The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×
Published Online:

Abstract

Rheumatic fever is an immunologically mediated disease that follows infection by group A β-hemolytic Streptococcus (GABHS). In rheumatic fever, antibodies generated against GABHS cross-react with the heart, joints, skin, and other sites, inducing an inflammatory, multisystem disease. Brain tissue-specific antibodies have been demonstrated in a subset of children with Sydenham chorea (a component of the Jones criteria for the diagnosis of rheumatic fever), and most Sydenham chorea patients manifest obsessive-compulsive symptoms very similar to those in traditional obsessive-compulsive disorder. The parallels drawn from the paradigm of Sydenham's chorea to Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal Infections (PANDAS) is an area of active controversy. Newly emerging information on the role of GABHS superantigens in the pathogenesis of rheumatic fever is of particular interest. In this article, we review the microbial characteristics of GABHS and the subsequent immune responses to GABHS as a possible etiology of PANDAS.

The relationship between group A β-hemolytic Streptococcus (GABHS) infection and subsequent development of diseases in skin (erythema marginatum, scarlet fever),1 the heart (rheumatic heart disease),2 joints (migratory polyarthritis),3,4 kidney (poststreptococcal glomerulonephritis),5 and the brain (Sydenham's chorea)6 has been documented in the medical literature for decades. Streptococcal peptides stimulate specific lymphocytic immune responses,7 and there is an association of B lymphocytes with a unique surface alloantigen and susceptibility to rheumatic heart disease (RHD).8 Humoral and/or cellular immune responses, initially mounted against GABHS cross-react to various cardiac tissue-specific epitopes.9,10 T lymphocytes also infiltrate cardiac tissue.11,12

Published evidence indicates a possible etiological link between GABHS infection and a subset of obsessive-compulsive disorder (OCD),1316 Tourette's syndrome (TS),13,1619 and autism,20 and suggests that antibodies that develop against a subgroup of Streptococcus pyogenes cross-react with human brain tissue in genetically susceptible children. Swedo et al. coined the acronym PANDAS (Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal Infections) to represent these disorders. The PANDAS criteria are: 1) presence of OCD or tic disorder, 2) onset between age 3 to puberty, 3) episodic course with dramatic symptom exacerbations, 4) association with GABHS infection, and 5) motoric hyperactivity.21 The PANDAS concept has recently been reviewed by Murphy et al.22 and Swedo et al.23,24 and challenged by others who conclude that currently there is insufficient evidence to establish an association between GABHS and PANDAS.2528 These authors argue that there is not clear-cut evidence for concordance of acute rheumatic fever (ARF) and PANDAS, no evidence of an increased incidence of ARF among family members of PANDAS cases, and no clear-cut association of PANDAS with GABHS infections. Further, they argue that antineuronal antibodies have not been found in all TS cases and antineuronal antibodies have been found in a substantial number of control subjects. They also point out that majority of TS cases do not meet PANDAS criteria and doubted if there is room to implicate nongenetic factors as being significant in the pathogenesis of TS.

The mechanism for anti-GABHS antibody cross-reactivity to brain tissue is attributed to molecular mimicry. Susceptibility to instigating an autoimmune reaction in response to an environmental trigger (GABHS or possibly other infectious agents) has been ascribed to a genetic predisposition. Genetic and environmental influences involved in the pathophysiology of rheumatic fever (RF) and PANDAS have not been elucidated, however, recently published papers provide intriguing clues (see below) that suggest that streptococcal superantigens play a crucial role. (The hypothesis that streptococcal superantigens play a critical role in the pathogenesis of OCD and PANDAS was originally advanced by K.A. Williams, co-author of this article, and is currently under analysis.)

Although the significance has been disputed, elevated levels of a unique surface alloantigen on B lymphocytes have been suggested to be characteristic in RF and PANDAS subjects (see below). Zabriskie et al. raised monoclonal antibodies against this alloantigen by immunizing mice with B cells from a patient with RHD.8 One of these monoclonal antibodies, D8/17, has been isolated in an effort to detect individuals at risk as well as individuals who manifest active illnesses. Children who are susceptible to or who have early onset OCD, TS and autism appear to be vulnerable or tend to overexpress this lymphocyte marker in their B cell population.

The putative lymphocyte alloantigen has not been characterized,29 and the role of the B cell alloantigen in the pathogenesis of the disease remains unknown. It is clear that more evidence is necessary before an immune-based mechanism against GABHS may be attributed to the development of pediatric neuropsychiatric disorders.

The goal of the present paper is to review: 1) microbiologic characteristics of GABHS, 2) immune responses to GABHS as one of the possible etiologies of PANDAS and other neuropsychiatric disorders, and 3) the D8/17 antibody as a potential marker of disease and/or risk for development of PANDAS.

Microbial Characteristics of Group A β-Hemolytic Streptococcus

Group A β-hemolytic streptococcus, or streptococcus pyogenes, is an extracellular, gram-positive bacterium. GABHS is a strictly human pathogen and is responsible for a wider variety of disease than perhaps any other bacterial species.30 There are over 100 serotypes of GABHS, some of which demonstrate selective tissue tropism; Class I strains are rheumatogenic, and Class II strains are nonrheumatogenic.31 Recent genome sequencing of a Class I strain of GABHS32 has determined that this organism has 1,752 predicted protein-encoding genes, including 40 putative virulence-associated genes and microbial molecular mimicry-associated genes.32

An important virulence factor of GABHS, the M protein, protrudes from the surface and interferes with phagocytosis by neutrophils.33 The M proteins of rheumatogenic strains include epitopes that cross-react with cardiac and other host tissues (trophomyosin).34,35 Class I strains have M proteins with a C-repeat domain region lacking in M proteins of nonrheumatogenic serotypes.36 Rheumatogenic GABHS strains are primarily tropic for the throat and cannot produce lipoproteinase, which is characteristic of skin strains.36 M types of GABHS suspected to cause PANDAS, if different from the ones that cause RF, are not known. Sequence analysis and protein prediction of GABHS virulence factors may provide evidence in support of a neural tissue-specific autoimmune phenomenon in the development of PANDAS.

Immune Responses to GABHS in the Etiology of PANDAS

Poststreptococcal diseases occur after a latent period of 1–4 weeks following a GABHS infection.30 Established diseases that follow poststreptococcal infections include acute glomerulonephritis and RF with possible complications of RHD and Sydenham's chorea (SC). The latent period before disease implies that a hypersensitivity response to GABHS infection may be responsible for causing disease, rather than the direct effect of disseminated bacteria. Thus, an acquired autoimmune response to pharyngitis caused by a rheumatogenic strain of GABHS may result from bouts of infection by additional rheumatogenic strains that contain the same cross-reactive epitopes.36 Nephritis is more common after a skin infection, while rheumatic fever is more commonly preceded by infection of the upper respiratory tract. The majority of acute pharyngitis cases are of viral etiology; only 5%–30% of cases are infected with GABHS.3739

The association of preceding streptococcal infection with development of PANDAS has implicated GABHS as a candidate for initiating a parallel neurospecific autoimmune phenomenon. The relationship between GABHS and PANDAS, however, remains unclear. Certain M protein epitopes have been shown to cross-react with heart tissues, synovium, and brain.3 Evidence of an interaction between M protein-induced antineuronal antibodies and cognate brain tissue (Table 1 and Table 2) have led investigators to link certain neurological and psychiatric symptoms to a possible autoimmune reaction within the brain. Preliminary evidence also suggests that heterologous M antigens trigger different antineuronal antibodies, each with a unique binding pattern in the brain. The role of B lymphocytes and streptococcal M protein-induced autoantibodies in PANDAS and SC has been examined recently.16,34,35,4043

Most recently, a prospective study provided convincing evidence establishing a temporal relationship between GABHS infections and PANDAS.44 In this study, authors proposed the role of GABHS-associated toxins in the development of pathophysiology of PANDAS instead of the role of traditional antibodies. GABHS-associated toxins act as superantigens within the host. By binding both the Class II major histocompatibility complex molecules on antigen presenting cells (HLA Class II) and specific Variable-β regions on the T cell receptor, superantigens can undermine immune function.45 Allelic changes within the HLA Class II can lead to proliferation of specific T cell clones at a far higher orders of magnitude than what would be expected in the absence of superantigens.46,47 These expanded T cells interact not only with the M protein epitope but also cross-react with the epitope within the host tissues.48,49

Successful plasma exchange therapy in childhood OCD cases provides supporting evidence of an antibody-mediated etiology of disease.50,51 Conversely, no response to plasmapheresis was reported in a limited number of children with OCD of a non-PANDAS origin.52

As mentioned previously, both the B cell-mediated mechanisms and T cell infiltration in heart and inflammatory cytokines in the CNS have been noted in RHD and OCD patients.11,5357 A recent study showed increased cell-mediated response in CSF of early-onset OCD patients but not in patients with schizophrenia.58 The cytokines involved in a cell-mediated immune response include IL-2, IFN, and TNF, which are responsible for activating T cells and macrophages.59 These results suggest that cell-mediated immune mechanisms participate in the pathogenesis of early onset of OCD in addition to the humoral (cross-reactive autoantibodies) mechanisms mentioned previously. Autoantibodies present in PANDAS patients may be the consequence of, rather than the cause of, PANDAS. It is possible that destruction of cells exposes previously masked epitopes that lead to the development of autoantibodies.

D8/17 Monoclonal Antibody as a Potential Marker for PANDAS

D8/17 is a monoclonal antibody developed against a B lymphocyte surface antigen by repeatedly injecting B cells from confirmed RHD patients into mice.8 Preincubation of the D8/17 antibody with the B cell line originally isolated from an RHD patient markedly diminished binding of D8/17 antibody to cardiac muscle. In Western blot analysis D8/17 antibodies did bind to recombinant type M6 protein, vimentin, and myosin.60 These findings indicate that this B cell alloantigen may be related to contractile proteins present in heart, skeletal, and smooth muscle and may also share epitopes with some components of GABHS. The D8/17 is not in the subset of autoantibodies potentially endogenous to these patients, nor is it a natural antibody present in these patients; rather, it was generated in mice as a monoclonal antibody. Therefore, since the D8/17 antibody is not produced by patients, it is not involved in the establishment of immune memory.

Subsequent studies have investigated expression levels of this B cell surface alloantigen on lymphocytes from RHD and PANDAS patients by using flow cytometry or indirect immunofluorescence. (Flow cytometry is a method for quantitating components or structural features of cells primarily by optical means. In this case, it can detect specific proteins on the surface of cells.) In RHD patients, the number of B lymphocytes with this unique surface alloantigen is increased regardless of the status of the disease, suggesting that this alloantigen may potentially serve as a trait marker.60 Studies demonstrate that the percentage of B lymphocyte populations that express this marker is increased in PANDAS patients (Table 3).

The control groups within the studies showed consistently lower rates of test positive individuals. One recent study that used flow cytometric analysis showed a sensitivity of 77% and specificity of 87% for correlation of D8/17 staining with OCD and TS.14 Earlier studies in RF cases showed that D8/17 binding is both highly sensitive (false negative in 0%–10% in RF) and specific (false positive in 5%–15% in healthy volunteers).60,61 In two studies, D8/17 binding was positive in over 60% of RHD cases and in less than 15% of the control groups.62,63

In these studies, a D8/17 positive B cell count that reached 12% of total B lymphocytes or higher was considered positive. An issue that has been raised concerns the use of isotype control to account for nonspecific antibody binding. Hoekstra et al.64 used an irrelevant IgM monoclonal antibody (MOC 32) as a negative control. In this study, D8/17 positivity was ascertained by mean fluorescence intensity. A shift in mean fluorescence intensity in test patients was reported, while no significant elevation in the percentage of B cells staining positive for the unique D8/17 responsive antigen was detected. This study concluded that B cells as an aggregate population did not segregate into D8/17-positive and D8/17-negative groups; rather, each B cell showed increased expression of the alloantigen.64 As stated above, Murphy et al.65 confirmed this finding in their recent report.

In the Swedo et al. study,12 all subjects met PANDAS criteria, while in other studies13,20 prepubertal onset with defined diagnoses (OCD, TS, autism) were sufficient to meet the study inclusion criteria. The rates of positive D8/17 test results, however, were not significantly different in these studies. Since it is estimated that about 10% of OCD is presumed to meet PANDAS criteria66 it may be that the latter two investigations enrolled study subjects who failed to meet PANDAS criteria. Incidentally, in the study by Hollander et al.,18 the increased repetitive behaviors among autistic children were associated with greater D8/17 expression. These early findings suggest that the PANDAS criteria may undergo modifications depending on the outcomes of the future studies. Presently, there are no published data indicating that OCD or TS patients who fail to meet PANDAS criteria are D8/17 negative. Although limited in scope, D8/17 has also been examined in anorexia nervosa6770 and trichotillomania.15

The discrepancies found among published studies cited above may be due, in part, to the differences in the methods used in these studies. For example, Murphy et al.65 stressed the importance of using a specific dilution of D8/17 (1:1000) to detect its specific epitope.

Until the molecular characteristics of this B cell alloantigen or its monoclonal antibody (D8/17) are known, the conflicting findings reported above, and the difficulties found working with this IgM monoclonal antibody, may remain elusive.

Discussion

Although the significance of D8/17 testing for OCD is in question, the relationship between recurrent streptococcal infections and development of sudden onset of OCD in a subset appears firmly established.(44) Streptococcal antibodies from SC subjects (and by inference OCD) that bind to basal ganglia but not to the rest of the brain tissue71 support above findings. Currently, superantigens from a specific streptococcal strain or strains are believed to play a crucial role in the expansion of specific clones of T cells that target a specific epitope on the M protein as well as host tissue (molecular mimicry)48,49 (see also Immune Responses to GABHS in the Etiology of PANDAS section).

Regarding the D7/18 monoclonal antibody, eight recent studies investigating D8/17 positivity on the surface of B cells demonstrated elevated D8/17-positive cases in a subset of neuropsychiatric disorders that includes PANDAS, SC, OCD, TS, trichotillomania, tics, autism, and anorexia nervosa (Table 3). In these studies, investigators did not use uniform standardized laboratory procedure, thus the reliability of these study results is in question. For example, recently Hoekstra et al. stressed the importance of using an isotype control to account for nonspecific antibody binding64 and Murphy et al. recommended a specific dilution of D8/17 (1:1000) to detect its specific epitope.65

The observation that obsessive compulsive (OC) symptoms from SC are indistinguishable from OC symptoms found in chronic childhood OCD cases72,73 has led Swedo et al. to hypothesize that SC and OCD share the same or similar pathophysiology or etiology.73 Sydenham's chorea (SC) has known pathologic findings (that date back nearly a century) in the corpus striatum,74 thus the etiology of OCD was presumed to follow corresponding anatomical and/or pathophysiological changes. Swedo et al. have subsequently conducted a series of studies that have redefined a subset of OCD, with an increased rate of OCD in first-degree relatives of PANDAS cases than those reported in the general population,75 which are distinct clinical characteristics21 and laboratory findings. Specifically, OCD children were found to have an expanded subset of B lymphocytes staining positive for the D8/17-specific antigen (Table 3), increased antineuronal antibody titers (Table 2), and enlarged corpus striatum.76 The increased basal ganglia size found in the study was similar to that found previously for subjects with SC compared with normal subjects.77

Regarding the relationship between PANDAS and GABHS infection, a recent study reported that an association between antistreptococcal antibodies and OCD/tics might have been confounded by the presence of attention deficit hyperactivity disorder (ADHD).27 The authors enrolled subjects ages 7 to 55 regardless of the status of GABHS infection. In this study, elevated antistreptolysin-O (ASO) and anti-DNase B titers were associated with ADHD but not with tics or OCD. Increased ASO and anti-DNase B titers, however, are nonspecific measurements of recent and repeated streptococcal infections, do not indicate immunity, and are not pathognomonic of complicated streptococcal infection. In the case of RF, when ASO and anti-DNase B levels are increased, the diagnosis of rheumatic fever has always been considered only presumptive.36 It is true that subjects in the above study were younger than the subjects in some OCD/tics studies, and thus may have affected the study outcomes.

Although OC symptoms from OCD and SC are similar, pathogenic strains responsible for SC may be different from serotypes associated with PANDAS. The PANDAS concept was introduced, in part, to allow for the characterization of particular strains of GABHS that are prone to the induction of PANDAS in the susceptible host.21 A relevant publication demonstrated two different immunofluorescence staining patterns in human brain by two separate streptococcal strains, one with M5 antigens and the other with M6 antigens. M5 staining was confined to the caudate region (SC) but M6 staining was more diffuse.42 These antibodies detected a number of brain antigens, but prior absorption by M5 or M6 antigens abolished the reactions.43 The findings suggest that heterologous M proteins trigger different antibrain antibodies that engender different symptoms or clinical courses.

A recent study has reported that serum antibodies from OCD patients bind to human brain tissue.78 In this study, antibody binding was most pronounced in the basal ganglia although binding in other regions was also detected.78 Postmortem studies have demonstrated evidence of vascular inflammation and perivascular infiltration,79 as well as neuronal damage within and outside of the basal ganglia tissue in children with SC.74,79,80 Vascular inflammation within the CNS was also reported in living SC patients.81,82 The neuronal damage is postulated to be associated with brain tissue antigens interacting with streptococcal antineuronal antibodies.41,71 Absorption with GABHS membranes abolished the immunofluorescent staining pattern in neuronal cells, indicating that the antineuronal antibodies were induced by GABHS.41 Although patients with systemic lupus erythematosus (SLE) also have elevated titers of antinuclear antibodies that bind to caudate cells, the antinuclear antibodies found in SLE patients are not absorbed by streptococcal antigens.41 The tissue-specific staining pattern was evidenced by the fact that only isolated neurons abolished the immunofluorescent-staining pattern.41

The association of SC as a constituent of RF with RHD sequela is also well established.83 In RHD, GABHS M protein induced antibodies interact with cardiac antigens.10,34,35 In SC, the GABHS-induced antibodies appear to interact with brain tissue antigens (Table 1 & Table 2). Antineuronal antibody binding to brain tissue, projected from the time of original GABHS infection, and development of SC are temporally mismatched. Whereas antigen-antibody binding occurs within minutes, development of SC often takes several months after GABHS infection.84

Additionally, if antineuronal antibody binds to basal ganglia tissue the manner through which it crosses the blood-brain barrier is not clear. Does an inflammatory process allow antineuronal antibodies to cross the blood-brain barrier? Swedo and Kiessling advanced several plausible possibilities that may allow such antibodies to cross the blood brain barrier.85

In SC, OCD, or TS, antineuronal antibody titers have been shown to correlate with symptom severity.16,17,73,85,86 Whether putative antineuronal antibody binding to brain tissue is associated with changes in underlying pathophysiology is unclear. Published reports also suggest that OC symptoms precede chorea and that OC symptoms resolve before SC symptoms.73 The temporal sequence of OC and SC symptom emergence and resolution as well as the ability of an underlying immune mechanism to effectuate clinical manifestation remains elusive.

In summary, emerging evidence strongly suggests an involvement of streptococcal superantigens in the pathogenesis of RF48 and Kawasaki disease.49 Changes in HLA class II alleles appear to affect superantigen functions, dramatically leading to the proliferation of specific T cell clones,46,47 which appear to target cardiac and vascular tissues.48,49 The pressing need at this time is to examine the role of superantigens from M18 strain (known to cause RF) in SC and PANDAS. We speculate that subsets of other psychiatric disorders such as anorexia nervosa, body dysmorphic disorder, and ADHD may also share pathophysiology similar to that of RF. Findings from these studies, if affirmative, would have a profound impact for the practicing physicians in psychiatry and pediatrics.

TABLE 1. Antineuronal Antibody and Brain Tissue (Basal Ganglia) Interaction in Normals and Nonpsychiatric Patients*,**
TABLE 2. Antineuronal Antibody Assays for OCD and/or Tourette's Syndrome, Sydenham's Chorea, Anorexia Nervosa, and for Healthy Subjects
TABLE 3. D8/17 Studies in PANDAS, Sydenham's Chorea, OCD, Tourette's Syndrome, Trichotillomania, Tics, Autism, Anorexia Nervosa, and Controls

Received May 22, 2002; revised December 15, 2002; accepted January 13, 2003. From the Department of Psychiatry, University of Minnesota Medical School, Minneapolis, Minnesota; the Department of Microbiology and Immunology, FUHS/The Chicago Medical School, Chicago, Illinois; the Division of Child and Adolescent Psychiatry, University of Minnesota Medical School, Minneapolis, Minnesota; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota; and the Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, Minnesota. Address Correspondence to Dr. Kim, Department of Psychiatry, University of Minnesota Medical School, F256/2A West 2450 Riverside Avenue, Minneapolis, MN 55454-1495; (E-mail).
References

1 Bisno AL, Stevens DL: Streptococcal infections of skin and soft tissues. N Engl J Med 1996; 334:240–245Crossref, MedlineGoogle Scholar

2 Goldstein I, Rebeyrotte P, Parlebas J, et al: Isolation from heart valves of glycopeptides which share immunological properties with Streptococcus haemolyticus group A polysaccharides. Nature 1968; 219:866–868Crossref, MedlineGoogle Scholar

3 Dale JB, Chiang EC: Intranasal immunization with recombinant group A streptococcal M fragment fused to the B subunit of Escherichia coli labile toxin protects mice against systemic challenge infections. J Infect Dis 1995; 171:1038–1041Crossref, MedlineGoogle Scholar

4 Gibofsky A, Zabriskie JB: Rheumatic fever and poststreptococcal reactive arthritis. Review. Current Opin Rheumatol 1995; 7:299–305Crossref, MedlineGoogle Scholar

5 Wannamaker LW: Medical progress: differences between streptococcal infections of the throat and of the skin. N Engl J Med 1970; 282:23–31Crossref, MedlineGoogle Scholar

6 Kingston D, Glynn LE: A cross-reaction between Str. pyogenes and human fibroblasts, endothelial cells and astrocytes. Immunol 1971; 21:1003–1016MedlineGoogle Scholar

7 Zabriskie JB, Falk RE: In vitro reactivity of lymphocytes to particulate and soluble antigens. Nature 1970; 226:943–945Crossref, MedlineGoogle Scholar

8 Patarroyo ME, Winchester RJ, Vejerano A, et al: Association of a B-cell alloantigen with susceptibility to rheumatic fever. Nature 1979; 278:173–174Crossref, MedlineGoogle Scholar

9 Cunningham MW: Pathogenesis of group A streptococcal infections. Clin Microbiol Rev 2000; 13:470–511Crossref, MedlineGoogle Scholar

10 Tontsch D, Pankuweit S, Maisch B: Autoantibodies in the sera of patients with rheumatic heart disease: characterization of myocardial antigens by two-dimensional immunoblotting and N-terminal sequence analysis. Clin Exp Immunol 2000; 121:270–274Crossref, MedlineGoogle Scholar

11 Zabriskie JB: T-cells and T-cell clones in rheumatic fever valvulitis. Getting to the heart of the matter? Circulation 1995; 92:281–282Crossref, MedlineGoogle Scholar

12 Swedo SE, Leonard HL, Mittleman BB, et al: Identification of children with pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections by a marker associated with rheumatic fever. Am J Psychiatry 1997; 154:110–112Crossref, MedlineGoogle Scholar

13 Murphy TK, Goodman WK, Fudge MW, et al: B lymphocyte antigen D8/17: a peripheral marker for childhood-onset obsessive-compulsive disorder and Tourette's syndrome? Am J Psychiatry 1997; 154:402–407Crossref, MedlineGoogle Scholar

14 Chapman F, Visvanathan K, Carreno-Manjarrez R, et al: A flow cytometric assay for D8/17 B cell marker in patients with Tourette's syndrome and obsessive compulsive disorder. J Immunol Methods 1998; 219:181–186Crossref, MedlineGoogle Scholar

15 Niehaus DJ, Knowles JA, van Kradenberg J, et al: D8/17 in obsessive-compulsive disorder and trichotillomania (letter). S African Med Journal 1999; 89:755–756MedlineGoogle Scholar

16 Kiessling LS, Marcotte AC, Culpepper L: Antineuronal antibodies in movement disorders. Pediatrics 1993; 92:39–43MedlineGoogle Scholar

17 Swedo SE, Leonard HL, Kiessling LS: Speculations on antineuronal antibody-mediated neuropsychiatric disorders of childhood. Pediatrics 1994; 93:323–326MedlineGoogle Scholar

18 Singer HS, Giuliano JD, Hansen BH, et al: Antibodies against human putamen in children with Tourette syndrome. Neurol 1998; 50:1618–1624Crossref, MedlineGoogle Scholar

19 Morshed SA, Parveen S, Leckman JF, et al: Antibodies against neural, nuclear, cytoskeletal and streptococcal epitopes in children and adults with Tourette's syndrome, Sydenham's chorea, and autoimmune disoders. Biol Psychiatry Biol Psychiatry 2001; 50:566–577Crossref, MedlineGoogle Scholar

20 Hollander E, DelGiudice-Asch G, Simon L, et al: B lymphocyte antigen D8/17 and repetitive behaviors in autism. Am J Psychiatry 1999; 156:317–320MedlineGoogle Scholar

21 Swedo SE, Leonard HL, Garvey M, et al: Pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections: clinical description of the first 50 cases. Am J Psychiatry 1998; 155:264–271MedlineGoogle Scholar

22 Murphy TK, Goodman WK, Ayoub EM, et al: On defining Sydenham's chorea: where do we draw the line? Biol Psychiatry 2000; 47:851–857Crossref, MedlineGoogle Scholar

23 Swedo SE, Garvey M, Snider L, et al: The PANDAS subgroup: recognition and treatment. CNS Spectrums 2001; 6:419–426Crossref, MedlineGoogle Scholar

24 Swedo SE: Pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections (PANDAS). Mol Psychiatry 2002; 7(Suppl)2:24–25Google Scholar

25 Kurlan R: Tourette's syndrome and ‘PANDAS’: will the relation bear out? Pediatric autoimmune neuropsychiatric disorders associated with streptococcal infection Neurol 1998; 50:1530–1534Google Scholar

26 Shulman ST: Pediatric autoimmune neuropsychiatric disorders associated with streptococci (PANDAS). Ped Infect Dis Journal 1999; 18:281–282Crossref, MedlineGoogle Scholar

27 Kaplan EL: PANDAS? or PAND? Or both? Or neither? Contemporary Pediatrics 2000; 17:81–96Google Scholar

28 Peterson BS, Leckman JF, Tucker D, et al: Preliminary findings of antistreptococcal antibody titers and basal ganglia volumes in tic, obsessive-compulsive, and attention deficit/hyperactivity disorders. Arch Gen Psychiatry 2000; 57:364–372Crossref, MedlineGoogle Scholar

29 Kumar D, Kaur S, Grover A, et al: Further observations and characterization of monoclonal antibodies reacting with B cell alloantigens associated with rheumatic fever and rheumatic heart disease. J Lab Clin Med 2000; 135:287–293Crossref, MedlineGoogle Scholar

30 Brooks GF, Butel JS, Ornston LN: Medical Microbiology, 20th ed. Norwalk, CT: Appleton and Lange, 1995; 195–199Google Scholar

31 Brandt ER, Sriprakash KS, Hobb RI, et al: New multi-determinant strategy for a group A streptococcal vaccine designed for the Australian Aboriginal population. Nature Med 2000; 6:455–459Crossref, MedlineGoogle Scholar

32 Ferretti JJ, McShan WM, Ajdic D, et al: Complete genome sequence of an M1 strain of Streptococcus pyogenes. PNAS 2001; 98:4658–4663.Crossref, MedlineGoogle Scholar

33 Baker CJ: Immunization to prevent group B streptococcal disease: victories and vexations. J Infect Dis 1990; 161:917–921Crossref, MedlineGoogle Scholar

34 Cunningham MW, Swerlick RA: Polyspecificity of antistreptococcal murine monoclonal antibodies and their implications in autoimmunity. J Exp Med 1986; 164:998–1012Crossref, MedlineGoogle Scholar

35 Cunningham MW, Hall NK, Krisher KK, et al: A study of anti-group A streptococcal monoclonal antibodies cross-reactive with myosin. J Immunol 1986; 136:293–298MedlineGoogle Scholar

36 Stollerman GH: Rheumatic fever. Lancet 1997; 349:935–42Crossref, MedlineGoogle Scholar

37 Komaroff AL, Pass TM, Aronson MD, et al: The prediction of streptococcal pharyngitis in adults. J Gen Int Med 1986; 1:1–7Crossref, MedlineGoogle Scholar

38 Joslyn SA, Hoekstra GL, Sutherland JE: Rapid antigen detection testing in diagnosing group A beta-hemolytic streptococcal pharyngitis. J Am Bd Family Prac 1995; 8:177–182MedlineGoogle Scholar

39 Dobbs F: A scoring system for predicting group A streptococcal throat infection. Br J Gen Prac 1996; 46:461–464MedlineGoogle Scholar

40 Dale JB, Beachey EH: Epitopes of streptococcal M proteins shared with cardiac myosin. J Exp Med 1985; 162:583–591Crossref, MedlineGoogle Scholar

41 Dorling J, Kingston D, Webb JA: Anti-streptococcal antibodies reacting with brain tissue. II. Utrastructural studies. Br J Exp Pathol 1976; 57:255–265MedlineGoogle Scholar

42 Husby G, van de Rijn I, Zabriskie JB, et al: Antibodies reacting with cytoplasm of subthalamic and caudate nuclei neurons in chorea and acute rheumatic fever. J Exp Med 1976; 144:1094–1110Crossref, MedlineGoogle Scholar

43 Bronze MS, Dale JB: Epitopes of streptococcal M proteins that evoke antibodies that cross-react with human brain. J Immunol 1993; 151:2820–2828MedlineGoogle Scholar

44 Murphy ML, Pichichero ME: Prospective indentification and treatment of children with Pediatric Autoimmune Neuropsychiatric Disorder Associated with Group A Streptococcal Infection (PANDAS). Arch Pediatr Adolesc Med 2002; 156:356–361Crossref, MedlineGoogle Scholar

45 McCormick JK, Yarwood JM, Schlievert PM: Toxic shock syndrome and bacterial superantigens: an update. Annu Rev Microbiol. 2001;55:77–104Google Scholar

46 Norrby-Teglund A, Nepom GT, Kotb M: Differential presentation of group A streptococcal superantigens by HLA class II DQ and DR alleles. Eur J Immunol. 2002 32:2570–2577Google Scholar

47 Kotb M, Norrby-Teglund A, McGeer A, et al: An immunogenetic and molecular basis for differences in outcomes of invasive group A streptococcal infections. Nat Med. 2002 Nov 18 (published on line ahead of print)Google Scholar

48 Smoot LM, McCormick JK, Smoot JC, et al: Characterization of two novel pyrogenic toxin superantigens made by an acute rheumatic fever clone of Streptococcus pyogenes associated with multiple disease outbreaks. Infect Immun 2002; 70:7095–7104Crossref, MedlineGoogle Scholar

49 Leung DY, Meissner HC, Shulman ST et al: Prevalence of superantigen-secreting bacteria in patients with Kawasaki disease. J Pediatr. 2002; 140:742–746Google Scholar

50 Garvey MA, Swedo SW, Shapiro MB, et al: Intravenous immunoglobulin and plasmapheresis as effective treatments of Sydenham's chorea. Neurol 1996; 46:A147Google Scholar

51 Perlmutter SJ, Leitman SF, Garvey MA, et al: Therapeutic plasma exchange and intravenous immunoglobulin for obsessive-compulsive disorder and tic disorders in childhood. Lancet 1999; 354:1153–1158Crossref, MedlineGoogle Scholar

52 Nicolson R, Swedo SE, Lenane M, et al: An open trial of plasma exchange in childhood-onset obsessive-compulsive disorder without poststreptococcal exacerbations. J Am Acad Child Adolesc Psychiatry 2000; 39:1313–1315Crossref, MedlineGoogle Scholar

53 Guilherme L, Dulphy N, Douay C, et al: Molecular evidence for antigen-driven immune responses in cardiac lesions of rheumatic heart disease patients. Int Immunol 2000; 12:1063–1074Crossref, MedlineGoogle Scholar

54 Tomai M, Kotb M, Majumdar G, et al: Superantigenicity of streptococcal M protein. J Exp Med 1990; 172:359–362Crossref, MedlineGoogle Scholar

55 Raizada V, Williams RC Jr, Chopra P, et al: Tissue distribution of lymphocytes in rheumatic heart valves as defined by monoclonal anti-T cell antibodies. Am J Med 1983; 74:90–96Crossref, MedlineGoogle Scholar

56 Read SE, Reid HF, Fischetti VA, et al: Serial studies on the cellular immune response to streptococcal antigens in acute and convalescent rheumatic fever patients in Trinidad. J Clin Immunol 1986; 6:433–441Crossref, MedlineGoogle Scholar

57 Yoshinaga M, Figueroa F, Wahid MR, et al: Antigenic specificity of lymphocytes isolated from valvular specimens of rheumatic fever patients. J Autoimmunity 1995; 8:601–613Crossref, MedlineGoogle Scholar

58 Mittleman BB: Cytokine networks in Sydenham's chorea and PANDAS. Adv Exp Med Biol 1997; 418:933–935Crossref, MedlineGoogle Scholar

59 Mittleman BB, Castellanos FX, Jacobsen LK, et al: Cerebrospinal fluid cytokines in pediatric neuropsychiatric disease. J Immunol 1997; 159:2994–2999MedlineGoogle Scholar

60 Khanna AK: Presence of a non-HLA B cell antigen in rheumatic fever patients and their families as defined by a monoclonal antibody. J Clin Invest 1989; 83:1710–171Crossref, MedlineGoogle Scholar

61 Herdy GV, Zabriskie JB, Chapman F, et al: A rapid test for the detection of a B-cell marker (D8/17) in rheumatic fever patients. Braz J Med Biol Res 1992; 25:789–794MedlineGoogle Scholar

62 Ganguly NK, Anand IS, Koicha M, et al: Frequency of D8/17 B lymphocyte alloantigen in north Indian patients with rheumatic heart disease. Immunol Cell Biol 1992; 70:9–14Crossref, MedlineGoogle Scholar

63 Taneja V, Mehra NK, Reddy KS, et al: HLA-DR/DQ antigens and reactivity to B cell alloantigen D8/17 in Indian patients with rheumatic heart disease. Circulation 1989; 80:335–340Crossref, MedlineGoogle Scholar

64 Hoekstra PJ, Bijzet J, Limburg PC, et al: Elevated D8/17 expression on B lymphocytes, a marker of rheumatic fever, measured with flow cytometry in tic disorder patients. Am J Psychiatry 2001; 158:605–610Crossref, MedlineGoogle Scholar

65 Murphy TK, Benson N, Zaytoun A, Yang M, Braylan R, Ayoub E, Goodman WK: Progress toward analysis of D8/17 binding to B cells in children with obsessive-compulsive disorder and /or chronic tic disorder J Neuroimmunol 2001; 120:146–151Google Scholar

66 Trifiletti RR, Packard AM: Immune mechanisms in pediatric neuropsychiatric disorders. Tourette's syndrome, OCD, and PANDAS. Child Adolesc Psychiatr Clin N Amer 1999; 8:767–775Crossref, MedlineGoogle Scholar

67 Henry MC, Perlmutter SJ, Swedo SE: Anorexia, OCD, and streptococcus. J Am Acad Child Adolesc Psychiatry 1999; 38:228–229Crossref, MedlineGoogle Scholar

68 Sokol MS, Gray NS: Case study: an infection-triggered, autoimmune subtype of anorexia nervosa. J Am Acad Child Adolesc Psychiatry 1997; 36:1128–1133Crossref, MedlineGoogle Scholar

69 Sokol MS: Infection-triggered anorexia nervosa in children: clinical description of four cases. J Child Adolesc Psychopharmacol 2000; 10:133–145Crossref, MedlineGoogle Scholar

70 Sokol MS, Ward PE, Tamiya H et al: D8/17 expression on B lymphocytes in anorexia nervosa. Am J Psychiatry 2002; 159:1430–1432Crossref, MedlineGoogle Scholar

71 Church et al. 2002 {Church AJ, Cardoso F, Dale RC et al: Anti-basal ganglia antibodies in acute and persistent Sydenham's chorea. Neurology. 2002; 59:227–231Google Scholar

72 Swedo SE, Rapoport JL, Cheslow DL, et al: High prevalence of obsessive-compulsive symptoms in patients with Sydenham's chorea. Am J Psychiatry 1989; 146:246–249Crossref, MedlineGoogle Scholar

73 Swedo SE, Leonard HL, Schapiro MB, et al: Sydenham's chorea: physical and psychological symptoms of St Vitus dance. Pediatrics 1993; 91:706–713MedlineGoogle Scholar

74 Wilson G, Winkelman NE: A clinicopathologic study of acute and chronic chorea. Arch Neurol 1923; 9:170–177CrossrefGoogle Scholar

75 Lougee L, Perlmutter SJ, Nicolson R, 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; 39:1120–1126Crossref, MedlineGoogle Scholar

76 Giedd JN, Rapoport JL, Garvey MA, et al: MRI assessment of children with obsessive-compulsive disorder or tics associated with streptococcal infection. Am J Psychiatry 2000; 157:281–283Crossref, MedlineGoogle Scholar

77 Giedd JN, Rapoport JL, Kruesi MJ, et al: Sydenham's chorea: magnetic resonance imaging of the basal ganglia. Neurology 1995; 45:2199–2202Crossref, MedlineGoogle Scholar

78 Visvanathan K, Trifilette RR, Altemus M, et al: Autoimmune mechanisms in movement disorders. Seminars in Pediatric Neurol 2000; 7:103–107Crossref, MedlineGoogle Scholar

79 Buchanan DN: Pathological changes in chorea. Am J Dis Children 1941; 62:443–445Google Scholar

80 Colony HS, Nalamud N: Sydenham's chorea: a clinicopathologic study. Neurol 1956; 6:672–676Crossref, MedlineGoogle Scholar

81 Castillo M, Kwock L, Arbelaez A: Sydenham's chorea: MRI and proton spectroscopy. Neuroradiol 1999; 41:943–945Crossref, MedlineGoogle Scholar

82 Ryan MM, Antony JH: Cerebral vasculitis in a case of Sydenham's chorea. J Child Neurol 1999; 14:815–818Crossref, MedlineGoogle Scholar

83 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: Guidelines for the diagnosis of rheumatic fever: Jones criteria update. JAMA 1992; 268:2069–2073Crossref, MedlineGoogle Scholar

84 Goldman L, Bennett JC: Cecil Textbook of Medicine, 21st ed. Philadelphia, PA: W. B. Saunders Company, 2000Google Scholar

85 Swedo SE: Sydenham's chorea. A model for childhood autoimmune neuropsychiatric disorders. JAMA 1994; 272:1788–1791Crossref, MedlineGoogle Scholar

86 Swedo SE, Kiessling LS, Leonard HL: A possible neuroimmunologic subtype of obsessive compulsive disorder (OCD), attention deficit hyperactive disorder (ADHD), and Tourette's syndrome (TS). Scientific Abstracts American College of Neuropsychopharmacology 31st Annual Meeting. San Juan, Puerto Rico: ACNP 1992Google Scholar

87 Eisen JL, Leonard HL, Swedo SE et al: The use of antibody D8/ 17 to identify B cells in adults with obsessive-compulsive disorder. Psychiatry Res. 2001; 104:221–225Google Scholar

88 Abraham S, O'Gorman M, Shulman ST: Anti-nuclear antibodies in Sydenham's chorea. Adv Exp Med Biology 1997; 418:153–156Crossref, MedlineGoogle Scholar

89 Kotby AA. El Badawy N. El Sokkary S. et al: Antineuronal antibodies in rheumatic chorea. Clinical & Diagnostic Laboratory Immunology 1998; 5:836–839MedlineGoogle Scholar

90 Harel Z, Hallett J, Riggs S, Vaz R, et al: Antibodies against human putamen in adolescents with anorexia nervosa. Int J Eat Disord 2001; 29463–469Google Scholar