Next Article in Journal
Insight into the Role of Epigenetic Processes in Abiotic and Biotic Stress Response in Wheat and Barley
Previous Article in Journal
CG200745, a Novel HDAC Inhibitor, Attenuates Kidney Fibrosis in a Murine Model of Alport Syndrome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Alterations in the Nervous System and Gut Microbiota after β-Hemolytic Streptococcus Group A Infection—Characteristics and Diagnostic Criteria of PANDAS Recognition

by
Jacek Baj
1,*,
Elżbieta Sitarz
2,
Alicja Forma
2,
Katarzyna Wróblewska
3 and
Hanna Karakuła-Juchnowicz
4,5
1
Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland
2
Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland
3
North London Forensic Service, Chase Farm Hospital, 127 The Ridgeway, Enfield, Middlesex EN2 8JL, UK
4
Chair and 1st Department of Psychiatry, Psychotherapy and Early Intervention, Medical University of Lublin, Gluska Street 1, 20-439 Lublin, Poland
5
Department of Clinical Neuropsychiatry, Medical University of Lublin, Gluska Street 1, 20-439 Lublin, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(4), 1476; https://doi.org/10.3390/ijms21041476
Submission received: 9 February 2020 / Revised: 16 February 2020 / Accepted: 18 February 2020 / Published: 21 February 2020
(This article belongs to the Section Molecular Neurobiology)

Abstract

:
The objective of this paper is to review and summarize conclusions from the available literature regarding Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal Infections (PANDAS). The authors have independently reviewed articles from 1977 onwards, primarily focusing on the etiopathology, symptoms, differentiation between similar psychiatric conditions, immunological reactions, alterations in the nervous system and gut microbiota, genetics, and the available treatment for PANDAS. Recent research indicates that PANDAS patients show noticeable alterations within the structures of the central nervous system, including caudate, putamen, globus pallidus, and striatum, as well as bilateral and lentiform nuclei. Likewise, the presence of autoantibodies that interact with basal ganglia was observed in PANDAS patients. Several studies also suggest a relationship between the presence of obsessive-compulsive disorders like PANDAS and alterations to the gut microbiota. Further, genetic predispositions—including variations in the MBL gene and TNF-α—seem to be relevant regarding PANDAS syndrome. Even though the literature is still scarce, the authors have attempted to provide a thorough insight into the PANDAS syndrome, bearing in mind the diagnostic difficulties of this condition.

1. From Acute Pharyngitis to Rheumatic Fever

Acute pharyngitis and tonsillitis may be induced by various bacterial and viral organisms, even though β-hemolytic Streptococci group A remains the most common causation so far [1,2,3]. Streptococcus pyogenes (β-hemolytic Streptococcus group A) (GAS) is responsible for the majority of bacterial infections in children [4]. The highest infection rate is estimated to occur during late autumn, winter, and early spring. An infected human constitutes a reservoir of pathogenic bacteria and a potential source of infection. GAS infection can be spread by airborne transmission or through direct contact with an infected individual. Streptococcal pharyngitis is characterized by the following symptoms: rapid and acute onset, sore throat, pain during swallowing, headache, nausea, vomiting, fever, intensely red-coloured oral mucosa with swelling, as well as painful and enlarged anterior cervical lymph nodes [5]. Coughing and rhinitis are not common symptoms of streptococcal infection. Post-infectious streptococcal complications mainly include peritonsillar abscesses, purulent otitis media, or paranasal sinusitis [6]. Further, streptococcal toxic shock syndrome, post-streptococcal acute glomerulonephritis, acute rheumatic fever, rheumatic heart disease, or post-streptococcal autoimmune neuropsychiatric disorders (so-called Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal Infections, or PANDAS) may be induced by autoimmune responses [7]. There are also incidents of non-streptococcal infections by the Bornavirus or Mycoplasma pneumoniae that are implicated in the onset of symptoms similar to those in PANDAS [8,9,10]. Exacerbations of PANDAS symptoms are observed in 30% of cases due to GAS infections; 20% are because of non-streptococcal illnesses and approximately half of the cases are because of a non-identified factor [11].

2. Rheumatic Fever and OCD

Rheumatic fever is an acute autoimmune disease constituting a relatively late complication of GAS infection [12,13]. The Jones criteria for rheumatic fever include major criteria (carditis, arthritis, chorea, erythema marginatum, subcutaneous nodules) and minor criteria (polyarthralgia, fever, prolonged PR interval, erythrocyte sedimentation rate (ESR) ≥60 mm, C-reactive protein (CRP) ≥3.0 mg/dL) [14]. The symptoms of rheumatic fever appear usually two-to-three weeks after pharyngitis and include the inflammation of large joints, myocarditis, marginal erythema, subcutaneous nodules, or even incidents of Sydenham’s chorea [15,16]. The number of acute rheumatic fever incidents occurring as a side effect of streptococcal infection equals 5%–6% and depends on the susceptibility of an infected individual, the duration of GAS exposure, and the type of treatment therapy (or even lack of appropriate treatment) [17]. The pathomechanism of rheumatic fever involves immunological reactions directed towards specific epitopes with a structure similar to the proteins present in the myocardium, heart valves, synovium, and skin, as well as in the hypothalamus and caudate nucleus [18,19].
Approximately 2%–4% of children with rheumatic fever are prone to developing OCD, which is also associated with the progressive damage of basal ganglia [20,21]. This phenomenon shows a male predominance and may also manifest as tics, Tourette’s syndrome, or attention-deficit/hyperactivity disorder (ADHD) [22]. Obsessive-compulsive disorders (OCD) might also accompany Sydenham’s chorea. Sydenham’s chorea manifests in the presence of unilateral involuntary movements—mainly of facial and limb muscles—general weakness, and emotional instability [23]. The psychiatric symptoms may appear insidiously and include emotional instability, irritability, anxiety, and inappropriate behavior in general. Additionally, affected children may present progressively worse results in school. In both PANDAS and Sydenham’s chorea, some new evidence supports the concept of autoantibody mimicry mechanisms [24,25]. The antibodies, which induce either rheumatic fever or PANDAS, are cross-reactive with the N-acetyl-βD-glucosamine dominant epitope specifically [26].

3. PANDAS and OCD—Where Is the Line?

The first description of PANDAS was presented in 1998 by Swedo et al. based on the 50 cases she reported, defining it as the sudden beginning of obsessive-compulsive or tic disorders’ symptoms due to the complications of GAS infection [27]. The rapid onset of such symptoms hinders the differentiation between OCD and PANDAS [28]. PANDAS involves various diagnostic implications since the profile of symptoms seems to be similar to other streptococcal-associated conditions, like OCD, tics, or motor hyperactivity [29,30]. Further, the majority of PANDAS cases described by Swedo included the presence of insignificant piano-playing choreiform movements of fingers and toes, which may be overlooked [31]. Symptoms similar to PANDAS may present not only in children but also among adults as a side effect of post-streptococcal disease [32]. Besides sore throats due to the GAS infection, there are cases of streptococcal skin infections, which may also be associated with the onset of PANDAS [33]. The OCD symptoms constitute the foundational criteria for PANDAS recognition, as well. It was estimated that the post-streptococcal autoimmunity mechanism is responsible for up to 10% of cases of childhood OCD [19]. To resolve the diagnostic issue, it was hypothesized that PANDAS constitutes a subtype of children’s OCD, with the etiology of GAS infection. The legitimacy of PANDAS as a subgroup of OCD was supported by the study performed by Jaspers-Fayer et al. in 2017 [34].

PANDAS Symptoms

PANDAS mostly occurs in children or adolescents and its prevalence is significantly higher in males [35,36,37,38,39,40,41]. PANDAS is characterized by impairments within basal ganglia, which are responsible for the switching of both motor and mental behaviours, resulting in novel behaviour [42]. PANDAS children present characteristic symptoms such as tics, hyperactivity, urinary urgency, impulsivity, anxiety, impulsiveness, eating disorders, and a significant decline in school performance along with the deterioration of handwriting [43,44]. Usually, children with PANDAS show significant and rapid (between 24 and 72 h) changes in behavior, shifting from high-functioning and well-adjusted to psychotic symptoms which may disrupt children’s functioning [45]. On average, anxiety and other mood symptoms are more severe compared to somatic and functional symptoms like stomach, muscle, or joint pain [46].
There is a positive correlation between upper respiratory infections, the sudden onset of OCD, and PANDAS symptoms [47]. Nonetheless, other risk factors including the age of onset, neurologic impairments, family history of either autoimmune disorders or rheumatic fever, the presence of comorbidities or the severity of OCD symptoms seem not to be correlated with the presence of upper respiratory infections [47,48,49].
According to Leon et al. (2017), a complete PANDAS remission is, on average, 3.3 years [11]. Moreover, it was observed that the majority of children (approximately 72%) showed at least one exacerbation of PANDAS symptoms throughout the period of gradual remission. Among PANDAS cohorts, psychiatric comorbidities included ADHD (40%), oppositional defiant disorder (40%), and depression (36%). Less common were enuresis (20%) or dysthymia (12%) [50]. ADHD-like behavioral changes are common side effects of concurrent or prior streptococcal infections [51]. Male predominance was observed in ADHD and PANDAS; males are also more prone to recurrent GAS infections [52]. PANDAS symptoms may be similar to those which are present in other psychiatric disorders, including OCD, Sydenham’s chorea, Tourette’s syndrome, or Tumor Necrosis Factor Receptor Associated Periodic Syndrome (TRAPS) [53].

4. PANDAS Diagnostic Guidelines

Guidelines regarding diagnostic criteria for PANDAS were improved in 2017, as it includes:
  • Presence of OCD and/or tics, mainly multiple ones, complex or not observable in other disorders
  • Specific period of childhood and age (symptoms of PANDAS are more common between the age of 3 years old and period of puberty)
  • Acute onset and episodic changes in behavior
  • Association with infection of Streptococci group A
  • Coexisting neurological impairments [54,55].

4.1. The Presence of OCD and/or Tics

Obsessions, compulsions and/or tics should be severe to meet either OCD or tics criteria and distort patients functioning abilities comparing to the state before the disease.

4.2. Specific Period of Childhood and Age

Symptoms are most frequently observed between the age of 3 and puberty. The early onset of PANDAS is associated with the time of the highest rate of GAS exposition (early childhood). The onset of PANDAS after puberty is possible, nevertheless, it forms only in a minor percentage of cases.

4.3. Acute Onset and Episodic Changes in Behavior

Clinical outcome is characterized by the rapid and dramatic onset of OCD, plus tics. The beginning of specific symptoms may usually be assigned to a particular day or week, which confirms that the onset is relatively rapid and obvious. The prolonged duration of the streptococcal infection could worsen the clinical outcome. Nevertheless, to comply with this criterion, deterioration of tics must be of such severity as to restrain the patient. The varying severity of symptoms depends on the episode of the disease. Moreover, because children are potential carriers of various range of pathogens, the symptomatic period might slightly be prolonged depending on the patient.

4.4. Association with Infection of Streptococci Group A

The exacerbation of symptoms must be associated with the infection of group A Streptococci, which needs to be confirmed with the positive swab from the throat and/or increased titers of antistreptolysin-O (ASO) or anti-DNase B [56,57]. In the case of PANDAS, GAS infection usually occurs without macroscopic changes in the throat and symptoms of pharyngitis. It is recommended to provide a 24–48 h agar culture to confirm the colonization of GAS [58]. Positive findings may indicate the carrier state of GAS in a patient without any specific immunological reactions. With regards to PANDAS, it was detected that neuropsychiatric symptoms are associated with the presence of immunological reactions, and can be eliminated after the removal of antibodies. The increase of the titers of either ASO or anti-DNase B can be used in the detection of previous infections of GAS.
Therefore, the usage of this diagnostic method could slightly be misleading due to a number of reasons:
  • Titers of either ASO or anti-DNase B can remain at a high level even for many months after GAS infection, which might present as a false positive result.
  • Approximately 40% of children with GAS infection do not present the increased levels of ASO or anti-DNase B which is a false negative result.
  • Time constitutes an essential critical factor in the determination of the increased levels (approximately 2–4 times higher) of ASO, and anti-DNase B after one-four and six-eight weeks correspondingly.
  • When a child with specific symptoms presents negative results from a throat swab, the levels of basic anti-streptococcal antibodies should be provided.
Studies have shown that after initial infection, disease exacerbations could be associated with factors other than GAS, such as other bacterial or viral infections, or internal stimuli like stress [59].

4.5. Coexisting Neurological Impairments

During the exacerbation of symptoms, patients show inappropriate results from the neurologic examination. The most common impairments include hyperactivity or involuntary movements (tics or chorea). It is essential to differentiate between PANDAS and Sydenham’s chorea since the last one is the common symptom of rheumatic fever and present different, separate characteristics and treatment strategies. Approximately 30% of patients with either OCD or PANDAS present choreiform movements that are similar to those in Sydenham’s chorea, which eventually might raise diagnostic issues (Table 1) [22].
Etiologically in both PANDAS and Sydenham’s chorea present as a side effect of GAS infection, nevertheless, besides the presence of tics, which is common for both diseases, Sydenham’s chorea appears with more severe obsessive-compulsive syndromes and definite chorea with the presence of hypotonia [93,94]. Moreover, Sydenham’s chorea usually presents with complete remissions and a duration less than one year, whereas the PANDAS course is likely to be more chronic [95]. Furthermore, PANDAS found to be more prevalent in men, whereas Sydenham’s chorea in women. Regarding the streptococcal M-protein subtypes—PANDAS is associated with M1, M3, M11, M12, and M13, whereas Sydenham’s chorea—M5, M6, and M19 [96].
Diagnostic criteria for PANDAS require the presence of OCD or tics in general, nonetheless, neuropsychiatric symptoms are also commonly present among patients. They appear simultaneously with OCD or tics, and similarly present sudden onset and exacerbation. It has been shown that the presence of somatic disturbances like higher frequency of urination, pupil dilatation, and insomnia aids in the differentiation between PANDAS and OCD, or Tourette’s syndrome [97]. The affected children may present signs of inattention, impulsiveness, emotional lability for days, weeks, or even months after remission of OCD and/or tics. In some cases, a child can experience constant anxiety and fear as well as OCD, whereas other symptoms can gradually disappear. Accurate recognition and diagnosis of PANDAS need to fulfill all of the 5 aforementioned criteria.

5. PANDAS vs. PANS

A Pediatric Acute-Onset Neuropsychiatric Syndrome (PANS), is a condition characterized by sudden onset of OCD, tics or disturbed food intake along with various psychiatric distortions, which is similar to PANDAS [71]. Unlike PANDAS, PANS syndrome does not always require previous GAS infection (Figure 1).
Actual criteria of PANS classification include:
  • Rapid onset or recurrence of OCD or abnormalities associated with food intake.
  • Rapid onset of coexisting neuropsychiatric disorders (at least two of them):
    • Increased anxiety levels and/or fear associated with the separation from parents
    • Increased motility or motor dysfunctions (including tics and dysgraphia)
    • Behavioral regress
    • Rapid decline of grades among children in school
    • Emotional lability (irritability, aggression and/or oppositional behaviours)
    • Dysuric symptoms
    • Somatic signs (including insomnia)
  • Present symptoms were not explained and associated with the known neurological or medical conditions [98].
PANS symptoms are induced by the immunological reactions associated with one to several bacteria, or viral infections [84]. These mainly include angina, atypical pneumonia caused by Mycoplasma pneumoniae, influenza, upper airway inflammation, or sinusitis [99]. Psychosocial stress may also contribute to the progression of symptoms [54,100].

The Pathophysiology of GAS Infection and PANDAS

Infectious agents such as GAS could play a role in the immune etiopathogenesis via the activation of various cytokine cascades [101]. Preliminary studies also showed altered IgM, IgA, and IgG subclasses profiles, and the decreased T cell count in PANDAS and Tourette’s syndrome [102]. It has also been reported by Morer et al. that genetic factors may be related to the increase in the D8/17 positive B lymphocytes subpopulation that could trigger the vulnerability to PANDAS [103]. Thus, authors have also suggested that D8/17 could be a potential marker of susceptibility to PANDAS. Sokol et al. observed that PANDAS anorexia nervosa patients were far more likely to have peripheral B lymphocytes expressing D8/17 comparing to patients without PANDAS-characteristics [104].
M protein is a major virulence factor of GAS, which contains brain-cross-reactive epitopes and involved in the pathogenesis of Sydenham’s chorea [105]. M5 and M6 antigens of two separate streptococcal strains were detected in the human brain via immunofluorescence staining patterns [106]. Additionally, M5 was mostly associated with the caudate region, whereas the presence of M6 was more diffuse. This finding suggests that M proteins may trigger the anti-brain antibodies enhancing the severity of symptoms and clinical course [107]. It was observed that neonatal corticosterone contributes to the increase of anti-inflammatory IL-9 in both peripheral and central nervous system in mice model, which may mitigate symptoms of streptococcal infection [108].
Recent research has shown that dopamine levels may contribute to the immunological mechanisms involved in PANDAS pathogenesis [109,110,111]. Altered levels of dopamine may also be an explanation of the decrease of Treg levels and elevations in proinflammatory cytokines [112,113,114,115]. Moreover, it was observed that among PANDAS patients, vitamin D deficiency was significantly more frequent in comparison to the control groups [78]. This finding may lead to the conclusion that vitamin D can be partially associated with the diagnostic criteria of PANDAS or OCD patients, nevertheless, more research associated with this topic should be completed.

6. PANDAS and Alterations in the Nervous System

The majority of OCD and PANDAS patients present neuropsychiatric symptoms associated with the impaired functions of either the central or peripheral nervous system, or both [116]. The pathophysiology of PANDAS is mainly associated with the cross-reactions of the antibodies with both streptococcal N-acetyl-beta-D-glucosamine and neuronal lysoganglioside and tubulin [117,118]. PANDAS involves the impairments within the cortico-basal ganglia circuitry [119].

6.1. Striatum and Striatal Interneurons

PANDAS patients exhibited significantly enlarged volumes of striatum in comparison to control groups, according to Giedd et al. [62]. The binding of serum antibodies obtained from children diagnosed with PANDAS to cholinergic interneurons in the striatum of mice was significantly higher in comparison to control groups [64]. This finding may present one of the potential loci of the pathophysiology of PANDAS. Cholinergic interneurons are involved in the pathophysiology of tic disorders as well as Tourette’s syndrome, which is indicated by the decreased number of ChAT-positive interneurons in the putamen and caudate [120]. Since the comorbidity of PANDAS and tic disorder is estimated at approximately 60%, the aforementioned finding may be applicable in PANDAS, as well. The reviewed literature also suggests the role of striatal cholinergic interneurons in the progression of PANDAS. The dysregulation within striatal interneurons, including antibody deposition, can lead to impairments in behavior because of the strict association (either direct or indirect) of the interneurons with the basal ganglia [121]. Moreover, it was reported that cholinergic interneurons express D2 dopamine receptors, which are also involved in the binding of antibodies present in PANDAS [122,123,124].

6.2. Microglia

Recent studies showed the relevance of microglia in the etiology of PANDAS. Microglia cells play a major role to mediate immune responses in the central nervous system. Nevertheless, it was shown that besides immunological reactions, microglia are involved in brain development, homeostasis, brain plasticity, and adult neurogenesis [125,126]. PET imaging with the usage of 11C-[R]-PK-11195, which is a ligand that binds to the transporter protein expressed by active microglia, showed the increased striatal volumes of children with PANDAS [65]. Furthermore, 11C-[R]-PK-11195 binding was far more intensified in the striatum of children with PANDAS compared to the group of healthy children. Interestingly, the inflammation was more broadly spread and intense mainly in the bilateral caudate and lentiform nucleus. Repeated intranasal GAS infections could result in an elevated number of fCD68+/Iba1+ activated microglia in the glomerular layer of the olfactory bulb [127]. Moreover, it was also hypothesized that GAS antigens could be presented to Th17 cells by the means of the microglia. Synaptic pruning may be elevated in PANDAS, nevertheless, recent research is still restricted to animal models [65].

6.3. Basal Ganglia and Antibodies

The first evidence of the presence of antineuronal antibodies in humans was reported by Husby et al. (1977), in the case of Sydenham’s chorea [128]. The presence of antineuronal antibodies and their impact on Sydenham’s chorea was supported by Kirvan et al. (2003) [129,130,131]. It was shown that monoclonal antibodies signal neuronal cells and stimulate them to further release of dopamine and activate CaM kinase II [129]. Several animal models demonstrated behavioral symptoms induced by the deposition of antibodies in the brain and basal ganglia in particular after the immunization with GAS or M18 strain of S. pyogenes [132,133]. Furthermore, it was proven that recipient mice or rats may develop PANDAS or Sydenham’s chorea by means of the anti-streptococcal antibodies transfer [134]. Animal models showed that antibodies were deposited in the basal ganglia and thalamus specifically, but not in the cerebellum or hippocampus. In a study of a human Sydenham’s chorea, Cox et al. (2013), demonstrated that putting human V genes for the chorea antibody into transgenic mice, which were then expressed in their B cells, produced the antibodies in serum of the mice [123]. The antibodies penetrated the blood-brain barrier and targeted the basal ganglia, substantia nigra or the ventral tegmental area.
The presence of anti-basal ganglia antibodies in sera of PANDAS patients is a highly specific and sensitive marker in comparison to control, non-affected groups [135]. Antineuronal antibodies that may contribute to the progression of PANDAS symptoms include antipyruvate kinase antibodies, anti-dopamine receptor antibody, and antilysoganglioside GM1 antibody [70,117,129,136,137,138]. In a study performed by Pavone et al. (2004), anti-basal ganglia antibodies were present in nearly two-thirds of patients affected by PANDAS [139]. Additionally, the samples of sera that were obtained from PANDAS patients showed significantly higher activation of CaM kinase II in comparison to healthy controls [140]. The higher activation of CaM kinase II can be associated with the progression of choreic movements in affected individuals. Interestingly, those children who have developed a sore throat due to the streptococcal infection and were treated with antibiotics such as penicillin or azithromycin, showed that they were not at the higher risk of the development of tics or obsessive-compulsive disorders [51].
In general, OCD children present with enlarged volumes of the corpus striatum and basal ganglia, as well as an increased number of antineuronal antibody titers [63]. The size of basal ganglia is strictly associated with the total number of antibodies present during GAS infection [141,142]. Furthermore, Cabrera et al. found a greater volume of grey matter within basal ganglia and a reduced volume of white matter in this area [143]. It was reported that the infection of GAS is correlated with the production of antineuronal antibodies. The presence of streptococcal antineuronal antibodies can be associated with the neuronal damage as these antibodies interact with structures of the nervous system, as a result of the antibody mimicry [12,144,145,146,147]. There is no significant correlation between the size of the basal ganglia and the severity of symptoms among the studied groups. The neuroimaging studies are consistent with the pathological immune reactions of the antibodies reacting with the basal ganglia leading to their altered volumes. Even some observable variations of the basal ganglia volumes are present and observable during examinations, the knowledge is still restricted to the potential functional changes, which may be present within basal ganglia due to the aforementioned impairments. Therefore, there was no study strictly associated with the alterations of the basal ganglia functions specifically; it could be the potential area of interest regarding future studies associated with PANDAS.

6.4. Other Structures in CNS

Giedd et al. (2000), demonstrated that children with streptococcal OCD present with increased sizes of the caudate, putamen, and globus pallidus, whereas the average size of thalamus and total cerebrum remains unchanged [62]. Further studies proved the aforementioned results showing that all of the previous structures besides thalamus are enlarged in the case of PANDAS patients. The caudate and putamen are specifically enlarged most extensively in the acute phase of PANDAS. This research may also indicate the potential localization of the pathophysiological mechanisms of this syndrome. Besides the alterations that were observable using Magnetic Resonance Imaging (MRI), the study with the usage of Positron Emission Tomography (PET) showed that PANDAS patients tend to present with a higher percentage of binding potential values in bilateral caudate and lentiform nuclei compared to the control group [148].

7. The Characterization of Gut Microbiota

7.1. The Impact of Gut Microbiota on the CNS

There is an increasing amount of research that presents the relationship between the gut microbiome and the development of psychiatric disorders [149,150]. The most recent data indicates that gastrointestinal homeostasis significantly affects the development and functioning of the central nervous system, which presents the so-called microbiota–gut–brain axis as the new paradigm in neuroscience [151]. Neuropsychiatric disorders where the etiopathology can be related to the gastrointestinal microbiota include anxiety, autism, anorexia nervosa, Parkinson’s disease, Alzheimer’s disease, ADHD, schizophrenia, alcohol dependence, bipolar disorders, or migraine pain [82,100,152,153,154,155,156,157,158,159,160,161]. Interestingly, the microbiota-gut-brain axis starts to develop during the intrauterine period of fetal development. The axis is affected by both intrauterine and extrauterine factors including antimicrobial treatments of the mother, vaccinations, increased contact with chemicals, diet, type of child delivery, feeding habits during infancy period, as well as exposure to bacterial, viral or parasite infections and maternal stress during pregnancy [162,163]. Moreover, the stages of gut microbiome development occur simultaneously with the neurodevelopment. The connection between gut microbiota and central nervous systems occurs by several pathways, which involve microbial metabolites (short-chain fatty acids), immune cells, tryptophan metabolism along with neural (vagus nerve), and endocrine pathways (the hypothalamic-pituitary-adrenal axis) [164,165]. Bacterial commensals in the gastrointestinal tract are also responsible for the production of γ-aminobutyric acid, which is the main inhibitory neurotransmitter in the central nervous system and may induce neuropsychiatric symptoms [166,167,168,169,170,171,172]. Dopamine, noradrenaline, and histamine, as well as brain-derived neurotrophic factor (BDNF) levels, can also be affected by gut microbiota [173].

7.2. Alterations in Gut Microbiota in PANDAS

Quagliariello et al. performed a study to investigate the bidirectional connection between the gut microbiota and the central nervous system in PANDAS patients [66]. A group of 30 PANS/PANDAS patients aged 4–16 (20 males and 10 females) were studied in terms of microbiota that colonizing their gastrointestinal tracts. The group of 4–8 years old patients presented the altered abundance of phylum levels including increased Bacteroidetes and lower Firmicutes. This group of patients also presented with lower distribution of Actinobacteria and the total absence of the TM7 phylum (Saccharibacteria). Regarding family levels, the group of 4–8 years old patients presented with higher levels of Bacteroidaceae, Rikenellaceae, and Odoribacteriaceae. Contrarily, some of the Firmicutes families including Turicibacteraceae, Tissierellaceae, Gemellaceae, and Carnobacteriaceae (Bacilli class), Corynebacteriaceae and Lachnospiracea (Clostridia class) were absent. Nevertheless, within Firmicutes phylum, Turicibacteriaceae, Erysipelotrichaceae, and Lachnospiraceae are considered to be potential characteristic gut microbiota in PANDAS patients even if their distribution was at a minor level. Furthermore, Bacteroidaceae were present greatly in the gastrointestinal tract of PANDAS patients. Moreover, a strong negative correlation between increased levels of Bacteroidaceae and other bacterial families has been stated. On the other hand, PANDAS patients above 9 years old presented with a greater abundance of Peptostreptococcaceae and Erysipelotrichaceae and lowered levels of Rikenellaceae and Barnesiellacea. The discrepancy between gut microbiota in two aforementioned groups may occur because of a different age of PANDAS patients as microbial diversity in the gastrointestinal tract significantly differs throughout the lifetime [174,175,176,177]. Many treatment therapies especially antibiotic drugs significantly affect gut microbiota and function, which constitute a limitation preventing the detection of possible microbial markers associated with PANDAS [178].
According to the authors’ knowledge, it is the first detailed study of gut microbiota specifically in PANDAS patients, therefore more research should be accomplished to compare the results of studies and provide further information.
Nevertheless, as it was stated in this study, alterations between levels of gut microbiota were clearly observable in cases of PANDAS patients of different age. This observation may lead to the conclusion that alterations in the gut microbiota constitute one of the factors enhancing PANDAS development, as well as the potential marker of further diagnostic procedures of this syndrome.

8. Genetic Approach to OCD, Including PANDAS

With regards to the genetic approach to the understanding of the pathophysiology of PANDAS, the studies are still scarce and usually limited. Murphy et al. in 2010 reported among examined biological mothers of 107 children with either OCD, tics or both, the rate of autoimmune diseases was 17.8%, whereas in the general population the rate was equal to 5% [179]. This finding may present the potential genetic aspect considering the development and possibly the progression of PANDAS patients, as well since PANDAS is considered to be the subtype of OCD. In other disorders like Tourette syndrome, which also present various intensity and severity of tics, several mutations including Slit and Trk-like family member 1 (SLITRK1) gene are present in rare cases [180]. Interestingly, the expression of this gene is strictly associated with the cholinergic interneurons in the adult striatum. Additionally, regarding Tourette syndrome, acetylcholine-related genes seem to correlate with the severity of this disorder [181]. Even though the aforementioned results were not studied in the case of PANDAS patients, it should be considered that the comorbidity of Tourette syndrome and OCD along with OCD and PANDAS is equal to 50% and 60% correspondingly. Though patients with Tourette syndrome present with elevated levels of immune gene expression restricted to basal ganglia specifically, these results are still not clear in the case of PANDAS patients. Mannose-binding lectin (MBL) plays an important role in immune responses. It was stated that GAS show a high susceptibility to bind to MBL [182]. Any variant in exon 1 of the MBL2 gene increases the probability of PANDAS [67]. Interestingly, recurrent infections may be associated with the polymorphism of codon 54 of the MBL2 gene [183]. MBL2 polymorphism is also suspected to play a role in the pathogenesis of rheumatic heart disease and rheumatic fever [184]. Therefore, the MBL2 gene is suspected to be one of the diagnostic markers of PANDAS. According to Luleyap et al., TNF-α −308 AA polymorphism can be regarded as a molecular indicator of PANDAS [68]. Altered levels of TNF-α can be associated with exacerbation of PANDAS symptoms and facilitating autoimmune responses within the central nervous system. There is not much research done regarding genetic aspects associated with PANDAS. Even though, several pieces of research indicate the relationships between particular genetic variations and the onset of PANDAS.

9. Treatment of PANDAS

9.1. Antibiotic Therapy

Treatment of PANDAS is to a large extent restricted to its severity including symptoms, exacerbations, and trajectory [79]. PANDAS treatment therapy mainly includes psychoactive drugs, immunotherapeutics with steroids, antibiotics, plasmapheresis as well as intravenous immunoglobins [80,185,186,187,188]. With regard to the proper treatment of PANDAS, it should be considered that the application of penicillin or other antibiotics alone does not constitute the only way of treatment of this disease [171]. It is because penicillin is useful only in the cases of active streptococcal infection to eradicate bacteria. Nevertheless, in cases of persistent and active infection of GAS, immediate elimination of the infection source should be provided to minimize the severity of PANDAS. Moreover, the most recent research showed that neuropsychiatric exacerbations can be minimized by the application of penicillin V and azithromycin even after the one-year duration of PANDAS [72]. The main antibiotic used in the streptococcal eradication is penicillin, but additional drugs such as amoxicillin or azithromycin are also applied [79]. Some research suggests that azithromycin can also be helpful in the alleviation of PANDAS symptoms, nevertheless, its application should be strictly controlled because of the potential side effects mainly in the form of cardiac risks [73]. According to the most recent research, it is more common to provide treatment in the form of antibiotics rather than the application of psychiatric treatments including cognitive-behavioral therapy [189,190].

9.2. Psychoactive Drugs

Treatment of PANDAS is a complex process that includes not only psychoactive medications but additionally behavioral-cognitive therapy which is offered mainly to those children who present with high levels of distress and anxiety [186,191]. The cognitive-behavioral intervention seems to be a less invasive and expensive way of treatment in comparison to antibiotic therapy or selective serotonin reuptake inhibitors (SSRIs) [77]. Even though, clinicians should take into consideration the possibility of combining the aforementioned techniques to obtain better clinical outcomes among PANDAS patients. Antipsychotic drugs including risperidone can be applied in cases of severe behavioral symptoms [74]. Unfortunately, only a small number of studies researched the effects of psychiatric therapies in PANDAS. It was stated that among examined patients, the usage of SSRIs resulted in the improvement of the OCD symptoms [192,193,194]. Patients with PANDAS who also present with the ADHD symptoms can be prescribed atomoxetine [195]. One of the relatives of benzodiazepine, lorazepam, shows the potential in PANDAS treatment since it improves the motor activity, expressive language, as well as the posturing of patients [75]. Such symptoms like mood lability or compulsion to void, which are not alleviated by lorazepam, appeared to be improved by the means of plasmapheresis application.

9.3. Nonsteroidal Anti-Inflammatory Drugs

In the observational study performed by Spartz et al. in 2017, it was reported that the usage of the nonsteroidal anti-inflammatory drugs (NSAIDs) in PANDAS pediatric patients, provided different and in some cases contradictory results. Approximately 31% of examined patients show improvements regarding OCD and tic symptoms. Contradictorily, 35% of patients present the aggravation of tics severity and patients’ mood. So far, it is unknown why the examined patients present different therapeutic effects of NSAIDs. However, both prophylactic and early usage of NSAIDs is associated with the shorter duration of the exacerbation of PANDAS symptoms [196]. Because of the wide range of the results which are available at the actual state of knowledge, nevertheless, additional research trials should be done to specify the role of NSAIDs application in PANDAS patients.

9.4. Tonsillectomy

There is only a small number of reports regarding the application of tonsillectomy to eradicate symptoms of PANDAS. In 2011, Alexander et al. reported a case of a 9-year-old boy with PANDAS who presented with frequent and recurrent streptococcal infections, which were resolved just after tonsillectomy [197]. The positive clinical outcomes after tonsillectomy could be observable not only because of the removal of tonsils but also because of proper medications provided just after the tonsillectomy procedure [198]. Tonsillectomy is a reasonable procedure, mainly in those cases when the usage of antibiotics does not bring positive clinical outcomes and in the recurrent GAS-associated tonsillitis. This can also be an alternative way of treatment in the severe cases of patients who present with multiple resistance to several groups of antibiotics. In the study performed by Demesh et al. in 2015, 33% of children who had a tonsillectomy, presented with the complete resolution of neuropsychiatric symptoms [199]. In those children, whose symptoms were not resolved completely, it was observed that the severity of symptoms was significantly reduced. Even though, there are still cases of increased morbidity even after tonsillectomy procedure [198]. Due to the limited research in this area, it is still unknown whether tonsillectomy should be treated as a preventive procedure of PANDAS symptoms, or rather than the second-line therapy for those patients who present with a poor clinical outcome during antibiotic treatment.

9.5. Other Treatment Strategies

There is still, there is a lack of consistent evidence in PANDAS treatment with antimicrobials and immunotherapy as well [200]. Intravenous immunoglobulin therapy (IVIG) was observed to decrease the number of pathogenic antibodies in PANDAS patients alleviating OCD symptoms [201]. It is advised to follow the immunotherapy as well, which could decrease the side effects of neuroinflammation and autoimmunity associated with streptococcal infection. Milder symptoms of the disease can be treated with the usage of non-steroidal anti-inflammatory drugs, whereas more severe symptoms are treated with intravenous immunoglobulins. Plasma exchange can be applied in cases when symptoms are of such intense severity that may be life-threatening [202]. Application of plasmapheresis may also lead to the stabilization of the basal ganglia volumes, which are altered during the disease [141,203,204,205]. Since vitamin D metabolism has been observed to play a role in the pathogenesis of PANDAS, supplements of this vitamin can be useful in the reduction of psychiatric symptoms of patients [206,207].

10. Conclusions

Even though the concept of PANDAS was first introduced in 1998 by Swedo et al. it is still difficult to unequivocally distinguish its symptoms from a typical OCD or a tic disorder. To provide the most accurate diagnosis, several aspects of PANDAS etiopathology should be taken into consideration, including the infection of GAS, rapid onset, genetic predispositions, environmental factors, or even the coexistence of other diseases—mainly psychiatric ones—which can enhance PANDAS symptoms [208]. Moreover, it should be noted that immune factors are not only the markers of the disease, but they seem to be pathogenic, as well [140]. PANDAS has a male predominance according to the studies. With regard to alterations in the nervous system in PANDAS patients, several important observations have been reported. Various neuronal proteins, including tubulin, lysoganglioside, and dopamine receptors are affected by the antibodies produced in the course of PANDAS. As a consequence, some alterations of cortico-basal ganglia circuity are observed [209]. The deposits of antibodies are also accumulated in the striatal interneurons. PANDAS patients present significantly enlarged volumes of corpus striatum, caudate, putamen, globus pallidus, and basal ganglia. Since the activated microglia were found in close proximity to CD4+ T cells, it was assumed that local microglia might present GAS antigens to Th17 cells [65]. Besides manifestations from the central nervous system, noticeable changes in the presence and distribution of specific microbiota in the gastrointestinal tract were observed in PANDAS patients [66]. This might have an indirect impact on behavior due to the reduced production of various metabolites involved in brain functions. Further, exon variants of the MBL gene and altered levels of TNF-α increase the susceptibility of PANDAS [67,68]. The treatment of PANDAS syndrome is still not obvious and specified, however, there are several alternatives that show promising results. Among them are antibiotics, immunomodulatory drugs, psychiatric drugs, and behavioral interventions.

Author Contributions

Conceptualization, J.B.; methodology, H.K.-J. and E.S.; writing—original draft preparation, J.B., E.S., A.F.; writing—review and editing, J.B., E.S., A.F., K.W., H.K.-J.; supervision, H.K.-J.; project administration, J.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

PANDASPediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal Infections
PANSPediatric Acute-Onset Neuropsychiatric Syndrome
MBLMannose-binding lectin
TNF-αTumor necrosis factor alpha
OCDObsessive-compulsive disorders
ADHDAttention-Deficit/Hyperactivity Disorder
GASGroup A Streptococci
CaM kinase IICalmodulin-dependent protein kinase II
IgMImmunoglobulin M
TRAPSTumor Necrosis Factor Receptor Associated Periodic Syndrome
ASOAntistreptolysin-O
SLITRK1Slit and Trk-like family member 1
CELSR3Cadherin EGF LAG Seven-Pass G-Type Receptor 3
FLT3FMS-like receptor tyrosine kinase-3
COL27A1Collagen type XXVII alpha 1 chain
SLITRK5SLIT and NTRK-like protein 5
HDCHistidine decarboxylase
NRXN1Neurexin 1
CNTN6Contactin 6
DRD3Dopamine Receptor D3
GDNFGlial cell-derived neurotrophic factor
KCNJ5Potassium Voltage-Gated Channel Subfamily J Member 5
AADACArylacetamide Deacetylase
HCN4Potassium/sodium hyperpolarization-activated cyclic nucleotide-gated channel 4
SSRISelective Serotonin Reuptake Inhibitor
MRIMagnetic Resonance Imaging
PETPositron Emission Tomography
IVIGIntravenous immunoglobulin therapy
NSAIDNonsteroidal anti-inflammatory drug
LPSLipopolysaccharide
NOX2NADPH oxidase 2
NDNo data

References

  1. Anjos, L.M.M.; Marcondes, M.B.; Lima, M.F.; Mondelli, A.L.; Okoshi, M.P. Streptococcal acute pharyngitis. Rev. Soc. Bras. Med. Trop. 2014, 47, 409–413. [Google Scholar] [CrossRef]
  2. Ivaska, L.; Niemela, J.; Lempainen, J.; Osterback, R.; Waris, M.; Vuorinen, T.; Hytonen, J.; Rantakokko-Jalava, K.; Peltola, V. Aetiology of febrile pharyngitis in children: Potential of myxovirus resistance protein A (MxA) as a biomarker of viral infection. J. Infect. 2017, 74, 385–392. [Google Scholar] [CrossRef]
  3. Bisno, A.L. Acute Pharyngitis. N. Engl. J. Med. 2001, 344, 205–211. [Google Scholar] [CrossRef]
  4. Soderholm, A.T.; Barnett, T.C.; Sweet, M.J.; Walker, M.J. Group a streptococcal pharyngitis: Immune responses involved in bacterial clearance and GAS-associated immunopathologies. J. Leukoc. Biol. 2017, 103, 193–213. [Google Scholar] [CrossRef]
  5. Shaikh, N.; Swaminathan, N.; Hooper, E.G. Accuracy and precision of the signs and symptoms of streptococcal pharyngitis in children: A systematic review. J. Pediatr. 2012, 160, 487–493. [Google Scholar] [CrossRef]
  6. Shulman, S.T.; Tanz, R.R. Group a streptococcal pharyngitis and immune-mediated complications: From diagnosis to management. Expert Rev. Anti Infect. Ther. 2010, 8, 137–150. [Google Scholar] [CrossRef]
  7. Walker, M.J.; Barnett, T.C.; McArthur, J.D.; Cole, J.N.; Gillen, C.M.; Henningham, A.; Sriprakash, K.S.; Sanderson-Smith, M.L.; Nizet, V. Disease manifestations and pathogenic mechanisms of group A Streptococcus. Clin. Microbiol. Rev. 2014, 27, 264–301. [Google Scholar] [CrossRef] [Green Version]
  8. Dietrich, D.E.; Zhang, Y.; Bode, L.; Munte, T.F.; Hauser, U.; Schmorl, P.; Richter-Witte, C.; Godecke-Koch, T.; Feutl, S.; Schramm, J.; et al. Brain potential amplitude varies as a function of Borna disease virus-specific immune complexes in obsessive-compulsive disorder. Mol. Psychiatry 2005, 10, 519–520. [Google Scholar] [CrossRef]
  9. Muller, N.; Riedel, M.; Blendinger, C.; Oberle, K.; Jacobs, E.; Abele-Horn, M. Mycoplasma pneumoniae infection and Tourette’s syndrome. Psychiatry Res. 2004, 129, 119–125. [Google Scholar] [CrossRef]
  10. Matsuo, M.; Tsuchiya, K.; Hamasaki, Y.; Singer, H.S. Restless legs syndrome: Association with streptococcal or Mycoplasma infection. Pediatr. Neurol. 2004, 31, 119–121. [Google Scholar] [CrossRef]
  11. Leon, J.; Hommer, R.; Grant, P.; Farmer, C.; D’Souza, P.; Kessler, R.; Williams, K.; Leckman, J.F.; Swedo, S. Longitudinal outcomes of children with pediatric autoimmune neuropsychiatric disorder associated with streptococcal infections (PANDAS). Eur. Child Adolesc. Psychiatry 2018, 27, 637–643. [Google Scholar] [CrossRef] [PubMed]
  12. Carapetis, J.R.; Beaton, A.; Cunningham, M.W.; Guilherme, L.; Karthikeyan, G.; Mayosi, B.M.; Sable, C.; Steer, A.; Wilson, N.; Wyber, R.; et al. Acute rheumatic fever and rheumatic heart disease. Nat. Rev. Dis. Primers 2016, 2, 1–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Coffey, P.M.; Ralph, A.P.; Krause, V.L. The role of social determinants of health in the risk and prevention of group a streptococcal infection, acute rheumatic fever and rheumatic heart disease: A systematic review. PLoS Negl. Trop. Dis. 2018, 12, 1–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Gewitz, M.H.; Baltimore, R.S.; Tani, L.Y.; Sable, C.A.; Shulman, S.T.; Carapetis, J.; Remenyi, B.; Taubert, K.A.; Bolger, A.F.; Beerman, L.; et al. Revision of the Jones Criteria for the Diagnosis of Acute Rheumatic Fever in the Era of Doppler Echocardiography. Circulation 2015, 131, 1806–1818. [Google Scholar] [CrossRef] [Green Version]
  15. Karthikeyan, G.; Guilherme, L. Acute rheumatic fever. Lancet 2018, 392, 161–174. [Google Scholar] [CrossRef]
  16. Webb, R.H.; Grant, C.; Harnden, A. Acute rheumatic fever. BMJ 2015, 351, h3443. [Google Scholar] [CrossRef] [Green Version]
  17. Carapetis, J.R.; Currie, B.J.; Mathews, J.D. Cumulative incidence of rheumatic fever in an endemic region: A guide to the susceptibility of the population? Epidemiol. Infect. 2000, 124, 239–244. [Google Scholar] [CrossRef]
  18. Guilherme, L.; Kalil, J. Rheumatic fever and rheumatic heart disease: Cellular mechanisms leading autoimmune reactivity and disease. J. Clin. Immunol. 2010, 30, 17–23. [Google Scholar] [CrossRef]
  19. Garcia, A.F.; Yamaga, K.M.; Shafer, L.A.; Bollt, O.; Tam, E.K.; Cunningham, M.W.; Kurahara, D.K. Cardiac myosin epitopes recognized by autoantibody in acute and convalescent rheumatic fever. Pediatr. Infect. Dis. J. 2016, 35, 1021–1026. [Google Scholar] [CrossRef] [Green Version]
  20. Perez-Vigil, A.; Cruz, L.F.D.L.; Brander, G.; Isomura, K.; Gromark, C.; Mataix-Cols, D. The link between autoimmune diseases and obsessive-compulsive and tic disorders: A systematic review. Neurosci. Biobehav. Rev. 2016, 71, 542–562. [Google Scholar] [CrossRef]
  21. Asbahr, F.R.; Negrao, A.B.; Gentil, V.; Zanetta, D.M.; Paz, J.A.D.; Marques-Dias, M.J.; Kiss, M.H. Obsessive-Compulsive and Related Symptoms in Children and Adolescents with Rheumatic Fever With and Without Chorea: A Prospective 6-Month Study. Am. J. Psychiatry 1998, 155, 1122–1124. [Google Scholar] [CrossRef] [PubMed]
  22. Grzanka, K.; Pieczyrak, R.; Kotulska, A.; Kucharz, E. Zmiany dotyczace ukladu nerwowego u chorych na goraczke reumatyczna Neurological involvement in patients with rheumatic fever. Reumatologia/Rheumatology 2005, 43, 211–215. [Google Scholar]
  23. Punukollu, M.; Mushet, N.; Linney, M.; Hennessy, C.; Morton, M. Neuropsychiatric manifestations of Sydenham’s chorea: A systematic review. Dev. Med. Child Neurol. 2016, 58, 16–28. [Google Scholar] [CrossRef] [PubMed]
  24. Galvin, J.E.; Hemric, M.E.; Ward, K.; Cunningham, M.W. Cytotoxic mAb from rheumatic carditis recognizes heart valves and laminin. J. Clin. Investig. 2000, 106, 217–224. [Google Scholar] [CrossRef] [Green Version]
  25. Cunningham, M.W. Rheumatic Fever, Autoimmunity, and Molecular Mimicry: The Streptococcal Connection. Int. Rev. Immunol. 2014, 33, 314–329. [Google Scholar] [CrossRef] [Green Version]
  26. Cunningham, M.W. Streptococcus and rheumatic fever. Curr. Opin. Rheumatol. 2012, 24, 408–416. [Google Scholar] [CrossRef]
  27. Swedo, S.E.; Leonard, H.L.; Garvey, M.; Mittleman, B.; Allen, A.J.; Perlmutter, S.; Lougee, L.; Dow, S.; Zamkoff, J.; Dubbert, B.K. Pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections: Clinical description of the first 50 cases. Am. J. Psychiatry 1998, 155, 264–271. [Google Scholar] [CrossRef]
  28. Murphy, T.K.; Gerardi, D.M.; Parker-Athill, E.C. The PANDAS Controversy: Why (and How) Is It Still Unsettled? Curr. Dev. Disord. Rep. 2014, 1, 236–244. [Google Scholar] [CrossRef] [Green Version]
  29. Swedo, S.E.; Leonard, H.L.; Rapoport, J.L. The pediatric autoimmune neuropsychiatric disorders associated with streptococcal infection (PANDAS) subgroup: Separating fact from fiction. Pediatrics 2004, 113, 907–911. [Google Scholar] [CrossRef]
  30. Chiarello, F.; Spitoni, S.; Hollander, E.; Matucci Cerinic, M.; Pallanti, S. An expert opinion on PANDAS/PANS: Highlights and controversies. Int. J. Psychiatry Clin. Pract. 2017, 21, 91–98. [Google Scholar] [CrossRef]
  31. Snider, L.A.; Swedo, S.E. PANDAS: Current status and directions for research. Mol. Psychiatry 2004, 9, 900–907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Bodner, S.M.; Morshed, S.A.; Peterson, B.S. The question of PANDAS in adults. Biol. Psychiatry 2001, 49, 807–810. [Google Scholar] [CrossRef]
  33. Carelli, R.; Pallanti, S. Streptococcal infections of skin and PANDAS. Dermatol. Ther. 2014, 27, 28–30. [Google Scholar] [CrossRef] [PubMed]
  34. Jaspers-Fayer, F.; Han, S.H.J.; Chan, E.; McKenney, K.; Simpson, A.; Boyle, A.; Ellwyn, R.; Stewart, S.E. Prevalence of Acute-Onset Subtypes in Pediatric Obsessive-Compulsive Disorder. J. Child Adolesc. Psychopharmacol. 2017, 27, 332–341. [Google Scholar] [CrossRef]
  35. Swedo, S.E.; Rapoport, J.L.; Cheslow, D.L.; Leonard, H.L.; Ayoub, E.M.; Hosier, D.M.; Wald, E.R. High prevalence of obsessive-compulsive symptoms in patients with Sydenham’s chorea. Am. J. Psychiatry 1989, 146, 246–249. [Google Scholar]
  36. Swedo, S.E. Sydenham’s chorea: A model for childhood autoimmune neuropsychiatric disorders. JAMA 1994, 272, 1788–1791. [Google Scholar] [CrossRef]
  37. Gilbert, D.L.; Kurlan, R. PANDAS: Horse or zebra? Neurology 2009, 73, 1252–1253. [Google Scholar] [CrossRef]
  38. De Oliveira, S.K.; Pelajo, C.F. Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal Infection (PANDAS): A Controversial Diagnosis. Curr. Infect. Dis. Rep. 2010, 12, 103–109. [Google Scholar] [CrossRef]
  39. Swedo, S.E.; Leckman, J.F.; Rose, N.R. From research subgroup to clinical syndrome: Modifying the PANDAS criteria to describe PANS (pediatric acute-onset neuropsychiatric syndrome). Pediatr. Ther. 2012, 2, 113. [Google Scholar] [CrossRef]
  40. Murphy, T.K.; Parker-Athill, E.C.; Lewin, A.B.; Storch, E.A.; Mutch, P.J. Cefdinir for recent-onset pediatric neuropsychiatric disorders: A pilot randomized trial. J. Child Adolesc. Psychopharmacol. 2015, 25, 57–64. [Google Scholar] [CrossRef] [Green Version]
  41. Jadah, R.H.S.; Mujeeb, A.A. Neuropsychiatric symptoms following sore throat in a young boy. BMJ Case Rep. 2019, 12, 1–4. [Google Scholar] [CrossRef] [PubMed]
  42. Hoekstra, P.J.; Minderaa, R.B. Tic disorders and obsessive-compulsive disorder: Is autoimmunity involved? Int. Rev. Psychiatry 2005, 17, 497–502. [Google Scholar] [CrossRef]
  43. Bernstein, G.A.; Victor, A.M.; Pipal, A.J.; Williams, K.A. Comparison of clinical characteristics of pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections and childhood obsessive-compulsive disorder. J. Child Adolesc. Psychopharmacol. 2010, 20, 333–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Calkin, C.V.; Carandang, C.G. Certain eating disorders may be a neuropsychiatric manifestation of PANDAS: Case report. J. Can. Acad. Child Adolesc. Psychiatry 2007, 16, 132–135. [Google Scholar] [PubMed]
  45. Murphy, T.K.; Kurlan, R.; Leckman, J. The immunobiology of Tourette’s disorder, pediatric autoimmune neuropsychiatric disorders associated with streptococcus, and related disorders: A way forward. J. Child Adolesc. Psychopharmacol. 2010, 20, 317–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Calaprice, D.; Tona, J.; Parker-Athill, E.C.; Murphy, T.K. A Survey of Pediatric Acute-Onset Neuropsychiatric Syndrome Characteristics and Course. J. Child Adolesc. Psychopharmacol. 2017, 27, 607–618. [Google Scholar] [CrossRef] [Green Version]
  47. Giulino, L.; Gammon, P.; Sullivan, K.; Franklin, M.; Foa, E.; Maid, R.; March, J.S. Is parental report of upper respiratory infection at the onset of obsessive-compulsive disorder suggestive of pediatric autoimmune neuropsychiatric disorder associated with streptococcal infection? J. Child Adolesc. Psychopharmacol. 2002, 12, 157–164. [Google Scholar] [CrossRef]
  48. Lin, H.; Williams, K.A.; Katsovich, L.; Findley, D.B.; Grantz, H.; Lombroso, P.J.; Kingb, R.A.; Bessenc, D.E.; Johnsond, D.; Kaplan, E.L.; et al. Streptococcal Upper Respiratory Tract Infections and Psychosocial Stress Predict Future Tic and Obsessive-Compulsive Symptom Severity in Children and Adolescents with Tourette Syndrome and Obsessive-Compulsive Disorder. Biol. Psychiatry 2010, 67, 684–691. [Google Scholar] [CrossRef] [Green Version]
  49. Orlovska, S.; Vestergaard, C.H.; Bech, B.H.; Nordentoft, M.; Vestergaard, M.; Benros, M.E. Association of Streptococcal Throat Infection with Mental Disorders: Testing Key Aspects of the PANDAS Hypothesis in a Nationwide Study. JAMA Psychiatry 2017, 74, 740–746. [Google Scholar] [CrossRef]
  50. Williams, K.A.; Swedo, S.E. Post-infectious autoimmune disorders: Sydenham’s chorea, PANDAS and beyond. Brain Res. 2015, 1617, 144–154. [Google Scholar] [CrossRef]
  51. Perrin, E.M.; Murphy, M.L.; Casey, J.R.; Pichichero, M.E.; Runyan, D.K.; Miller, W.C.; Snider, L.A.; Swedo, S.E. Does group a beta-hemolytic streptococcal infection increase risk for behavioral and neuropsychiatric symptoms in children? Arch. Pediatr. Adolesc. Med. 2004, 158, 848–856. [Google Scholar] [CrossRef] [Green Version]
  52. Murphy, T.K.; Snider, L.A.; Mutch, P.J.; Harden, E.; Zaytoun, A.; Edge, P.J.; Storch, E.A.; Yang, M.C.; Mann, G.; Goodman, W.K.; et al. Relationship of Movements and Behaviors to Group A Streptococcus Infections in Elementary School Children. Biol. Psychiatry 2007, 61, 279–284. [Google Scholar] [CrossRef]
  53. Garcia-Delgar, B.; Morer, A.; Luber, M.J.; Coffey, B.J. Obsessive-compulsive disorder, tics, and autoinflammatory diseases: Beyond PANDAS. J. Child Adolesc. Psychopharmacol. 2016, 26, 847–850. [Google Scholar]
  54. Thienemann, M.; Murphy, T.; Leckman, J.; Shaw, R.; Williams, K.; Kapphahn, C.; Frankovich, J.; Geller, D.; Bernstein, G.; Chang, K.; et al. Clinical Management of Pediatric Acute-Onset Neuropsychiatric Syndrome: Part I—Psychiatric and Behavioral Interventions. J. Child Adolesc. Psychopharmacol. 2017, 27, 566–573. [Google Scholar] [CrossRef]
  55. Chang, K.; Frankovich, J.; Cooperstock, M.; Cunningham, M.W.; Latimer, M.E.; Murphy, T.K.; Pasternack, M.; Thienemann, M.; Williams, K.; Walter, J.; et al. Clinical Evaluation of Youth with Pediatric Acute-Onset Neuropsychiatric Syndrome (PANS): Recommendations from the 2013 PANS Consensus Conference. J. Child Adolesc. Psychopharmacol. 2015, 25, 3–13. [Google Scholar] [CrossRef] [Green Version]
  56. Sen, E.S.; Ramanan, A.V. How to use antistreptolysin O titre. Arch. Dis. Child Educ. Pract. Ed. 2014, 99, 231–238. [Google Scholar] [CrossRef]
  57. Delice, S. Detection of anti- DNase B antibody upper normal values in children’s age groups who were admitted to hospital with noninfectious reasons. North Clin. Istanbul. 2015, 2, 136–141. [Google Scholar] [CrossRef] [Green Version]
  58. Cohen, J.F.; Bertille, N.; Cohen, R.; Chalumeau, M. Rapid antigen detection test for group A streptococcus in children with pharyngitis (Review). Cochrane Database Syst. Rev. 2016, 7, CD010502. [Google Scholar]
  59. Katz, B.Z. Streptococcal Infections and Exacerbations in Pediatric Autoimmune Neuropsychiatric Disorder Associated With Streptococcal Infection: A Systematic Review and Meta-Analysis. Pediatr. Infect. Dis. J. 2019, 38, e190–e191. [Google Scholar] [CrossRef]
  60. Frick, L.; Pittenger, C. Microglial Dysregulation in OCD, Tourette Syndrome, and PANDAS. J. Immunol. Res. 2016, 2016, 1–8. [Google Scholar] [CrossRef]
  61. Zibordi, F.; Zorzi, G.; Carecchio, M.; Nardocci, N. CANS: Childhood acute neuropsychiatric syndromes. Eur. J. Paediatr. Neurol. 2018, 22, 316–320. [Google Scholar] [CrossRef] [PubMed]
  62. Giedd, J.N.; Rapoport, J.L.; Garvey, M.A.; Perlmutter, S.; Swedo, S.E. MRI assessment of children with obsessive-compulsive disorder or tics associated with streptococcal infection. Am. J. Psychiatry 2000, 157, 281–283. [Google Scholar] [CrossRef] [PubMed]
  63. Giedd, J.N.; Rapoport, J.L.; Kruesi, M.J.; Parker, C.; Schapiro, M.B.; Allen, A.J.; Leonard, H.L.; Kaysen, D.; Dickstein, D.P.; Marsch, W.L.; et al. Sydenham’s chorea: Magnetic resonance imaging of the basal ganglia. Neurology 1996, 45, 2199–2202. [Google Scholar] [CrossRef] [PubMed]
  64. Frick, L.R.; Rapanelli, M.; Jindachomthong, K.; Grant, P.; Leckman, J.F.; Swedo, S.; Williams, K.; Pittenger, C. Differential binding of antibodies in PANDAS patients to cholinergic interneurons in the striatum. Brain Behav. Immun. 2018, 69, 304–311. [Google Scholar] [CrossRef]
  65. Tay, T.L.; Bechade, C.; D’Andrea, I.; St-Pierre, M.K.; Henry, M.S.; Roumier, A.; Tremblay, M.E. Microglia Gone Rogue: Impacts on Psychiatric Disorders across the Lifespan. Front. Mol. Neurosci. 2018, 4, 421. [Google Scholar] [CrossRef] [Green Version]
  66. Quagliariello, A.; Del Chierico, F.; Russo, A.; Reddel, S.; Conte, G.; Lopetuso, L.R.; Ianiro, G.; Dallapiccola, B.; Cardona, F.; Gasbarrini, A.; et al. Gut microbiota profiling and gut-brain crosstalk in children affected by pediatric acute-onset neuropsychiatric syndrome and pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections. Front. Microbiol. 2018, 9, 1–15. [Google Scholar] [CrossRef]
  67. Celik, G.G.; Tas, D.A.; Tahiroglu, A.Y.; Erken, E.; Seydaoglu, G.; Ray, P.C.; Avci, A. Mannose-Binding Lectin2 gene polymorphism in PANDAS patients. Noro Psikiyatri Arsivi 2018, 56, 99–105. [Google Scholar]
  68. Luleyap, H.; Onatoglu, D.; Yilmaz, M.; Alptekin, D.; Tahiroglu, A.; Cetiner, S.; Pazarbasi, A.; Unal, I.; Avci, A.; Comertpay, G. Association between pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections disease and tumor necrosis factor- gene-308 g/a, -850 c/t polymorphisms in 4-12-year-old children in Adana/Turkey. Indian J. Hum. Genet. 2013, 19, 196. [Google Scholar] [CrossRef] [Green Version]
  69. Morris-Berry, C.M.; Pollard, M.; Gao, S.; Thompson, C.; Singer, H.S. Anti-streptococcal, tubulin, and dopamine receptor 2 antibodies in children with PANDAS and Tourette syndrome: Single-point and longitudinal assessments. J. Neuroimmunol. 2013, 264, 106–113. [Google Scholar] [CrossRef]
  70. Macerollo, A.; Martino, D. Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal Infections (PANDAS): An Evolving Concept. Tremor Other Hyperkinetic Mov. 2013, 3. [Google Scholar] [CrossRef]
  71. Hesselmark, E.; Bejerot, S. Biomarkers for diagnosis of Pediatric Acute Neuropsychiatric Syndrome (PANS)-Sensitivity and specificity of the Cunningham Panel. J. Neuroimmunol. 2017, 312, 31–37. [Google Scholar] [CrossRef] [Green Version]
  72. Snider, L.A.; Lougee, L.; Slattery, M.; Grant, P.; Swedo, S.E. Antibiotic prophylaxis with azithromycin or penicillin for childhood-onset neuropsychiatric disorders. Biol. Psychiatry 2005, 57, 788–792. [Google Scholar] [CrossRef]
  73. Murphy, T.K.; Brennan, E.M.; Johnco, C.; Parker-Athill, E.C.; Miladinovic, B.; Storch, E.A.; Lewin, A.B. A Double-Blind Randomized Placebo-Controlled Pilot Study of Azithromycin in Youth with Acute-Onset Obsessive-Compulsive Disorder. J. Child Adolesc. Psychopharmacol. 2017, 27, 640–651. [Google Scholar] [CrossRef]
  74. Aman, M.; Rettiganti, M.; Nagaraja, H.N.; Hollway, J.A.; Mccracken, J.; Mcdougle, C.J.; Tierney, E.; Scahill, L.; Arnold, L.E.; Hellings, J.; et al. Tolerability, safety, and benefits of risperidone in children and adolescents with autism: 21-month follow-up after 8-week placebo-controlled trial. J. Child Adolesc. Psychopharmacol. 2015, 25, 482–493. [Google Scholar] [CrossRef]
  75. Elia, J.; Dell, M.L.; Friedman, D.F.; Zimmerman, R.A.; Balamuth, N.; Ahmed, A.A.; Pati, S. PANDAS with catatonia: A case report. Therapeutic response to lorazepam and plasmapheresis. J. Am. Acad. Child Adolesc. Psychiatry 2005, 44, 1145–1150. [Google Scholar] [CrossRef]
  76. Kovacevic, M.; Grant, P.; Swedo, S.E. Use of Intravenous Immunoglobulin in the Treatment of Twelve Youths with Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal infections. J. Child Adolesc. Psychopharmacol. 2015, 25, 65–69. [Google Scholar] [CrossRef] [Green Version]
  77. Storch, E.A.; Murphy, T.K.; Geffken, G.R.; Mann, G.; Adkins, J.J.; Merlo, L.; Duke, D.; Munson, M.; Swaine, Z.; Goodman, W.K. Cognitive-behavioral therapy for PANDAS-related obsessive-compulsive disorder: Findings from a preliminary waitlist controlled open trial. J. Am. Acad. Child Adolesc. Psychiatry 2006, 45, 1171–1178. [Google Scholar] [CrossRef] [Green Version]
  78. Celik, G.; Taa, D.; Tahiroglu, A.; Avci, A.; Yuksel, B.; Cam, P. Vitamin D deficiency in obsessive-compulsive disorder patients with pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections: A case control study. Noro Psikiyatri Arsivi 2016, 53, 31–34. [Google Scholar] [CrossRef]
  79. Sigra, S.; Hesselmark, E.; Bejerot, S. Treatment of PANDAS and PANS: A systematic review. Neurosci. Biobehav. Rev. 2018, 86, 51–65. [Google Scholar] [CrossRef]
  80. Cooperstock, M.S.; Swedo, S.E.; Pasternack, M.S.; Murphy, T.K.; Consortium, F.T.P. Clinical Management of Pediatric Acute-Onset Neuropsychiatric Syndrome: Part III-Treatment and Prevention of Infections. J. Child Adolesc. Psychopharmacol. 2017, 27, 594–606. [Google Scholar] [CrossRef]
  81. Moore, D.P. Neuropsychiatric aspects of Sydenham’s chorea: A comprehensive review. J. Clin. Psychiatry 1996, 57, 407–414. [Google Scholar]
  82. Winter, G.; Hart, R.A.; Charlesworth, R.P.G.; Sharpley, C.F. Gut microbiome and depression: What we know and what we need to know. Rev. Neurosci. 2018, 29, 629–643. [Google Scholar] [CrossRef]
  83. Hermann, A.; Walker, R.H. Diagnosis and Treatment of Chorea Syndromes. Curr. Neurol. Neurosci. Rep. 2015, 15, 514. [Google Scholar] [CrossRef]
  84. Stern, J.S. Tourette’s syndrome and its borderland. Pract. Neurol. 2018, 18, 262–270. [Google Scholar] [CrossRef]
  85. Martino, D.; Ganos, C.; Pringsheim, T.M. Tourette Syndrome and Chronic Tic Disorders: The Clinical Spectrum Beyond Tics. Int. Rev. Neurobiol. 2017, 134, 1461–1490. [Google Scholar]
  86. Hallett, M. Tourette Syndrome: Update. Brain Dev. 2015, 37, 651–655. [Google Scholar] [CrossRef] [Green Version]
  87. Cavanna, A.E.; Black, K.J.; Hallett, M.; Voon, V. Neurobiology of the Premonitory Urge in Tourette’s Syndrome: Pathophysiology and Treatment Implications. J. Neuropsychiatry Clin. Neurosci. 2017, 29, 95–104. [Google Scholar] [CrossRef] [Green Version]
  88. Scharf, J.; Yu, D.; Huang, A.; Tsetsos, F.; Paschou, P.; Coppola, G.; Mathews, C. Collaborative Genome-Wide Association and Copy Number Variation Analysis of Tourette Syndrome. Eur. Neuropsychopharmacol. 2019, 29, S736–S737. [Google Scholar] [CrossRef]
  89. Hirschtritt, M.E.; Darrow, S.M.; Illmann, C.; Osiecki, L.; Grados, M.; Sandor, P.; Dion, Y.; King, R.A.; Pauls, D.; Budman, C.L.; et al. Genetic and phenotypic overlap of specific obsessive-compulsive and attention-deficit/hyperactive subtypes with Tourette syndrome. Psychol. Med. 2017, 48, 279–293. [Google Scholar] [CrossRef] [Green Version]
  90. Church, A.J. Tourettes syndrome: A cross sectional study to examine the PANDAS hypothesis. J. Neurol. Neurosurg. Psychiatry 2003, 74, 602–607. [Google Scholar] [CrossRef] [Green Version]
  91. Spinello, C.; Laviola, G.; Macri, S. Pediatric Autoimmune Disorders Associated with Streptococcal Infections and Tourettes Syndrome in Preclinical Studies. Front. Neurosci. 2016, 10, 310. [Google Scholar] [CrossRef]
  92. Suhs, K.-W.; Skripuletz, T.; Pul, R.; Alvermann, S.; Schwenkenbecher, P.; Stangel, M.; Muller-Vahl, K. Gilles de la Tourette syndrome is not linked to contactin-associated protein receptor 2 antibodies. Mol. Brain 2015, 8, 62. [Google Scholar] [CrossRef] [Green Version]
  93. Oosterveer, D.M.; Overweg-Plandsoen, W.C.; Roos, R.A. Sydenhams Chorea: A Practical Overview of the Current Literature. Pediatr. Neurol. 2010, 43, 1–6. [Google Scholar] [CrossRef]
  94. Crealey, M.; Allen, N.M.; Webb, D.; Bouldin, A.; Sweeney, N.M.; Peake, D.; Tirupathi, S.; Butler, K.; King, M.D. Sydenhams chorea: Not gone but perhaps forgotten. Arch. Dis. Child. 2015, 100, 1160–1162. [Google Scholar] [CrossRef]
  95. Murphy, T.K.; Goodman, W.K.; Ayoub, E.M.; Voeller, K.K. On defining Sydenham’s chorea: Where do we draw the line? Biol. Psychiatry 2000, 47, 851–857. [Google Scholar] [CrossRef]
  96. Walker, K.G.; De Vries, P.J.; Stein, D.J.; Wilmshurst, J.M. Sydenham chorea and PANDAS in South Africa: Review of evidence and recommendations for management in resource-poor countries. J. Child Neurol. 2015, 30, 850–859. [Google Scholar] [CrossRef]
  97. Morris, C.M.; Pardo-Villamizar, C.; Gause, C.D.; Singer, H.S. Serum autoantibodies measured by immunofluorescence confirm a failure to differentiate PANDAS and Tourette syndrome from controls. J. Neurol. Sci. 2009, 276, 45–48. [Google Scholar] [CrossRef]
  98. Gamucci, A.; Uccella, S.; Sciarretta, L.; D’Apruzzo, M.; Calevo, M.G.; Mancardi, M.M.; Veneselli, E.; De Grandis, E. PANDAS and PANS: Clinical, Neuropsychological, and Biological Characterization of a Monocentric Series of Patients and Proposal for a Diagnostic Protocol. J. Child Adolesc. Psychopharmacol. 2019, 29, 305–312. [Google Scholar] [CrossRef] [Green Version]
  99. Lehman, M.; Navarro, V.; Suchecki, D.; Handa, R. Introduction to the PANS special issue. J. Neuroendocrinol. 2018, 30, 12612. [Google Scholar] [CrossRef]
  100. Frankovich, J.; Thienemann, M.; Rana, S.; Chang, K. Five Youth with Pediatric Acute-Onset Neuropsychiatric Syndrome of Differing Etiologies. J. Child Adolesc. Psychopharmacol. 2015, 25, 31–37. [Google Scholar] [CrossRef] [Green Version]
  101. Muller, N.; Schwarz, M.J. Neuroimmune-endocrine crosstalk in schizophrenia and mood disorders. Expert Rev. Neurother. 2006, 6, 1017–1038. [Google Scholar] [CrossRef]
  102. Hornig, M. The role of microbes and autoimmunity in the pathogenesis of neuropsychiatric illness. Curr. Opin. Rheumatol. 2013, 25, 488–495. [Google Scholar] [CrossRef]
  103. Morer, A.; Viñas, O.; Lázaro, L.; Bosch, J.; Toro, J.; Castro, J. D8/17 Monoclonal Antibody: An Unclear Neuropsychiatric Marker. Behav. Neurol. 2005, 16, 1–8. [Google Scholar] [CrossRef] [Green Version]
  104. Sokol, M.S.; Ward, P.E.; Tamiya, H.; Kondo, D.G.; Houston, D.; Zabriskie, J.B. D8/17 Expression on B Lymphocytes in Anorexia Nervosa. Am. J. Psychiatry 2002, 159, 1430–1432. [Google Scholar] [CrossRef]
  105. Bronze, M.S.; Dale, J.B. Epitopes of streptococcal M proteins that evoke antibodies that cross-react with human brain. J. Immunol. 1993, 151, 2820–2828. [Google Scholar]
  106. Kim, S.W.; Grant, J.E.; Kim, S.I.; Swanson, T.A.; Bernstein, G.A.; Jaszcz, W.B.; Williams, K.A.; Schlievert, P.M. A Possible Association of Recurrent Streptococcal Infections and Acute Onset of Obsessive-Compulsive Disorder. J. Neuropsychiatry Clin. Neurosci. 2004, 16, 252–260. [Google Scholar] [CrossRef]
  107. Marazziti, D.; Albert, U.; Mucci, F.; Piccinni, A. The Glutamate and the Immune Systems: New Targets for the Pharmacological Treatment of OCD. Curr. Med. Chem. 2018, 25, 5731–5738. [Google Scholar] [CrossRef]
  108. Macri, S.; Spinello, C.; Widomska, J.; Magliozzi, R.; Poelmans, G.; Invernizzi, R.W.; Creti, R.; Roessner, V.; Bartolini, E.; Margarit, I.; et al. Neonatal corticosterone mitigates autoimmune neuropsychiatric disorders associated with streptococcus in mice. Sci. Rep. 2018, 8, 1–12. [Google Scholar] [CrossRef]
  109. Kipnis, J.; Cardon, M.; Avidan, H.; Lewitus, G.M.; Mordechay, S.; Rolls, A.; Shani, Y.; Schwartz, M. Dopamine, through the extracellular signal-regulated kinase pathway, downregulates CD4+CD25+ regulatory T-cell activity: Implications for neurodegeneration. J. Neurosci. 2004, 24, 6133–6143. [Google Scholar] [CrossRef] [Green Version]
  110. Besser, M.J.; Ganor, Y.; Levite, M. Dopamine by itself activates either D2, D3 or D1/D5 dopaminergic receptors in normal human T-cells and triggers the selective secretion of either IL-10, TNFalpha or both. J. Neuroimmunol. 2005, 169, 161–171. [Google Scholar] [CrossRef]
  111. Ben-Pazi, H.; Stoner, J.A.; Cunningham, M.W. Dopamine Receptor Autoantibodies Correlate with Symptoms in Sydenham’s Chorea. PLoS ONE 2013, 8, e73516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Ravindran, A.V.; Griffiths, J.; Merali, Z.; Anisman, H. Circulating lymphocyte subsets in obsessive compulsive disorder, major depression and normal controls. J. Affect. Disord. 1999, 52, 1–10. [Google Scholar] [CrossRef]
  113. Lamothe, H.; Baleyte, J.-M.; Smith, P.; Pelissolo, A.; Mallet, L. Individualized Immunological Data for Precise Classification of OCD Patients. Brain Sci. 2018, 8, 149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Murphy, T.K.; Storch, E.A.; Lewin, A.B.; Edge, P.J.; Goodman, W.K. Clinical factors associated with pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections. J. Pediatr. 2012, 160, 314–319. [Google Scholar] [CrossRef] [Green Version]
  115. Levite, M. Dopamine and T Cells: Dopamine Receptors and Potent Effects on T Cells, Dopamine Production in T Cells, and Abnormalities in the Dopaminergic System in T Cells in Autoimmune, Neurological and Psychiatric Diseases. Acta Physiol. 2016, 216, 42–89. [Google Scholar] [CrossRef]
  116. Gruner, P.; Pittenger, C. Cognitive inflexibility in Obsessive-Compulsive Disorder. Neuroscience 2017, 345, 243–255. [Google Scholar] [CrossRef] [Green Version]
  117. Kirvan, C.A.; Swedo, S.E.; Snider, L.A.; Cunningham, M.W. Antibody-mediated neuronal cell signaling in behavior and movement disorders. J. Neuroimmunol. 2006, 179, 173–179. [Google Scholar] [CrossRef]
  118. Cunningham, M.W. Rheumatic fever revisited. Nat. Rev. Cardiol. 2014, 11, 123. [Google Scholar] [CrossRef] [Green Version]
  119. Hughes, R.; MedSci, F.; Bensa, S.; Willison, H.; Van Den Bergh, P.; Comi, G.; IIIa, I.; Nobile-Orazio, E.; van Doorn, P.; Dalakas, M.; et al. Randomized controlled trial of intravenous immunoglobulin versus oral prednisolone in chronic inflammatory demyelinating polyradiculoneuropathy. Ann. Neurol. 2001, 50, 195–201. [Google Scholar] [CrossRef]
  120. Kataoka, Y.; Kalanithi, P.S.A.; Grantz, H.; Schwartz, M.L.; Saper, C.; Leckman, J.F.; Vaccarino, F.M. Decreased number of parvalbumin and cholinergic interneurons in the striatum of individuals with Tourette syndrome. J. Comp. Neurol. 2010, 518, 277–291. [Google Scholar] [CrossRef]
  121. Bonsi, P.; Cuomo, D.; Martella, G.; Madeo, G.; Schirinzi, T.; Puglisi, F.; Ponterio, G.; Pisani, A. Centrality of Striatal Cholinergic Transmission in Basal Ganglia Function. Front. Neuroanat. 2011, 5, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Swedo, S.; Williams, K. PANDAS as a post-Streptococcal autoimmune neuropsychiatric form of OCD. In Obsessive-Compulsive Disorder: Phenomenology, Pathophysiology, and Treatment; Pittenger, C., Ed.; Oxford University Press: New York, NY, USA, 2017; pp. 311–321. [Google Scholar]
  123. Cox, C.J.; Sharma, M.; Leckman, J.F.; Zuccolo, J.; Zuccolo, A.; Kovoor, A.; Swedo, S.E.; Cunningham, M.W. Brain Human Monoclonal Autoantibody from Sydenham Chorea Targets Dopaminergic Neurons in Transgenic Mice and Signals Dopamine D2 Receptor: Implications in Human Disease. J. Immunol. 2013, 191, 5524–5541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Sciamanna, G.; Napolitano, F.; Pelosi, B.; Bonsi, P.; Vitucci, D.; Nuzzo, T.; Punzo, D.; Ghiglieri, V.; Ponterio, G.; Pasqualetti, M.; et al. Rhes regulates dopamine D2 receptor transmission in striatal cholinergic interneurons. Neurobiol. Dis. 2015, 78, 146–161. [Google Scholar] [CrossRef] [PubMed]
  125. Tremblay, M.; Stevens, B.; Sierra, A.; Wake, H.; Bessis, A.; Nimmerjahn, A. The role of microglia in the healthy brain. J. Neurosci. 2011, 31, 16064–16069. [Google Scholar] [CrossRef]
  126. Du, L.; Zhang, Y.; Chen, Y.; Zhu, J.; Yang, Y.; Zhang, H.L. Role of Microglia in Neurological Disorders and Their Potentials as a Therapeutic Target. Mol. Neurobiol. 2017, 54, 7567–7584. [Google Scholar] [CrossRef]
  127. Dileepan, T.; Smith, E.D.; Knowland, D.; Hsu, M.; Platt, M.; Bittner-Eddy, P.; Cohen, B.; Southern, P.; Latimer, E.; Harley, E.; et al. Group A Streptococcus intranasal infection promotes CNS infiltration by streptococcal-specific Th17 cells. J. Clin. Investig. 2016, 126, 303–317. [Google Scholar] [CrossRef] [Green Version]
  128. Husby, G.; Rijn, I.D.; Zabriskie, J.; Abdin, Z.; Williams, R. Anti-Neuronal Antibody in Sydenhams Chorea. Lancet 1977, 309, 1208. [Google Scholar] [CrossRef]
  129. Kirvan, C.A.; Swedo, S.E.; Heuser, J.S.; Cunningham, M.W. Mimicry and autoantibody-mediated neuronal cell signaling in Sydenham chorea. Nat. Med. 2003, 9, 914–920. [Google Scholar] [CrossRef]
  130. Kirvan, C.A.; Cox, C.J.; Swedo, S.E.; Cunningham, M.W. Tubulin Is a Neuronal Target of Autoantibodies in Sydenham’s Chorea. J. Immunol. 2007, 178, 7412–7421. [Google Scholar] [CrossRef]
  131. Kirvan, C.A.; Swedo, S.E.; Kurahara, D.; Cunningham, M.W. Streptococcal mimicry and antibody-mediated cell signaling in the pathogenesis of Sydenhams chorea. Autoimmunity 2006, 39, 21–29. [Google Scholar] [CrossRef]
  132. Hoffman, K.L. A Murine Model for Neuropsychiatric Disorders Associated with Group A -Hemolytic Streptococcal Infection. J. Neurosci. 2004, 24, 1780–1791. [Google Scholar] [CrossRef] [PubMed]
  133. Brimberg, L.; Benhar, I.; Mascaro-Blanco, A.; Alvarez, K.; Lotan, D.; Winter, C.; Klein, J.; Moses, A.E.; Somnier, F.E.; Leckman, J.F. Behavioral, Pharmacological, and Immunological Abnormalities after Streptococcal Exposure: A Novel Rat Model of Sydenham Chorea and Related Neuropsychiatric Disorders. Neuropsychopharmacology 2012, 37, 2076–2087. [Google Scholar] [CrossRef] [PubMed]
  134. Yaddanapudi, K.; Hornig, M.; Serge, R.; Miranda, J.D.; Baghban, A.; Villar, G.; Lipkin, W.I. Passive transfer of streptococcus-induced antibodies reproduces behavioral disturbances in a mouse model of pediatric autoimmune neuropsychiatric disorders associated with streptococcal infection. Mol. Psychiatry 2009, 15, 712–726. [Google Scholar] [CrossRef] [PubMed]
  135. Church, A.J.; Dale, R.C.; Giovannoni, G. Anti-basal ganglia antibodies: A possible diagnostic utility in idiopathic movement disorders? Arch. Dis. Child 2004, 89, 611–614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Perlmutter, S.J.; Leitman, S.F.; Garvey, M.A.; Hamburger, S.; Feldman, E.; Leonard, H.L.; Swedo, S.E. Therapeutic plasma exchange and intravenous immunoglobulin for obsessive-compulsive disorder and tic disorders in childhood. Lancet 1999, 354, 1153–1158. [Google Scholar] [CrossRef]
  137. Ben-Pazi, H.; Sadan, O.; Offen, D. Striatal Microinjection of Sydenham Chorea Antibodies: Using a Rat Model to Examine the Dopamine Hypothesis. J. Mol. Neurosci. 2012, 46, 162–166. [Google Scholar] [CrossRef]
  138. Lotan, D.; Benhar, I.; Alvarez, K.; Mascaro-Blanco, A.; Brimberg, L.; Frenkel, D.; Cunningham, M.W.; Joel, D. Behavioral and neural effects of intra-striatal infusion of anti-streptococcal antibodies in rats. Brain Behav. Immun. 2014, 38, 249–262. [Google Scholar] [CrossRef] [Green Version]
  139. Pavone, P.; Bianchini, R.; Parano, E.; Incorpora, G.; Rizzo, R.; Mazzone, L.; Trifiletti, R.R. Anti-brain antibodies in PANDAS versus uncomplicated streptococcal infection. Pediatr. Neurol. 2004, 30, 107–110. [Google Scholar] [CrossRef] [Green Version]
  140. da Rocha, F.F.; Correa, H.; Teixeira, A.L. Obsessive-compulsive disorder and immunology: A review. Prog. Neuro Psychopharmacol. Biol. Psychiatry 2008, 32, 1139–1146. [Google Scholar] [CrossRef]
  141. Peterson, B.S.; Leckman, J.F.; Tucker, D.; Scahill, L.; Staib, L.; Zhang, H.; King, R.; Cohen, D.J.; Gore, J.C.; Lombroso, P. Preliminary findings of antistreptococcal antibody titers and basal ganglia volumes in tic, obsessive-compulsive, and attention- deficit/hyperactivity disorders. Arch. Gen. Psychiatry 2000, 57, 364–372. [Google Scholar] [CrossRef] [Green Version]
  142. Rizzo, R.; Gulisano, M.; Pavone, P.; Fogliano, F.; Robertson, M.M. Increased antistreptococcal antibody titers and anti-basal ganglia antibodies in patients with Tourette syndrome: Controlled cross-sectional study. J. Child Neurol. 2006, 21, 747–753. [Google Scholar] [CrossRef] [PubMed]
  143. Cabrera, B.; Romero-Rebollar, C.; Jimenez-Angeles, L.; Genis-Mendoza, A.D.; Flores, J.; Lanzagorta, N.; Arroyo, M.; de la Fuente-Sandoval, C.; Santana, D.; Medina-Banuelos, V.; et al. Neuroanatomical features and its usefulness in classification of patients with PANDAS. CNS Spectr. 2018, 24, 533–543. [Google Scholar] [CrossRef]
  144. Snider, L.A.; Swedo, S.E. Post-streptococcal Autoimmune Disorders of the Central Nervous System. Curr. Opin. Neurol. 2003, 16, 359–365. [Google Scholar] [PubMed]
  145. Bellini, S.; Fleming, K.E.; De, M.; McCauley, J.P.; Petroccione, M.A.; D’Brant, L.Y.; Tkachenko, A.; Kwon, S.; Jones, L.A.; Scimemi, A. Neuronal Glutamate Transporters Control Dopaminergic Signaling and Compulsive Behaviors. J. Neurosci. 2018, 38, 937–961. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Stollerman, G.H. Rheumatic Fever in the 21st Century. Clin. Infect. Dis. 2001, 33, 806–814. [Google Scholar] [CrossRef] [PubMed]
  147. Cunningham, M.W. Molecular Mimicry, Autoimmunity, and Infection: The Cross-Reactive Antigens of Group A Streptococci and their Sequelae. Gram Posit. Pathog. 2019, 7, 86–107. [Google Scholar]
  148. Kumar, A.; Williams, M.T.; Chugani, H.T. Evaluation of Basal Ganglia and Thalamic Inflammation in Children with Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal Infection and Tourette Syndrome: A Positron Emission Tomographic (PET) Study Using 11C-[R]-PK11. J. Child Neurol. 2015, 30, 749–756. [Google Scholar] [CrossRef]
  149. Malan-Muller, S.; Valles-Colomer, M.; Raes, J.; Lowry, C.A.; Seedat, S.; Hemmings, S.M.J. The gut microbiome and mental health: Implications for anxiety- and trauma-related disorders. Omics J. Integr. Biol. 2018, 22, 90–107. [Google Scholar] [CrossRef]
  150. Lach, G.; Schellekens, H.; Dinan, T.G.; Cryan, J.F. Anxiety, Depression, and the Microbiome: A Role for Gut Peptides. Neurotherapeutics 2018, 15, 36–59. [Google Scholar] [CrossRef] [Green Version]
  151. Gulas, E.; Wysiadecki, G.; Strzelecki, D.; Gawlik-Kotelnicka, O.; Polguj, M. Can microbiology affect psychiatry? A link between gut microbiota and psychiatric disorders. Psychiatr. Pol. 2018, 52, 1023–1039. [Google Scholar]
  152. Foster, J.A.; McVey Neufeld, K.A. Gut-brain axis: How the microbiome influences anxiety and depression. Trends Neurosci. 2013, 36, 305–312. [Google Scholar] [CrossRef] [PubMed]
  153. Fowlie, G.; Cohen, N.; Ming, X. The perturbance of microbiome and gut-brain axis in autism spectrum disorders. Int. J. Mol. Sci. 2018, 19, 2251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Seitz, J.; Trinh, S.; Herpertz-Dahlmann, B. The Microbiome and Eating Disorders. Psychiatr. Clin. N. Am. 2019, 42, 93–103. [Google Scholar] [CrossRef] [PubMed]
  155. Caputi, V.; Giron, M.C. Microbiome-gut-brain axis and toll-like receptors in parkinson’s disease. Int. J. Mol. Sci. 2018, 19, 1689. [Google Scholar] [CrossRef] [Green Version]
  156. Sochocka, M.; Donskow-Lysoniewska, K.; Diniz, B.S.; Kurpas, D.; Brzozowska, E.; Leszek, J. The Gut Microbiome Alterations and Inflammation-Driven Pathogenesis of Alzheimer’s Disease—A Critical Review. Mol. Neurobiol. 2019, 56, 1841–1851. [Google Scholar] [CrossRef] [Green Version]
  157. Aarts, E.; Ederveen, T.H.A.; Naaijen, J.; Zwiers, M.P.; Boekhorst, J.; Timmerman, H.M.; Smeekens, S.P.; Netea, M.G.; Buitelaar, J.K.; Franke, B.; et al. Gut microbiome in ADHD and its relation to neural reward anticipation. PLoS ONE 2017, 12, 1–17. [Google Scholar]
  158. Kanji, S.; Fonseka, T.M.; Marshe, V.S.; Sriretnakumar, V.; Hahn, M.K.; Muller, D.J. The microbiome-gut-brain axis: Implications for schizophrenia and antipsychotic induced weight gain. Eur. Arch. Psychiatry Clin. Neurosci. 2018, 268, 3–15. [Google Scholar] [CrossRef]
  159. Leclercq, S.; Matamoros, S.; Cani, P.D.; Neyrinck, A.M.; Jamar, F.; Starkel, P.; Windey, K.; Trematoli, V.; Backhed, F.; Verbeke, K.; et al. Intestinal permeability, gut-bacterial dysbiosis, and behavioral markers of alcohol-dependence severity. Proc. Natl. Acad. Sci. USA 2014, 111, E4485–E4493. [Google Scholar] [CrossRef] [Green Version]
  160. Gondalia, S.; Parkinson, L.; Stough, C.; Scholey, A. Gut microbiota and bipolar disorder: A review of mechanisms and potential targets for adjunctive therapy. Psychopharmacology 2019, 236, 1433–1443. [Google Scholar] [CrossRef]
  161. Tang, Y.; Liu, S.; Shu, H.; Yanagisawa, L.; Tao, F. Gut Microbiota Dysbiosis Enhances Migraine-Like Pain Via TNF Upregulation. Mol. Neurobiol. 2019, 57, 461–468. [Google Scholar] [CrossRef]
  162. Milliken, S.; Allen, R.M.; Lamont, R.F. The role of antimicrobial treatment during pregnancy on the neonatal gut microbiome and the development of atopy, asthma, allergy and obesity in childhood. Expert Opin. Drug Saf. 2019, 18, 173–185. [Google Scholar] [CrossRef] [PubMed]
  163. Jasarevic, E.; Howard, C.D.; Misic, A.M.; Beiting, D.P.; Bale, T.L. Stress during pregnancy alters temporal and spatial dynamics of the maternal and offspring microbiome in a sex-specific manner. Sci. Rep. 2017, 7, 44182. [Google Scholar] [CrossRef] [PubMed]
  164. Kelly, J.R.; Clarke, G.; Cryan, J.F.; Dinan, T.G. Brain-gut-microbiota axis: Challenges for translation in psychiatry. Ann. Epidemiol. 2016, 26, 366–372. [Google Scholar] [CrossRef] [PubMed]
  165. Rudzki, L.; Szulc, A. Immune Gate of Psychopathology-The Role of Gut Derived Immune Activation in Major Psychiatric Disorders. Front. Psychiatry 2018, 9, 205. [Google Scholar] [CrossRef] [Green Version]
  166. Lener, M.S.; Niciu, M.J.; Ballard, E.D.; Park, M.; Park, L.T.; Nugent, A.C.; Zarate, C.A., Jr. Glutamate and Gamma-Aminobutyric Acid Systems in the Pathophysiology of Major Depression and Antidepressant Response to Ketamine. Biol. Psychiatry 2017, 81, 886–897. [Google Scholar] [CrossRef] [Green Version]
  167. Lydiard, R.B. The role of GABA in anxiety disorders. J. Clin. Psychiatry 2003, 64 (Suppl. 3), 21–27. [Google Scholar]
  168. Romeo, B.; Choucha, W.; Fossati, P.; Rotge, J.-Y. Meta-analysis of central and peripheral Y-aminobutyric acid levels in patients with unipolar and bipolar depression. J. Psychiatry Neurosci. 2018, 43, 58–66. [Google Scholar] [CrossRef] [Green Version]
  169. Freed, R.D.; Coffey, B.J.; Mao, X.; Weiduschat, N.; Kang, G.; Shungu, D.C.; Gabbay, V. Decreased Anterior Cingulate Cortex Y-Aminobutyric Acid in Youth with Tourettes Disorder. Pediatr. Neurol. 2016, 65, 64–70. [Google Scholar] [CrossRef]
  170. Gabbay, V.; Bradley, K.A.; Mao, X.; Ostrover, R.; Kang, G.; Shungu, D.C. Anterior cingulate cortex Y-aminobutyric acid deficits in youth with depression. Transl. Psychiatry 2017, 7, e1216. [Google Scholar] [CrossRef]
  171. Chiu, P.; Lui, S.S.; Hung, K.S.; Chan, R.C.; Chan, Q.; Sham, P.; Cheung, E.F.C.; Mak, H.K.F. In vivo gamma-aminobutyric acid and glutamate levels in people with first-episode schizophrenia: A proton magnetic resonance spectroscopy study. Schizophr. Res. 2018, 193, 295–303. [Google Scholar] [CrossRef]
  172. Gubellini, P.; Pisani, A.; Centonze, D.; Bernardi, G.; Calabresi, P. Metabotropic glutamate receptors and striatal synaptic plasticity: Implications for neurological diseases. Prog. Neurobiol. 2004, 74, 271–300. [Google Scholar] [CrossRef] [PubMed]
  173. Sherwin, E.; Sandhu, K.V.; Dinan, T.G.; Cryan, J.F. May the Force Be with You: The Light and Dark Sides of the Microbiota-Gut-Brain Axis in Neuropsychiatry. CNS Drugs 2016, 30, 1019–1041. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Rodriguez, J.M.; Murphy, K.; Stanton, C.; Ross, R.P.; Kober, O.I.; Juge, N.; Avershina, E.; Rudi, K.; Narbad, A.; Jenmalm, M.C.; et al. The composition of the gut microbiota throughout life, with an emphasis on early life. Microb. Ecol. Health Dis. 2015, 26, 26050. [Google Scholar] [CrossRef]
  175. Otoole, P.W.; Claesson, M.J. Gut microbiota: Changes throughout the lifespan from infancy to elderly. Int. Dairy J. 2010, 20, 281–291. [Google Scholar] [CrossRef]
  176. Aleman, F.D.D.; Valenzano, D.R. Microbiome evolution during host aging. PLoS Pathog. 2019, 15, e1007727. [Google Scholar] [CrossRef] [PubMed]
  177. Derrien, M.; Alvarez, A.-S.; Vos, W.M.D. The Gut Microbiota in the First Decade of Life. Trends Microbiol. 2019, 27, 997–1010. [Google Scholar] [CrossRef] [Green Version]
  178. Becattini, S.; Taur, Y.; Pamer, E.G. Antibiotic-induced changes in the intestinal microbiota and disease. Trends Mol. Med. 2016, 22, 458–478. [Google Scholar] [CrossRef] [Green Version]
  179. Murphy, T.K.; Storch, E.A.; Turner, A.; Reid, J.M.; Tan, J.; Lewin, A.B. Maternal history of autoimmune disease in children presenting with tics and/or obsessive-compulsive disorder. J. Neuroimmunol. 2010, 229, 243–247. [Google Scholar] [CrossRef] [Green Version]
  180. O’Roak, B.J.; Morgan, T.M.; Fishman, D.O.; Saus, E.; Alonso, P.; Gratacos, M.; Estivill, X.; Teltsh, O.; Kohn, Y.; Kidd, K.K.; et al. Additional support for the association of SLITRK1 var321 and Tourette syndrome. Mol. Psychiatry 2010, 15, 447–450. [Google Scholar] [CrossRef]
  181. Tian, Y.; Gunther, J.R.; Liao, I.H.; Liu, D.; Ander, B.P.; Stamova, B.S.; Lit, L.; Jickling, G.C.; Xu, H.; Zhan, X.; et al. GABA- and acetylcholine-related gene expression in blood correlate with tic severity and microarray evidence for alternative splicing in Tourette syndrome: A pilot study. Brain Res. 2011, 1381, 228–236. [Google Scholar] [CrossRef]
  182. Neth, O.; Jack, D.L.; Dodds, A.W.; Holzel, H.; Klein, N.J.; Turner, M.W. Mannose-binding lectin binds to a range of clinically relevant microorganisms and promotes complement deposition. Infect. Immun. 2000, 68, 688–693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Chen, J.; Xu, Z.; Ou, X.; Wang, M.; Yang, X.; Li, Q. Mannose-binding lectin polymorphisms and recurrent respiratory tract infection in Chinese children. Eur. J. Pediatr. 2009, 168, 1305–1313. [Google Scholar] [CrossRef] [PubMed]
  184. Gomaa, M.H.; Ali, S.S.; Fattouh, A.M.; Hamza, H.S.; Badr, M.M. MBL2 gene polymorphism rs1800450 and rheumatic fever with and without rheumatic heart disease: An Egyptian pilot study. Pediatr. Rheumatol. 2018, 16, 1–7. [Google Scholar] [CrossRef] [PubMed]
  185. Blackburn, J.S. Tic Disorders and PANDAS. Semin. Pediatr. Neurol. 2018, 25, 25–33. [Google Scholar] [CrossRef]
  186. Farhood, Z.; Ong, A.A.; Discolo, C.M. PANDAS: A systematic review of treatment options. Int. J. Pediatr. Otorhinolaryngol. 2016, 89, 149–153. [Google Scholar] [CrossRef]
  187. Frankovich, J. 23.3 Clinical Management of Pediatric Acute-Onset Neuropsychiatric Syndrome: Use of Immunomodulatory Therapies. J. Am. Acad. Child Adolesc. Psychiatry 2017, 56, S339. [Google Scholar] [CrossRef]
  188. Brown, K.D.; Farmer, C.; Freeman, G.M.; Spartz, E.J.; Farhadian, B.; Thienemann, M.; Frankovich, J. Effect of Early and Prophylactic Nonsteroidal Anti-Inflammatory Drugs on Flare Duration in Pediatric Acute-Onset Neuropsychiatric Syndrome: An Observational Study of Patients Followed by an Academic Community-Based Pediatric Acute-Onset Neuropsychiatric Syndrome Clinic. J. Child Adolesc. Psychopharmacol. 2017, 27, 619–628. [Google Scholar]
  189. Hesselmark, E.; Bejerot, S. Patient Satisfaction and Treatments Offered to Swedish Patients with Suspected Pediatric Acute-Onset Neuropsychiatric Syndrome and Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal Infections. J. Child Adolesc. Psychopharmacol. 2019, 29, 634–641. [Google Scholar] [CrossRef] [Green Version]
  190. Mahapatra, A.; Panda, P.K.; Sagar, R. Pediatric autoimmune neuropsychiatric disorders associated with streptococcal infection treated successfully with a course of oral antibiotics. Asian J. Psychiatr. 2017, 25, 256–257. [Google Scholar] [CrossRef]
  191. Swedo, S.E.; Frankovich, J.; Murphy, T.K. Overview of Treatment of Pediatric Acute-Onset Neuropsychiatric Syndrome. J. Child Adolesc. Psychopharmacol. 2017, 27, 562–565. [Google Scholar] [CrossRef] [Green Version]
  192. Hirschtritt, M.E.; Bloch, M.H.; Mathews, C.A. Obsessive-Compulsive Disorder. JAMA 2017, 317, 1358. [Google Scholar] [CrossRef] [PubMed]
  193. Cognitive-Behavior Therapy, Sertraline, and Their Combination for Children and Adolescents with Obsessive-Compulsive Disorder. JAMA 2004, 292, 1969. [CrossRef] [PubMed] [Green Version]
  194. Ost, L.-G.; Riise, E.N.; Wergeland, G.J.; Hansen, B.; Kvale, G. Cognitive behavioral and pharmacological treatments of OCD in children: A systematic review and meta-analysis. J. Anxiety Disord. 2016, 43, 58–69. [Google Scholar] [CrossRef] [PubMed]
  195. Demirkaya, S.K.; Demirkaya, M.; Yusufo_lu, C.; Akln, E. Atomoxetine Use in Attention-Deficit/Hyperactivity Disorder and Comorbid Tic Disorder in Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal Infections. J. Child Adolesc. Psychopharmacol. 2017, 27, 104–105. [Google Scholar] [CrossRef] [Green Version]
  196. Spartz, E.J.; Freeman, G.M., Jr.; Brown, K.; Farhadian, B.; Thienemann, M.; Frankovich, J. Course of Neuropsychiatric Symptoms After Introduction and Removal of Nonsteroidal Anti-Inflammatory Drugs: A Pediatric Observational Study. J. Child Adolesc. Psychopharmacol. 2017, 27, 652–659. [Google Scholar] [CrossRef]
  197. Alexander, A.A.Z.; Patel, N.J.; Southammakosane, C.A.; Mortensen, M.M. Pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections (PANDAS): An indication for tonsillectomy. Int. J. Pediatr. Otorhinolaryngol. 2011, 75, 872–873. [Google Scholar] [CrossRef]
  198. Windfuhr, J.P.; Hilf, K.M. Tonsillectomy remains a questionable option for pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections (PANDAS). GMS Curr. Top. Otorhinolaryngol. Head Neck Surg. 2016, 15, 110–115. [Google Scholar]
  199. Demesh, D.; Virbalas, J.M.; Bent, J.P. The role of tonsillectomy in the treatment of Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal Infections (PANDAS). JAMA Otolaryngol. Head Neck Surg. 2015, 141, 272–275. [Google Scholar] [CrossRef] [Green Version]
  200. Wilbur, C.; Bitnun, A.; Kronenberg, S.; Laxer, R.M.; Levy, D.M.; Logan, W.J.; Shouldice, M.; Yeh, E.A. PANDAS/PANS in childhood: Controversies and evidence. Paediatr. Child Health 2019, 24, 85–91. [Google Scholar] [CrossRef]
  201. Pavone, P.; Falsaperla, R.; Nicita, F.; Zecchini, A.; Battaglia, C.; Spalice, A.; Iozzi, L.; Parano, E.; Vitaliti, G.; Verrotti, A.; et al. Pediatric Autoimmune Neuropsychiatric Disorder Associated with Streptococcal Infection (PANDAS): Clinical Manifestations, IVIG Treatment Outcomes, Results from a Cohort of Italian Patients. Neuropsychiatry 2018, 8, 854–860. [Google Scholar] [CrossRef]
  202. Latimer, M.E.; L’Etoile, N.; Seidlitz, J.; Swedo, S.E. Therapeutic plasma apheresis as a treatment for 35 severely Ill children and adolescents with pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections. J. Child Adolesc. Psychopharmacol. 2015, 25, 70–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Moretti, G.; Pasquini, M.; Mandarelli, G.; Tarsitani, L.; Biondi, M. What every psychiatrist should know about PANDAS: A review. Clin. Pract. Epidemiol. Ment. Health 2008, 4, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Dale, R. Autoimmunity and the basal ganglia: New insights into old diseases. QJM 2003, 96, 183–191. [Google Scholar] [CrossRef]
  205. Singer, H.S.; Loiselle, C.R.; Lee, O.; Minzer, K.; Swedo, S.; Grus, F.H. Anti-basal ganglia antibodies in PANDAS. Mov. Disord. 2004, 19, 406–415. [Google Scholar] [CrossRef] [PubMed]
  206. Yazici, K.U.; Percinel Yazici, I.; Ustundag, B. Vitamin D levels in children and adolescents with obsessive-compulsive disorder. Nord. J. Psychiatry 2018, 72, 173–178. [Google Scholar] [CrossRef] [PubMed]
  207. Stagi, S.; Lepri, G.; Rigante, D.; Cerinic, M.M.; Falcini, F. Cross-Sectional Evaluation of Plasma Vitamin D Levels in a Large Cohort of Italian Patients with Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal Infections. J. Child Adolesc. Psychopharmacol. 2018, 28, 124–129. [Google Scholar] [CrossRef] [PubMed]
  208. Boileau, B. A review of obsessive-compulsive disorder in children and adolescents. Dialogues Clin. Neurosci. 2011, 13, 401–411. [Google Scholar] [PubMed]
  209. Centonze, D.; Gubellini, P.; Pisani, A.; Bernardi, G.; Calabresi, P. Dopamine, Acetylcholine and Nitric Oxide Systems Interact to Induce Corticostriatal Synaptic Plasticity. Rev. Neurosci. 2003, 14, 207–216. [Google Scholar] [CrossRef]
Figure 1. Common and distinct symptoms characteristic to PANDAS and PANS.
Figure 1. Common and distinct symptoms characteristic to PANDAS and PANS.
Ijms 21 01476 g001
Table 1. Characteristics of Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal Infections (PANDAS), Pediatric Acute-Onset Neuropsychiatric Syndrome (PANS), Sydenham’s chorea, and Tourette syndrome.
Table 1. Characteristics of Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal Infections (PANDAS), Pediatric Acute-Onset Neuropsychiatric Syndrome (PANS), Sydenham’s chorea, and Tourette syndrome.
Psychiatric Syndrome SymptomsChanges in CNSChanges in MicrobiotaGenetic Alterations/MutationsPresence of AntibodiesAvailable Treatment
PANDASOCD, tics, obsessions, compulsions [60], anxiety, agitation, aggression, insomnia, impulsiveness, emotional lability, depression, hyperactivity, suicidality [54], dysuric symptoms, dilated pupils, inattention, behavioral regress, dysgraphia, visual and auditory hallucinations, irritation [61] Enlarged striatum, caudate, putamen, basal ganglia and globus pallidus [62,63], dysregulation within striatal interneurons [64], overactivation of microglia [65], increased number of antineuronal antibody titers [63]Higher percentage of Bacteroidetes, Rikenellaceae and Odoribacteriaceae; lower of Firmicutes and Actinobacteria; absence of Saccharibacteria and Turicibacteraceae, Tissierellaceae, Gemellaceae, and Carnobacteriaceae (Bacilli class), Corynebacteriaceae and Lachnospiracea (Clostridia class) [66]MBL2 [67], TNF-α −308 AA polymorphism [68]Autoantibodies against: dopamine D1, D2 receptor, tubulin, lysoganglioside, antipyruvate kinase [69,70], calcium calmodulin dependent kinase II [71]Penicillin V, amoxicillin, azithromycin [72,73], steroids, antipsychoactive drugs (risperidone) [74], immunotherapeutic, plasmapheresis [75], intravenous immunoglobulins [76], cognitive-behavioral therapy, SSRIs [77], vitamin D supplementation [78]
PANSOCD, tics, impaired food intake, anxiety, separation fear, motor dysfunctions, dysgraphia, behavioral regress, regress in school outcome, emotional lability, irritability, aggression, oppositional behaviours, dysuric symptoms, insomniaNDHigher percentage of Bacteroidaceae, Rikenellaceae, and Odoribacteriaceae
lower level of Firmicutes, absence of Turicibacteraceae, Tissierellaceae, Gemellaceae, and Carnobacteriaceae (Bacilli class); Corynebacteriaceae and Lachnospiraceae (Clostridia class); Bifidobacteriaceae (Actinobacteria) and Erysipelotrichaceae [66]
NDAutoantibodies against: dopamine D1, D2 receptor, tubulin, lysoganglioside, calcium calmodulin dependent kinase II [71]First-line antibiotics: penicillin, amoxicillin
Alternative antibiotics: azithromycin, cefadroxil, cephalexin, cefpodoxime
Other:
Intravenous immunoglobulin, therapeutic plasma exchange, corticosteroids, SSRIs, non-SSRI-antidepressants, ADHD medication, antipsychotics, anxiolytics, mood-stabilizers, cognitive-behaviour therapy [79,80]
Sydenham’s choreaOCD, tics, ADHD, generalized anxiety, mood disorders, psychotic features, emotional lability, irritability, regressive behaviour, separation anxiety, panic disorder, phobias, social phobia, agoraphobia, executive dysfunction [81]Permanent basal ganglia damage, dysfunction in the connection between the basal ganglia and the superior colliculi [56]NDNDAutoantibodies against: dopamine receptor D2, anti-basal ganglia antibodies [82]Penicillin, neuroleptics (risperidone, haloperidol), sodium valproate, corticosteroids, plasma exchange, intravenous immunoglobulins, dopamine depleters (tetrabenazine, deutetrabenazine, valbenazine), dopamine antagonists [83]
Tourette’s syndromeOCD, tics, ADHD, depression, bipolar disorder, anxiety, personality disorder, learning disability, speech and language disorder, intellectual disability, trichotillomania, sleep disorders, pathologic nail-biting, simple phobia, social phobia, agoraphobia, impulse control disorders (intermittent), explosive disorder, self-injurious behavior, impulsive-compulsive sexual behavior [84,85]Increased functional connectivity between the basal ganglia nuclei (right and left caudate, putamen and left pallidum) and cortex (superior temporal gyrus, medial temporal gyrus, paracingulate gyrus, precuneus, angular gyrus, insular cortex)
Increased connectivity between left thalamus and the cortex (right planum temporale, right superior temporal gyrus) [86,87]
ND CELSR3, FLT3, COL27A1, SLITRK1, SLITRK5, HDC, NRXN1, CNTN6, DRD3, GDNF, KCNJ5, AADAC [88,89] Antibodies against: rheumatogenic GAS serotypes (M12, M19), anti-neuronal antibodies, antibodies to membrane isoforms of glycolytic enzymes (aldolase C, enolase, pyruvate kinase M1), anti-HCN4 antibodies [90,91,92] First line: aripiprazole, sulpiride, risperidone
Second line: clonidine, guanfacine
Third line: topiramate, pimozide, tetrabenazine
Fourth line: clonazepam, haloperidol
Also: botulinum toxin, SSRIs (sertraline), tricyclics (clomipramine), stimulants (methylphenidate, dextroamphetamin), non-stimulants (atomoxetine, clonidine, guanfacine)
Other: behavioural treatments, deep brain stimulation [87]

Share and Cite

MDPI and ACS Style

Baj, J.; Sitarz, E.; Forma, A.; Wróblewska, K.; Karakuła-Juchnowicz, H. Alterations in the Nervous System and Gut Microbiota after β-Hemolytic Streptococcus Group A Infection—Characteristics and Diagnostic Criteria of PANDAS Recognition. Int. J. Mol. Sci. 2020, 21, 1476. https://doi.org/10.3390/ijms21041476

AMA Style

Baj J, Sitarz E, Forma A, Wróblewska K, Karakuła-Juchnowicz H. Alterations in the Nervous System and Gut Microbiota after β-Hemolytic Streptococcus Group A Infection—Characteristics and Diagnostic Criteria of PANDAS Recognition. International Journal of Molecular Sciences. 2020; 21(4):1476. https://doi.org/10.3390/ijms21041476

Chicago/Turabian Style

Baj, Jacek, Elżbieta Sitarz, Alicja Forma, Katarzyna Wróblewska, and Hanna Karakuła-Juchnowicz. 2020. "Alterations in the Nervous System and Gut Microbiota after β-Hemolytic Streptococcus Group A Infection—Characteristics and Diagnostic Criteria of PANDAS Recognition" International Journal of Molecular Sciences 21, no. 4: 1476. https://doi.org/10.3390/ijms21041476

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop