WO2013133708A1 - Compositions and methods for diagnosing and treating intellectual disability syndrome, autism and autism related disorders - Google Patents

Compositions and methods for diagnosing and treating intellectual disability syndrome, autism and autism related disorders Download PDF

Info

Publication number
WO2013133708A1
WO2013133708A1 PCT/NL2013/050147 NL2013050147W WO2013133708A1 WO 2013133708 A1 WO2013133708 A1 WO 2013133708A1 NL 2013050147 W NL2013050147 W NL 2013050147W WO 2013133708 A1 WO2013133708 A1 WO 2013133708A1
Authority
WO
WIPO (PCT)
Prior art keywords
auts2
mlpa
gene
deletion
syndrome
Prior art date
Application number
PCT/NL2013/050147
Other languages
French (fr)
Inventor
Alexander Jacobus Adrianus GROFFEN
Erik André SISTERMANS
Original Assignee
Stichting Vu-Vumc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Stichting Vu-Vumc filed Critical Stichting Vu-Vumc
Publication of WO2013133708A1 publication Critical patent/WO2013133708A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6827Hybridisation assays for detection of mutation or polymorphism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers

Definitions

  • the disclosure relates to methods of diagnosing and treating intellectual disability (ID) syndrome, autism and autism related disorders.
  • the disclosure provides diagnostic methods based on identifying alterations in the AUTS2 gene, in particular in exons 9- 19 of the AUTS2 gene and preferably in the AUTS2 short isoform.
  • the disclosure further relates to therapeutic interventions utilizing the newly identified alternative transcript.
  • Autism is a neuropsychiatric disorder characterized by abnormal social behavior and communication skills associated with repetitive behavior. It belongs to the group of pervasive developmental disorders ("autism spectrum” disorders) which includes autism, Asperger syndrome, Childhood disintegrative disorder, and Pervasive
  • DSM IV Diagnostic and Statistical Manual of Mental Disorders uses the following criteria to diagnose autism: (I) A total of six (or more) items from (A), (B), and (C), with at least two from (A), and one each from (B) and (C)
  • a method is provided of classifying an individual comprising detecting an alteration in an AUTS2 gene of said individual.
  • the presence of an alteration in the AUTS2 gene indicates that the individual has or is at risk of developing AUTS2 syndrome.
  • the alteration is a chromosome abnormality, more preferably an intragenic deletion.
  • said individual is first selected as having at least one symptom of the AUTS2 syndrome.
  • AUTS2 syndrome includes autism and intellectual disability.
  • a preferred form of intellectual disability (syndrome) is Rett syndrome.
  • the alteration is located between exons 9- 19 (inclusive of exons 9 and 19) of the AUTS2 gene.
  • the alteration affects the AUTS2 short isoform, e.g., by altering expression or modifying the amino acid sequence.
  • the alteration located between exons 9-19 or affecting the AUTS2 short isoform indicates the severity of the AUTS2 syndrome.
  • the detection is performed using multiplex ligation dependent probe amplification (MLPA).
  • MLPA multiplex ligation dependent probe amplification
  • said MLPA is performed using one or more probe pairs selected from MLPA 102, MLPA 110, MLPA 118, MLPA 126, MLPA 130, MLPA 096, MLPA 096, MLPA 136, MLPA 140, MLPA144, MLPA 100, MLPA 108, MLPA 112, MLPA 116, MLPA 120, MLPA 124, MLPA 128, MLPA 132, MLPA 134, MLPA 105, MLPA 138, MLPA 107, and MLPA 136.
  • the present disclosure also provides multiplex ligation dependent probe amplification (MLPA) probe sets specific for the AUTS2 gene.
  • a polypeptide comprising an AUTS2 sequence having at least 95% identity to SEQ ID NO:2 is provided.
  • the N- terminus of said amino acid sequence is encoded by exon 9 of the AUTS2 gene.
  • an isolated nucleic acid encoding said polypeptide is provided.
  • a vector comprising said nucleic acid is provided provided.
  • the vector is for use in gene therapy, preferably in the treatment of AUTS2 syndrome.
  • the disclosure further provides a method of treating an individual in need thereof comprising administering a therapeutically effective amount of a composition comprising the polypeptides, nucleic acids, vectors, or cells as described herein.
  • an anti-AUTS2 antibody which specifically binds to the AUTS2 short isoform is provided.
  • a mammalian cell comprising a nucleic acid or vector as described herein is provided.
  • Figure 1 Overview oiAUTS2 aberrations. Overview of all AUTS2 aberrations in the probands that were included in this study. The location of the deletions is indicated by the bars, the inversion breakpoint is indicated by an arrowhead, the area in which the translocation breakpoint is located is indicated by '-' . On the right you can appreciate a visualization of the AUTS2 syndrome severity score, where the darker shades indicate a more severe and/or more specific phenotype (lightest gray: ⁇ 6; light gray: 6- 10; gray: 11-15; dark gray: 16-20; darkest gray >20).
  • Figure 2 Pictures of the AUTS2 aberration patients, a: Patient 5 at the age of 3 years shows no evident dysmorphic features, b+i: Patient 8 at the age of 2,5 years has a repaired cleft lip, mild proptosis, short and mild upslanting palpebral fissures, c: The mother of patient 8 also shows a repaired cleft lip, ptosis and retrognatia. d:
  • Patient 9 at the age of 3 years shows highly arched eyebrows, mild downslanting palpebral fissures, epicanthal folds and a short philtrum.
  • e+m patient 10 at the age of about 6 years. She is hyperteloric, has a ptosis and down slanting palpebral features, a short philtrum and narrow mouth as does her brother shown in f+n at the ages of 10 years
  • g+o Patient 13 at the age of 32. You can appreciate hypertelorism, proptosis, upslanting palpebral fissures, a short upturned philtrum an a narrow mouth, h:
  • i+p Patient 17 at the age of 5,5 years. He has a hypertelorism, ptosis, a broad nasal bridge, a short and upturned philtrum and a narrow mouth
  • j+k+q Patient 19 at the age of about 1 year (j) and 4,8 years (k). He has a broad nasal bridge, short palpebral fissures and a short philtrum and narrow mouth.
  • Figure 3 Evolutionary conservation of exons that constitute the main AUTS2 transcript in humans and a novel transcriptional start site (TSS) detected in exon 9 of human AUTS2 using 5'RACE to amplify capped 5' mRNA ends in a human brain mRNA sample.
  • TSS transcriptional start site
  • AUTS2 orthologs were detected in gorilla (gorGor3), macaque (Mmul_l), dog (Broadd2), cow (Btau_4.0), pig (Sscrofa9), mouse (NCBIM37), chicken (Washuc2), clawed frog (JGI_4.2) and zebrafish (Zf9) by tblastn searches using the human amino acid sequence as a query against the corresponding genome databases, a) Compared exon-intron organization oiAUTS2 orthologs in humans and zebrafish. Red arrows indicate the location of two transcriptional start sites (TSS) used in human brain mRNA. The alternative TSS is located 1, 17 Mbp downstream in the cluster that contains exons 7- 19.
  • TSS transcriptional start sites
  • Amino acid alignment (determined by ClustalW2) of human AUTS2 orthologs in gorilla, macaque, dog, cow, pig, mouse, chicken, clawed frog and zebrafish. Exon boundaries are indicated above the alignment. Fully conserved or similar residues are marked by symbols at below the alignment. Grey lowercase letters indicate low-complexity or repetitive segments of the human amino acid sequence according to the XNU + SEG algorithms 11 13 .
  • FIG. 6 NM_015570 Homo sapiens autism susceptibility candidate 2 (AUTS2) mRNA
  • the autism susceptibility candidate 2 (AUTS2) gene was identified in a translocation breakpoint analysis in twins with autism, developmental delay and epilepsy 1 .
  • the authors of this study concluded that AUTS2 is unlikely an autism susceptibility gene for idiopathic autism.
  • five patients with a translocation breakpoint and one patient with an inversion breakpoint disrupting AUTS2 were described; these patients have intellectual disability (ID) and/or developmental delay (all), autism features (in four reported cases), epilepsy and skeletal abnormalities (reported in three cases each) 2 4 .
  • ID intellectual disability
  • all autism features
  • epilepsy epilepsy
  • skeletal abnormalities reported in three cases each
  • Disruptions in either neighboring genes or distantly located genes may also be responsible for the patient phenotypes.
  • an intragenic deletion in AUTS2 has any affect on neurodevelopment.
  • the function of the protein encoded by AUTS2 is largely unknown.
  • the longest transcript oiAUTS2 has 19 exons and spans 1.2 Mb.
  • the wild-type human AUTS2 gene sequence corresponds to Gene ID: 26053 located on human chromosome 7ql l.22. It encodes a 1,295 amino acid protein that contains 2 potential proline-rich protein- binding domains 1 .
  • the mouse Auts2 ortholog is highly expressed in the developing mouse brain, specifically in the frontal cortex, the hippocampus and the cerebellum— areas known to be affected in autistic patients.
  • the Auts2 protein is located in the nucleus and co-localizes in the cortex with Tbrl, a transcription factor critical for corticogenesis 8 .
  • AUTS2 syndrome The 5' end (exon 1-4) oiAUTS2 was flagged for having the strongest statistical signal for a putative positive selective sweep in early modern humans 7 .
  • the frequency of derived alleles for human polymorphic sites is less than expected in Neanderthals.
  • the present invention is based, in part, on results which conclusively demonstrate that that disruption of the AUTS2 gene is associated with a complex phenotype that includes not only autism, but also a series of anatomical and neurodevelopmental defects including intellectual disability, which are referred to herein as AUTS2 syndrome.
  • Table 1 provides the phenotypes associated with AUTS2 syndrome and their prevalence.
  • Embodiments of the invention are also based on the discovery that the AUTS2 gene encodes an alternatively spliced isoform in the human brain. The inventors further demonstrate that disruptions in the genomic region encoding the alternatively spliced isoform are associated with more severe form of AUTS2 syndrome.
  • AUTS2 syndrome is characterized by intellectual disability, microcephaly, mild short stature, feeding difficulties, hypotonia, cerebral palsy and dysmorphic features. It is understood that an affected individual will usually not present with all of the symptoms and may show varying degrees of severity for each symptom.
  • AUTS2 syndrome is characterized by at least two symptoms selected from intellectual disability, microcephaly, mild short stature, feeding difficulties, hypotonia, cerebral palsy and dysmorphic features.
  • AUTS2 syndrome is characterized by at least the presence of autism as defined by the DSM IV.
  • AUTS2 syndrome is synonymous with autism.
  • the AUTS2 syndrome is synonymous with intellectual disability.
  • One aspect of the disclosure provides a method of classifying an individual comprising detecting an alteration in an AUTS2 gene of said individual. Alterations may be detected at the level of DNA, RNA, or protein, preferably said alteration is detected in a nucleic acid. Said methods may detect a change in expression level (including haploinsufficiency caused either by a deletion or by nonsense mediated decay) or change in amino acid sequence.
  • Alterations include the insertion or deletion of a single nucleotide, or of more than one nucleotide, resulting in a frame shift; the change of at least one nucleotide, resulting in a change in the encoded amino acid; the change of at least one nucleotide, resulting in the generation of a premature stop codon; the deletion of several nucleotides, resulting in a deletion of one or more amino acids encoded by the nucleotides; the insertion of one or several nucleotides, such as by unequal recombination or gene conversion, resulting in an interruption of the coding sequence of a reading frame; duplication of all or a part of a sequence; transposition; or a rearrangement of a nucleotide sequence.
  • sequence changes can alter the polypeptide encoded by the nucleic acid.
  • the change in the nucleic acid sequence causes a frame shift
  • the frame shift can result in a change in the encoded amino acids, and/or can result in the generation of a premature stop codon, causing generation of a truncated polypeptide.
  • An alteration can also alter splice sites, affect the stability or transport of mRNA, or otherwise affect the transcription or translation of an encoded polypeptide. It can also alter DNA to increase the possibility that structural changes, such as amplifications or deletions, occur at the somatic level. These alterations can be detected by any number of methods known to a skilled person at the level of DNA, RNA, or protein.
  • Detection of the expression levels can be performed using immunological assays, including, Western Blot, immunohistochemistry, FACS analysis, radio immuno assay (RIA), immunofluorescence.
  • Methods for the detection of particular mRNAs are well known and include, for example, hybridization assays using complementary DNA probes (such as in situ hybridization using labeled riboprobes, Northern blot and related techniques) and various nucleic acid amplification assays (such as RT-PCR using complementary primers specific for the AUTS2 short isoform, and other amplification type detection methods).
  • Chromosome abnormality refers to a deviation between the structure of the subject chromosome and a normal homologous chromosome.
  • the term "normal” refers to the predominate structure or nucleic acid sequence found in healthy individuals. Chromosome abnormalities include insertions, deletions, duplications, translocations, and inversions. Preferably, the abnormality is an intragenic deletion of the AUTS2 gene.
  • an intragenic deletion is a less severe chromosomal abnormality than, e.g., large deletions or inversions. While duplications, translocations, and inversions, can affect several genes, the effects of an intragenic deletion are normally confined to the specific gene.
  • the intragenic deletion is less than 150kb, more preferably less than lOOkb.
  • the method of the invention permits the classification of an individual as to the risk of developing AUTS2 syndrome or of the likelihood of suffering from AUTS2 syndrome.
  • the method of the invention also permits the classification of an individual as to the risk that an offspring of said individual may be of risk of developing AUTS2 syndrome.
  • the current diagnosis of autism and autism related disorders is based on behaviour. This manner of diagnosis suffers from the disadvantage that the symptoms may widely vary among individuals and that the diagnosis can be complicated by other conditions, such as blindness, which can result in difficulties in socializing or communicating.
  • the method of the invention provides objective criteria by which to assess the presence or risk of developing AUTS2 syndrome.
  • the presence of at least one alteration in the AUTS2 gene is an indicator that the individual is at risk of developing AUTS2 syndrome, is likely suffering from AUTS2 syndrome, or is at risk of producing an offspring having AUTS2 syndrome.
  • the assessment of risk of developing or likelihood of suffering from AUTS2 syndrome may also include determining whether the individual has intellectual disability, multiple congenital anomaly (MCA), or difficulties in social skills, language and behaviour that are known to be associated with autism.
  • MCA multiple congenital anomaly
  • the method of the invention can be used in combination with symptomatic diagnostic criteria, e.g. based on DSM IV autism diagnosis.
  • symptomatic diagnostic criteria e.g. based on DSM IV autism diagnosis.
  • Both the risk of and likelihood of having AUTS2 syndrome also increase when an individual also exhibits one or more of the following features: microcephaly, mild short stature, feeding difficulties, hypotonia, cerebral palsy and dysmorphic features.
  • the risk of and likelihood of having AUTS2 syndrome may also increase when an individual does not have any symptoms, but has a family history of autism.
  • any individual may be classified using a method of the invention. It is preferred that said individual is tested as soon as possible, preferably before or at birth. In another preferred embodiment the individual is first selected based on behavioural criteria, such as having at least one symptom of the AUTS2 syndrome.
  • behavioural criteria such as having at least one symptom of the AUTS2 syndrome.
  • an individual is selected who presents with intellectual disability, multiple congenital anomaly (MCA), or difficulties in social skills, language and behaviour that are known to be associated with autism.
  • MCA congenital anomaly
  • the individual has been diagnosed with autism using symptomatic diagnostic criteria, e.g. based on DSM IV.
  • said individual also exhibits one or more of the following features: microcephaly, mild short stature, feeding difficulties, hypotonia, cerebral palsy and dysmorphic features.
  • the AUTS2 syndrome is characterized by intellectual disability and at least one other symptom selected from microcephaly, mild short stature, feeding difficulties, hypotonia, cerebral palsy, and dysmorphic
  • the individual does not have any symptoms but has a family history of autism.
  • said individuals are humans and may be adults, children, or fetuses.
  • a UTS2 encodes, in addition to the full- length message, a shorter isoform in the human brain.
  • This shorter isoform uses an alternative transcription start site in the center of exon 9 followed by a start codon in the same reading frame as the full length AUTS2 transcript.
  • the sequence of the short isoform is highly conserved across species. Consistent with a critical functional role for this isoform, deletion of this region is associated with significantly more severe phenotypes.
  • the nucleic acid coding sequence of the shorter isoform is:
  • the protein sequence of the shorter isoform is predicted to be:
  • R (SEQ ID NO:2).
  • the present disclosure provides methods as described herein for classifying an individual comprising detecting an alteration in the AUTS2 short isoform.
  • the AUTS2 short isoform refers to SEQ ID NO:2.
  • An alteration includes a mutation affecting the expression and/or amino acid sequence of the AUTS2 short isoform. Alterations indicate an increased risk of developing or having AUTS2 syndrome.
  • the methods disclosed herein comprise detecting at least one alteration, preferably a chromosomal abnormality, between exons 9- 19 (inclusive of exons 9 and 19) of the AUTS2 gene of said individual. Exons 9-19 correspond to nucleotide positions 2212-6426 of NM_015570.
  • the alteration may affect exons 9-19 or their corresponding introns. These exons encode the AUTS2 short isoform and alterations in either the intervening introns or 5' and 3' genomic regions may affect the expression and/or proper splicing of the short isoform.
  • the alteration is an intragenic deletion deleting at least one or a part of exons 9-19.
  • the alteration between exons 9-19 of the AUTS2 gene is preferably in the genomic region which encodes the short isoform and/or affects translation or splicing. Identification of an alteration between exons 9-19 or in the genomic region encoding the AUTS2 short isoform may also indicate that the severity of AUTS2 syndrome is likely to be greater than the severity resulting from an abnormality in the 5' region of AUTS2. This finding is contrary to what one would have predicted. A disruption in the 3' end of a gene often results in a truncated protein which may retain a small amount of function.
  • disruptions occurring in the 5' end of the AUTS2 gene may have a lesser effect on the shorter isoform, which can then perform its normal function.
  • the detection of the chromosomal abnormalities and alterations described herein may be performed on any sample from an individual which comprises nucleic acid, in particular genomic DNA. Suitable samples include bodily fluids (e.g., blood, urine, serum, cerebral spinal fluid, amniotic fluid), cells (e.g., brain cells, white blood cells), or body tissue. Standard methods are known for extracting DNA from samples.
  • Nucleic acid may be used in a purified or unpurified form.
  • a number of methods for detecting chromosome abnormalities are known in the art, including karyotyping, FISH, array-CGH, MAPH, sequencing assays, quantitative PCR (q-PCR), QMPSF (Quantitative Multiplex PCR of Short Fluorescent Fragments, see Casilli et all, 2001 Hum Mutat 20:218-26) and MLPA. (see U.S. Publication US2011/0281759)
  • Karyotyping is a conventional procedure for determining the chromosome complement of an individual, defined both by the number and morphology of the chromosomes. Stains are used to produce banding patterns on the chromosomes which permits the identification of the chromosome and the visualization of structural abberations.
  • GTG banding (G-bands by trypsin using Giemsa) is a standard cytogenetic technique.
  • Hybridization assays such as fluorescence in situ hybridization (FISH) are also common techniques for detecting chromosomal abnormalities.
  • FISH fluorescence in situ hybridization
  • chromosomes are hybridized with "painting probes" labeled by combinations of different fluorophores. Details of this technique and variations of the techniqe are found, e.g., in Liehr et al. Histol Histopathol 2004 19:229-237.
  • Array comparative genomic hybridization (array-CGH) and Multiplex Amplifiable Probe Hybridization (MAPH) are both molecular-cytogenetic methods for the analysis of copy number variation (Kousoulidou et al. Methods Mol Biol 2010 653:47-71).
  • Copy number variation refers to the number of copies of a particular gene.
  • Amplification assays are also known to the skilled person and are suitable for detecting chromosome abnormalities.
  • 3C-qPCR is one exemplary technique (Hagege et al. 2007 Nature Protoc 2" 1722-33).
  • a chromosomal abnormality in the AUTS2 gene is detected using MLPA.
  • MLPA is a technique to determine copy number (Schouten et al. 2002 Nucleic Acids Res. 30:e57).
  • MLPA probes are designed to hybridize to a region of interest.
  • MLPA probes comprise a set of two probes. Each probe of the probe set is designed to hybridize to the target DNA in close proximity to each other.
  • Each probe in a probe set has between 15-100 nucleic acids complementary the target sequence, preferably between 20-50 nucleotides (referred to as LHS and RHS, left and right hybridizing sequence, respectively.)
  • Each probe has, in addition the LHS and RHS, a primer recognition sequence.
  • LHS and RHS a primer recognition sequence
  • a "stuffer" sequence may be inserted between the LHS/RHS and the primer recognition sequences.
  • the stuffer sequence may be used to alter the length of the amplified probe in order to distinguish the particular probe set from other probe sets. Further details of this method are described in U.S. Patent No. 6,955,901 which is hereby incorporated by reference in its entirety.
  • the MLPA technique has the advantage that small deletions and duplications can be detected and the location of the abnormality can be finely mapped to a specific region. MLPA permits the high resolution detection of abnormalities in specific gene candidates. It also is able to measure copy number changes with high quantitative power. In the case of the AUTS2 gene, these advantages are of importance as we have discovered that relatively small deletions can be pathogenic and the severity of the resulting disorder can be predicted based on the location of the abnormality. While MLPA is a well-known technique, its application in determining alterations in the AUTS2 gene is surprising as it was not known before the present disclosure that intragenic deletions of AUTS2 could lead to the phenotypes described in Table 1.
  • One aspect of the disclosure provides multiplex ligation dependent probe
  • MLPA amplification amplification
  • “Specificity” for the AUTS2 gene refers to the ability of each probe in the set of two to bind adjacently to each other to the AUTS2 gene.
  • Each probe of the set may also bind to other genomic sequences, however, for amplification to occur, the probes need to bind next to each other in order to ligate.
  • the MPLA probe set is specific for a genomic region between exons 9-19 of the AUTS2 region, more preferably for the genomic region which encodes the short isoform.
  • the probe sets may be designed to either the sense or anti-sense strand. While exon specific probes are generally preferred, some exons may not contain suitable sequences with which to design MLPA probes.
  • MLPA probe pairs useful in the present invention include MLPA 102, MLPA 110, MLPA 118, MLPA 126, MLPA 130, MLPA 096, MLPA 096, MLPA 136, MLPA 140, MLPA144, MLPA 100, MLPA 108, MLPA 112, MLPA 116, MLPA 120, MLPA 124, MLPA 128, MLPA 132, MLPA 134, MLPA 105, MLPA 138, MLPA 107, and MLPA 136.
  • Probe sequences are provided in Table 6. Table 6A provides the "left" probe and Table 6B provides the "right" probe for each probe pair.
  • MLPA 102 probe refers to a pair of sequences comprising:
  • MLPA probe design is described, e.g., in Pantano et al. BMC Genomics 2008 9:573, Zhi J. BMC Research Notes 2010 3: 137 and MRC-Holland B.V. provides a detailed probe design protocol at www.mlpa.com.
  • One aspect of the disclosure provides antibodies specific for the AUTS2 short isoform of SEQ ID NO:2.
  • “Specific for the AUTS2 short form” refers to antibodies which recognize SEQ ID NO:2, but not exons 1-8 of AUTS2.
  • the antibody specific for the AUTS2 short isoform of SEQ ID NO:2 does not recognize the protein encoded by the longer transcript.
  • the AUTS2 specific antibody is typically specific for a three dimensional epitope not present in the protein encoded by the longer transcript.
  • the antibody specific for the AUTS2 short isoform of SEQ ID NO:2 can also be specific towards the unique 5' end of the shorter isoform.
  • Such antibodies are useful to detect not only the presence of the AUTS2 short isoform, but also changes in expression level. Suitable antibodies include monoclonal and polyclonal antibodies as well as fragments containing the antigen binding domain and/or one or more
  • an antibody fragment is defined as at least a portion of the variable region of the immunoglobulin molecule that binds to its target, i.e., the antigen binding region.
  • the present invention also provides new therapeutic applications for treating AUTS2 syndrome and in particular autism.
  • the disclosure demonstrates that loss of auts2 in zebrafish embryos causes microcephaly and decreases neuronal staining. This phenotype is rescued by the C-terminal AUTS2 human mRNA. The AUTS2 short isoform is therefore sufficient for rescuing the loss of AUTS2 in zebrafish and is useful in treating disorders mediated by AUTS2
  • the disclosure provides a polypeptide comprising the AUTS2 sequence having at least 80, at least 85, preferably at least 90, more preferably at least 95% identical to SEQ ID NO:2.
  • polypeptide may include additional sequences, such as protein tags (His, Flag, Myc, HA, GST, V5, GFP, etc.), it does not comprise additional AUTS2 sequences, namely from exons 1-8.
  • additional AUTS2 sequences namely from exons 1-8.
  • the N-terminus of said polypeptide is encoded by exon 9 of the AUTS2 gene.
  • said polypeptide may include additional sequences, such as protein tags (His, Flag, Myc, HA, GST, V5, GFP, etc.), it does not comprise additional AUTS2 sequences, namely from exons 1-8.
  • the N-terminus of said polypeptide is encoded by exon 9 of the AUTS2 gene.
  • polypeptide comprises the alternatively spliced amino acids VRTPGRN.
  • the disclosure provides a nucleic acid molecule encoding the polypeptides disclosed herein.
  • the nucleic acid sequence encodes the amino acid sequence of SEQ ID NO:2 or an amino acid sequence at least 80, at least 85, preferably at least 90, more preferably at least 95% identical to SEQ ID NO:2.
  • the nucleic acid molecule comprises SEQ ID NO: l or a nucleic acid molecule at least 80, at least 85, preferably at least 90, more preferably at least 95% identical to SEQ ID NO: 1.
  • the nucleic acid molecule may comprise additional sequences, for example, promoter or expression sequences, but does not comprise additional sequences of AUTS2, e.g., exons 1-8.
  • the nucleic acid encodes a polypeptide encoded by exon 9 of the AUTS2 gene.
  • the nucleic acid molecule may be provided in a vector.
  • a "vector” is a recombinant nucleic acid construct, such as plasmid, phase genome, virus genome, cosmid, or artificial chromosome, to which another DNA segment may be attached.
  • the term “vector” includes both viral and nonviral means for introducing the nucleic acid into a cell in vitro, ex vivo or in vivo.
  • Non-viral vectors include plasmids, liposomes, electrically charged lipids (cytofectins), DNA-protein complexes, and biopolymers.
  • Viral vectors include retrovirus, adeno-associated virus, pox, baculovirus, vaccinia, herpes simplex, Epstein-Barr and adenovirus vectors, as set forth in greater detail below.
  • Vector sequences may also contain one or more regulatory regions, and/or selectable markers useful in selecting, measuring, and monitoring nucleic acid transfer results (transfer to which tissues, duration of expression, etc.). Cells comprising said nucleic acids or vectors comprising nucleic acids are also provided.
  • the method of introduction is largely dictated by the targeted cell type include, e.g., CaP04 precipitation, liposome fusion, lipofectin, electroporation, dextran-mediated transfection, calcium phosphate precipitation, polybrene mediated transfection, protoplast fusion, viral infection, encapsulation of the polynucleotide (s) in liposomes, and direct microinjection of the DNA into nuclei.
  • the nucleic acids may stably integrate into the genome of the host cell (for example, with retroviral introduction, outlined below), or may exist either transiently or stably in the cytoplasm (i.e. through the use of traditional plasmids, utilizing standard regulatory sequences, selection markers, etc.).
  • AUTS2 short isoform polypeptides as described herein may be produced by culturing a host cell transformed with an expression vector containing nucleic acid encoding a dominant negative polypeptide.
  • host cells include yeast, bacteria, archaebacteria, fungi, and insect and animal cells, including mammalian cells. Of particular interest are Drosophila melangaster cells, Saccharomyces cerevisiae and other yeasts, E. coli, Bacillus subtilis, SF9 cells, C129 cells, 293 cells, Neurospora, BHK, CHO, COS, Pichia pastoris, etc.
  • said polypeptides are expressed in mammalian cells. Mammalian expression systems are also known in the art, and include retroviral systems.
  • Suitable cell types include tumor cells, Jurkat T cells, NIH3T3 cells, CHO, and Cos, cells.
  • the cells are culturable cells and more preferred the cells are in vitro cells.
  • the nucleic acid sequence encoding the AUTS2 short isoform may be provided to a cell as part of a gene delivery vehicle.
  • a gene delivery vehicle is preferably a liposome or a viral gene delivery vehicle. Liposomes are well known in the art and many variants are available for gene transfer purposes.
  • Various viral gene delivery are currently used to transfer genes into target cells. In the present disclosure it is preferred to use those viral vectors that do not express their own genes but only the transferred genes.
  • the nucleic acid compound is preferably provided as an expression cassette wherein the expression cassette encodes a transcript comprising said compound.
  • a further aspect of the present disclosure is the provision of a vector comprising a nucleic acid molecule encoding the AUTS2 short isoform for use in gene therapy.
  • Such therapy is useful in the treatment of AUTS2 syndrome.
  • Methods are therefore provided for treating an individual afflicted with or at risk of developing AUTS2 syndrome comprising administering to an individual in need thereof a nucleic acid molecule encoding the AUTS2 short isoform.
  • the nucleic acid molecule is preferably provided in a viral vector suitable for gene therapy.
  • Appropriate vectors and delivery methods are known to a skilled person and are described, e.g., in Schlachetzki et al. Neurology, Gene therapy of the brain. (2004) 62: 1275- 1281 and Richardson et al. Neurosurg Clin N Am. 2009 20(2):205-10.
  • a further aspect of the disclosure provides pharmaceutical composition
  • a pharmaceutically acceptable excipient may for instance be found in Remington: The Science and Practice of Pharmacy, 20th Edition. Baltimore, MD: Lippincott Williams & Wilkins, 2000.
  • the compound is dissolved in a solution that is compatible with the delivery method.
  • the solution is a physiological salt solution.
  • excipients capable of forming complexes, vesicles and/or liposomes that deliver such a compound as defined herein in a vesicle or liposome through a cell membrane.
  • Suitable excipients comprise polyethylenimine (PEI) or similar cationic polymers, including polypropyleneimine or polyethylenimine copolymers (PECs) and derivatives, ExGen 500, synthetic amphiphils (SAINT- 18), lipofectinTM, DOTAP and/or viral capsid proteins that are capable of self assembly into particles that can deliver such compounds, to a cell.
  • the compositions can be administered by controlled release means or by delivery devices that are well known to those of ordinary skill in the art. Examples include, but are not limited to, those described in U.S. Pat. Nos. 3,845,770; 3,916,899;
  • Such dosage forms can be used to provide slow or controlled-release of one or more active ingredients using, for example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions.
  • Suitable controlled-release formulations known to those of ordinary skill in the art, including those described herein, can be readily selected for use with the active ingredients of the invention.
  • the invention thus encompasses single unit dosage forms suitable for oral administration such as, but not limited to, tablets, capsules, gelcaps, and caplets that are adapted for controlled-release.
  • Actual dosage levels of the pharmaceutical preparations described herein may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • the selected dosage level will depend upon a variety of factors including the activity of the particular compound, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • the disclosure provides a method of treating an individual, preferably a human, in need thereof comprising administering a therapeutically effective amount of a pharmaceutical composition as disclosed herein.
  • the composition is administered intracerebrally or into the spinal cord of the individual.
  • the disclosure further provides non-human animals, preferably mammals, comprising nucleic acids encoding AUTS2 short isoform.
  • Methods for introducing nucleic acids into animals are known to one of skill in the art and include standard transgenic techniques such as introducing said nucleic acid into an undifferentiated cell type, e.g., an embryonic stem (ES) cell.
  • ES embryonic stem
  • the ES cell is injected into a mammalian embryo, where it integrates into the developing embryo. Insertion of the nucleic acid construct into the ES cells can be accomplished using a variety of methods well known in the art including for example, electroporation, microinjection, and calcium phosphate treatment.
  • the embryo is implanted into a foster mother for the duration of gestation.
  • Transgenic animals comprise a heterologous nucleic acid sequence present as an extrachromosomal element or stably integrated in all or a portion of its cells, especially in germ cells.
  • chimeras or “chimeric animals” are generated, in which only a subset of cells have the altered genome. Chimeras are primarily used for breeding purposes in order to generate the desired transgenic animal. Animals having a heterozygous alteration are generated by breeding of chimeras. Male and female heterozygotes are typically bred to generate homozygous animals.
  • transgenic animals particularly animals such as mice or rats
  • Methods for generating transgenic animals have become conventional in the art and are described, for example, in U.S. Pat. Nos. 4,736,866 and 4,870,009.
  • the invention further provides a non-human animal, preferably a non-human vertebrate wherein the AUTS2 short isoform is knockout on at least one and preferable two of the AUTS2 chromosomes in the cell.
  • a non-human animal preferably a non-human vertebrate wherein the AUTS2 short isoform is knockout on at least one and preferable two of the AUTS2 chromosomes in the cell.
  • the invention provides a non-human animal, preferably a non-human vertebrate, wherein the AUTS2 short isoform is silenced, preferably by means of an siRNA specific for the AUTS short isoform.
  • said non- human animal is a mammal or fish.
  • to comprise and its conjugations is used in its non-limiting sense to mean that items following the word are included, but items not specifically mentioned are not excluded.
  • verb "to consist” may be replaced by "to consist essentially of meaning that a compound or adjunct compound as defined herein may comprise additional component(s) than the ones specifically identified, said additional component(s) not altering the unique characteristic of the invention.
  • an element means one element or more than one element.
  • treating includes prophylactic and/or therapeutic treatments.
  • prophylactic or therapeutic treatment is art-recognized and includes administration to the host of one or more of the subject compositions. If it is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the host animal) then the treatment is prophylactic (i.e., it protects the host against developing the unwanted condition), whereas if it is administered after manifestation of the unwanted condition, the treatment is therapeutic, (i.e., it is intended to diminish, ameliorate, or stabilize the existing unwanted condition or side effects thereof).
  • an inversion (patient 20) and a translocation (patient 21) with one of the breakpoints in AUTS2 were found by conventional karyotyping from two different laboratories; detailed genotypic and phenotypic data were available for both of these patients. In total 25 patients from 21 unrelated families were included in our analyses (see figure 1, figure 6, and table 7).
  • the deletion involved a downstream gene, WBSCR17, while in patients 17 and 19 two downstream genes, WBSCR17 and CALN1 were also included in the deletion (see figure 1).
  • probands had intellectual disability or a developmental delay (patient 1, 2, 4-21); only patient 3 had normal intellectual development.
  • Two of the parents carrying an AUTS2 deletion had a normal intelligence (the mother of patient 3 and the father of patient 5), one had experienced learning difficulties (the mother of patient 8) and one had mild intellectual disability (mother of patient 10 and her sibling).
  • Ten probands were diagnosed with AS or showed autistic behavior (patient 3, 4, 7, 9, 13, 16, 17, 20 and 21). The mother of patient 3 reported herself to have
  • the phenotype caused by the observed AUTS2 aberrations is variable.
  • microcephaly is strongly associated with ASD (Shinawi, M. et al. J Med Genet 47, 332-41 (2010)). Likewise, microcephaly is the most representative clinical feature amongst our patients with an AUTS2 deletion (16/43).
  • sb-MOs splice-blocking morpholinos
  • Table 1 Phenotypic features characterizing the AUTS2 syndrome patients
  • Table S2 Control cohorts. The control cohort tested for CNV's in AUTS2 with the number of controls per cohort and the references that give more information on these cohorts. The number of deletions identified in the controls at the AUTS2 locus is indicated. Datasets 1, 2, 4, and 5 were determined with Affymetrix 6.0; dataset 3 was determined with Illumina 1M; dataset 6 was determined with Illumina 240K-650K; and dataset 7 was determined with agillent 105K/180K.
  • Table S4 Overview of the location of the probes used for MLPA analysis. All features are sorted for the start position with numbering according to genome build HG18.
  • Table S5 FISH probes used for breakpoint mapping on chromosome 7 and chromosome 22. Showing probe name, chromosome band, start position and end position according to the GRCh37 genome assembly.
  • exon 18 L107_ _auts2 gggttccctaagggttggaTGTCTCAGTCGCAGTTATACATCAGCACGTC
  • R104_auts2 /5Phos/GAACATGACAGCCAGGATGCAGGGCCGATTtctagattggatcttgctggcgcgtcc
  • R116_auts2 /5Phos/TTACCATGCTACACATTGAATGTAACTGCTTTGCCtctagattggatcttgctggcgcgtcc
  • R120_auts2 /5Phos/ATGGCATCAAACTGAGATTACGTGGCTTGCTCAtctagattggatcttgctggcgcgtcc
  • R124_auts2 /5Phos/GTCCCACACACTTTACTCCAAAAGGACCCGAGGGTACGTGCAAAGTCtctagattggatcttgctggcgcgt.cc
  • R128_auts2 /5Phos/CCAGTTAGTAGACAGCTTTAAAAGCCAGGGGTCTGAAGGCCATTtctagattggatcttgctggcgcgtcc
  • R132_auts2 /5Phos/AATGTCTTCCTCCTAACCACGTTGCTCTTTCTTGTTCCAGAAACCAGGGAAGTGtctagattggatcttgctggcgcgtcc
  • R136_auts2 /5Phos/CCGACCCTAGTAACAAAAGTCTCTCAGTTACCCAGGGCACattatcaggagcatttctagattggatcttgctggcgcgtcc
  • R140_auts2 /5Phos/TCCAGGCTACATGAGACATGCAGACATGCTCCAGTGTCATGAACCTCCattatcaggtctagattggatcttgctggcgcgtcc
  • R144_auts2 /5Phos/AGTCCAGCCGTTGTCCTAAGGAGGGAGCAGGCCACCTCAGACAGAAAATGCAGCTGGAGtctagattggatcttgctggcgcgtcc
  • R102_auts2 /5Phos/CCTCGiGCAGAAGAGGACATCATTGATGGatttctagattggatcttgctggcgcgtcc
  • R118_auts2 /5Phos/ATCCAAGGGCTTCCACCGGAGCAGCTCatatttctagattggatcttgctggcgcgtcc
  • R126_auts2 /5Phos/CTGGCCATGTAAGGAGATACAGGGAACgattatcaggtattctagattggatcttgctggcgcgtcc
  • R130_auts2 /5Phos/GCACCTTGCTGGACTGACCTGAGGAAGAGCttgaatgcgagttattatctctagattggatcttgctggcgcgtcc
  • R134_auts2 /5Phos/TGTTCCCCTTGCTGTGTAGAAACAGATGCAGTCAGACCCACATAAGCTtctagattggatcttgctggcgcgtcc
  • R105_auts2 /5Phos/GTTTCAGTACATCCCGTCCAGCTTCCAGATTctctagattggatcttgctggcgcgtcc
  • R138_auts2 /5Phos/CTACATTCACAGGCCTAGCAGCAGTTGGTGGCAATGCCTTCGGGGGACTTGGAAATCCTTCCtctagattggatcttgctggcgcgtcc
  • R136_auts2 /5Phos/CGTTTCCTGATGTGACGCACTGTATTCCAGCTGGTGATCAAGTCTGGGAAtctagattggatcttgctggcgcgtcc
  • NimbleGen 135k custom (custom design 135k (custom design signature) signature) design signature
  • Brain disorder ND (assymetric) ND cerebral palsy/spasticity +(assymetric)
  • Huang 2 CA M09-0400 DGAP201 1
  • Routine diagnostic array CGH and karyotyping was performed for intellectual disability (ID)/ multiple congenital anomalies (MCA) for a total of about 50,000 patients in ten diagnostic centers in the Netherlands, Belgium, Great Britain, the USA and Canada.
  • ID intellectual disability
  • MCA multiple congenital anomalies
  • Peripheral blood samples of patients with an AUTS2 aberration were obtained for further studies after informed consent of the patient or parents where appropriate. Clinical information was collected through the referring physicians and with approval of the local medical ethical committee. Informed consent was obtained from parents for publishing photographs. Patients were included in this study when detailed phenotypic an genotypic data were available.
  • the clinical data of patients and carrier parents were used to identify features that occurred in at least two unrelated patients. These features were added to the list of clinical features characterizing the AUTS2 syndrome for which all patients were scored positive or negative. Asymmetrically occurring features were not counted as positive. The sum of positive features was counted for each patient and was defined as the AUTS2 syndrome severity score, which was used to investigate a genotype- phenotype correlation.
  • Chromosome analysis was performed on standard synchronized cultures of peripheral blood lymphocytes. In general GTG-banded chromosomes were analyzed at the 550- band level from five metaphases and the chromosomes were counted from ten metaphases. Array CGH
  • the relative DNA copy numbers at the SNP/CNV loci from the Affymetrix platforms were determined by comparison of the normalized array signal intensity data for the proband's DNA sample against the HapMap270 reference file provided by Affymetrix, using Genotyping Console or ChAS software (default settings). All rare CNV's are checked using Nexus 20 . Analysis and intra-chip normalization of the Illumina image files was performed using lllumina's GenomeStudio Genotyping Module software v.2010.3 with default parameters.
  • Genotype calls were generated using the Illumina-provided genotype cluster definitions file (HumanOmni2.5-4vl_B.egt, generated using HapMap project DNA samples). CNV analysis was performed using a multi- algorithm approach, as described elsewhere 25 . Data analysis of the NimbleGen arrays was performed as described previously 26 .
  • Probes were designed for exons 1 to 19 of the reference sequence oiAUTS2 (supplementary table S4). Oligonucleotides were synthesized by Integrated DNA Technologies (Leuven, Belgium); all other reagents were from MRC- Holland (Amsterdam, The Netherlands). The samples were separated on a 3730 automated sequencer (ABI Systems) and analyzed using Gene marker vl.95 software (Soft genetics).
  • DNA from patient 20 was sequenced using a customized whole-genome large insert jumping library for Illumina HiSeq2000 sequencing based on the mate-pair methods of Applied Biosystems SOLiD sequencing (Applied
  • Biosystems Inc 23 > 24 .
  • the method sequences the ends of fragments separated by large inserts ( ⁇ 3kb in this case) of genomic sequence, yielding very high physical coverage of the genome by mapping the insert between paired-end reads.
  • 20 ⁇ g of DNA was sheared and size selected around 3.5 kb, cap adaptors with EcoP15I restriction sites were ligated to the ends, and fragments were circularized with an internal oligonucleotide adaptor containing a subject- specific barcode and a single biotinylated thymine.
  • BAM files were processed to identify rearrangement breakpoints by a freely available C++ program, BamStat (http://mappingtools.chgr.org), we developed to tabulate mapping statistics and output lists of anomalous read-pairs (defined as having ends that map to two different chromosomes, an abnormal insert size, or unexpected strand orientations).
  • the breakpoint positions were converted to the GRCh36/hgl8 genome built to allow comparison with other patients in this article. Complete details of all protocols and bioinformatic analysis are available 23 .
  • FISH analysis was used for translocation breakpoint mapping in patient 21.
  • Locus specific FISH probes for chromosome 7qll.22 and 22ql2.1 were selected using the Ensembl and UCSC genome browser database (http://www.ensembl.org,
  • RNA rescue experiments the human wild type mRNAs, full-length and short isoform, were cloned into the pCS2 vector and transcribed in vitro using the SP6 Message Machine kit (Ambion). All the
  • Proband one is born prematurely at 34 weeks of gestation and had a birth weight of 1361 gram ( ⁇ p3). His mother has used alcohol in this pregnancy and is mildly intellectual disabled. His father is diagnosed with schizophrenia. The little boy (now six years and 4 months old) started talking at 2,5 years of age. He is mildly
  • Proband two is a five years and eight months old girl. She is born at term with a normal birth weight. She has a delayed psychomotor development and epilepsy. She had gastro-oesophageal reflux. Physical examination at the age of 5 years revealed borderline short stature (height at p3), microcephaly (headcircumference 45.5cm, ⁇ p2), a nasal deromid sinus and left sided muscle weakness and right sided spasticity. She is said to have soft cleft palate, but this was not objectified. A MRI of the brain was performed, which revealed colpocephaly, corpus callosum agenesis,
  • hypoplasia of the brain stem and an asymmetry of the cerebral hemispheres with a smaller hemisphere and an enlarged ventricle at the right.
  • ophthalmologic evaluation a tear duct hypoplasia and right sided hypoplastic optic nerve were noticed. She has amblyopia and exotropia of the right eye.
  • the proband in family three is a five years and three months old boy that is the first child of non-consanguineous parents. His mother used alcohol in the first two months and antidepressants in all months of this pregnancy. He was born 5,5 weeks early. He had a birth weight of 2353 grams (p50). He has aggressive and autistic behavior. The best fitting diagnose would be pervasive developmental delay with suspicion to progress to Asperger syndrome. Physical examination at the age of 5 years and 3 months revealed a height of 106 cm (p50), a head circumference of 51,5cm (p50) and a weight of 18,5 kg (p3- 10). He has short palpebral fissures, a low nasal bridge with a bulbous nasal tip and a highly arched palate. His neck is short, and he has a shawl scrotum. Examination of his extremities was remarkable for a clinodactyly of his little finger, lax ligaments, fat pad on the pointed finger and an inversion of the foot.
  • Echocardiogram revealed a small patent formamen ovale that closed spontaneously.
  • Microarray revealed two variants: a deletion in intron 2 of the AUTS2 gene (chr7:69134595-69202305) and a duplication of Xp ll. l (FAAH2 and ZXDA, chrX:57356516-57658457).
  • the AUTS2 deletion was confirmed by FISH and high density array (Roche Nimblegen 135K, high density custom designed by... .) ⁇ Both variants were found to be inherited from his mother, who thinks that she herself has Asperger syndrome characteristics.
  • Proband four is the first child of non-consanguineous parents. He was born at term after a normal pregnancy and delivery. Anthropometric measurements at birth were within normal limits. His early developmental milestones were normal. At the age of three years he was noticed to present significant opposite behavior. Now at the age of eight years he has been diagnosed with an autism spectrum disorder (ASD). He is quiet and needs a well structured environment, but overall his communication skills are better than those of his younger brother who has also been diagnosed with ASD. He receives special education, because of his autistic behavior. His intellectual development is delayed, with a mild discrepancy between performal and verbal skills.
  • ASSD autism spectrum disorder
  • Array CGH (Agilent 180K) analysis revealed a de novo intragenic deletion in the AUTS2 gene. No other aberrations were noted. The deletion spans 187 to 242 kb in intron 4 of the largest A UTS2 transcript (and deletes exon 5 of the shorter alternative transcript: AUTS2-003
  • This proband is the third child of non-consanguineous healthy parents.
  • Both parents have no intellectual deficits nor autistic features and followed normal education. Pregnancy and delivery were uneventful. He was born at a gestational age of 41 weeks and 2 days with a birth weight of 4170 gram (p85). Early developmental milestones were mildly delayed. He started walking at the age of 18 months. At the age of 28 months he had a speech development comparable with a 16 month old child. There are (no) features of autism. Now at the age of 36 months, he is functioning at a developmental age of 23 months. Physical examination revealed a height 100 cm (p25) and a head circumference of 50.8 cm (p50). No dysmorphic features are evident (see figure 2A). Physical examination of the father showed no dysmorphic features and normal body proportions. Genetic analysis
  • Array-CGH using a Lowlands consortium custom designed Agilent 105K platform, revealed an intragenic deletion of 150 kb with breakpoints at 68,900,000 (Probe A16_P17972091) and 69,050,000 (Probe A16_P17972516) removing exon 2 of the AUTS2-gene. This deletion leaves the reading frame in tact and causes a deletion of 71 amino acids. The deletion was inherited from his normal farther, who inherited the deletion from his normal mother/father.
  • Patient six is a three year old girl. She is the second child of healthy non- consanguineous parents. The family history is unremarkable. She is born at term after an uneventful pregnancy and delivery. She has a delay in motor and speech development. She could sit up at the age of 10 months and started walking at 18 months of age. She received early intervention services including speech therapy and physical therapy, but has made little progress. Currently, her behavior has been most concerning as she has severe temper tantrums which might be related to her limited ability to express herself. Physical examination at the age of three years revealed a head circumference of 48.5 cm (p25). Height and weight measurements are not available. No dysmorphic features except for a left eye ptosis.
  • Brain MM has been performed and showed, by report, diffuse symmetric white matter abnormality in the periventricular region with some subcortical involvement.
  • Proband 7 is an 2 year and 6 months old boy. He is the second child of healthy non consanguineous parents. His older brother was diagnosed with pervasive
  • This proband is the second child of non-consanguineous parents.
  • the mother of this girl was born with a non-midline upper cleft lip and although she was said to have had normal early childhood development, she struggled at mainstream school with reading and writing and needed extra help.
  • Antenatally, cleft lip and probable cleft palate was detected with ultrasound. She was born at 42 +2 weeks by normal vaginal delivery with birth weight 3.3 kg (p25-50). At birth, the cleft lip and palate were confirmed and a heart murmur was detected. Echocardiography revealed a atrial septal defect with left to right flow.
  • the developmental milestones were delayed with smiling at 4.4 months, sitting up unsupported at 10.5 months, and walking at 22 months. Her speech was also delayed for which she was referred to speech and language therapy.
  • Dysmorphic features were noted consisting of plagiocephaly, brachycephaly, facial asymmetry with the right eyes appearing larger than the left (which improved with age), left sided unilateral cleft lip repair, prominent forehead with thin lips, a relatively flattened nasal tip, short upslanting palpebral fissures and proptosis, overlapping 2 nd , 3 rd and 4 th toes, capillary haemangioma over the occipital region of the head and over the sacrum.
  • Heart sounds confirmed the presence of a quiet mid- systolic murmur over the left sternal edge.
  • Her mother had evidence of her cleft lip repair, ptosis, retrognatia and a narrow mouth.
  • Array analysis performed using the Affymetrix 2.7M array showed an intragenic AUTS2 deletion.
  • the deletion spans 52 kb and deletes exon 3 and 4 of the AUTS2 gene (chr7:69,211,000-69,263,000).
  • Q-PCR analysis the deletion was shown to be inherited from her mother. Mosaicism in the mother cannot be excluded. We are awaiting samples to arrive from the maternal grandparents for further analysis.
  • Patient 9 de novo deletion overlapping exon 1-4 of the AUTS2 gene, no other genes involved
  • Proband nine is born at term. She is the first child of healthy non-consanguineous parents. Family history reveals no relevant diseases, birth defects or developmental problems. Pregnancy and delivery were unremarkable. Her birth weight was: 3090g (pl0-p25), and her head circumference at birth was 32 cm (p3). Her early motor milestones were obtained at a normal age; she was rolling over at six months, sitting alone at seven months and crawling at eight to nine months. Around the age of 15 months, however, a delay in development and signs of autism were noted. She was tested to have a developmental level of 5-9 months in most areas as well as decreased eye contact and was given the diagnosis autism. She has a history of low muscle tone and feeding problems, including reflux and constipation. She is sensitive to loud sounds.
  • Neurological evaluation revealed normal muscle tone. A MM scan of the brain (without contrast) was normal.
  • Array CGH (Signature OS Chip) revealed a de novo microdeletion of 2.3 Mb at 7ql l.22, overlapping exon 1-4 of the AUTS2 gene (chr7:67056711-69323591). The deletion is not overlapping any other genes and was confirmed by FISH and custom high density array.
  • the proband in this family is a little girl of now six years of age that is the second born child of non-consanguineous parents.
  • the mother has had learning difficulties and feeding difficulties. She has short stature and mild dysmorphic features.
  • the pregnancy of the proband was complicated by intrauterine growth retardation and polyhydramniom. She is born by normal vaginal delivery. Her birth weight was 2.38 kgs (p2-9).
  • Her height and head circumference were below the 0.4 th centile. She is hyperteloric, has a ptosis and a downslant of the palpebral fissures, a mild proptosis and has highly arched eyebrows. She has a broad nasal bridge, a bulbous nasal tip, simple ears, low columella, strabismus and retrognatia. Her older brother, now ten years old, had a similar antenatal and postnatal course. His birth weight was 2.26 kg (p0.4-2). He fed poorly and failed to thrive with difficulty weaning onto solids. His early developmental milestones were delayed. He smiled at four to five months, crawled at 18 months and walked at two years.
  • Proband eleven is a 5,5 year old boy. He is born after an uneventful pregnancy and had a birth weight of (p ). He did not begin to talk until he was 3 years of age. He is hyperactive and shows a global development delay. He is now able to talk in three to four word sentences. Physical examination at the age of 5 years and 6 months reveals a short stature (height: 106.5 cm, p lO) a weight of 14.7 kg (p25) and microcephaly (head circumference: 45.5 cm «p2, p50 for a 9-month-old).
  • Chromosome analysis revealed a normal male karyogram: 46, XY. Methylation testing for Angelman syndrome was normal and Fragile X DNA testing was normal too.
  • Array CGH Alent 105K identified an intragenic AUTS2 deletion (chr7:69612584- 69827896) deleting exon 6. This deletion causes the AUTS2 sequence to go out frame from exon 7 onwards. It is unknown if the deletion occurred de novo as the parents are not yet tested.
  • Proband 12 was born at term and has healthy non-consanguineous parents.
  • hyperteloric has epicanthal folds and micrognatia. His ears are large and low set, and he has large hands and feet. Neurologic examination was remarkable for increased deep tendon reflexes and hypertonia of both legs. A MM of the brain was normal.
  • Array CGH (Roche NimbleGen 135k, a Signature Genetics custom design) was performed and showed a small intragenic deletion of the AUTS2 gene involving exon 6 (chr7:69654259-69828289). This deletion causes the AUTS2 sequence to go out frame from exon 7 onwards. The deletion was visualized by FISH and confirmed by high density array (custom design Roche Nimblgen 135K). Both parents of the boy tested negative for the deletion, which implies the deletion to be de novo.
  • Proband 13 is the first child of non-consanguineous parents. She was born at a gestational age of 42 weeks after an uneventful pregnancy, but had a slow start (with an apgar score of 5 after 1 minute). Blood gas parameters were normalised one hour after birth. She had a birth weight of 3690 gram (p55). In the neonatal period she was tachypnoeic, hypotonic and inactive. In the first months of her life she had feeding difficulties, failure to thrive and needed tube feeding. Physical examination at that time revealed low muscle tone, an upslant of the palpebral fissures, strabism and an expressionless face. At childhood (7- 10 years) she suffered from recurrent upper airway infections.
  • Proband 14 is a 2 years and 11 months old girl with developmental delay. Family history was unremarkable. She was born at term and had a birth weight of 2,55 kg ( ⁇ p3) She had an inguinal hernia, which was successfully operated at the age of 3 months. At the age of 5 months there was a choking incident for which she was evaluated in hospital. TORCH- and metabolic screen was normal, as was the EEC. At the age of 11 months an infected preauricular sinus was operated. Although autism was suspected at a younger age, during evaluation at the age of 25 months there was no clinical suspicion of autism but she had a mild global developmental delay of 3/9 months.
  • Her tone and reflexes were normal throughout, with only mild restriction of dorsiflexion in left ankle remaining. Brain MRI at 13 months showed nonspecific hyper intense signal changes in the white matter in the periventricular zones at fron to-parietal areas. The lateral and third ventricles were prominent. MRS and MRA were normal.
  • Rett syndrome testing (by sequencing of MeCP2) was negative.
  • Array analysis using Affymetrix SNP array 6.0 showed a deletion of 7qll.22.
  • the minimal deletion spans 330 kb of the AUTS2 gene and includes exons 6- 11 (chr7:69,544,241-69,874,867).
  • the deletion was confirmed to be de novo by Q-PCR and FISH of the proband and her parents.
  • the 15th patient is an 1 year and 11 months old girl with a mild developmental delay. She was born at term and had a normal birth weight. Her medical history was remarkable for a metopic suture stenosis. She is mildly delayed in general
  • microcephaly with a head circumference of 44 cm, ( ⁇ p l). She had some mild dysmorphic features, namely elevated eyebrows, proptosis and epicanthal folds.
  • Array CGH (Agilent 105k custom designed by Signature Genetics) revealed an intragenic microdeletion in AUTS2, deleting at least exon 6 (chr7:69699592- 69828036). The deletion was confirmed by FISH and High density Array (custom design Roche Nimblgen 135K). With High density array it was clarified that the deletion contains exon 6 to 18 of the AUTS2 gene.
  • Patient 16 (deletion exon 7-19, de novo):
  • Proband 16 is a 7 years and 2 months old girl. This little girl was born premature. In the neonatal period an intraventricular hemorrhage was noticed. She had some feeding difficulties and development was delayed. She has had one febrile seizure, but no signs of epilepsy at this moment. At the age of 6 years she was almost non-verbal, using some signs and communication aids. She was functioning at a developmental level of a 2.5 years old child. She meets the criteria for PDD-NOS. She is a picky eater and just stays inside the normal range with her weight with a lot of effort from her mom. Physical examination at the age of 6 years revealed a height of 110.7 cm (p20), her weight was 17.5 kg (plO), and her head circumference 50 cm (p50).
  • Metabolic screening revealed an elevated C5-DC acylcarnitine, which suggests the diagnosis of glutaric aciduria type I (GA1). This was however excluded with DNA testing of the Glutaryl Co-A Dehydrogenase gene. MRI of the brain showed somewhat large ventricles and a mammillary body atrophy. No white matter abnormalities were observed. There was no evidence of neonatal brain damage.
  • Microarray was performed (using the Agilent 244K array CHG) and demonstrated a deletion of exon 7-19 of the AUTS2 gene (Chr7:69842425-69895671). Additional testing did not show this deletion in both parents, indicating that it is a de novo deletion.
  • the 17th patient is a boy of now 5 years and 7 months of age. He is the child of non- consanguineous healthy parents. His mother is born premature and had Rhesus disease at birth and a developmental delay. He was born full term and had a birth weight 3520 gram (p50). Pregnancy and delivery were uneventful. He has been successfully operated for an umbilical and an inguinal hernia. A g-tube was placed, because of significant failure to thrive. The developmental milestones were
  • Cardiovascular examination revealed a murmur that appeared to be functional in nature and a hemodynamically stable patent foramen ovale with small left to right shunt. The boy was diagnosed with mild cerebral palsy with severe cognitive deficits. MRI results revealed a mild prominence of the ventricular system and extra-axial spaces but were otherwise unremarkable.
  • Proband 18 is a 9 years and 1 month old girl. She was the third child of healthy non- consanguineous parents. She has two healthy siblings. Apart for seizures treated with medication in a paternal cousin there are no relevant disorders in the family. The pregnancy was uncomplicated except for small fetal size. At birth, she weighted 2523 gram and was determined to be small for gestational age. Weight gain was followed closely and she did well. She has a developmental delay with relative strength in gross motor skills. She has a history of hypersensitivity to loud sounds. Physical examination reveals a head circumference of 50.5 cm (p25). Her height was 131 cm and weight was 27.3 kg, both at the 30th percentile for age. She has a large tongue and tends to hold her mouth open.
  • She has a flat nasal bridge, incurving 5th finger on the left and very small fingernails. She shows asymmetric developmental posturing on stressed gaits (left-sided). Muscle and strength were normal. Heel cords were slightly tight but both ankles could be flexed to neutral position. Tone throughout was otherwise within normal limits. Deep tendon reflexes were 2+ throughout and symmetrical. Toes were down-going.
  • Array CGH (Agilent 244K) revealed a deletion of 2.4 mb, deleting AUTS2 in total and part of WBSCR17 (chr7:67, 771,230-70,363,865). Parental analysis revealed the deletion to be de novo. (This patient was submitted to the Decipher database, https://decipher.sanger.ac.uk/patient/251781, 9 November 2011) Patient 19 (de novo deletion of the complete AUTS2 gene and the WBSCR17 and CALN1 genes)
  • Patient 19 involves a little boy of now 4 years old. He was born at term after a normal pregnancy and delivery and had a good start. His birth weight was 3658 gram (p50). Because of severe feeding problems he was admitted to the special care baby unit. He needed tube feeding. He was then discovered to have a diaphragm eventration for which he was operated when he was two weeks old. Cardiologic evaluation revealed a atrium septum defect. Further more distal hypospadias was noticed. Because of severe feeding problems a percutaneous endoscopic gastrostomy (PEG) tube was placed. On physical examination at the age of 21 months his height is 81 cm (p9), his weight 11, 1 kg (p9-25) and his head circumference 47,6 (p2). He is slightly
  • hyperteloric has short upslanting palpebral fissures, a broad nasal bridge, a short philtrum, a narrow mouth and a micrognatia. He is hypotonic. Evaluation at the age of two years revealed that he is still struggling with his feeding and is still being fed through his PEG tube. He seems to be delayed in motor and speech development. He was cruising but not walking yet and he had about 10-15 words. He is sensitive to loud sounds. He was tip toeing especially on the left and an increased tone around his left ankle and slightly brisk reflexes were noted at that time. Further neurologic examination was unremarkable. During re-evaluation at three years of age he showed improvement. He does however show some evidence of dystonic stiffness of his left ankle. Dysmorphic features were more or less unchanged. An MM of the brain showed no abnormalities.
  • Neurological evaluation the reflexes are present at knees, but hard to obtain in upper limbs and ankles. She has mild dysmorphic features, namely: short palpebral fissures with a mild upslant and a short philtrum and narrow mouth. Genetic analysis:
  • Chromosomal analysis revealed a paracentric inversion of chromosome 7
  • the 21st proband is the first child of non-consanguineous healthy parents.
  • the pregnancy and delivery were uneventful. She was born at 37 weeks of gestation and had Apgar scores of 9 and 10 after 1 and 5 minutes, respectively.
  • birth weight was 2740 grams (p30). Due to hypoglycemia and feeding problems she was admitted in the neonatal intensive care unit for three weeks. Feeding remained difficult during the first months after birth. At the age of five months she had an axial hypotonia and a failure to thrive. Physical examination at that age revealed mild dysmorphic features; low-implanted ears, highly arched eyebrows, a broad nasal bridge, hypertelorism, short palpebral fissures and epicanthic folds and a full lower lip (see figure 2D).
  • chromosomes a systematic series of FISH analyses was performed. On chromosome 7, the clones RP11-689B18 (location on chr7:69996264- 70178753, including exon 6) map proximally and RP11-575M4 (location on chr7: 70185711-70372702, including exons 7-19) spans the breakpoint.
  • G248P84061D2 all map distally to the breakpoint, clearly indicating that the translocation interrupts the AUTS2 gene, starting in intron 6 of the gene.
  • clones RP11-699H18 and RP11-772E17 flank the breakpoint, while clone RP11-1056M20 spans the breakpoint (table S4).
  • Ref ID 94 Livak,K.J. & Schmittgen,T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2(- Delta Delta C(T)) Method.

Abstract

The disclosure relates to methods of diagnosing and treating autism. The disclosure provides diagnostic methods based on identifying alterations in the AUTS2 gene, in particular in the AUTS2 short isoform. The disclosure further relates to therapeutic interventions utilizing the newly identified alternative transcript.

Description

Title- COMPOSITIONS AND METHODS FOR DIAGNOSING AND TREATING INTELLECTUAL DISABILITY
SYNDROME, AUTISM AND AUTISM RELATED DISORDERS
FIELD OF THE INVENTION
The disclosure relates to methods of diagnosing and treating intellectual disability (ID) syndrome, autism and autism related disorders. The disclosure provides diagnostic methods based on identifying alterations in the AUTS2 gene, in particular in exons 9- 19 of the AUTS2 gene and preferably in the AUTS2 short isoform. The disclosure further relates to therapeutic interventions utilizing the newly identified alternative transcript.
BACKGROUND OF THE INVENTION
Autism is a neuropsychiatric disorder characterized by abnormal social behavior and communication skills associated with repetitive behavior. It belongs to the group of pervasive developmental disorders ("autism spectrum" disorders) which includes autism, Asperger syndrome, Childhood disintegrative disorder, and Pervasive
Developmental Disorder Not Otherwise Specified. The Diagnostic and Statistical Manual of Mental Disorders (DSM IV) uses the following criteria to diagnose autism: (I) A total of six (or more) items from (A), (B), and (C), with at least two from (A), and one each from (B) and (C)
(A) qualitative impairment in social interaction, as manifested by at least two of the following:
1. marked impairments in the use of multiple nonverbal behaviors such as eye-to-eye gaze, facial expression, body posture, and gestures to regulate social interaction
2. failure to develop peer relationships appropriate to developmental level
3. a lack of spontaneous seeking to share enjoyment, interests, or achievements with other people, (e.g., by a lack of showing, bringing, or pointing out objects of interest to other people)
4. lack of social or emotional reciprocity ( note: in the description, it gives the following as examples: not actively participating in simple social play or games, preferring solitary activities, or involving others in activities only as tools or
"mechanical" aids ) (B) qualitative impairments in communication as manifested by at least one of the following:
1. delay in, or total lack of, the development of spoken language (not accompanied by an attempt to compensate through alternative modes of communication such as gesture or mime)
2. in individuals with adequate speech, marked impairment in the ability to initiate or sustain a conversation with others
3. stereotyped and repetitive use of language or idiosyncratic language
4. lack of varied, spontaneous make-believe play or social imitative play appropriate to developmental level
(C) restricted repetitive and stereotyped patterns of behavior, interests and activities, as manifested by at least two of the following:
1. encompassing preoccupation with one or more stereotyped and restricted patterns of interest that is abnormal either in intensity or focus
2. apparently inflexible adherence to specific, nonfunctional routines or rituals
3. stereotyped and repetitive motor mannerisms (e.g. hand or finger flapping or twisting, or complex whole-body movements)
4. persistent preoccupation with parts of objects
(II) Delays or abnormal functioning in at least one of the following areas, with onset prior to age 3 years:
(A) social interaction
(B) language as used in social communication
(C) symbolic or imaginative play.
Recent studies suggest that autism often has a genetic cause or background.
Classically, discovery of causative genes has involved family -based linkage, case- control association studies, copy number variants or translocations observed in affected patients. The identification of a gene involved in the predisposition to autism/ID offers the potential to develop a diagnostic tool. Diagnosis of autism is currently based on behavior and while there is no cure, there is some evidence that early behavioral training and/or antipsychiotic medication may lessen the associated deficits. It is an object of the present disclosure to provide methods and compositions for the diagnosis and treatment of autism, autism related disorders and intellectual disability (ID) (both syndromal and non-syndromal). It is also an object of the present disclosure to provide methods and compositions for developing and using in vitro and in vivo models for diagnosis, treatment, as well as testing.
SUMMARY OF THE INVENTION
In one aspect of the disclosure, a method is provided of classifying an individual comprising detecting an alteration in an AUTS2 gene of said individual. Preferably, the presence of an alteration in the AUTS2 gene indicates that the individual has or is at risk of developing AUTS2 syndrome. Preferably the alteration is a chromosome abnormality, more preferably an intragenic deletion. Preferably, said individual is first selected as having at least one symptom of the AUTS2 syndrome. Preferably, AUTS2 syndrome includes autism and intellectual disability. A preferred form of intellectual disability (syndrome) is Rett syndrome.
Preferably the alteration is located between exons 9- 19 (inclusive of exons 9 and 19) of the AUTS2 gene. Preferably the alteration affects the AUTS2 short isoform, e.g., by altering expression or modifying the amino acid sequence. In some embodiments, the alteration located between exons 9-19 or affecting the AUTS2 short isoform indicates the severity of the AUTS2 syndrome.
In preferred embodiments the detection is performed using multiplex ligation dependent probe amplification (MLPA). Preferably said MLPA is performed using one or more probe pairs selected from MLPA 102, MLPA 110, MLPA 118, MLPA 126, MLPA 130, MLPA 096, MLPA 096, MLPA 136, MLPA 140, MLPA144, MLPA 100, MLPA 108, MLPA 112, MLPA 116, MLPA 120, MLPA 124, MLPA 128, MLPA 132, MLPA 134, MLPA 105, MLPA 138, MLPA 107, and MLPA 136. The present disclosure also provides multiplex ligation dependent probe amplification (MLPA) probe sets specific for the AUTS2 gene.
In a further aspect of the disclosure, a polypeptide comprising an AUTS2 sequence having at least 95% identity to SEQ ID NO:2 is provided. Preferably wherein the N- terminus of said amino acid sequence is encoded by exon 9 of the AUTS2 gene. In a further aspect of the disclosure, an isolated nucleic acid encoding said polypeptide is provided. In a further aspect of the disclosure, a vector comprising said nucleic acid is provided provided. Preferably, the vector is for use in gene therapy, preferably in the treatment of AUTS2 syndrome.
The disclosure further provides a method of treating an individual in need thereof comprising administering a therapeutically effective amount of a composition comprising the polypeptides, nucleic acids, vectors, or cells as described herein.
In a further aspect of the disclosure, an anti-AUTS2 antibody which specifically binds to the AUTS2 short isoform is provided.
In a further aspect of the disclosure, a mammalian cell comprising a nucleic acid or vector as described herein is provided.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1: Overview oiAUTS2 aberrations. Overview of all AUTS2 aberrations in the probands that were included in this study. The location of the deletions is indicated by the bars, the inversion breakpoint is indicated by an arrowhead, the area in which the translocation breakpoint is located is indicated by '-' . On the right you can appreciate a visualization of the AUTS2 syndrome severity score, where the darker shades indicate a more severe and/or more specific phenotype (lightest gray: <6; light gray: 6- 10; gray: 11-15; dark gray: 16-20; darkest gray >20).
Figure 2: Pictures of the AUTS2 aberration patients, a: Patient 5 at the age of 3 years shows no evident dysmorphic features, b+i: Patient 8 at the age of 2,5 years has a repaired cleft lip, mild proptosis, short and mild upslanting palpebral fissures, c: The mother of patient 8 also shows a repaired cleft lip, ptosis and retrognatia. d:
Patient 9 at the age of 3 years shows highly arched eyebrows, mild downslanting palpebral fissures, epicanthal folds and a short philtrum. e+m: patient 10 at the age of about 6 years. She is hyperteloric, has a ptosis and down slanting palpebral features, a short philtrum and narrow mouth as does her brother shown in f+n at the ages of 10 years, g+o: Patient 13 at the age of 32. You can appreciate hypertelorism, proptosis, upslanting palpebral fissures, a short upturned philtrum an a narrow mouth, h:
Patient 14 at the age of 2 years. She shows a prominent nasal tip, anteverted nares and short philtrum. i+p: Patient 17 at the age of 5,5 years. He has a hypertelorism, ptosis, a broad nasal bridge, a short and upturned philtrum and a narrow mouth, j+k+q: Patient 19 at the age of about 1 year (j) and 4,8 years (k). He has a broad nasal bridge, short palpebral fissures and a short philtrum and narrow mouth.
Figure 3: Evolutionary conservation of exons that constitute the main AUTS2 transcript in humans and a novel transcriptional start site (TSS) detected in exon 9 of human AUTS2 using 5'RACE to amplify capped 5' mRNA ends in a human brain mRNA sample. AUTS2 orthologs were detected in gorilla (gorGor3), macaque (Mmul_l), dog (Broadd2), cow (Btau_4.0), pig (Sscrofa9), mouse (NCBIM37), chicken (Washuc2), clawed frog (JGI_4.2) and zebrafish (Zf9) by tblastn searches using the human amino acid sequence as a query against the corresponding genome databases, a) Compared exon-intron organization oiAUTS2 orthologs in humans and zebrafish. Red arrows indicate the location of two transcriptional start sites (TSS) used in human brain mRNA. The alternative TSS is located 1, 17 Mbp downstream in the cluster that contains exons 7- 19. b) Phylogenetic tree based on clustalW2 alignment of the full-length amino acid sequences11, c) To assess the conservation of individual exons, the percentage amino acid identity was calculated. For comparison, conservation of the full-length protein is given in the last column. Italic numbers indicate the number of amino acids encoded by each human exon. Grey numbers correspond to cases where conserved sequences are not included in the longest annotated protein (the respective accession numbers are:
ENSGGOP00000011519, ENSMMUP00000023254, ENSBTAP00000002697, ENSSSCP00000008253, ENSCAFP00000016549, ENSMUSP00000062515,
ENSGALP00000001729, ENSXETP00000007747, ENSDARP00000073379). A complete multiple sequence alignment is provided in the supplementary data, d) Sequence analysis of a novel AUTS2 transcript amplified from human brain mRNA by 5' RACE (rapid amplification of cDNA ends). The alternative transcript started in the center of exon 9 (asterisk) and contained the indicated cDNA sequence (italic font). The mRNA was spliced to exon 10 using the second of two known splice donor sites in exon 9, thus resulting in the incorporation of 7 alternatively spliced amino acids (rectangle). The alternative mRNA uses the same reading frame as the conventional transcript. Conventional exons are in uppercase, introns in lowercase. Figure 4: Suppression of auts2 in zebrafish leads to head size defects.
a) Lateral views of representative control embryos and embryos injected with AUTS2 morpholinos (MO), b) Quantification of microcephaly in embryo batches injected with AUTS2 morpholinos: 4.5 ng 5'MO (targeting exon 2 donor splice) or 6 ng 3'MO (targeting exon 10 donor splice) plus 100 pg wild type human AUTS2 full length (PL) or short isofhrm (3') niRNAs (n~ 56-91 embryos per injection). P-values are denoted on the bar graph. NS, non-significant. Figure 5: Amino acid alignment oiAUTS2 orthologs. Amino acid alignment (determined by ClustalW2) of human AUTS2 orthologs in gorilla, macaque, dog, cow, pig, mouse, chicken, clawed frog and zebrafish. Exon boundaries are indicated above the alignment. Fully conserved or similar residues are marked by symbols at below the alignment. Grey lowercase letters indicate low-complexity or repetitive segments of the human amino acid sequence according to the XNU + SEG algorithms11 13.
Figure 6: NM_015570 Homo sapiens autism susceptibility candidate 2 (AUTS2) mRNA
DETAILED DESCRIPTION OF THE DISCLOSED EMBODIMENTS
The autism susceptibility candidate 2 (AUTS2) gene was identified in a translocation breakpoint analysis in twins with autism, developmental delay and epilepsy1. The authors of this study concluded that AUTS2 is unlikely an autism susceptibility gene for idiopathic autism. Subsequently, five patients with a translocation breakpoint and one patient with an inversion breakpoint disrupting AUTS2 were described; these patients have intellectual disability (ID) and/or developmental delay (all), autism features (in four reported cases), epilepsy and skeletal abnormalities (reported in three cases each) 2 4 . In addition, WO2009/089464 describes a patient with social and cognitive delays having a chromosomal inversion affecting the AUTS2 and the CNTNAP2 gene.
Due to the small number of patients examined and large extent of the chromosomal abnormalites in these patients, it has not been possible to decisively conclude that disruption of the AUTS2 gene is responsible for the symptoms in these patients.
Disruptions in either neighboring genes or distantly located genes (e.g., resulting from the translocation of a chromosomal segment comprising AUTS2) may also be responsible for the patient phenotypes. In particular, it was not known that an intragenic deletion in AUTS2 has any affect on neurodevelopment.
The function of the protein encoded by AUTS2 is largely unknown. The longest transcript oiAUTS2 has 19 exons and spans 1.2 Mb. The wild-type human AUTS2 gene sequence corresponds to Gene ID: 26053 located on human chromosome 7ql l.22. It encodes a 1,295 amino acid protein that contains 2 potential proline-rich protein- binding domains1. The mouse Auts2 ortholog is highly expressed in the developing mouse brain, specifically in the frontal cortex, the hippocampus and the cerebellum— areas known to be affected in autistic patients. The Auts2 protein is located in the nucleus and co-localizes in the cortex with Tbrl, a transcription factor critical for corticogenesis8.
Recently, the 5' end (exon 1-4) oiAUTS2 was flagged for having the strongest statistical signal for a putative positive selective sweep in early modern humans7. In this region the frequency of derived alleles for human polymorphic sites is less than expected in Neanderthals. This suggests that different parts of the AUTS2 gene may have been subjected to different selective pressures during hominid evolution compared to Neanderthals. The present invention is based, in part, on results which conclusively demonstrate that that disruption of the AUTS2 gene is associated with a complex phenotype that includes not only autism, but also a series of anatomical and neurodevelopmental defects including intellectual disability, which are referred to herein as AUTS2 syndrome. Table 1 provides the phenotypes associated with AUTS2 syndrome and their prevalence. Embodiments of the invention are also based on the discovery that the AUTS2 gene encodes an alternatively spliced isoform in the human brain. The inventors further demonstrate that disruptions in the genomic region encoding the alternatively spliced isoform are associated with more severe form of AUTS2 syndrome.
As used herein, AUTS2 syndrome is characterized by intellectual disability, microcephaly, mild short stature, feeding difficulties, hypotonia, cerebral palsy and dysmorphic features. It is understood that an affected individual will usually not present with all of the symptoms and may show varying degrees of severity for each symptom. Preferably, AUTS2 syndrome is characterized by at least two symptoms selected from intellectual disability, microcephaly, mild short stature, feeding difficulties, hypotonia, cerebral palsy and dysmorphic features. Preferably, AUTS2 syndrome is characterized by at least the presence of autism as defined by the DSM IV. In preferred embodiments of the disclosure, AUTS2 syndrome is synonymous with autism. In another preferred embodiment the AUTS2 syndrome is synonymous with intellectual disability.
One aspect of the disclosure provides a method of classifying an individual comprising detecting an alteration in an AUTS2 gene of said individual. Alterations may be detected at the level of DNA, RNA, or protein, preferably said alteration is detected in a nucleic acid. Said methods may detect a change in expression level (including haploinsufficiency caused either by a deletion or by nonsense mediated decay) or change in amino acid sequence. Alterations include the insertion or deletion of a single nucleotide, or of more than one nucleotide, resulting in a frame shift; the change of at least one nucleotide, resulting in a change in the encoded amino acid; the change of at least one nucleotide, resulting in the generation of a premature stop codon; the deletion of several nucleotides, resulting in a deletion of one or more amino acids encoded by the nucleotides; the insertion of one or several nucleotides, such as by unequal recombination or gene conversion, resulting in an interruption of the coding sequence of a reading frame; duplication of all or a part of a sequence; transposition; or a rearrangement of a nucleotide sequence. Such sequence changes can alter the polypeptide encoded by the nucleic acid. For example, if the change in the nucleic acid sequence causes a frame shift, the frame shift can result in a change in the encoded amino acids, and/or can result in the generation of a premature stop codon, causing generation of a truncated polypeptide. An alteration can also alter splice sites, affect the stability or transport of mRNA, or otherwise affect the transcription or translation of an encoded polypeptide. It can also alter DNA to increase the possibility that structural changes, such as amplifications or deletions, occur at the somatic level. These alterations can be detected by any number of methods known to a skilled person at the level of DNA, RNA, or protein.
Detection of the expression levels can be performed using immunological assays, including, Western Blot, immunohistochemistry, FACS analysis, radio immuno assay (RIA), immunofluorescence. Methods for the detection of particular mRNAs are well known and include, for example, hybridization assays using complementary DNA probes (such as in situ hybridization using labeled riboprobes, Northern blot and related techniques) and various nucleic acid amplification assays (such as RT-PCR using complementary primers specific for the AUTS2 short isoform, and other amplification type detection methods). Methods for detecting mutations at the level of DNA are well-known in the art and include Southern Blot hybridization, comparative genomic hybridization (CGH), SNP arrays, MLPA, next generation gene sequencing and DNA sequencing. Preferably the alteration is a chromosomal abnormality. Chromosome abnormality refers to a deviation between the structure of the subject chromosome and a normal homologous chromosome. The term "normal" refers to the predominate structure or nucleic acid sequence found in healthy individuals. Chromosome abnormalities include insertions, deletions, duplications, translocations, and inversions. Preferably, the abnormality is an intragenic deletion of the AUTS2 gene. As is clear to a skilled person, an intragenic deletion is a less severe chromosomal abnormality than, e.g., large deletions or inversions. While duplications, translocations, and inversions, can affect several genes, the effects of an intragenic deletion are normally confined to the specific gene. Preferably the intragenic deletion is less than 150kb, more preferably less than lOOkb.
The method of the invention permits the classification of an individual as to the risk of developing AUTS2 syndrome or of the likelihood of suffering from AUTS2 syndrome. The method of the invention also permits the classification of an individual as to the risk that an offspring of said individual may be of risk of developing AUTS2 syndrome. The current diagnosis of autism and autism related disorders is based on behaviour. This manner of diagnosis suffers from the disadvantage that the symptoms may widely vary among individuals and that the diagnosis can be complicated by other conditions, such as blindness, which can result in difficulties in socializing or communicating. The method of the invention provides objective criteria by which to assess the presence or risk of developing AUTS2 syndrome. The presence of at least one alteration in the AUTS2 gene is an indicator that the individual is at risk of developing AUTS2 syndrome, is likely suffering from AUTS2 syndrome, or is at risk of producing an offspring having AUTS2 syndrome.
The assessment of risk of developing or likelihood of suffering from AUTS2 syndrome may also include determining whether the individual has intellectual disability, multiple congenital anomaly (MCA), or difficulties in social skills, language and behaviour that are known to be associated with autism. The method of the invention can be used in combination with symptomatic diagnostic criteria, e.g. based on DSM IV autism diagnosis. Both the risk of and likelihood of having AUTS2 syndrome also increase when an individual also exhibits one or more of the following features: microcephaly, mild short stature, feeding difficulties, hypotonia, cerebral palsy and dysmorphic features. The risk of and likelihood of having AUTS2 syndrome may also increase when an individual does not have any symptoms, but has a family history of autism.
Any individual may be classified using a method of the invention. It is preferred that said individual is tested as soon as possible, preferably before or at birth. In another preferred embodiment the individual is first selected based on behavioural criteria, such as having at least one symptom of the AUTS2 syndrome. Preferably, an individual is selected who presents with intellectual disability, multiple congenital anomaly (MCA), or difficulties in social skills, language and behaviour that are known to be associated with autism. Preferably the individual has been diagnosed with autism using symptomatic diagnostic criteria, e.g. based on DSM IV. Preferably, said individual also exhibits one or more of the following features: microcephaly, mild short stature, feeding difficulties, hypotonia, cerebral palsy and dysmorphic features. Preferably the AUTS2 syndrome is characterized by intellectual disability and at least one other symptom selected from microcephaly, mild short stature, feeding difficulties, hypotonia, cerebral palsy, and dysmorphic features. In some
embodiments, the individual does not have any symptoms but has a family history of autism. Preferably, said individuals are humans and may be adults, children, or fetuses.
The present disclosure also demonstrates that A UTS2 encodes, in addition to the full- length message, a shorter isoform in the human brain. This shorter isoform uses an alternative transcription start site in the center of exon 9 followed by a start codon in the same reading frame as the full length AUTS2 transcript. In contrast to the 5'region of the predicted protein, the sequence of the short isoform is highly conserved across species. Consistent with a critical functional role for this isoform, deletion of this region is associated with significantly more severe phenotypes.
The nucleic acid coding sequence of the shorter isoform is:
atgccgacgccagcacctcccatgtttgacaaataccctacaaaagttgacccattctaccggcacagtctcttccattcctat cctcctgcagtgtcgggcatcccccctatgatcccacccactggcccttttggttcactacaaggagcatttcagccgaagttg acagatcctttcagacctatgttaaggaaaccagggaagtggtgtgctatgcatgttcacatcgcctggcagatttaccacc accaacagaaagtcaagaaacagatgcagtcagacccacataagctggactttggactgaaacctgagttcctgagccgc cctccaggccccagtctttttggagccatccaccacccccatgacctggcacggccttcaactttgttctctgccgctggtgctgc acacccaactgggaccccttttgggccacctcctcatcacagcaacttcctcaaccctgctgcccacctagagccttttaatcgg ccgtctacattcacaggcctagcagcagttggtggcaatgccttcgggggacttggaaatccttccgttacacccaactcaat gttcggccacaaggatggccccagtgtgcagaactttagcaaccctcacgaaccctggaaccggctgcaccgaacgcctccg tcgttcccgacccctccgccctggctgaagccaggggagctggagcgcagcgcgtccgctgcagctcatgacagagatagag atgtagataaacgagactcatctgttagtaaagatgacaaagaaagggaaagcgtcgagaagagacactccagccaccc ttcaccagcacctgtcctcccggtgaatgccctgggacatacccgcagctccactgaacagatccgggctcatctgaacactg aggctcgggagaaggacaaacccaaagagagggagagagaccactcggaatcccgcaaggacctggccgccgacgagc acaaggcgaaagagggccacctgcccgagaaggacgggcacggccacgaggggcgcgccgcgggcgaagaggccaagc agctggcccgggtgccgtctccctacgtgcggaccccggtggtggagagtgccaggcccaacagcacctcgagccgggagg ccgagccgcgcaagggtgagccggcctacgagaaccccaagaagagctccgaggtcaaggtgaaggaggagcggaagg aagaccatgacctgcctccagaggccccgcagacccaccgggcctcggagccgccgcctcccaactcctcgtccagcgtgcac ccggggcccctggcctcgatgcccatgacggtgggggtgacgggcattcaccccatgaacagcatcagcagcctggacagg actcgcatgatgacccccttcatgggcatcagccccctcccgggcggagagcgcttcccgtacccttctttccactgggacccc atccgggaccccttgagggatccttaccgagaacttgacattcaccggagagacccgctgggcagggacttcctgctaagg aacgacccgctccaccggctctcgactccccggctgtacgaagccgaccgctccttcagggaccgggagcctcacgactaca gccaccaccaccaccaccaccaccacccgctgtctgtggaccctcggcgggagcacgagcggggaggccacctggacgagc gggagcgcttgcacatgctcagagaagactacgagcacacgcggctccactccgtgcaccccgcctccctcgacggacacct cccccaccccagcctcatcaccccgggactccccagcatgcactatccccgcatcagccccaccgcgggcaaccagaacggac tcctcaacaagacccctccgacagcagcgctgagcgcacctcccccgctcatctccacgctggggggccgcccggtctctccca gaaggacgactcc tctgtccgcagagataagggagaggcccccttcccacacgctgaaggatatcgaggcccgataa (SEQ ID N0:1).
The protein sequence of the shorter isoform is predicted to be:
MPTPAPPMFDKYPTKVDPFYRHSLFHSYPPAVSGIPPMIPP TGPFGSLQGAFQPKLTDPFRPMLRKPGKWCAMHVHIAWQ IYHHQQKVKKQMQSDPHKLDFGLKPEFLSRPPGPSLFGAI HHPHDLARPSTLFSAAGAAHPTGTPFGPPPHHSNFLNPAA HLEPFNRPSTFTGLAAVGGNAFGGLGNPSVTPNSMFGHKD GPSVQNFSNPHEPWNRLHRTPPSFPTPPPWLKPGELERSA SAAAHDRDRDVDKRDSSVSKDDKERESVEKRHSSHPSPAP VLPVNALGHTRSSTEQIRAHLNTEAREKDKPKERERDHSE SRKDLAADEHKAKEGHLPEKDGHGHEGRAAGEEAKQLAR VPSPYVRTPVVESARPNSTSSREAEPRKGEPAYENPKKSS EVKVKEERKEDHDLPPEAPQTHRASEPPPPNSSSSVHPGP LASMPMTVGVTGIHPMNSISSLDRTRMMTPFMGISPLPGG ERFPYPSFHWDPIRDPLRDPYRELDIHRRDPLGRDFLLRN DPLHRLSTPRLYEADRSFRDREPHDYSHHHHHHHHPLSV DPRREHERGGHLDERERLHMLREDYEHTRLHSVHPASLD GHLPHPSLITPGLPSMHYPRISPTAGNQNGLLNKTPPTAA LSAPPPLISTLGGRPVSPRRTTPLSAEIRERPPSHTLKDIEA
R (SEQ ID NO:2).
Accordingly, the present disclosure provides methods as described herein for classifying an individual comprising detecting an alteration in the AUTS2 short isoform. As used herein, the AUTS2 short isoform refers to SEQ ID NO:2. An alteration includes a mutation affecting the expression and/or amino acid sequence of the AUTS2 short isoform. Alterations indicate an increased risk of developing or having AUTS2 syndrome. Preferably, the methods disclosed herein comprise detecting at least one alteration, preferably a chromosomal abnormality, between exons 9- 19 (inclusive of exons 9 and 19) of the AUTS2 gene of said individual. Exons 9-19 correspond to nucleotide positions 2212-6426 of NM_015570. The alteration may affect exons 9-19 or their corresponding introns. These exons encode the AUTS2 short isoform and alterations in either the intervening introns or 5' and 3' genomic regions may affect the expression and/or proper splicing of the short isoform.
Preferably the alteration is an intragenic deletion deleting at least one or a part of exons 9-19. As used herein, the alteration between exons 9-19 of the AUTS2 gene is preferably in the genomic region which encodes the short isoform and/or affects translation or splicing. Identification of an alteration between exons 9-19 or in the genomic region encoding the AUTS2 short isoform may also indicate that the severity of AUTS2 syndrome is likely to be greater than the severity resulting from an abnormality in the 5' region of AUTS2. This finding is contrary to what one would have predicted. A disruption in the 3' end of a gene often results in a truncated protein which may retain a small amount of function. Although not wishing to be bound by theory, disruptions occurring in the 5' end of the AUTS2 gene may have a lesser effect on the shorter isoform, which can then perform its normal function. The detection of the chromosomal abnormalities and alterations described herein may be performed on any sample from an individual which comprises nucleic acid, in particular genomic DNA. Suitable samples include bodily fluids (e.g., blood, urine, serum, cerebral spinal fluid, amniotic fluid), cells (e.g., brain cells, white blood cells), or body tissue. Standard methods are known for extracting DNA from samples.
Nucleic acid may be used in a purified or unpurified form.
A number of methods for detecting chromosome abnormalities are known in the art, including karyotyping, FISH, array-CGH, MAPH, sequencing assays, quantitative PCR (q-PCR), QMPSF (Quantitative Multiplex PCR of Short Fluorescent Fragments, see Casilli et all, 2001 Hum Mutat 20:218-26) and MLPA. (see U.S. Publication US2011/0281759)
Karyotyping is a conventional procedure for determining the chromosome complement of an individual, defined both by the number and morphology of the chromosomes. Stains are used to produce banding patterns on the chromosomes which permits the identification of the chromosome and the visualization of structural abberations. GTG banding (G-bands by trypsin using Giemsa) is a standard cytogenetic technique.
Hybridization assays such as fluorescence in situ hybridization (FISH) are also common techniques for detecting chromosomal abnormalities. In multicolor-FISH, chromosomes are hybridized with "painting probes" labeled by combinations of different fluorophores. Details of this technique and variations of the techniqe are found, e.g., in Liehr et al. Histol Histopathol 2004 19:229-237. Array comparative genomic hybridization (array-CGH) and Multiplex Amplifiable Probe Hybridization (MAPH) are both molecular-cytogenetic methods for the analysis of copy number variation (Kousoulidou et al. Methods Mol Biol 2010 653:47-71). Copy number variation (CNV) refers to the number of copies of a particular gene.
Amplification assays are also known to the skilled person and are suitable for detecting chromosome abnormalities. 3C-qPCR is one exemplary technique (Hagege et al. 2007 Nature Protoc 2" 1722-33). In a preferred embodiment, a chromosomal abnormality in the AUTS2 gene is detected using MLPA. MLPA is a technique to determine copy number (Schouten et al. 2002 Nucleic Acids Res. 30:e57). MLPA probes are designed to hybridize to a region of interest. MLPA probes comprise a set of two probes. Each probe of the probe set is designed to hybridize to the target DNA in close proximity to each other. When both probes hybridize to the target DNA they are ligated together and thus form a complete DNA template. Each probe in a probe set has between 15-100 nucleic acids complementary the target sequence, preferably between 20-50 nucleotides (referred to as LHS and RHS, left and right hybridizing sequence, respectively.) Each probe has, in addition the LHS and RHS, a primer recognition sequence. In the MLPA technique, it is not the target sequence, but rather the ligated probe set which is amplified. The amount of PCR product formed will be more than in a control sample if a duplication is present in the region of interest. A "stuffer" sequence may be inserted between the LHS/RHS and the primer recognition sequences. The stuffer sequence may be used to alter the length of the amplified probe in order to distinguish the particular probe set from other probe sets. Further details of this method are described in U.S. Patent No. 6,955,901 which is hereby incorporated by reference in its entirety. The MLPA technique has the advantage that small deletions and duplications can be detected and the location of the abnormality can be finely mapped to a specific region. MLPA permits the high resolution detection of abnormalities in specific gene candidates. It also is able to measure copy number changes with high quantitative power. In the case of the AUTS2 gene, these advantages are of importance as we have discovered that relatively small deletions can be pathogenic and the severity of the resulting disorder can be predicted based on the location of the abnormality. While MLPA is a well-known technique, its application in determining alterations in the AUTS2 gene is surprising as it was not known before the present disclosure that intragenic deletions of AUTS2 could lead to the phenotypes described in Table 1.
One aspect of the disclosure provides multiplex ligation dependent probe
amplification (MLPA) sets specific for the AUTS2 gene. "Specificity" for the AUTS2 gene refers to the ability of each probe in the set of two to bind adjacently to each other to the AUTS2 gene. Each probe of the set may also bind to other genomic sequences, however, for amplification to occur, the probes need to bind next to each other in order to ligate. Preferably, the MPLA probe set is specific for a genomic region between exons 9-19 of the AUTS2 region, more preferably for the genomic region which encodes the short isoform. The probe sets may be designed to either the sense or anti-sense strand. While exon specific probes are generally preferred, some exons may not contain suitable sequences with which to design MLPA probes.
MLPA probe pairs useful in the present invention include MLPA 102, MLPA 110, MLPA 118, MLPA 126, MLPA 130, MLPA 096, MLPA 096, MLPA 136, MLPA 140, MLPA144, MLPA 100, MLPA 108, MLPA 112, MLPA 116, MLPA 120, MLPA 124, MLPA 128, MLPA 132, MLPA 134, MLPA 105, MLPA 138, MLPA 107, and MLPA 136. Probe sequences are provided in Table 6. Table 6A provides the "left" probe and Table 6B provides the "right" probe for each probe pair. Capital letters indicate the sequence which hybridizes to AUTS2 and lower case letters indicate the primer recognition and stuffer sequences. It is clear to a skilled person that the necessary sequence for MLPA probes is the sequence indicated by capital letters in Tables 6A and 6B. Any primer recognition and stuffer sequence can be used. Thus, e.g., MLPA 102 probe refers to a pair of sequences comprising:
1) CGGAGGAAGCGGAGAGAGTCCA and
2) CCTCGGCAGAAGAGGACATCATTGATGG. It is clear to a skilled person that additional MLPA probe pairs can be designed to the AUTS2 genomic regions. MLPA probe design is described, e.g., in Pantano et al. BMC Genomics 2008 9:573, Zhi J. BMC Research Notes 2010 3: 137 and MRC-Holland B.V. provides a detailed probe design protocol at www.mlpa.com.
One aspect of the disclosure provides antibodies specific for the AUTS2 short isoform of SEQ ID NO:2. "Specific for the AUTS2 short form" refers to antibodies which recognize SEQ ID NO:2, but not exons 1-8 of AUTS2. The antibody specific for the AUTS2 short isoform of SEQ ID NO:2, does not recognize the protein encoded by the longer transcript. The AUTS2 specific antibody is typically specific for a three dimensional epitope not present in the protein encoded by the longer transcript. The antibody specific for the AUTS2 short isoform of SEQ ID NO:2 can also be specific towards the unique 5' end of the shorter isoform. Such antibodies are useful to detect not only the presence of the AUTS2 short isoform, but also changes in expression level. Suitable antibodies include monoclonal and polyclonal antibodies as well as fragments containing the antigen binding domain and/or one or more
complementarity determining regions of these antibodies. As used herein, an antibody fragment is defined as at least a portion of the variable region of the immunoglobulin molecule that binds to its target, i.e., the antigen binding region.
In addition to the diagnostic applications, the present invention also provides new therapeutic applications for treating AUTS2 syndrome and in particular autism. The disclosure demonstrates that loss of auts2 in zebrafish embryos causes microcephaly and decreases neuronal staining. This phenotype is rescued by the C-terminal AUTS2 human mRNA. The AUTS2 short isoform is therefore sufficient for rescuing the loss of AUTS2 in zebrafish and is useful in treating disorders mediated by AUTS2
disruption, such as AUTS2 syndrome and in particular autism. Although it is preferred that the AUTS2 short isoform is used to treat patients having a disruption in the AUTS2 short isoform (e.g., reduced expression, sequence mutation), the disclosure also contemplates treating patients with any disruption if the AUTS2 gene which affects the expression or function of the gene. Accordingly, the disclosure provides a polypeptide comprising the AUTS2 sequence having at least 80, at least 85, preferably at least 90, more preferably at least 95% identical to SEQ ID NO:2. While the polypeptide may include additional sequences, such as protein tags (His, Flag, Myc, HA, GST, V5, GFP, etc.), it does not comprise additional AUTS2 sequences, namely from exons 1-8. Preferably, the N-terminus of said polypeptide is encoded by exon 9 of the AUTS2 gene. Preferably, said
polypeptide comprises the alternatively spliced amino acids VRTPGRN.
Accordingly, the disclosure provides a nucleic acid molecule encoding the polypeptides disclosed herein. Preferably the nucleic acid sequence encodes the amino acid sequence of SEQ ID NO:2 or an amino acid sequence at least 80, at least 85, preferably at least 90, more preferably at least 95% identical to SEQ ID NO:2.
Preferably, the nucleic acid molecule comprises SEQ ID NO: l or a nucleic acid molecule at least 80, at least 85, preferably at least 90, more preferably at least 95% identical to SEQ ID NO: 1.
The nucleic acid molecule may comprise additional sequences, for example, promoter or expression sequences, but does not comprise additional sequences of AUTS2, e.g., exons 1-8. Preferably, the nucleic acid encodes a polypeptide encoded by exon 9 of the AUTS2 gene.
The nucleic acid molecule may be provided in a vector. A "vector" is a recombinant nucleic acid construct, such as plasmid, phase genome, virus genome, cosmid, or artificial chromosome, to which another DNA segment may be attached. The term "vector" includes both viral and nonviral means for introducing the nucleic acid into a cell in vitro, ex vivo or in vivo. Non-viral vectors include plasmids, liposomes, electrically charged lipids (cytofectins), DNA-protein complexes, and biopolymers. Viral vectors include retrovirus, adeno-associated virus, pox, baculovirus, vaccinia, herpes simplex, Epstein-Barr and adenovirus vectors, as set forth in greater detail below. Vector sequences may also contain one or more regulatory regions, and/or selectable markers useful in selecting, measuring, and monitoring nucleic acid transfer results (transfer to which tissues, duration of expression, etc.). Cells comprising said nucleic acids or vectors comprising nucleic acids are also provided. The method of introduction is largely dictated by the targeted cell type include, e.g., CaP04 precipitation, liposome fusion, lipofectin, electroporation, dextran-mediated transfection, calcium phosphate precipitation, polybrene mediated transfection, protoplast fusion, viral infection, encapsulation of the polynucleotide (s) in liposomes, and direct microinjection of the DNA into nuclei. The nucleic acids may stably integrate into the genome of the host cell (for example, with retroviral introduction, outlined below), or may exist either transiently or stably in the cytoplasm (i.e. through the use of traditional plasmids, utilizing standard regulatory sequences, selection markers, etc.).
AUTS2 short isoform polypeptides as described herein may be produced by culturing a host cell transformed with an expression vector containing nucleic acid encoding a dominant negative polypeptide. Appropriate host cells include yeast, bacteria, archaebacteria, fungi, and insect and animal cells, including mammalian cells. Of particular interest are Drosophila melangaster cells, Saccharomyces cerevisiae and other yeasts, E. coli, Bacillus subtilis, SF9 cells, C129 cells, 293 cells, Neurospora, BHK, CHO, COS, Pichia pastoris, etc. Preferably, said polypeptides are expressed in mammalian cells. Mammalian expression systems are also known in the art, and include retroviral systems.
Suitable cell types include tumor cells, Jurkat T cells, NIH3T3 cells, CHO, and Cos, cells. Preferably, the cells are culturable cells and more preferred the cells are in vitro cells.
The nucleic acid sequence encoding the AUTS2 short isoform may be provided to a cell as part of a gene delivery vehicle. Such a vehicle is preferably a liposome or a viral gene delivery vehicle. Liposomes are well known in the art and many variants are available for gene transfer purposes. Various viral gene delivery are currently used to transfer genes into target cells. In the present disclosure it is preferred to use those viral vectors that do not express their own genes but only the transferred genes. In a viral vector, the nucleic acid compound is preferably provided as an expression cassette wherein the expression cassette encodes a transcript comprising said compound.
A further aspect of the present disclosure is the provision of a vector comprising a nucleic acid molecule encoding the AUTS2 short isoform for use in gene therapy. Such therapy is useful in the treatment of AUTS2 syndrome. Methods are therefore provided for treating an individual afflicted with or at risk of developing AUTS2 syndrome comprising administering to an individual in need thereof a nucleic acid molecule encoding the AUTS2 short isoform. The nucleic acid molecule is preferably provided in a viral vector suitable for gene therapy. Appropriate vectors and delivery methods are known to a skilled person and are described, e.g., in Schlachetzki et al. Neurology, Gene therapy of the brain. (2004) 62: 1275- 1281 and Richardson et al. Neurosurg Clin N Am. 2009 20(2):205-10.
A further aspect of the disclosure provides pharmaceutical composition comprising a polypeptide, nucleic acid, vector, or cell as described herein and a pharmaceutically acceptable excipient. Such pharmaceutically acceptable excipients may for instance be found in Remington: The Science and Practice of Pharmacy, 20th Edition. Baltimore, MD: Lippincott Williams & Wilkins, 2000. When administering the pharmaceutical preparations thereof to an individual, it is preferred that the compound is dissolved in a solution that is compatible with the delivery method. For intravenous, subcutaneous, intramuscular, intrathecal and/or intraventricular administration it is preferred that the solution is a physiological salt solution. Preferred are excipients capable of forming complexes, vesicles and/or liposomes that deliver such a compound as defined herein in a vesicle or liposome through a cell membrane. Many of these excipients are known in the art. Suitable excipients comprise polyethylenimine (PEI) or similar cationic polymers, including polypropyleneimine or polyethylenimine copolymers (PECs) and derivatives, ExGen 500, synthetic amphiphils (SAINT- 18), lipofectin™, DOTAP and/or viral capsid proteins that are capable of self assembly into particles that can deliver such compounds, to a cell. The compositions can be administered by controlled release means or by delivery devices that are well known to those of ordinary skill in the art. Examples include, but are not limited to, those described in U.S. Pat. Nos. 3,845,770; 3,916,899;
3,536,809; 3,598, 123; and 4,008,719, 5,674,533, 5,059,595, 5,591,767, 5, 120,548, 5,073,543, 5,639,476, 5,354,556, and 5,733,566, each of which is incorporated herein by reference. Such dosage forms can be used to provide slow or controlled-release of one or more active ingredients using, for example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions. Suitable controlled-release formulations known to those of ordinary skill in the art, including those described herein, can be readily selected for use with the active ingredients of the invention. The invention thus encompasses single unit dosage forms suitable for oral administration such as, but not limited to, tablets, capsules, gelcaps, and caplets that are adapted for controlled-release.
Actual dosage levels of the pharmaceutical preparations described herein may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. The selected dosage level will depend upon a variety of factors including the activity of the particular compound, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start with doses of the compounds described herein at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved. Accordingly, the disclosure provides a method of treating an individual, preferably a human, in need thereof comprising administering a therapeutically effective amount of a pharmaceutical composition as disclosed herein. Preferably the composition is administered intracerebrally or into the spinal cord of the individual.
The disclosure further provides non-human animals, preferably mammals, comprising nucleic acids encoding AUTS2 short isoform. Methods for introducing nucleic acids into animals are known to one of skill in the art and include standard transgenic techniques such as introducing said nucleic acid into an undifferentiated cell type, e.g., an embryonic stem (ES) cell. The ES cell is injected into a mammalian embryo, where it integrates into the developing embryo. Insertion of the nucleic acid construct into the ES cells can be accomplished using a variety of methods well known in the art including for example, electroporation, microinjection, and calcium phosphate treatment. The embryo is implanted into a foster mother for the duration of gestation.
Transgenic animals comprise a heterologous nucleic acid sequence present as an extrachromosomal element or stably integrated in all or a portion of its cells, especially in germ cells. During the initial construction of the animal, "chimeras" or "chimeric animals" are generated, in which only a subset of cells have the altered genome. Chimeras are primarily used for breeding purposes in order to generate the desired transgenic animal. Animals having a heterozygous alteration are generated by breeding of chimeras. Male and female heterozygotes are typically bred to generate homozygous animals.
Methods for generating transgenic animals, particularly animals such as mice or rats, have become conventional in the art and are described, for example, in U.S. Pat. Nos. 4,736,866 and 4,870,009.
The invention further provides a non-human animal, preferably a non-human vertebrate wherein the AUTS2 short isoform is knockout on at least one and preferable two of the AUTS2 chromosomes in the cell. In another preferred
embodiment the invention provides a non-human animal, preferably a non-human vertebrate, wherein the AUTS2 short isoform is silenced, preferably by means of an siRNA specific for the AUTS short isoform. In a preferred embodiment said non- human animal is a mammal or fish.
As used herein, "to comprise" and its conjugations is used in its non-limiting sense to mean that items following the word are included, but items not specifically mentioned are not excluded. In addition the verb "to consist" may be replaced by "to consist essentially of meaning that a compound or adjunct compound as defined herein may comprise additional component(s) than the ones specifically identified, said additional component(s) not altering the unique characteristic of the invention.
The articles "a" and "an" are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, "an element" means one element or more than one element.
The word "approximately" or "about" when used in association with a numerical value (approximately 10, about 10) preferably means that the value may be the given value of 10 more or less 1% of the value.
The term "treating" includes prophylactic and/or therapeutic treatments. The term "prophylactic or therapeutic" treatment is art-recognized and includes administration to the host of one or more of the subject compositions. If it is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the host animal) then the treatment is prophylactic (i.e., it protects the host against developing the unwanted condition), whereas if it is administered after manifestation of the unwanted condition, the treatment is therapeutic, (i.e., it is intended to diminish, ameliorate, or stabilize the existing unwanted condition or side effects thereof).
All patent and literature references cited in the present specification are hereby incorporated by reference in their entirety.
The invention is further explained in the following examples. These examples do not limit the scope of the invention, but merely serve to clarify the invention. EXAMPLES
Example 1
Patients
We studied a total of 49,684 probands with ID and/or MCA on oligo-based arrays with coverage of AUTS2. We found forty-three deletions including at least part oiAUTS2 and a maximum of two other genes: fourteen were found in AUTS2 introns, and twenty-five included at least one exon. For the remaining five deletions, it was uncertain whether they included an exon due to gaps between array probes. We were able to obtain detailed genotypic and phenotypic data from 19 of these 43 probands (patient 1-19), as well as from 3 affected family members that were shown to carry the familial deletion. Further, an inversion (patient 20) and a translocation (patient 21) with one of the breakpoints in AUTS2 were found by conventional karyotyping from two different laboratories; detailed genotypic and phenotypic data were available for both of these patients. In total 25 patients from 21 unrelated families were included in our analyses (see figure 1, figure 6, and table 7).
Ten of the AUTS2 aberrations found were proven to be de novo (patient 4, 9, 12, 14, 16, 18, 19, 20 and 21). One of these de novo deletions was inconclusive with regard to AS and ID causality in the family, because an affected brother was not carrying the AUTS2 deletion (patient 4). Four probands had an AUTS2 deletion that was inherited from one of the parents (patient 3, 5, 8 and 10) and in eight probands the inheritance status of the AUTS2 deletion was unknown, because one or both parents were unavailable for testing (patient 1, 2, 6, 7, 11, 13, 15 and 17).
Mapping of the breakpoints indicated that four of the deletions were intronic (patient 1-4) and that fifteen deletions encompassed at least one exon of the AUTS2 trascript (patient 5-19). Four probands carried a deletion that is predicted to cause a frame -shift of the full-length A UTS2 transcript (patient 10, 11, 12 and 15). In patient 18 the deletion involved a downstream gene, WBSCR17, while in patients 17 and 19 two downstream genes, WBSCR17 and CALN1 were also included in the deletion (see figure 1).
Twenty of the probands had intellectual disability or a developmental delay (patient 1, 2, 4-21); only patient 3 had normal intellectual development. Two of the parents carrying an AUTS2 deletion had a normal intelligence (the mother of patient 3 and the father of patient 5), one had experienced learning difficulties (the mother of patient 8) and one had mild intellectual disability (mother of patient 10 and her sibling). Ten probands were diagnosed with AS or showed autistic behavior (patient 3, 4, 7, 9, 13, 16, 17, 20 and 21). The mother of patient 3 reported herself to have
Asperger syndrome characteristics, but diagnostic testing on autism spectrum disorder was lacking. Other clinical features were (numbers of patients in brackets): microcephaly (16), short stature (14), feeding difficulties (10), hypotonia (8), cerebral palsy (9) and dysmorphic features; hypertelorism (10), proptosis (6), short palpebral fissures (9), epicanthal folds (7), hypertelorism (7), a short and/or upturned philtrum (10), micrognatia (7) and a narrow mouth (12). Less frequent occurring features are skeletal abnormalities including (signs of) arthrogryposis (3), umbilical or inguinal herniation (2) and heart defects (4) (see tables 1, 7 and figure 2). Taken together, these observations suggested that genomic deletions of the AUTS2 locus likely drive a complex phenotype that includes both neurocognitive impairment but also anatomical defects, includes facial dysmorphic features.
Notably, the phenotype caused by the observed AUTS2 aberrations is variable.
Several patients had mild neurocognitive phenotypes that were readily
distinguishable from the remainder of our A[/ <¾?-positive cohort: Patients 1-4, all of whom had an intronic AUTS2 deletion, as well as patient 5, with an in frame deletion of exon 2, showed a mild phenotype. The AUTS2 syndrome severity scores in these patients did not exceed 5 (maximum score was 32). As can be seen from figure 1, the AUTS2 syndrome severity score in patients with deletions of exons 3 and 4 (patients 6-8) was generally lower than seen in patients with deletions of downstream exons, a whole gene deletion or the deletion of exon 1 to 4 (patient 9- 19).
Controls
In total, 16,784 healthy controls from 12 control cohorts (supplementary table S2) were analyzed using arrays with coverage of the AUTS2 locus. Four deletions involving A UTS2 were identified in the WTCCC2 cohort; no AUTS2 deletions were found in any of the other cohorts. The four deletions identified in controls were all intronic, not disrupting any annotated or predicted exons (supplementary table S3). The number oiAUTS2 deletions found in controls (4/16,784) compared to the number oiAUTS2 deletions found in cases (43/49,651) is significantly different: p
(Fisher exact test, OR = 3.63; 95% CI = 1.32- 13.95).
Example 2
Evolutionary conservation
As the strong phenotype associated with deletions in the 3' exons oiAUTS2 suggests an important role for this part of the gene, we determined the degree of evolutionary conservation for each exon (figure 3 and supplementary figure S2). First, amino acid sequence alignments were performed for AUTS2 gene products annotated in the current Ensembl genome database for each species. In species where multiple proteins are annotated we selected the longest protein (e.g. many splice variants are annotated in humans and mice). This strategy successfully detected the conservation of exons 8- 19 in all species tested, whereas exons 1-7 were found only in annotations of the macaque, mouse and chicken genome (black numbers in figure 3c). As an alternative strategy to deal with possible incomplete annotation, we also scanned the complete genomes for conserved open reading frames using the tblastn algorithm10. This method revealed the presence of additional conserved sequences in all species tested including zebrafish (see grey exons in figure 3a). Although the conserved genomic sequences do not necessarily represent exons we compared their degree of conservation (see grey numbers in figure 3c). Also in this comparison, exons 8- 19 are more strongly conserved than exons 1-7.
Example 3
Expression of a short 3' AUTS2 variant in human brain
In view of the relatively mild phenotype of patients with a deletion in the 5' half of the AUTS2 gene, and the relatively low degree of sequence conservation in the 5' exons, we explored if the 5' and 3' exons may be differentially expressed in human brain mRNA. Using 5'-RACE, we identified a short AUTS2 mRNA variant starting in the centre of exon 9. This alternative transcription start site is followed by a start codon in the same reading frame as the full length AUTS2 transcript, and is predicted to encode a polypeptide of 704 amino acids instead of the 1,295 amino acids of the full length protein (figure 3d). Example 4
AUTS2 knockdown and rescue in the zebrafish
A recent post hoc analysis of autistic spectrum disorder (ASD) loci has revealed that microcephaly is strongly associated with ASD (Shinawi, M. et al. J Med Genet 47, 332-41 (2010)). Likewise, microcephaly is the most representative clinical feature amongst our patients with an AUTS2 deletion (16/43).
Given the association between autism and changes in head size, we investigated the role of AUTS2 in brain development in vivo. Using reciprocal BLAST, we identified a single Danio rerio ortholog oiAUTS2 (auts2 on chromosome 10; 62% identity with the long isoform of human AUTS2) and we were able to detect endogenous auts2 message by RT-PCR as early as the embryonic 5-somite stage (data not shown). Next, we designed two splice-blocking morpholinos (sb-MOs) targeting auts2, a 5' MO targeting the splice donor site of exon 2 and a 3' MO targeting the splice donor site of exon 10 (corresponding to the short human AUTS2 transcript based on sequence conservation, see Figure 3a), which we injected into two-cell-stage embryos (n = 50- 100 embryos per injection dose). Masked scoring of embryos at 3 days post- fertilization (3 dpi) showed reproducible microcephaly phenotype, 53% and 48% for 5' and 3' sb-MO respectively (Figure 4a) that were concomitant with the efficiency of splice blocking of the two sb-MOs, as established by RT-PCR. Morphants had a normal appearance in regards to their pigment cells and their body length was not statistically different from the control fish from the same clutch, excluding any developmental delay. Importantly, the phenotype was specific; the observed microcephaly driven by the two sb-MOs could be rescued efficiently with co-injection of wildtype human full-length and short AUTS2 mRNAs (Figure 4b).
Table 1: Phenotypic features characterizing the AUTS2 syndrome patients
This table shows the most important phenotypic features of the AUTS2 syndrome and in what percentage they are seen in patients with a pathogenic AUTS2 aberration. For the calculation of the percentages all patients described here (n=18) with a likely pathogenic AUTS2 deletion/disruption (see Table 7, patient 6-21 and their affected family members) and the seven patients described in literature, with a translocation or inversion breakpoint in AUTS2, were taken into account2 4. Table 1
Phenotypic features of the AUTS2 % syndrome (in Total, n=27)
Growth and feeding
Low birth weight 33 Short stature <pl0 63 Microcephaly <p2 59 Feeding difficulties 52
Neurodevelopmental disorders
Mental retardation/Developmental
delay 100
Autism/Autistic behavior 44
Neurological disorders
Generalised hypotonia 44
Brain disorder 30
Cerebral palsy /spasticity 41
Dysmorphic features
Highly arched eyebrows 33
Hypertelorism 37
Proptosis 37
Short palpebral fissures 37
Ptosis 33
Epicanthal fold 30
Strabismus 33
Short/upturned philtrum 56
Micro/retrognatia 37
Low set ears 30
Narrow mouth 56
Skeletal disorders
Kyphosis/ scoliosis 15 Arthrogryposis/ shallow palmar
creases 15
Tight heel cords 19
Congenital malformations
Hernia umbilicalis/ inguinalis 11
Heart defect 11 Table S2: Control cohorts. The control cohort tested for CNV's in AUTS2 with the number of controls per cohort and the references that give more information on these cohorts. The number of deletions identified in the controls at the AUTS2 locus is indicated. Datasets 1, 2, 4, and 5 were determined with Affymetrix 6.0; dataset 3 was determined with Illumina 1M; dataset 6 was determined with Illumina 240K-650K; and dataset 7 was determined with agillent 105K/180K.
Control dataset # individuals deletions
Ottawa Heart Institute (OHI)
1 controls from Canada 1,234 0
2 POPGEN controls from Germany 1, 123 0
3 SAGE controls from USA 1,287 0
4 Wellcome Trust (WTCCC) controls 4,783 4
5 HapMap phase 3 controls 1,056 0
combined adult controls set
(HGDP, NINDS, PARC/PARC2,
London, FHCRC, In CHIANTI)
6 (excluding the WTCCC2 cohort) 6,320 0
7 lowlands concortium controls 981 0 References for the dataset and analysis described as follows:
1: Dataset and analysis described in Lionel et al. 2011 (PMID: 21832240) 2: Dataset and analysis described in Lionel et al. 2011 (PMID: 21832240) 3: Dataset and analysis described in Pinto et al. 2010 (PMID: 20531469) 4: http s : //www . wtccc . or g. uk/ccc2 /
5: Dataset and analysis described in International HapMap 3 Consortium et al. 2010 (PMID: 20811451) raw data available at
http : //h apm ap . ncbi . nlm . nih . go v/do wnlo ads/ r a w_dat a/h ap m ap 3_affy 6.0/ 6: Dataset and analysis described in Cooper et al. 2011 (PMID: 21841781) 7: personal communication with Kok K., UMCG, Department of Genetics, Groningen, the Netherlands
Table S3: Deletions in AUTS2 found in controls.
Four deletions were found in the WTTTC2 cohort. All four deletions were intragenic deletions. The breakpoint and effect of the deletions on AUTS2 are depicted here. Exons involved in deletion: - (in x): no exons involved but intron x deletion; basepair: number of basepairs deleted; probe: number of probes deleted. id number breakpoints AUTS2 exons involved basepairs probe
69,665,498-
BC1088 69,777,260 - (in 5) -111763 bp 111
68,857,867-
BC0081 68,892, 176 - (in 1) -34310 bp 19
69,328,576- - (in 4)/
NBS1906 69,469,301 alternative -140726 bp 70
69,067,672- NBS1607 69, 194,966 - (in 2) -127295 bp 72
Table S4: Overview of the location of the probes used for MLPA analysis. All features are sorted for the start position with numbering according to genome build HG18.
Hybridizing region (HG18)
MLPA
Exon probe Start End
exon 1 68702256 68702885
MLPA 102 68702795 68702847
exon 2 69002209 69002421
MLPA 110 69002227 69002281
exon 3 69221055 69221156
MLPA 118 69221075 69221136
exon 4 69237459 69237494
MLPA 126 69237710 69237775
MLPA 130 69538370 69538429
exon 5 69538675 69538704
MLPA 096 69542823 69542872
69596454 69596513
MLPA 136 69598011 69598086
MLPA 140 69604531 69604615
MLPA 144 69608563 69608660
69612586 69612645
MLPA 100 69801299 69801351
exon 6 69801492 69801543
69858624 69858683
MLPA 104 69865791 69865850
exon 7 69865793 69866264
exon 8 69867675 69867928
MLPA 108 69867966 69868027
exon 9 69869037 69869257
MLPA 112 69869343 69869403
exon 10 69870947 69870991
MLPA 116 69870967 69871035
MLPA 120 69874308 69874382
exon 11 69874472 69874567
exon 12 69876951 69877022
MLPA 124 69876959 69877036
MLPA 128 69877860 69877941
69878248 69878307
exon 13 69878280 69878309
MLPA 132 69879948 69880038
exon 14 69880026 69880097
MLPA 134 69884478 69884565 exonl5 69884538 69884679
exonl6 69887865 69887942
MLPA 105 69888063 69888120
MLPA 138 69888864 69888956
exonl7 69888878 69888961
exonl8 69890132 69890354
MLPA 107 69890571 69890635
exonl9 69892671 69895991
MLPA 136 69895018 69895108
Table S5: FISH probes used for breakpoint mapping on chromosome 7 and chromosome 22. Showing probe name, chromosome band, start position and end position according to the GRCh37 genome assembly.
Name Chr Start (bp) End (bp)
RP4-736H5 chr7ql l.22 67029247 67175902
RP11-358M3 chr7ql l.22 67400859 67525501
RP11-156A14 chr7ql l.22 67909795 68062117
RP11-3P22 chr7ql l.22 68613015 68778577
RP4-715F13 chr7ql l.22 69466935 69610812
RP11-290M1 chr7ql l.22 69819029 69993539
RP11-689B18 chr7qll.22 69996264 70178753
G248P87196G1 (WI2- chr7qll.22 70298275 2151M1) 70259728
G248P82441H1 (WI2- chr7qll.22 70247727 1017P1) 70206291
G248P84061D2 (WI2- chr7qll.22 70279409 1371H3) 70239978
RP11-575M4 chr7qll.22 70185711 70372702
RP11-26L10 chr7ql l.22 70525506 70703364
RP11-409J21 chr7ql l.22 71049273 71204976
RP4-562A11 chr7ql l.23 77133285 77277250 RP4-560O14 chr7q21.11 81518832 81663290 CTA-115F6 chr22ql l.l 17779327 17965779 CTA-433F6 chr22ql l.21 20729567 20874531 CTA-322B1 chr22ql l.23 24315264 24392055 CTA-125H2 chr22ql2.1 26230801 26404213 RP11-259P1 chr22ql2.1 26612062 26660045
RP11-322L06 chr22ql2.1 26964530 27132594 CTA-992D9 chr22ql2.1 27411607 27566652
RP11-263G19 chr22ql2.1 27629570 27813359
RP11-699H18 chr22ql2.1 27816054 27979854 RP11-1056M20 chr22ql2.1 27932064 28119112
RP11-772E17 chr22ql2.1 28105947 28295835 RP3-353E16 chr22ql2.1 28167384 28357044 RP11-329J7 chr22ql2.1 28914764 28987692
RP11-664C16 chr22ql2.1 29341003 29489465 chr22ql2.1-
CTA-57G9 29626406 ql2.2 29512541
RP1-76B20 chr22ql2.2 30049384 30221065 RP4-539M6 chr22ql2.2 30787513 30946905 CTA-221H1 chr22ql2.3 34571571 34574493 CTA-150C2 chr22ql3.1 39280299 39481341
Table 6A "left" primer probe exon5 L096_ _auts2 gggttccctaagggttggaTATAGCATGTGACTGAGCTGATTGGAAG
exon6 L100_ _auts2 gggttccctaagggttggatGCAAATAGCTGCACTGCAGGTCTC
exon7 L104_ _auts2 gggttccctaagggtt ggaTC C GGAGTTAGGTGTTGGC AC GCTAC C A
exon8 L108_ _auts2 gggttccctaagggttggatACCGCCCCTCGCTGTGACCTCACCCTACCTA
exon9 L112_ _auts2 gggttccctaagggttggattataCCACCAGAGATCGAATGGGGACTGACGGCT
exonlO L116_ _auts2 gggttccctaagggtt ggatGTTGAC C C ATTCT AC C GGC AC AGTGTGAGTTTC A exonl l L120_ _auts2 gggttccctaagggttggaTGGCACGTCTGTGTGATCATTCCATCATGGCTTCTTGACTA exonl2 L124_ _auts2 gggttccctaagggttggatCCTATCGATGTCGCTGCTCGGCCTGGGACA
exonl3 L128_ _auts2 gggttccctaagggttggaTCTCCTGGATGGATGGAAAGGACACCTCTCCCCAGATA exonl4 L132_ _auts2 gggttccctaagggttggatCTTTGGGGAAATTGAAGGTGGTTTTCTCTGA
intron5a L136_ _auts2 gggttccctaagggttggaTAGACAAGTAGGGTGATATTTTCCTTGGTCTCGTG intron5b L140_ _auts2 gggttccctaagggttggaTGGGTAGGGGTGGTGGTGTTGCCTCCCGGAACCCTCA intron5c L144_ _auts2 gggttccctaagggttggaTGCACGTGGCTCCTGTGAACCGTGGTTATACCCAGGCTA exon 1 L102_ _auts2 gggttccctaagggttggaattCGGAGGAAGCGGAGAGAGTCCA
exon 2 L110_ _auts2 gggttccctaagggttggaattatattCTCAGGAACGTGTGGAGAAACGCCA
exon 3 L118_ _auts2 gggttccctaagggttggattaatTGCAGGGACAGTGACAGTGAAAGTGCCAGTGGAGA exon 4 L126_ _auts2 gggttccctaagggttggaattatcggaTTCCTAACGCCCAGGACATTAAGCTCCTTAC exon 5 L130_ _auts2 gggttccctaagggttggattaatTGGGAGGGAAGCGCTCACTTGTGTGATTGA exon 15 L134_ _auts2 gggttccctaagggtt ggaTAAC AC CTTTATTC CTTGCTGTTGGC CTTGGGAC C CTT AC exon 16 L105_ _auts2 gggttccctaagggttggaTAGGCAGGAGATGGTGCTCTCTTAGGA
exon 17 L138_ _auts2 gggttccctaagggtt ggaTCTTGTTTGC AG AGC CTTTTAATC GGC C GT
exon 18 L107_ _auts2 gggttccctaagggttggaTGTCTCAGTCGCAGTTATACATCAGCACGTC
exon 19 L136_ _auts2 gggttccctaagggttggaTGTTTTCACTGTATTCAATAACTGACGGATGTAAGGTGCA
Table 6B "righf'primer probe
R096_auts2 /5Phos/GCATATAGCCCAGTGGCCAAGCtctagattggatcttgctggcgcgtcc
R100_auts2 /5Phos/TCTCAAGCAGTCAACTAGCACCACCTTATtctagattggatcttgctggcgcgtcc
R104_auts2 /5Phos/GAACATGACAGCCAGGATGCAGGGCCGATTtctagattggatcttgctggcgcgtcc
R108_auts2 /5Phos/TGTTGTCCCGATTGCAAGGTTGCATTTGCCtctagattggatcttgctggcgcgtcc
R 112_auts2 /5Phos/GTTGGCTGGAGAGAAGGGAATGTGTCCTAGTtctagattggatcttgctggcgcgtcc
R116_auts2 /5Phos/TTACCATGCTACACATTGAATGTAACTGCTTTGCCtctagattggatcttgctggcgcgtcc
R120_auts2 /5Phos/ATGGCATCAAACTGAGATTACGTGGCTTGCTCAtctagattggatcttgctggcgcgtcc
R124_auts2 /5Phos/GTCCCACACACTTTACTCCAAAAGGACCCGAGGGTACGTGCAAAGTCtctagattggatcttgctggcgcgt.cc
R128_auts2 /5Phos/CCAGTTAGTAGACAGCTTTAAAAGCCAGGGGTCTGAAGGCCATTtctagattggatcttgctggcgcgtcc
R132_auts2 /5Phos/AATGTCTTCCTCCTAACCACGTTGCTCTTTCTTGTTCCAGAAACCAGGGAAGTGtctagattggatcttgctggcgcgtcc
R136_auts2 /5Phos/CCGACCCTAGTAACAAAAGTCTCTCAGTTACCCAGGGCACattatcaggagcatttctagattggatcttgctggcgcgtcc
R140_auts2 /5Phos/TCCAGGCTACATGAGACATGCAGACATGCTCCAGTGTCATGAACCTCCattatcaggtctagattggatcttgctggcgcgtcc
R144_auts2 /5Phos/AGTCCAGCCGTTGTCCTAAGGAGGGAGCAGGCCACCTCAGACAGAAAATGCAGCTGGAGtctagattggatcttgctggcgcgtcc
R102_auts2 /5Phos/CCTCGiGCAGAAGAGGACATCATTGATGGatttctagattggatcttgctggcgcgtcc
R 110_auts2 /5Phos/GACGCCCCTGAC C AAGAAGAAACGAGAAGCctctagattggatcttgctggcgcgtcc
R118_auts2 /5Phos/ATCCAAGGGCTTCCACCGGAGCAGCTCatatttctagattggatcttgctggcgcgtcc
R126_auts2 /5Phos/CTGGCCATGTAAGGAGATACAGGGAACgattatcaggtattctagattggatcttgctggcgcgtcc
R130_auts2 /5Phos/GCACCTTGCTGGACTGACCTGAGGAAGAGCttgaatgcgagttattatctctagattggatcttgctggcgcgtcc
R134_auts2 /5Phos/TGTTCCCCTTGCTGTGTAGAAACAGATGCAGTCAGACCCACATAAGCTtctagattggatcttgctggcgcgtcc
R105_auts2 /5Phos/GTTTCAGTACATCCCGTCCAGCTTCCAGATTctctagattggatcttgctggcgcgtcc
R138_auts2 /5Phos/CTACATTCACAGGCCTAGCAGCAGTTGGTGGCAATGCCTTCGGGGGACTTGGAAATCCTTCCtctagattggatcttgctggcgcgtcc
R107_auts2 /5Phos/CCAAGTCCCTGTTAGCGTGTCCAAACAGAGtctagattggatcttgctggcgcgtcc
R136_auts2 /5Phos/CGTTTCCTGATGTGACGCACTGTATTCCAGCTGGTGATCAAGTCTGGGAAtctagattggatcttgctggcgcgtcc
Table 7
Overview of all the clinical and genotypic data, including the published translocation patients. This table gives an overview of all genotypic an phenotypic features of all probands of which detailed information was available and their family members carrying the AUTS2 deletion, idel: intragenic deletion, del: deletion that includes (a part of) AUTS2, trans:
translocation with one of the breakpoints in AUTS2, inv: inversion with one of the breakpoints in AUTS2, - (in x): no exons involved but intron x deletion, br. in x: breakpoint of the translocation or inversion in intron x. x y x m: x years and x months old, NA: not applicable, ND: not determined, m: male, f: female. The numbers in the table from 1-94 correspond to the row description (e.g., row 1 corresponds to "first author") centrum/first author ohio ame
CI 1-728 GC43007 (son of local reference number/code AUTS2_009 AUTS2_014) former reference number manscript
vl 21 26
4
5 family number (new) 2
6 METHODS
7 Array
array platform (initial) Roche Roche NimbleGen Roche NimbleGen 135k
NimbleGen 135k (custom (custom design 135k (custom design signature) signature) design signature)
9 GPL number (see GEO website)
10
11 conformational studies
12 confirmed by MLP A
13 confirmation by FISH
14 confirmation by QPCR
15 confirmation by high density array
16
17 GENETIC RESULTS
18 AUTS2 defect
19 type of AUTS 2 abberation idel idel idel mean breakpoints array chr7:68820750- chr7:69041764- chr7:69134595- 68912659 69186314 69202305 minimal breapoints array chr7:68,820,750- 68,912,599 maximal breakpoints array chr7:68,809,325- 68.917,095
Exons involved (MLP A/high
density array) (in2) (in2) outframe deletion causing
frameshift
aminoaccids missing family testing results
de novo ? (not mat)
segregation with phenotype in the
family ? other genetic defects
Translocatie/inversie partner NA NA NA
Other genes involved other CNV's/second hits 9q24.3 del Xpl l . l dup (inherited)
CLINICAL FEATURES
general
Age at examination(yrs) 6y4m 5y8m 5y3m Sex m F m growth and feeding
Low birth weight
Short stature <pl0
microcephaly <p2 +
Feeding difficulties (reflux)
Neurodevelopmental disorders
Intellectual
disability/Developmental delay
Autism/ Autistic behaviour +
Sound sensitivity ND ND hyperactivity/ ADHD
other neurodevelopmental disorder
Neurological disorders
generalysed hypotonia
Epilepsy
Brain disorder ND +(assymetric) ND cerebral palsy/spasticity +(assymetric)
other neurological disorder ACC, brain
assymetry,
hydrocephalus Dysmorphic features
Highly arched eyebrows ND
Hypertelorism ND
Proptosis ND
Short palpebral fissures ND
Upslanting palpebral fissures ND
Ptosis ND
Epicanthal fold ND
Strabismus
Prominent nasal tip ND
Ante verted nares ND
deep/broad nasal bridge ND
Short/upturned philtrum ND
Micro/retrognatia ND
Low set ears ND
narrow mouth ND
other dysmorphic feature nasal dermoid short neck, shawl sinus scrotum
Skeletal disorders
Kyphosis/ scoliosis
Arthrogryposis/shallow palmar
creases
tight heel cords ND other skeletal feature . Foot, clinodactyly Congenital malformations
hernia umbilicalis/inguinalis
heart defect patent foramen ovale
other features premature birth soft palate cleft exposure to alcohol and alcohol exp. (not objectified) antidepressants prenataly
recurrent tear duct
earinfections hypoplasia optic nerve
hypoplasy (ri)
hip dysplasia
AUTS2 disruption likely pathogenic 93 AUTS2 syndrome severity score/ 3/16 5/30 5/29 total number of scored items
94 (featurs with other in title not (assymetric + not
scored) counted)
Table 7, continued. The row heading will be indicated in the continuing table only by the reference number. ame gent utr utr harvard AUTS2_014=mother of
GC43007 2 victor fath CHB23
26 5 2 2 33 mother of p3 4 5 father of p5 6
Roche NimbleGen 135k Agilent 180K Agilent 105K Agilent 105K Agilent 244K (custom design (custom design (custom design
signature) Oxford) Oxford)
9
10
11
12
13
14
15
16
idel idel idel idel idel chr7:69134595- chr7:69288622- chr7:68900000- chr7:68900000- chr7:69153079- 69202305 69475350 69050000 69050000 69248071
21 chr7:69,288,622- chr7:68,906,975- chr7:69,158,443- 69,475,409 69,053,679 69,251,631
22 chr7:69,266,221- chr7:68,890,372- chr7:69,147,715- 69,507,743 69,070,952 69,257,044
23 (in2) (in4) 2 2 3-4 24
25 0 0 71 71 46 26
27
28
29 NA NA NA NA NA
Xpll.l dup
28y 8y 3y ND 3y f m m m F
ND ND ND
mild
+(self-described) +
ND ND
ND ND ND +
+ white matter abn.
- . . . . - . . . . . . . . . - . . . . - . . . . . . . . Left . . . . . . . +
. . . . - . . . . - - . . . - . . . . - - . . . 75
76
77
78
79
80 . - - ND
81 . - - ND
82 ND ND - - ND 83
84
85
86
87
88
89
90
91
92 + 93 1/29 3/31 1/31 0/31 4/26
94 (assymetric + not counted)
Table 7, continued.
1 ohio eng eng ame Eng 2 C10-330 case 3 case 3mother AUTS2_018 case 2sib 3 31 7 4
5 mother of p8 10 6
7
Roche Affy 2.7M Roche Affy 6.0 NimbleGen 135k NimbleGen
(custom design 135k (custom
signature) design signature) +
+ +
+ +
idel idel idel del Idel chr7:69186254- chr7:69211000- chr7:69211000- chr7:67056711- chr7:69495000- 69407271 69263000 69263000 69323591 69827000 chr7:69, 186,254- chr7:69,211,398- chr7:67,063,120- chr7:69478104- 69,407,211 69,263,338 69,321,364 69827159 chr7:69, 180,268 chr7:69,209,046- chr7:67,001,077- chr7:'69475784- 69,413,687 69,264,563 69,322,899 69827248
3-4 3-4 3-4 1-4
+
46 46 46 230
+
+
NA NA NA NA NA
2y6m 2y4m 22y ly7m 2y3m m f f f F
+ ND + + (p20) + + ND + +
+ + + + borderline/mild + Mild + - - +
ND ND - ND ND - - - - -
- +
- - - - ND ND - ND - - - - -
+ +
+ + + + +
+ +
+
+ + + + ND + ND + + + plachiocephaly, low columella brachycephaly,
facial assymmetry,
prominent forehead
ND ND ND ND
mild pectus
carinatum
atrial septum defect 89 unilateral cleft lip cleft lip
90
91
92 + + + + +
93 6/31 7/30 5/27 11/29 16/29
94
Table 7, continued.
1 eng eng ohio ame Vu 2 case 2 mother sibs C09-3214 AUTS2_013 Jesica 3 7 7 25 1 4
5 sibling of plO mother of plO 11 12 13
6
7
Affy 6.0 customized agilent Roche Agilent 105K
105k (custom design NimbleGen (custom design signature) 135k (custom oxford) design
signature)
9
10
11
12
13
14
15
16
17
18
19 idel idel idel idel Idel 20 chr7:69495000- chr7:69495000- chr7:69612584- chr7:69654259- chr7:69596512- 69827000 69827001 69827896 69828289 69878246
21 chr7:69478104- chr7:69,612,584- chr7:69,612,584- 69827159 69,827,895 69,858,682 chr7:'69475784- chr7:69,548,807- chr7:69,596,512- 69827248 69,895.670 69.878,246
5-6 5-6 6-9 + + + +
27,3 27,3 333
+ ? (not pat)
+ + +
NA NA NA NA NA
6y 31y 5y6m 11m 32 m m m F
+ ND ND
+ + + + + + + + + + (reflux) + mild/moderate + Moderate
-? + ND ND ND
+
ND ND ND ND
+
+ + + + + +
+ + downslant +
+
ND - +
+ + + + + - +
large hand and short forehead feet
. Limitation
le arm - - - +
ND ND + ND ND
sensorineural deafness
90
91
92 + + + + +
93 16/29 8/28 9/27 8/31 22/30
94
Table 7, continued.
1 can ame harvard can Harvard
2 case 1 AUTS_002 CHB44 case 2 CHB30 10 14 34 11 32 14 15 16 17 18 Affy 6.0 Agilent 105k Agilent 244K Illumina Omni Agilent 244K
(custom design 2.5M array signature)
idel idel idel del Del chr7:69544241- chr7:69699592- chr7:69842425 chr7:69861129- chr7:67771230-
69874867 69828036 69895671 71574626 70363865 chr7:69,544,961- chr7:69,842,425- chr7:67,771,230- 69,869,585 69,895,730 chr7:69,855,702- 70,370,488
70,057,754
chr7:69,544,254- chr7:69,827,895- chr7:67,703,093- 69,874,855 69,902,739 chr7:69,855,618- 70,376,527
70,059,580
6-11 6-18 7-19 7-19 All
+ NA NA NA
380 612,3 1011 1011 1259
+ + ? (not mat) + + + + +
NA NA NA NA NA WBSCR17
CALN1 WBSCR17 dup 2.. Inh. 2ylm lyl lm 7y2m 5y7m 9ylm f f f m F
+ - + - +
+ + - (p20) + - (p25)
+ + - + - (p30)
- - + + - mild/moderate mild + severe +
- - + + -
ND ND ND ND +
- - - - +
allmost no
speech
- + + + -
- - - - -
+ - + - -
+ + + + + hyperintense ataxic gait
signal WM
periventricular
- + - + -
- - + + -
- + - + -
- - - - -
- - - - -
- - - + -
+ + - + -
+ - - - -
+ - - - -
+ - - - -
- - - + +
- - - + -
- - - + -
- - - - -
+ - - + - earpit prominent lips large torn
mild 82 ND ND
83 metopic synostosis clinodactily dig
V
84
85
86
87 patent foramen
ovale
88
89 sacral dimple premature birth
90 intraventricular
hemorrhage
91
92 + + + + +
93 14/31 8/30 9/31 18/30 7/32
94
Table 7, continued.
1 liverpool harvard gent Bakk. Huang 2 CA M09-0400 DGAP201 1
3 0 4
4
5 19 20
6
7
Bluegnome Agilent 180K
Cytochip ISCA
60K
NA
9
10
11
12 NA
+
13 NA
+
14 NA
15 NA del inv trans trans chr7:68059522- chr7:69323903- 71060526 69323913
NA
NA NA
NA NA
all br. in 4 br. in 6 5/6 1/2
NA NA NA
1259 NA NA
+ + + + + + + + + +
NA 7q36.3 22ql2.1 7q35 6ql4 WBSCR17
CALN1 PTPRN2 CNTNP2 HTR1B?
4y9m 6y8m ly8m 3,5 6 m f f m m
+
+ +
+ + + p 10/25 + + + mild mild/moderate moderate mo? mi/mo?
+ + + + ND ND +
ND + + + + - - - - - - ND - + ND + - - + - delayed
myelination
+
+
+ +
+ + downslant
+
+ +
ND
+ + + +
+ + + + + + +
+ +
+ +
prominent fetal pads toes
cheeks
ND - - ND ND - + - ND ND + (li) ND ND + ND camptodactyly dig V
- - - + - patent foramen - - ovale, ASD Hypospadias conductive
hearing loss eventration of
diafragm (re) 91
92 + + + + + 93 15/31 1 1/29 17/31 9/28 10/27
94 (assymetric +
not counted)
Table 7, continued.
Sultana, d/1 Sultana, d/1
Kalscheuer Kalscheuer Kalscheuer Barra Barra
2 1 Ί 3 TwinA twinC 3
4
5
6
7
9
10
11
12
13
14
15
16
17
18
19 trans trans trans trans trans 20
21
22
23 5/6 2/5 5/7 2/3 2/3
24
25 + + + + + + + + + +
3p21.3 13q22 llpll.2 20qll.2 20qll.2
LYZL4 KLF5/PIBF PRDM11/SYT13 ND ND
6 27 10 16 16 m f m f f + + - - - + - + + + ND ND ND + ND + + + ND ND se mi mo se se ND ND ND + +
+ - ND ND - - + ND ND
+ + + ND ND + - - + + + ND ND + + ND ND ND ND ND
- + - ND ND
- - ND ND + + - ND ND - + +? ND ND - + downslant ND ND - + + ND ND
- - ND ND - - + + + - ND ND - + - ND ND - - - ND ND + + - + + + - - ND ND + - - ND ND + + + ND ND
+ +? ND ND + ND ND ND ND ND ND ND
optic nerve juvenile catheract
hypoplasia,
microphthalmia
hearing loss hypospadias
+ + + + +
15/28 15/27 8/27 8/12 7/11
Subjects and methods
Patients
Routine diagnostic array CGH and karyotyping was performed for intellectual disability (ID)/ multiple congenital anomalies (MCA) for a total of about 50,000 patients in ten diagnostic centers in the Netherlands, Belgium, Great Britain, the USA and Canada. Peripheral blood samples of patients with an AUTS2 aberration were obtained for further studies after informed consent of the patient or parents where appropriate. Clinical information was collected through the referring physicians and with approval of the local medical ethical committee. Informed consent was obtained from parents for publishing photographs. Patients were included in this study when detailed phenotypic an genotypic data were available.
AUTS2 syndrome severity score
The clinical data of patients and carrier parents were used to identify features that occurred in at least two unrelated patients. These features were added to the list of clinical features characterizing the AUTS2 syndrome for which all patients were scored positive or negative. Asymmetrically occurring features were not counted as positive. The sum of positive features was counted for each patient and was defined as the AUTS2 syndrome severity score, which was used to investigate a genotype- phenotype correlation.
Controls
Several control groups were used to assess the frequency oiAUTS2 deletions within a larger general population. A total of 4,783 DNA samples from the Welcome Trust Case Control Consortium 2 (WTCCC2) were analyzed with SNP array. This control group includes individuals from the 1958 Birth cohort and the UK Blood service collection (https://www.wtccc.org.uk/ccc2/, 10-26-2011) that have been nationally ascertained and regarded healthy14. Furthermore control CNV data from 8,329 cell line and blood-derived controls were obtained primarily from genome-wide association studies of non-neurological phenotypes. As these include 2009 controls from the UK Blood service collection this set adds only 6,320 unique controls. Although these data were not ascertained specifically for neurological disorders, they consist of adult individuals providing informed consent as described previously15. In addition publicly available data from Hapmap phase 3, which consist of 1,056 healthy controls from 11 different populations were checked for deletions involving A UTS216
(http://hapmap.ncbi.nlm.nih.gOv/downloads/raw_data/hapmap3_affy6.0/, 10-26-2011). CNV data from four control sets from respectively Canada, Germany, the USA and the Netherlands were available: The Ottawa Heart Institute (OHI) controls (n=1.234); POPGEN controls (n=1.123), SAGE controls (n=1.287) and the Low-Lands- Consortium controls (n=981, a Dutch control cohort)16 18. See supplementary table S2.
Chromosome analysis
Chromosome analysis was performed on standard synchronized cultures of peripheral blood lymphocytes. In general GTG-banded chromosomes were analyzed at the 550- band level from five metaphases and the chromosomes were counted from ten metaphases. Array CGH
For the initial CNV analysis ten different designs were used on six analogous platforms. All signal intensities were analyzed in hgl8 (build 36). See supplementary table SI for platforms used per patient. In a subset of the patients validation and high resolution breakpoint delineation was performed using two types of custom Roche Nimblegen 135K microarrays with probes tiled across 7ql l.22 (hgl8; chr7:65, 992,311- 72,003,221) at a median density of 1000 bp (patient 9) and 75 bp (patients 2, 3, 12, and 15)19, see supplementary figure SI.
Array CGH labelling, hybridization and data analysis
Labelling and hybridization of Agilent, Affymetrix and Roche NimbleGen arrays were performed as described previously20 24. With the Infinium HumanOmni2.5-Quad vl.O BeadChip (Illumina Inc., San Diego, CA, USA) 2,443, 177 markers were genotyped according to the manufacturer's protocol and scanned with default settings using the Illumina iScan.
Data analysis for the Agilent platforms was performed with Agilent analysis software (CGH Analytics or Genomic Workbench Standard Edition 5.0.14, Agilent
Technologies, Inc.) or an in-house developed software tool arrayCGHbase
(http://medgen.ugent.be/arrayCGHbase/). The relative DNA copy numbers at the SNP/CNV loci from the Affymetrix platforms were determined by comparison of the normalized array signal intensity data for the proband's DNA sample against the HapMap270 reference file provided by Affymetrix, using Genotyping Console or ChAS software (default settings). All rare CNV's are checked using Nexus20. Analysis and intra-chip normalization of the Illumina image files was performed using lllumina's GenomeStudio Genotyping Module software v.2010.3 with default parameters.
Genotype calls were generated using the Illumina-provided genotype cluster definitions file (HumanOmni2.5-4vl_B.egt, generated using HapMap project DNA samples). CNV analysis was performed using a multi- algorithm approach, as described elsewhere25. Data analysis of the NimbleGen arrays was performed as described previously26.
MLPA
The exon content of intragenic deletions was verified by MLPA in all patients that were not confirmed otherwise. Probes were designed for exons 1 to 19 of the reference sequence oiAUTS2 (supplementary table S4). Oligonucleotides were synthesized by Integrated DNA Technologies (Leuven, Belgium); all other reagents were from MRC- Holland (Amsterdam, The Netherlands). The samples were separated on a 3730 automated sequencer (ABI Systems) and analyzed using Gene marker vl.95 software (Soft genetics).
Q-PCR
Presence of the intragenic deletion found in patient 8 was tested by Q-PCR on parental DNA samples. Primer3 was used to design the Q-PCR primers, that were ordered from Eurofins MWG Operon. DNA copy number changes were measured using the Rotor-Gene™ 6000 and Sensimix SYBR Green (QT650-05) from Bioline Ltd. The Q-PCR data were analyzed using the 2-AACt (comparative quantification) method as described previously21. The final dosage value for each experimental replicate was assessed from the 2_AACt values: scores < 0.75 are indicative of a deletion. Breakpoint sequencing
DNA from patient 20 (paracentric inversion) was sequenced using a customized whole-genome large insert jumping library for Illumina HiSeq2000 sequencing based on the mate-pair methods of Applied Biosystems SOLiD sequencing (Applied
Biosystems Inc)23> 24. The method sequences the ends of fragments separated by large inserts (~3kb in this case) of genomic sequence, yielding very high physical coverage of the genome by mapping the insert between paired-end reads. In brief, 20 μg of DNA was sheared and size selected around 3.5 kb, cap adaptors with EcoP15I restriction sites were ligated to the ends, and fragments were circularized with an internal oligonucleotide adaptor containing a subject- specific barcode and a single biotinylated thymine. Restriction enzyme digestion was performed, and biotinylated fragments containing the circularization junction were bound to streptavidin beads followed by standard library preparation using NEBnext reagents (New England Biolabs, Inc) and Illumina paired-end adaptors. Bioinformatic analyses were performed by aligning reads to the human GRCh37/hgl9 reference sequencing using BWA27, modified to accommodate long-insert outward facing reads and SAMtools28. BAM files were processed to identify rearrangement breakpoints by a freely available C++ program, BamStat (http://mappingtools.chgr.org), we developed to tabulate mapping statistics and output lists of anomalous read-pairs (defined as having ends that map to two different chromosomes, an abnormal insert size, or unexpected strand orientations). The breakpoint positions were converted to the GRCh36/hgl8 genome built to allow comparison with other patients in this article. Complete details of all protocols and bioinformatic analysis are available23. FISH
FISH analysis was used for translocation breakpoint mapping in patient 21. Locus specific FISH probes for chromosome 7qll.22 and 22ql2.1 were selected using the Ensembl and UCSC genome browser database (http://www.ensembl.org,
http://genome.ucsc.edu, 6 December 2010) (genome build GRCh37) (supplementary table S5). Using nick translation the probes were either labeled with SpectrumGreen or SpectrumOrange (Abbott Molecular, Belgium). Metaphase FISH with probes from the region shown to be deleted by array CGH was also used to visualize the deletions in patients 1, 2, 8, 9, 11 and 12, and their parents, where available, according to previously described methods29.
Evolutionary conservation
To determine the evolutionary conservation of human AUTS2 exonic sequences we used the following species for comparison: gorilla (gorGor3), macaque (Mmul_l), dog (Broadd2), cow (Btau_4.0), pig (Sscrofa9), mouse (NCBIM37), chicken (Washuc2), clawed frog (JGI_4.2) and zebrafish (Zf9). Two methods were used. We first aligned the predicted protein of the longest isoform in humans to the predicted amino-acid sequences of the orthologous species using the muscle software30. Therefore sequences from the latest builds were downloaded from Ensembl. The second method aimed to also detect similarity in non-annotated or non-coding genomic DNA and used the tblastn algorithm with the human amino acid sequence as query10. The degree of homology was calculated as the percentage of identical amino acids.
Identification of alternative transcription start sites
To investigate putative alternative transcription initiation sites we used human brain mRNA from the caudate nucleus and the medial frontal gyrus from one person provided by the Dutch Brain Bank. Procedures for rapid amplification of 5' cDNA ends (5'RACE) were performed with the Ambion FirstChoice® RLM-RACE kit according to the manufacturer's instructions. Nested PCR amplification was performed with 5'-atgtcttcggctgaaatgct-3' as the outer, and 5'-ggaagagactgtgccggtag-3' as the inner A UTS 2- specific reverse primer. Morpholino and embryo manipulations
Zebrafish (Danio rerio) embryos were raised and fish were maintained as described31. Splice blocker morpholinos (MOs) against auts2 were designed and obtained from Gene Tools, LLC (sequences available on request). We injected lnl of diluted MOs (4.5 ng for 5'MO targeting exon 2 donor splice, and 6ng for 3'MO targeting exon 10 donor splice) and/or RNA (100 pg) into wild type zebrafish embryos at the 1- to 2-cell stage. Injected embryos were scored at 3dpf and classified into two groups, i.e. normal and microcephalic, on the basis of the relative head size compared with age-matched controls from the same clutch. For RNA rescue experiments, the human wild type mRNAs, full-length and short isoform, were cloned into the pCS2 vector and transcribed in vitro using the SP6 Message Machine kit (Ambion). All the
experiments were repeated three times and we ran a t-test to determine the significance of the morphant phenotype and the wild type rescue.
Supplementary patient information
Patient 1 (intron 2 deletion, inheritance status is unknown)
Clinical features
Proband one is born prematurely at 34 weeks of gestation and had a birth weight of 1361 gram (<p3). His mother has used alcohol in this pregnancy and is mildly intellectual disabled. His father is diagnosed with schizophrenia. The little boy (now six years and 4 months old) started talking at 2,5 years of age. He is mildly
intellectual disabled and has behavioral problems. He was diagnosed with ADHD and Oppositional Defiant Disorder. He does not meet the criteria for autism. His medical history revealed recurrent ear infections and a result of this some conductive hearing loss of his right ear. Physical examination at the age of 6 years and 4 months showed a height of 120 cm (p68.7), a head circumference of 49 cm (p8.85 ) and obesity with a weight of 31.7 kg (p96.8). He walks with his toes turned inward.
Genetic analysis:
Diagnostic array (Roche Nimblegen 135K, custom design of signature genetics) revealed an small intronic deletion of the AUTS2 gene (chr7:68820750-68912659) and a 9q24.3 deletion (chr9:l, 802,429- 1,866,421, max) not containing any genes. Parents were not available for testing.
Patient 2 (intron 2 deletion; father not tested, mother: neg.)
Clinical features
Proband two is a five years and eight months old girl. She is born at term with a normal birth weight. She has a delayed psychomotor development and epilepsy. She had gastro-oesophageal reflux. Physical examination at the age of 5 years revealed borderline short stature (height at p3), microcephaly (headcircumference 45.5cm, <p2), a nasal deromid sinus and left sided muscle weakness and right sided spasticity. She is said to have soft cleft palate, but this was not objectified. A MRI of the brain was performed, which revealed colpocephaly, corpus callosum agenesis,
hydrocephalus, hypoplasia of the brain stem and an asymmetry of the cerebral hemispheres with a smaller hemisphere and an enlarged ventricle at the right. With ophthalmologic evaluation a tear duct hypoplasia and right sided hypoplastic optic nerve were noticed. She has amblyopia and exotropia of the right eye.
Genetic analysis:
With array analysis (Roche Nimblegen 135K, custom designed by Signature Genetics) a deletion in intron 2 of the AUTS2 gene was detected (chr7:69041764-69186314). The deletion was confirmed by FISH and high density array (Roche Nimblegen 135K, high density custom designed by ). The mother tested negative for this deletion, but the father was not available for testing. Patient 3: (intron 2 deletion, maternally inherited):
Clinical features
The proband in family three is a five years and three months old boy that is the first child of non-consanguineous parents. His mother used alcohol in the first two months and antidepressants in all months of this pregnancy. He was born 5,5 weeks early. He had a birth weight of 2353 grams (p50). He has aggressive and autistic behavior. The best fitting diagnose would be pervasive developmental delay with suspicion to progress to Asperger syndrome. Physical examination at the age of 5 years and 3 months revealed a height of 106 cm (p50), a head circumference of 51,5cm (p50) and a weight of 18,5 kg (p3- 10). He has short palpebral fissures, a low nasal bridge with a bulbous nasal tip and a highly arched palate. His neck is short, and he has a shawl scrotum. Examination of his extremities was remarkable for a clinodactyly of his little finger, lax ligaments, fat pad on the pointed finger and an inversion of the foot.
Neurological examination was normal. Echocardiogram revealed a small patent formamen ovale that closed spontaneously.
Genetic analysis:
His karyogram was normal, and a DNA test for fragile X was normal as well.
Microarray revealed two variants: a deletion in intron 2 of the AUTS2 gene (chr7:69134595-69202305) and a duplication of Xp ll. l (FAAH2 and ZXDA, chrX:57356516-57658457). The AUTS2 deletion was confirmed by FISH and high density array (Roche Nimblegen 135K, high density custom designed by... .)· Both variants were found to be inherited from his mother, who thinks that she herself has Asperger syndrome characteristics.
Patient 4 (deletion intron 4; de novo):
Clinical features
Proband four is the first child of non-consanguineous parents. He was born at term after a normal pregnancy and delivery. Anthropometric measurements at birth were within normal limits. His early developmental milestones were normal. At the age of three years he was noticed to present significant opposite behavior. Now at the age of eight years he has been diagnosed with an autism spectrum disorder (ASD). He is quiet and needs a well structured environment, but overall his communication skills are better than those of his younger brother who has also been diagnosed with ASD. He receives special education, because of his autistic behavior. His intellectual development is delayed, with a mild discrepancy between performal and verbal skills. Physical examination at age eight revealed a height of 134 cm (p50), a weight of 25,5 kg (p3) and a head circumference of 52 cm (p50). No obvious dysmorphic features were seen. An MM scan of the brain was normal. Additional imaging studies, including cardiac and abdominal ultrasounds, were also without abnormalities.
Genetic analysis
Array CGH (Agilent 180K) analysis revealed a de novo intragenic deletion in the AUTS2 gene. No other aberrations were noted. The deletion spans 187 to 242 kb in intron 4 of the largest A UTS2 transcript (and deletes exon 5 of the shorter alternative transcript: AUTS2-003
http://www.ensembl.org/Homo_sapiens/Gene/Summary?g=ENSG0000015832 l;r=7:69 063905-70258054, 16/7/2011) (chr7:69.288.622-69.475.350). Array CGH analysis of his parents and his affected brother yielded normal results. Patient 5 (deletion exon 2; paternally inherited):
Clinical features
This proband is the third child of non-consanguineous healthy parents. The first pregnancy of his mother ended in an IUVD eci. Both parents have no intellectual deficits nor autistic features and followed normal education. Pregnancy and delivery were uneventful. He was born at a gestational age of 41 weeks and 2 days with a birth weight of 4170 gram (p85). Early developmental milestones were mildly delayed. He started walking at the age of 18 months. At the age of 28 months he had a speech development comparable with a 16 month old child. There are (no) features of autism. Now at the age of 36 months, he is functioning at a developmental age of 23 months. Physical examination revealed a height 100 cm (p25) and a head circumference of 50.8 cm (p50). No dysmorphic features are evident (see figure 2A). Physical examination of the father showed no dysmorphic features and normal body proportions. Genetic analysis
Array-CGH, using a Lowlands consortium custom designed Agilent 105K platform, revealed an intragenic deletion of 150 kb with breakpoints at 68,900,000 (Probe A16_P17972091) and 69,050,000 (Probe A16_P17972516) removing exon 2 of the AUTS2-gene. This deletion leaves the reading frame in tact and causes a deletion of 71 amino acids. The deletion was inherited from his normal farther, who inherited the deletion from his normal mother/father.
Patient 6 (an exon 3-4 deletion, inheritance status unknown)
Clinical features
Patient six is a three year old girl. She is the second child of healthy non- consanguineous parents. The family history is unremarkable. She is born at term after an uneventful pregnancy and delivery. She has a delay in motor and speech development. She could sit up at the age of 10 months and started walking at 18 months of age. She received early intervention services including speech therapy and physical therapy, but has made little progress. Currently, her behavior has been most concerning as she has severe temper tantrums which might be related to her limited ability to express herself. Physical examination at the age of three years revealed a head circumference of 48.5 cm (p25). Height and weight measurements are not available. No dysmorphic features except for a left eye ptosis. She had a normal muscle-tone, but high deep tendon reflexes, with little asymmetry. Her triceps-reflex was +3 on the left and 2+ on the right, and the knee reflex was 3+ on the left and 2++ on the right. Brain MM has been performed and showed, by report, diffuse symmetric white matter abnormality in the periventricular region with some subcortical involvement.
Genetic analysis
Array analysis (244K Agilent array) showed a deletion of 0,5 Mb of exon 3 and 4 of AUTS2 (chr7:69, 153,079-69,248,071). Parents were not analysed so the inheritance status of the deletion is unknown. The array did not reveal any other rare CNV's.
Patient 7 (exon 3-4 deletion, inheritance status is unknown)
Clinical features
Proband 7 is an 2 year and 6 months old boy. He is the second child of healthy non consanguineous parents. His older brother was diagnosed with pervasive
developmental disorder. Except for recurrent ear infections and strabismus he had an unremarkable medical history. He is delayed in speech and language skills. He spoke his first words at 2 years of age and at this moment (2 years and 6 months old) he is still using single words. His motor milestones were normal. He started to walk at the age of 1 year. He shows repetitive stereotyped behavior and has impairment of social interaction and meets the criteria for autism. Physical examination at the age of 2,5 years revealed a slightly short stature (height at p lO), a weight at the 10th percentile and microcephaly with a head circumference at the 2nd percentile. He showed stereotyped behavior. There were no dysmorphic features noticed.
Genetic analysis
A Signature Genetics custom design Roche NimbleGen 135k array CGH was performed and showed an intragenic deletion of the AUTS2 gene involving exon 3 and 4 (chr7:69186254-69407271). Both parents and the brother of the boy were not tested, therefore it is unknown if the deletion is segregating with the autism spectrums disorder phenotype in this family. Patient 8 (exon 3-4 deletion; maternally inherited):
Clinical features
This proband is the second child of non-consanguineous parents. The mother of this girl was born with a non-midline upper cleft lip and although she was said to have had normal early childhood development, she struggled at mainstream school with reading and writing and needed extra help. Antenatally, cleft lip and probable cleft palate was detected with ultrasound. She was born at 42+2 weeks by normal vaginal delivery with birth weight 3.3 kg (p25-50). At birth, the cleft lip and palate were confirmed and a heart murmur was detected. Echocardiography revealed a atrial septal defect with left to right flow. The developmental milestones were delayed with smiling at 4.4 months, sitting up unsupported at 10.5 months, and walking at 22 months. Her speech was also delayed for which she was referred to speech and language therapy. Her behavior was not considered a problem and she did not display any autistic tendencies. Despite a good appetite and feeding well, there were early concerns regarding her growth, which resolved as she grew older. At the age of 15 months, physical examination showed a weight of 8.1kg (p i) and a head circumference of 46 cm (p 10-25). At the age of 2 years 4 months her height was 86.5 cm (p25-50), weight of 10.5kg (p3) and a head circumference of 47.3cm (p2-10).
Dysmorphic features were noted consisting of plagiocephaly, brachycephaly, facial asymmetry with the right eyes appearing larger than the left (which improved with age), left sided unilateral cleft lip repair, prominent forehead with thin lips, a relatively flattened nasal tip, short upslanting palpebral fissures and proptosis, overlapping 2nd, 3rd and 4th toes, capillary haemangioma over the occipital region of the head and over the sacrum. Heart sounds confirmed the presence of a quiet mid- systolic murmur over the left sternal edge. Her mother had evidence of her cleft lip repair, ptosis, retrognatia and a narrow mouth.
Genetic analysis
Array analysis performed using the Affymetrix 2.7M array showed an intragenic AUTS2 deletion. The deletion spans 52 kb and deletes exon 3 and 4 of the AUTS2 gene (chr7:69,211,000-69,263,000). By Q-PCR analysis the deletion was shown to be inherited from her mother. Mosaicism in the mother cannot be excluded. We are awaiting samples to arrive from the maternal grandparents for further analysis. Patient 9 (de novo deletion overlapping exon 1-4 of the AUTS2 gene, no other genes involved)
Clinical features
Proband nine is born at term. She is the first child of healthy non-consanguineous parents. Family history reveals no relevant diseases, birth defects or developmental problems. Pregnancy and delivery were unremarkable. Her birth weight was: 3090g (pl0-p25), and her head circumference at birth was 32 cm (p3). Her early motor milestones were obtained at a normal age; she was rolling over at six months, sitting alone at seven months and crawling at eight to nine months. Around the age of 15 months, however, a delay in development and signs of autism were noted. She was tested to have a developmental level of 5-9 months in most areas as well as decreased eye contact and was given the diagnosis autism. She has a history of low muscle tone and feeding problems, including reflux and constipation. She is sensitive to loud sounds. Physical examination at the age of 19 months showed a weight of 8.4 kg (<p3), a height of 72,5 cm (p20) and a head circumference of 43 cm (pO.l). She has highly arched eyebrows and short palpebral fissures. Her ears are somewhat large but normally placed. She has a wide nasal bridge, a short philtrum and a narrow mouth. She has a slight pectus carinatum. Extremities do not show any abnormalities.
Neurological evaluation revealed normal muscle tone. A MM scan of the brain (without contrast) was normal.
Genetic analysis:
Array CGH (Signature OS Chip) revealed a de novo microdeletion of 2.3 Mb at 7ql l.22, overlapping exon 1-4 of the AUTS2 gene (chr7:67056711-69323591). The deletion is not overlapping any other genes and was confirmed by FISH and custom high density array.
Patient 10 (exon 5-6 deletion; maternally inherited):
Clinical features
Family ten concerns two siblings with developmental delay, failure to thrive and dysmorphic features. The proband in this family is a little girl of now six years of age that is the second born child of non-consanguineous parents. The father had a Tetralogy of Fallot. The mother has had learning difficulties and feeding difficulties. She has short stature and mild dysmorphic features. The pregnancy of the proband was complicated by intrauterine growth retardation and polyhydramniom. She is born by normal vaginal delivery. Her birth weight was 2.38 kgs (p2-9). She had feeding difficulties as a baby. She achieved her milestones slightly delayed and at two years and three months she was speaking at a 12 - 15 month level. She attends mainstream school with additional support. Physical examination of her height and head circumference were below the 0.4th centile. She is hyperteloric, has a ptosis and a downslant of the palpebral fissures, a mild proptosis and has highly arched eyebrows. She has a broad nasal bridge, a bulbous nasal tip, simple ears, low columella, strabismus and retrognatia. Her older brother, now ten years old, had a similar antenatal and postnatal course. His birth weight was 2.26 kg (p0.4-2). He fed poorly and failed to thrive with difficulty weaning onto solids. His early developmental milestones were delayed. He smiled at four to five months, crawled at 18 months and walked at two years. At six years his speech was equivalent to a three to four year old and he attends a school for children with learning disabilities. He has Attention Deficit Disorder which has responded well to medication. Physical examination revealed short stature and microcephaly with a height and head circumference below the 0.4th centile, bilateral ptosis, strabismus, right preauricular ear pit, small mouth, short philtrum, retrognatia and limited supination of the left arm as a result of a radio-ulnar synostosis.
Genetic analysis
Array analysis was performed using the Affymetrix SNP6.0 genotyping array in the girl. An intragenic deletion of 332 kb involving exon 6 and 7 oiAUTS2 was found (chr7:69,495,000-69,827,000). The same 7ql l.2 deletion in the older brother was confirmed by Q-PCR. The mother carries the same deletion, confirmed by Affymetrix SNP6.0 array, and the father has a normal Q-PCR result. Patient 11 (exon 6 deletion, inheritance status is unknown)
Clinical features:
Proband eleven is a 5,5 year old boy. He is born after an uneventful pregnancy and had a birth weight of (p ). He did not begin to talk until he was 3 years of age. He is hyperactive and shows a global development delay. He is now able to talk in three to four word sentences. Physical examination at the age of 5 years and 6 months reveals a short stature (height: 106.5 cm, p lO) a weight of 14.7 kg (p25) and microcephaly (head circumference: 45.5 cm «p2, p50 for a 9-month-old). He has minor epicanthal folds bilaterally, mild downslant of the palpebral fissures a flattened nasal bridge, a slightly tented upper lip and a narrow mouth with down turned corners. He has tapering fingers but the creases are normal. He wears orthotics bilaterally at the ankles. He has spasticity noted at the ankles bilaterally with limited plantar and dorsiflexion. His right ankle appears to be more spastic than his left. His reflexes in the upper extremities are 3/4. His reflexes in the lower extremities are 4/4. Despite his ankle spasticity, his reflexes at the bilateral ankles are 2/4. He has a wide-based gait and tends to walk on his toes. He still is unsteady in his gait but showed progression. His muscle strength is grossly intact. Genetic analysis
Chromosome analysis revealed a normal male karyogram: 46, XY. Methylation testing for Angelman syndrome was normal and Fragile X DNA testing was normal too. Array CGH (Agilent 105K) identified an intragenic AUTS2 deletion (chr7:69612584- 69827896) deleting exon 6. This deletion causes the AUTS2 sequence to go out frame from exon 7 onwards. It is unknown if the deletion occurred de novo as the parents are not yet tested.
Patient 12 (de novo deletion of exon 6):
Clinical features
Proband 12 was born at term and has healthy non-consanguineous parents.
Pregnancy and delivery were uneventful. His birth weight is 3033 gram (which is in the normal range). This little boy has reflux but furthermore feeds well. He was seen because of spasticity and developmental delay and was placed on a muscle relaxant. He is too young to determine autism or autism spectrum disorder. Physical examination at the age of 11 months revealed a short stature (height:... . p..), microcephaly (head circumference: ... p..) and dysmorphic features. He is
hyperteloric, has epicanthal folds and micrognatia. His ears are large and low set, and he has large hands and feet. Neurologic examination was remarkable for increased deep tendon reflexes and hypertonia of both legs. A MM of the brain was normal.
Genetics:
Array CGH (Roche NimbleGen 135k, a Signature Genetics custom design) was performed and showed a small intragenic deletion of the AUTS2 gene involving exon 6 (chr7:69654259-69828289). This deletion causes the AUTS2 sequence to go out frame from exon 7 onwards. The deletion was visualized by FISH and confirmed by high density array (custom design Roche Nimblgen 135K). Both parents of the boy tested negative for the deletion, which implies the deletion to be de novo.
Patient 13 (deletion exon 6-9, father tested negative, mother not tested):
Clinical features
Proband 13 is the first child of non-consanguineous parents. She was born at a gestational age of 42 weeks after an uneventful pregnancy, but had a slow start (with an apgar score of 5 after 1 minute). Blood gas parameters were normalised one hour after birth. She had a birth weight of 3690 gram (p55). In the neonatal period she was tachypnoeic, hypotonic and inactive. In the first months of her life she had feeding difficulties, failure to thrive and needed tube feeding. Physical examination at that time revealed low muscle tone, an upslant of the palpebral fissures, strabism and an expressionless face. At childhood (7- 10 years) she suffered from recurrent upper airway infections. Furthermore autistic behavior was noticed and she had a developmental delay. At the age of 11 years a scoliosis was diagnosed. In the following years she developed progressive kyphosis as well. She came to our attention at the age of 32 for a diagnostic evaluation of the intellectual disability. At that time she made no eye contact and used few words and short sentences. Clinical evaluation revealed a low frontal and dorsal hairline, a short forehead, highly arched eyebrows, short palpebral fissures, ptosis, a prominent nasal tip, a short and prominent philtrum, retrognatia and a kypho-scoliosis. Furthermore she has slender hands and feet, a sandal gap and absent creases at DIP joints of digitus IV, the other palmar creases of the hands are very shallow (see figure 2A). Her height is 153.5 (pO. l), her weight: 56 kg (p80) and her head circumference is 51.5 cm (pi). Genetic analysis
Chromosomal analysis was repeated as a karyogram performed just after birth was said to have shown a mosaic monosomy 21, the original results were however not preserved. The karyogram did not reveal any abnormalities in 30 cells. Further genomic analysis by array CGH revealed an intragenic deletion of the AUTS2 gene on chromosome 7qll.22 (chr7:69596512-69878246). Because of a gap between the deleted and not deleted probes the exact distal breakpoint could not be defined. Further breakpoint analysis by MLPA revealed a deletion of exon 6 to 9, which results in a deletion of 333 amino acids of the Auts2 protein (999 bp) leaving the reading frame in tact. Array CGH of her normal father showed no deletion in AUTS2, material from the mother was not available.
Patient 14 (exon 6- 11 deletion; de novo):
Clinical features
Proband 14 is a 2 years and 11 months old girl with developmental delay. Family history was unremarkable. She was born at term and had a birth weight of 2,55 kg (<p3) She had an inguinal hernia, which was successfully operated at the age of 3 months. At the age of 5 months there was a choking incident for which she was evaluated in hospital. TORCH- and metabolic screen was normal, as was the EEC. At the age of 11 months an infected preauricular sinus was operated. Although autism was suspected at a younger age, during evaluation at the age of 25 months there was no clinical suspicion of autism but she had a mild global developmental delay of 3/9 months. On physical examination at that age a height of 80.5 cm (plO), a weight of 9.8 kg (p3) and a head circumference of 44.4 cm (just below the p3) was measured. She drooled intermittently. Aside from mild epicanthus, she was not dysmorphic.
Intermittent right esotropia was noted. Her neurologic exam was remarkable for increased tone in the left upper extremity, Modified Ashworth Score of 1+, hamstrings 1+ on the left and 1 on the right, adductors 1+ and left gastrocs 1. Deep tendon reflexes were 3+ in the lower extremities and 2+ in the upper extremities. No clonus was present at the ankles, and plantar responses were downgoing. She has full range of motion of all joints except the left heel cord which is very mildly restricted in dorsiflexion. Now at the age of 35 months she uses gestures and a few signs. She has fewer than 10 spontaneous words and was noted to be highly distractible. Her tone and reflexes were normal throughout, with only mild restriction of dorsiflexion in left ankle remaining. Brain MRI at 13 months showed nonspecific hyper intense signal changes in the white matter in the periventricular zones at fron to-parietal areas. The lateral and third ventricles were prominent. MRS and MRA were normal.
Genetic analysis
Rett syndrome testing (by sequencing of MeCP2) was negative. Array analysis using Affymetrix SNP array 6.0 showed a deletion of 7qll.22. The minimal deletion spans 330 kb of the AUTS2 gene and includes exons 6- 11 (chr7:69,544,241-69,874,867). The deletion was confirmed to be de novo by Q-PCR and FISH of the proband and her parents.
Patient 15 (deletion exon 6-18, inheritance status is unknown)
Clinical features
The 15th patient is an 1 year and 11 months old girl with a mild developmental delay. She was born at term and had a normal birth weight. Her medical history was remarkable for a metopic suture stenosis. She is mildly delayed in general
development with a more prominent speech delay. Her hearing was tested normally. She has no behavioral problems. Physical examination at the age of 1 year and 11 months revealed short stature with a height on the 3rd- 10th percentile and
microcephaly with a head circumference of 44 cm, (<p l). She had some mild dysmorphic features, namely elevated eyebrows, proptosis and epicanthal folds.
Neurological examination revealed mild hypertonia. A CT-scan of the brain did not reveal any abnormalities.
Genetic analysis
Array CGH (Agilent 105k custom designed by Signature Genetics) revealed an intragenic microdeletion in AUTS2, deleting at least exon 6 (chr7:69699592- 69828036). The deletion was confirmed by FISH and High density Array (custom design Roche Nimblgen 135K). With High density array it was clarified that the deletion contains exon 6 to 18 of the AUTS2 gene. Patient 16 (deletion exon 7-19, de novo):
Clinical features
Proband 16 is a 7 years and 2 months old girl. This little girl was born premature. In the neonatal period an intraventricular hemorrhage was noticed. She had some feeding difficulties and development was delayed. She has had one febrile seizure, but no signs of epilepsy at this moment. At the age of 6 years she was almost non-verbal, using some signs and communication aids. She was functioning at a developmental level of a 2.5 years old child. She meets the criteria for PDD-NOS. She is a picky eater and just stays inside the normal range with her weight with a lot of effort from her mom. Physical examination at the age of 6 years revealed a height of 110.7 cm (p20), her weight was 17.5 kg (plO), and her head circumference 50 cm (p50). She has only fleeting eye contact. Her extra ocular movements were intact. She is hyperteloric. Neurological examination showed an increased muscle tone in her lower extremities. Her DTRs were difficult to elicit in her lower extremities but were 1+ and symmetric in the upper extremities. There was no clonus and her toes were down going.
Metabolic screening revealed an elevated C5-DC acylcarnitine, which suggests the diagnosis of glutaric aciduria type I (GA1). This was however excluded with DNA testing of the Glutaryl Co-A Dehydrogenase gene. MRI of the brain showed somewhat large ventricles and a mammillary body atrophy. No white matter abnormalities were observed. There was no evidence of neonatal brain damage.
Genetic analysis :
Microarray was performed (using the Agilent 244K array CHG) and demonstrated a deletion of exon 7-19 of the AUTS2 gene (Chr7:69842425-69895671). Additional testing did not show this deletion in both parents, indicating that it is a de novo deletion.
Patient 17 (exon 7- 19 deletion incl WBSCR17 and CALN1; mother tested negative,, father was not tested):
Clinical features
The 17th patient is a boy of now 5 years and 7 months of age. He is the child of non- consanguineous healthy parents. His mother is born premature and had Rhesus disease at birth and a developmental delay. He was born full term and had a birth weight 3520 gram (p50). Pregnancy and delivery were uneventful. He has been successfully operated for an umbilical and an inguinal hernia. A g-tube was placed, because of significant failure to thrive. The developmental milestones were
delayed/normal, he walked at the age of 2 years and 8 months. Now at the age of 5 years and 7 months he suffers from severe developmental delay with autistic features. He has no spoken language, but uses non-verbal communication including babbling and pointing to items. Physical examination (at the age of 5 years and 7 months) revealed a height of 104.8 cm (p5), a weight of 14.9 kg (just below p3) and a head circumference: 47.8 cm (<p2). Patient was non-communicative and moderately co- operative. He was noticed to have hypertelorism, high arched brows, mild epicanthal folds and mild ptosis. He has a broad nasal bridge. The philtrum is somewhat short and the mouth is narrow, the lips prominent and he has large central incisors.
Neurological examination was remarkable for abnormally low central muscle tone. Cardiovascular examination revealed a murmur that appeared to be functional in nature and a hemodynamically stable patent foramen ovale with small left to right shunt. The boy was diagnosed with mild cerebral palsy with severe cognitive deficits. MRI results revealed a mild prominence of the ventricular system and extra-axial spaces but were otherwise unremarkable. Genetic analysis
He was tested negative for Smith-Lemli-Optiz syndrome (normal 7- dehydrocholesterol), Angelman/Prader Willi syndrome (methylation analysis), and for subtelomeric screening and Fragile X DNA testing. Test results were also negative for defects in peroxisomal fatty acid metabolism. Array analysis (Illumina Omni 2.5M array) however showed a deletion of 1,65 Mb disrupting exons 7- 19 oiAUTS2 and the two downstream genes WBSCR17 and CALN1 (chr7:69,895,671-71,547,312). The mother did not have the deletion. The father was not available for testing.
Patient 18 (a de novo deletion of the complete AUTS2 gene and the WBSCR17 gene) Clinical features
Proband 18 is a 9 years and 1 month old girl. She was the third child of healthy non- consanguineous parents. She has two healthy siblings. Apart for seizures treated with medication in a paternal cousin there are no relevant disorders in the family. The pregnancy was uncomplicated except for small fetal size. At birth, she weighted 2523 gram and was determined to be small for gestational age. Weight gain was followed closely and she did well. She has a developmental delay with relative strength in gross motor skills. She has a history of hypersensitivity to loud sounds. Physical examination reveals a head circumference of 50.5 cm (p25). Her height was 131 cm and weight was 27.3 kg, both at the 30th percentile for age. She has a large tongue and tends to hold her mouth open. She has a flat nasal bridge, incurving 5th finger on the left and very small fingernails. She shows asymmetric developmental posturing on stressed gaits (left-sided). Muscle and strength were normal. Heel cords were slightly tight but both ankles could be flexed to neutral position. Tone throughout was otherwise within normal limits. Deep tendon reflexes were 2+ throughout and symmetrical. Toes were down-going.
Genetic analysis:
Array CGH (Agilent 244K) revealed a deletion of 2.4 mb, deleting AUTS2 in total and part of WBSCR17 (chr7:67, 771,230-70,363,865). Parental analysis revealed the deletion to be de novo. (This patient was submitted to the Decipher database, https://decipher.sanger.ac.uk/patient/251781, 9 November 2011) Patient 19 (de novo deletion of the complete AUTS2 gene and the WBSCR17 and CALN1 genes)
Clinical features
Patient 19 involves a little boy of now 4 years old. He was born at term after a normal pregnancy and delivery and had a good start. His birth weight was 3658 gram (p50). Because of severe feeding problems he was admitted to the special care baby unit. He needed tube feeding. He was then discovered to have a diaphragm eventration for which he was operated when he was two weeks old. Cardiologic evaluation revealed a atrium septum defect. Further more distal hypospadias was noticed. Because of severe feeding problems a percutaneous endoscopic gastrostomy (PEG) tube was placed. On physical examination at the age of 21 months his height is 81 cm (p9), his weight 11, 1 kg (p9-25) and his head circumference 47,6 (p2). He is slightly
hyperteloric, has short upslanting palpebral fissures, a broad nasal bridge, a short philtrum, a narrow mouth and a micrognatia. He is hypotonic. Evaluation at the age of two years revealed that he is still struggling with his feeding and is still being fed through his PEG tube. He seems to be delayed in motor and speech development. He was cruising but not walking yet and he had about 10-15 words. He is sensitive to loud sounds. He was tip toeing especially on the left and an increased tone around his left ankle and slightly brisk reflexes were noted at that time. Further neurologic examination was unremarkable. During re-evaluation at three years of age he showed improvement. He does however show some evidence of dystonic stiffness of his left ankle. Dysmorphic features were more or less unchanged. An MM of the brain showed no abnormalities.
Genetic evaluation
Chromosomal analysis was normal, as was a methylation test of the prader willi region on 15q. With qPCR for subtelomeric deletions or duplications a
deletion/duplication of the 4q/p was found, which she inherited from her normal farther. It is therefore unlikely to be involved in the clinical features of this proband. BAC array however revealed a deletion of 3 to 3.28 Mb on 7qll.22. Confirmation with a Bluegnome Cytochip ISCA 60K showed that this deletion involved AUTS2, the WBSCR17 gene and the CALN1 gene (chr7: 68059522-71060526). This deletion was not found in one of the parents suggesting that it is de novo.
Patient 20 (de novo paracentric inversion of chromosome 7: 46,XX,inv(7)(ql l.23q36.3), one
breakpoint in intron 5 of the AUTS2 gene and in the PTPRN2 gene) Clinical features
This little girl, now 6 years and 8 months, was born at full term with a birth weight of 3200 gram (p25). She is the second child of healthy non-consanguineous parents and has a older brother and younger sister that are both healthy. Apart from the fact that her mother has had three miscarriages the family history is unremarkable. She had feeding difficulties characterized by poor suck and reflux. At the age of 17 months she still had a failure to thrive, her growth parameters remaining below the 3rd percentile. Mild generalized hypotonia was noted by her physiotherapist. Her milestones:
smiling at 3 months, sitting at 11 months, standing at 12 months and walking at 27 months were (mildly) delayed. At the age of two years and five months she had a vocabulary of about seven words. Her head circumference was about 3rd percentile, weight 9.8 kg (<p3), height 85.7 cm (p25). She was diagnosed with mild autistic spectrum disorder and mild attention deficit disorder and she is having hearing problems. At the age of 6 years and 8 months here gait was evaluated because of frequent falls. Physical examination at that time revealed a height of 118 cm (p20), a weight of 16.8 kg (<p3). The head appeared microcephalic with a head circumference of 49 cm (p2- 10). She has loose jointed upper and lower limbs except for Achilles tendons, which are tight. She has a flexion contracture of the little finger on both hands. Her feet have high arches, they are internally rotated, and she is toeing.
Neurological evaluation: the reflexes are present at knees, but hard to obtain in upper limbs and ankles. She has mild dysmorphic features, namely: short palpebral fissures with a mild upslant and a short philtrum and narrow mouth. Genetic analysis:
Chromosomal analysis revealed a paracentric inversion of chromosome 7
(46,XX,inv(7)(qll.22q36.3)), which was found to be de novo after testing both parents. The exact breakpoint was sequenced. One breakpoint deleted 5 nucleotides in intron 9 of the PTPRN2-gene (157577848- 157577842) and the other breakpoint was in intron 5 of the AUTS2 gene, deleting 9 nucleotides (6968596-69685977).
Patient 21 (translocation (7;22), intron 6; de novo):
Clinical features
The 21st proband is the first child of non-consanguineous healthy parents. The pregnancy and delivery were uneventful. She was born at 37 weeks of gestation and had Apgar scores of 9 and 10 after 1 and 5 minutes, respectively. Birth weight was 2740 grams (p30). Due to hypoglycemia and feeding problems she was admitted in the neonatal intensive care unit for three weeks. Feeding remained difficult during the first months after birth. At the age of five months she had an axial hypotonia and a failure to thrive. Physical examination at that age revealed mild dysmorphic features; low-implanted ears, highly arched eyebrows, a broad nasal bridge, hypertelorism, short palpebral fissures and epicanthic folds and a full lower lip (see figure 2D). She had a height of 58 cm (pi), a weight of 5070 grams (pi) and a head circumference of 39 cm (p2). Clinical examination at the age of 21 months revealed a height, weight and head circumference of respectively 74 cm (<p l), 8006 g (p3) and 44,9 cm (p2), overall hypotonia, pelvic instability and dysmorphic features as described before. She had a developmental delay and some features of autism.
Genetic analysis
Conventional karyotyping revealed a de novo balanced translocation between chromosome 7 and 22; 46 XX, t(7;22)(qll.22;ql2.1). Array CGH analysis (Agilent platform 180K) was normal. To localize the breakpoint on both derivative
chromosomes, a systematic series of FISH analyses was performed. On chromosome 7, the clones RP11-689B18 (location on chr7:69996264- 70178753, including exon 6) map proximally and RP11-575M4 (location on chr7: 70185711-70372702, including exons 7-19) spans the breakpoint. Fosmids G248P87196G1, G248P82441H1 and
G248P84061D2 all map distally to the breakpoint, clearly indicating that the translocation interrupts the AUTS2 gene, starting in intron 6 of the gene. On chromosome 22, clones RP11-699H18 and RP11-772E17 flank the breakpoint, while clone RP11-1056M20 spans the breakpoint (table S4). There are no protein coding genes in the breakpoint region on chromosome 22.
Reference List
1. Sultana,R. et al. Identification of a novel gene on chromosome 7qll.2 interrupted by a translocation breakpoint in a pair of autistic twins.
Genomics 80, 129-134 (2002).
Ref ID: 13
2. Bakkaloglu,B. et al. Molecular cytogenetic analysis and resequencing of contactin associated protein-like 2 in autism spectrum disorders. Am. J. Hum. Genet. 82, 165-173 (2008).
Ref ID: 10
3. Huang,X.L., Zou,Y.S., Maher,T.A., Newton,S., & Milunsky,J.M. A de novo balanced translocation breakpoint truncating the autism susceptibility candidate 2 (AUTS2) gene in a patient with autism. Am. J. Med. Genet. A 152A, 2112-2114 (2010).
Ref ID: 2
4. Kalscheuer,V.M. et al. Mutations in autism susceptibility candidate 2 (AUTS2) in patients with mental retardation. Hum. Genet. 121, 501-509 (2007).
Ref ID: 11
5. Mefford,H.C. et al. Genome-wide copy number variation in epilepsy: novel susceptibility loci in idiopathic generalized and focal epilepsies. PLoS. Genet. 6, e 1000962 (2010).
Ref ID: 4
6. Firth,H.V. et al. DECIPHER: Database of Chromosomal Imbalance and Phenotype in Humans Using Ensembl Resources. Am. J. Hum. Genet. 84,
524-533 (2009).
Ref ID: 224
7. Green,R.E. et al. A draft sequence of the Neandertal genome. Science
328, 710-722 (2010).
Ref ID: 91
8. Bedogni,F. et al. Tbrl regulates regional and laminar identity of
postmitotic neurons in developing neocortex. Proc. Natl. Acad. Sci. U. S. A 107, 13129-13134 (2010).
Ref ID: 3
9. Migeon,B.R. et al. Lack of X inactivation associated with maternal X isodisomy: evidence for a counting mechanism prior to X inactivation during human embryogenesis. Am. J. Hum. Genet. 58, 161-170 (1996).
Ref ID: 236 10. Altschul,S.F. et al. Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Res. 25, 3389-3402 (1997). Ref ID: 240
11. Larkin,M.A. et al. Clustal W and Clustal X version 2.0. Bioinformatics.
23, 2947-2948 (2007).
Ref ID: 239
12. Claverie, J.M. & States,D.J. Information Enhancement Methods for Large-Scale Sequence-Analysis. Computers & Chemistry 17, 191-201 (1993). Ref ID: 242
13. Wootton, J.C. & Federhen,S. Statistics of Local Complexity in Amino-
Acid-Sequences and Sequence Databases. Computers & Chemistry 17, 149- 163 (1993).
Ref ID: 28
14. Conrad,D.F. et al. Origins and functional impact of copy number
variation in the human genome. Nature 464, 704-712 (2010).
Ref ID: 237
15. Cooper,G.M. et al. A copy number variation morbidity map of
developmental delay. Nat. Genet. 43, 838-846 (2011).
Ref ID: 2
16. Altshuler,D.M. et al. Integrating common and rare genetic variation in diverse human populations. Nature 467, 52-58 (2010).
Ref ID: 232
17. Lionel,A.C. et al. Rare copy number variation discovery and cross- disorder comparisons identify risk genes for ADHD. Sci. Transl. Med. 3, 95ra75 (2011).
Ref ID: 234
18. Pinto,D. et al. Comprehensive assessment of array-based platforms and calling algorithms for detection of copy number variants. Nat. Biotechnol. 29, 512-520 (2011).
Ref ID: 233
19. Selzer,R.R. et al. Analysis of chromosome breakpoints in neuroblastoma at sub-kilobase resolution using fine-tiling oligonucleotide array CGH. Genes Chromosomes. Cancer 44, 305-319 (2005).
Ref ID: 202
20. Laura Bernardini High-resolution SNP arrays in mental retardation diagnostics: how much do we gain? European Journal of Human Genetics 18, 178- 185 (2010).
Ref ID: 94 Livak,K.J. & Schmittgen,T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2(- Delta Delta C(T)) Method.
Methods 25, 402-408 (2001).
Ref ID: 95
Pinto,D. et al. Functional impact of global rare copy number variation in autism spectrum disorders. Nature 466, 368-372 (2010).
Ref ID: 229
Talkowski,M.E. et al. Next- generation sequencing strategies enable routine detection of balanced chromosome rearrangements for clinical diagnostics and genetic research. Am. J. Hum. Genet. 88, 469-481 (2011). Ref ID: 230
Talkowski,M.E. et al. Assessment of 2q23.1 Microdeletion Syndrome Implicates MBD5 as a Single Causal Locus of Intellectual Disability,
Epilepsy, and Autism Spectrum Disorder. Am. J. Hum. Genet. 89, 551-563 (2011).
Ref ID: 235
Pinto,D. et al. Functional impact of global rare copy number variation in autism spectrum disorders. Nature 466, 368-372 (2010).
Ref ID: 1
Girirajan,S. et al. A recurrent 16pl2.1 microdeletion supports a t model for severe developmental delay. Nat. Genet. 42, 203-209 (2010).
Ref ID: 244
Li,H. & Durbin,R. Fast and accurate short read alignment with
Burrows-Wheeler transform. Bioinformatics. 25, 1754-1760 (2009).
Ref ID: 3
Li,H. et al. The Sequence Alignment/Map format and SAMtools.
Bioinformatics. 25, 2078-2079 (2009).
Ref ID: 2
Traylor,R.N. et al. Microdeletion of 6ql6.1 encompassing EPHA7 in a child with mild neurological abnormalities and dysmorphic features: case report. Mol. Cytogenet. 2, 17 (2009).
Ref ID: 38
Edgar,R.C. MUSCLE: multiple sequence alignment with high accuracy and high throughput. Nucleic Acids Res. 32, 1792- 1797 (2004).
Ref ID: 238
Westerfield,M., Doerry,E., Kirkpatrick,A.E., Driever,W., & Douglas,S.A.
An on-line database for zebrafish development and genetics research. Semin. Cell Dev. Biol. 8, 477-488 (1997).
Ref ID: 246

Claims

Claims
1. A method of classifying an individual comprising detecting at least one alteration, preferably a chromosomal abnormality, in an AUTS2 gene of said individual.
2. The method of claim 1, wherein said alteration disrupts the AUTS2 short isoform depicted in SEQ ID NO:2.
3. The method of claim 1 or 2 wherein said alteration is located between exons 9- 19 of the AUTS2 gene.
4. The method of any one of the proceeding claims, wherein the presence of at least one alteration in the AUTS2 gene indicates that the individual has or is at risk of developing AUTS2 syndrome.
5. The method of any one of the proceeding claims, wherein the presence of an alteration disrupting the AUTS2 short isoform depicted in SEQ ID NO:2 indicates the severity of the AUTS2 syndrome.
6. The method of any one of the proceeding claims, wherein the alteration is an intragenic deletion.
7. The method of any one of the proceeding claims, wherein said individual is first selected as having at least one symptom of the AUTS2 syndrome.
8. The method of any one of the proceeding claims, wherein said detection is performed using multiplex ligation dependent probe amplification (MLPA), preferably wherein said MLPA is performed using one or more probe pairs selected from MLPA 102, MLPA 110, MLPA 118, MLPA 126, MLPA 130, MLPA 096, MLPA 096, MLPA 136, MLPA 140, MLPA144, MLPA 100, MLPA 108, MLPA 112, MLPA 116, MLPA 120, MLPA 124, MLPA 128, MLPA 132, MLPA 134, MLPA 105, MLPA 138, MLPA 107, and MLPA 136.
9. A multiplex ligation dependent probe amplification (MLPA) probe set specific for the AUTS2 gene.
10. An isolated polypeptide comprising an amino acid sequence having at least 95% identity to SEQ ID NO:2, wherein the N-terminus of said amino acid sequence is encoded by exon 9 of the AUTS2 gene.
11. An isolated nucleic acid comprising a nucleic acid sequence encoding the polypeptide of claim 10.
12. A vector comprising the nucleic acid of claim 11.
13. The vector of claim 12 for use in gene therapy, preferably in the treatment of AUTS2 syndrome.
14. The method of any one of claims 4, 5, or 7, , wherein AUTS2 syndrome is autism.
15. The method of any one of claims 4, 5, 7, or 14, wherein AUTS2 syndrome is characterized by intellectual disability and at least one other symptom selected from microcephaly, mild short stature, feeding difficulties, hypotonia, cerebral palsy, and dysmorphic features.
16. An anti-AUTS2 antibody which binds to SEQ ID NO:2.
17. A mammalian cell comprising the nucleic acid of claim 11 or the vector of claim 12.
18. A non-human mammal comprising the cell of claim 17.
PCT/NL2013/050147 2012-03-07 2013-03-07 Compositions and methods for diagnosing and treating intellectual disability syndrome, autism and autism related disorders WO2013133708A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP12158474.2 2012-03-07
EP12158474 2012-03-07

Publications (1)

Publication Number Publication Date
WO2013133708A1 true WO2013133708A1 (en) 2013-09-12

Family

ID=47902326

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/NL2013/050147 WO2013133708A1 (en) 2012-03-07 2013-03-07 Compositions and methods for diagnosing and treating intellectual disability syndrome, autism and autism related disorders

Country Status (1)

Country Link
WO (1) WO2013133708A1 (en)

Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2443177A (en) 1944-02-28 1948-06-15 Submarine Signal Co Submarine signaling apparatus
US3536809A (en) 1969-02-17 1970-10-27 Alza Corp Medication method
US3598123A (en) 1969-04-01 1971-08-10 Alza Corp Bandage for administering drugs
US3845770A (en) 1972-06-05 1974-11-05 Alza Corp Osmatic dispensing device for releasing beneficial agent
US3916899A (en) 1973-04-25 1975-11-04 Alza Corp Osmotic dispensing device with maximum and minimum sizes for the passageway
US4008719A (en) 1976-02-02 1977-02-22 Alza Corporation Osmotic system having laminar arrangement for programming delivery of active agent
US4736866B1 (en) 1984-06-22 1988-04-12 Transgenic non-human mammals
US4870009A (en) 1982-11-22 1989-09-26 The Salk Institute For Biological Studies Method of obtaining gene product through the generation of transgenic animals
US5059595A (en) 1989-03-22 1991-10-22 Bioresearch, S.P.A. Pharmaceutical compositions containing 5-methyltetrahydrofolic acid, 5-formyltetrahydrofolic acid and their pharmaceutically acceptable salts in controlled-release form active in the therapy of organic mental disturbances
US5073543A (en) 1988-07-21 1991-12-17 G. D. Searle & Co. Controlled release formulations of trophic factors in ganglioside-lipsome vehicle
US5120548A (en) 1989-11-07 1992-06-09 Merck & Co., Inc. Swelling modulated polymeric drug delivery device
US5354556A (en) 1984-10-30 1994-10-11 Elan Corporation, Plc Controlled release powder and process for its preparation
US5591767A (en) 1993-01-25 1997-01-07 Pharmetrix Corporation Liquid reservoir transdermal patch for the administration of ketorolac
US5639476A (en) 1992-01-27 1997-06-17 Euro-Celtique, S.A. Controlled release formulations coated with aqueous dispersions of acrylic polymers
US5674533A (en) 1994-07-07 1997-10-07 Recordati, S.A., Chemical And Pharmaceutical Company Pharmaceutical composition for the controlled release of moguisteine in a liquid suspension
US5733566A (en) 1990-05-15 1998-03-31 Alkermes Controlled Therapeutics Inc. Ii Controlled release of antiparasitic agents in animals
US6955901B2 (en) 2000-02-15 2005-10-18 De Luwe Hoek Octrooien B.V. Multiplex ligatable probe amplification
WO2006097462A2 (en) * 2005-03-15 2006-09-21 Ares Trading S.A. Compositions and methods for treating inflammatory cns disorders
WO2009089464A1 (en) 2008-01-09 2009-07-16 Yale University Mutations in contaction associated protein 2 (cntnap2) are associated with increased risk for ideopathic autism
US20110281759A1 (en) 2008-01-02 2011-11-17 Children's Medical Center Corporation Method for diagnosing autism spectrum disorder

Patent Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2443177A (en) 1944-02-28 1948-06-15 Submarine Signal Co Submarine signaling apparatus
US3536809A (en) 1969-02-17 1970-10-27 Alza Corp Medication method
US3598123A (en) 1969-04-01 1971-08-10 Alza Corp Bandage for administering drugs
US3845770A (en) 1972-06-05 1974-11-05 Alza Corp Osmatic dispensing device for releasing beneficial agent
US3916899A (en) 1973-04-25 1975-11-04 Alza Corp Osmotic dispensing device with maximum and minimum sizes for the passageway
US4008719A (en) 1976-02-02 1977-02-22 Alza Corporation Osmotic system having laminar arrangement for programming delivery of active agent
US4870009A (en) 1982-11-22 1989-09-26 The Salk Institute For Biological Studies Method of obtaining gene product through the generation of transgenic animals
US4736866B1 (en) 1984-06-22 1988-04-12 Transgenic non-human mammals
US4736866A (en) 1984-06-22 1988-04-12 President And Fellows Of Harvard College Transgenic non-human mammals
US5354556A (en) 1984-10-30 1994-10-11 Elan Corporation, Plc Controlled release powder and process for its preparation
US5073543A (en) 1988-07-21 1991-12-17 G. D. Searle & Co. Controlled release formulations of trophic factors in ganglioside-lipsome vehicle
US5059595A (en) 1989-03-22 1991-10-22 Bioresearch, S.P.A. Pharmaceutical compositions containing 5-methyltetrahydrofolic acid, 5-formyltetrahydrofolic acid and their pharmaceutically acceptable salts in controlled-release form active in the therapy of organic mental disturbances
US5120548A (en) 1989-11-07 1992-06-09 Merck & Co., Inc. Swelling modulated polymeric drug delivery device
US5733566A (en) 1990-05-15 1998-03-31 Alkermes Controlled Therapeutics Inc. Ii Controlled release of antiparasitic agents in animals
US5639476A (en) 1992-01-27 1997-06-17 Euro-Celtique, S.A. Controlled release formulations coated with aqueous dispersions of acrylic polymers
US5591767A (en) 1993-01-25 1997-01-07 Pharmetrix Corporation Liquid reservoir transdermal patch for the administration of ketorolac
US5674533A (en) 1994-07-07 1997-10-07 Recordati, S.A., Chemical And Pharmaceutical Company Pharmaceutical composition for the controlled release of moguisteine in a liquid suspension
US6955901B2 (en) 2000-02-15 2005-10-18 De Luwe Hoek Octrooien B.V. Multiplex ligatable probe amplification
WO2006097462A2 (en) * 2005-03-15 2006-09-21 Ares Trading S.A. Compositions and methods for treating inflammatory cns disorders
US20110281759A1 (en) 2008-01-02 2011-11-17 Children's Medical Center Corporation Method for diagnosing autism spectrum disorder
WO2009089464A1 (en) 2008-01-09 2009-07-16 Yale University Mutations in contaction associated protein 2 (cntnap2) are associated with increased risk for ideopathic autism

Non-Patent Citations (48)

* Cited by examiner, † Cited by third party
Title
"CGH Analytics or Genomic Workbench Standard Edition 5.0.14", AGILENT TECHNOLOGIES, INC.
"Remington: The Science and Practice of Pharmacy", 2000, LIPPINCOTT WILLIAMS & WILKINS
ALTSCHUL,S.F. ET AL.: "Gapped BLAST and PSI-BLAST: a new generation of protein database search programs", NUCLEIC ACIDS RES., vol. 25, 1997, pages 3389 - 3402, XP002905950, DOI: doi:10.1093/nar/25.17.3389
ALTSHULER,D.M. ET AL.: "Integrating common and rare genetic variation in diverse human populations", NATURE, vol. 467, 2010, pages 52 - 58, XP055247268, DOI: doi:10.1038/nature09298
BAKKALOGLU,B. ET AL.: "Molecular cytogenetic analysis and resequencing of contactin associated protein-like 2 in autism spectrum disorders", AM. J. HUM. GENET., vol. 82, 2008, pages 165 - 173
BEDOGNI F ET AL: "Autism susceptibility candidate 2 (Auts2) encodes a nuclear protein expressed in developing brain regions implicated in autism neuropathology", GENE EXPRESSION PATTERNS, ELSEVIER, AMSTERDAM, NL, vol. 10, no. 1, 1 January 2010 (2010-01-01), pages 9 - 15, XP026853300, ISSN: 1567-133X, [retrieved on 20091203] *
BEDOGNI,F. ET AL.: "Tbrl regulates regional and laminar identity of postmitotic neurons in developing neocortex", PROC. NATL. ACAD. SCI. U. S. A, vol. 107, 2010, pages 13129 - 13134
CASILLI, HUM MUTAT, vol. 20, 2001, pages 218 - 26
CLAVERIE,J.M.; STATES,D.J.: "Information Enhancement Methods for Large-Scale Sequence-Analysis", COMPUTERS & CHEMISTRY, vol. 17, 1993, pages 191 - 201, XP026488515, DOI: doi:10.1016/0097-8485(93)85010-A
CONRAD,D.F. ET AL.: "Origins and functional impact of copy number variation in the human genome", NATURE, vol. 464, 2010, pages 704 - 712, XP055146767, DOI: doi:10.1038/nature08516
COOPER,G.M. ET AL.: "A copy number variation morbidity map of developmental delay", NAT. GENET., vol. 43, 2011, pages 838 - 846
DATABASE EMBL [Online] 23 September 2008 (2008-09-23), "SubName: Full=cDNA FLJ58510, highly similar to Autism susceptibility gene 2 protein;", XP002681191, retrieved from EBI accession no. TR:B4DLG0_HUMAN Database accession no. B4DLG0 *
EDGAR,R.C.: "MUSCLE: multiple sequence alignment with high accuracy and high throughput", NUCLEIC ACIDS RES., vol. 32, 2004, pages 1792 - 1797, XP008137003, DOI: doi:10.1093/nar/gkh340
FIRTH,H.V. ET AL.: "DECIPHER: Database of Chromosomal Imbalance and Phenotype in Humans Using Ensembl Resources", AM. J. HUM. GENET., vol. 84, 2009, pages 524 - 533
GIRIRAJAN,S. ET AL.: "A recurrent 16p12.1 microdeletion supports a two-hit model for severe developmental delay", NAT. GENET., vol. 42, 2010, pages 203 - 209
GREEN,R.E. ET AL.: "A draft sequence of the Neandertal genome", SCIENCE, vol. 328, 2010, pages 710 - 722
HAGEGE ET AL., NATURE PROTOC 2, 2007, pages 1722 - 33
HUANG,X.L.; ZOU,Y.S.; MAHER,T.A.; NEWTON,S.; MILUNSKY,J.M.: "A de novo balanced translocation breakpoint truncating the autism susceptibility candidate 2 (AUTS2) gene in a patient with autism", AM. J. MED. GENET. A 152A, 2010, pages 2112 - 2114, XP055034459, DOI: doi:10.1002/ajmg.a.33497
KALSCHEUER,V.M. ET AL.: "Mutations in autism susceptibility candidate 2 (AUTS2) in patients with mental retardation", HUM. GENET., vol. 121, 2007, pages 501 - 509, XP019517847, DOI: doi:10.1007/s00439-006-0284-0
KOUSOULIDOU ET AL., METHODS MOL BIOL, vol. 653, 2010, pages 47 - 71
LARKIN,M.A. ET AL.: "Clustal W and Clustal X version 2.0.", BIOINFORMATICS, vol. 23, 2007, pages 2947 - 2948
LAURA BERNARDINI: "High-resolution SNP arrays in mental retardation diagnostics: how much do we gain?", EUROPEAN JOURNAL OF HUMAN GENETICS, vol. 18, 2010, pages 178 - 185
LI,H. ET AL.: "The Sequence Alignment/Map format and SAMtools", BIOINFORMATICS., vol. 25, 2009, pages 2078 - 2079, XP055229864, DOI: doi:10.1093/bioinformatics/btp352
LI,H.; DURBIN,R.: "Fast and accurate short read alignment with Burrows-Wheeler transform", BIOINFORMATICS., vol. 25, 2009, pages 1754 - 1760, XP055287430, DOI: doi:10.1093/bioinformatics/btp324
LIEHR ET AL., HISTOL HISTOPATHOL, vol. 19, 2004, pages 229 - 237
LIONEL,A.C. ET AL.: "Rare copy number variation discovery and cross- disorder comparisons identify risk genes for ADHD", SCI. TRANSL. MED., vol. 3, 2011, pages 95RA75
LIVAK,K.J.; SCHMITTGEN,T.D.: "Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method", METHODS, vol. 25, 2001, pages 402 - 408
MEFFORD,H.C. ET AL.: "Genome-wide copy number variation in epilepsy: novel susceptibility loci in idiopathic generalized and focal epilepsies", PLOS. GENET., vol. 6, 2010, pages E1000962
MIGEON,B.R. ET AL.: "Lack of X inactivation associated with maternal X isodisomy: evidence for a counting mechanism prior to X inactivation during human embryogenesis", AM. J. HUM. GENET., vol. 58, 1996, pages 161 - 170
PANTANO ET AL., BMC GENOMICS, vol. 9, 2008, pages 573
PINTO,D. ET AL.: "Comprehensive assessment of array-based platforms and calling algorithms for detection of copy number variants", NAT. BIOTECHNOL., vol. 29, 2011, pages 512 - 520, XP002735142, DOI: doi:10.1038/nbt.1852
PINTO,D. ET AL.: "Functional impact of global rare copy number variation in autism spectrum disorders", NATURE, vol. 466, 2010, pages 368 - 372, XP055033303, DOI: doi:10.1038/nature09146
RICHARDSON ET AL., NEUROSURG CLIN N AM., vol. 20, no. 2, 2009, pages 205 - 10
SCHLACHETZKI ET AL.: "Neurology", GENE THERAPY OF THE BRAIN., vol. 62, 2004, pages 1275 - 1281
SCHOUTEN ET AL., NUCLEIC ACIDS RES., vol. 30, 2002, pages E57
SCHOUTEN JAN P ET AL: "Relative quantification of 40 nucleic acid sequences by multiplex ligation-dependent probe amplification", NUCLEIC ACIDS RESEARCH, OXFORD UNIVERSITY PRESS, GB, vol. 30, no. 12, 15 June 2002 (2002-06-15), XP009145839, ISSN: 1362-4962, DOI: 10.1093/NAR/GNF056 *
SELZER,R.R. ET AL.: "Analysis of chromosome breakpoints in neuroblastoma at sub-kilobase resolution using fine-tiling oligonucleotide array CGH", GENES CHROMOSOMES. CANCER, vol. 44, 2005, pages 305 - 319, XP009057632
SHINAWI, M. ET AL., J MED GENET, vol. 47, 2010, pages 332 - 41
SULTANA RAZIA ET AL: "Identification of a novel gene on chromosome 7q11.2 interrupted by a translocation breakpoint in a pair of autistic twins", GENOMICS, ACADEMIC PRESS, SAN DIEGO, US, vol. 80, no. 2, 1 August 2002 (2002-08-01), pages 129 - 134, XP002385351, ISSN: 0888-7543 *
SULTANA,R. ET AL.: "Identification of a novel gene on chromosome 7qll.2 interrupted by a translocation breakpoint in a pair of autistic twins", GENOMICS, vol. 80, 2002, pages 129 - 134, XP002385351
TALKOWSKI,M.E. ET AL.: "Assessment of 2q23.1 Microdeletion Syndrome Implicates MBD5 as a Single Causal Locus of Intellectual Disability, Epilepsy, and Autism Spectrum Disorder", AM. J. HUM. GENET., vol. 89, 2011, pages 551 - 563, XP028310093, DOI: doi:10.1016/j.ajhg.2011.09.011
TALKOWSKI,M.E. ET AL.: "Next-generation sequencing strategies enable routine detection of balanced chromosome rearrangements for clinical diagnostics and genetic research", AM. J. HUM. GENET., vol. 88, 2011, pages 469 - 481, XP028195310, DOI: doi:10.1016/j.ajhg.2011.03.013
TRAYLOR,R.N. ET AL.: "Microdeletion of 6q16.1 encompassing EPHA7 in a child with mild neurological abnormalities and dysmorphic features: case report", MOL. CYTOGENET., vol. 2, 2009, pages 17, XP021060645, DOI: doi:10.1186/1755-8166-2-17
VERA M KALSCHEUER ET AL: "Mutations in autism susceptibility candidate 2 (AUTS2) in patients with mental retardation", HUMAN GENETICS, SPRINGER, BERLIN, DE, vol. 121, no. 3-4, 9 January 2007 (2007-01-09), pages 501 - 509, XP019517847, ISSN: 1432-1203, DOI: 10.1007/S00439-006-0284-0 *
WESTERFIELD,M.; DOERRY,E.; KIRKPATRICK,A.E.; DRIEVER,W.; DOUGLAS,S.A.: "An on-line database for zebrafish development and genetics research", SEMIN. CELL DEV. BIOL., vol. 8, 1997, pages 477 - 488
WOOTTON,J.C.; FEDERHEN,S.: "Statistics of Local Complexity in Amino-Acid-Sequences and Sequence Databases", COMPUTERS & CHEMISTRY, vol. 17, 1993, pages 149 - 163, XP026488511, DOI: doi:10.1016/0097-8485(93)85006-X
XIN-LI HUANG ET AL: "A de novo balanced translocation breakpoint truncating the autism susceptibility candidate 2 (AUTS2) gene in a patient with autism", AMERICAN JOURNAL OF MEDICAL GENETICS PART A, vol. 152A, no. 8, 15 July 2010 (2010-07-15), pages 2112 - 2114, XP055034459, ISSN: 1552-4825, DOI: 10.1002/ajmg.a.33497 *
ZHI J., BMC RESEARCH NOTES, vol. 3, 2010, pages 137

Similar Documents

Publication Publication Date Title
US11920199B2 (en) Methods and compositions for screening and treating developmental disorders
JP6078211B2 (en) Genetic changes associated with autism and the phenotype of autism and its use for diagnosis and treatment of autism
Beleza-Meireles et al. Oculo-auriculo-vertebral spectrum: clinical and molecular analysis of 51 patients
Korenberg et al. VI. Genome structure and cognitive map of Williams syndrome
Clayton-Smith et al. Whole-exome-sequencing identifies mutations in histone acetyltransferase gene KAT6B in individuals with the Say-Barber-Biesecker variant of Ohdo syndrome
Gueneau et al. KIAA1109 variants are associated with a severe disorder of brain development and arthrogryposis
US20140162894A1 (en) Methods and compositions for screening and treating developmental disorders
Heimer et al. TECPR2 mutations cause a new subtype of familial dysautonomia like hereditary sensory autonomic neuropathy with intellectual disability
Barber et al. 8p23. 1 duplication syndrome; common, confirmed, and novel features in six further patients
Taylor et al. Novel PEX11B mutations extend the peroxisome biogenesis disorder 14B phenotypic spectrum and underscore congenital cataract as an early feature
Wexler et al. Molecular approaches to hereditary diseases of the nervous system: Huntington's disease as a paradigm
Boyle et al. Deletion of 11q12. 3–11q13. 1 in a patient with intellectual disability and childhood facial features resembling Cornelia de Lange syndrome
Schmidt et al. Congenital myasthenic syndrome due to a novel missense mutation in the gene encoding choline acetyltransferase
Wang et al. Phenotypic heterogeneity of post-lingual and/or milder hearing loss for the patients with the GJB2 c. 235delC homozygous mutation
Ryu et al. Identification of a novel splicing mutation within SLC17A8 in a Korean family with hearing loss by whole-exome sequencing
Bykhovskaya et al. TSC1 mutations in keratoconus patients with or without tuberous sclerosis
KR20190043845A (en) Marker TMEM43 for diagnosing sensorineural hearing loss and uses thereof
Müller et al. A newly identified chromosomal microdeletion of the rapsyn gene causes a congenital myasthenic syndrome
Bateman et al. A new locus for autosomal dominant cataract on chromosome 12q13
JP2008537486A (en) Human autism susceptibility genes encoding transmembrane proteins and uses thereof
Yu et al. Apparent homozygosity for a novel splicing variant in EPS8 causes congenital profound hearing loss
Lee et al. Noncanonical splice site and deep intronic Frmd7 variants activate cryptic exons in X-linked infantile nystagmus
WO2013133708A1 (en) Compositions and methods for diagnosing and treating intellectual disability syndrome, autism and autism related disorders
AU2003285153B2 (en) Organic anion transport polypeptide related protein-4 (OATPRP4) gene in tourette syndrome and related disorders
Domingues et al. The genetics of stuttering

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13710909

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13710909

Country of ref document: EP

Kind code of ref document: A1