You are on page 1of 12

REPORT

De Novo Variants in the F-Box Protein FBXO11


in 20 Individuals with a Variable
Neurodevelopmental Disorder
Anne Gregor,1 Lynette G. Sadleir,2 Reza Asadollahi,3,4 Silvia Azzarello-Burri,3,4 Agatino Battaglia,5
Lilian Bomme Ousager,6 Paranchai Boonsawat,3,4 Ange-Line Bruel,7 Rebecca Buchert,8
Eduardo Calpena,9 Benjamin Cogné,10,11 Bruno Dallapiccola,12 Felix Distelmaier,13 Frances Elmslie,14
Laurence Faivre,7,15 Tobias B. Haack,8 Victoria Harrison,16 Alex Henderson,17 David Hunt,16
Bertrand Isidor,10 Pascal Joset,3,4 Satoko Kumada,18 Augusta M.A. Lachmeijer,19 Melissa Lees,20
Sally Ann Lynch,21 Francisco Martinez,22 Naomichi Matsumoto,23 Carey McDougall,24
Heather C. Mefford,25 Noriko Miyake,23 Candace T. Myers,25 Sébastien Moutton,7,15 Addie Nesbitt,26
Antonio Novelli,12 Carmen Orellana,22 Anita Rauch,3,4 Monica Rosello,22 Ken Saida,23
Avni B. Santani,26,27 Ajoy Sarkar,28 Ingrid E. Scheffer,29 Marwan Shinawi,30 Katharina Steindl,3,4
Joseph D. Symonds,31 Elaine H. Zackai,24 University of Washington Center for Mendelian Genomics,
DDD Study,32 André Reis,1 Heinrich Sticht,33 and Christiane Zweier1,*

Next-generation sequencing combined with international data sharing has enormously facilitated identification of new disease-associ-
ated genes and mutations. This is particularly true for genetically extremely heterogeneous entities such as neurodevelopmental disor-
ders (NDDs). Through exome sequencing and world-wide collaborations, we identified and assembled 20 individuals with de novo
variants in FBXO11. They present with mild to severe developmental delay associated with a range of features including short (4/20)
or tall (2/20) stature, obesity (5/20), microcephaly (4/19) or macrocephaly (2/19), behavioral problems (17/20), seizures (5/20), cleft
lip or palate or bifid uvula (3/20), and minor skeletal anomalies. FBXO11 encodes a member of the F-Box protein family, constituting
a subunit of an E3-ubiquitin ligase complex. This complex is involved in ubiquitination and proteasomal degradation and thus in con-
trolling critical biological processes by regulating protein turnover. The identified de novo aberrations comprise two large deletions, ten
likely gene disrupting variants, and eight missense variants distributed throughout FBXO11. Structural modeling for missense variants
located in the CASH or the Zinc-finger UBR domains suggests destabilization of the protein. This, in combination with the observed
spectrum and localization of identified variants and the lack of apparent genotype-phenotype correlations, is compatible with loss of
function or haploinsufficiency as an underlying mechanism. We implicate de novo missense and likely gene disrupting variants in
FBXO11 in a neurodevelopmental disorder with variable intellectual disability and various other features.

Neurodevelopmental disorders (NDDs), including intellec- and genetically extremely heterogeneous. Currently, path-
tual disability (ID), autism spectrum disorders, and devel- ogenic variants in more than 1,000 genes have been linked
opmental and epileptic encephalopathies, are clinically to NDDs (SysID database).1 In non-consanguineous
1
Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; 2Department of Paediatrics and Child
Health, University of Otago, Wellington 6242, New Zealand; 3Institute of Medical Genetics, University of Zurich, 8952 Schlieren-Zurich, Switzerland;
4
radiz – ‘‘Rare Disease Initiative Zurich, Clinical Research Priority Program for Rare Diseases University of Zurich,’’ 8032 Zurich, Switzerland; 5Stella Maris
Clinical Research Institute for Child and Adolescent Neurology and Psychiatry, 56128 Calambrone, Pisa, Italy; 6Department of Clinical Genetics, Odense
University Hospital, 5000 Odense, Denmark; 7INSERM U1231, LNC UMR1231 GAD, Burgundy University, 21079 Dijon, France; 8Institute of Medical
Genetics and Applied Genomics, University of Tübingen, 72076 Tübingen, Germany; 9Clinical Genetics Group, MRC Weatherall Institute of Molecular
Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK; 10Department of Medical Genetics, CHU Nantes, 44093 Nantes, France;
11
l’Institut du Thorax, INSERM, CNRS, UNIV Nantes, 44007 Nantes, France; 12Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino
Gesù, IRCCS, Rome 00146, Italy; 13Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children’s Hospital Düsseldorf,
Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany; 14South West Thames Regional Genetics Service, St. George’s, University of
London, London SW17 0RE, UK; 15Reference Center for Developmental Anomalies, Department of Medical Genetics, Dijon University Hospital, 21000
Dijon, France; 16Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton SO16 5YA, UK; 17Northern Genetics Service, Newcastle upon
Tyne Hospitals, Newcastle upon Tyne NE1 3BZ, UK; 18Department of Neuropediatrics, Tokyo Metropolitan Neurological Hospital, Tokyo 183-0042, Japan;
19
Department of Genetics, University Medical Center Utrecht, PO Box 85090, 3508 AB Utrecht, the Netherlands; 20North East Thames Regional Genetics
Service, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street Hospital, London WC1N 3JH, UK; 21Dept of Clinical Ge-
netics, Temple Street Children’s Hospital Dublin 1, D12 V004 Dublin, Ireland; 22Unidad de Genética, Hospital Universitario y Politécnico La Fe, Avda Fer-
nando Abril Martorell 106, 46026 Valencia, Spain; 23Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama
236-0004, Japan; 24Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA; 25Division of Genetic Medicine,
Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; 26Division of Genomic Diagnostics, Children’s Hospital of Philadelphia, Phil-
adelphia, PA 19104, USA; 27Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
19104, USA; 28Nottingham Regional Genetics Service, City Hospital Campus, Nottingham University Hospitals NHS Trust, The Gables, Hucknall Road,
Nottingham NG5 1PB, UK; 29Departments of Medicine and Paediatrics, Austin Health and Royal Children’s Hospital, The University of Melbourne, Park-
ville, VIC 3050, Australia; 30Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis,
MO 63110, USA; 31Paediatric Neurosciences Research Group, Fraser of Allander Neurosciences Unit, Royal Hospital for Children, Glasgow G51 4TF, UK;
32
Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK; 33Institute of Biochemistry, Emil-Fischer Center,
Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
*Correspondence: christiane.zweier@uk-erlangen.de
https://doi.org/10.1016/j.ajhg.2018.07.003.
Ó 2018 American Society of Human Genetics.

The American Journal of Human Genetics 103, 305–316, August 2, 2018 305
from the parents or legal guardians of the affected individ-
Table 1. Summary of Clinical Details
uals. In two individuals, chromosomal microarray analysis
Missense Del/LGD Total
(n ¼ 8) (n ¼ 12) (n ¼ 20) was performed. In 13 individuals, trio exome sequencing
with filtering for de novo variants was performed, and in
Developmental delay/ID mild DD, mild DD, 100%
profound ID severe ID four individuals, affected-only exome sequencing with
filtering against lists including confirmed and candidate
Walking age 12 mo–5 y 13 mo–6 y 12 mo–6 y
genes such as SysID1 was carried out. Panel sequencing
Age first words delay–no delay–no delay–no in individual 18 has been described previously6 (for details
speech speech speech
see Table S1). De novo occurrence of the FBXO11 variant
Short stature 3 1 20% was shown for all case subjects by trio exome and/or
Tall stature 1 1 10% Sanger sequencing (see also Table S1). Clinical information
Obesity 1 4 25%
is summarized in Tables 1 and 2. All identified individuals
had developmental delay manifested by delayed motor
Microcephaly 1 4 25%
and/or speech milestones and a variable degree of ID
Macrocephaly 2 0 10% ranging from normal IQ with difficulties in specific areas
Seizures 4 (þ1) 1 25% to severe or profound ID. Individuals of school age were
attending either special or mainstream schools with addi-
Behavioral anomalies 7 10 85%
tional support. Behavioral abnormalities including autistic
Sleeping difficulties 5 4 45% features and attention deficits as well as sleeping diffi-
Cleft lip/palate/ bifid uvula 2 1 15% culties were reported in most of the individuals. Addition-
Skeletal/hand/feet 5 7 60%
ally, variable aspects such as growth and vision anomalies,
anomalies hypotonia, and skeletal anomalies occurred. Epilepsy in at
Vision anomalies 5 6 55%
least five individuals ranged from developmental and
epileptic encephalopathy in individual 7 to a single seizure
Recurrent infections/otitis 2 4 30%
media
in individual 15. Facial dysmorphisms were frequently
noted. Though certain individuals resembled each other
Abbreviations: mo, months; y, years
(e.g., individuals I3, I9, and I13) and aspects such as
long or downslanting palpebral fissures with laterally
everted lids or long eyelashes were frequently observed,
populations, de novo mutations have been highlighted as overall no distinctive facial gestalt could be delineated
an important contributor.2–4 World-wide data sharing ap- (Figure 1A). De novo FBXO11 variants are therefore associ-
proaches facilitated by various platforms have further ated with a variable neurodevelopmental disorder and a
increased the rate of novel disease-gene discovery and range of additional anomalies but without a distinct,
the delineation of associated phenotypes. recognizable presentation.
By trio exome sequencing on an Illumina HiSeq 2500 FBXO11 encodes a 927 amino acid F-Box protein which
platform and data analysis with an in-house analysis pipe- is highly conserved in evolution, reflected in a 100%
line as described previously,5 we identified a de novo amino acid identity with Pan troglodytes, 99.5% with
missense variant in FBXO11 (MIM: 607871) in a non-ver- mice, and 60% with C. elegans. According to GTEX, it is
bal individual with severe ID and short stature. So far, broadly expressed across various tissues with particularly
only two variants in FBXO11 have been reported as associ- high levels in the brain. FBXO11 contains an F-Box
ated with NDDs, a likely gene-disrupting (LGD) de novo domain, three carbohydrate binding and hydrolase
variant (c.2738_2739delAT [p.Tyr913*]) in an individual (CASH) domains, and a zinc finger-UBR domain (Fig-
with developmental delay, microcephaly, and other fea- ure 1B). The F-Box motif is shared by more than 60 human
tures6 and a homozygous missense variant (c.2596G>A F-Box proteins, with FBXL denoting proteins additionally
[p.Val866Met]) in another individual with severe ID and containing leucine-rich repeats, FBXW denoting proteins
muscular hypotonia.7 Searching the DECIPHER Data- with additional WD repeats, and FBXO denoting F-Box
base,3,8 using GeneMatcher9 and personal communication proteins with either another or no other motif.11 Several
with colleagues enabled us to assemble clinical and muta- other genes encoding F-Box proteins have been implicated
tional details on a total of 20 individuals with de novo var- in NDDs so far (Figure S1). Recessive mutations in FBXL4
iants in FBXO11, including the previously published (MIM: 605654) cause an encephalomyopathic, mitochon-
variant in individual 18.6 Testing in collaborating centers drial DNA depletion syndrome (MIM: 615471), a homozy-
was performed either in the setting of routine diagnostic gous LGD variant in FBXO31 (MIM: 609102) was identified
testing without the requirement for institutional ethics in one family with non-syndromic mild to moderate ID
approval or within research settings approved by the (MIM: 615979),12 and a homozygous missense variant in
ethical review boards of the respective institutions (e.g., FBXO47 (MIM: 609498) has been found in four affected
University Erlangen-Nuremberg, Yokohama City Univer- members of one family with non-syndromic ID.13 The
sity School of Medicine). Informed consent was obtained only dominant variant in another F-box gene reported so

306 The American Journal of Human Genetics 103, 305–316, August 2, 2018
far is a de novo missense variant in FBXO10 (MIM: 609092) bic interactions with Val551 (Figure 2B). These interactions
identified in a single individual with autism-spectrum dis- cannot be formed by the shorter sidechain in the
order and an IQ of 67.14 Interestingly, FBXO10 is the pro- p.Ile538Val variant (Figure 2C), thereby probably destabi-
tein most closely related to FBXO11, and the reported de lizing the first CASH domain. The Thr623 sidechain points
novo FBXO10 variant is in close proximity to the F-Box to the outside of the b-helical structure and forms polar in-
(Figure S1), similarly positioned to the missense FBXO11 teractions with the sidechains of Tyr598 and His600
variants in individuals I2 and I3. (Figure 2D). Due to the different length, the Arg623 side-
We assembled two whole or partial gene deletions, eight chain can form only a weak interaction with Tyr598,
missense variants, and ten LGD variants. One of the whereas electrostatic repulsion emerges between the two
missense variants in I3 was detected in a possible 60% positively charged residues Arg623 and His600 (Figure 2E),
mosaic. All identified LGD variants are distributed which destabilizes the second CASH domain.
throughout the gene/protein, with only one of them The missense variants p.Ser840Pro and p.Ala892Asp are
located within a functional domain (Figure 1B). They located in the UBR-domain, which is characteristic for E3
comprise nonsense, splice site, and frameshift variants ubiquitin ligases and binds directly to N-terminal degrada-
and most are likely to lead to nonsense-mediated mRNA tion signals in substrate proteins.22 The Ser840 stabilizes
decay, except for the variants in individuals 16, 17, and the N terminus of an a helix. Its sidechain interacts with
18,6 which are located in the last exon of FBXO11 and the backbone amide groups of residues 839 and 840 via a
thus potentially escape nonsense-mediated mRNA decay. bridging water molecule (Figure 2G). This bridging is no
In combination with observations of larger de novo dele- longer possible in the p.Ser840Pro variant, because proline
tions of FBXO11 in individuals with a phenotype of devel- lacks both the sidechain hydroxyl group and the backbone
opmental delay or intellectual disability and variable amide hydrogen present in the wild-type serine. In addi-
additional features, our findings are compatible with a tion, the cyclic proline sidechain results in steric clashes
loss-of-function mechanism causing haploinsufficiency, with the bridging water, thus leading to an entire
though other mechanism such as gain-of-function or disruption of this helix-stabilizing structural element
dominant-negative effects are possible, particularly for (Figure 2G). Ala892 forms hydrophobic interactions with
missense variants. This is in accordance with constraint Phe887 (Figure 2H), which stabilizes the orientation of
scores from ExAC,15 which indicate that FBXO11 is a the connecting loop that contains two cysteines (Cys888,
gene highly intolerant to loss-of-function variants with a Cys890) coordinating a zinc ion. The p.Ala892Asp ex-
pLI score of 1. With a z-score of 4.02 it is also intolerant change places the negatively charged aspartate sidechain
to missense variation. The identified missense variants do in the immediate vicinity of the hydrophobic Phe887
not cluster within a particular domain or region of sidechain (Figure 2I). This unfavorable interaction is
FBXO11 but are also distributed across the protein (Fig- expected to cause a conformational rearrangement of
ure 1B). Interpretation regarding pathogenicity is more the entire sequence stretch, which will also affect the cys-
challenging than for LGD variants. All missense variants teines required for zinc coordination, consequently desta-
affect highly conserved amino acids (Figure 2A), are not bilizing the domain structure. Variants p.Pro905Arg and
contained in GnomAD, and are predicted to be deleterious p.Asp910Gly are located downstream of the zf-UBR
by at least three in silico prediction programs (Table S1). To domain (aa 833–904), thereby preventing structural
further assess possible consequences of the missense vari- modeling. Interestingly, this C-terminal region seems to
ants on protein stability and function, we performed be a hotspot for variants with two LGD and three missense
structural modeling. Variants were modeled with Swiss- variants.
Model,23 and RasMol24 was used for structure analysis Two missense variants, p.Arg138Ser and p.Gln156Arg,
and visualization. Missense variants for which structural are located upstream or within the F-Box like domain
information was available are likely to lead to a destabiliza- (aa 153–201), respectively. F-Boxes are interaction sites
tion of the protein, which supports loss of function as the for SKP1 (MIM: 601434), and both proteins together
most likely disease mechanism. form the substrate specificity module of the E3 ubiquitin
The missense variants p.Ile538Val and p.Thr623Arg are ligase. SKP1 then interacts with CUL1 (MIM: 603134).28
located in the first and second CASH domain of FBXO11, No structural information was available for this region.
respectively. CASH domains are frequently found in carbo- One might speculate that mutations in the F-Box domain
hydrate binding proteins and hydrolases and are impor- could impair interaction with SKP1 and therefore render
tant for substrate recognition in F-Box proteins.25,26 Dele- this E3 ubiquitin ligase complex non-functional.
tion of the CASH domain in FBXO11 leads to impaired Seizures occurred more frequently in individuals with
interaction with proto-oncogene BCL6 (MIM: 109565). missense variants, and microcephaly and obesity occurred
Therefore, BCL6 is aberrantly stabilized in cells lacking more frequently in individuals with LGD variants (Table 1).
FBXO11 and in cells with FBXO11 CASH domain muta- Apart from that, the phenotype of individuals with
tions.25 CASH domains exhibit an elongated structure missense variants is not apparently distinct from that
that consists of right-handed b helices.27 Ile538 points to associated with LGD variants. This is in accordance with
the interior of this b-helical structure and forms hydropho- loss of function as the most likely mechanism. However,

The American Journal of Human Genetics 103, 305–316, August 2, 2018 307
Table 2. Clinical Details of Individuals with De Novo FBXO11 Variants
Individual 1 2 3 4 5 6 7 8 9 10

Gender male female male male female female male male male female

Age at last 9 y 3 mo 8y 7 y 5 mo 8y 29 y 10 y 10 mo 15 y 7 y 6 mo 3 y 10 mo 5y
investigation

De novo c.1612A>G c.414A>T c.467A>G c.1868C>G c.2518T>C c.2675C>A c.2714C>G c.2729A>G c.319_320del c.506_507del
variant (p.Ile538Val) (p.Arg138Ser) (p.Gln156Arg); (p.Thr623Arg) (p.Ser840Pro) (p.Ala892Asp) (p.Pro905Arg) (p.Asp910Gly) (p.Leu107Alafs* (p.Ser169Leufs*9)
mosaic (60%) 45)

B. weight (SD) 3,230 g (0.9) 4,900 g (3.4) 4,000 g (0.9) 2,850 g (1.8) 3,175 g (0.71) 3,495 g (0) normal NA 2,580 g (2.4) 3,624 g (0.4)

B. length (SD) 51 cm (0.7) NA normal 52 cm (0.2) NA 51 cm (0.3) normal NA 45 cm (3.3) 51 cm (0.3)

B. OFC (SD) 36 cm (0.3) small normal 35 cm (0.5) NA 35 cm (0.1) normal NA 33 cm (2) 34 cm (0.7)

Height (SD) 122 cm (2.7) 136.8 cm (1.3) (0.3) 107 cm (4.6) 174 cm (1.29) 133 cm (2) (>2) (tall 119.4 cm (1.6) 105.3 cm (0.5) 115.5 cm (1.1)
parents)

Weight (SD) 20 kg (3.5) 31.4 kg (0.8) (1.65) 17.1 kg (4.1) 59.3 kg (NA) 25 kg (2.3) (>2) 23.6 kg (0.7) 21.6 kg (1.9) 29.5 kg (2.7)

OFC (SD) 53 cm (0.3) 51 cm (0.7) (1.6) 47 cm (4) 59 cm (2.44) 50.5 cm (1.9) (>2) NA 48 cm (2.1) 50 cm (0.4)

Walking 17 mo 22 mo 18 mo 5y 16 mo 2 y 2 mo 12 mo 2 y 6 mo 13 mo 15 mo
at age

First words no speech, 2 y/speech mild speech 8 y/2 words, 20 mo 2 y/oral 18 mo/short 3 y: no words/ 23 mo/speech NA
at age/ limit. therapy delay/speech compreh. motor sentences signs, good therapy,
speech compreh. therapy better dyspraxia non-verbal rhinolalia
abilities commun. aperta

DD/ID severe moderate global DD, profound mild-moderate moderate mild- moderate uneven profile mild-moderate
(tested) (22 mo: WPPSI-III-NL: moderate (IQ 75-88)
12 mo delay) TIQ 98,
VIQ 106,
PIQ 96

Seizures no yes (7 y) no yes (3 y) yes (4 y) possibly yes (2 y no no no


(onset) (focal, (GTCS, (atonic, GTCS) 1 (14 mo) 10 mo)
(type) nightly) absence) (myoclonic
atonic,
myoclonic,
GTCS,
absence)

Regression no no no no no NA yes (3 y) NA no no

MRI no reduced no no craniosynostosis NA parenchymal NA NA NA


anomalies white cysts right
matter perioccipital
bulk white matter

Hypotonia yes yes yes severe yes mild yes severe in infancy

Sleeping needs little sleep sleep awakes each no sleep initiation no sleep no frequent sleep initiation
difficulties initiation night difficulty initiation awakenings difficulty
difficulty difficulty

Behavioral autistic feat., autistim short 4 y: little autism, anxiety no autistic poor emotional short attention
anomalies little social spectrum concentration social features concentration, immaturity, span, ‘‘social and
interaction, disorder, span, attention interaction, sociable, some low attention emotional
no eye anxiety, panic deficit, limited intermittent behavioral span delays’’,
contact, attacks expression, hand flapping issues hand wringing
(auto-) lacks social
aggressivity intuition

(Continued on next page)

308 The American Journal of Human Genetics 103, 305–316, August 2, 2018
6
11 12 13 14 15 16 17 18 19 20

male female male male male female male male male female

14 y 1 y 6 mo 8 y 4 mo 9 y 10 mo 11 y 5 mo 19 y 2 y 4 mo 9 y 2 mo 13 y 6 mo 15 y 4 mo

c.10421G> c.1260þ1G> c.1825_ c.2084 c.20846_ c.2700_2703dup c.2709dup c.2738_ deletiona deletionb
C (p.?) C (p.?) 1829del 1G>A (p.?) 2085dup (p.Ala902Ilefs*4) (p.Glu904*) 2739del exons plus 3
(p.Glu609*) (p.Gly697*) (p.Tyr913*) 1–18 genes

3,900 g (0.6) 1,795 g (4) 2,670 g (1.7) 3,100 g (1.2) 3232 g (0.89) 2325 g (2.3) 2178 g (2.6) 2500 g (2.6) 2610 g (2.3) 3175 g (0.7)

52 cm (0.2) 40 cm (5.3) 47 cm (2) 52 cm (0.2) NA NA 45.0 cm (2.6) 48 cm (2) 47 cm (2.4) NA

36 cm (0.3) 28.5 cm (4.9) 32 cm (2.3) 33.5 cm (1.6) NA NA 32.0 cm (1.1) 32 cm (2.8) 32.4 cm (2.5) (0.66)

194 cm (3.3) 79 cm (- 0.9) 134 cm (1.3) 146 cm (0.8) 152.5 cm (0.82) 157.7 cm (1.3) 84.5 cm (1.0) 102 cm (2.1) 172 cm (1.1) 164.9 cm (0)
at 5 y

93 kg (2.1) 12.4 kg (1.3) 44.1 kg (5.4) 88 kg (3) 73 kg (2.3) 69.9 kg (0.9) 11.9 kg (0.4) 14.4 kg (2.6) 80 kg (1.7) 71.85 kg (1.2)
at 5 y

55 cm (0.3) 44.2 cm (2.2) 50 cm (2.2) 56 cm (1.6) 54.5 cm (0.32) 57.5 cm (1.7) 47.5 cm (0.8) 47.5 cm (2.9) 54 cm (0.7) 55 cm (0.1)
at 5 y

24 mo not yet 18 mo 4y 14 mo 2–3 y 2 y 4 mo 6y 18 mo 2 y 2 mo

speech delay 13 mo/ 2–3 y/limit. no speech limit. vocabulary, sentences no words, 28 mo/only 12 mo/long speech delay
stagnation, vocabulary, short sentences, several signs disyllabic stagnation,
2 words saccadic speech single words words, limit. mild oral
compreh. motor
dyspraxia

mild-moderate moderate DD, no ID in severe moderate moderate moderate severe mild- mild-
WISC-V, reduced moderate moderate
comprehension
and speed,
learning
difficulties

no no no no yes (single no no no no no
seizure 5 y)

no NA no no yes (1 y) no NA no no NA

no no NA, CT normal NA NA prominent no no right NA


posterior opercular
cerebral sulci, focal
low brainstem dysplasia
volume

NA yes mild yes NA yes yes yes facial yes

NA sleep no no sleep initiation NA no no no no


initiation difficulty, awakes
difficulty, each night
frequent
awakenings

hyperactivity, hyperactivity no anxiety, autism, ADHD, disruptive no autistic feat., repetitive autism,
mood disorder agressive anxiety, behavior stereotypies and ritual anxiety
behavior aggressive disorder, sentences,
outbursts, picking difficulties
difficulty with behaviors in social
social interaction, interaction
food seeking

(Continued on next page)

The American Journal of Human Genetics 103, 305–316, August 2, 2018 309
Table 2. Continued
Individual 1 2 3 4 5 6 7 8 9 10

Facial triangular deep set eyes periorbital brachy- and long facies, downslanting no plagiocephallus, large eyes, deep-set,
dysmorphism face, broad fullness, mild plagiocephalus, micrognathia palpebral narrow palpebral arched hooded eyes,
forehead, mild hypertelorism deep set eyes, fissures, long fissures, eyebrows, long broad nasal
exophthalmos, broad eyebrows, philtrum, low blepharochalasis, eyelashes, bridge, ear
long eyelashes, long eyelashes, set ears, large full cheeks, downslanting pit on left,
mild narrow eyebrows pointed chin palpebral high arched
hypertelorism, palpebral fissures, palate
large incisors fissures, small hypertelorism,
nose, bulbous prominent
nasal tip, nasal bridge,
anteverted broad nasal tip,
nares, smooth, smooth, short
long philtrum, philtrum, thin
narrow mouth, upper lip,
thin upper and pointed chin,
everted lower prominent
vermillion, lower
pointed chin, vermillion,
large pinnae large pinnae,
rotated ears,
prominent
tragus

Abnormalities fetal fingerpads intoeing mild small hands, arachnodactyly, NA no bilateral small hands, no
of hands and clinodactyly V, broad feet, pes planus camptodactyly broad distal
feet mild fetal short finger V, sandal phalanges,
finger pads fingersþtoes, gap, broad fetal finger
narrow end thumbsþhalluces, pads, broad
phalanges, unilat. single feet, sandal gap
clinodactyly V, palmar crease
pits on the
back of hands,
hyperconvex
nails

Cleft lip or no no no uvula bifida no short uvula no cleft palate no no


palate

Skeletal joint laxity no hypermobility slender long sagittal no no no NA no


anomalies bones, craniosynostosis
2 fractures,
osteopenia,
delayed
bone age

Recurrent no no respiratory respiratory no no no no NA no


infections/ (early tract (early
otitis media childhood childhood)
adeno
tonsillectomy)

Vision strabismus right no convergent NA strabismus no convergent convergent no


anomalies divergent strabismus squint, strabismus
squint alternans hypermetropia

Other hairy elbows, severe NA feeding tube syringomyelia, NA no atrial septal pectus tongue tie
anomalies constipation hyperemesis until age 8 y increased defect, dry excavatum (repaired), 1
gravidarum, sweating, feet with fine of inferior hypopigmented
low APGARs, pneumothorax, creases sternum spot on right
brief marfanoid shoulder
resuscitation features
required

Variant nomenclature based on GenBank: NM_001190274.1. Abbreviations: y, years; mo, months; NA, not available or not applicable; SD, standard deviation;
limit. compreh., limited comprehension; commun., communication; DD, developmental delay; feat., features; ADHD, attention deficit hyperactivity disorder;
GORD, gastresophageal reflux disease; GTCS, generalized tonic-clonic seizures.
a
hg19:chr2:g.48045596-48418922; 373,327 bp
b
hg19:chr2:g.48032122-48698515; 666,394 bp

a broader spectrum of mutational consequences including these individuals, however, cannot be excluded. Of note,
dominant-negative or gain-of-function effects that might the deletion in I20 also affects the last five exons of MSH6
explain clinical characteristics unique to certain individ- (MIM: 600678), leading to a possible additional diagnosis
uals is possible. In four individuals, exome sequencing of Lynch syndrome (MIM: 120435). Generally, additional
revealed additional (de novo) variants of unknown signifi- modifying factors have to be considered in regard to the
cance (Table S1). To our knowledge, neither TCTEX1D4 large phenotypic variability observed in this cohort.
(MIM: 611713), TP73 (MIM: 601990), nor SMARCAD1 All variants described here occurred de novo. Except for
(MIM: 612761) have been linked to NDDs and GPT2 (MIM: an unclear, maternally inherited variant in I7 with a low
138210) has been linked only to recessive ID (MRT49 frequency in ExAC and which was not phased, there was
[MIM: 616281]). A contributory effect to the phenotype of no indication of additional, likely pathogenic variants on

310 The American Journal of Human Genetics 103, 305–316, August 2, 2018
6
11 12 13 14 15 16 17 18 19 20

long face, low-set ears, sharp, tapered long angular eyebrows, large nose, frontal bossing, epicanthus, downslanting flat philtrum,
epicanthic large eye corners, palpebral broad mouth large lips, curled hair, sparse palpebral cowlick
folds, eyebrows, long eyelashes, fissures, and philtrum hooded highly arched eyebrows, fissures,
retrognathia, downslanting supernumerary simple with full lower eyelids, and sparse large everted everted lower
mandibular palpebral left superior prominent lip, full cheeks, coarse facies eyebrows, long ears, thick lips lip, mild
hypoplasia fissures incisor ears slightly uplifted eyelashes, prognathism
ear lobes downslanting
palpebral fissures,
depressed nasal
ridge,
tented upper lip

tapering no fetal NAD clinodactyly V bilateral deep plantar small and cold no toe
fingers, flat feet, fingerpads, morton’s toe, creases handsþfeet, syndactyly
sandal gap short hands small left 3rd polysyndactyly II/III, sandal
and fingers toe, broad digit V left foot gaps,
fingers with ingrown toe
hyperconvex nails
nails

NA no cleft lip and no no no no no no no


alveolar ridge

metopic no no no no no no no no no
craniosynostosis,
joint
hypermobility

yes no no no recurrent recurrent otitis 2 mo–1 y: no no NA


otitis media media, low- several otitis
frequency media
hearing loss

hypermetropia, normal hypermetropia normal bilateral bilateral no convergent normal normal


strabismus Duane degenerative starbismus
anomaly progressive
high myopia,
strabismus

cubitus valgus, recurrent NA constipation GORD in asthma, feeding cryptorchidism, no constipation,


striae, mild vomiting infancy primary difficulities renal ectasia hirsutism
aortic dilatation, amenorrhea
luxation of patella,
trichonocephaly

the second allele in the herewith reported individuals. [MIM: 617183]).30 However, the evidence for autosomal-
Given the report of a homozygous missense variant,7 recessive inheritance of FBXO11 variants is based on a sin-
FBXO11 might belong to the genes in which both domi- gle case subject and therefore limited so far.
nant and recessive variants can be associated with NDDs, Initially, FBXO11 (or PRMT9) was identified as a type II
as has been reported for other genes, for example NALCN protein arginine methyltransferase, but with a structure
(MIM: 611549) (causing congenital contractures of the different to other protein arginine methyltransferases.31
limbs and face, hypotonia, and developmental delay F-Box proteins constitute a subunit of the SKP1-Cullin-
[MIM: 616266] and hypotonia, infantile, with psychomo- F-Box (SCF) complex, an E3-ubiquitin ligase complex
tor retardation and characteristic facies 1 [MIM: 615419])29 involved in phosphorylation-dependent ubiquitina-
or ATAD3A (MIM: 612316) (causing Harel-Yoon syndrome tion.28 Ubiquitin-mediated proteasomal degradation is a

The American Journal of Human Genetics 103, 305–316, August 2, 2018 311
Figure 1. De Novo Variants Identified in FBXO11
(A) Clinical pictures of individuals with pathogenic variants in FBXO11, displaying minor and nonspecific dysmorphic features such as
epicanthal folds, down-slanting palpebral fissures, and a prominent nasal tip.
(B) Schematic drawing of genomic and protein structure of FBXO11 (isoform GenBank: NM_001190274.1, genomic coordinates
FBXO11 (hg19): chr2:48,034,059–48,132,932). Domains are color-coded according to InterPro.10 Genomic deletions are depicted
with black arrows, missense variants are highlighted in black, and likely gene-disrupting (LGD) variants are shown in red. The
herewith published variants are spread out across the protein. Corresponding positions of mutations in FBXO11 mutant mice are shown
in green.

conserved cellular mechanism that controls critical biolog- athy, early infantile, 64 [MIM: 618004]). These disorders
ical processes by regulating protein turnover. FBXO11 are associated with variable ID and syndromic or non-syn-
therefore also adds to the number of NDD-associated genes dromic features.
involved in ubiquitination and protein degradation such So far, the role of FBXO11 has been studied mainly in the
as UBE3A32 (MIM: 601623) (causing Angelman syndrome context of tumorigenesis. Somatic inactivating mutations
[MIM: 105830]), UBE3B33 (MIM: 608047) (causing Kauf- in FBXO11 have been identified in various B cell malig-
man oculocerebrofacial syndrome [MIM: 244450]), nancies, where its loss resulted in increased stability
HUWE134 (MIM: 300697) (causing mental retardation, of proto-oncogene BCL6.25 In gastric cancer, overexpres-
X-linked syndromic, Turner type [MIM: 300706]), sion of FBXO11 was found to result in increased prolifera-
USP9X35 (MIM: 300072) (causing mental retardation, tion, migration, and invasion through suppression of
X-linked 99 [MIM: 300919] and mental retardation, PTEN (MIM: 601728) and activation of the PI3K/AKT
X-linked 99, syndromic, female restricted [MIM: pathway.39 Additionally, FBXO11 promotes neddylation,
300968]), CUL4B36 (MIM: 300304) (causing mental retar- another post-translational modification analogous to ubiq-
dation, X-linked, syndromic (Cabezas type) [MIM: uitination.40
300354]), PSMD1237 (MIM: 604450) (causing Stankie- Intronic variants in FBXO11 have been associated with
wicz-Isidor syndrome [MIM: 617516]), and very recently otitis media susceptibility in humans.41–43 Similarly, a het-
RHOBTB238 (MIM: 607352) (causing epileptic encephalop- erozygous mouse model, Jeff (Jf), carrying a semi-dominant

312 The American Journal of Human Genetics 103, 305–316, August 2, 2018
Figure 2. Conservation and Structural Modeling of FBXO11 Missense Variants
(A) Conservation of amino acids affected by FBXO11 missense variants depicted with clustal omega.16,17 All herewith reported missense
variants affect highly conserved amino acid residues.
(B–E) Structural effect of FBXO11 missense variants. To evaluate the effect of the p.Ile538Val and p.Thr623Arg variants, the first and sec-
ond CASH domain of FBXO11 were modeled based on the crystal structure of the alginate epimerase AlgG (PDB: 4OZZ, 4NK818). The
template for modeling was identified using LOMETS19 and HHpred,20 and modeling was performed with Modeler 9.16.21
(B) Ile538 is located in the first CASH domain and forms hydrophobic interactions with Val551 (green arrow).
(C) These interactions cannot be formed by the shorter sidechain in the p.Ile538Val variant.
(D) Thr623 is located in the second CASH domain and forms polar interactions (green lines) with the Tyr598 and His600 sidechains.
(E) Arg623 can form only weak polar interactions with Tyr598 and instead an electrostatic repulsion between Arg623 and His600 is
observed (magenta arrow).
(F–I) The effects of the variants p.Ser840Pro and p.Ala892Asp were evaluated based on the crystal structure of the FBXO11 Zf-UBR
domain (PDB: 5VMD22).
(F) Ser840 is located in the UBR domain and coordinates a bridging water molecule (blue ball) that forms a total of three hydrogen bonds
(green lines).
(G) In the p.Ser840Pro variant, two of these hydrogen bonds are lost due to the lack of the respective hydrogens in proline and a steric
clash (magenta arrow) is caused by the cyclic proline sidechain.
(H and I) Ala892 (H) is located in the UBR domain and forms stabilizing hydrophobic interactions with Phe887 (green line), whereas in
the p.Ala892Asp variant (I), an unfavorable interaction is observed between the negatively charged Asp892 and the hydrophobic Phe887
(magenta arrow).

The American Journal of Human Genetics 103, 305–316, August 2, 2018 313
missense variant located between the two CASH domains through the Juniorverbund in der Systemmedizin ‘‘mitOmics’’
of Fbxo11,44 had chronic otitis media and conductive hear- (01ZX1405C). N. Matsumoto is supported by grants from AMED
ing loss. Additionally, heterozygous mice were smaller (JP18ek0109280, JP18dm0107090, and JP18ek0109301). C.Z. is
than wild-type littermates and displayed mild craniofacial supported by grants from the DFG (ZW184/1-2, ZW184/3-1, and
GRK2162) and by the IZKF Erlangen (E26). E.C. was supported by
abnormalities,44,45 in agreement with growth anomalies
the National Institute for Health Research (NIHR) Oxford Biomed-
and facial dysmorphisms found in several of the herewith
ical Research Centre Programme.
reported individuals. Otitis media, however, was described
in only three individuals of the herewith reported group.
Another mouse model, Mutt, carrying a missense variant Declaration of Interests
between F-box and CASH domain (Figure 1B), showed I.E.S. has served on scientific advisory boards for UCB, Eisai,
mild craniofacial anomalies but did not develop otitis me- GlaxoSmithKline, Biomarin, and Nutricia; editorial boards of the
dia, thus pointing to a milder hypomorphic effect of that Annals of Neurology, Neurology, and Epileptic Disorders; may
mutation.44 Interestingly, both mouse models in a homo- accrue future revenue on pending patent WO61/010176 (filed:
zygous state showed various degrees of perinatal lethality 2008): Therapeutic Compound; has received speaker honoraria
and displayed facial clefting and cleft palate.44 Cleft palate from GlaxoSmithKline, Athena Diagnostics, UCB, Eisai, and
or a bifid uvula were observed in three of the individuals Transgenomics; has received funding for travel from Athena Diag-
nostics, UCB, Biocodex, GlaxoSmithKline, Biomarin, and Eisai;
reported here. These findings in humans support a role
and receives/has received research support from the National
of FBXO11 in the closure of the palatal shelves.46
Health and Medical Research Council of Australia, National Insti-
To conclude, we identified de novo missense and tutes of Health, Australian Research Council, Health Research
likely gene-disrupting variants in the E3-ubiquitin ligase Council of New Zealand, CURE, and March of Dimes.
FBXO11 in individuals with a neurodevelopmental disor- All other authors declare no conflicts of interest.
der comprising a variable degree of intellectual disability
and various other features. Received: April 9, 2018
Accepted: June 29, 2018
Published: July 26, 2018
Accession Numbers
Variants were submitted to LOVD (accession numbers: 00165141- Web Resources
158).
ExAC Browser, http://exac.broadinstitute.org/
GenBank, https://www.ncbi.nlm.nih.gov/genbank/
Supplemental Data gnomAD Browser, http://gnomad.broadinstitute.org/
GTEx Portal, https://www.gtexportal.org/home/
Supplemental Data include one figure, one table, and Supple-
LOVD, http://www.lovd.nl/3.0/home
mental Acknowledgements and can be found with this article on-
OMIM, http://www.omim.org/
line at https://doi.org/10.1016/j.ajhg.2018.07.003.
RCSB Protein Data Bank, http://www.rcsb.org/pdb/home/
home.do
UCSC Genome Browser, https://genome.ucsc.edu
Acknowledgments
We thank all affected individuals and their families for participating
in this study. We thank Laila Distel for excellent technical assis-
References
tance. We thank Christian T. Thiel for the in-house NGS tool and
Arif Ekici and Steffen Uebe from the NGS facility in the Institute 1. Kochinke, K., Zweier, C., Nijhof, B., Fenckova, M., Cizek, P.,
of Human Genetics, Erlangen. This study makes use of data gener- Honti, F., Keerthikumar, S., Oortveld, M.A., Kleefstra, T., Kramer,
ated by the DECIPHER community (for additional information see J.M., et al. (2016). Systematic phenomics analysis deconvolutes
Supplemental Acknowledgements). Studies regarding individuals genes mutated in intellectual disability into biologically
18 and 11 were supported by grant PI14/00350 (Instituto de Salud coherent modules. Am. J. Hum. Genet. 98, 149–164.
Carlos III -Acción Estratégica en Salud 2013–2016; FEDER -Fondo 2. de Ligt, J., Willemsen, M.H., van Bon, B.W., Kleefstra, T.,
Europeo de Desarrollo Regional) and by PARI 2012 from Regional Yntema, H.G., Kroes, T., Vulto-van Silfhout, A.T., Koolen,
Council of Burgundy / Dijon University Hospital, respectively. D.A., de Vries, P., Gilissen, C., et al. (2012). Diagnostic exome
Exome sequencing for individual 7 was performed at the University sequencing in persons with severe intellectual disability.
of Washington Center for Mendelian Genomics (UW-CMG) and N. Engl. J. Med. 367, 1921–1929.
was funded by the NHRGI and the NHLBI (grant HG006493). 3. Deciphering Developmental Disorders, S.; and Deciphering
L.G.S. is supported by grants from the Health Research Council Developmental Disorders Study (2017). Prevalence and archi-
and Cure Kids New Zealand. A. Rauch was supported by the ERA- tecture of de novo mutations in developmental disorders.
NET grant ‘‘Euromicro’’ (SNF 31ER30_154238). A. Reis was sup- Nature 542, 433–438.
ported by the German Ministry of Education and Research 4. Rauch, A., Wieczorek, D., Graf, E., Wieland, T., Endele, S.,
(01GS08160) and the Interdisciplinary Center for Clinical Research Schwarzmayr, T., Albrecht, B., Bartholdi, D., Beygo, J., Di Do-
(IZKF) Erlangen (E16). F.D. was supported by a grant of the German nato, N., et al. (2012). Range of genetic mutations associated
Research Foundation (DFG) (DI 1731/2-1). T.B.H. is supported by with severe non-syndromic sporadic intellectual disability:
the German Federal Ministry of Education and Research (BMBF) an exome sequencing study. Lancet 380, 1674–1682.

314 The American Journal of Human Genetics 103, 305–316, August 2, 2018
5. Hauer, N.N., Popp, B., Schoeller, E., Schuhmann, S., Heath, 19. Wu, S., and Zhang, Y. (2007). LOMETS: a local meta-thread-
K.E., Hisado-Oliva, A., Klinger, P., Kraus, C., Trautmann, U., ing-server for protein structure prediction. Nucleic Acids Res.
Zenker, M., et al. (2018). Clinical relevance of systematic phe- 35, 3375–3382.
notyping and exome sequencing in patients with short stat- 20. Zimmermann, L., Stephens, A., Nam, S.Z., Rau, D., Kübler, J.,
ure. Genet. Med. 20, 630–638. Lozajic, M., Gabler, F., Söding, J., Lupas, A.N., and Alva, V.
6. Martı́nez, F., Caro-Llopis, A., Roselló, M., Oltra, S., Mayo, S., (2018). A completely reimplemented MPI bioinformatics tool-
Monfort, S., and Orellana, C. (2017). High diagnostic yield kit with a new HHpred server at its core. J. Mol. Biol. 430,
of syndromic intellectual disability by targeted next-genera- 2237–2243.
tion sequencing. J. Med. Genet. 54, 87–92. 21. Webb, B., and Sali, A. (2017). Protein structure modeling with
7. Reuter, M.S., Tawamie, H., Buchert, R., Hosny Gebril, O., MODELLER. Methods Mol. Biol. 1654, 39–54.
Froukh, T., Thiel, C., Uebe, S., Ekici, A.B., Krumbiegel, M., Zwe- 22. Muñoz-Escobar, J., Kozlov, G., and Gehring, K. (2017). Crystal
ier, C., et al. (2017). Diagnostic yield and novel candidate structure of the UBR-box from UBR6/FBXO11 reveals domain
genes by exome sequencing in 152 consanguineous families swapping mediated by zinc binding. Protein Sci. 26, 2092–
with neurodevelopmental disorders. JAMA Psychiatry 74, 2097.
293–299. 23. Guex, N., and Peitsch, M.C. (1997). SWISS-MODEL and the
8. Firth, H.V., Richards, S.M., Bevan, A.P., Clayton, S., Corpas, M., Swiss-PdbViewer: an environment for comparative protein
Rajan, D., Van Vooren, S., Moreau, Y., Pettett, R.M., and Carter, modeling. Electrophoresis 18, 2714–2723.
N.P. (2009). DECIPHER: database of chromosomal imbalance 24. Sayle, R.A., and Milner-White, E.J. (1995). RASMOL: biomole-
and phenotype in humans using Ensembl resources. Am. J. cular graphics for all. Trends Biochem. Sci. 20, 374.
Hum. Genet. 84, 524–533. 25. Duan, S., Cermak, L., Pagan, J.K., Rossi, M., Martinengo, C., di
9. Sobreira, N., Schiettecatte, F., Valle, D., and Hamosh, A. Celle, P.F., Chapuy, B., Shipp, M., Chiarle, R., and Pagano, M.
(2015). GeneMatcher: a matching tool for connecting investi- (2012). FBXO11 targets BCL6 for degradation and is inacti-
gators with an interest in the same gene. Hum. Mutat. 36, vated in diffuse large B-cell lymphomas. Nature 481, 90–93.
928–930. 26. Jin, J., Cardozo, T., Lovering, R.C., Elledge, S.J., Pagano, M.,
10. Finn, R.D., Attwood, T.K., Babbitt, P.C., Bateman, A., Bork, P., and Harper, J.W. (2004). Systematic analysis and nomencla-
Bridge, A.J., Chang, H.Y., Dosztányi, Z., El-Gebali, S., Fraser, ture of mammalian F-box proteins. Genes Dev. 18, 2573–
M., et al. (2017). InterPro in 2017-beyond protein family and 2580.
domain annotations. Nucleic Acids Res. 45 (D1), D190–D199. 27. Ciccarelli, F.D., Copley, R.R., Doerks, T., Russell, R.B., and Bork,
11. Kipreos, E.T., and Pagano, M. (2000). The F-box protein fam- P. (2002). CASH–a beta-helix domain widespread among car-
ily. Genome Biol. 1, reviews3002.1–reviews3002.7. bohydrate-binding proteins. Trends Biochem. Sci. 27, 59–62.
12. Mir, A., Sritharan, K., Mittal, K., Vasli, N., Araujo, C., Jamil, T., 28. Cardozo, T., and Pagano, M. (2004). The SCF ubiquitin ligase:
Rafiq, M.A., Anwar, Z., Mikhailov, A., Rauf, S., et al. (2014). Trun- insights into a molecular machine. Nat. Rev. Mol. Cell Biol. 5,
cation of the E3 ubiquitin ligase component FBXO31 causes 739–751.
non-syndromic autosomal recessive intellectual disability in a 29. Chong, J.X., McMillin, M.J., Shively, K.M., Beck, A.E., Marvin,
Pakistani family. Hum. Genet. 133, 975–984. C.T., Armenteros, J.R., Buckingham, K.J., Nkinsi, N.T., Boyle,
13. Harripaul, R., Vasli, N., Mikhailov, A., Rafiq, M.A., Mittal, K., E.A., Berry, M.N., et al.; University of Washington Center for
Windpassinger, C., Sheikh, T.I., Noor, A., Mahmood, H., Mendelian Genomics (2015). De novo mutations in NALCN
Downey, S., et al. (2018). Mapping autosomal recessive intel- cause a syndrome characterized by congenital contractures
lectual disability: combined microarray and exome sequencing of the limbs and face, hypotonia, and developmental delay.
identifies 26 novel candidate genes in 192 consanguineous Am. J. Hum. Genet. 96, 462–473.
families. Mol. Psychiatry 23, 973–984. 30. Harel, T., Yoon, W.H., Garone, C., Gu, S., Coban-Akdemir, Z.,
14. O’Roak, B.J., Vives, L., Girirajan, S., Karakoc, E., Krumm, N., Eldomery, M.K., Posey, J.E., Jhangiani, S.N., Rosenfeld, J.A.,
Coe, B.P., Levy, R., Ko, A., Lee, C., Smith, J.D., et al. (2012). Cho, M.T., et al.; Baylor-Hopkins Center for Mendelian Geno-
Sporadic autism exomes reveal a highly interconnected pro- mics; and University of Washington Center for Mendelian Ge-
tein network of de novo mutations. Nature 485, 246–250. nomics (2016). Recurrent de novo and biallelic variation of
15. Lek, M., Karczewski, K.J., Minikel, E.V., Samocha, K.E., Banks, ATAD3A, encoding a mitochondrial membrane protein, re-
E., Fennell, T., O’Donnell-Luria, A.H., Ware, J.S., Hill, A.J., sults in distinct neurological syndromes. Am. J. Hum. Genet.
Cummings, B.B., et al.; Exome Aggregation Consortium 99, 831–845.
(2016). Analysis of protein-coding genetic variation in 31. Cook, J.R., Lee, J.H., Yang, Z.H., Krause, C.D., Herth, N., Hoff-
60,706 humans. Nature 536, 285–291. mann, R., and Pestka, S. (2006). FBXO11/PRMT9, a new pro-
16. Goujon, M., McWilliam, H., Li, W., Valentin, F., Squizzato, S., tein arginine methyltransferase, symmetrically dimethylates
Paern, J., and Lopez, R. (2010). A new bioinformatics analysis arginine residues. Biochem. Biophys. Res. Commun. 342,
tools framework at EMBL-EBI. Nucleic Acids Res. 38, W695-9. 472–481.
17. Sievers, F., Wilm, A., Dineen, D., Gibson, T.J., Karplus, K., Li, 32. Kishino, T., Lalande, M., and Wagstaff, J. (1997). UBE3A/E6-AP
W., Lopez, R., McWilliam, H., Remmert, M., Söding, J., et al. mutations cause Angelman syndrome. Nat. Genet. 15, 70–73.
(2011). Fast, scalable generation of high-quality protein mul- 33. Basel-Vanagaite, L., Dallapiccola, B., Ramirez-Solis, R., Segref,
tiple sequence alignments using Clustal Omega. Mol. Syst. A., Thiele, H., Edwards, A., Arends, M.J., Miró, X., White,
Biol. 7, 539. J.K., Désir, J., et al. (2012). Deficiency for the ubiquitin ligase
18. Wolfram, F., Kitova, E.N., Robinson, H., Walvoort, M.T., UBE3B in a blepharophimosis-ptosis-intellectual-disability
Codée, J.D., Klassen, J.S., and Howell, P.L. (2014). Catalytic syndrome. Am. J. Hum. Genet. 91, 998–1010.
mechanism and mode of action of the periplasmic alginate 34. Froyen, G., Corbett, M., Vandewalle, J., Jarvela, I., Lawrence,
epimerase AlgG. J. Biol. Chem. 289, 6006–6019. O., Meldrum, C., Bauters, M., Govaerts, K., Vandeleur, L.,

The American Journal of Human Genetics 103, 305–316, August 2, 2018 315
Van Esch, H., et al. (2008). Submicroscopic duplications of the hibits its transcriptional activity. J. Biol. Chem. 282, 1797–
hydroxysteroid dehydrogenase HSD17B10 and the E3 ubiqui- 1804.
tin ligase HUWE1 are associated with mental retardation. Am. 41. Bhutta, M.F., Lambie, J., Hobson, L., Goel, A., Hafrén, L., Ei-
J. Hum. Genet. 82, 432–443. narsdottir, E., Mattila, P.S., Farrall, M., Brown, S., and Burton,
35. Homan, C.C., Kumar, R., Nguyen, L.S., Haan, E., Raymond, M.J. (2017). A mouse-to-man candidate gene study identifies
F.L., Abidi, F., Raynaud, M., Schwartz, C.E., Wood, S.A., association of chronic otitis media with the loci TGIF1 and
Gecz, J., and Jolly, L.A. (2014). Mutations in USP9X are associ- FBXO11. Sci. Rep. 7, 12496.
ated with X-linked intellectual disability and disrupt neuronal 42. Rye, M.S., Wiertsema, S.P., Scaman, E.S., Oommen, J., Sun, W.,
cell migration and growth. Am. J. Hum. Genet. 94, 470–478. Francis, R.W., Ang, W., Pennell, C.E., Burgner, D., Richmond,
36. Vulto-van Silfhout, A.T., Nakagawa, T., Bahi-Buisson, N., Haas, P., et al. (2011). FBXO11, a regulator of the TGFb pathway, is
S.A., Hu, H., Bienek, M., Vissers, L.E., Gilissen, C., Tzschach, associated with severe otitis media in Western Australian chil-
A., Busche, A., et al. (2015). Variants in CUL4B are associated dren. Genes Immun. 12, 352–359.
with cerebral malformations. Hum. Mutat. 36, 106–117. 43. Segade, F., Daly, K.A., Allred, D., Hicks, P.J., Cox, M., Brown,
37. Küry, S., Besnard, T., Ebstein, F., Khan, T.N., Gambin, T., Doug- M., Hardisty-Hughes, R.E., Brown, S.D., Rich, S.S., and Bow-
las, J., Bacino, C.A., Craigen, W.J., Sanders, S.J., Lehmann, A., den, D.W. (2006). Association of the FBXO11 gene with
et al. (2017). De novo disruption of the proteasome regulatory chronic otitis media with effusion and recurrent otitis media:
subunit PSMD12 causes a syndromic neurodevelopmental dis- the Minnesota COME/ROM Family Study. Arch. Otolaryngol.
order. Am. J. Hum. Genet. 100, 689. Head Neck Surg. 132, 729–733.
38. Straub, J., Konrad, E.D.H., Grüner, J., Toutain, A., Bok, L.A., 44. Hardisty-Hughes, R.E., Tateossian, H., Morse, S.A., Romero,
Cho, M.T., Crawford, H.P., Dubbs, H., Douglas, G., Jobling, M.R., Middleton, A., Tymowska-Lalanne, Z., Hunter, A.J.,
R., et al.; Deciphering Developmental Disorders Study Cheeseman, M., and Brown, S.D. (2006). A mutation in the
(2018). Missense variants in RHOBTB2 cause a developmental F-box gene, Fbxo11, causes otitis media in the Jeff mouse.
and epileptic encephalopathy in humans, and altered levels Hum. Mol. Genet. 15, 3273–3279.
cause neurological defects in Drosophila. Am. J. Hum. Genet. 45. Hardisty, R.E., Erven, A., Logan, K., Morse, S., Guionaud, S.,
102, 44–57. Sancho-Oliver, S., Hunter, A.J., Brown, S.D., and Steel, K.P.
39. Sun, C., Tao, Y., Gao, Y., Xia, Y., Liu, Y., Wang, G., and Gu, Y. (2003). The deaf mouse mutant Jeff (Jf) is a single gene model
(2018). F-box protein 11 promotes the growth and metastasis of otitis media. J. Assoc. Res. Otolaryngol. 4, 130–138.
of gastric cancer via PI3K/AKT pathway-mediated EMT. Bio- 46. Tateossian, H., Hardisty-Hughes, R.E., Morse, S., Romero,
med. Pharmacother. 98, 416–423. M.R., Hilton, H., Dean, C., and Brown, S.D. (2009). Regulation
40. Abida, W.M., Nikolaev, A., Zhao, W., Zhang, W., and Gu, W. of TGF-beta signalling by Fbxo11, the gene mutated in the Jeff
(2007). FBXO11 promotes the Neddylation of p53 and in- otitis media mouse mutant. PathoGenetics 2, 5.

316 The American Journal of Human Genetics 103, 305–316, August 2, 2018

You might also like