Ultrarare Variants in DNA Damage Repair Genes in Pediatric Acute-Onset Neuropsychiatric Syndrome or Acute Behavioral Regression in Neurodevelopmental Disorders =============================================================================================================================================================== * Janet L. Cunningham * Jennifer Frankovich * Robert A. Dubin * Erika Pedrosa * Refıa Nur Baykara * Noelle Cathleen Schlenk * Shahina B. Maqbool * Hedwig Dolstra * Jacqueline Marino * Jacob Edinger * Julia M. Shea * Gonzalo Laje * Sigrid M.A. Swagemakers * Siamala Sinnadurai * Peter J. van der Spek * Herbert M. Lachman ## Abstract Acute onset of severe psychiatric symptoms or regression may occur in children with premorbid neurodevelopmental disorders, although typically developing children can also be affected. Infections or other stressors are likely triggers. The underlying causes are unclear, but a current hypothesis suggests the convergence of genes that influence neuronal and immunological function. We previously identified 11 genes in Pediatric Acute-Onset Neuropsychiatry Syndrome (PANS), in which two classes of genes related to either synaptic function or the immune system were found. Among the latter, three affect the DNA damage response (DDR): *PPM1D, CHK2,* and *RAG1*. We now report an additional 17 cases with mutations in *PPM1D* and other DDR genes in patients with acute onset of psychiatric symptoms and/or regression that were classified by their clinicians as PANS or another inflammatory brain condition. The genes include clusters affecting p53 DNA repair (*PPM1D*, *ATM, ATR*, *53BP1,* and *RMRP*), and the Fanconi Anemia Complex (*FANCE, SLX4/FANCP, FANCA, FANCI,* and *FANCC*). We hypothesize that defects in DNA repair genes, in the context of infection or other stressors, could lead to an increase in cytosolic DNA in immune cells triggering DNA sensors, such as cGAS-STING and AIM2 inflammasomes. These findings could lead to new treatment strategies. ## Introduction We recently reported the first whole exome and whole genome sequencing (WES and WGS, respectively) analysis in Pediatric Acute-Onset Neuropsychiatry Syndrome (PANS) in which ultrarare variants were found in 11 genes in 21 cases1. The genes harboring ultrarare variants (minor allele frequency <0.001) showed extensive heterogeneity but clustered to those that affect innate and adaptive immunity; *PPM1D* (3 cases), *NLRC4* (4 cases), *RAG1* (3 cases), *PLCG2,* and *CHK2*, or genes that affect synaptic function; *SGCE* (2 cases), *CACNA1B* (2 cases), *SHANK3* (3 cases), and one case each for *GRIN2A, GABRG2,* and *SYNGAP1.* Of the 21 subjects, five were diagnosed with autism spectrum disorder (ASD) before their deterioration that met PANS criteria. In addition to ASD, four cases were diagnosed with another neurodevelopmental disorder (NDD), one of whom had Jansen de Vries Syndrome (JdVS), which is caused by truncating mutations in *PPM1D* exons 5 or 6 that cause a gain-of-function effect by suppressing p53 and other proteins involved in the DNA damage repair response (DDR) (e.g., MDM2, ATM, CHK1, CHK2, ATR, and H2AX)2–6. The high prevalence of PANS superimposed on pre-existing NDD is consistent with the observations of other groups7, 8. PANS is thought to be an inflammatory disorder that primarily affects the deep brain grey matter (primarily the basal ganglia but the thalamus and amygdala may be involved in more extensive cases). This hypothesis is based on the cardinal symptoms of PANS and is supported by four neuroimaging studies showing the following: swelling of the basal ganglia during the acute stage, microglial activation in the caudate and globus pallidus, grey/white matter differences in the basal ganglia, and microstructural changes throughout the brain but most prominently in the basal ganglia9–12. The clinical diagnosis PANS has core symptoms of abrupt onset obsessive-compulsive disorder (OCD) and/or restricted eating (food aversion), with two or more secondary symptoms: anxiety, emotional lability, irritability, rage, cognitive regression, sleep disturbance, sensory dysregulation, movement abnormalities, urinary symptoms including new onset dysuria or urinary frequency13. While the criteria require 2 secondary symptoms, most patients have 5 to 6 secondary symptoms that start abruptly alongside the OCD and/or eating restriction. Autonomic instability (POTS, dilated pupils, increased urinary frequency, enuresis) and hypermobility have also been reported as co-morbid with PANS13–18. A deterioration in school performance occurs, exemplified by the loss of previously learned skills and/or new onset procedural learning challenges presumably relating to the basal ganglia inflammation PANS13–18. Activities of daily living, extracurricular activities, and social interactions are severely affected during relapses. PANS can occur in children with or without premorbid ASD or other NDD. For parents, the abrupt onset is a telling feature with a rapid decline in behavior and functioning that is sometimes, literally, overnight or emerging over a period of a week. The course is typically relapsing and remitting, but not all relapses are hyperacute/acute. Relapses are frequently associated with an infection and data in support of a neuroinflammatory etiology for PANS is accumulating. In the subgroup of cases following Streptococcus infections, a range of group A streptococcal (GAS) autoantibodies and antibodies to striatal cholinergic interneurons can be detected15, 19–21. Recently, an increased prevalence of folate receptor alpha autoantibodies associated with cerebral folate deficiency and ASD were detected in PANS22. Additionally, cross-sectional prevalence data show that approximately one-third of patients meeting PANS criteria eventually develop enthesitis-related arthritis (ERA) and/or inflammatory back pain further supporting a role for inflammation in this disorder18, 23. Furthermore, inflammatory/autoimmune disorders are prevalent in first-degree family members8, 24, 25. As is true in other rare pediatric rheumatological conditions (including ERA and Sydenham chorea), observational studies in PANS suggest that non-steroidal anti-inflammatory drugs (NSAIDs) and corticosteroids may alleviate symptoms and/or shorten the length of the relapses26–28. More potent immunomodulators, such as intravenous immunoglobulin (IVIg) and a B-cell inhibitor Rituximab have mixed results, likely due to the genetic heterogeneity and the difficulty in appropriately powering randomized trials in relapsing/remitting conditions29, 30. An immune-based etiology is also suggested in some cases of ASD and NDD, especially acute and subacute regression, which have some symptoms that overlap with PANS and respond to immunomodulators31–34. In addition, a strong family history of autoimmune disorders is associated with an increased risk for ASD35–37. We are interested in identifying genetic factors that underlie the development of PANS and acute behavioral regression using next-generation sequencing. In our original paper, two of the ultrarare variants we found are in genes that affect DDR (*PPM1D* and *CHK2*), and one induces DNA breaks (*RAG1*). *RAG1*, along with *RAG2*, codes for endonucleases that induce DNA double-strand breaks responsible for the somatic recombination that generates immune cell diversity in B- and T-lymphocytes1, 38, 39. RAG1/RAG2 also affects NK cell differentiation and plays a role in DNA transposition40, 41. Double-stranded breaks induced by the RAG complex are repaired by ATM, a serine/threonine kinase that orchestrates DNA repair at double-strand breaks39, 42–44. Our overarching aim is to identify genetic variations that converge on common biological pathways within the heterogeneous population of children with PANS or acute-onset regression with or without premorbid NDD. Given the findings in our first PANS genetic study, one common pathway is DNA repair. This hypothesis is supported by the findings in this paper in which many other genes involved in DNA repair were identified that may contribute to genetic vulnerability. ## Subjects and Methods ### Ethical considerations This study was conducted in accordance with the Declaration of Helsinki. Parents signed informed consent approved by the Albert Einstein College of Medicine IRB (2022-14636). #### Subjects The subjects were identified through connections between the senior investigators and a PANS group called EXPAND, a non-profit European advocacy organization for families of children, adolescents, and adults with immune-mediated neuropsychiatric disorders ([https://expand.care/](https://expand.care/)), through the Neuroimmune Foundation, a non-profit organization dedicated to neuroimmune and inflammatory brain conditions ([https://neuroimmune.org/about/](https://neuroimmune.org/about/)), The Louisa Adelynn Johnson Fund for Complex Disease ([https://tlajfundforcomplexdisease.com/](https://tlajfundforcomplexdisease.com/)), and the Jansen de Vries Syndrome Foundation ([https://jansen-devries.org/](https://jansen-devries.org/)). Histories were obtained by the participating physicians and confirmed and collated by one of the senior investigators who interviewed every family member (H.M.L). The cases met the criteria for PANS with or without co-morbid ASD or NDD, or individuals with ASD or other NDDs who had a history of rapid-onset behavioral regression. ### Genetic analyses Whole exome sequencing (WES) data was available for six cases (**Table**). WES was carried out by different companies Courtagen Diagnostics Laboratory, GeneDx, Baylor Genetics, and for case 10, WES was conducted locally in the Genetics Core at the Albert Einstein College of Medicine, Epigenetics Shared Facility. For the remaining cases, data was available from commercially available panels: Invitae (Primary Immunodeficiency Panel), NHS North West (NDD panel). The genetic variants for cases 1 and 4-12 were verified using Sanger sequencing (see **Table** and **Supplementary Methods**). A comprehensive description of the library preparation and analysis of sequencing data for the Einstein sample (case 10) can be found in the **Supplementary Methods** section. Parents were genotyped for variants found in all cases except cases 10, and 13-16. View this table: [Table.](http://medrxiv.org/content/early/2024/02/23/2024.02.20.24302984.1/T1) Table. Description of all primary DNA repair variants. *Age range at diagnosis; Dx: diagnosis (JdVS: Jansen de Vries Syndrome, AR: acute regression, PANS: pediatric acute-onset neuropsychiatric syndrome, AE: autoimmune encephalitis, DD: developmental delay, CVID: common variable immune deficiency); variant shows the cDNA and amino acid changes; type shows how the mutation affects gene function; CADD: Combined Annotation score; N/A not available. ClinVar is the predicted pathogenicity based on the ClinVar database; AlphaMissense is the predicted pathogenicity based on amino acid change (see main text); Inheritance pattern shows that the variant in question was either de novo or transmitted from a parent. The sex of the parent was omitted to maintain anonymity; MAF: Minor allele frequency; Map position shows coordinates of variants based on hg38 build; rs number is based on SNP database; Additional variants were found in each case (see Supplemental table for descriptions). Genome sequencing shows the different methods used to detect variants, For case 17, Invitae AAS, IEIC, and MIBD panels are Autoinflammatory and Autoimmunity Syndromes, Inborn errors of immunity and Cytopenia, and Monogenic Inflammatory Bowel Disease, respectively. ### Evaluation of Variants Minor allele frequencies (MAFs) were obtained from the GnomAD database on the UCSC Genome Browser ([https://genome.ucsc.edu/](https://genome.ucsc.edu/)). The ClinVar database, Combined Annotation-Dependent Depletion (CADD), and AlphaMissense were used to predict the pathogenicity of mutations45–48. [https://www.ncbi.nlm.nih.gov/clinvar/](https://www.ncbi.nlm.nih.gov/clinvar/). RNAfold was used to predict the secondary structure of RMRP. A detailed explanation is in the **Supplementary Methods** section. ## Results Ultrarare variants (MAF <0.001) affecting DNA repair genes were found in 17 cases (11 males and 6 females; specific sex of cases are not included to maintain anonymity) including four families with two or more affected individuals. The identified genes are involved in the p53 repair pathway: *PPM1D* (2 cases), *ATM* (5 cases,), *ATR* (2 cases), and *53BP1* (one case) (**Figure 1**), and the Fanconi Anemia Complex (FC): *FANCE* (2 cases), *FANCP/ SLX4* (2 cases), *FANCI*, *FANCA*, and *FANCC* (one case each) (**Figure 2**; see **Table** for details on all p53 and FC gene variants). The FC pathway repairs DNA interstrand crosslinks and interacts with ATM/ATR/p53 DNA repair pathways **(Figure 2)**49–54. In addition, ultrarare variants were found in 34 other genes, many of which have effects on DNA repair and innate immune function (see **Supplementary Table** for details on all additional variants described in this paper). STRING was used to show the connections between the DNA repair genes and other variants found in this study, which centered around ATM and p53 (**Figure 3**). The cases and genetic variants with their potential impact on the phenotypes are described below. ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/02/23/2024.02.20.24302984.1/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2024/02/23/2024.02.20.24302984.1/F1) Figure 1. p53 DNA repair pathway. A simplified depiction showing the regulation of p53 tumor suppressor activity by phosphorylation triggered by ATM, ATR, CHK1, and CHK2 kinases, and dephosphorylation by PPM1D. P53 activation can induce apoptosis or cell cycle arrest, which will eliminate DNA-damaged cells or provide an opportunity to repair the damage, respectively. Loss of function mutations in *ATM* or gain of function mutations in *PPM1D*, such as those found in patients with acute neuropsychiatric decompensation in cases 1-7, would be expected to lead to a decrease in p53 activity. ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/02/23/2024.02.20.24302984.1/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2024/02/23/2024.02.20.24302984.1/F2) Figure 2. Fanconi Core Complex. The Fanconi Core Complex of proteins repairs interstrand crosslink breaks (Figure adapted from reference 49). The letters are abbreviations for the Fanconi complex proteins (i.e., “A” is FANCA; “E” is FANCE, etc.). Monoubiquitinated (Ub) FANCD2 and FANCI form a dimer that binds to crosslink repair sites, which leads to the recruitment of nucleases that repair the DNA lesion (reference 53). All of the Fanconi complex genes described in this paper are depicted in bold type (FANCE, SLX4, FANCE, and FANCA). Interactions between Fanconi proteins with ATM/ATR are shown. The image was generated using biorender [https://www.biorender.com/](https://www.biorender.com/)). ![Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/02/23/2024.02.20.24302984.1/F3.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2024/02/23/2024.02.20.24302984.1/F3) Figure 3. Protein-Protein Interaction Network: STRING. A connectivity network was generated for the candidate genes using IPA software. Central to the network are ATM and p53. Genes described in the paper that do not fit into the connectivity network are not shown (C7, DBH, SLC13AS, SRPK3, NLRX1, MTO1, CACNA1H, CACNA1S, CLPB, AHNAK2). SLX4 = FANCA/SLX4, TP53=p53, and TP53BP1 = 53BP1. ### p53 DNA Repair Pathway (*PPM1D, ATM, ATR, 53BP1*) ### PPM1D (Protein phosphatase, Mg2+/Mn2+dependent 1D) Case 1 has JdVS with a 1:1 mosaicism for a de novo truncating *PPM1D* variant in exon 6. The patient was diagnosed with mild intellectual disability but was high functioning, requiring minimal support until high school when the patient had an abrupt onset of OCD, anxiety, cognitive regression, insomnia, and psychosis with rapid decline over approximately three weeks. The deterioration coincided with an upper respiratory infection and post-infectious vasculitis. Further regression occurred with the loss of previously mastered global abilities and the eventual development of mutism, which has persisted for several years, unresponsive to psychiatric medications and other clinical and behavioral interventions. Two additional ultrarare de novo variants were found and are likely pathogenic, one of which is relevant to the DDR hypothesis: a variant in *CSNK1a1,* which codes for Casein Kinase 1 Alpha 1, a p53 inhibitor55, 56. Another was found in *SRPK3,* an SRSF Protein Kinase that has no known effect on DDR, although interestingly, expression is inversely correlated with alpha-synuclein levels in mouse models of Parkinson’s disease57. Finally, an inherited ultrarare, nonsense mutation was found in the citrate transporter, *SLC13A5* as well as a splice acceptor variant in *CACNA1S*. Case 2 also has a typical JdVS variant (a truncating mutation in *PPM1D* exon 6) and was diagnosed with ASD and a learning disability. The patient has periods of abrupt onset behavioral regression lasting several days to months, characterized by eating refusal, rage, cognitive deficits, loss of previously learned skills, an inability to walk, and enuresis. The most severe relapses have occurred following infection with SARS-CoV2 and a Group A Streptococcal infection concurrent with shingles. ### ATM (Ataxia-telangiectasia mutated or serine/threonine kinase) Case 3 carries an ultrarare and pathological frameshift mutation in *ATM*. The patient was typically developing and excelled in school until adolescence when diagnosed with Postural Orthostatic Tachycardia Syndrome (POTS), irritable bowel syndrome gastroparesis, and enthesitis-related arthritis. Soon thereafter, an episode of gastroenteritis led to acute-onset OCD, anxiety, impulsivity, and severe rage; the first of many such episodes. An MRI scan was negative. Relapses responded to IVIg and steroid bursts; however, due to frequent relapses and the development of chronic symptoms, a trial of Rituximab was undertaken, which was helpful. While the patient appeared to respond to immunomodulation, severe symptoms returned when these therapies were tapered or discontinued. Multiple hospitalizations for uncontrolled psychiatric symptoms followed. A parent carries the same ATM variant and has a history of dysautonomia and an inflammatory skin condition. Other ultrarare variants found in this patient include a de novo variant in *AHNAK2* and compound heterozygosity for two ultrarare *CACNA1H* variants. While there are no obvious connections to DNA repair, AHNAK2 has been implicated in systemic lupus erythematosus (SLE) in two studies and *CACNA1H* mutations are found in ASD and intellectual disabilities58–61. Cases 4-7 are a family with four affected children who all carry a pathogenic parental transmitted ATM splice donor mutation. There is a bilneal parental history of severe autoimmune disorders and hypermobile joints, as well as an extended family history of early-onset cancer, and autoimmune disorders. This is consistent with the increased risk of cancer and autoimmune disorders in individuals with loss-of-function mutations in *ATM* 62–65. Case 4, the proband, was diagnosed with ASD and experienced a typical PANS relapsing-remitting course with abrupt relapses of OCD, food aversion, anxiety, irritability, impulse control problems, oppositional behaviors, tics, and fatigue. Relapses were commonly coincident with infections and were effectively managed by addressing infections with antibiotics and using IVIg. The proband also has a history of Hashimoto’s thyroiditis, joint hypermobility, and consistently low immunoglobulin levels which likely contributed to frequent infections. Alongside the *ATM* variant, the patient inherited rare and ultrarare variants affecting DNA repair and innate immunity including a likely pathogenic variant in *RNASEH2B*, which codes for Ribonuclease H2, an enzyme that removes ribonucleotides that have been incorporated into replicating DNA, affecting double-stranded break repair kinetics66. Homozygosity for this variant leads to Aicardi-Goutières syndrome (AGS), a type I interferonopathy characterized by neurodevelopmental defects and upregulation of type I interferon signaling and neuroinflammation due to the effects of DNA damage66, 67. The patient also has ultrarare nonsynonymous variants of unknown significance in *RAG2, NOD2*, and a 24-base pair microduplication in *SP110*. Each of these genes has effects on innate immunity, but RAG2 is the only one with a strong connection to DNA repair, as described in the introduction1, 38, 39 68. An *NOD2* variant was transmitted from one of the parents. A second ultrarare variant in *NOD2* was identified in the other parent (see **Supplementary Table**). Cases 5-7 are siblings to case 4 who were all diagnosed with ASD, dysautonomia, and joint hypermobility. They all have a relapsing-remitting course characterized by abrupt onset OCD, anxiety, emotional lability, increased urinary frequency, oppositional behavior, and reduced social interactions, along with cognitive and behavioral regression. Relapses are typically coincident with infection. Case 5 had an MRI that showed right temporal sclerosis. Cases 6 and 7 have epilepsy, low levels of IgM and IgA, and relapses improve after clearance of infection and treatment with IVIg and/or a corticosteroid burst. Although they were not analyzed by WES, each was analyzed by Sanger sequencing for the variants found in the proband. Each sibling has inherited the *ATM* variant, and cases 5 and 6, the *RNASEH2B* variant. They have also inherited one of the two *NOD2* variants. ### ATR (Ataxia telangiectasia and rad3-related protein) Cases 8 and 9 are siblings with regressive ASD and a relapsing-remitting course with relapses commonly coincident with infections. They both have a unique, transmitted ultrarare *ATR* variant of unknown significance. One parent has a history of Hashimoto’s thyroiditis and a family history of SLE, and the other parent has a history of joint hypermobility and childhood seizure disorder. ATR is a serine/threonine kinase and DNA damage sensor that functions as a master regulator of the DDR69–73. The proband, case 8, was a typically developing child with hypermobility who was well until abrupt behavioral regression occurred following “croup” at which time a diagnosis of ASD was made. The patient subsequently developed a relapsing-remitting course characterized by abrupt onset OCD, aggression, and mutism. Relapses appeared to be responsive to NSAIDs. The proband’s sibling, Case 9, was well until the development of an abrupt behavioral regression and loss of previously achieved milestones following a herpesvirus 6 infection, after which a diagnosis of ASD was made. The patient also has a relapsing-remitting course characterized by abrupt onset of OCD, restricted eating, rage, and depression. Relapses are typically coincident with infections. Symptoms typically respond to glucocorticoid bursts. MRI showed atrophy of the hippocampus, amygdala, caudate, and putamen. WES analysis of both cases 8 and 9 also revealed several other ultrarare variants of interest: *NLRX1*, *CENPJ, CACNA1S, DBH, and CLPB.* The most interesting from the DNA repair perspective is a pathogenic frameshift mutation in *CENPJ,* which codes for centromere protein J. Centromere regulation is critical for maintaining genomic stability, and defects lead to aberrant mitosis and activation of the cGAS-STING pathway73, 74. It is interesting to note that biallelic mutations in *CENPJ* and *ATR* can both cause Seckel syndrome, which is associated with cell-cycle checkpoint signaling abnormalities75, 76. Another variant was found in *NLRX1,* which codes for a key player in the innate immune response that regulates NF-κB expression and has been implicated in autoimmune and inflammatory diseases77–80. A third sibling is typically developing and did not inherit the *ATR* variant but carries the *CENPJ, CACNA1S,* and *CLPB* variants. Thus, the two affected siblings with ASD and acute-onset neuropsychiatric symptoms have ultrarare variants in two genes that affect DNA repair, *ATR,* and *CENPJ,* as well as *NLRX1 DBH, CACNA1S,* and *CLPB*. ### 53BP1 (Tumor suppressor p53-binding protein 1) Case 10 has a history of developmental delay, and experienced severe regression as a young child characterized by loss of the ability to walk, loss of verbal skills, and new-onset enuresis. Upon WES, we found an ultrarare missense mutation in *53BP1*, a p53-binding protein that regulates DDR 81–83. A preceding infection to initial deterioration was not ascertainable. However, the patient developed a relapsing-remitting course with relapses (abrupt onset of OCD, eating refusal, severe rage, and oppositional behavior) coincident with infections with improvement following treatment infections with antibiotics. The patient also developed Hashimoto’s thyroiditis, scoliosis, joint hypermobility, and gastroparesis. A parent has a history of Raynaud’s and fibromyalgia, and there is a family history of thyroid disease and Parkinson’s Disease. Parents have not been genotyped. In addition to 53BP1, several other ultrarare variants were found that could be playing a role in the clinical state via DNA repair. One is *MED1*, which codes for a mediator complex protein that plays a major role in homologous recombination, base excision repair, the maintenance of genomic stability, and p53-dependant apoptosis84. It has also been implicated in SLE85. Another is *KMT2D*, which codes for a histone methyltransferase that plays a role in DNA repair and has been implicated in autoimmune disorders86, 87. Finally, we found a nonsense mutation in *MTO1*, which regulates oxidative phosphorylation and has been found to affect DNA repair and recombination in yeast 88, 89. ### Fanconi Anemia Complex (FANCE, FANCP/SLX4, FANCA, FANCI, and FANCC) ### FANCE (Fanconi anemia complementation group E) Cases 11 and 12, a sibling pair with common variable immune deficiency (CVID) and acute onset of neuropsychiatric symptoms, both carry an ultrarare missense mutation in *FANCE*. Case 11 met the diagnostic criteria for PANS, characterized by a relapsing-remitting course of abrupt onset OCD, anxiety, and rage. Episodes were commonly coincident with group A streptococcal infection and other infections and always improved after treating the infection followed by a corticosteroid burst. IVIg (aimed to treat the underlying CVID) was associated with a reduced frequency of relapses but did not reduce the severity of relapses. Case 12 was diagnosed with ASD as a child after abrupt loss of speech and eye contact following an infection. The patient regained eye contact and sociability, but apraxia of speech continued. He regressed again later in childhood and soon thereafter was diagnosed with PANS, which was characterized by abrupt onset of OCD, anxiety, aggression (self-injurious behavior and property destruction), and tics. Symptoms improved after treating the infection followed by corticosteroids. The patient subsequently developed acute onset dysarthria and mutism and further evaluation revealed CSF pleocytosis, intracranial hypertension, and GAD antibodies (three times the upper limit of normal). Subsequent relapses responded to addressing infection followed by a corticosteroid burst. Relapse frequency (but not severity) was reduced following the introduction of IVIg to treat CVID. Due to a chronic progressive course, Rituximab was added, which was followed by documented improvements by the speech pathologist that were lost when B cells were repopulated (the speech pathologist was blinded to treatments and B cell repopulation). Consequently, Rituximab was scheduled quarterly. The clinical trajectory improved but deteriorations coincident with infections occurred, so Leflunomide was added as adjunct therapy which has led to further stabilization for 2 years. There is a positive history of inflammatory skin disease on both sides of the family, and a family history of immunodeficiency syndrome, arthritis, lupus-related autoantibodies, recurrent sepsis, and recurrent septic joints. Both cases carry an ultrarare variant of unknown significance in *VPS13B*. Case 11 carries a maternally transmitted *NOD2* variant that has been described as a risk allele in Crohn’s disease89, 90. Case 12 carries another ultrarare, pathogenic, novel variant involved in the DNA repair gene, *THRAP3*, which codes for thyroid hormone receptor-associated protein 3, an RNA processing factor involved in ATR kinase-dependent DDR and the expression of several DNA repair proteins, including FANCL 91, 92. ### FANCI (Fanconi anemia, complementation group I) Case 13 is a neurotypical child who developed a sudden onset of OCD, fatigue, restricted eating, severe anxiety, enuresis, and insomnia following an upper respiratory infection. Autoimmune thyroiditis was diagnosed based on having high titers of anti-thyroid peroxidase and anti-thyroglobulin antibodies, an elevated TSH, and an ultrasound. A nonsense mutation in *FANCI* was identified. There was a partial response to intravenous glucocorticoids, IVIg, and Rituximab. Case 11 also carries ultrarare variants in two other genes that affect DNA repair. One is a non-synonymous variant in another FC gene, *FANCP/SLX4*, which may be a benign variant. However, cases 14 and 15 also have an ultrarare variant in this gene (see below). The other is a variant in the DNA repair gene, *LIG4,* which codes for DNA Ligase IV, a key enzyme involved in repairing DNA double-strand breaks through non-homologous end joining and sealing DNA breaks that occur during V(D)J recombination93, 94. *LIG4* mutations cause autoimmune and immunodeficiency disorders94, 95. ### FANCP/SLX4 Fanconi anemia, complementation group P/Structure-Specific endonuclease subunit) Cases 14 and 15 are siblings diagnosed with CVID who carry a pathogenic mutation in *TNFRSF13B*, which is one of the more common variants found in immune deficiencies96, 97. The same variant has been found in several other PANS cases (unpublished observations). They also share an ultrarare variant in *FANCP/SLX4*, but the relatively low CADD score suggests that the gene alone is not sufficient to explain the severe clinical presentation. Case 14 has a history of acute neuropsychiatric decompensation consistent with PANS, who initially responded to treatment with corticosteroid bursts and IVIg and further stabilized on Anakinra. Case 15 initially presented with anxiety but has regressed substantially, developing a seizure disorder and autoimmune encephalitis (AE) that has partially responded to Rituximab, glucocorticoids, and IVIg. Anakinra has not been effective. MRI indicates atrophy. The siblings also have an ultrarare missense mutation adjacent to a splice acceptor site in *DOCK8*, which has been implicated in immune deficiency and autoimmune disorders64, 98. Both also share a variant in *MEFV* (pyrin), an inflammasome regulator gene that is commonly mutated in autoinflammatory disorders99–102. Finally, they both have an ultrarare variant in *RMRP*, a noncoding RNA that functions as the RNA component of mitochondrial RNA processing endoribonuclease. This is a very intriguing candidate according to our DNA repair model as it acts as an inhibitor of p53103, 104. A comparison between wild-type and mutant alleles using RNAfold shows differences in the secondary structure, suggesting that it could be functional (**Supplementary Figure**). ### FANCA Fanconi Anemia, Complementation Group A Case 16 is a previously healthy and neurotypical child who presented with abrupt-onset OCD, eating restriction, severe irritability, hyperactivity, aggression, mood swings, sensory amplification, and headaches. Physical exam showed truncal instability, subtle choreiform movements, folliculitis, and findings consistent with enthesitis and arthritis (confirmed on joint ultrasound). Labs indicate persistently elevated vasculitis markers (high vWF Ag and D-dimers after COVID-19), persistently elevated C4, and persistent microcytic anemia thought to be secondary to chronic inflammation. The patient has had a secondary progressive course with relapses coincident with infections (influenza, mycoplasma, COVID-19, and sinusitis) with partial improvement after bacterial infections were treated. Psychotropics and CBT have been minimally helpful. The patient did not tolerate oral or IV steroids, IVIg, or immunomodulation aimed at treating arthritis and systemic inflammation (e.g., methotrexate, sulfasalazine, and azathioprine). There has been a partial response to NSAIDs, and more recently, a solid trajectory of improvement since introducing colchicine. An ultrarare pathogenic variant (according to AlphaMissense) was found in *FANCA*. In addition, ultrarare variants were found in five other genes that have effects on innate immune function, but no direct effect on DNA repair. The most important is a pathogenic variant in *UNC93B1*, a regulator of nucleic acid-sensing toll-like receptors (e.g., TLR3, TLR7, and TLR8) and interferon and STING signaling, suggesting a synergistic interaction with FANCA-related DNA repair defects105–107. The others are ultrarare variants of unknown significance in *NLRP1, TICAM1, TYK2*, and *TCN2*. NLRP1, TICAM1, and TYK2 have regulatory effects on the innate immune system, including nucleic acid sensing, and have been implicated in autoimmune disorders108–111. First-degree relatives have migraines, recurrent sinusitis, recurrent pancreatitis, and spondylarthritis. First-degree family members also have anxiety, OCD, ADHD, and depression. ### FANCC Fanconi Anemia, Complementation Group C Case 17 had a typically developing childhood but developed OCD, ADHD, anxiety symptoms in pre-school, followed by three significant deteriorations over the next couple of years resulting in massive escalation of psychiatric symptoms - OCD, eating restriction, anxiety, irritability, extreme levels of oppositionality, poor sleep, polyuria, school refusal, and refusal to engage in basic routine personal care. Each deterioration resulted in prolonged absences from school and uncontrollable behavior when able to attend. Treatment with an SSRI increased rage and resulted in the emergence of suicidality, which led to the addition of a mood stabilizer. Eventually, he was diagnosed with ASD. A medical work-up revealed: low C3, low C4, anti-histone antibodies, microcytic anemia, livedo reticularis, enthesitis related arthritis, mild proteinuria, sinusitis, and mild hypothyroidism. Treatment of suspected sinusitis followed by high dose IVIg and a steroid burst resulted in a clear reduction in all psychiatric symptoms, the first improvement seen in two years. Based on the arthritis diagnosis, and concern for relapse after being weaned from steroids, the patient was started on standard of care for arthritis (NSAIDS, methotrexate, sulfasalazine) and was able to return to school. Ongoing ADHD, OCD, and anxiety, which have been relatively mild compared to previous levels, have been controlled with pharmacological and behavioral therapy. There have been minor flares in psychiatric symptoms and enthesitis after significant illnesses (e.g., flu, sinusitis) which are treated with standard-of-care antibiotics for bacterial infections and early introduction of NSAIDs according to PANS treatment guidelines. He has had no further major relapses and has become a good student who is socially engaged at school and home. There is a history of acute rheumatic fever, spondylarthritis, enthesitis related arthritis, psoriasis, and mood disorders in first degree relatives. A pathogenic frameshift variant was found in *FANCC.* FANCC deficiency has been found to mediate microglial pyroptosis and neuronal apoptosis in a mouse spinal cord contusion model, and regulate mitophagy by interacting with Parkin112, 113. Interestingly, we have identified several PANS and regressive ASD cases with *PRKN* copy variants (unpublished observations). Ultrarare variants were also found in *PARN*, and *IGLL1,* neither of which has significant direct effects on DNA repair pathways, although *PARN*, which codes for Poly(A)-Specific Ribonuclease, regulates mRNA 3’ end cleavage reactions under DNA-damaging conditions114. IGLL1 is a regulator of B cell differentiation and deficiency leads to autosomal recessive agammaglobulinemia115, 116. Both variants were scored as benign according to AM, but the CADD score for the *PARN* variant was 26.8 (**Supplementary Table**). ## Discussion Sixteen cases were identified with ultrarare variants in protein-coding genes related to p53 and FC DNA repair pathways*: PPM1D, ATM, ATR, 53BP1, FANCE, FANCI, FANCP/SLX4, FANCA,* and *FANCC*. These were often accompanied by additional rare variants related to DNA repair and/or immune deficiencies. Most individuals in this study have an underlying neurodevelopmental disorder, with approximately half diagnosed with ASD. The unifying feature for these cases is the distinct, acute onset change in behavior, severe psychiatric symptoms, and loss of function in association with infections and other stressors. Many cases have symptoms consistent with PANS, while others had acute, severe neuropsychiatric decompensation (e.g., mutism, behavioral regression, loss of previously learned skills, cognitive dysfunction) that did not meet clinical criteria for PANS. Co-morbid inflammatory conditions were common, including vasculitis signs (n=2), enthesitis and/or arthritis (n=4), thyroiditis, and/or elevated thyroid antibodies (n=3). Inflammatory conditions were also common among first-degree family members. Three cases had abnormal MRI scans (cases 5, 9, and 15), and one (case 3) had a normal study. Pleocytosis and moderately elevated anti-GAD antibodies were found in case 12. Clinical improvement was observed in most cases following the resolution of infections and the application of immunomodulation (NSAIDS, corticosteroids, IVIg, Rituximab, colchicine, etc.). The findings are congruent with our previous work and provide further support for our hypothesis that DNA repair deficits are a vulnerability factor for neuropsychiatric decompensation. The DNA repair hypothesis first emerged from our original genetic analysis in which three out of the five immune genes we identified have strong connections to this process: *PPM1D, CHK2*, and *RAG1*1. The idea that DNA repair deficits underlie PANS and acute decompensation in NDD is novel, although studies linking DDR to behavior have been described. For example, postmitotic genome instability in neurons can lead to behavioral alterations and neurodegenerative disorders, and genetic studies show that DNA repair genes are involved in genetic subgroups of ASD and NDDs including most of the genes described in this report (e.g., *PPM1D, ATM, ATR, 53BP1, FANCE, FANCI,* and *FANCP/SLX4*)117–124. The two major gene families affecting DNA repair described in this paper code for proteins involved in the p53 and FC pathways. So far, between the current study and our previous report, we have found ultrarare variants in five protein-coding genes that directly affect p53: *PPM1D, ATM*, *CHK2*, *ATR*, and *53BP1*. Among these genes, pathogenic mutations were found in *PPM1D, ATM*, and, in our previous study, *CHK2.* The ultrarare *ATR* and *53BP1* variants described in this paper, however, are variants of unknown significance that will require replication and functional validation. Remarkably, we also found an ultrarare variant in the long non-coding RNA gene, *RMRP*, a p53 inhibitor103, 104. *RMRP* expression is induced by immune activators and is increased in autoimmune/autoinflammatory disorders125. As described in the results section, structural differences between the mutant and wild-type alleles were predicted by RNAfold, but again, functional validation would be needed to determine whether the *RMRP* variant we identified affects p53 function and DNA repair. Ultrarare variants were found in five different FC genes: *FANCE, FANCP*/*SLX4*, *FANCI, FANCA,* and *FANCC.* Among these, all are predicted to be pathogenic, except the *FANCP/SLX4* variants. However, the fact that two different ultrarare variants were found in this gene in three cases from two families increases the likelihood they are playing a role. Interestingly, FC proteins interact with components of the p53 DDR pathway, including ATM and ATR (**Figure 2**)126–129. Some researchers have suggested that autoimmune and/or autoinflammatory processes underlie the development of behavioral regression in PANS and in a subgroup of individuals with ASD34, 130–132. Impaired DDR may be one mechanism for these associations as it can lead to leakage of nuclear DNA into the cytosol, which can trigger the cGAS-STING pathway, a key component of the type I interferon (IFN-I) anti-viral innate immune response, especially following an infectious disease trigger65, 133–135. Mitochondrial DNA breaks and oxidized mitochondrial DNA released into the cytosol can also trigger cGAS-STING136–138. AIM2 inflammasome signaling, which leads to the induction of IL-1β and IL-18, is also triggered by cytosolic DNA139, 140. Physiological activation of cGAS-STING is central for the response to foreign viral DNA, and perhaps viral RNA as well40. However, studies show that over-activation can lead to autoimmune, autoinflammatory, and neurodegenerative disorders141–144. For example, increased IFN-I responses mediated by cGAS-STING due to abnormal clearance of cytosolic DNA and RNA have been implicated in the pathogenesis of autoimmune disorders including SLE and aggressive rheumatoid arthritis145–148. Circulating type I IFN levels are elevated in approximately 50% of patients with SLE and IFN receptor inhibition is an approved therapy149, 150. A substantial fraction of SLE patients have neuropsychiatric symptoms151, 152. It has also been proposed that some neurodegenerative disorders involve cGAS-STING signaling, including Alzheimer’s Disease (AD), Parkinson’s Disease (PD), Amyotrophic Lateral Sclerosis (ALS), Huntington’s disease (HD), and multiple sclerosis (MS)153–155. Markers for neurodegenerative disorders are elevated in 27% of patients with psychiatric disease preselected for suspected immunological involvement and phenotypes including catatonia, agitation, and acute onset of symptoms similar to the cases described here, although the genetic data is not available ([https://doi.org/10.21203/rs.3.rs-3491787/v1](https://doi.org/10.21203/rs.3.rs-3491787/v1)). Early immunological interventions may mitigate some damage, but this needs to be confirmed in clinical trials. However, other forms of damage may accumulate over time that are not responsive to immunological interventions. In addition to nuclear DNA, disruption of mitochondrial integrity can also lead to the release of mitochondrial DNA into the cytoplasm, which is a potent cGAS-STING trigger136–138. A few of the genes described in this report have effects on mitochondria (e.g., *MTO1, SLC13A5, RMRP*). Some FC proteins also regulate mitochondrial DNA replication fork stability that can activate the mtDNA-dependent cGAS/STING response when dysfunctional, and FANCI has been found to regulate PRKN-mediated mitophagy156, 157. A large number of cases had more than one DNA repair gene mutation. This could be due to ascertainment bias since most of the cases we studied had severe symptoms and accompanying neurodevelopmental problems, which could have prompted genetic testing by their physicians. Gene panels rather than WGS per se were performed in many cases, which could cause ascertainment bias due to the selection of genes represented on the commercially available gene panels (**Table**). However, the histories of autoimmune disorders from both parents in many cases suggest true bilineal inheritance of risk variants. The current paper explores a genetic subgroup linked to impaired DNA repair with acute onset of severe psychiatric symptoms or regression in children associated with infections. It is important to note that non-infectious cellular stressors can also induce DNA damage and activate cGAS-STING, such as cellular injury, heat shock, and oxidative stress158, 159. In addition, recent studies suggest that faulty DNA repair could disrupt several cellular processes beyond an innate immune response, such as autophagy, senescence, and apoptosis160–162. Clinically, if our findings are confirmed, they could lead to specific diagnostic tools and new treatments for infection-associated psychiatric episodes and behavioral decompensation in PANS, ASD, and other NDDs in those with DNA repair mutations, targeting putative over-stimulated immune pathways (e.g., type I interferons and AIM2 inflammasomes). To validate our findings, more studies are needed to integrate longitudinal clinical data, genetics, and immunophenotypes. Functional studies on immune cells from patients, and microglia from animal models and induced pluripotent stem cells (iPSCs) are also needed. Studies using iPSC-derived microglia with genetic variants in DNA repair genes are ongoing. ## Supporting information supplemental methods [[supplements/302984_file06.docx]](pending:yes) supplementary table [[supplements/302984_file07.xlsx]](pending:yes) supplementary figure [[supplements/302984_file08.docx]](pending:yes) ## Author contributions J.L.C. co-wrote the paper, and was involved in the study concept and design; J.F. contributed cases, provided genetic data for cases 16, and 17, and co-wrote the manuscript); R,A.B. carried out WES on case 10); E.P. processed DNA, validated variants, edited manuscript, created figures 1,3; R.F.A. validated variants; N.C.S. contributed case 16; S.B.M. made the WES library for case 10; H.D. involved in study concept; J.M. validated variants); J.E. validated variants; J.M.S. validated variants carried out Alphamissense analysis; G.L. contributed case 1, edited manuscript); S.M.A.S. bioinformatics of variants, created Figure 4; S.S. edited manuscript;P.J.S. study concept and design, bioinformatics; H.M.L.co-wrote paper, study concept and design, analyzed variants, confirmed diagnoses, supervised study. All authors reviewed the manuscript. ## Data availability statement Variants reported in this paper will be submitted to ClinVar. WES data on case 10 will be made available at GenBank ® ## Competing interests J.L.C. has received lecturing fees from Otsuka Pharma Scandinavia, Janssen-Cilag AB and H. Lundbeck AB. ## Supplementary Data **Supplementary Methods.** An expanded description of the methods and primers used for Sanger DNA sequencing. **Supplementary Table**. Description of additional gene variants found upon whole exome sequencing or the analysis of gene panels. **Supplementary Figure. RMRP secondary structures.** The RNAfold web server was used to predict secondary structures for wild-type and mutant (c. C126T) RMRP RNAs. Differences between the two, especially in the centroid secondary structures, are seen. ## Acknowledgments H.M.L. is supported by the National Institute of Child Health and Human Development NIH/NICHD; P30 HD071593 to the Albert Einstein College of Medicine’s Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, and the National Institute of Mental Health, R21MH131740. The Lachman lab also receives support from the Janice C. Blanchard Family Fund and the iPS Cell Research for Ryan Stearn Fund. This work was funded by grants to the J.L.C Swedish Research Council (Grant No. 2019-06082). P.J. van der Spek is supported by EU H2020 grants, an ImmunAID grant (ID: 7792950), and a MOODSTRATIFICATION grant (ID: 754740). The Bioinformatics infrastructure and team are supported by grants from KWF, NWO/ZonMW, and the Dutch Heart Foundation through the BDVA-initiated H2020 Bigmedilytics program on Personalized Medicine. Janet Cunningham is a Gullstrand Fellow at Uppsala University Hospital. Jennifer Frankovich’s research is supported by several foundations including the Neuroimmune Foundation, Lucile Packard Foundation for Children’s Health, the Dollinger Biomarker Core, Stanford Spark, and collaborations on NIH grants. Funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. We thank the Albert Einstein College of Medicine Epigenomics Shared Core Facility RRID: SCR_023284 for Library preparation, QC, and sequencing, and Yongmei Zhao for technical assistance with the library preps. The authors want to thank the participating families. ## Footnotes * We included an additional case; case 17, which has a pathogenic mutation in FANCC. This lends further support to our hypothesis that mutations in Fanconi complex genes increases the risk of acute neuropsychiatric decompensation in children * Received February 20, 2024. * Revision received February 23, 2024. * Accepted February 23, 2024. * © 2024, Posted by Cold Spring Harbor Laboratory The copyright holder for this pre-print is the author. All rights reserved. The material may not be redistributed, re-used or adapted without the author's permission. ## References 1. 1.Trifiletti, R., et al. Identification of ultra-rare genetic variants in pediatric acute onset neuropsychiatric syndrome (PANS) by exome and whole genome sequencing. Sci. Rep. 12, 11106–3 (2022). 2. 2.Bhattacharya, D., Hiregange, D. & Rao, B. J. ATR kinase regulates its attenuation via PPM1D phosphatase recruitment to chromatin during recovery from DNA replication stress signalling. J. Biosci. 43, 25–47 (2018). 3. 3.Jansen, S., et al. De Novo Truncating Mutations in the Last and Penultimate Exons of PPM1D Cause an Intellectual Disability Syndrome. Am. J. Hum. Genet. 100, 650–658 (2017). 4. 4.Gräf, J. F., et al. Substrate spectrum of PPM1D in the cellular response to DNA double-strand breaks. iScience 25, 104892 (2022). 5. 5.Nahta, R. & Castellino, R. C. Phosphatase magnesium-dependent 1 δ (PPM1D), serine/threonine protein phosphatase and novel pharmacological target in cancer. Biochem. Pharmacol. 184, 114362 (2021). 6. 6.Zekavat, S. M., et al. TP53-mediated clonal hematopoiesis confers increased risk for incident atherosclerotic disease. *Nat*. Cardiovasc. Res. 2, 144–158 (2023). 7. 7.Jyonouchi, H., Geng, L. & Davidow, A. L. Cytokine profiles by peripheral blood monocytes are associated with changes in behavioral symptoms following immune insults in a subset of ASD subjects: an inflammatory subtype? J. Neuroinflammation 11, 187–2 (2014). 8. 8.O’Dor, S. L., et al. A Survey of Demographics, Symptom Course, Family History, and Barriers to Treatment in Children with Pediatric Acute-Onset Neuropsychiatric Disorders and Pediatric Autoimmune Neuropsychiatric Disorder Associated with Streptococcal Infections. J. Child Adolesc. Psychopharmacol. 32, 476–487 (2022). 9. 9.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 157, 281–283 (2000). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1176/appi.ajp.157.2.281&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10671403&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000085169000024&link_type=ISI) 10. 10.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]-PK11195. J. Child Neurol. 30, 749–756 (2015). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1177/0883073814543303&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25117419&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) 11. 11.Cabrera, B., et al. Neuroanatomical features and its usefulness in classification of patients with PANDAS. CNS Spectr. 24, 533–543 (2019). 12. 12.Zheng, J., et al. Association of Pediatric Acute-Onset Neuropsychiatric Syndrome With Microstructural Differences in Brain Regions Detected via Diffusion-Weighted Magnetic Resonance Imaging. *JAMA Netw*. Open 3, e204063 (2020). 13. 13.Chang, K., et al. Clinical evaluation of youth with pediatric acute-onset neuropsychiatric syndrome (PANS): recommendations from the 2013 PANS Consensus Conference. J. Child Adolesc. Psychopharmacol. 25, 3–13 (2015). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1089/cap.2014.0084&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25325534&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) 14. 14.Vreeland, A., et al. Neuroinflammation in Obsessive-Compulsive Disorder: Sydenham Chorea, Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal Infections, and Pediatric Acute Onset Neuropsychiatric Syndrome. Psychiatr. Clin. North Am. 46, 69–88 (2023). 15. 15.Vreeland, A., et al. Post-infectious inflammation, autoimmunity, and OCD: Sydenham Chorea, Pediatric Autoimmune Neuropsychiatric Disorder Associated with Streptococcal infection (PANDAS), and Pediatric Acute-onset Neuropsychiatric Disorder (PANS). Dev. Neurosci. (2023). 16. 16.Thienemann, M., et al. Clinical Management of Pediatric Acute-Onset Neuropsychiatric Syndrome: Part I-Psychiatric and Behavioral Interventions. J. Child Adolesc. Psychopharmacol. 27, 566–573 (2017). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1089/cap.2016.0145&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) 17. 17.Chan, A., et al. Children With PANS May Manifest POTS. Front. Neurol. 13, 819636 (2022). 18. 18.Ma, M., et al. Arthritis in children with psychiatric deteriorations: a case series. Dev. Neurosci., 1 (2023). 19. 19.Swedo, S. E., et al. Pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections: clinical description of the first 50 cases. Am. J. Psychiatry 155, 264–271 (1998). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9464208&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000071809700018&link_type=ISI) 20. 20.Cunningham, M. W. Molecular Mimicry, Autoimmunity, and Infection: The Cross-Reactive Antigens of Group A Streptococci and their Sequelae. Microbiol. Spectr. 7, doi:10.1128/microbiolspec.GPP3-2018 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1128/microbiolspec.GPP3-2018&link_type=DOI) 21. 21.Hyman, S. E. PANDAS: Too Narrow a View of the Neuroimmune Landscape. Am. J. Psychiatry 178, 5–7 (2021). 22. 22.Wells, L., O’Hara, N., Frye, R. E., Hullavard, N. & Smith, E. Folate Receptor Alpha Autoantibodies in the Pediatric Acute-Onset Neuropsychiatric Syndrome (PANS) and Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal Infections (PANDAS) Population. Journal of personalized medicine 14, 166 (2024). 23. 23.Frankovich, J., et al. Multidisciplinary clinic dedicated to treating youth with pediatric acute-onset neuropsychiatric syndrome: presenting characteristics of the first 47 consecutive patients. J. Child Adolesc. Psychopharmacol. 25, 38–47 (2015). 24. 24.Masterson, E. E. & Gavin, J. M. Baseline characteristics of children in the International PANS Registry (IPR) Epidemiology Study. BMJ Open 14, e072743–072743 (2024). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYm1qb3BlbiI7czo1OiJyZXNpZCI7czoxMjoiMTQvMS9lMDcyNzQzIjtzOjQ6ImF0b20iO3M6NTI6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDIvMjMvMjAyNC4wMi4yMC4yNDMwMjk4NC4xLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 25. 25.Chan, A., et al. Familial Clustering of Immune-Mediated Diseases in Children with Abrupt-Onset Obsessive Compulsive Disorder. J. Child Adolesc. Psychopharmacol. 30, 345–346 (2020). 26. 26.Brown, K., et al. Pediatric Acute-Onset Neuropsychiatric Syndrome Response to Oral Corticosteroid Bursts: An Observational Study of Patients in an Academic Community-Based PANS Clinic. J. Child Adolesc. Psychopharmacol. 27, 629–639 (2017). 27. 27.Brown, K. D., et al. 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. 27, 619–628 (2017). 28. 28.Spartz, E. J., et al. Course of Neuropsychiatric Symptoms After Introduction and Removal of Nonsteroidal Anti-Inflammatory Drugs: A Pediatric Observational Study. J. Child Adolesc. Psychopharmacol. 27, 652–659 (2017). 29. 29.Perlmutter, S. J., et al. Therapeutic plasma exchange and intravenous immunoglobulin for obsessive-compulsive disorder and tic disorders in childhood. Lancet 354, 1153–1158 (1999). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(98)12297-3&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10513708&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000082954100010&link_type=ISI) 30. 30.Williams, K. A., et al. Randomized, Controlled Trial of Intravenous Immunoglobulin for Pediatric Autoimmune Neuropsychiatric Disorders Associated With Streptococcal Infections. J. Am. Acad. Child Adolesc. Psychiatry 55, 860–867.e2 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jaac.2016.06.017&link_type=DOI) 31. 31.Hart, S. J., Worley, G., Kishnani, P. S. & Van Mater, H. Case Report: Improvement Following Immunotherapy in an Individual With Seronegative Down Syndrome Disintegrative Disorder. Front. Neurol. 12, 621637 (2021). 32. 32.Bey, A. L., et al. Subacute Neuropsychiatric Syndrome in Girls With SHANK3 Mutations Responds to Immunomodulation. Pediatrics 145, e20191490. doi: 10.1542/peds.2019-1490 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1542/peds.2019-1490&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32015180&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) 33. 33.Zheng, J., et al. Association of Pediatric Acute-Onset Neuropsychiatric Syndrome With Microstructural Differences in Brain Regions Detected via Diffusion-Weighted Magnetic Resonance Imaging. *JAMA Netw*. Open 3, e204063 (2020). 34. 34.Malek, M., et al. Prednisolone as Adjunctive Treatment to Risperidone in Children With Regressive Type of Autism Spectrum Disorder: A Randomized, Placebo-Controlled Trial. Clin. Neuropharmacol. 43, 39–45 (2020). 35. 35.Mohebalizadeh, M., et al. Role of Maternal Immune Factors in Neuroimmunology of Brain Development. Mol. Neurobiol. (2023). 36. 36.Croen, L. A., et al. Family history of immune conditions and autism spectrum and developmental disorders: Findings from the study to explore early development. Autism Res. 12, 123–135 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/aur.1979&link_type=DOI) 37. 37.Arenella, M., et al. Immunogenetics of autism spectrum disorder: A systematic literature review. Brain Behav. Immun. 114, 488–499 (2023). 38. 38.Braams, M., Pike-Overzet, K. & Staal, F. J. T. The recombinase activating genes: architects of immune diversity during lymphocyte development. Front. Immunol. 14, 1210818 (2023). 39. 39.Christie, S. M., Fijen, C. & Rothenberg, E. V(D)J Recombination: Recent Insights in Formation of the Recombinase Complex and Recruitment of DNA Repair Machinery. Front. Cell. Dev. Biol. 10, 886718 (2022). 40. 40.Ni, G., Ma, Z. & Damania, B. cGAS and STING: At the intersection of DNA and RNA virus-sensing networks. PLoS Pathog. 14, e1007148 (2018). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.ppat.1007148&link_type=DOI) 41. 41.Chen, X., et al. How mouse RAG recombinase avoids DNA transposition. Nat. Struct. Mol. Biol. 27, 127–133 (2020). 42. 42.Helmink, B. A. & Sleckman, B. P. The response to and repair of RAG-mediated DNA double-strand breaks. Annu. Rev. Immunol. 30, 175–202 (2012). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1146/annurev-immunol-030409-101320&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22224778&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000304198100008&link_type=ISI) 43. 43.Johnston, R., et al. Nuclease-independent functions of RAG1 direct distinct DNA damage responses in B cells. EMBO Rep., e55429 (2022). 44. 44.Chu, S. H., et al. Loss of H3K36 Methyltransferase SETD2 Impairs V(D)J Recombination during Lymphoid Development. iScience 23, 100941 (2020). 45. 45.Rentzsch, P., Witten, D., Cooper, G. M., Shendure, J. & Kircher, M. CADD: predicting the deleteriousness of variants throughout the human genome. Nucleic Acids Res. 47, D886–D894 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/gky1016&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) 46. 46.Niroula, A. & Vihinen, M. How good are pathogenicity predictors in detecting benign variants? PLoS Comput. Biol. 15, e1006481 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pcbi.1006481&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) 47. 47.Kircher, M., et al. A general framework for estimating the relative pathogenicity of human genetic variants. Nat. Genet. 46, 310–315 (2014). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.2892&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24487276&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) 48. 48.Cheng, J., et al. Accurate proteome-wide missense variant effect prediction with AlphaMissense. Science 381, eadg7492 (2023). 49. 49.Ceccaldi, R., Sarangi, P. & D’Andrea, A. D. The Fanconi anaemia pathway: new players and new functions. Nat. Rev. Mol. Cell Biol. 17, 337–349 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nrm.2016.48&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27145721&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) 50. 50.Jones, M. J. K. & Huang, T. T. The Fanconi anemia pathway in replication stress and DNA crosslink repair. Cell Mol. Life Sci. 69, 3963–3974 (2012). 51. 51.Traband, E. L., et al. Mitotic DNA Synthesis in Untransformed Human Cells Preserves Common Fragile Site Stability via a FANCD2-Driven Mechanism That Requires HELQ. J. Mol. Biol. 435, 168294 (2023). 52. 52.Bona, N. & Crossan, G. P. Fanconi anemia DNA crosslink repair factors protect against LINE-1 retrotransposition during mouse development. Nat. Struct. Mol. Biol. (2023). 53. 53.Kupculak, M., et al. Phosphorylation by ATR triggers FANCD2 chromatin loading and activates the Fanconi anemia pathway. Cell. Rep. 42, 112721 (2023). 54. 54.Alcón, P., et al. FANCD2-FANCI is a clamp stabilized on DNA by monoubiquitination of FANCD2 during DNA repair. Nat. Struct. Mol. Biol. 27, 240–248 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41594-020-0380-1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32066963&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) 55. 55.Xu, W., et al. Casein kinase 1α inhibits p53 downstream of MDM2-mediated autophagy and apoptosis in acute myeloid leukemia. Oncol. Rep. 44, 1895–1904 (2020). 56. 56.Liu, X., et al. Tumor-derived CK1α mutations enhance MDMX inhibition of p53. Oncogene 39, 176–186 (2020). 57. 57.Seo, M. H. & Yeo, S. Association Between Decreased Srpk3 Expression and Increased Substantia Nigra Alpha-Synuclein Level in an MPTP-Induced Parkinson’s Disease Mouse Model. Mol. Neurobiol. 60, 780–788 (2023). 58. 58.Akizuki, S., et al. PLD4 is a genetic determinant to systemic lupus erythematosus and involved in murine autoimmune phenotypes. Ann. Rheum. Dis. 78, 509–518 (2019). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6ImFubnJoZXVtZGlzIjtzOjU6InJlc2lkIjtzOjg6Ijc4LzQvNTA5IjtzOjQ6ImF0b20iO3M6NTI6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDIvMjMvMjAyNC4wMi4yMC4yNDMwMjk4NC4xLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 59. 59.Wen, L., et al. Exome-wide association study identifies four novel loci for systemic lupus erythematosus in Han Chinese population. Ann. Rheum. Dis. 77, 417–211823. Epub 2017 Dec 12 (2018). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6ImFubnJoZXVtZGlzIjtzOjU6InJlc2lkIjtzOjg6Ijc3LzMvNDE3IjtzOjQ6ImF0b20iO3M6NTI6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDIvMjMvMjAyNC4wMi4yMC4yNDMwMjk4NC4xLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 60. 60.Kessi, M., et al. Calcium channelopathies and intellectual disability: a systematic review. Orphanet J. Rare Dis. 16, 219– (2021). 61. 61.Viggiano, M., et al. Contribution of CACNA1H Variants in Autism Spectrum Disorder Susceptibility. Front. Psychiatry. 13, 858238 (2022). 62. 62.Woodward, E. R., et al. Germline testing of BRCA1, BRCA2, PALB2 and CHEK2 c.1100delC in 1514 triple negative familial and isolated breast cancers from a single centre, with extended testing of ATM, RAD51C and RAD51D in over 400. J. Med. Genet. (2023). 63. 63.Stubbins, R. J., Korotev, S. & Godley, L. A. Germline CHEK2 and ATM Variants in Myeloid and Other Hematopoietic Malignancies. Curr. Hematol. Malig Rep. 17, 94–104 (2022). 64. 64.Azizi, G., et al. The Autoimmune Manifestations in Patients with Genetic Defects in the B Cell Development and Differentiation Stages. J. Clin. Immunol. 43, 819–834 (2023). 65. 65.Pezone, A., et al. Inflammation and DNA damage: cause, effect or both. Nat. Rev. Rheumatol. 19, 200–211 (2023). 66. 66.Ghosh, D., Kumari, S. & Raghavan, S. C. Depletion of RNASEH2 Activity Leads to Accumulation of DNA Double-strand Breaks and Reduced Cellular Survivability in T Cell Leukemia. J. Mol. Biol. 434, 167617 (2022). 67. 67.Viguera-Elías, D., de la Iglesia-Nagore, I., Toledo-Gotor, C., Domínguez-Garrido, E. & Poch-Olivé, M. L. Aicardi-Goutieres syndrome: a family case due to alteration of the RNASEH2B gene. Rev. Neurol. 72, 407–409 (2021). 68. 68.Walia, J. & Mujahid, R. Clinical Disease States Related to Mutations of the NOD2 Gene: A Case Report and Literature Review. Cureus 15, e38584 (2023). 69. 69.Priya, B., Ravi, S. & Kirubakaran, S. Targeting ATM and ATR for cancer therapeutics: Inhibitors in clinic. Drug Discov. Today 28, 103662 (2023). 70. 70.Maréchal, A. & Zou, L. DNA damage sensing by the ATM and ATR kinases. Cold Spring Harb Perspect. Biol. 5, a012716. doi: 10.1101/cshperspect.a012716 (2013). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6ImNzaHBlcnNwZWN0IjtzOjU6InJlc2lkIjtzOjExOiI1LzkvYTAxMjcxNiI7czo0OiJhdG9tIjtzOjUyOiIvbWVkcnhpdi9lYXJseS8yMDI0LzAyLzIzLzIwMjQuMDIuMjAuMjQzMDI5ODQuMS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 71. 71.Saxena, S. & Zou, L. Hallmarks of DNA replication stress. Mol. Cell 82, 2298–2314 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/J.MOLCEL.2022.05.004&link_type=DOI) 72. 72.Paull, T. T. Mechanisms of ATM Activation. Annu. Rev. Biochem. 84, 711–738 (2015). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1146/annurev-biochem-060614-034335&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25580527&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) 73. 73.Lee, J. & Paull, T. T. Activation and regulation of ATM kinase activity in response to DNA double-strand breaks. Oncogene 26, 7741–7748 (2007). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/sj.onc.1210872&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18066086&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000251537900004&link_type=ISI) 74. 74.Huang, Y., et al. DNAJA2 deficiency activates cGAS-STING pathway via the induction of aberrant mitosis and chromosome instability. Nat. Commun. 14, 5246– (2023). 75. 75.Brown, A. D., et al. ATR suppresses endogenous DNA damage and allows completion of homologous recombination repair. PLoS One 9, e91222 (2014). 76. 76.McIntyre, R. E., et al. Disruption of mouse Cenpj, a regulator of centriole biogenesis, phenocopies Seckel syndrome. PLoS Genet. 8, e1003022 (2012). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pgen.1003022&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23166506&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) 77. 77.Nagai-Singer, M. A., Morrison, H. A. & Allen, I. C. NLRX1 Is a Multifaceted and Enigmatic Regulator of Immune System Function. Front. Immunol. 10, 2419 (2019). 78. 78.Chen, L., Cao, S., Lin, Z., He, S. & Zuo, J. NOD-like receptors in autoimmune diseases. Acta Pharmacol. Sin. 42, 1742–1756 (2021). 79. 79.Liu, M., et al. The regulatory role of NLRX1 in innate immunity and human disease. Cytokine 160, 156055 (2022). 80. 80.Chu, X., Wu, S. & Raju, R. NLRX1 Regulation Following Acute Mitochondrial Injury. Front. Immunol. 10, 2431 (2019). 81. 81.Leriche, M., et al. 53BP1 interacts with the RNA primer from Okazaki fragments to support their processing during unperturbed DNA replication. Cell. Rep. 42, 113412 (2023). 82. 82.Arnould, C., et al. Chromatin compartmentalization regulates the response to DNA damage. Nature 623, 183–192 (2023). 83. 83.Vincendeau, E., et al. SHLD1 is dispensable for 53BP1-dependent V(D)J recombination but critical for productive class switch recombination. Nat. Commun. 13, 3707–3 (2022). 84. 84.Honjoh, H., et al. MED1, a novel binding partner of BRCA1, regulates homologous recombination and R-loop processing. Sci. Rep. 12, 17140–8 (2022). 85. 85.Armstrong, D. L., et al. GWAS identifies novel SLE susceptibility genes and explains the association of the HLA region. Genes Immun. 15, 347–354 (2014). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/gene.2014.23&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24871463&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) 86. 86.Adam, M. P., Hudgins, L. & Hannibal, M. in GeneReviews(®) (ed Adam, M. P. et al.) (University of Washington, Seattle. GeneReviews is a registered trademark of the University of Washington, Seattle. All rights reserved, Seattle (WA), 1993). 87. 87.Dermawan, J. K., et al. Distinct genomic landscapes in radiation-associated angiosarcoma compared with other radiation-associated sarcoma histologies. J. Pathol. 260, 465–477 (2023). 88. 88.Zhurinsky, J., et al. Effects of the microtubule nucleator Mto1 on chromosomal movement, DNA repair, and sister chromatid cohesion in fission yeast. Mol. Biol. Cell 30, 2695–2708 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1091/mbc.E19-05-0301&link_type=DOI) 89. 89.Zhou, C., et al. Clinical and genetic analysis of combined oxidative phosphorylation defificiency-10 caused by MTO1 mutation. Clin. Chim. Acta 526, 74–80 (2022). 90. 90. Mushtaq Hashim Al-Bderee, N., Faaz Nassir Al-Saad, N., Jawad Al-Imari, M., Mudhaher Habbeb, S. & Mizal Azoz, A. Genetic Polymorphisms of NOD2 and ATG16L1 in Different Types of Digestive Tract Inflammation. Arch. Razi Inst. 78, 493–498 (2023). 91. 91.Vohhodina, J., et al. The RNA processing factors THRAP3 and BCLAF1 promote the DNA damage response through selective mRNA splicing and nuclear export. Nucleic Acids Res. 45, 12816–12833 (2017). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/gkx1046&link_type=DOI) 92. 92.Beli, P., et al. Proteomic investigations reveal a role for RNA processing factor THRAP3 in the DNA damage response. Mol. Cell 46, 212–225 (2012). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.molcel.2012.01.026&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22424773&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000303365300011&link_type=ISI) 93. 93.Kumari, N., Antil, H., Kumari, S. & Raghavan, S. C. Deficiency of ligase IV leads to reduced NHEJ, accumulation of DNA damage, and can sensitize cells to cancer therapeutics. Genomics 115, 110731 (2023). 94. 94.Jauch, A. J., et al. Autoimmunity and immunodeficiency associated with monoallelic LIG4 mutations via haploinsufficiency. J. Allergy Clin. Immunol. 152, 500–516 (2023). 95. 95. Yilmaz Demirdag, Y. & Gupta, S. Infections in DNA Repair Defects. Pathogens 12, 440. doi: 10.3390/pathogens12030440 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3390/pathogens12030440&link_type=DOI) 96. 96.Ramirez, N., et al. Multi-omics analysis of naïve B cells of patients harboring the C104R mutation in TACI. Front. Immunol. 13, 938240 (2022). 97. 97.Kakkas, I., et al. TACI Mutations in Primary Antibody Deficiencies: A Nationwide Study in Greece. Medicina (Kaunas*)* 57, 827. doi: 10.3390/medicina57080827 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3390/medicina57080827&link_type=DOI) 98. 98.de Valles-Ibáñez, G., et al. Evaluating the Genetics of Common Variable Immunodeficiency: Monogenetic Model and Beyond. Front. Immunol. 9, 636 (2018). 99. 99.Nomani, H., et al. Implications of combined NOD2 and other gene mutations in autoinflammatory diseases. Front. Immunol. 14, 1265404 (2023). 100.100.Karamanakos, A., et al. Autoinflammatory syndromes with coexisting variants in Mediterranean FeVer and other genes: Utility of multiple gene screening and the possible impact of gene dosage. Semin. Arthritis Rheum. 56, 152055 (2022). 101.101.Wouters, F., Bogie, J., Wullaert, A. & van der Hilst, J. Recent Insights in Pyrin Inflammasome Activation: Identifying Potential Novel Therapeutic Approaches in Pyrin-Associated Autoinflammatory Syndromes. J. Clin. Immunol. 44, 8–5 (2023). 102.102.Nomani, H., et al. Implications of combined NOD2 and other gene mutations in autoinflammatory diseases. Front. Immunol. 14, 1265404 (2023). 103.103.Hao, Q., Lu, H. & Zhou, X. A potential synthetic lethal strategy with PARP inhibitors: perspective on ’Inactivation of the tumor suppressor p53 by long noncoding RNA RMRP’. J. Mol. Cell. Biol. 13, 690–692 (2021). 104.104.Huang, R., et al. The p53/RMRP/miR122 signaling loop promotes epithelial-mesenchymal transition during the development of silica-induced lung fibrosis by activating the notch pathway. Chemosphere 263, 128133 (2021). 105.105.Wolf, C., et al. UNC93B1 variants underlie TLR7-dependent autoimmunity. Sci. Immunol., eadi9769 (2024). 106.106.He, Z., Ye, S., Xing, Y., Jiu, Y. & Zhong, J. UNC93B1 curbs cytosolic DNA signaling by promoting STING degradation. Eur. J. Immunol. 51, 1672–1685 (2021). 107.107.Zhu, H., Zhang, R., Yi, L., Tang, Y. & Zheng, C. UNC93B1 attenuates the cGAS-STING signaling pathway by targeting STING for autophagy-lysosome degradation. J. Med. Virol. 94, 4490–4501 (2022). 108.108.Ciążyńska, M., et al. The Role of NLRP1, NLRP3, and AIM2 Inflammasomes in Psoriasis: Review. *Int. J. Mol. Sci.* **22**, 5898. doi: 10.3390/ijms22115898 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3390/ijms22115898&link_type=DOI) 109.109.Docherty, C. A., et al. A novel dual NLRP1 and NLRP3 inflammasome inhibitor for the treatment of inflammatory diseases. Clin. Transl. Immunology 12, e1455 (2023). 110.110.Miyashita, Y., et al. TICAM-1/TRIF associates with Act1 and suppresses IL-17 receptor-mediated inflammatory responses. Life. Sci. Alliance 5, e202101181. doi: 10.26508/lsa.202101181. Print 2022 Feb (2021). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoibHNhIjtzOjU6InJlc2lkIjtzOjE0OiI1LzIvZTIwMjEwMTE4MSI7czo0OiJhdG9tIjtzOjUyOiIvbWVkcnhpdi9lYXJseS8yMDI0LzAyLzIzLzIwMjQuMDIuMjAuMjQzMDI5ODQuMS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 111.111.Jensen, L. T., Attfield, K. E., Feldmann, M. & Fugger, L. Allosteric TYK2 inhibition: redefining autoimmune disease therapy beyond JAK1-3 inhibitors. EBioMedicine 97, 104840 (2023). 112.112.Sumpter, R. J., et al. Fanconi Anemia Proteins Function in Mitophagy and Immunity. Cell 165, 867–881 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2016.04.006&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27133164&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) 113.113.Xia, M., et al. FANCC deficiency mediates microglial pyroptosis and secondary neuronal apoptosis in spinal cord contusion. Cell. Biosci. 12, 82–4 (2022). 114.114.Cevher, M. A., et al. Nuclear deadenylation/polyadenylation factors regulate 3’ processing in response to DNA damage. EMBO J. 29, 1674–1687 (2010). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiZW1ib2pubCI7czo1OiJyZXNpZCI7czoxMDoiMjkvMTAvMTY3NCI7czo0OiJhdG9tIjtzOjUyOiIvbWVkcnhpdi9lYXJseS8yMDI0LzAyLzIzLzIwMjQuMDIuMjAuMjQzMDI5ODQuMS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 115.115.Gemayel, K. T., Litman, G. W. & Sriaroon, P. Autosomal recessive agammaglobulinemia associated with an IGLL1 gene missense mutation. Ann. Allergy Asthma Immunol. 117, 439–441 (2016). 116.116.Lu, X., et al. MTA2/NuRD Regulates B Cell Development and Cooperates with OCA-B in Controlling the Pre-B to Immature B Cell Transition. Cell. Rep. 28, 472–485.e5 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.celrep.2019.06.029&link_type=DOI) 117.117.Gevezova, M., et al. Autistic spectrum disorder (ASD) - Gene, molecular and pathway signatures linking systemic inflammation, mitochondrial dysfunction, transsynaptic signalling, and neurodevelopment. Brain Behav. Immun. Health. 30, 100646 (2023). 118.118.Pollina, E. A., et al. A NPAS4-NuA4 complex couples synaptic activity to DNA repair. Nature 614, 732–741 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-023-05711-7&link_type=DOI) 119.119.Zhou, X., et al. Integrating de novo and inherited variants in 42,607 autism cases identifies mutations in new moderate-risk genes. Nat. Genet. 54, 1305–1319 (2022). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=35982159&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) 120.120.Wilfert, A. B., et al. Recent ultra-rare inherited variants implicate new autism candidate risk genes. Nat. Genet. 53, 1125–1134 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41588-021-00899-8&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=34312540&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) 121.121.Fu, J. M., et al. Rare coding variation provides insight into the genetic architecture and phenotypic context of autism. Nat. Genet. 54, 1320–1331 (2022). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=35982160&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) 122.122.Mueller, F. S., et al. Deficient DNA base-excision repair in the forebrain leads to a sex- specific anxiety-like phenotype in mice. BMC Biol. 20, 170–1 (2022). 123.123.Shen, X., et al. The role of Gadd45b in neurologic and neuropsychiatric disorders: An overview. Front. Mol. Neurosci. 15, 1021207 (2022). 124.124.Dileep, V., et al. Neuronal DNA double-strand breaks lead to genome structural variations and 3D genome disruption in neurodegeneration. Cell 186, 4404–4421.e20 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2023.08.038&link_type=DOI) 125.125.Yang, C., et al. Expression signature of inflammation-associated long non-coding RNAs in adult-onset Still’s disease. Clin. Exp. Rheumatol. 39 **Suppl 132**, 67–74 (2021). 126.126.Jaber, S., Toufektchan, E., Lejour, V., Bardot, B. & Toledo, F. p53 downregulates the Fanconi anaemia DNA repair pathway. Nat. Commun. 7, 11091 (2016). 127.127.Rakotopare, J. & Toledo, F. p53 in the Molecular Circuitry of Bone Marrow Failure Syndromes. Int. J. Mol. Sci. 24, 14940. doi: 10.3390/ijms241914940 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3390/ijms241914940&link_type=DOI) 128.128.Parvin, S., et al. ATM depletion induces proteasomal degradation of FANCD2 and sensitizes neuroblastoma cells to PARP inhibitors. BMC Cancer 23, 313-y (2023). 129.129.Ishiai, M. Regulation of the Fanconi Anemia DNA Repair Pathway by Phosphorylation and Monoubiquitination. Genes (Basel*)* 12, 1763. doi: 10.3390/genes12111763 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3390/genes12111763&link_type=DOI) 130.130.Tammimies, K. Genetic mechanisms of regression in autism spectrum disorder. Neurosci. Biobehav. Rev. 102, 208–220 (2019). 131.131.Golla, S. & Sweeney, J. A. Corticosteroid therapy in regressive autism: Preliminary findings from a retrospective study. BMC Med. 12, 79–79 (2014). 132.132.Rossignol, D. A. & Frye, R. E. A Systematic Review and Meta-Analysis of Immunoglobulin G Abnormalities and the Therapeutic Use of Intravenous Immunoglobulins (IVIG) in Autism Spectrum Disorder. J. Pers. Med. 11, 488. doi: 10.3390/jpm11060488 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3390/jpm11060488&link_type=DOI) 133.133.Hopfner, K. & Hornung, V. Molecular mechanisms and cellular functions of cGAS-STING signalling. Nat. Rev. Mol. Cell Biol. 21, 501–521 (2020). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) 134.134.Phelan, T., Little, M. A. & Brady, G. Targeting of the cGAS-STING system by DNA viruses. Biochem. Pharmacol. 174, 113831 (2020). 135.135.Fan, Y. M., Zhang, Y. L., Luo, H. & Mohamud, Y. Crosstalk between RNA viruses and DNA sensors: Role of the cGAS-STING signalling pathway. Rev. Med. Virol. 32, e2343 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/rmv.2343&link_type=DOI) 136.136.Wang, C., et al. Ultrasound-responsive low-dose doxorubicin liposomes trigger mitochondrial DNA release and activate cGAS-STING-mediated antitumour immunity. Nat. Commun. 14, 3877-x (2023). 137.137.Kim, J., Kim, H. & Chung, J. H. Molecular mechanisms of mitochondrial DNA release and activation of the cGAS-STING pathway. Exp. Mol. Med. 55, 510–519 (2023). 138.138.West, A. P., et al. Mitochondrial DNA stress primes the antiviral innate immune response. Nature 520, 553–557 (2015). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature14156&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25642965&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) 139.139.Lammert, C. R., et al. AIM2 inflammasome surveillance of DNA damage shapes neurodevelopment. Nature 580, 647–652 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-020-2174-3&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32350463&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) 140.140.Dawson, R. E., et al. The cytosolic DNA sensor AIM2 promotes Helicobacter-induced gastric pathology via the inflammasome. Immunol. Cell Biol. 101, 444–457 (2023). 141.141.Vinuesa, C. G., Grenov, A. & Kassiotis, G. Innate virus-sensing pathways in B cell systemic autoimmunity. Science 380, 478–484 (2023). 142.142.Hu, Y., et al. Emerging role of the cGAS-STING signaling pathway in autoimmune diseases: Biologic function, mechanisms and clinical prospection. Autoimmun. Rev. 21, 103155 (2022). 143.143.Kivanc, D. & Dasdemir, S. The relationship between defects in DNA repair genes and autoinflammatory diseases. Rheumatol. Int. 42, 1–13 (2022). 144.144.Miner, J. J. & Fitzgerald, K. A. A path towards personalized medicine for autoinflammatory and related diseases. Nat. Rev. Rheumatol. 19, 182–189 (2023). 145.145.Li, R., et al. cGAS/STING signaling in the regulation of rheumatoid synovial aggression. Ann. Transl. Med. 10, 431–4533 (2022). 146.146.Hagiwara, A. M., Moore, R. E., Wallace, D. J., Ishimori, M. & Jefferies, C. A. Regulation of cGAS-STING Pathway - Implications for Systemic Lupus Erythematosus. *Rheumatol*. Immunol. Res. 2, 173–184 (2021). 147.147.Wobma, H., Shin, D. S., Chou, J. & Dedeoğlu, F. Dysregulation of the cGAS-STING Pathway in Monogenic Autoinflammation and Lupus. Front. Immunol. 13, 905109 (2022). 148.148.Iourov, I. Y., et al. Causes and Consequences of Genome Instability in Psychiatric and Neurodegenerative Diseases. Mol. Biol. (Mosk*)* 55, 42–53 (2021). 149.149.Loncharich, M. F. & Anderson, C. W. Interferon Inhibition for Lupus with Anifrolumab: Critical Appraisal of the Evidence Leading to FDA Approval. ACR Open Rheumatol. 4, 486–491 (2022). 150.150.Weckerle, C. E., et al. Network analysis of associations between serum interferon-α activity, autoantibodies, and clinical features in systemic lupus erythematosus. Arthritis Rheum. 63, 1044–1053 (2011). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/art.30187&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21162028&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F23%2F2024.02.20.24302984.1.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000289421100023&link_type=ISI) 151.151.Nikolaidou, A., Beis, I., Dragoumi, P. & Zafeiriou, D. Neuropsychiatric manifestations associated with Juvenile Systemic Lupus Erythematosus: An overview focusing on early diagnosis. Brain Dev. (2023). 152.152.Van der Heijden, H., et al. Implications of Inflammatory Processes on a Developing Central Nervous System in Childhood-onset Systemic Lupus Erythematosus. Arthritis Rheumatol. (2023). 153.153.Ferecskó, A. S., et al. STING-Triggered CNS Inflammation in Human Neurodegenerative Diseases. Biomedicines 11, 1375. doi: 10.3390/biomedicines11051375 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3390/biomedicines11051375&link_type=DOI) 154.154.Gulen, M. F., et al. cGAS-STING drives ageing-related inflammation and neurodegeneration. Nature 620, 374–380 (2023). 155.155.Chen, K., et al. cGAS-STING-mediated IFN-I Response in Host Defense and Neuroinflammatory Diseases. Curr. Neuropharmacol. 20, 362–371 (2022). 156.156.Beesetti, S., et al. FANCL supports Parkin-mediated mitophagy in a ubiquitin ligase- independent manner. Biochim. Biophys. Acta Mol. Basis Dis. 1868, 166453 (2022). 157.157.Luzwick, J. W., et al. MRE11-dependent instability in mitochondrial DNA fork protection activates a cGAS immune signaling pathway. Sci. Adv. 7, eabf9441 (2021). 158.158.Habibi, P., et al. Effect of heat stress on DNA damage: a systematic literature review. Int. J. Biometeorol. 66, 2147–2158 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00484-022-02351-w&link_type=DOI) 159.159.Couillin, I. & Riteau, N. STING Signaling and Sterile Inflammation. Front. Immunol. 12, 753789 (2021). 160.160.Yang, J., et al. STING in tumors: a focus on non-innate immune pathways. Front. Cell. Dev. Biol. 11, 1278461 (2023). 161.161.He, X., et al. IUPHAR ECR review: The cGAS-STING pathway: novel functions beyond innate immune and emerging therapeutic opportunities. Pharmacol. Res., 107063 (2024). 162.162.Xun, J., et al. A conserved ion channel function of STING mediates noncanonical autophagy and cell death. EMBO Rep. (2024).