PT - JOURNAL ARTICLE AU - Bassani, Sissy AU - Chrast, Jacqueline AU - Ambrosini, Giovanna AU - Voisin, Norine AU - Schütz, Frédéric AU - Brusco, Alfredo AU - Sirchia, Fabio AU - Turban, Lydia AU - Schubert, Susanna AU - Jamra, Rami Abou AU - Schlump, Jan-Ulrich AU - DeMille, Desiree AU - Bayrak-Toydemir, Pinar AU - Nelson, Gary Rex AU - Wong, Kristen Nicole AU - Duncan, Laura AU - Mosera, Mackenzie AU - Gilissen, Christian AU - Vissers, Lisenka E.L.M. AU - Pfundt, Rolph AU - Kersseboom, Rogier AU - Yttervik, Hilde AU - Hansen, Geir Åsmund Myge AU - Smeland, Marie Falkenberg AU - Butler, Kameryn M. AU - Lyons, Michael J. AU - Carvalho, Claudia M.B. AU - Zhang, Chaofan AU - Lupski, James R. AU - Potocki, Lorraine AU - Flores-Gallegos, Leticia AU - Morales-Toquero, Rodrigo AU - Petit, Florence AU - Yalcin, Binnaz AU - Tuttle, Annabelle AU - Elloumi, Houda Zghal AU - Mccormick, Lane AU - Kukolich, Mary AU - Klaas, Oliver AU - Horvath, Judit AU - Scala, Marcello AU - Iacomino, Michele AU - Operto, Francesca AU - Zara, Federico AU - Writzl, Karin AU - Maver, Ales AU - Haanpää, Maria K. AU - Pohjola, Pia AU - Arikka, Harri AU - Iseli, Christian AU - Guex, Nicolas AU - Reymond, Alexandre TI - Variant-specific pathophysiological mechanisms of <em>AFF3</em> differently influence transcriptome profiles AID - 10.1101/2024.01.14.24301100 DP - 2024 Jan 01 TA - medRxiv PG - 2024.01.14.24301100 4099 - http://medrxiv.org/content/early/2024/01/17/2024.01.14.24301100.short 4100 - http://medrxiv.org/content/early/2024/01/17/2024.01.14.24301100.full AB - Background We previously described the KINSSHIP syndrome, an autosomal dominant disorder associated with intellectual disability (ID), mesomelic dysplasia and horseshoe kidney,caused by de novo variants in the degron of AFF3. Mouse knock-ins and overexpression in zebrafish provided evidence for a dominant-negative (DN) mode-of-action, wherein an increased level of AFF3 resulted in pathological effects.Methods Evolutionary constraints suggest that other mode-of-inheritance could be at play. We challenged this hypothesis by screening ID cohorts for individuals with predicted-to-be deleterious variants in AFF3. We used both animal and cellular models to assess the deleteriousness of the identified variants.Results We identified an individual with a KINSSHIP-like phenotype carrying a de novo partial duplication of AFF3 further strengthening the hypothesis that an increased level of AFF3 is pathological. We also detected seventeen individuals displaying a milder syndrome with either heterozygous LoF or biallelic missense variants in AFF3. Consistent with semi-dominance, we discovered three patients with homozygous LoF and one compound heterozygote for a LoF and a missense variant, who presented more severe phenotypes than their heterozygous parents. Matching zebrafish knockdowns exhibit neurological defects that could be rescued by expressing human AFF3 mRNA, confirming their association with the ablation of aff3. Conversely, some of the human AFF3 mRNAs carrying missense variants identified in affected individuals did not complement. Overexpression of mutated AFF3 mRNAs in zebrafish embryos produced a significant increase of abnormal larvae compared to wild-type overexpression further demonstrating deleteriousness. To further assess the effect of AFF3 variation, we profiled the transcriptome of fibroblasts from affected individuals and engineered isogenic cells harboring +/+, DN/DN, LoF/+, LoF/LoF or DN/LoF AFF3 genotypes. The expression of more than a third of the AFF3 bound loci is modified in either the DN/DN or the LoF/LoF lines. While the same pathways are affected, only about one-third of the differentially expressed genes are common to these homozygote datasets, indicating that AFF3 LoF and DN variants largely modulate transcriptomes differently, e.g. the DNA repair pathway displayed opposite modulation.Conclusions Our results and the high pleiotropy shown by variation at this locus suggest that minute changes in AFF3 function are deleterious.Competing Interest StatementAnnabelle Tuttle, Houda Zghal Elloumi and Chaofan Zhang are employees of GeneDx and Desiree DeMille works for ARUP Laboratories. James R. Lupski has stock ownership in 23andMe and is a paid consultant for Genome International. Claudia M.B. Carvalho provides consulting services for Ionis Pharmaceuticals. The other authors have no competing interests to declare.Funding StatementThis work was supported by grants from the Swiss National Science Foundation (31003A_182632 and IZSTZ0_216615 to AR), the Lejeune Foundation (#1838-2019A to AR), the Blackswan Foundation (to AR), a PRIN 2020 grant from the Italian Ministry of Universities and Research (20203P8C3X to AB), and the US National Institutes of Health (NS105078 and HG011758 to JRL). This study makes use of data generated by the DECIPHER community. Funding for the DECIPHER project was provided by the Wellcome Trust [grant number WT223718/Z/21/Z]. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.Author DeclarationsI confirm all relevant ethical guidelines have been followed, and any necessary IRB and/or ethics committee approvals have been obtained.YesThe details of the IRB/oversight body that provided approval or exemption for the research described are given below:Informed consent forms were obtained for all affected individuals or their guardians participating in this study. The current study was approved by the CER (Commission d'éthique de la recherche) of the canton of Vaud (Protocol number: CER-VD 2021-01400). This research complies with the principles of the Declaration of Helsinki.I confirm that all necessary patient/participant consent has been obtained and the appropriate institutional forms have been archived, and that any patient/participant/sample identifiers included were not known to anyone (e.g., hospital staff, patients or participants themselves) outside the research group so cannot be used to identify individuals.YesI understand that all clinical trials and any other prospective interventional studies must be registered with an ICMJE-approved registry, such as ClinicalTrials.gov. I confirm that any such study reported in the manuscript has been registered and the trial registration ID is provided (note: if posting a prospective study registered retrospectively, please provide a statement in the trial ID field explaining why the study was not registered in advance).YesI have followed all appropriate research reporting guidelines, such as any relevant EQUATOR Network research reporting checklist(s) and other pertinent material, if applicable.YesData produced in the present study are available upon reasonable request to the authors. The HEK293T and fibroblast RNA-seq reads are deposited in GEO under accession GSE241621 (token otezqcsslbctbyx) and GSE246554 (token wxibcyqatdyphkl), and will be available from August 2024.IDintellectual disabilitiesDDdevelopmental delayDNdominant-negativeLoFloss-of-function+wild-type alleleNHDN-terminal homology domainCHDC-terminal homology domainsALFAF4-LAF4-FMR2 domainTADtransactivation domainNLSnuclear/nucleolar localization sequenceSECstranscriptional super elongation complexesP-TEFpositive transcription elongation factorMLLTmyeloid/lymphoid or mixed-lineage leukemia; translocated toELLElongation Factor for RNA Polymerase IIELLbowELL-binding domainpLIprobability of being loss-of-function intolerantpLOEUFloss-of-function observed/expected upper bound fractionPTU1-phenyl 2-thioureaK22-K26KINSSHIP patients 22 to 26DUPpatient with partial duplication of AFF3L1-L14patients 1 to 14 with loss-of-function variantsB1-B7patients 1 to 7 with biallelic missense variantsADHDattention deficit hyperactivity disorderEEGElectroencephalographyASDautism spectrum disorderKDknock-downDpfdays post fertilizationIODinter-ocular distanceHWhead widthUnUntreatedMmockWtwild-typeMAFminor allele frequencyDEGdifferentially expressed genesMTAG-associatedassociated by multi-trait analysis of GWASBMIbody mass index