Elsevier

Autoimmunity Reviews

Volume 14, Issue 8, August 2015, Pages 710-725
Autoimmunity Reviews

Review
Post-infectious group A streptococcal autoimmune syndromes and the heart

https://doi.org/10.1016/j.autrev.2015.04.005Get rights and content

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.

Introduction

Human infections with Streptococcus pyogenes (group A streptococcus, GAS) constitute a major, worldwide health problem, with up to 700 million cases annually [1]. GAS is an anaerobic, Gram-positive coccus and its only known reservoir is in humans. The oropharynx and skin are the primary colonization sites for GAS and around 12% of apparently normal individuals harbour GAS as a commensal organism in these locations [2].

GAS has a long history with human disease. Intriguingly, it can cause both infectious and post-infectious, immune-mediated diseases. The former includes non-invasive infections, such as pharyngitis and impetigo; invasive infections, such as pneumonia, septic arthritis and necrotising fasciitis; and toxin-mediated syndromes, such as toxic shock syndrome and scarlet fever. Immune syndromes following GAS infection include acute rheumatic fever (ARF), rheumatic heart disease (RHD), post-streptococcal glomerulonephritis (PSGN) and possibly, paediatric autoimmune neuropsychiatric disorders associated with streptococcal infections (PANDAS) (Fig. 1). This review focuses on the clinical and pathological features of these sterile, post-infectious GAS syndromes in order to gain insight into host immunopathogenic mechanisms and hopefully, to suggest new approaches to diagnosis and treatment.

Section snippets

Acute rheumatic fever (ARF) and rheumatic heart disease (RHD)

ARF occurs at a median of two weeks after an antecedent GAS infection. The diagnosis is based on the Jones criteria, comprising major and minor manifestations. The five major manifestations in the Jones criteria reflect target tissue involvement: synovium (inflammatory arthritis), heart valves (endocarditis), brain (chorea), skin (erythema marginatum) and subcutaneous tissue (nodules). A diagnosis of ARF is made in the presence of two major, or one major and two minor, manifestations, together

Genetics of host susceptibility to post-streptococcal immune syndromes

Only a few studies have explored genetic susceptibility to PSGN and PANDAS. Increased frequencies of HLA-DRW4 [48] and HLA-DRB1*0311 [49] were reported in Venezuelan and Egyptian cohorts with PSGN compared to healthy controls. In a Turkish study, children with homozygote ‘a’ alleles, or heterozygote ‘a’/‘b’ alleles, of the endothelial nitric oxide synthase gene intron 4a/b (eNOS4a/b) variable number of tandem repeats polymorphism, had a greater risk for PSGN than those carrying homozygote ‘b’

Bacterial factors that contribute to post-streptococcal immune syndromes

At a molecular level, GAS can be typed by sequencing the hypervariable N-terminal regions of the emm gene encoding the M protein, the major cell surface glycoprotein of GAS. GAS strains can now be classified into 223 emm-types [115]. A number of studies have attempted to link GAS strains or emm-types to disease patterns. Particular emm-types are commonly associated with pharyngitis in high-income countries [116] and also appear to be responsible for invasive disease and ARF in the same

Complement and immune complex activation

PSGN is considered an immune complex disorder, during which streptococcal proteins and anti-GAS antibodies deposit in renal glomeruli, either as pre-formed immune complexes or forming in situ. Immune complex deposition leads to classical pathway complement activation, triggering inflammatory cell recruitment and tissue damage [160], [161]. Two GAS antigens have been implicated as initiators of PGSN-NAPlr, a streptococcal form of glyceraldehyde 3-phosphate dehydrogenase, and SpeB, a cysteine

Where to next in understanding post-streptococcal immune syndromes?

Surprisingly, there are currently no definitive laboratory-based tests for the various post-streptococcal clinical syndromes and diagnosis remains largely clinical. As outlined above, traditional immunological approaches have yielded limited results. A way forward may lie in the application of the new “omics” technologies – genomics, transcriptomics, proteomics, and metabolomics – to these ancient syndromes. Omics approaches provide an unprecedented breadth of information on disease states due

Immunopathogenesis of post-streptococcal immune syndromes: shared or specific mechanisms?

The clinically distinctive post-streptococcal immune syndromes may result from preferential activation of different components of the immune response, acting within distinct anatomical compartments of hosts with variable genetic and environmentally determined risk. PSGN is caused by glomerular deposition of immune complexes, complement activation and temporary amplification of the actions of plasmin. PSGN appears to be a ‘one hit’ immune complex disorder that typically resolves without

Take-home messages

  • In susceptible hosts, GAS engages immune pathways that result in post-infectious sequelae, affecting various organs in distinctive ways.

  • Genetic risk factors play a role in post-GAS sequelae, but the mechanisms involved remain poorly understood.

  • Molecular mimicry between GAS M protein and host cardiac myosin is the most widely accepted cause of ARF and RHD, but additional mechanisms involving a range of alpha coiled-coil endogenous molecules, or other cross reactive targets are likely.

  • Immune

Acknowledgements

Supported by the Reid Charitable Trusts, the National Health and Medical Research Council of Australia (grants 1023407 and 1016647 to Dr. Wicks and grant 0123462 to Dr. Inouye and Prof. Carapetis), and the Victorian State Government (Operational Infrastructure Grant). Many thanks to the key support of Prof. Doug Hilton of the Walter and Eliza Hall Institute of Medical Research.

References (293)

  • S. Stagi et al.

    Evaluation of autoimmune phenomena in patients with pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections (PANDAS)

    Autoimmun Rev

    (2014)
  • S.J. Perlmutter et al.

    Therapeutic plasma exchange and intravenous immunoglobulin for obsessive–compulsive disorder and tic disorders in childhood

    Lancet

    (1999)
  • M.A. Garvey et al.

    A pilot study of penicillin prophylaxis for neuropsychiatric exacerbations triggered by streptococcal infections

    Biol Psychiatry

    (1999)
  • Z. Layrisse et al.

    Family studies of the HLA system in acute post-streptococcal glomerulonephritis

    Hum Immunol

    (1983)
  • L. Lougee et al.

    Psychiatric disorders in first-degree relatives of children with pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections (PANDAS)

    J Am Acad Child Adolesc Psychiatry

    (2000)
  • D.D. Adams et al.

    Autoimmune diseases: solution of the environmental, immunological and genetic components with principles for immunotherapy and transplantation

    Autoimmun Rev

    (2010)
  • M.I. Anastasiou-Nana et al.

    HLA-DR typing and lymphocyte subset evaluation in rheumatic heart disease: a search for immune response factors

    Am Heart J

    (1986)
  • G. Hernandez-Pacheco et al.

    Tumor necrosis factor-alpha promoter polymorphisms in Mexican patients with rheumatic heart disease

    J Autoimmun

    (2003)
  • H.T. Chou et al.

    Association of HLA DRB1-DQA1-DQB1 haplotypes with rheumatic heart disease in Taiwan

    Int J Cardiol

    (2008)
  • W. Weidebach et al.

    HLA class II antigens in rheumatic fever. Analysis of the DR locus by restriction fragment-length polymorphism and oligotyping

    Hum Immunol

    (1994)
  • T. Koyanagi et al.

    DNA typing of HLA class II genes in Japanese patients with rheumatic heart disease

    J Mol Cell Cardiol

    (1996)
  • I.J. Messias Reason et al.

    The association between mannose-binding lectin gene polymorphism and rheumatic heart disease

    Hum Immunol

    (2006)
  • S. Rehman et al.

    A study on the association of TNF-alpha(-308), IL-6(-174), IL-10(-1082) and IL-1Ra(VNTR) gene polymorphisms with rheumatic heart disease in Pakistani patients

    Cytokine

    (2013)
  • P.M. Azevedo et al.

    Interleukin-1 receptor antagonist gene (IL1RN) polymorphism possibly associated to severity of rheumatic carditis in a Brazilian cohort

    Cytokine

    (2010)
  • S.J. Catarino et al.

    Association of MASP2 polymorphisms and protein levels with rheumatic fever and rheumatic heart disease

    Hum Immunol

    (2014)
  • R. Ramasawmy et al.

    Association of polymorphisms within the promoter region of the tumor necrosis factor-alpha with clinical outcomes of rheumatic fever

    Mol Immunol

    (2007)
  • N. Sallakci et al.

    TNF-alpha G-308A polymorphism is associated with rheumatic fever and correlates with increased TNF-alpha production

    J Autoimmun

    (2005)
  • N. Shaikh et al.

    Prevalence of streptococcal pharyngitis and streptococcal carriage in children: a meta-analysis

    Pediatrics

    (2010)
  • Guidelines for the diagnosis of rheumatic fever. Jones criteria, 1992 update. Special Writing Group of the Committee on Rheumatic Fever, Endocarditis, and Kawasaki Disease of the Council on Cardiovascular Disease in the Young of the American Heart Association

    JAMA

    (1992)
  • RHD Australia (ARF/RHD writing group)

    The Australian guideline for prevention, diagnosis and management of acute rheumatic heart disease

    (2012)
  • J.R. Carapetis et al.

    Rheumatic fever in a high incidence population: the importance of monoarthritis and low grade fever

    Arch Dis Child

    (2001)
  • J.R. Carapetis et al.

    Cumulative incidence of rheumatic fever in an endemic region: a guide to the susceptibility of the population?

    Epidemiol Infect

    (2000)
  • Disease WECoRFaRH

    Rheumatic fever and rheumatic heart disease: a report of a WHO Expert Consultation

    (2001)
  • H.C. Lue et al.

    The natural history of rheumatic fever and rheumatic heart disease in the Orient

    Jpn Heart J

    (1979)
  • C. Armstrong

    AHA guidelines on prevention of rheumatic fever and diagnosis and treatment of acute streptococcal pharyngitis

    Am Fam Physician

    (2010)
  • M.G. Parnaby et al.

    Rheumatic fever in indigenous Australian children

    J Paediatr Child Health

    (2010)
  • M. McDonald et al.

    Preventing recurrent rheumatic fever: the role of register based programmes

    Heart

    (2005)
  • A.C. Steer et al.

    Prevention and treatment of rheumatic heart disease in the developing world

    Nat Rev Cardiol

    (2009)
  • E. Marijon et al.

    Prevalence of rheumatic heart disease detected by echocardiographic screening

    N Engl J Med

    (2007)
  • J.R. Carapetis et al.

    Evaluation of a screening protocol using auscultation and portable echocardiography to detect asymptomatic rheumatic heart disease in Tongan schoolchildren

    Nat Clin Pract Cardiovasc Med

    (2008)
  • L.Y. Tani et al.

    Rheumatic fever in children younger than 5 years: is the presentation different?

    Pediatrics

    (2003)
  • A. Nordstrand et al.

    Pathogenic mechanism of acute post-streptococcal glomerulonephritis

    Scand J Infect Dis

    (1999)
  • C.C. Blyth et al.

    Post-streptococcal glomerulonephritis in Sydney: a 16-year retrospective review

    J Paediatr Child Health

    (2007)
  • H.C. Dillon

    Post-streptococcal glomerulonephritis following pyoderma

    Rev Infect Dis

    (1979)
  • C.L. Streeton et al.

    An epidemic of acute post-streptococcal glomerulonephritis among aboriginal children

    J Paediatr Child Health

    (1995)
  • T. Kearns et al.

    Outbreak of acute post streptococcal glomerulonephritis in the Northern Territory — 2000

    NT Dis Control Bull.

    (2001)
  • C.S. Marshall et al.

    Acute post-streptococcal glomerulonephritis in the Northern Territory of Australia: a review of 16 years data and comparison with the literature

    Am J Trop Med Hyg

    (2011)
  • R.B. Jennings et al.

    Post-streptococcal glomerulo-nephritis: histopathologic and clinical studies of the acute, subsiding acute and early chronic latent phases

    J Clin Invest

    (1961)
  • R.J. Wyatt et al.

    Complement profiles in acute post-streptococcal glomerulonephritis

    Pediatr Nephrol

    (1988)
  • S.E. Swedo et al.

    Pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections: clinical description of the first 50 cases

    Am J Psychiatry

    (1998)
  • Cited by (86)

    • Rheumatic cardiopathies and its risk factors: about 50 cases

      2024, Annales de Cardiologie et d'Angeiologie
    • Rheumatic Heart Disease: JACC Focus Seminar 2/4

      2023, Journal of the American College of Cardiology
    View all citing articles on Scopus
    View full text