222, Australia
| | - Michael R Batzloff
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland 4222, Australia
| | - Jes Dietrich
- Department of Infectious Disease Immunology, Statens Serum Institut, 2300 Copenhagen, Denmark; and
| | - Michael F Good
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland 4222, Australia;
| |
Collapse
|
28
|
Abstract
Autoimmune carditis is associated with many human rheumatic conditions, including rheumatic fever, systemic lupus erythematosus, and rheumatoid arthritis. The immune mechanisms that mediate the cardiovascular pathology connected to these diseases are poorly defined. Several animal models are used to recapitulate human pathophysiology in order to better characterize the immunopathogenic mechanisms driving autoimmune endocardial inflammation. These animal models point toward common mechanisms mediating autoimmune endocarditis; in particular, CD4+ T cells and pro-inflammatory macrophages play critical roles in directing the disease process. The goals of this review are to discuss the prevailing animal models of autoimmune endocarditis and their underlying immunologic mechanisms and to provide insight regarding potential therapeutic targets in humans.
Collapse
|
29
|
Martin WJ, Steer AC, Smeesters PR, Keeble J, Inouye M, Carapetis J, Wicks IP. Post-infectious group A streptococcal autoimmune syndromes and the heart. Autoimmun Rev 2015; 14:710-25. [PMID: 25891492 DOI: 10.1016/j.autrev.2015.04.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 04/10/2015] [Indexed: 12/16/2022]
Abstract
There is a pressing need to reduce the high global disease burden of rheumatic heart disease (RHD) and its harbinger, acute rheumatic fever (ARF). ARF is a classical example of an autoimmune syndrome and is of particular immunological interest because it follows a known antecedent infection with group A streptococcus (GAS). However, the poorly understood immunopathology of these post-infectious diseases means that, compared to much progress in other immune-mediated diseases, we still lack useful biomarkers, new therapies or an effective vaccine in ARF and RHD. Here, we summarise recent literature on the complex interaction between GAS and the human host that culminates in ARF and the subsequent development of RHD. We contrast ARF with other post-infectious streptococcal immune syndromes - post-streptococcal glomerulonephritis (PSGN) and the still controversial paediatric autoimmune neuropsychiatric disorders associated with streptococcal infections (PANDAS), in order to highlight the potential significance of variations in the host immune response to GAS. We discuss a model for the pathogenesis of ARF and RHD in terms of current immunological concepts and the potential for application of in depth "omics" technologies to these ancient scourges.
Collapse
Affiliation(s)
- William John Martin
- Inflammation Division, Water and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia.
| | - Andrew C Steer
- Centre for International Child Health, Department of Pediatrics, University of Melbourne and Murdoch Childrens Research Institute, Parkville, VIC 3052, Australia; Group A Streptococcus Laboratory, Murdoch Childrens Research Institute, Parkville, VIC 3052, Australia
| | - Pierre Robert Smeesters
- Centre for International Child Health, Department of Pediatrics, University of Melbourne and Murdoch Childrens Research Institute, Parkville, VIC 3052, Australia; Group A Streptococcus Laboratory, Murdoch Childrens Research Institute, Parkville, VIC 3052, Australia
| | - Joanne Keeble
- Inflammation Division, Water and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Michael Inouye
- Medical Systems Biology, Department of Pathology and Department of Microbiology and Immunology, University of Melbourne, VIC 3010, Australia
| | | | - Ian P Wicks
- Inflammation Division, Water and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia; Rheumatology Unit, Royal Melbourne Hospital, Parkville, VIC 3052, Australia.
| |
Collapse
|
30
|
Cunningham MW. Rheumatic fever, autoimmunity, and molecular mimicry: the streptococcal connection. Int Rev Immunol 2014; 33:314-29. [PMID: 24892819 DOI: 10.3109/08830185.2014.917411] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The group A streptococcus, Streptococcus pyogenes, and its link to autoimmune sequelae, has acquired a new level of understanding. Studies support the hypothesis that molecular mimicry between the group A streptococcus and heart or brain are important in directing immune responses in rheumatic fever. Rheumatic carditis, Sydenham chorea and a new group of behavioral disorders called pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections are reviewed with consideration of autoantibody and T cell responses and the role of molecular mimicry between the heart, brain and group A streptococcus as well as how immune responses contribute to pathogenic mechanisms in disease. In rheumatic carditis, studies have investigated human monoclonal autoantibodies and T cell clones for their crossreactivity and their mechanisms leading to valve damage in rheumatic heart disease. Although studies of human and animal sera from group A streptococcal diseases or immunization models have been crucial in providing clues to molecular mimicry and its role in the pathogenesis of rheumatic fever, study of human monoclonal autoantibodies have provided important insights into how antibodies against the valve may activate the valve endothelium and lead to T cell infiltration. Passive transfer of anti-streptococcal T cell lines in a rat model of rheumatic carditis illustrates effects of CD4+ T cells on the valve. Although Sydenham chorea has been known as the neurological manifestation of rheumatic fever for decades, the combination of autoimmunity and behavior is a relatively new concept linking brain, behavior and neuropsychiatric disorders with streptococcal infections. In Sydenham chorea, human mAbs and their expression in transgenic mice have linked autoimmunity to central dopamine pathways as well as dopamine receptors and dopaminergic neurons in basal ganglia. Taken together, the studies reviewed provide a basis for understanding streptococcal sequelae and how immune responses against group A streptococci influence autoimmunity and inflammatory responses in the heart and brain.
Collapse
Affiliation(s)
- Madeleine W Cunningham
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Biomedical Research Center , Oklahoma City, OK , USA
| |
Collapse
|
31
|
Rush CM, Govan BL, Sikder S, Williams NL, Ketheesan N. Animal models to investigate the pathogenesis of rheumatic heart disease. Front Pediatr 2014; 2:116. [PMID: 25414841 PMCID: PMC4220098 DOI: 10.3389/fped.2014.00116] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 10/07/2014] [Indexed: 11/16/2022] Open
Abstract
Rheumatic fever (RF) and rheumatic heart disease (RHD) are sequelae of group A streptococcal (GAS) infection. Although an autoimmune process has long been considered to be responsible for the initiation of RF/RHD, it is only in the last few decades that the mechanisms involved in the pathogenesis of the inflammatory condition have been unraveled partly due to experimentation on animal models. RF/RHD is a uniquely human condition and modeling this disease in animals is challenging. Antibody and T cell responses to recombinant GAS M protein (rM) and the subsequent interactions with cardiac tissue have been predominantly investigated using a rat autoimmune valvulitis model. In Lewis rats immunized with rM, the development of hallmark histological features akin to RF/RHD, both in the myocardial and in valvular tissue have been reported, with the generation of heart tissue cross-reactive antibodies and T cells. Recently, a Lewis rat model of Sydenham's chorea and related neuropsychiatric disorders has also been described. Rodent models are very useful for assessing disease mechanisms due to the availability of reagents to precisely determine sequential events following infection with GAS or post-challenge with specific proteins and or carbohydrate preparations from GAS. However, studies of cardiac function are more problematic in such models. In this review, a historical overview of animal models previously used and those that are currently available will be discussed in terms of their usefulness in modeling different aspects of the disease process. Ultimately, cardiologists, microbiologists, immunologists, and physiologists may have to resort to diverse models to investigate different aspects of RF/RHD.
Collapse
Affiliation(s)
- Catherine M Rush
- Infectious Disease and Immunopathogenesis Research Group, Australian Institute of Tropical Health and Medicine, James Cook University , Townsville, QLD , Australia
| | - Brenda L Govan
- Infectious Disease and Immunopathogenesis Research Group, Australian Institute of Tropical Health and Medicine, James Cook University , Townsville, QLD , Australia
| | - Suchandan Sikder
- Infectious Disease and Immunopathogenesis Research Group, Australian Institute of Tropical Health and Medicine, James Cook University , Townsville, QLD , Australia
| | - Natasha L Williams
- Infectious Disease and Immunopathogenesis Research Group, Australian Institute of Tropical Health and Medicine, James Cook University , Townsville, QLD , Australia
| | - Natkunam Ketheesan
- Infectious Disease and Immunopathogenesis Research Group, Australian Institute of Tropical Health and Medicine, James Cook University , Townsville, QLD , Australia
| |
Collapse
|
32
|
Kirvan CA, Galvin JE, Hilt S, Kosanke S, Cunningham MW. Identification of streptococcal m-protein cardiopathogenic epitopes in experimental autoimmune valvulitis. J Cardiovasc Transl Res 2013; 7:172-81. [PMID: 24346820 DOI: 10.1007/s12265-013-9526-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 11/20/2013] [Indexed: 11/26/2022]
Abstract
The M protein of rheumatogenic group A streptococci induces carditis and valvulitis in Lewis rats and may play a role in pathogenesis of rheumatic heart disease. To identify the epitopes of M5 protein that produce valvulitis, synthetic peptides spanning A, B, and C repeat regions contained within the extracellular domain of the streptococcal M5 protein were investigated. A repeat region peptides NT4, NT5/6, and NT7 induced valvulitis similar to the intact pepsin fragment of M5 protein. T cell lines from rats with valvulitis recognized M5 peptides NT5/6 and NT6. Passive transfer of an NT5/6-specific T cell line into naïve rats produced valvulitis characterized by infiltration of CD4+ cells and upregulation of VCAM-1, while an NT6-specific T cell line did not target the valve. Our new data suggests that M protein-specific T cells may be important mediators of valvulitis in the Lewis rat model of rheumatic carditis.
Collapse
Affiliation(s)
- Christine A Kirvan
- Department of Biological Sciences, California State University, Sacramento, CA, USA
| | | | | | | | | |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW To give an overview of the current hypotheses of the pathogenesis of rheumatic fever and group A streptococcal autoimmune sequelae of the heart valve and brain. RECENT FINDINGS Human monoclonal antibodies (mAbs) derived from rheumatic heart disease have provided evidence for crossreactive autoantibodies that target the dominant group A streptococcal epitope of the group A carbohydrate, N-acetyl-beta-D-glucosamine (GlcNAc), and heart valve endothelium, laminin and laminar basement membrane. T cells in peripheral blood and in rheumatic heart valves revealed the presence of T cells crossreactive with streptococcal M protein and cardiac myosin. For initiation of disease, evidence suggests a two-hit hypothesis for antibody attack on the valve endothelium with subsequent extravasation of T cells through activated endothelium into the valve to form granulomatous lesions and Aschoff bodies. Autoantibodies against the group A streptococcal carbohydrate epitope GlcNAc and cardiac myosin and its peptides appear during progression of rheumatic heart disease. However, autoantibodies against collagen that are not crossreactive may form because of the release of collagen from damaged valve or to responses to collagen bound in vitro by certain serotypes of streptococci. In Sydenham chorea, human mAbs derived from disease target the group A carbohydrate epitope GlcNAc and gangliosides and dopamine receptors found on the surface of neuronal cells in the brain. Human mAbs and autoantibodies in Sydenham chorea were found to signal neuronal cells and activate calcium calmodulin-dependent protein kinase II (CaMKII) in neuronal cells and recognize the intracellular protein biomarker tubulin. SUMMARY To summarize, pathogenic mechanisms of crossreactive autoantibodies which target the valve in rheumatic heart disease and the neuronal cell in Sydenham chorea share a common streptococcal epitope GlcNAc and target intracellular biomarkers of disease including cardiac myosin in the myocardium and tubulin, a protein abundant in the brain. However, intracellular antigens are not believed to be the basis for disease. The theme of molecular mimicry in streptococcal autoimmune sequelae is the recognition of targeted intracellular biomarker antigens such as cardiac myosin and brain tubulin, while targeting extracellular membrane antigens such as laminin on the valve surface endothelium or lysoganglioside and dopamine receptors in the brain. Antibody binding to these cell surface antigens may lead to valve damage in rheumatic heart disease or neuropsychiatric behaviors and involuntary movements in Sydenham chorea.
Collapse
|
34
|
Animal models used to examine the role of the environment in the development of autoimmune disease: findings from an NIEHS Expert Panel Workshop. J Autoimmun 2012; 39:285-93. [PMID: 22748431 DOI: 10.1016/j.jaut.2012.05.020] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 05/24/2012] [Indexed: 11/23/2022]
Abstract
Autoimmunity is thought to result from a combination of genetics, environmental triggers, and stochastic events. Environmental factors, such as chemicals, drugs or infectious agents, have been implicated in the expression of autoimmune disease, yet human studies are extremely limited in their ability to test isolated exposures to demonstrate causation or to assess pathogenic mechanisms. In this review we examine the research literature on the ability of chemical, physical and biological agents to induce and/or exacerbate autoimmunity in a variety of animal models. There is no single animal model capable of mimicking the features of human autoimmune disease, particularly as related to environmental exposures. An objective, therefore, was to assess the types of information that can be gleaned from the use of animal models, and how well that information can be used to translate back to human health. Our review notes the importance of genetic background to the types and severity of the autoimmune response following exposure to environmental factors, and emphasizes literature where animal model studies have led to increased confidence about environmental factors that affect expression of autoimmunity. A high level of confidence was reached if there were multiple studies from different laboratories confirming the same findings. Examples include mercury, pristane, and infection with Streptococcus or Coxsackie B virus. A second level of consensus identified those exposures likely to influence autoimmunity but requiring further confirmation. To fit into this category, there needed to be significant supporting data, perhaps by multiple studies from a single laboratory, or repetition of some but not all findings in multiple laboratories. Examples include silica, gold, TCE, TCDD, UV radiation, and Theiler's murine encephalomyelitis virus. With the caveat that researchers must keep in mind the limitations and appropriate applications of the various approaches, animal models are shown to be extremely valuable tools for studying the induction or exacerbation of autoimmunity by environmental conditions and exposures.
Collapse
|
35
|
Roosens B, Bala G, Droogmans S, Van Camp G, Breyne J, Cosyns B. Animal models of organic heart valve disease. Int J Cardiol 2012; 165:398-409. [PMID: 22475840 DOI: 10.1016/j.ijcard.2012.03.065] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 02/18/2012] [Accepted: 03/03/2012] [Indexed: 01/23/2023]
Abstract
Heart valve disease is a frequently encountered pathology, related to high morbidity and mortality rates in industrialized and developing countries. Animal models are interesting to investigate the causality, but also underlying mechanisms and potential treatments of human valvular diseases. Recently, animal models of heart valve disease have been developed, which allow to investigate the pathophysiology, and to follow the progression and the potential regression of disease with therapeutics over time. The present review provides an overview of animal models of primary, organic heart valve disease: myxoid age-related, infectious, drug-induced, degenerative calcified, and mechanically induced valvular heart disease.
Collapse
Affiliation(s)
- Bram Roosens
- Centrum Voor Hart- en Vaatziekten (CHVZ), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Brussels, Belgium.
| | | | | | | | | | | |
Collapse
|
36
|
Guerino MT, Postol E, Demarchi LMF, Martins CO, Mundel LR, Kalil J, Guilherme L. HLA class II transgenic mice develop a safe and long lasting immune response against StreptInCor, an anti-group A streptococcus vaccine candidate. Vaccine 2011; 29:8250-6. [PMID: 21907752 DOI: 10.1016/j.vaccine.2011.08.113] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 08/24/2011] [Accepted: 08/28/2011] [Indexed: 11/19/2022]
Abstract
Streptococcus pyogenes infections remain a health problem in several countries because of post-streptococcal sequelae, such as rheumatic fever and rheumatic heart disease. We developed a vaccine epitope (StreptInCor) composed of 55 amino acid residues of the C-terminal portion of the M protein that encompasses both T and B cell protective epitopes. Recently, by using human blood samples, we showed that the StreptInCor epitope is able to bind to different HLA class II molecules and that it could be considered a universal vaccine epitope. In the present work, we evaluated the immune response of HLA class II transgenic mice against aluminum hydroxide-absorbed StreptInCor. After a period of one year, several organs were analyzed histologically to verify the safety of the candidate vaccine epitope. Our results showed that StreptInCor is able to induce robust and safe and long lasting immune response without deleterious reactions in several organs. In conclusion, the results presented here indicate that StreptInCor could be considered a safe vaccine against severe streptococcus-induced diseases.
Collapse
|
37
|
Abstract
Pathogenesis of rheumatic heart disease (RHD) remains incompletely understood. Several genes associated with RHD have been described; most of these are involved with immune responses. Single nucleotide polymorphisms in a number of genes affect patients with RHD compared to controls. Molecular mimicry between streptococcal antigens and human proteins, including cardiac myosin epitopes, vimentin and other intracellular proteins is central to the pathogenesis of RHD. Autoreactive T cells migrate from the peripheral blood to the heart and proliferate in the valves in response to stimulation with specific cytokines. The types of cells involved in the inflammation as well as different cytokine profiles in these patients are being investigated. High TNF alpha, interferon gamma, and low IL4 are found in the rheumatic valve suggesting an imbalance between Th1 and Th2 cytokines and probably contributing to the progressive and permanent valve damage. Animal model of ARF in the Lewis rat may further contribute towards understanding the ARF.
Collapse
Affiliation(s)
- L Guilherme
- Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo; Brazil
| | | | | | | |
Collapse
|
38
|
Fitzgerald KT, Holladay CA, McCarthy C, Power KA, Pandit A, Gallagher WM. Standardization of models and methods used to assess nanoparticles in cardiovascular applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2011; 7:705-717. [PMID: 21319299 DOI: 10.1002/smll.201001347] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Revised: 09/22/2010] [Indexed: 05/30/2023]
Abstract
Nanotechnology has the potential to revolutionize the management and treatment of cardiovascular disease. Controlled drug delivery and nanoparticle-based molecular imaging agents have advanced cardiovascular disease therapy and diagnosis. However, the delivery vehicles (dendrimers, nanocrystals, nanotubes, nanoparticles, nanoshells, etc.), as well as the model systems that are used to mimic human cardiac disease, should be questioned in relation to their suitability. This review focuses on the variations of the biological assays and preclinical models that are currently being used to study the biocompatibility and suitability of nanomaterials in cardiovascular applications. There is a need to standardize appropriate models and methods that will promote the development of novel nanomaterial-based cardiovascular therapies.
Collapse
Affiliation(s)
- Kathleen T Fitzgerald
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | | | | | | |
Collapse
|
39
|
Gorton D, Blyth S, Gorton JG, Govan B, Ketheesan N. An alternative technique for the induction of autoimmune valvulitis in a rat model of rheumatic heart disease. J Immunol Methods 2010; 355:80-5. [PMID: 20206182 DOI: 10.1016/j.jim.2010.02.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2009] [Revised: 02/23/2010] [Accepted: 02/24/2010] [Indexed: 11/19/2022]
Abstract
We currently use a rat model in our investigations into human rheumatic heart disease (RHD). This model traditionally involves footpad immunization with antigen emulsified in complete Freund's adjuvant (CFA). Trials to find an alternative adjuvant to CFA which produced a Th1 type response in the rats resulting in carditis were unsuccessful. However, hock immunization was found to produce the desired valvular pathology without the adverse inflammatory side-effects associated with CFA. We therefore consider the hock an ideal site for immunization, particularly when using CFA.
Collapse
Affiliation(s)
- D Gorton
- Microbiology and Immunology, School of Veterinary and Biomedical Sciences, James Cook University, Townsville 4811, Australia.
| | | | | | | | | |
Collapse
|