WO2019229113A1 - Detection of tubb1 mutations for diagnosing thyroid dysgenesis - Google Patents

Detection of tubb1 mutations for diagnosing thyroid dysgenesis Download PDF

Info

Publication number
WO2019229113A1
WO2019229113A1 PCT/EP2019/063933 EP2019063933W WO2019229113A1 WO 2019229113 A1 WO2019229113 A1 WO 2019229113A1 EP 2019063933 W EP2019063933 W EP 2019063933W WO 2019229113 A1 WO2019229113 A1 WO 2019229113A1
Authority
WO
WIPO (PCT)
Prior art keywords
thyroid
tubb1
mutation
tubulin
mutations
Prior art date
Application number
PCT/EP2019/063933
Other languages
French (fr)
Inventor
Michel Polak
Aurore Carre
Athanasia STOUPA
Dulanjalee KARIYAWASAM
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Université Paris Descartes
Centre National De La Recherche Scientifique (Cnrs)
Assistance Publique-Hôpitaux De Paris (Aphp)
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 INSERM (Institut National de la Santé et de la Recherche Médicale), Université Paris Descartes, Centre National De La Recherche Scientifique (Cnrs), Assistance Publique-Hôpitaux De Paris (Aphp) filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Publication of WO2019229113A1 publication Critical patent/WO2019229113A1/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
    • C12Q1/6813Hybridisation 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
    • C12Q2600/112Disease subtyping, staging or classification

Definitions

  • the present invention relates to a method for diagnosing or predicting thyroid dysgenesis by detecting a mutation in Tubulin, Beta 1 Class VI ( TUBB1 ).
  • Thyroid dysgenesis is a feature in 65% of patients with congenital hypothyroidism (CH), which is the most common neonatal endocrine disorder (1/3,500 neonates) 1,2 .
  • CH congenital hypothyroidism
  • TD includes a vast spectrum of developmental thyroid anomalies encompassing athyreosis, thyroid ectopia, hypoplasia of an orthotopic gland, and hemiagenesis 1,3 .
  • the midline thyroid strom appears on embryonic day E.8.5 in mice and at 3 gestational weeks (GW) in humans.
  • the midline strom and ultimobranchial bodies migrate and fuse in the definitive pretracheal position on El 3.5 in mice and at 7 GW in humans 4,5 .
  • the cells differentiate into thyrocytes organized in follicles or C-cells 6 .
  • Abnormalities at any step of thyroid development may result in TD with variable degrees of hypothyroidism or rarely normal thyroid function 7 .
  • Studies of sporadic and familial TD covering a wide clinical spectrum identified mutations in eight genes: PAX8, NKX2-1, FOXE1, NKX2-5, TSHR, GLIS3, NTN1, and BOREALIN 8-14 .
  • mutations in these genes are found in only 5% of all patients with TD and identification of causative mutations remains a challenging task.
  • TUBB1 Tebulin, Beta 1 Class VI encodes a member of the b-tubulin protein family.
  • b-tubulins are one of two core protein families that heterodimerise a/b-tubulin dimers, which assemble into microtubules, one of the major cytoskeletal structures. Microtubules are involved in crucial processes such as cell division, the growth polarity, and migration; intracellular trafficking; and cell communication 17,18 .
  • the b ⁇ isotype of tubulin (TUBB1) has been described as specifically expressed in platelets and megakaryocytes and involved in proplatelet formation and platelet release 19 .
  • the present invention relates to a method for diagnosing or predicting thyroid dysgenesis by detecting a mutation in Tubulin, Beta 1 Class VI (TUBB1 ).
  • the present invention is defined by the claims.
  • TUBB1 thyroid dysgenesis
  • the first object of the present invention relates to a method of diagnosing thyroid dysgenesis in a subject, comprising detecting a mutation in the TUBB1 gene in a sample obtained from said subject, wherein detecting the presence of a mutation in the TUBB1 gene is considered to be indicative of thyroid dysgenesis.
  • thyroid dysgenesis has its general meaning in the art and refers to a permanent thyroid hormone deficiency present from birth and resulting from an abnormality in the development of thyroid. Thyroid dysgenesis comprises e.g. thyroid ectopy, athyreosis and thyroid hypoplasia.
  • the term“subject” is preferably a human. More particularly, the subject is a new-bom or a foetus. As used herein, a“new-born” is a child who is less than 2 weeks old, particularly less than 1 week old, more particularly less than 3 days old. Thus, when the method is method is performed“just after birth”, it means that the method is performed within 2 weeks, particularly 1 week, more particularly 3 days after birth.
  • thyroid dysgenesis results from an abnormality of the development of thyroid during embryo development.
  • the present invention is thus particularly interesting for detecting thyroid dysgenesis just after birth, i.e. wherein the subject is a new-bom.
  • the method according to the present invention is performed just after birth and after having suspected thyroid dysgenesis during embryo development, e.g. by observing an abnormality in the development of the embryo’s thyroid during an ultrasonography, particularly obstetric ultrasonography.
  • the method according to the present invention is performed in a child, preferably a new-born, whose parents (1 or both of them) present thyroid dysgenesis.
  • the method according to the present invention is performed in a child, preferably a new-born, whose parents (1 or both of them) are identified has displaying a mutation in the TUBB1 gene.
  • the method according to the present invention is interesting for detecting an abnormality in the thyroid development at the embryo stage.
  • the present invention relates to a prenatal method for diagnosing or predicting thyroid dysgenesis in a foetus by detecting a mutation in the TUBB1 gene in a sample obtained from said foetus or from said foetus’s mother.
  • TD is suspected by routine screening which shows thyroid stimulating hormone (TSH) elevation and low T4 level.
  • TSH thyroid stimulating hormone
  • Other tests, such as thyroid radionuclide uptake and scan, thyroid sonography, or serum thyroglobulin determination may be carried out before performing the method of the present invention.
  • TUBB1 has its general mean in the art and refers to gene that encodes a member of the beta tubulin protein family: tubulin beta 1 class VI.
  • Beta tubulins are one of two core protein families (alpha and beta tubulins) that heterodimerize and assemble to form microtubules tubulin beta 1 class VI is encoded by the TUBB1 gene (available under the reference ENSG00000101162 (gene) in the Ensembl Gene Database) or referenced as GENE ID : 81027 in the NCBI database.
  • An exemplary nucleic acid sequence is represented by SEQ ID NO: l and an exemplary amino acid sequence is represented by SEQ ID NO:2.
  • mutation has its general meaning in the art and refers to any detectable change in genetic material, e.g. DNA, RNA, cDNA, or in an amino acid sequence encoded by such a genetic material.
  • genetic material e.g. DNA, RNA, cDNA
  • a mutation is identified in a subject by comparing the sequence of a nucleic acid or of a polypeptide expressed by said subject with the corresponding nucleic acid or polypeptide expressed in a control population.
  • dbSNP Single Nucleotide Polymorphism Database
  • NCBI National Center for Biotechnology Information
  • NHGRI National Human Genome Research Institute
  • the present inventors have identified different mutations of the TUBB1 gene involved in thyroid dysgenesis.
  • One of these mutations is a missense homozygous TUBB1 mutation (c.479C>T, p.Pl60L, rs759l 17911) which is a substitution of a cytosine residue into a thymine residue at position 479 of the TUBB1 gene.
  • This mutation encodes for the substitution of the proline residue at position 160 by a leucine residue in the mature protein.
  • Another mutation identified by the present inventors is a heterozygous TUBB1 mutation (c.3 l8C>G, p.Yl06X) which is the substitution of a cytosine residue into a guanine residue at position 318 of the TUBB1 gene. This mutation leads to the creation a premature stop codon at amino acid 106.
  • Another mutation identified by the inventors is a heterozygous frameshift TUBB1 mutation (c.35delG, p.Cysl 2Lcufs* 12, rs77324804) that created a premature stop codon at amino acid 23.
  • All three mutations are located in the first part of TUBB1, i.e., in the N-terminal domain needed for guanosine triphosphate (GTP) activity, i.e. the domain ranging from the amino acid residue at position 1 to the amino acid residue at position 206 in the protein.
  • All three TUBB 1 mutations identified lead to nonfunctional a/b -tubulin dimers that cannot be incorporated into microtubules.
  • the mutations lead to an early abnormal proliferation of progenitors, delayed thyroid migration, defective thyroid tissue differentiation, and impaired thyroid hormone release. More interestingly, the mutations also affect b ⁇ -tubulin expression in platelets and result in abnormally large platelet size, probably as a consequence of proplatelet abnormal formation.
  • the p.Pl60L and r.U106C mutations induce significant basal platelet activation and hyperaggregation in response to agonists, whereas the c.35delG mutation does not seem to affect platelet function.
  • sample refers to sample liable to contain nucleic acid molecules including mRNA, genomic DNA and cDNA derived from mRNA.
  • DNA or RNA can be single stranded or double stranded. These may be utilized for detection by amplification and/or hybridization with a probe, for instance.
  • the nucleic acid sample may be obtained from any cell source or tissue biopsy. Non-limiting examples of cell sources available include without limitation blood cells, buccal cells, epithelial cells, fibroblasts, or any cells present in a tissue obtained by biopsy. Cells may also be obtained from body fluids, such as blood, plasma, serum, lymph, etc. In some embodiments, the sample is an amniotic fluid sample.
  • the sample may further be any biological sample wherein foetal DNA may be detected.
  • the biological sample may be a sample obtained from the mother but wherein foetal DNA can be found.
  • foetal DNA is detectable in maternal serum or plasma samples. Abnormalities in the foetus genetic material can thus be detected by directly analysing the foetal DNA present in the mother’s blood.
  • the sample according to the present invention is a maternal serum or plasma sample.
  • DNA may be extracted using any methods known in the art, such as described in Sambrook et ah, 1989.
  • RNA may also be isolated, for instance from tissue biopsy, using standard methods well known to the one skilled in the art such as guanidium thiocyanate- phenol-chloroform extraction.
  • TUBB1 mutations may be detected in a RNA or DNA sample, preferably after amplification.
  • the isolated RNA may be subjected to coupled reverse transcription and amplification, such as reverse transcription and amplification by polymerase chain reaction (RT-PCR), using specific oligonucleotide primers that are specific for a mutated site or that enable amplification of a region containing the mutated site.
  • RT-PCR polymerase chain reaction
  • conditions for primer annealing may be chosen to ensure specific reverse transcription (where appropriate) and amplification; so that the appearance of an amplification product be a diagnostic of the presence of a particular TUBB1 mutation.
  • RNA may be reverse-transcribed and amplified, or DNA may be amplified, after which a mutated site may be detected in the amplified sequence by hybridization with a suitable probe or by direct sequencing, or any other appropriate method known in the art.
  • a cDNA obtained from RNA may be cloned and sequenced to identify a mutation in TUBB1 sequence.
  • numerous strategies for genotype analysis are available (Antonarakis et ah, 1989; Cooper et ah, 1991; Grompe, 1993). Briefly, the nucleic acid molecule may be tested for the presence or absence of a restriction site.
  • a base substitution mutation creates or abolishes the recognition site of a restriction enzyme, this allows a simple direct PCR test for the mutation.
  • Further strategies include, but are not limited to, direct sequencing, restriction fragment length polymorphism (RFFP) analysis; hybridization with allele-specific oligonucleotides (ASO) that are short synthetic probes which hybridize only to a perfectly matched sequence under suitably stringent hybridization conditions; allele-specific PCR; PCR using mutagenic primers; ligase- PCR, HOT cleavage; denaturing gradient gel electrophoresis (DGGE), temperature denaturing gradient gel electrophoresis (TGGE), single-stranded conformational polymorphism (SSCP) and denaturing high performance liquid chromatography (Kuklin et ah, 1997).
  • RFFP restriction fragment length polymorphism
  • ASO allele-specific oligonucleotides
  • Direct sequencing may be accomplished by any method, including without limitation chemical sequencing, using the Maxam-Gilbert method ; by enzymatic sequencing, using the Sanger method ; mass spectrometry sequencing; sequencing using a chip-based technology; and real- time quantitative PCR.
  • DNA from a subject is first subjected to amplification by polymerase chain reaction (PCR) using specific amplification primers.
  • PCR polymerase chain reaction
  • RCA rolling circle amplification
  • InvaderTMassay or oligonucleotide ligation assay (OLA).
  • OLA may be used for revealing base substitution mutations.
  • oligonucleotides are constructed that hybridize to adjacent sequences in the target nucleic acid, with the join sited at the position of the mutation.
  • DNA ligase will covalently join the two oligonucleotides only if they are perfectly hybridized. Therefore, useful nucleic acid molecules, in particular oligonucleotide probes or primers, according to the present invention include those which specifically hybridize the regions where the mutations are located.
  • Oligonucleotide probes or primers may contain at least 10, 15, 20 or 30 nucleotides. Their length may be shorter than 400, 300, 200 or 100 nucleotides.
  • the TUBB1 mutations of the present invention may be detected by Next Generation Sequencing or NGS.
  • NGS Next Generation Sequencing
  • genomic DNA or cDNA library is first prepared, and common adaptors may then be ligated to the fragmented genomic DNA or cDNA. Different protocols may be used to generate jumping libraries of mate-paired tags with controllable distance distribution.
  • An array of millions of spatially immobilized PCR colonies or "polonies" is generated with each polonies consisting of many copies of a single shotgun library fragment. Because the polonies are tethered to a planar array, a single micro liter- scale reagent volume can be applied to manipulate the array features in parallel, for example, for primer hybridization or for enzymatic extension reactions. Imaging-based detection of fluorescent labels incorporated with each extension may be used to acquire sequencing data on all features in parallel. Successive iterations of enzymatic interrogation and imaging may also be used to build up a contiguous sequencing read for each array feature.
  • the mutation may be also detected at a protein level (e.g. for loss of function mutation) according to any appropriate method known in the art.
  • a biological sample such as a tissue biopsy, obtained from a subject may be contacted with antibodies specific of a mutated form of TUBB1 protein, i.e. antibodies that are capable of distinguishing between a mutated form of TUBB1 and the wild-type protein, to determine the presence or absence of a TUBB1 specified by the antibody.
  • the antibodies may be monoclonal or polyclonal antibodies, single chain or double chain, chimeric antibodies, humanized antibodies, or portions of an immunoglobulin molecule, including those portions known in the art as antigen binding fragments Fab, Fab', F(ab')2 and F(v). They can also be immunoconjugated, e.g. with a toxin, or labelled antibodies. Whereas polyclonal antibodies may be used, monoclonal antibodies are preferred for they are more reproducible in the long run. Procedures for raising“polyclonal antibodies” are also well known. Alternatively, binding agents other than antibodies may be used for the purpose of the invention.
  • aptamers are a class of molecule that represents an alternative to antibodies in term of molecular recognition.
  • Aptamers are oligonucleotide or oligopeptide sequences with the capacity to recognize virtually any class of target molecules with high affinity and specificity.
  • ligands may be isolated through Systematic Evolution of Ligands by Exponential enrichment (SELEX) of a random sequence library.
  • the method according to the present invention is performed after having suspected thyroid dysgenesis during embryo development and treated the foetus thereby identified as presenting an abnormality in the thyroid development.
  • An example of treatment which can be administered to a foetus identified as presenting an abnormality in the development of thyroid is intra-amniotic injection of Levothyroxine.
  • Levothyroxine is intra-amniotic injection of Levothyroxine.
  • Such treatment is e.g. disclosed in Leger et al (ESPE-PES-SLEP-JSPE-APEG-APPES-ISPAE; Congenital Hypothyroidism Consensus Conference Group. European Society for Paediatric Endocrinology consensus guidelines on screening, diagnosis, and management of congenital hypothyroidism. J Clin Endocrinol Metab.
  • a therapy is administered.
  • Levothyroxine is the therapy of choice. Treatment is typically initiated in any infant with a positive screening result. The dose and timing of thyroid hormone replacement are important in achieving optimal neurocognitive outcome. Thus, the goal of treatment should be to restore the serum T4 to > 129 mmol/L (> 10 pg/dl) as rapidly as possible.
  • the recommended initial l-thyroxine dose set forth by the American Academy of Pediatrics (AAP) and the European Society for Paediatric Endocrinology (ESPE) is 10-15 mcg/kg per day. In term infants this amounts to an average of 37.5 to 50 meg per day.
  • a prophylactic treatment mays also be prescribed.
  • said treatment may consist in antagonists of platelet activation and particularly of ADP receptors such as thienopyridines (eg, clopidogrel, prasugrel) or non-thienopyridine ADP inhibitors (eg, ticagrelor), or any newer anti-platelet agent (eg, inhibitors of the thrombin receptor).
  • FIGURES are a diagrammatic representation of FIGURES.
  • Genomic DNA was isolated from whole blood. Exome capture and sequencing were performed at the genomics platform of the Imagine Institute. WES libraries were prepared from 3 Lig genomic DNA per individual, which was sheared by ultrasonication (Covaris S220 Ultrasonicator, Wobum, MA, USA). Exome capture was performed using the SureSelect Human All Exon V6 kit (Agilent Technologies, Santa Clara, CA, USA). The resulting libraries were sequenced on a HiSeq 2,500 HT device (Illumina, San Diego, CA, USA) according to the manufacturer’s recommendations. Paired-end (2x130) 76-bp reads were generated and mapped on the human reference genome. More than 97% of the exome was covered at least 30 times.
  • the variant prioritisation strategy was as follows: (i) selection of functional (protein-altering) variants (removal of intergenic and 375’ UTR variants, non-splice related intronic variants, and synonymous variants); (ii) variants with a frequency below 1% in public databases (dbSNP, 1,000 Genomes, EVS, ExAC; release date, January 2018), and (iii) variants previously identified in fewer than five individuals contributing 11,811 in-house exomes (data not shown).
  • the HypothySeq NGS panel included 78 genes known to be associated with CH (TD; dyshormonogenesis; defects in thyroid hormone (TH) transport proteins; and inborn errors in TH membrane transport, metabolism, or action) and candidate genes validated in animal models (mouse and zebrafish knock-out models) or by microarray assays but not yet validated in humans.
  • This panel was previously validated using controls including samples from positive controls with known thyroid disease-causing mutations, to assess sensitivity (false-negative rate); and from healthy individuals screened by WES for another research study, to test specificity (false-positive rate).
  • Genomic DNA libraries were created using SureSelectXT Target Enrichment Reagent Kit (Agilent Technologies) and subjected to custom targeted DNA panel enrichment.
  • Burden test Rare variant burden testing was performed for the TUBB1 gene using the CAST collapsing method 55 in 270 patients with TD, including 193 with ectopia, hemi-agenesis or hypoplasia and 77 with athyreosis.
  • Logistic regression was performed to compare the prevalence of deleterious variants in the patients with TD and in 406 Caucasian controls from the 1,000 Genomes project phase 3 56 .
  • Disruptive in- frame, frame-shift, missense, splice- acceptor, splice-donor, start-lost, stop-gained, or stop-lost variants were considered deleterious. All deleterious variants with a minor allele frequency ⁇ 1% in the ExAC database were included in the analysis. We first studied all 270 TD patients then only the 193 TD patients without athyreosis.
  • embryonic thyroid tissue was obtained from products of elective termination of pregnancy and adult thyroid tissues from patients undergoing thyroid surgery.
  • Tubbl / mice were previously generated by replacing exons 3 and 4, encoding amino acids 56 to 451, with a neomycin-resistance gene cassette as previously described 23 .
  • Tubbl +/ mice on a mixed l29/Sv-BALB/c background were interbred with C57BL/6J mice over ten generations to generate homozygous null mutants (Tubbl ⁇ ) with the C57BL/6J background (Bb.CG-b 1 tubulinTM). All experiments were conducted in accordance with French regulations and were approved by the France regional ethics committee for animal experimentation (C.R.E.M.E.A.S., CEEA 35). Thyroids at different embryonic stages from El3.5 to E17.5 and adult thyroids at 3 months of age were obtained from wild-type and Tubbl mice and microdissected as described previously 34 .
  • Aortic blood samples were collected from 3-month-old wild-type and Tubbl mice. Radioimmunoassays were used to measure serum TSH and serum total T4 after iodothyronine extraction (Dr. S. Refetoff, Chicago, IL, USA) as previously described 57 .
  • the cells were acquired on a BD FACSAria II flow cytometer (Becton Dickinson, Franklin Lakes, NJ, USA).
  • the following monoclonal antibodies were used: EpCam/CD326 (G8.8), PDGFRa/CDl40a (APA5), CD45 (30-F11), and Pecam/CD3 l (390) from BioLegend (San Diego, CA, USA); and CD41 from Abeam (Cambridge, UK).
  • the secondary antibody was goat anti-rabbit A647 from Life Technologies (Carlsbad, CA, USA).
  • Each pool of sorted cells was collected in RLT buffer from the Qiagen RNeasy MicroKit (Qiagen, Valencia, CA, USA) for RNA extraction experiments.
  • RNA of sorted cells or thyroid tissue was isolated using the Qiagen RNeasy Microkit or Minikit (Qiagen).
  • the Maxima First Strand cDNA Synthesis Kit (Thermo Fisher Scientific) was used for reverse transcription of 250 ng of each RNA sample.
  • the synthesised cDNA was diluted to 1/20, and 5 mE was used for each PCR reaction.
  • Each reaction consisted of TaqMan Universal PCR Master Mix or SybrGreen PCR Master Mix (Thermo Fisher Scientific) and primers.
  • Peptidylpropyl isomerase A served as an endogenous control.
  • Real-time PCR was performed using the QuantStudio 3 Real-Time PCR System (Thermo Fisher Scientific). The data were analysed using the comparative cycle threshold method and reported as the fold change in gene expression, normalised for a calibrator of value 1.
  • the primary antibodies were used at the following dilutions: rabbit antibody to human or mouse b ⁇ -tubulin, 1 : 1 000 (donated by Frangois Fanza), rabbit anti- Ecadherin, 1 : 100 (Becton Dickinson), mouse anti-TG, 1 : 100 (DakoCytomation, Glostrup, Denmark), rabbit anti-Nkx2-l, 1 :2500 (Biopat, Italy), mouse anti-T4, 1 : 10 000 (AbD Serotec, Raleigh, NC, USA), rabbit anti-calcitonin, 1 :400 (DakoCytomation), mouse anti-Ki67, 1 :20 (Becton Dickinson), and rabbit anti-KDEF, 1 : 1500 (Thermo Fisher Scientific).
  • the fluorescent secondary antibodies were Alexa Fluor 594 goat anti-rabbit and Alexa Fluor 488 goat anti mouse antibodies (1/400, Thermo Fisher Scientific).
  • the nuclei were stained using the Hoechst 33 342 fluorescent stain (0.3 mg/mL; Thermo Fisher Scientific). Photographs were taken using a fluorescence microscope (Leitz DMRB; Leica, Wetzlar, Germany) and digitised using a chilled 3CCD camera (C5810; Hamamatsu Photonics, Hamamatsu City, Japan).
  • the sections were then analysed using Image J l.32s (freeware, www.rsbweb.nih.gov/ij) as previously described 34,58 .
  • the Nkx2-l -positive surface areas per section allowed us to draw the total thyroid surface area in pm 2 .
  • the surface areas positive for calcitonin and T4 two markers of late thyroid differentiation, were normalised for total thyroid surface area.
  • For stained surface quantification we used one of every two sections at E9.5 and El 1.5, one of every five sections at E13.5, and five sections per adult thyroid (3 months of age). We determined the surface area to obtain an estimate of the total stained surface for each thyroid and each marker.
  • Proliferation of Nkx2-l -positive cells at E9.5 was estimated by counting Ki67-positive nuclei among Nkx2-l -positive cells on every other section throughout the entire tissue sample at E9.5. At least three thyroids were analysed per genotype. The results are reported as mean ⁇ SEM.
  • the first immunohistochemistry steps were as described above. After application of the primary antibody, the sections were incubated with biotinylated secondary antibody for 1 hour. Immunostaining was then performed using the Vectastain ABC kit (Vector Laboratories, Burlingame, CA, USA) according to the manufacturer’s instructions. The sections were then incubated in 3,39- diaminobenzidine tetrahydrochloride and counterstained with hemalum-eosin.
  • Proteins prepared from mouse thyroid tissue collected in RIPA buffer and sonicated were quantified using the BCA protein assay (Thermo Fisher Scientific). Then, 20 pg of total protein was separated on BisTris polyacrylamide gel with a 4%-l2% gradient (Thermo Fisher Scientific) and transferred onto PVDF membranes (Thermo Fisher Scientific). The membranes were incubated with the primary antibodies mouse anti-Chop (1 :1,000, Cell Signalling Technology, Danvers, MA, USA) or rabbit anti-actin (Sigma- Aldrich) antibodies, followed by horseradish peroxidase-conjugated goat anti-mouse or anti-rabbit antibodies.
  • mouse anti-Chop 1 :1,000, Cell Signalling Technology, Danvers, MA, USA
  • rabbit anti-actin Sigma- Aldrich
  • Binding of secondary antibodies was revealed using the Amersham ECL Prime Detection Reagent Kit (GE Healthcare, Chicago, IL, USA). The protein bands on the membranes were scanned with the ImageQuant LAS 4,000 Station (GE Healthcare) then analysed using ImageJ 1 32s to determine the protein levels, with actin protein serving as an internal control.
  • the wild-type human TUBB1 sequence (accession number: Q9H4B7) was downloaded from the UniProt database, and the P160L mutation introduced into it. Both the wild-type and mutant TUBB1 sequences were modelled using Modeller 9.18 software 59 with PDB 4I4T chain B as the template 60 . The models were analysed using PyMOL visualisation software 61 .
  • Mutant P 160 L- 77/55/ was generated using a PCR-based site-directed mutagenesis method as described previously, using the Stratagene QuikchangeVR kit (Agilent Technologies) 62 .
  • Nthy (Nthy-ori 3.1) immortalised human thyroid cell lines were cultured as previously described 63 .
  • the Nthy cells were plated at 0.4 ⁇ l0 5 /well on poly-L-lysine-coated slides in l2-well plates 24 h before transfection then transfected with 500 ng of vectors containing wild-type or P160L mutant TUBB1 using XtremeGENE-HP-DNA, as recommended by the manufacturer (Roche Applied Science, Penzberg, Germany). After 24 h, cells were used for immunofluorescence as already described 64 . The cells were washed with pre-warmed PHEM buffer, fixed with 4% PFA, 0.2% glutaraldehyde, and 0.5% Triton, and permeabilised with PBS-Triton 0.1%.
  • Immunostaining was performed with rabbit anti-Myc antibody (1 :500, Cell Signaling Technology) and mouse anti-a-tubulin (DM1 A, 1 : 1,000, Sigma- Aldrich, Saint-Louis, MI, USA) then with Alexa Fluor 647 goat anti-rabbit and Alexa Fluor 555 goat anti-mouse antibodies (1 :400, Thermo Fisher Scientific).
  • CD34 + cells were isolated from peripheral blood using an immunomagnetic technique (Miltenyi Biotec, Bergisch Gladbach, Germany). The remaining population was cultured at 37°C in 5% C0 2 in Iscove Modified Dulbecco’s Medium (IMDM; Thermo Fisher Scientific) supplemented with 15% BIT 9500 serum substitute (Stemcell Technologies, Vancouver, Canada), a-monothioglycerol (Sigma-Aldrich), and liposomes (phosphatidylcholine, cholesterol, and oleic acid; all from Sigma-Aldrich), in the presence of human recombinant stem cell factor (SCF, 20 ng/mL, Miltenyi Biotec) and human thrombopoeitin (50 nM, Miltenyi Biotec) added once on day 0 to the culture medium, followed by 20 nM thrombopoeitin alone on day 6 with no further SCF addition.
  • SCF human recombinant stem cell factor
  • SCF human
  • megakaryocytes were plated on a BSA-coated surface (chamber slide, Ibidi, Martinsried, Germany) on day 10. On day 13 or 14, the megakaryocytes were fixed using 4% paraformaldehyde and stained for b- tubulin.
  • venous blood from healthy donors or patients was collected in 10% ACD/A buffer (75 mM sodium citrate, 44 mM citric acid, 136 mM dextrose, pH 4.5). Platelets were washed as previously described 65 in the presence of apyrase (100 mU/mL) and prostaglandin El (1 mM) to minimise platelet activation.
  • Platelet counts in patients and controls were adjusted to similar levels (3 ⁇ 0 8 platelets/mL) in Tyrode's buffer (137 mM NaCl, 2 mM KC1, 0.3 mM NaH 2 P0 4 , 1 mM MgCh, 5.5 mM glucose, 5 mM N-2-hydroxyethylpiperazine- N’-2-ethanesulfonic acid, 12 mM NaHCCE, and 2 mM CaCl 2 , pH 7.3).
  • Tyrode's buffer 137 mM NaCl, 2 mM KC1, 0.3 mM NaH 2 P0 4 , 1 mM MgCh, 5.5 mM glucose, 5 mM N-2-hydroxyethylpiperazine- N’-2-ethanesulfonic acid, 12 mM NaHCCE, and 2 mM CaCl 2 , pH 7.3.
  • Platelet aggregation was monitored by measuring light transmission through a stirred suspension of washed platelets (3 ⁇ l0 8 /mL) at 37°C using a Chrono-Log aggregometer (Chrono- Log Corporation, Havertown, PA, USA), as previously described 66 . Platelet aggregation was triggered by ADP and type I collagen.
  • Washed platelets (300 uL; 3 - l0 8 /mL) were lysed in Laemmli sample buffer (10 mM HEPES, 2% SDS, 10% glycerol, 5 mM EDTA). The proteins were separated by sodium dodecyl sulphate (SDS)-polyacrylamide gel electrophoresis and transferred to nitrocellulose membranes, which were incubated with the primary antibodies rabbit anti-a tubulin (Clone EP1332Y; Merck Millipore, Billerica, MA, USA) or rabbit anti-b! -tubulin antibody (donated by Frangois Lanza).
  • Laemmli sample buffer 10 mM HEPES, 2% SDS, 10% glycerol, 5 mM EDTA.
  • SDS sodium dodecyl sulphate
  • nitrocellulose membranes which were incubated with the primary antibodies rabbit anti-a tubulin (Clone EP1332Y; Merck Millipore, Billerica
  • Immunoreactive bands were visualised with enhanced chemiluminescence detection reagents (Perbio Science, Thermo Fisher Scientific) using a G:BOX Chemi XT 16 Image System then quantified using Gene Tools version 4.03.05.0 (Syngene, Cambridge, UK).
  • WES whole exome sequencing
  • Thyroid scintigraphy showed an ectopic thyroid.
  • the father carried the same heterozygous mutation but was not able to undergo thyroid ultrasonography.
  • Thyroid ultrasonography and scintigraphy showed thyroid hemi-agenesis with absence of the left lobe.
  • a patient II.1, P6 with CH and an ectopic thyroid was shown by targeted NGS to have a heterozygous frameshift TUBB1 mutation (c.35delG, p.Cys 12Lcufs* 12, rs77324804) that created a premature stop codon at amino acid 23 (Fig. la).
  • CH was diagnosed neonatally based on TSH elevation (476 m ⁇ u/mL) and low free T4 and T3 levels (8 pmol/L and 5.6 pmol/L, respectively).
  • L-thyroxine therapy was started at 11 days of age. Both parents were of French descent.
  • the father (1.1, P7) carried the same heterozygous mutation and had normal thyroid function with mild thyroid lobe asymmetry by ultrasonography (right lobe, 6.9 mL; left lobe, 5 mL).
  • the other siblings and mother had normal thyroid function and morphology and did not carry the mutation.
  • a Burden test was applied to determine whether TUBB1 was significantly enriched in rare variants in the 270 patients with CH and TD versus 406 Caucasian controls from the 1,000 Genomes project.
  • the patients had athyreosis, ectopia, hemi-agenesis, or hypoplasia.
  • the three amino acids affected by the TUBB1 mutations are strictly conserved across species, from humans to zebrafish, and across all b-tubulins (data not shown). All three mutations were located in the first part of TUBB1, i.e., in the N-terminal domain needed for guanosine triphosphate (GTP) activity (Fig. lb).
  • GTP guanosine triphosphate
  • Fig. lb The c.3 l8C>G and c.35delG mutations created a premature stop codon, thereby removing the intermediate and C-terminal domains required for microtubule-associated protein (MAP) binding. 25
  • b ⁇ -tubulin is expressed in the developing thyroid in humans and mice
  • b 1 -tubulin expression has so far been reported only in megakaryocytes and platelets 26,27 .
  • Our finding of TUBB1 mutations in patients with TD prompted us to look for b ⁇ -tubulin expression in thyroid tissue.
  • TUBB1 mRNA was expressed at 8, 10, and 12 GW and in adulthood (data not shown).
  • Tubbl was expressed at E13.5 and strongly at E15.5, E17.5, and adulthood (data not shown).
  • To refine our study of Tubbl expression we used cells sorted from mice thyroid tissue based on well-accepted markers 28 . As expected, expression was strongest in platelets sorted using the specific megakaryocyte lineage marker CD41 (data not shown). However, Tubbl was also expressed in EpCam-positive epithelial-cell populations containing thyrocytes, at El 7.5 and adulthood (data not shown).
  • the sole missense mutation that should give rise to a full-length protein we first compared the configurations of the mutated and wild-type proteins.
  • the P160L mutation is located at the end of helix H4 (data not shown).
  • the proline residue stabilises loop H4- S5 and places R156 in a position that promotes a salt bridge interaction with D197 in the b- sheet S6.
  • R162 in loop H4-S5 and N195 in the b-sheet S6 establish hydrogen bonds with R156.
  • pro line 160 to leucine might affect the loop conformation of the H4-S5 loop and thus disrupting the interaction network mediated by R156.
  • the P160L mutation is most likely affecting the conformation of the B -tubulin, which could lead to dysfunctions of the a/b-tubulin dimer.
  • Tubbl knock-out in mice affects thyroid development and function
  • Thyroid gland morphology in mice To study thyroid gland development from the early stages of budding and migration of the median strom (MA) and ultimobranchial bodies (UB) to the late stages of differentiation, we used immunohistochemistry, surface quantification, and quantitative PCR (data not shown). For most of the experiments, we used Nkx2-l as a marker of progenitor and differentiated thyroid cells. At E9.5, thyroid strom surface area and proliferation ratio were significantly greater in TubbT 1 mice than in wild-type mice (data not shown.
  • T4-positive thyroid surface area relative to total thyroid surface area was shown by immunohistochemistry to be significantly increased at E17.5 in TubbT /_ versus wild-type embryos (data not shown). Calcitonin-positive thyroid surface area relative to total thyroid surface area was also significantly greater in the mutants. Thus, final thyroid differentiation was abnormal in TubbT /_ embryos, with increases in T4 and calcitonin that probably reflected impaired hormone secretion.
  • Thyroid gland function and structure in adult mice We compared thyroid hormone status in adult Tubbl ' and wild-type mice. At 3 months of age, serum TSH levels were higher and T4 levels lower in Tubbl 1 than in wild-type mice, suggesting hypothyroidism in the mutants data not shown). These results are consistent with the finding of CH in our patients carrying TUBB1 mutations.
  • ER dilation was seen in Tubbl 1 compared to wild-type thyrocytes (data not shown), indicating ER stress, which was confirmed by the findings of increased Chop and XBPs expression by quantitative PCR and of increased Chop protein levels by western blotting in Tubbl 1 thyroids (data not shown).
  • the ER marker KDEL co- localized with TG in Tubbl 1 and wild-type mice but was especially abundant in disorganized areas of adult Tubbl 1 thyroids (data not shown).
  • Haematological parameters Thrombocytopenia was a feature in patients with TUBB1 mutations studied by other groups 20-22 .
  • MPV mean platelet volume
  • a blood smear analysis (data not shown) demonstrated variations in platelet size, with the presence of macroplatelets.
  • parents 1.1 and 1.2 of the family Fl had MPV values at the top of the normal range (10.2 fL and 1 1.3 fL, respectively; normal, 7.5-11.2 fL) with many macroplatelets. Electron microscopy confirmed the large platelet size (data not shown).
  • the increased platelet size could be related to defects in their biogenesis.
  • Pl and P4 from peripheral blood progeniors and analysed proplatelet formation (data not shown).
  • shaft thickness and coiled element diameter of the future platelets were significantly increased compared to controls, indicating that the TUBB1 mutations affected proplatelet formation and platelet size.
  • a discoid shape is a normal feature of circulating or non-activated platelets.
  • Pl and P3 Fl
  • Pl and P3 indicating abnormal platelet activation.
  • P6 and P7 family 3
  • the marker levels were comparable to those in controls (data not shown).
  • Tubbl 2 mice Provide the first evidence of a role for b ⁇ -tubulin in TD and hypothyroidism.
  • the thyroid glands of Tubbl 2 mice exhibited deficient proliferation at E9.5, abnormal migration at El 1.5 and E13.5, and hormone secretion failure at E17.5 and adulthood. All these mechanisms require normal microtubule organisation and function.
  • the idea that microtubule functions are fine-tuned, and thus adapted to specific cellular role by the expression of specific tubulin isotypes is an emerging concept known as the‘tubulin code’ 17 .
  • Tubulin mutations have been more and more linked to different human disorders, in particular to neurological disorders 30 .
  • Microtubules are essential for mitosis to unfold normally 31 .
  • b ⁇ -tubulin is the most divergent isotype of tubulin, and its incorporation into microtubule is expected to change the properties of these filaments. Losing b ⁇ -tubulin thus certainly alters microtubule properties, and our data provide strong evidence that these particular properties are essential for adequate thyroid development and function.
  • Thyroid dysfunction can affect the haemostatic balance 48,49 , with clinical effects that vary across thyroid disorders 50 .
  • MPV correlated positively with the TSH level 51 .
  • a TUBB1 -mutation screening study in patients with hypothyroidism and altered MPV and/or a history of thrombotic disease would be of interest.
  • the clinical symptoms to accompany platelet hyperaggregability include unexplained arterial thrombosis, complications during pregnancy, and less frequently, venous thromboembolism.
  • Patients bearing a TUBB1 mutation with cardiovascular risk factors could be monitored and the question arises whether antiplatelet drugs might be effective as treatment for and/or prophylaxis 52 .
  • the c.35delG mutation in P6 and P7 did not cause hyperaggregation.
  • the patients with the c.35delG mutation had the same thyroid phenotype as other families with other mutations. At least one patient per family had thyroid gland ectopia with CH. Neither thyroid nor platelet phenotype severity correlated with the type of mutation. Further investigations are needed to elucidate the difference between the effects of c.35delG and pl60L/pYl06X on platelet function.
  • TUBB1 mutations constitute a model of dominant inheritance of CH with TD. Most known mutations responsible for TD and previously described TUBB1 mutations causing macrothrombocytopenia are also dominant 10,13,20 22,53 . Our data indicate high penetrance for platelet alterations and variable expressivity for TD, ranging from TD without CH to TD with CH and leading to variable types of TD (ectopia, hypoplasia, hemi-agenesis, or asymmetric thyroid gland). No patient had athyreosis. TUBB1 mutations were found in only 1.1% of our cohort with CH and TD patients. The Burden test showed enrichment in rare TUBB1 variants carriers in the cohort versus controls. Our data increment the number of causative genes for thyroid dysgenesis but with a novel phenotype associating platelets disorder.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • Physics & Mathematics (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The genetic causes of congenital hypothyroidism due to thyroid dysgenesis (TD) remain largely unknown. The inventors identified three novel TUBB1 gene mutations that co-segregated with TD in three affected families. TUBB1 (Tubulin, Beta 1 Class VI) encodes a member of the β-tubulin protein family. In mice, Tubb1 knock-out in vivo disrupted microtubule integrity by preventing β1-tubulin incorporation and impaired thyroid migration and thyroid hormone secretion. In addition, TUBB1 mutations caused hyperaggregation of human platelets. The data highlight unexpected roles for β1-tubulin in thyroid development and function and in platelet physiology. Accordingly, the present invention relates to a method of diagnosing thyroid dysgenesis in a subject, comprising detecting a mutation in the TUBB1 gene in a sample obtained from said subject, wherein detecting the presence of a mutation in the TUBB1 gene is considered to be indicative of thyroid dysgenesis.

Description

DETECTION OF TUBB1 MUTATIONS FOR DIAGNOSING THYROID
DYSGENESIS
FIELD OF THE INVENTION:
The present invention relates to a method for diagnosing or predicting thyroid dysgenesis by detecting a mutation in Tubulin, Beta 1 Class VI ( TUBB1 ).
BACKGROUND OF THE INVENTION:
Thyroid dysgenesis (TD) is a feature in 65% of patients with congenital hypothyroidism (CH), which is the most common neonatal endocrine disorder (1/3,500 neonates)1,2. TD includes a vast spectrum of developmental thyroid anomalies encompassing athyreosis, thyroid ectopia, hypoplasia of an orthotopic gland, and hemiagenesis1,3. During embryogenesis, the midline thyroid anlage appears on embryonic day E.8.5 in mice and at 3 gestational weeks (GW) in humans. The midline anlage and ultimobranchial bodies migrate and fuse in the definitive pretracheal position on El 3.5 in mice and at 7 GW in humans4,5. The cells differentiate into thyrocytes organized in follicles or C-cells 6. Abnormalities at any step of thyroid development may result in TD with variable degrees of hypothyroidism or rarely normal thyroid function7. Studies of sporadic and familial TD covering a wide clinical spectrum identified mutations in eight genes: PAX8, NKX2-1, FOXE1, NKX2-5, TSHR, GLIS3, NTN1, and BOREALIN 8-14. However, mutations in these genes are found in only 5% of all patients with TD and identification of causative mutations remains a challenging task.
TUBB1 (Tubulin, Beta 1 Class VI) encodes a member of the b-tubulin protein family. b-tubulins are one of two core protein families that heterodimerise a/b-tubulin dimers, which assemble into microtubules, one of the major cytoskeletal structures. Microtubules are involved in crucial processes such as cell division, the growth polarity, and migration; intracellular trafficking; and cell communication17,18. The bΐ isotype of tubulin (TUBB1) has been described as specifically expressed in platelets and megakaryocytes and involved in proplatelet formation and platelet release19. Three mutations in the intermediate and the C-terminal domain of TUBB1 have been identified in patients with a rare autosomal dominant disease congenital macrothrombocytopenia, in which impaired microtubule assembly results in low platelet counts and macroplatelets20-22. Patients with these TUBB1 mutations had mild bleeding, which could be due to thrombocytopenia. Features reported to date in Tubbl -knockout mice include thrombocytopenia and spherical platelets23, but not thyroid abnormalities. SUMMARY OF THE INVENTION:
The present invention relates to a method for diagnosing or predicting thyroid dysgenesis by detecting a mutation in Tubulin, Beta 1 Class VI ( TUBB1 ). In particular, the present invention is defined by the claims.
DETAILED DESCRIPTION OF THE INVENTION:
The genetic causes of congenital hypothyroidism due to thyroid dysgenesis (TD) remain largely unknown. The inventors identified three novel TUBB1 gene mutations that co- segregated with TD in three affected families. TUBB1 (Tubulin, Beta 1 Class VI) encodes a member of the b-tubulin protein family. In mice, Tubbl knock-out in vivo disrupted microtubule integrity by preventing bΐ -tubulin incorporation and impaired thyroid migration and thyroid hormone secretion. In addition, TUBB1 mutations caused hyperaggregation of human platelets. The data highlight unexpected roles for bΐ -tubulin in thyroid development and function and in platelet physiology.
Accordingly, the first object of the present invention relates to a method of diagnosing thyroid dysgenesis in a subject, comprising detecting a mutation in the TUBB1 gene in a sample obtained from said subject, wherein detecting the presence of a mutation in the TUBB1 gene is considered to be indicative of thyroid dysgenesis.
As used herein, the term“thyroid dysgenesis” has its general meaning in the art and refers to a permanent thyroid hormone deficiency present from birth and resulting from an abnormality in the development of thyroid. Thyroid dysgenesis comprises e.g. thyroid ectopy, athyreosis and thyroid hypoplasia.
As used herein, the term“subject” is preferably a human. More particularly, the subject is a new-bom or a foetus. As used herein, a“new-born” is a child who is less than 2 weeks old, particularly less than 1 week old, more particularly less than 3 days old. Thus, when the method is method is performed“just after birth”, it means that the method is performed within 2 weeks, particularly 1 week, more particularly 3 days after birth.
As disclosed above, thyroid dysgenesis results from an abnormality of the development of thyroid during embryo development. The present invention is thus particularly interesting for detecting thyroid dysgenesis just after birth, i.e. wherein the subject is a new-bom. In some embodiments, the method according to the present invention is performed just after birth and after having suspected thyroid dysgenesis during embryo development, e.g. by observing an abnormality in the development of the embryo’s thyroid during an ultrasonography, particularly obstetric ultrasonography. In some embodiments, the method according to the present invention is performed in a child, preferably a new-born, whose parents (1 or both of them) present thyroid dysgenesis. Particularly, the method according to the present invention is performed in a child, preferably a new-born, whose parents (1 or both of them) are identified has displaying a mutation in the TUBB1 gene.
The method according to the present invention is interesting for detecting an abnormality in the thyroid development at the embryo stage. In some embodiments, the present invention relates to a prenatal method for diagnosing or predicting thyroid dysgenesis in a foetus by detecting a mutation in the TUBB1 gene in a sample obtained from said foetus or from said foetus’s mother.
In some embodiments, TD is suspected by routine screening which shows thyroid stimulating hormone (TSH) elevation and low T4 level. Other tests, such as thyroid radionuclide uptake and scan, thyroid sonography, or serum thyroglobulin determination may be carried out before performing the method of the present invention.
As used herein, the term“TUBB1” has its general mean in the art and refers to gene that encodes a member of the beta tubulin protein family: tubulin beta 1 class VI. Beta tubulins are one of two core protein families (alpha and beta tubulins) that heterodimerize and assemble to form microtubules tubulin beta 1 class VI is encoded by the TUBB1 gene (available under the reference ENSG00000101162 (gene) in the Ensembl Gene Database) or referenced as GENE ID : 81027 in the NCBI database. An exemplary nucleic acid sequence is represented by SEQ ID NO: l and an exemplary amino acid sequence is represented by SEQ ID NO:2.
SEQ ID NO: 1
1 ggggcagtat tctgtgttga gggaggaaaa acactccctt ccaaaagcat gacaggcaga
61 aagcagagaa gggccaggac tggctgaggg cggggagctg ggcctctggg gtggacacac
121 ccttggtcac attgtgaggg tagcttggtt ggccagtccc accactgcag tgaccacagt
181 tgtgttgggc tcacaccagt gaaccgaagc tctggattct gagagtctga ggattccgtg
241 aagatctcag acttgggctc agagcaagga tgcgtgaaat tgtccatatt cagattggcc
301 agtgtggcaa ccagatcgga gccaagttct gggagatgat tggtgaggaa cacgggatcg
361 acttggctgg gagcgaccgc ggggcctcgg ccttgcagct ggagagaatc agcgtgtact
421 acaacgaagc ctacggtagg aaatatgtgc cccgagcagt cttggtggac ctagaacctg
481 ggacgatgga cagcattcga tctagcaaat taggagctct ctttcaaccc gacagttttg
541 tccatggtaa ctctggggct ggcaacaact gggccaaagg ccactacacg gagggagccg
601 agctgatcga gaatgtccta gaggtggtga ggcacgagag tgagagctgt gactgcctgc
661 agggcttcca gatcgtccac tccctgggcg ggggcacagg ctccgggatg ggcactctgc
721 tcatgaacaa gattagagag gagtacccgg accggatcat gaattccttc agcgtcatgc
781 cttctcccaa ggtgtcggac actgtggtgg agccctacaa cgcggttctg tctatccacc
841 agctgattga gaatgcagat gcctgtttct gcattgacaa tgaggccctc tatgacatct
901 gcttccgtac cctgaagctg acgacaccca cctatgggga tctcaaccac ctagtgtcct
961 tgaccatgag cggcataacc acctccctcc ggttcccggg tcagctcaac gcagacctgc
1021 gcaagctggc ggtgaacatg gtccccttcc cccgcctgca cttctttatg cccggctttg
1081 ccccactcac ggcccagggc agccagcagt accgagccct ctccgtggcc gagctcaccc
1141 agcagatgtt cgatgcccgc aataccatgg ctgcctgtga cctccgccgt ggccgctacc
1201 tcacagtggc ctgcattttc cggggcaaga tgtccaccaa ggaagtggac cagcaactgc
1261 tctccgtgca gaccaggaac agcagctgct ttgtggagtg gattcccaac aacgtcaagg
1321 tggctgtctg cgacatcccg ccccgggggc tgagcatggc cgccaccttc attggcaaca 1381 acacggccat ccaagagatc tttaataggg tctctgagca tttctcagcc atgttcaaaa 1441 ggaaagcttt tgtgcactgg tacaccagcg aagggatgga cataaacgaa tttggggaag 1501 ctgaaaataa catccatgat ttggtatccg agtaccaaca atttcaagat gccaaagcag 1561 ttctagagga agatgaagag gtcacggagg aggcagaaat ggagccagaa gataagggac 1621 attaactgtg agagaagctg tgccgcggag tcgcttacag aacagtttct cattagatga 1681 gtgtttctcc tgcagcactc caaaacccac tctgcactgc agcacagtga atgatatgca 1741 ctcaccatta gcttcgacac agggactgag ggagacaggt ggggagcagc tgacaggcat 1801 tagggtcttt gctgacatct actaaccttg aagagtttga tgttcagtgc atacttatta 1861 acttaaaaaa atagcaaatt tattgtaaag tgctcccttt gtttcaaagt gtttgccagg 1921 catccagact acacgtgtgg atttgcaggg agccactgga gttggtgtta catttttata 1981 ctttagcagc actgataggc accctggaat cctcacttgg tatccgaggg ctactaagac 2041 tctttcctta ggttctttcc tctgagcaaa cactgactgg catcctgctt tccagtgcct 2101 gccagcctcc agaagagcca ggtgcctgac tagtacatgg ggagctacag agccaaggtc 2161 aatgtgagtc aacatccact agaaatatcc atgttgtgta gacctgtgca tacaacatgc 2221 taactggaaa agaggaaaaa agaaaagcca cagtcctctc cacaaaaata cctggtccaa 2281 acaagaaaaa caaaaagaca agcaaaacta aagaactgca gtcttctgat ctttatttct 2341 gaagagctag cctttaacat atatgtttat atagtttaaa tttcttacta ctgttagatc 2401 ccaggaattc attaataatc atccttggct ttccttttaa aggctatttt gaaatggtct 2461 tttcactttc attcagtcat caccccccaa aatgctctgc agcctctctg ctctttgaga 2521 aagggcacac catgcgctcg gcaaccattc aaatgcagga attaagcagc aatggctgca 2581 gtgtccttct cagttatgga ggacatcgtc tcattaggga acttttacag ttcaaattaa 2641 tttgcagaag ttgccataaa tgtttgcata atgacatagc tttaagcact acatgatttt 2701 aatctgctca cattataaca ggaccaaata cacaagagcg taatcaaatc atctgtaact 2761 tcttaattac agtttaccta tttctgacat gcagcactgc catctcttcc agcaccatca 2821 gggttttaat ggccctctag aattaccact gagatacact atttgatcca tggataaccg 2881 gtaatgggaa aatgctccga ccctcaatgc agtaaatatt tacttgcagg caactgggtt 2941 ctcatctctt gatttgcttt tgtaatcagc aataataaaa tagcaggtag atggatgaca 3001 gttgctcatt ctgagaaact tcactctttt cacttatgca tcacgaggaa ataactaaaa 3061 tacataccaa gagaaaaata ccttgccatc ggatcatcaa caagtcttct atttacaaac 3121 ttcaaaaaac acaaaacaac attcatgttt taaatgcttt ctacttgtgg ttcaagaagc 3181 actagattta gtaagaaact ctacctatat acttagtttg aagttagtaa cttcctgaga 3241 tgctaaagac ttacagcctg cgattataca aggatttaca catgcttcct ctggtgcttt 3301 acttcccaaa cctaaaaaag caatgaaata gatgtaagga aggagggatt taaacctttt 3361 aaaaaacttt tgctgactta tattactgta aagatttgtt tgctcaatag taatcattaa 3421 actacaaagt aattcaattt taaatggcaa aattgcttta tttcagacta aataaattcc 3481 ttttcttgaa gcctaa
SEQ ID NO:2:
1 mreivhiqig qcgnqigakf wemigeehgi dlagsdrgas alqlerisvy yneaygrkyv
61 pravlvdlep gtmdsirssk lgalfqpdsf vhgnsgagnn wakghytega elienvlevv 121 rhesescdcl qgfqivhslg ggtgsgmgtl lmnkireeyp drimnsfsvm pspkvsdtvv 181 epynavlsih qlienadacf cidnealydi cfrtlklttp tygdlnhlvs ltmsgittsl 241 rfpgqlnadl rklavnmvpf prlhffmpgf apltaqgsqq yralsvaelt qqmfdarntm 301 aacdlrrgry ltvacifrgk mstkevdqql lsvqtrnssc fvewipnnvk vavcdipprg 361 lsmaatfign ntaiqeifnr vsehfsamfk rkafvhwyts egmdinefge aennihdlvs 421 eyqqfqdaka vleedeevte eaemepedkg h
As used herein, the term "mutation" has its general meaning in the art and refers to any detectable change in genetic material, e.g. DNA, RNA, cDNA, or in an amino acid sequence encoded by such a genetic material. This includes gene mutations, in which the structure (e.g. DNA sequence) of a gene is altered any gene as well as protein mutations, in which the amino- acid structure of the protein is altered. Generally a mutation is identified in a subject by comparing the sequence of a nucleic acid or of a polypeptide expressed by said subject with the corresponding nucleic acid or polypeptide expressed in a control population. Typically, mutations are accessible in the Single Nucleotide Polymorphism Database (dbSNP), which is a free public archive for genetic variation within and across different species developed and hosted by the National Center for Biotechnology Information (NCBI) in collaboration with the National Human Genome Research Institute (NHGRI).
The present inventors have identified different mutations of the TUBB1 gene involved in thyroid dysgenesis. One of these mutations is a missense homozygous TUBB1 mutation (c.479C>T, p.Pl60L, rs759l 17911) which is a substitution of a cytosine residue into a thymine residue at position 479 of the TUBB1 gene. This mutation encodes for the substitution of the proline residue at position 160 by a leucine residue in the mature protein. Another mutation identified by the present inventors is a heterozygous TUBB1 mutation (c.3 l8C>G, p.Yl06X) which is the substitution of a cytosine residue into a guanine residue at position 318 of the TUBB1 gene. This mutation leads to the creation a premature stop codon at amino acid 106. Another mutation identified by the inventors is a heterozygous frameshift TUBB1 mutation (c.35delG, p.Cysl 2Lcufs* 12, rs77324804) that created a premature stop codon at amino acid 23. All three mutations are located in the first part of TUBB1, i.e., in the N-terminal domain needed for guanosine triphosphate (GTP) activity, i.e. the domain ranging from the amino acid residue at position 1 to the amino acid residue at position 206 in the protein. All three TUBB 1 mutations identified lead to nonfunctional a/b -tubulin dimers that cannot be incorporated into microtubules. The mutations lead to an early abnormal proliferation of progenitors, delayed thyroid migration, defective thyroid tissue differentiation, and impaired thyroid hormone release. More interestingly, the mutations also affect bΐ -tubulin expression in platelets and result in abnormally large platelet size, probably as a consequence of proplatelet abnormal formation. Moreover, the p.Pl60L and r.U106C mutations induce significant basal platelet activation and hyperaggregation in response to agonists, whereas the c.35delG mutation does not seem to affect platelet function.
As used herein, the term“sample” refers to sample liable to contain nucleic acid molecules including mRNA, genomic DNA and cDNA derived from mRNA. DNA or RNA can be single stranded or double stranded. These may be utilized for detection by amplification and/or hybridization with a probe, for instance. The nucleic acid sample may be obtained from any cell source or tissue biopsy. Non-limiting examples of cell sources available include without limitation blood cells, buccal cells, epithelial cells, fibroblasts, or any cells present in a tissue obtained by biopsy. Cells may also be obtained from body fluids, such as blood, plasma, serum, lymph, etc. In some embodiments, the sample is an amniotic fluid sample. The sample may further be any biological sample wherein foetal DNA may be detected. For instance, in this case, the biological sample may be a sample obtained from the mother but wherein foetal DNA can be found. As disclosed e.g. in Hixson et al (J Lab Autom; 20(5):562-73, 2015) or in the reference EP patent N° 0994963, foetal DNA is detectable in maternal serum or plasma samples. Abnormalities in the foetus genetic material can thus be detected by directly analysing the foetal DNA present in the mother’s blood. Thus, in some embodiments, the sample according to the present invention is a maternal serum or plasma sample.
DNA may be extracted using any methods known in the art, such as described in Sambrook et ah, 1989. RNA may also be isolated, for instance from tissue biopsy, using standard methods well known to the one skilled in the art such as guanidium thiocyanate- phenol-chloroform extraction.
TUBB1 mutations may be detected in a RNA or DNA sample, preferably after amplification. For instance, the isolated RNA may be subjected to coupled reverse transcription and amplification, such as reverse transcription and amplification by polymerase chain reaction (RT-PCR), using specific oligonucleotide primers that are specific for a mutated site or that enable amplification of a region containing the mutated site. According to a first alternative, conditions for primer annealing may be chosen to ensure specific reverse transcription (where appropriate) and amplification; so that the appearance of an amplification product be a diagnostic of the presence of a particular TUBB1 mutation. Otherwise, RNA may be reverse- transcribed and amplified, or DNA may be amplified, after which a mutated site may be detected in the amplified sequence by hybridization with a suitable probe or by direct sequencing, or any other appropriate method known in the art. For instance, a cDNA obtained from RNA may be cloned and sequenced to identify a mutation in TUBB1 sequence. Actually numerous strategies for genotype analysis are available (Antonarakis et ah, 1989; Cooper et ah, 1991; Grompe, 1993). Briefly, the nucleic acid molecule may be tested for the presence or absence of a restriction site. When a base substitution mutation creates or abolishes the recognition site of a restriction enzyme, this allows a simple direct PCR test for the mutation. Further strategies include, but are not limited to, direct sequencing, restriction fragment length polymorphism (RFFP) analysis; hybridization with allele-specific oligonucleotides (ASO) that are short synthetic probes which hybridize only to a perfectly matched sequence under suitably stringent hybridization conditions; allele-specific PCR; PCR using mutagenic primers; ligase- PCR, HOT cleavage; denaturing gradient gel electrophoresis (DGGE), temperature denaturing gradient gel electrophoresis (TGGE), single-stranded conformational polymorphism (SSCP) and denaturing high performance liquid chromatography (Kuklin et ah, 1997). Direct sequencing may be accomplished by any method, including without limitation chemical sequencing, using the Maxam-Gilbert method ; by enzymatic sequencing, using the Sanger method ; mass spectrometry sequencing; sequencing using a chip-based technology; and real- time quantitative PCR. Preferably, DNA from a subject is first subjected to amplification by polymerase chain reaction (PCR) using specific amplification primers. However several other methods are available, allowing DNA to be studied independently of PCR, such as the rolling circle amplification (RCA), the InvaderTMassay, or oligonucleotide ligation assay (OLA). OLA may be used for revealing base substitution mutations. According to this method, two oligonucleotides are constructed that hybridize to adjacent sequences in the target nucleic acid, with the join sited at the position of the mutation. DNA ligase will covalently join the two oligonucleotides only if they are perfectly hybridized. Therefore, useful nucleic acid molecules, in particular oligonucleotide probes or primers, according to the present invention include those which specifically hybridize the regions where the mutations are located. Oligonucleotide probes or primers may contain at least 10, 15, 20 or 30 nucleotides. Their length may be shorter than 400, 300, 200 or 100 nucleotides. In some embodiments, the TUBB1 mutations of the present invention may be detected by Next Generation Sequencing or NGS. As used herein, the term "Next Generation Sequencing" or“NGS” refers to a relatively new sequencing technique as compared to the traditional Sanger sequencing technique. For review, see Shendure et al, Nature Biotech., 26(10): 1135-45 (2008), which is hereby incorporated by reference into this disclosure. For purpose of this disclosure, NGS may include cyclic array sequencing, micro electrophoretic sequencing, sequencing by hybridization, among others. By way of example, in a typical NGS using cyclic-array methods, genomic DNA or cDNA library is first prepared, and common adaptors may then be ligated to the fragmented genomic DNA or cDNA. Different protocols may be used to generate jumping libraries of mate-paired tags with controllable distance distribution. An array of millions of spatially immobilized PCR colonies or "polonies" is generated with each polonies consisting of many copies of a single shotgun library fragment. Because the polonies are tethered to a planar array, a single micro liter- scale reagent volume can be applied to manipulate the array features in parallel, for example, for primer hybridization or for enzymatic extension reactions. Imaging-based detection of fluorescent labels incorporated with each extension may be used to acquire sequencing data on all features in parallel. Successive iterations of enzymatic interrogation and imaging may also be used to build up a contiguous sequencing read for each array feature.
The mutation may be also detected at a protein level (e.g. for loss of function mutation) according to any appropriate method known in the art. In particular a biological sample, such as a tissue biopsy, obtained from a subject may be contacted with antibodies specific of a mutated form of TUBB1 protein, i.e. antibodies that are capable of distinguishing between a mutated form of TUBB1 and the wild-type protein, to determine the presence or absence of a TUBB1 specified by the antibody. The antibodies may be monoclonal or polyclonal antibodies, single chain or double chain, chimeric antibodies, humanized antibodies, or portions of an immunoglobulin molecule, including those portions known in the art as antigen binding fragments Fab, Fab', F(ab')2 and F(v). They can also be immunoconjugated, e.g. with a toxin, or labelled antibodies. Whereas polyclonal antibodies may be used, monoclonal antibodies are preferred for they are more reproducible in the long run. Procedures for raising“polyclonal antibodies” are also well known. Alternatively, binding agents other than antibodies may be used for the purpose of the invention. These may be for instance aptamers, which are a class of molecule that represents an alternative to antibodies in term of molecular recognition. Aptamers are oligonucleotide or oligopeptide sequences with the capacity to recognize virtually any class of target molecules with high affinity and specificity. Such ligands may be isolated through Systematic Evolution of Ligands by Exponential enrichment (SELEX) of a random sequence library.
In some embodiments, the method according to the present invention is performed after having suspected thyroid dysgenesis during embryo development and treated the foetus thereby identified as presenting an abnormality in the thyroid development. An example of treatment which can be administered to a foetus identified as presenting an abnormality in the development of thyroid is intra-amniotic injection of Levothyroxine. Such treatment is e.g. disclosed in Leger et al (ESPE-PES-SLEP-JSPE-APEG-APPES-ISPAE; Congenital Hypothyroidism Consensus Conference Group. European Society for Paediatric Endocrinology consensus guidelines on screening, diagnosis, and management of congenital hypothyroidism. J Clin Endocrinol Metab. 2014 Feb;99(2):363-84) and in Ribault et al (French Fetal Goiter Study Group. Experience with intraamniotic thyroxine treatment in nonimmune fetal goitrous hypothyroidism in 12 cases. J Clin Endocrinol Metab. 2009 Oct;94(lO):373 l-9).
In some embodiments, when it is concluded that the subject suffers from TD, a therapy is administered. Levothyroxine (l-thyroxine) is the therapy of choice. Treatment is typically initiated in any infant with a positive screening result. The dose and timing of thyroid hormone replacement are important in achieving optimal neurocognitive outcome. Thus, the goal of treatment should be to restore the serum T4 to > 129 mmol/L (> 10 pg/dl) as rapidly as possible. The recommended initial l-thyroxine dose set forth by the American Academy of Pediatrics (AAP) and the European Society for Paediatric Endocrinology (ESPE) is 10-15 mcg/kg per day. In term infants this amounts to an average of 37.5 to 50 meg per day.
Since the subjects harbouring said mutations are also at risk of having thrombosis, in particular subject harbouring the p.Pl60L or r.UIObC mutations, a prophylactic treatment mays also be prescribed. Typically said treatment may consist in antagonists of platelet activation and particularly of ADP receptors such as thienopyridines (eg, clopidogrel, prasugrel) or non-thienopyridine ADP inhibitors (eg, ticagrelor), or any newer anti-platelet agent (eg, inhibitors of the thrombin receptor).
The invention will be further illustrated by the following figures and examples. However, these examples and figures should not be interpreted in any way as limiting the scope of the present invention.
FIGURES:
Figure 1: Molecular genetics
a) Pedigrees of three families with TUBB1 mutations. Family Fl has three affected individuals with homozygous mutations, family F2 has two affected individuals with heterozygous mutations, and family F3 has two affected individuals with heterozygous mutations. Thus, all seven patients (Pl to P7) carry at least one mutated allele and have thyroid dysgenesis (TD) and macroplatelets. The patients are represented with filled symbols and unaffected family members with open symbols. N, not mutated, m, mutated
b) Location of TUBB1 mutations in the cDNA and of the corresponding changes in the protein. Exons are represented by boxes numbered from 1 to 4. The black box represents the protein domain responsible for encoding guanosine triphosphate (GTP) and the box points represents the domain for microtubule-associated protein (MAP) binding. The arrows show the consequences of the three TUBB1 mutations in our patients, all of which are in the GTP domain.
EXAMPLE:
Methods:
Subjects
A consanguineous family in which two children had TD and CH and another had TD with normal thyroid function was studied. Subsequently, genetic testing was performed in 270 patients (184 girls and 86 boys) with CH and TD (ectopic thyroid gland, n=l67; athyreosis, n=77; hemi-agenesis, n=20; and thyroid hypoplasia, n=6). This study was approved by our institutional review board and informed consent were collected.
Detection of mutations in humans
Genomic DNA was isolated from whole blood. Exome capture and sequencing were performed at the genomics platform of the Imagine Institute. WES libraries were prepared from 3 Lig genomic DNA per individual, which was sheared by ultrasonication (Covaris S220 Ultrasonicator, Wobum, MA, USA). Exome capture was performed using the SureSelect Human All Exon V6 kit (Agilent Technologies, Santa Clara, CA, USA). The resulting libraries were sequenced on a HiSeq 2,500 HT device (Illumina, San Diego, CA, USA) according to the manufacturer’s recommendations. Paired-end (2x130) 76-bp reads were generated and mapped on the human reference genome. More than 97% of the exome was covered at least 30 times. Raw data were analysed as described54, using an in-house software system (Polyquery). The variant prioritisation strategy was as follows: (i) selection of functional (protein-altering) variants (removal of intergenic and 375’ UTR variants, non-splice related intronic variants, and synonymous variants); (ii) variants with a frequency below 1% in public databases (dbSNP, 1,000 Genomes, EVS, ExAC; release date, January 2018), and (iii) variants previously identified in fewer than five individuals contributing 11,811 in-house exomes (data not shown).
The HypothySeq NGS panel included 78 genes known to be associated with CH (TD; dyshormonogenesis; defects in thyroid hormone (TH) transport proteins; and inborn errors in TH membrane transport, metabolism, or action) and candidate genes validated in animal models (mouse and zebrafish knock-out models) or by microarray assays but not yet validated in humans. This panel was previously validated using controls including samples from positive controls with known thyroid disease-causing mutations, to assess sensitivity (false-negative rate); and from healthy individuals screened by WES for another research study, to test specificity (false-positive rate). Genomic DNA libraries were created using SureSelectXT Target Enrichment Reagent Kit (Agilent Technologies) and subjected to custom targeted DNA panel enrichment. In the 78 genes associated with CH, 1006 regions of interest were captured by the corresponding l20-bp cRNA baits, using SureDesign software (Agilent Technologies) (H. sapiens, hgl9, GRCh37, February 2,009). The 233,l03-bp targeted DNA regions (protein coding exons of the main iso form and supplementary coding exons of each gene, including 25- bp flanking intronic sequences) were sequenced on Illumina HiSeq 2,500 (Illumina). This step generated 2x130 paired-end reads. Bioinformatic analyses included alignment against the reference genome, variant calling and annotation, and copy number variation (CNV) detection. All data were integrated in the dedicated interface Polydiag developed by the bioinformatics platform at the IMAGINE Institute to check coverage of the targeted regions, to sort and filter the called variants by impact and frequency, and to identify relevant candidate mutations and/or CNVs for molecular diagnosis.
Sanger sequencing was performed to validate and segregate the identified TUBB1 (NM_030773) variants (3,500xL Genetic Analyzer, Thermo Fisher Scientific, Waltham, MA, USA). Primer sequences and PCR conditions are available on request.
Burden test Rare variant burden testing was performed for the TUBB1 gene using the CAST collapsing method55 in 270 patients with TD, including 193 with ectopia, hemi-agenesis or hypoplasia and 77 with athyreosis. Logistic regression was performed to compare the prevalence of deleterious variants in the patients with TD and in 406 Caucasian controls from the 1,000 Genomes project phase 356. Disruptive in- frame, frame-shift, missense, splice- acceptor, splice-donor, start-lost, stop-gained, or stop-lost variants were considered deleterious. All deleterious variants with a minor allele frequency <1% in the ExAC database were included in the analysis. We first studied all 270 TD patients then only the 193 TD patients without athyreosis.
Human thyroid tissue samples
After approval by our institutional review board of the experimental design and protocols, embryonic thyroid tissue was obtained from products of elective termination of pregnancy and adult thyroid tissues from patients undergoing thyroid surgery.
Animals
Tubbl / mice were previously generated by replacing exons 3 and 4, encoding amino acids 56 to 451, with a neomycin-resistance gene cassette as previously described23. Tubbl+/ mice on a mixed l29/Sv-BALB/c background were interbred with C57BL/6J mice over ten generations to generate homozygous null mutants (Tubbl· ) with the C57BL/6J background (Bb.CG-b 1 tubulin™). All experiments were conducted in accordance with French regulations and were approved by the Strasbourg regional ethics committee for animal experimentation (C.R.E.M.E.A.S., CEEA 35). Thyroids at different embryonic stages from El3.5 to E17.5 and adult thyroids at 3 months of age were obtained from wild-type and Tubbl mice and microdissected as described previously34.
Assays on mouse serum samples
Aortic blood samples were collected from 3-month-old wild-type and Tubbl mice. Radioimmunoassays were used to measure serum TSH and serum total T4 after iodothyronine extraction (Dr. S. Refetoff, Chicago, IL, USA) as previously described57.
Flow cytometry of mouse thyroid cells
Mouse thyroid tissue from El 7.5 embryos (15 pooled thyroids per sample) and adults (4 thyroids per sample) were microdissected, cleansed of fat and connective tissue, and placed in ice-cold phosphate-buffer saline (PBS) containing 2% foetal calf serum (FCS). Cells were prepared and sorted by flow cytometry as previously described28. Briefly, single-cell suspensions were obtained by enzymatic digestion with 1 mg/mL collagenase/dispase and 2 Lig/mL DNase I (Roche Diagnostics, Basel, Switzerland) at 37°C for 20 min. The cells were then centrifuged with PBS containing 2% FCS and stained with cell surface markers for 20 min. Finally, the cells were acquired on a BD FACSAria II flow cytometer (Becton Dickinson, Franklin Lakes, NJ, USA). The following monoclonal antibodies were used: EpCam/CD326 (G8.8), PDGFRa/CDl40a (APA5), CD45 (30-F11), and Pecam/CD3 l (390) from BioLegend (San Diego, CA, USA); and CD41 from Abeam (Cambridge, UK). The secondary antibody was goat anti-rabbit A647 from Life Technologies (Carlsbad, CA, USA). Each pool of sorted cells was collected in RLT buffer from the Qiagen RNeasy MicroKit (Qiagen, Valencia, CA, USA) for RNA extraction experiments.
RNA extraction and quantitative RT-PCR
The thyroids were microdissected and immediately snap-frozen and stored at -80°C. Total RNA of sorted cells or thyroid tissue was isolated using the Qiagen RNeasy Microkit or Minikit (Qiagen). The Maxima First Strand cDNA Synthesis Kit (Thermo Fisher Scientific) was used for reverse transcription of 250 ng of each RNA sample. The synthesised cDNA was diluted to 1/20, and 5 mE was used for each PCR reaction. Each reaction consisted of TaqMan Universal PCR Master Mix or SybrGreen PCR Master Mix (Thermo Fisher Scientific) and primers. Peptidylpropyl isomerase A served as an endogenous control. Real-time PCR was performed using the QuantStudio 3 Real-Time PCR System (Thermo Fisher Scientific). The data were analysed using the comparative cycle threshold method and reported as the fold change in gene expression, normalised for a calibrator of value 1.
Immunohistochemistry and quantification
Human or mouse tissues were fixed by immersion in 3.7% buffered formalin then embedded in paraffin. Subsequently, 4 pm-thick sections were mounted on StarFrost adhesive slides (Knittel Glaser, Braunschweig, Germany) and processed for immunohistochemistry, as previously described34. The primary antibodies were used at the following dilutions: rabbit antibody to human or mouse bΐ -tubulin, 1 : 1 000 (donated by Frangois Fanza), rabbit anti- Ecadherin, 1 : 100 (Becton Dickinson), mouse anti-TG, 1 : 100 (DakoCytomation, Glostrup, Denmark), rabbit anti-Nkx2-l, 1 :2500 (Biopat, Italy), mouse anti-T4, 1 : 10 000 (AbD Serotec, Raleigh, NC, USA), rabbit anti-calcitonin, 1 :400 (DakoCytomation), mouse anti-Ki67, 1 :20 (Becton Dickinson), and rabbit anti-KDEF, 1 : 1500 (Thermo Fisher Scientific). The fluorescent secondary antibodies were Alexa Fluor 594 goat anti-rabbit and Alexa Fluor 488 goat anti mouse antibodies (1/400, Thermo Fisher Scientific). The nuclei were stained using the Hoechst 33 342 fluorescent stain (0.3 mg/mL; Thermo Fisher Scientific). Photographs were taken using a fluorescence microscope (Leitz DMRB; Leica, Wetzlar, Germany) and digitised using a chilled 3CCD camera (C5810; Hamamatsu Photonics, Hamamatsu City, Japan).
The sections were then analysed using Image J l.32s (freeware, www.rsbweb.nih.gov/ij) as previously described34,58. The Nkx2-l -positive surface areas per section allowed us to draw the total thyroid surface area in pm2. The surface areas positive for calcitonin and T4, two markers of late thyroid differentiation, were normalised for total thyroid surface area. For stained surface quantification, we used one of every two sections at E9.5 and El 1.5, one of every five sections at E13.5, and five sections per adult thyroid (3 months of age). We determined the surface area to obtain an estimate of the total stained surface for each thyroid and each marker. Proliferation of Nkx2-l -positive cells at E9.5 was estimated by counting Ki67-positive nuclei among Nkx2-l -positive cells on every other section throughout the entire tissue sample at E9.5. At least three thyroids were analysed per genotype. The results are reported as mean±SEM.
For Nkx2-l staining of adult mouse thyroid glands, the first immunohistochemistry steps were as described above. After application of the primary antibody, the sections were incubated with biotinylated secondary antibody for 1 hour. Immunostaining was then performed using the Vectastain ABC kit (Vector Laboratories, Burlingame, CA, USA) according to the manufacturer’s instructions. The sections were then incubated in 3,39- diaminobenzidine tetrahydrochloride and counterstained with hemalum-eosin.
Western Blot studies of mouse thyroid tissue
Proteins prepared from mouse thyroid tissue collected in RIPA buffer and sonicated were quantified using the BCA protein assay (Thermo Fisher Scientific). Then, 20 pg of total protein was separated on BisTris polyacrylamide gel with a 4%-l2% gradient (Thermo Fisher Scientific) and transferred onto PVDF membranes (Thermo Fisher Scientific). The membranes were incubated with the primary antibodies mouse anti-Chop (1 :1,000, Cell Signalling Technology, Danvers, MA, USA) or rabbit anti-actin (Sigma- Aldrich) antibodies, followed by horseradish peroxidase-conjugated goat anti-mouse or anti-rabbit antibodies. Binding of secondary antibodies was revealed using the Amersham ECL Prime Detection Reagent Kit (GE Healthcare, Chicago, IL, USA). The protein bands on the membranes were scanned with the ImageQuant LAS 4,000 Station (GE Healthcare) then analysed using ImageJ 1 32s to determine the protein levels, with actin protein serving as an internal control.
Molecular modelling of the P160L mutated protein
The wild-type human TUBB1 sequence (accession number: Q9H4B7) was downloaded from the UniProt database, and the P160L mutation introduced into it. Both the wild-type and mutant TUBB1 sequences were modelled using Modeller 9.18 software59 with PDB 4I4T chain B as the template60. The models were analysed using PyMOL visualisation software61.
Electron microscopy
Samples were fixed for 1 hour in 3% glutaraldehyde in PBS buffer, washed, and embedded in Epon. 90 nm sections were collected on nickel grids and contrasted with uranyl acetate and lead citrate. Acquisitions were performed with a Gatan Orius 1000 CCD camera (Gatan, Pleasanton, CA, United States) on a JEOL 1011 transmission electron microscope (JEOL, Tokyo, Japan).
Plasmids, cell cultures, transfection, and immunofluorescence
We used the phumanTUBBl-tagged-Myc vector described by Kunishima et al20. Mutant P 160 L- 77/55/ was generated using a PCR-based site-directed mutagenesis method as described previously, using the Stratagene QuikchangeVR kit (Agilent Technologies)62. Nthy (Nthy-ori 3.1) immortalised human thyroid cell lines were cultured as previously described63. The Nthy cells were plated at 0.4· l05/well on poly-L-lysine-coated slides in l2-well plates 24 h before transfection then transfected with 500 ng of vectors containing wild-type or P160L mutant TUBB1 using XtremeGENE-HP-DNA, as recommended by the manufacturer (Roche Applied Science, Penzberg, Germany). After 24 h, cells were used for immunofluorescence as already described64. The cells were washed with pre-warmed PHEM buffer, fixed with 4% PFA, 0.2% glutaraldehyde, and 0.5% Triton, and permeabilised with PBS-Triton 0.1%. Immunostaining was performed with rabbit anti-Myc antibody (1 :500, Cell Signaling Technology) and mouse anti-a-tubulin (DM1 A, 1 : 1,000, Sigma- Aldrich, Saint-Louis, MI, USA) then with Alexa Fluor 647 goat anti-rabbit and Alexa Fluor 555 goat anti-mouse antibodies (1 :400, Thermo Fisher Scientific).
Human megakaryocytes and proplatelet formation
CD34+ cells were isolated from peripheral blood using an immunomagnetic technique (Miltenyi Biotec, Bergisch Gladbach, Germany). The remaining population was cultured at 37°C in 5% C02 in Iscove Modified Dulbecco’s Medium (IMDM; Thermo Fisher Scientific) supplemented with 15% BIT 9500 serum substitute (Stemcell Technologies, Vancouver, Canada), a-monothioglycerol (Sigma-Aldrich), and liposomes (phosphatidylcholine, cholesterol, and oleic acid; all from Sigma-Aldrich), in the presence of human recombinant stem cell factor (SCF, 20 ng/mL, Miltenyi Biotec) and human thrombopoeitin (50 nM, Miltenyi Biotec) added once on day 0 to the culture medium, followed by 20 nM thrombopoeitin alone on day 6 with no further SCF addition. For proplatelet formation assays, megakaryocytes were plated on a BSA-coated surface (chamber slide, Ibidi, Martinsried, Germany) on day 10. On day 13 or 14, the megakaryocytes were fixed using 4% paraformaldehyde and stained for b- tubulin.
Preparation of washed platelets
To obtain human platelets, venous blood from healthy donors or patients was collected in 10% ACD/A buffer (75 mM sodium citrate, 44 mM citric acid, 136 mM dextrose, pH 4.5). Platelets were washed as previously described65 in the presence of apyrase (100 mU/mL) and prostaglandin El (1 mM) to minimise platelet activation. Platelet counts in patients and controls were adjusted to similar levels (3 Ί08 platelets/mL) in Tyrode's buffer (137 mM NaCl, 2 mM KC1, 0.3 mM NaH2P04, 1 mM MgCh, 5.5 mM glucose, 5 mM N-2-hydroxyethylpiperazine- N’-2-ethanesulfonic acid, 12 mM NaHCCE, and 2 mM CaCl2, pH 7.3).
Platelet aggregation
Platelet aggregation was monitored by measuring light transmission through a stirred suspension of washed platelets (3 · l08/mL) at 37°C using a Chrono-Log aggregometer (Chrono- Log Corporation, Havertown, PA, USA), as previously described66. Platelet aggregation was triggered by ADP and type I collagen.
Flow cytometry of human platelets
Whole blood from healthy donors or patients was diluted in PBS to obtain a platelet concentration of 2.5 · l07/mL. Diluted whole blood was then incubated with phycocrythrin (PE)- anti-human CD62P (clone AK-4; eBioscience, Thermo Fisher Scientific) or fluorescein isothiocyanate (FITC) anti-human activated api,b^ integrin (PAC1; Becton Dickinson) for 20 minutes at room temperature. The samples were then analysed directly with an Accuri C6 flow cytometer (Becton Dickinson).
Western blotting study of human platelets
Washed platelets (300 uL; 3 - l08/mL) were lysed in Laemmli sample buffer (10 mM HEPES, 2% SDS, 10% glycerol, 5 mM EDTA). The proteins were separated by sodium dodecyl sulphate (SDS)-polyacrylamide gel electrophoresis and transferred to nitrocellulose membranes, which were incubated with the primary antibodies rabbit anti-a tubulin (Clone EP1332Y; Merck Millipore, Billerica, MA, USA) or rabbit anti-b! -tubulin antibody (donated by Frangois Lanza). Immunoreactive bands were visualised with enhanced chemiluminescence detection reagents (Perbio Science, Thermo Fisher Scientific) using a G:BOX Chemi XT 16 Image System then quantified using Gene Tools version 4.03.05.0 (Syngene, Cambridge, UK).
Statistics Results are reported as mean±SEM for the number of experiments indicated in the figure legends. Continuous data were compared using the two-tailed Student’s /-test or, for multiple comparisons, one-way A OVA followed by Dunnett’s test, as indicated in the figure legends. Statistical analyses were performed using GraphPad Prism4 (GraphPad, La Jolla, CA, USA).
Results:
Identification of TUBB1 mutations in a family (FI) with thyroid dysgenesis (TD)
Family Fl is a consanguineous family of Algerian descent. The parents are first cousins (1.1 , 1.2) with five children including two females (II.1 [patient Pl] and II.2 [patient P2]) with CH. Both patients were bom at full term and diagnosed with CH by routine neonatal screening (Fig. la), which showed thyroid stimulating hormone (TSH) elevation (164 and 177 LiIU/mL in Pl and P2, respectively). On days 13 and 11, TSH was 67 and 202 uIU/mL in Pl and P2, respectively (normal: 0.3-7 uIU/mL), and free thyroxine (T4) was 14 and 13.3 pmol in Pl and P2, respectively (normal: 9.5-25 pmol) (Data not shown). L-thyroxine therapy was initiated. I123 scintigraphy showed thyroid ectopia in both siblings. Another sibling (II.5, P3), aged 12 years, had thyroid hypoplasia (thyroid volume, 3.1 mL; normal: 7±3 mL) with a pyramidal lobe and normal thyroid function tests. The parents had normal thyroid function and two other siblings (II.3 and II.4) had normal thyroid function but were not able to undergo thyroid ultrasonography.
To look for genetic causes of CH in Pl and P2, we performed whole exome sequencing (WES) using the variant filtering and prioritisation strategy. Using the recessive transmission model, WES identified a novel missense homozygous TUBB1 mutation (c.479C>T, p.Pl60L, rs759117911) in both siblings with CH (Pl and P2) and in the sibling with thyroid hypoplasia (P3) (Data not shown). Both parents and sibling II.3 were carriers. The remaining sibling (II.4) did not carry the mutation. WES identified no variants in genes known to be associated with TD or thyroid dyshormonogenesis.
Search for TUBB1 mutations in a cohort with thyroid dysgenesis (TD) and congenital hypothyroidism (CH)
After identification of the above-described novel TUBB1 mutation, we used targeted next generation sequencing (NGS) to assess TUBB1 in a cohort of 270 patients with CH and TD. In a second family (F2) with a father (1.2) of Moroccan and a mother (1.1) of French descent, a female with CH and thyroid gland ectopia (P4, II.1) had a heterozygous TUBB1 mutation (c.3 l8C>G, r.U106C) (Fig. la). CH was diagnosed upon routine neonatal screening (TSH, 250 mΐu/mL) and confirmed on day 15 (TSH, 1,100 qfU/mL; free T4, 3.5 pmol/L; and free T3, 2.45 pmol/L). Thyroid scintigraphy showed an ectopic thyroid. The father carried the same heterozygous mutation but was not able to undergo thyroid ultrasonography. In a paternal aunt (1.3, P5), an evaluation at 26 years of age for obesity and depression showed mild hypothyroidism (TSH, 6.6 uIU/mL; normal, 0.1-5.5 mΐu/mL; free T4, 8.7 pmol/L; normal, 9.8- 23.1 pmol/L). Thyroid ultrasonography and scintigraphy showed thyroid hemi-agenesis with absence of the left lobe.
In a third family (F3), a patient (II.1, P6) with CH and an ectopic thyroid was shown by targeted NGS to have a heterozygous frameshift TUBB1 mutation (c.35delG, p.Cys 12Lcufs* 12, rs77324804) that created a premature stop codon at amino acid 23 (Fig. la). CH was diagnosed neonatally based on TSH elevation (476 mΐu/mL) and low free T4 and T3 levels (8 pmol/L and 5.6 pmol/L, respectively). L-thyroxine therapy was started at 11 days of age. Both parents were of French descent. The father (1.1, P7) carried the same heterozygous mutation and had normal thyroid function with mild thyroid lobe asymmetry by ultrasonography (right lobe, 6.9 mL; left lobe, 5 mL). The other siblings and mother had normal thyroid function and morphology and did not carry the mutation.
By targeted NGS, neither P4 nor P6 had any variants in genes known to cause CH (with TD or dyshormonogenesis).
In the Exome Aggregation Consortium (ExAC) database, estimated allele frequencies are 0.000008 for the c.479C>T mutation (rs759l 17911, 20:57598961 C/T) and 0.000025 for the c35delG (rs773248042, 20:57594611 TG/T) mutation. Neither mutation has been reported in homozygous form. The c.3 l8C>G variation has not been reported in public databases. The in silico prediction tools, Polyphen-2, SIFT, predict that c.479C>T is probably damaging and deleterious, respectively, with a CADD score of 32 (damaging: >l5)24. The other two mutations create a premature stop codon and have CADD score of 35.
A Burden test was applied to determine whether TUBB1 was significantly enriched in rare variants in the 270 patients with CH and TD versus 406 Caucasian controls from the 1,000 Genomes project. The patients had athyreosis, ectopia, hemi-agenesis, or hypoplasia. Compared to the control group, the TD group had a significantly higher proportion of individuals exhibiting at least one rare functional variant (p= 0.0227). None of the patients in our cohort with TUBB1 mutations had athyreosis. Performing the Burden test after excluding the 77 patients with athyreosis increased the significance of the difference of TD group (n=l93) versus controls 6/^=0.0095 ). The three amino acids affected by the TUBB1 mutations are strictly conserved across species, from humans to zebrafish, and across all b-tubulins (data not shown). All three mutations were located in the first part of TUBB1, i.e., in the N-terminal domain needed for guanosine triphosphate (GTP) activity (Fig. lb). The c.3 l8C>G and c.35delG mutations created a premature stop codon, thereby removing the intermediate and C-terminal domains required for microtubule-associated protein (MAP) binding.25
In sum, we identified three TUBB1 mutations in three independent families of patients with CH and TD chiefly consisting in thyroid gland ectopia.
bΐ-tubulin is expressed in the developing thyroid in humans and mice
b 1 -tubulin expression has so far been reported only in megakaryocytes and platelets26,27. Our finding of TUBB1 mutations in patients with TD prompted us to look for bΐ -tubulin expression in thyroid tissue. In human thyroid tissue, TUBB1 mRNA was expressed at 8, 10, and 12 GW and in adulthood (data not shown). In mouse thyroid tissue, Tubbl was expressed at E13.5 and strongly at E15.5, E17.5, and adulthood (data not shown). To refine our study of Tubbl expression, we used cells sorted from mice thyroid tissue based on well-accepted markers28. As expected, expression was strongest in platelets sorted using the specific megakaryocyte lineage marker CD41 (data not shown). However, Tubbl was also expressed in EpCam-positive epithelial-cell populations containing thyrocytes, at El 7.5 and adulthood (data not shown).
Similarly, in human thyroid tissue, immunohistochemistry showed bΐ -tubulin expression in the cytoplasm of thyroglobulin (TG)-producing thyrocytes at 12 GW (data not shown). Comparable findings were obtained with mouse thyroid tissue (data not shown). These data established that bΐ -tubulin is expressed in thyrocytes.
Functional in vitro analysis of disease-causing mutations
To further investigate the implication of TUBB1 gene mutations in thyroid disease, we transfected the mutations into the Nthy cell line. Only the c479C>T (p.Pl60L) mutation could be studied in this model. The c3 l8C>G (p.Yl06X) and c.35delG (p.Cys l 2Lcufs* 12) mutations created premature stop codons that yielded truncated proteins that cannot form functional a/b- tubulin dimers, and thus cannot get incorporated into microtubules25,29. The pathogenic role of these mutations is thus loss-of-fimction.
Structural modelling of p.P160L mutant bΐ-tubulin
To assess the consequences of the P160L mutation on bΐ -tubulin function, the sole missense mutation that should give rise to a full-length protein, we first compared the configurations of the mutated and wild-type proteins. The P160L mutation is located at the end of helix H4 (data not shown). In wild-type bΐ -tubulin, the proline residue stabilises loop H4- S5 and places R156 in a position that promotes a salt bridge interaction with D197 in the b- sheet S6. Furthermore, R162 in loop H4-S5 and N195 in the b-sheet S6 establish hydrogen bonds with R156. Mutation of pro line 160 to leucine might affect the loop conformation of the H4-S5 loop and thus disrupting the interaction network mediated by R156. Hence, the P160L mutation is most likely affecting the conformation of the B -tubulin, which could lead to dysfunctions of the a/b-tubulin dimer.
In vitro consequences of the p.P160L bΐ -tubulin mutation on microtubule incorporation
Following the predictions from the modelling, we examined the ability of bΐ -tubulin P160L protein to incorporate within the microtubule network in the Nthy cell line. We transfected Nthy cells with the wild-type and P160L mutant then compared the distribution and location of the protein by double-label immunofluorescence (data not shown). While the overexpressed wild-type bΐ -tubulin coassembled incorporates into microtubules, not incorporation was seen for bΐ -tubulin P160L. Thus, the P160L mutation clearly affects the capacity of bΐ -tubulin to incorporate into microtubules, which is most likely related to defects on the structural level, thus forming a dysfunctional a/B-tubulin dimer.
In summary, all three TUBB1 mutations identified here lead to nonfunctional a/B-tubulin dimers that cannot be incorporated into microtubules.
Tubbl knock-out in mice affects thyroid development and function
The phenotype of patients bearing TUBB1 mutations suggests that bΐ -tubulin may contribute to thyroid development and function. We assessed this hypothesis in Tubbl knock out mice ( TubbT ).
As expected, we found significant increases in expression levels of the other b-tubulin iso forms (Tubb2a, Tubb5, Tubb2b, and Tubb3) in E17.5 thyroids of TubbT/ mice compared to wild-type mice (data not shown). Tubb2b and Tubb3 expression levels were also increased in adult TubbT mice. In addition, Tuba3 and Tuba4 expression in TubbT mouse thyroids were diminished at El 7.5 and in adults. Interestingly, these compensatory changes in expression levels seen in the thyroid gland mirror those described in platelets of TubbT mice (data not shown)23.
Thyroid gland development and differentiation
Thyroid gland morphology in mice To study thyroid gland development from the early stages of budding and migration of the median anlage (MA) and ultimobranchial bodies (UB) to the late stages of differentiation, we used immunohistochemistry, surface quantification, and quantitative PCR (data not shown). For most of the experiments, we used Nkx2-l as a marker of progenitor and differentiated thyroid cells. At E9.5, thyroid anlage surface area and proliferation ratio were significantly greater in TubbT1 mice than in wild-type mice (data not shown. At El 1.5, thyroid migration was slightly delayed in 3 of 6 TubbT1 embryos, a few Nkx2-l -positive cells were visible along the migration tract of the mutants, and thyroid surface area was not different between mutant and wild-type embryos (data not shown). During late thyroid development, at E13.5, fusion of the median anlage and ultimobranchial bodies was slightly delayed in 3 of 4 TubbT /_ embryos (data not shown). After E13.5, Tubbl 1 thyroids were significantly hypoplastic (data not shown). Moreover, at El 7.5, a supplementary pyramidal lobe was visible near the normal lobe in 2 of 6 embryos (data not shown). In keeping with this finding, patient P3 in family 1 described above, who was homozygous for the p.Pl60L mutation, had a hypoplastic thyroid with a pyramidal lobe. In sum, the thyroid phenotype of TubbT1 mice indicates that bΐ -tubulin is required for normal thyroid migration and morphology.
Thyroid gene ontogeny in mice
We used quantitative PCR to assess the ontogeny of genes involved in thyroid function and development (data not shown). Expression levels of mRNAs for Tg, thyroid peroxidase (Tpo), TSH receptor (Tshr), and Calca), late differentiation markers, were significantly decreased at E15.5, as were those of Tg, Tpo, and Calca at E17.5, in TubbT/ compared to wild- type embryos. Significant decreases were also observed at E17.5 in the TubbT/ embryos in the early thyroid development markers Nkx2-l, Pax8, and Foxel . These results indicate impaired thyroid gland development and differentiation in Tubb /_ mice.
Endocrine signature at completion of thyroid gland development
T4-positive thyroid surface area relative to total thyroid surface area was shown by immunohistochemistry to be significantly increased at E17.5 in TubbT /_ versus wild-type embryos (data not shown). Calcitonin-positive thyroid surface area relative to total thyroid surface area was also significantly greater in the mutants. Thus, final thyroid differentiation was abnormal in TubbT /_ embryos, with increases in T4 and calcitonin that probably reflected impaired hormone secretion.
These results establish a central role for bΐ -tubulin in thyroid development and differentiation.
Thyroid gland function and structure in adult mice We compared thyroid hormone status in adult Tubbl ' and wild-type mice. At 3 months of age, serum TSH levels were higher and T4 levels lower in Tubbl 1 than in wild-type mice, suggesting hypothyroidism in the mutants data not shown). These results are consistent with the finding of CH in our patients carrying TUBB1 mutations.
In sum, the increase in T4-positive surface area combined with low serum T4 suggest trapping of T4 hormone within the thyrocyte cytoplasm.
Finally, we examined thyroid structure in adult mice (3 months of age). Surface area was not different between the Tubbl 1 and wild-type thyroids (data not shown). However, the thyroid tissue was disorganised in the mutants with large regions without organized follicles (data not shown). When we used electron microscopy to examine thyrocyte ultrastructure (data not shown), we found larger numbers of dense and rod-shaped vesicles in Tubbl 1 versus wild-type thyrocytes. These data suggest that the T4 retention in El 7.5 thyroids of Tubbl 1 mice was related to impaired T4 release responsible for hypothyroidism. Marked endoplasmic reticulum (ER) dilation was seen in Tubbl 1 compared to wild-type thyrocytes (data not shown), indicating ER stress, which was confirmed by the findings of increased Chop and XBPs expression by quantitative PCR and of increased Chop protein levels by western blotting in Tubbl 1 thyroids (data not shown). By immunohistochemistry, the ER marker KDEL co- localized with TG in Tubbl 1 and wild-type mice but was especially abundant in disorganized areas of adult Tubbl 1 thyroids (data not shown).
These data indicated thyroid tissue disorganization, vesicle accumulation, and ER stress in TubbT/ mice.
In sum, our findings in Tubbl-/- mice demonstrate a complex mechanism of hypothyroidism involving early abnormal proliferation of progenitors, delayed thyroid migration, defective thyroid tissue differentiation, and impaired thyroid hormone release. The abnormal thyroid migration may explain the thyroid phenotype found in patients carrying TUBB1 mutations, especially those with thyroid gland ectopia. Additional contributors to hypothyroidism are structural disorganization of the thyroid tissue and retention of thyroid hormone.
Analysis of human platelets of patients bearing TUBB1 mutations
Until now, bΐ -tubulin expression had been reported only in the megakaryocyte lineage19. TUBB1 mutations have been previously reported to cause macrothrombocytopenia20- 22. We therefore studied the platelets of the above-described patients with TUBB1 mutations and thyroid gland abnormalities.
Haematological parameters Thrombocytopenia was a feature in patients with TUBB1 mutations studied by other groups20-22. However, when we used an automated haematology analyser to measure the platelet count and mean platelet volume (MPV) in our seven patients, we consistently found normal platelet counts. In contrast, MPV was above the normal range in Pl and near the upper limit of normal in P4 and P7. A blood smear analysis (data not shown) demonstrated variations in platelet size, with the presence of macroplatelets. Moreover, parents 1.1 and 1.2 of the family Fl had MPV values at the top of the normal range (10.2 fL and 1 1.3 fL, respectively; normal, 7.5-11.2 fL) with many macroplatelets. Electron microscopy confirmed the large platelet size (data not shown).
The increased platelet size could be related to defects in their biogenesis. To study this we cultured megakaryocytes from patients Pl and P4 from peripheral blood progeniors and analysed proplatelet formation (data not shown). Interestingly, shaft thickness and coiled element diameter of the future platelets were significantly increased compared to controls, indicating that the TUBB1 mutations affected proplatelet formation and platelet size.
Functional analysis of human platelets
To investigate whether the TUBB1 mutations affected platelet function, we first quantified bΐ -tubulin expression in the platelets of our patients and found significant decreases ( O.OOl), of45.0±3.7% and 36.5±3.4% in Pl and P3 (Fl), respectively; 45.0±3.8% in P4 (F2); and 29. l±3.3% and 30.6±6.2% in P6 and P7 (F3), respectively. Expression of a-tubulin was normal (data not shown). The bΐ -tubulin antibody used in our study detects only the C- terminal part of the protein. Consequently, in the patients of families F2 and F3, whose mutations created premature stop codons, only the wild-type protein was detected. In the Fl patients, who were homozygous for the mutation, the results suggested either diminished protein expression or protein instability.
A discoid shape is a normal feature of circulating or non-activated platelets. By electron microscopy, discoid platelets were less numerous in Pl and P3 (Fl) (data not shown), consistent with either abnormal platelet sphericity or platelet activation. We therefore assessed basal platelet activation, using flow cytometry to investigate the platelet activation markers P- selectin exposure and integrin api,b ^ activation in whole blood. Both markers were significantly increased in Pl and P3, indicating abnormal platelet activation. In P6 and P7 (family 3), in contrast, the marker levels were comparable to those in controls (data not shown). We were unable to evaluate P4 (F2). Finally, we investigated platelet aggregation upon activation by ADP or collagen, two key platelet agonists (data not shown). Aggregation in response to ADP was normal in P6 and P7 (F3) but increased in Pl and P3 (Fl) and in P4 (F2) compared to controls (data not shown). Similarly, with collagen, platelet aggregation was normal in P6 and P7 but was increased in Pl and P4, even with low doses. These abnormal platelet aggregations were not related to hypothyroidism or patient’s treatment by L-Thyroxine, since they were not found in three patients without TUBB1 mutations but under the same treatment (data not shown). Moreover, it should be noted that these results were confirmed by at least three independent investigations for Pl and P3 and two for P6. The other patients were studied once. None of the 9 controls exhibited similar hyperaggregation profil,
In sum, these results indicate that p.Pl60L (Fl), r.UIObC (F2), and c.35delG (F3) TUBB1 mutations affect bΐ -tubulin expression in platelets and result in abnormally large platelet size, probably as a consequence of proplatelet abnormal formation. Moreover, the p.Pl60L mutation induces significant basal platelet activation and hyper aggregation in response to agonists, whereas the c.35delG mutation does not seem to affect platelet function.
Discussion:
We identified three TUBB1 mutations in patients with TD and macroplatelets. In a consanguineous family (Fl), two females with CH and thyroid gland ectopia had the same homozygous TUBB1 mutation, and their brother had thyroid gland hypoplasia with normal function. Of 270 patients with CH and TD, two probands from unrelated families had heterozygous TUBB1 mutations. All patients with heterozygous or homozygous TUBB1 mutations had TD and macroplatelets. Thyroid gland ectopia was the most common form of TD, but thyroid asymmetry, thyroid hypoplasia, and thyroid hemi-agenesis were seen also. All three TUBB1 mutations impaired bΐ -tubulin protein function.
This is the first time that CH with TD was associated with macroplatelets and we demonstrate in our study that TUBB1 mutations are the common cause bΐ -tubulin expression has heretofore been described as confined to megakaryocytes and platelets. Our findings demonstrate that bΐ -tubulin is also expressed in developing and adult thyroid tissue. Furthermore, CH and TD were found in humans with TUBB1 mutations and in Tubbl mice. These data indicate that normal thyroid development and function require bΐ -tubulin incorporation into the microtubule network. Furthermore, our patients with TUBB1 mutations had normal platelet counts contrasting with abnormal platelet morphology, and two of the three mutations (p.Pl60L and r.U106C) were associated with abnormal platelet function. In contrast, in previous studies, humans with TUBB1 mutations and Tubbl mice had macrothrombocytopenia but no reported thyroid disorders20-22.
The previously reported TUBB1 mutations affect the intermediate or C-terminal domain of bΐ -tubulin, whereas the three novel mutations described here modify the N-terminal domain. Strikingly, all three mutations are loss-of- function mutations: two of the mutations lead to truncated bΐ -tubulin, which cannot form functional a/B-tubulin dimers and thus cannot be incorporated into microtubules. The third is a missense mutation (P160L), which, despite expressing full-length B-tubulin, still does not allow the formation of functional a/B-tubulin dimers to be integrated into the microtubules network. Hence, the three TUBB1 mutations were deleterious for the bΐ -tubulin function, which is confirmed on the functional level as their phenotypes are similar to those of the Tubbl-/- mouse. Consequently, Tubbl 2 mice emerged as a useful tool for studying the impact of impaired bΐ -tubulin function on the thyroid gland.
Our experiments in Tubbl 2 mice provided the first evidence of a role for bΐ -tubulin in TD and hypothyroidism. The thyroid glands of Tubbl 2 mice exhibited deficient proliferation at E9.5, abnormal migration at El 1.5 and E13.5, and hormone secretion failure at E17.5 and adulthood. All these mechanisms require normal microtubule organisation and function. The idea that microtubule functions are fine-tuned, and thus adapted to specific cellular role by the expression of specific tubulin isotypes is an emerging concept known as the‘tubulin code’17. Tubulin mutations have been more and more linked to different human disorders, in particular to neurological disorders30. Microtubules are essential for mitosis to unfold normally31. Here we show that invalidation of B-tubulin isotype Tubbl perturbs mitosis of progenitors at E9.5, thus inducing hyperproliferation. Mitotic phenotypes have already described during neuronal development in mice with loss of another B-tubulin isotype, TiibbS'2. Moreover, normal thyroid development requires that progenitor proliferation occur during a specific time window33. Early proliferation at E9.5, as described by our group in HesT1 mice34, results in abnormal thyroid development, probably via impairment of the pool of progenitors dedicated to thyroid development. Furthermore, microtubules are required for cell orientation during migration35. Our data in the Tubbl-/- mice show a delay of thyroid progenitors cells migration during thyroid development, which again is mirrored by Tubb5-/- that show impaired neuronal migration32. In our patients with TUBB1 mutations, thyroid gland ectopia was the most common form of TD (4/7 patients), further supporting a role for bΐ -tubulin in thyroid gland migration. Finally, endosome/lysosome trafficking is taking place on microtubules36-38. Endosome-to-lysosome transport of thyroid hormones via a vesicular transport system has been described in the thyroid gland39,40. That thyroid microtubule integrity is required for thyroid hormone secretion was established by studies done in the l970s and l980s41,42. While so far nothing is known on the precise role of tubulin isotypes and microtubules organization in trafficking of thyroid hormone vesicles, there is strong evidence that transport along microtubules is affected by the tubulin code43. Our data are the first to demonstrate that a specific tubulin isotype, b 1 -tubulin, is required for proper vesicle trafficking and thyroid hormone release into the bloodstream, and they extend our knowledge about the association between microtubules and secretion vesicles in the thyroid gland. We found that thyroid hormone retention suggested in ER stress in the thyroid gland. Previous studies demonstrated ER stress with activation of the unfolded protein response in association with reduced TG synthesis and TG accumulation within the ER44,45. bΐ -tubulin is the most divergent isotype of tubulin, and its incorporation into microtubule is expected to change the properties of these filaments. Losing bΐ -tubulin thus certainly alters microtubule properties, and our data provide strong evidence that these particular properties are essential for adequate thyroid development and function.
Our patients with TUBB1 mutations had macroplatelets but no thrombocytopenia, in contrast to earlier reports20-22. The abnormal platelet size is further evidence that TUBB1 mutations adversely affect platelet morphology probably as a consequence of proplatelets coiled element diameter increase, as already suggested. In these previous described mutations, only the p.R3 l8W mutation has been investigated in platelet aggregations and it did not induce defect of platelet function20. For the other mutations (p.F260S and p.Q423X)21,22, no bleeding tendency have been reported. An unexpected finding in our patients with p.Pl60L and p.Yl06X mutations was basal activation and exaggerated aggregation of platelets, which had not been described previously in patients with TUBB1 mutations or in Tubbl mice. Whether these platelet function abnormalities result in clinical symptoms such as thrombosis requires evaluation, particularly in older patients who may be at increased risk. Platelets play an important pathophysiological role in thrombosis and inflammation in atherosclerosis. Large platelets have greater haemostatic and enzymatic potency and contain larger amounts of prothrombotic molecules chemokines and growth factors affecting both pathophysiological processes46 and increased MPV was previously reported to be associated to cardiovascular disease47. Thyroid dysfunction can affect the haemostatic balance48,49, with clinical effects that vary across thyroid disorders50. Moreover, MPV correlated positively with the TSH level51. A TUBB1 -mutation screening study in patients with hypothyroidism and altered MPV and/or a history of thrombotic disease would be of interest. The clinical symptoms to accompany platelet hyperaggregability include unexplained arterial thrombosis, complications during pregnancy, and less frequently, venous thromboembolism. Patients bearing a TUBB1 mutation with cardiovascular risk factors could be monitored and the question arises whether antiplatelet drugs might be effective as treatment for and/or prophylaxis52.
The c.35delG mutation in P6 and P7 did not cause hyperaggregation. The patients with the c.35delG mutation had the same thyroid phenotype as other families with other mutations. At least one patient per family had thyroid gland ectopia with CH. Neither thyroid nor platelet phenotype severity correlated with the type of mutation. Further investigations are needed to elucidate the difference between the effects of c.35delG and pl60L/pYl06X on platelet function.
TUBB1 mutations constitute a model of dominant inheritance of CH with TD. Most known mutations responsible for TD and previously described TUBB1 mutations causing macrothrombocytopenia are also dominant10,13,20 22,53. Our data indicate high penetrance for platelet alterations and variable expressivity for TD, ranging from TD without CH to TD with CH and leading to variable types of TD (ectopia, hypoplasia, hemi-agenesis, or asymmetric thyroid gland). No patient had athyreosis. TUBB1 mutations were found in only 1.1% of our cohort with CH and TD patients. The Burden test showed enrichment in rare TUBB1 variants carriers in the cohort versus controls. Our data increment the number of causative genes for thyroid dysgenesis but with a novel phenotype associating platelets disorder.
Taken together, our data indicate heretofore unsuspected roles for a specific isotype of b-tubulin, Tubbl, in thyroid development and function, while also confirming its importance for microtubule integrity and platelet function. Loss-of-fimction of TUBB1 mutations impair bΐ -tubulin incorporation into microtubules. Our results confirm that normal thyroid-cell proliferation and thyroid migration are essential to thyroid gland development. Thyroid hormone secretion requires bΐ -tubulin incorporation into the microtubules, suggesting a specific function of this tubulin isotype in intracellular transport of vesicles. Thus, our work provides novel insights into the role of the Tubbl isotype in thyroid physiopathology and in platelet function and therefore expands the spectrum of the rare pediatric diseases related to tubulin mutations and microtubule malfunction.
REFERENCES:
Throughout this application, various references describe the state of the art to which this invention pertains. The disclosures of these references are hereby incorporated by reference into the present disclosure. 1. Barry Y, Bonaldi C, Goulet V, Coutant R, Leger J, Paty AC, Delmas D, Cheillan D, Roussey M. Increased incidence of congenital hypothyroidism in France from 1982 to 2012: a nationwide multicenter analysis. Ann Epidemiol 26, 26, 100-5 (2016).
2. Deladoey J, Ruel J, Giguere Y, Van Vliet G. Is the incidence of congenital hypothyroidism really increasing? A 20-year retrospective population-based study in Quebec. J Clin Endocrinol Metab 96, (2011).
3. Stoupa A, Kariyawasam D, Carre A, Polak M. Update of Thyroid Developmental Genes. Endocrinol Metab Clin North Am 45, 243-54 (2016).
4. Trueba SS, Auge J, Mattel G, Etchevers H, Martinovic J, Czemichow P, Vekemans M, Polak M, Attie-Bitach T. PAX8, TITF1, and FOXE1 gene expression patterns during human development: new insights into human thyroid development and thyroid dysgenesis-associated malformations. J Clin Endocrinol Metab 90, 455-62 (2005).
5. Nilsson M, Fagman H. Mechanisms of thyroid development and dysgenesis: an analysis based on developmental stages and concurrent embryonic anatomy. Curr Top Dev Biol. 106, 123-170 (2013).
6. Szinnai G, Lacroix L, Carre A, Guimiot F, Talbot M, Martinovic J, Delezoide AL, Vekemans M, Michiels S, Caillou B, Schlumberger M, Bidart JM, Polak M. Sodium/iodide symporter (NIS) gene expression is the limiting step for the onset of thyroid function in the human fetus. J Clin Endocrinol Metab 92, 70-6 (2007).
7. Maiorana R, Carta A, Floriddia G, Leonardi D, Buscema M, Sava L, Calaciura F, Vigneri R. Thyroid hemiagenesis: prevalence in normal children and effect on thyroid function. J Clin Endocrinol Metab 88, 1534-6 (2003).
8. Sura-Trueba S, Aumas C, Carre A, Durif S, Leger J, Polak M, de Roux N. An inactivating mutation within the first extracellular loop of the thyrotropin receptor impedes normal posttranslational maturation of the extracellular domain. Endocrinology. 150, 1043-50 (2009).
9. Carre A, Hamza RT, Kariyawasam D, Guillot L, Teissier R, Tron E, Castanet M, Dupuy C, El Kholy M, Polak M. A novel FOXE1 mutation (R73S) in Bamforth-Lazarus syndrome causing increased thyroidal gene expression. Thyroid. 24, 24, 649-54 (2014).
10. Carre A, Stoupa A, Kariyawasam D, Gueriouz M, Ramond C, Monus T, Leger J, Gaujoux S, Sebag F, Glaser N, Zenaty D, Nitschke P, Bole-Feysot C, Hubert L, Lyonnet S, Scharfmann R, Munnich A, Besmond C, Taylor W, Polak M. Mutations in BOREALIN cause thyroid dysgenesis. Horn Mol Genet 26, 599-610 (2017). 11. Carre A, Szinnai G, Castanet M, Sura-Trueba S, Tron E, Broutin-L’Hermite I, Barat P, Goizet C, Lacombe D, Moutard ML, Raybaud C, Raynaud-Ravni C, Romana S, Ythier H, Leger J, Polak M. Five new TTF1/NKX2.1 mutations in brain-lung-thyroid syndrome: rescue by PAX8 synergism in one case. Hum Mol Genet 18, 18, 2266-76 (2009).
12. Senee V, Chelala C, Duchatelet S, Feng D, Blanc H, Cossec JC, Charon C, Nicolino M, Boileau P, Cavener DR, Bougneres P, Taha D, Julier C. Mutations in GLIS3 are responsible for a rare syndrome with neonatal diabetes mellitus and congenital hypothyroidism. Nat Genet 38, 682-687 (2006).
13. Ramos HE, Carre A, Chevrier L, Szinnai G, Tron E, Cerqueira TL, Leger J, Cabrol S, Puel O, Queinnec C, De Roux N, Guillot L, Castanet M, Polak M. Extreme phenotypic variability of thyroid dysgenesis in six new cases of congenital hypothyroidism due to PAX8 gene loss-of-fimction mutations. Eur J Endocrinol 171, 499-507 (2014).
14. Opitz R, Hitz MP, Vandemoot I, Trubiroha A, Abu-Khudir R, Samuels M, Desilets V, Costagliola S, Andelfmger G, Deladoey J. Functional zebrafish studies based on human genotyping point to netrin-l as a link between aberrant cardiovascular development and thyroid dysgenesis. Endocrinology 156, 377-88 (2015).
15. Hildebrandt F, Heeringa SF, Ruschendorf F, Attanasio M,Numberg G, Becker C, Seelow D, Huebner N, Chemin G, Vlangos CN, Zhou W, O’Toole JF, Hoskins BE, Wolf MT, Hinkes BG, Chaib H, Ashraf S, Schoeb DS, Ovunc B, Allen SJ, Vega-Warner V, Wise E, Harville HM, Lyons RH, Washburn J, Macdonald J, Numberg P, Otto EA. A systematic approach to mapping recessive disease genes in individuals from outbred populations. PLoS Genet 5, 5 (2009).
16. Choi M, Scholl UI, Ji W, Liu T, Tikhonova IR, Zumbo P, Nayir A, Bakkaloglu A, Ozen S, Sanjad S, Nelson-Williams C, Farhi A, Mane S, Litton RP. Genetic diagnosis by whole exome capture and massively parallel DNA sequencing. Proc Natl Acad Sci U A 106, 19096-101 (2009).
17. Janke C. The tubulin code: molecular components, readout mechanisms, and functions. J Cell Biol 206, 461-72 (2014).
18. Gadadhar S, Bodakuntla S, Natarajan K, Janke C. The tubulin code at a glance. J Cell Sci 130, 1347-1353 (2017).
19. Patel SR, Richardson JL, Schulze H, Kahle E, Galjart N, Drabek K, Shivdasani RA, Hartwig JH, Italiano JE Jr. Differential roles of microtubule assembly and sliding in proplatelet formation by megakaryocytes. Blood 106, 4076-85 (2015). 20. Kunishima S,Kobayashi R, Itoh TJ, et a. Mutation of the beta- 1 -tubulin gene associated with congenital macrothrombocytopenia affecting microtubule assembly. Blood 113, 458-461 (2009).
21. Kunishima S, Nishimura S, Suzuki H, Imaizumi M, Saito H. TUBB1 mutation disrupting microtubule assembly impairs proplatelet formation and results in congenital macrothrombocytopenia. Eur J Haematol 92, 276-82 (2014).
22. Fiore M, Goulas C, Pillois X. A new mutation in TUBB1 associated with thrombocytopenia confirms that C-terminal part of bΐ -tubulin plays a role in microtubule assembly. Clin Genet 91, 924-926 (2017).
23. Schwer HD, Lecine P, Tiwari S, Italiano JE Jr, Hartwig JH, Shivdasani RA. A lineage-restricted and divergent beta-tubulin isoform is essential for the biogenesis, structure and function of blood platelets. Curr Biol 11, 579-86 (2001).
24. Kircher M, Witten DM, Jain P, O’Roak BJ, Cooper GM, Shendure J. A general framework for estimating the relative pathogenicity of human genetic variants. Nat Genet 46, 310-5 (2014).
25. Nogales E, Wolf SG, Downing KH. Structure of the alpha beta tubulin dimer by electron crystallography. Nature 391, 199-203 (1998).
26. Wang D, Villasante A, Lewis SA, Cowan NJ. The mammalian beta-tubulin repertoire: hematopoietic expression of a novel, heterologous beta-tubulin isotype. J Cell Biol 103, 1903-10 (1986).
27. Lecine P, Italiano JE Jr, Kim SW, Villeval JL, Shivdasani RA. Hematopoietic- specific beta 1 tubulin participates in a pathway of platelet biogenesis dependent on the transcription factor NF-E2. Blood 96, 1366-73 (2000).
28. Gawade S, Mayer C, Hafen K, Barthlott T, Krenger W, Szinnai G. Cell Growth Dynamics in Embryonic and Adult Mouse Thyroid Revealed by a Novel Approach to Detect Thyroid Gland Subpopulations. Thyroid 26, 591-9 (2016).
29. Joe PA, Banerjee A, Luduena RF. Roles of beta-tubulin residues Ala428 and Thr429 in microtubule formation in vivo. J Biol Chem 284, 4283-91 (2009).
30. Chakraborti S, Natarajan K, Curiel J, Janke C, Liu J. The emerging role of the tubulin code: From the tubulin molecule to neuronal function and disease. Cytoskelet. Hoboken 73, 521-550 (2016).
31. Prosser SL, Pelletier L. Mitotic spindle assembly in animal cells: a fine balancing act. Nat Rev Mol Cell Biol 18, 187-201 (2017). 32. Breuss M, Heng JI, Poirier K, Tian G, Jaglin XH, Qu Z, Braun A, Gstrein T, Ngo L, Haas M, Bahi-Buisson N, Moutard ML, Passemard S, Verloes A, Gressens P, Xie Y, Robson KJ, Rani DS, Thangaraj K, Clausen T, Chelly J, Cowan NJ, Keays DA. Mutations in the b-tubulin gene TUBB5 cause microcephaly with structural brain abnormalities. Cell Rep 2, 1554-62 (2012).
33. Nilsson M, Fagman H. Development of the thyroid gland. Development 144, 2123-2140 (2017).
34. Carre A, Rachdi L, Tron E, Richard B, Castanet M, Schlumberger M, Bidart JM, Szinnai G, Polak M. Hesl is required for appropriate morphogenesis and differentiation during mouse thyroid gland development. PLoS One 6, 6 (2011).
35. Ladoux B, Mege RM, Trepat X. Front-Rear Polarization by Mechanical Cues: From Single Cells to Tissues. Trends Cell Biol 26, 420-433 (2016).
36. Bonifacino JS, Neefjes J. Moving and positioning the endolysosomal system. Curr Opin Cell Biol 47, 1-8 (2017).
37. Raiborg C, Wenzel EM, Stenmark H. ER-endosome contact sites: molecular compositions and functions. EMBO J 34, 1848-58 (2015).
38. Huotari J, Helenius A. Endosome maturation. EMBO J 30, 3481-500 (2011).
39. Rousset B, Dupuy C, Miot F, Dumont J. Chapter 2 Thyroid Hormone Synthesis And Secretion in Endotext (2015).
40. Carvalho DP, Dupuy C. Thyroid hormone biosynthesis and release. Mol Cell Endocrinol 458, 6-15 (2017).
41. Neve P, Ketelbant-Balasse P, Willems C, Dumont JE. Effect of inhibitors of microtubules and micro filaments on dog thyroid slices in vitro. Exp Cell Res 74, 227-44
(1972).
42. Wolff J, Bhattacharyya B. Microtubules and thyroid hormone mobilization. Ann N Acad Sci 253, 763-70 (1975).
43. Nirschl JJ, Magiera MM, Lazarus JE, Janke C, Holzbaur EL. a-Tubulin
Tyrosination and CLIP- 170 Phosphorylation Regulate the Initiation of Dynein-Driven
Transport in Neurons. Cell Rep 14, 2637-52 (2016).
44. Di Jeso B, Arvan P. Thyroglobulin From Molecular and Cellular Biology to Clinical Endocrinology. Endocr Rev 37, 2-36 (2016).
45. Gaide Chevronnay HP, Janssens V, Van Der Smissen P, Liao XH, Abid Y, Nevo N, Antignac C, Refetoff S, Cherqui S, Pierreux CE, Courtoy PJ. A mouse model suggests two mechanisms for thyroid alterations in infantile cystinosis: decreased thyroglobulin synthesis due to endoplasmic reticulum stress/unfolded protein response and impaired lysosomal processing. Endocrinology. 156, 2349-64 (2015).
46. Gasparyan AY, Ayvazyan L, Mikhailidis DP, Kitas GD. Mean platelet volume: a link between thrombosis and in- flammation? Curr Pharm Des 17, 47-58 (2011).
47. Chu SG, Becker RC, Berger PB, Bhatt DL, Eikelboom JW, Konkle B, Mohler ER, Reilly MP, Berger JS. Mean platelet volume as a predictor of cardiovascular risk: a systematic review and meta-analysis. J Thromb Haemost 8, 148-56 (2010).
48. Squizzato A, Romualdi E, Buller HR, Gerdes VE. Clinical Review: Thyroid Dysfunction and Effects on Coagulation and Fibrinolysis: A Systematic Review. Squizzato Romualdi E Buller HR Gerdes VE 92, 2415-20 (2007).
49. Kyriakakis N, Lynch J, Ajjan R, Murray RD. The effects of pituitary and thyroid disorders on haemostasis: potential clinical implications. Clin Endocrinol Oxf 84, 473-84 (2016).
50. Franchini M, Lippi G, Manzato F, Vescovi PP, Targher G. Hemostatic abnormalities in endocrine and metabolic disorders. Eur J Endocrinol 162, 439-51
51. Kim JH, Park JH, Kim SY, Bae HY. The mean platelet volume is positively correlated with serum thyrotropin concentrations in a population of healthy subjects and subjects with unsuspected subclinical hypothyroidism. Thyroid 23, 31-7 (2013).
52. Kuhli-Hattenbach C, Hellstem P, Kohnen T, Hattenbach LO. Platelet activation by ADP is increased in selected patients with anterior ischemic optic neuropathy or retinal vein occlusion. Platelets 28, (2017).
53. Guillot L, Carre A, Szinnai G, Castanet M, Tron E, Jaubert F, Broutin I, Counil F, Feldmann D, Clement A, Polak M, Epaud R. NKX2-1 mutations leading to surfactant protein promoter dysregulation cause interstitial lung disease in‘Brain- Lung-Thyroid Syndrome’. Hum Mutat 31, El 146-62 (2010).
54. Gordon CT, Petit F, Kroisel PM, Jakobsen L, Zechi-Ceide RM, Oufadem M, Bole-Feysot C, Pruvost S, Masson C, Tores F, Hieu T, Nitschke P, Lindholm P, Pellerin P, Guion-Almeida ML, Kokitsu-Nakata NM, Vendramini-Pittoli S, Munnich A, Lyonnet S, Holder-Espinasse M, Amiel J. Mutations in endothelin 1 cause recessive auriculocondylar syndrome and dominant isolated question-mark ears. Am J Hum Genet 93, 1118-1125 (2013).
55. Morgenthaler S, Thilly WG. A strategy to discover genes that carry multi-allelic or mono-allelic risk for common diseases: a cohort allelic sums test (CAST). Mutat Res 615, 28-56 (2007). 56. The 1000 Genomes Project Consortium. A global reference for human genetic variation. Nature 526, 68-74 (2015).
57. Pohlenz J, Maqueem A, Cua K, Weiss RE, Van Sande J, Refetoff S. Improved radioimmunoassay for measurement of mouse thyrotro- pin in serum: strain differences in thyrotropin concentration and thyrotroph sensitivity to thyroid hormone. Thyroid J Am Thyroid Assoc 9, 1265-1271 (1999).
58. Kariyawasam D, Rachdi L, Carre A, Martin M, Houlier M, Janel N, Delabar JM, Scharfmann R, Polak M. DYRK1 A BAC transgenic mouse: a new model of thyroid dysgenesis in Down syndrome. Endocrinology. 156, 171-80 (2015).
59. Sali, A., & Blundell, T. L. Comparative protein modelling by satisfaction of spatial restraints. J. Mol. Biol. 234, 779-815 (1993).
60. Prota, A. E., Bargsten, K., Zurwerra, D., Field, J. J., Diaz, J. F., Altmann, K. H., & Steinmetz, M. O. Molecular mechanism of action of microtubule-stabilizing anticancer agents. Science 339, 587-590 (2013).
61. DeLano, W.L. The PyMOL Molecular Graphics System. (2002).
62. Carre A, Castanet M, Sura-Trueba S, Szinnai G, Van Vliet G, Trochet D, Amiel J, Leger J, Czemichow P, Scotet V, Polak M. Polymorphic length of FOXE1 alanine stretch: evidence for genetic susceptibility to thyroid dysgenesis. Hum Genet. 467-76 (2007).
63. Lemoine NR, Mayall ES, Jones T, Sheer D, McDermid S, Kendall-Taylor P, Wynford-Thomas D. Characterisation of human thyroid epithelial cells immortalised in vitro by simian virus 40 DNA transfection. Br J Cancer 60, 897-903 (1989).
64. Bourg N, Mayet C, Dupuis G, Barroca T, Bon P, Lecart S, Fort E, Leveque-Fort S. Direct optical nano scopy with axially localized detection. Nat. Photonics 9, 587-93
65. Adam F, Verbeuren TJ, Fauchere JL, Guillin MC, Jandrot-Perrus M. Thrombin- induced platelet PAR4 activation: role of glycoprotein lb and ADP. J Thromb Haemost 1, 798- 804 (2003).
66. Adam F, Kauskot A, Nurden P, Sulpice E, Hoylaerts MF, Davis RJ, Rosa JP, Bryckaert M. Platelet JNK1 is involved in secretion and thrombus formation. Blood 115, 4083- 92 (2010).

Claims

CLAIMS:
1. A method of diagnosing thyroid dysgenesis in a subject, comprising detecting a mutation in the TUBB1 gene in a sample obtained from said subject, wherein detecting the presence of a mutation in the TUBB 1 gene is considered to be indicative of thyroid dysgenesis.
2. The method of claim 1 wherein the subject is a new-born or a foetus.
3. The method of claim 1 wherein the sample is a blood sample.
4. The method of claim 1 wherein the mutation is located in the N-terminal domain needed for guanosine triphosphate (GTP) activity, i.e. the domain ranging from the amino acid residue at position 1 to the amino acid residue at position 206 in the protein (SEQ ID NO:2).
5. The method of claim 1 wherein the mutation leads to nonfunctional a/B-tubulin dimers that cannot be incorporated into microtubules.
6. The method of claim 1 wherein the mutation leads to an early abnormal proliferation of progenitors, delayed thyroid migration, defective thyroid tissue differentiation, and impaired thyroid hormone release.
7. The method of claim 1 wherein the mutation affects bΐ 1 -tubulin expression in platelets and result in abnormally large platelet size, probably as a consequence of proplatelet abnormal formation.
8. The method of claim 1 wherein the mutation is a missense homozygous TUBB1 mutation (c.479C>T, p.Pl60L, rs759l 17911) which is a substitution of a cytosine residue into a thymine residue at position 479 of the TUBB1 gene.
9. The method of claim 1 wherein the mutation is a heterozygous TUBB1 mutation (c.3 l8C>G, p.Yl06X) which is the substitution of a cytosine residue into a guanine residue at position 318 of the TUBB1 gene
10. The method of claim 1 wherein the mutation is a heterozygous frameshift TUBB1 mutation (c.35delG, p.Cysl 2Lcufs* 12, rs77324804) that created a premature stop codon at amino acid 23.
PCT/EP2019/063933 2018-05-30 2019-05-29 Detection of tubb1 mutations for diagnosing thyroid dysgenesis WO2019229113A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP18305656 2018-05-30
EP18305656.3 2018-05-30

Publications (1)

Publication Number Publication Date
WO2019229113A1 true WO2019229113A1 (en) 2019-12-05

Family

ID=62631037

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2019/063933 WO2019229113A1 (en) 2018-05-30 2019-05-29 Detection of tubb1 mutations for diagnosing thyroid dysgenesis

Country Status (1)

Country Link
WO (1) WO2019229113A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0994963A1 (en) 1997-03-04 2000-04-26 Isis Innovation Limited Non-invasive prenatal diagnosis
WO2018002147A1 (en) * 2016-06-30 2018-01-04 INSERM (Institut National de la Santé et de la Recherche Médicale) Detection of borealin mutations for diagnosing thyroid dysgenesis

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0994963A1 (en) 1997-03-04 2000-04-26 Isis Innovation Limited Non-invasive prenatal diagnosis
WO2018002147A1 (en) * 2016-06-30 2018-01-04 INSERM (Institut National de la Santé et de la Recherche Médicale) Detection of borealin mutations for diagnosing thyroid dysgenesis

Non-Patent Citations (75)

* Cited by examiner, † Cited by third party
Title
"The 1000 Genomes Project Consortium. A global reference for human genetic variation", NATURE, vol. 526, 2015, pages 68 - 74
ADAM FKAUSKOT ANURDEN PSULPICE EHOYLAERTS MFDAVIS RJROSA JPBRYCKAERT M: "Platelet JNK1 is involved in secretion and thrombus formation", BLOOD, vol. 115, 2010, pages 4083 - 92
ADAM FVERBEUREN TJFAUCHERE JLGUILLIN MCJANDROT-PERRUS M: "Thrombin-induced platelet PAR4 activation: role of glycoprotein Ib and ADP", J THROMB HAEMOST, vol. 1, 2003, pages 798 - 804
BARRY YBONALDI CGOULET VCOUTANT RLEGER JPATY ACDELMAS DCHEILLAN DROUSSEY M: "Increased incidence of congenital hypothyroidism in France from 1982 to 2012: a nationwide multicenter analysis", ANN EPIDEMIOL, vol. 26, no. 26, 2016, pages 100 - 5
BONIFACINO JSNEEFJES J: "Moving and positioning the endolysosomal system", CURR OPIN CELL BIOL, vol. 47, 2017, pages 1 - 8
BOURG NMAYET CDUPUIS GBARROCA TBON PLECART SFORT E: "Leveque-Fort S. Direct optical nanoscopy with axially localized detection", NAT. PHOTONICS, vol. 9, pages 587 - 93
BREUSS MHENG JIPOIRIER KTIAN GJAGLIN XHQU ZBRAUN AGSTREIN TNGO LHAAS M: "Mutations in the 0-tubulin gene TUBB5 cause microcephaly with structural brain abnormalities", CELL REP, vol. 2, 2012, pages 1554 - 62
CARRE ACASTANET MSURA-TRUEBA SSZINNAI GVAN VLIET GTROCHET DAMIEL JLEGER JCZERNICHOW PSCOTET V: "Polymorphic length of FOXE1 alanine stretch: evidence for genetic susceptibility to thyroid dysgenesis", HUM GENET, 2007, pages 467 - 76, XP002466387, DOI: doi:10.1007/s00439-007-0420-5
CARRE AHAMZA RTKARIYAWASAM DGUILLOT LTEISSIER RTRON ECASTANET MDUPUY CEL KHOLY MPOLAK M.: "A novel FOXE1 mutation (R73S) in Bamforth-Lazarus syndrome causing increased thyroidal gene expression", THYROID, vol. 24, no. 24, 2014, pages 649 - 54
CARRE ARACHDI LTRON ERICHARD BCASTANET MSCHLUMBERGER MBIDART JMSZINNAI GPOLAK M.: "Hesl is required for appropriate morphogenesis and differentiation during mouse thyroid gland development", PLOS ONE, vol. 6, 2011, pages 6
CARRE ASTOUPA AKARIYAWASAM DGUERIOUZ MRAMOND CMONUS TLEGER JGAUJOUX SSEBAG FGLASER N: "Mutations in BOREALIN cause thyroid dysgenesis", HORN MOL GENET, vol. 26, 2017, pages 599 - 610
CARRE ASZINNAI GCASTANET MSURA-TRUEBA STRON EBROUTIN-L'HERMITE IBARAT PGOIZET CLACOMBE DMOUTARD ML: "Five new TTFl/NKX2.1 mutations in brain-lung-thyroid syndrome: rescue by PAX8 synergism in one case", HUM MOL GENET, vol. 18, no. 18, 2009, pages 2266 - 76
CARVALHO DPDUPUY C: "Thyroid hormone biosynthesis and release", MOL CELL ENDOCRINOL, vol. 458, 2017, pages 6 - 15, XP085269443, DOI: doi:10.1016/j.mce.2017.01.038
CHAKRABORTI SNATARAJAN KCURIEL JJANKE CLIU J: "The emerging role of the tubulin code: From the tubulin molecule to neuronal function and disease", CYTOSKELET, vol. 73, 2016, pages 521 - 550
CHOI MSCHOLL UIJI WLIU TTIKHONOVA IRZUMBO PNAYIR ABAKKALOGLU AOZEN SSANJAD S: "Genetic diagnosis by whole exome capture and massively parallel DNA sequencing", PROC NATL ACAD SCI U A, vol. 106, 2009, pages 19096 - 101, XP055082340, DOI: doi:10.1073/pnas.0910672106
CHU SGBECKER RCBERGER PBBHATT DLEIKELBOOM JWKONKLE BMOHLER ERREILLY MPBERGER JS: "Mean platelet volume as a predictor of cardiovascular risk: a systematic review and meta-analysis", J THROMB HAEMOST, vol. 8, 2010, pages 148 - 56
DELADOEY JRUEL JGIGUERE YVAN VLIET G: "Is the incidence of congenital hypothyroidism really increasing? A 20-year retrospective population-based study in Quebec", J CLIN ENDOCRINOL METAB, vol. 96, 2011
DELANO, W.L., THE PYMOL MOLECULAR GRAPHICS SYSTEM, 2002
DI JESO BARVAN P: "Thyroglobulin From Molecular and Cellular Biology to Clinical Endocrinology", ENDOCR REV, vol. 37, 2016, pages 2 - 36
FENG SUN ET AL: "The genetic characteristics of congenital hypothyroidism in China by comprehensive screening of 21 candidate genes", EUROPEAN JOURNAL OF ENDOCRINOLOGY, vol. 178, no. 6, 11 April 2018 (2018-04-11), GB, pages 623 - 633, XP055514599, ISSN: 0804-4643, DOI: 10.1530/EJE-17-1017 *
FIORE MGOULAS CPILLOIS X: "A new mutation in TUBB1 associated with thrombocytopenia confirms that C-terminal part of 01-tubulin plays a role in microtubule assembly", CLIN GENET, vol. 91, 2017, pages 924 - 926
FRANCHINI MLIPPI GMANZATO FVESCOVI PPTARGHER G: "Hemostatic abnormalities in endocrine and metabolic disorders", EUR J ENDOCRINOL, vol. 162, pages 439 - 51
GADADHAR SBODAKUNTLA SNATARAJAN KJANKE C: "The tubulin code at a glance", J CELL SCI, vol. 130, 2017, pages 1347 - 1353
GAIDE CHEVRONNAY HPJANSSENS VVAN DER SMISSEN PLIAO XHABID YNEVO NANTIGNAC CREFETOFF SCHERQUI SPIERREUX CE: "A mouse model suggests two mechanisms for thyroid alterations in infantile cystinosis: decreased thyroglobulin synthesis due to endoplasmic reticulum stress/unfolded protein response and impaired lysosomal processing", ENDOCRINOLOGY, vol. 156, 2015, pages 2349 - 64
GASPARYAN AYAYVAZYAN LMIKHAILIDIS DPKITAS GD: "Mean platelet volume: a link between thrombosis and in- flammation?", CURR PHARM DES, vol. 17, 2011, pages 47 - 58
GAWADE SMAYER CHAFEN KBARTHLOTT TKRENGER WSZINNAI G: "Cell Growth Dynamics in Embryonic and Adult Mouse Thyroid Revealed by a Novel Approach to Detect Thyroid Gland Subpopulations", THYROID, vol. 26, 2016, pages 591 - 9
GORDON CTPETIT FKROISEL PMJAKOBSEN LZECHI-CEIDE RMOUFADEM MBOLE-FEYSOT CPRUVOST SMASSON CTORES F: "Mutations in endothelin 1 cause recessive auriculocondylar syndrome and dominant isolated question-mark ears", AM J HUM GENET, vol. 93, 2013, pages 1118 - 1125
GUILLOT LCARRE ASZINNAI GCASTANET MTRON EJAUBERT FBROUTIN ICOUNIL FFELDMANN DCLEMENT A: "NKX2-1 mutations leading to surfactant protein promoter dysregulation cause interstitial lung disease in 'Brain-Lung-Thyroid Syndrome", HUM MUTAT, vol. 31, 2010, pages E 1146 - 62
HILDEBRANDT FHEERINGA SFRUSCHENDORF FATTANASIO MNURNBERG GBECKER CSEELOW DHUEBNER NCHERNIN GVLANGOS CN: "A systematic approach to mapping recessive disease genes in individuals from outbred populations", PLOS GENET, vol. 5, 2009, pages 5
HIXSON ET AL., J LAB AUTOM, vol. 20, no. 5, 2015, pages 562 - 73
HUOTARI JHELENIUS A: "Endosome maturation", EMBO J, vol. 30, 2011, pages 3481 - 500
JANKE C: "The tubulin code: molecular components, readout mechanisms, and functions", J CELL BIOL, vol. 206, 2014, pages 461 - 72, XP055467428, DOI: doi:10.1083/jcb.201406055
JOE PABANCRJCC ALUDUENA RF: "Roles of beta-tubulin residues Ala428 and Thr429 in microtubule formation in vivo", J BIOL CHEM, vol. 284, 2009, pages 4283 - 91
K. FRESON ET AL: "The TUBB1 Q43P functional polymorphism reduces the risk of cardiovascular disease in men by modulating platelet function and structure", BLOOD, vol. 106, no. 7, 1 October 2005 (2005-10-01), US, pages 2356 - 2362, XP055514704, ISSN: 0006-4971, DOI: 10.1182/blood-2005-02-0723 *
KARIYAWASAM DRACHDI LCARRE AMARTIN MHOULIER MJANEL NDELABAR JMSCHARFMANN RPOLAK M: "DYRK1A BAC transgenic mouse: a new model of thyroid dysgenesis in Down syndrome", ENDOCRINOLOGY, vol. 156, 2015, pages 171 - 80
KATE BURLEY ET AL: "TUBB1 variants and human platelet traits", PLATELETS (LONDON), vol. 29, no. 2, 15 January 2018 (2018-01-15), GB, pages 209 - 211, XP055514566, ISSN: 0953-7104, DOI: 10.1080/09537104.2017.1411587 *
KIM JHPARK JHKIM SYBAE HY: "The mean platelet volume is positively correlated with serum thyrotropin concentrations in a population of healthy subjects and subjects with unsuspected subclinical hypothyroidism", THYROID, vol. 23, 2013, pages 31 - 7, XP055457678, DOI: doi:10.1089/thy.2012.0033
KIRCHER MWITTEN DMJAIN PO'ROAK BJCOOPER GMSHENDURE J: "A general framework for estimating the relative pathogenicity of human genetic variants", NAT GENET, vol. 46, 2014, pages 310 - 5, XP055541282, DOI: doi:10.1038/ng.2892
KUHLI-HATTENBACH CHELLSTERN PKOHNEN THATTENBACH LO: "Platelet activation by ADP is increased in selected patients with anterior ischemic optic neuropathy or retinal vein occlusion", PLATELETS, vol. 28, 2017
KUNISHIMA SKOBAYASHI RITOH TJ: "Mutation of the beta-1-tubulin gene associated with congenital macrothrombocytopenia affecting microtubule assembly", BLOOD, vol. 113, 2009, pages 458 - 461
KUNISHIMA SNISHIMURA SSUZUKI HIMAIZUMI MSAITO H: "TUBB1 mutation disrupting microtubule assembly impairs proplatelet formation and results in congenital macrothrombocytopenia", EUR J HAEMATOL, vol. 92, 2014, pages 276 - 82
KYRIAKAKIS NLYNCH JAJJAN RMURRAY RD: "The effects of pituitary and thyroid disorders on haemostasis: potential clinical implications", CLIN ENDOCRINOL OXF, vol. 84, 2016, pages 473 - 84
LADOUX BMEGE RMTREPAT X: "Front-Rear Polarization by Mechanical Cues: From Single Cells to Tissues", TRENDS CELL BIOL, vol. 26, 2016, pages 420 - 433, XP029552410, DOI: doi:10.1016/j.tcb.2016.02.002
LECINE PITALIANO JE JRKIM SWVILLEVAL JLSHIVDASANI RA: "Hematopoietic-specific beta 1 tubulin participates in a pathway of platelet biogenesis dependent on the transcription factor NF-E2", BLOOD, vol. 96, 2000, pages 1366 - 73
LEGER ET AL.: "ESPE-PES-SLEP-JSPE-APEG-APPES-ISPAE; Congenital Hypothyroidism Consensus Conference Group. European Society for Paediatric Endocrinology consensus guidelines on screening, diagnosis, and management of congenital hypothyroidism", J CLIN ENDOCRINOL METAB., vol. 99, no. 2, February 2014 (2014-02-01), pages 363 - 84
LEMOINE NRMAYALL ESJONES TSHEER DMCDERMID SKENDALL-TAYLOR PWYNFORD-THOMAS D: "Characterisation of human thyroid epithelial cells immortalised in vitro by simian virus 40 DNA transfection", BR J CANCER, vol. 60, 1989, pages 897 - 903
LUCA PERSANI ET AL: "Genetics and management of congenital hypothyroidism", BAILLIERE'S BEST PRACTICE AND RESEARCH. CLINICAL ENDOCRINOLOGYAND METABOLISM, vol. 32, no. 4, 19 May 2018 (2018-05-19), GB, pages 387 - 396, XP055514540, ISSN: 1521-690X, DOI: 10.1016/j.beem.2018.05.002 *
MAIORANA RCARTA AFLORIDDIA GLEONARDI DBUSCEMA MSAVA LCALACIURA FVIGNERI R: "Thyroid hemiagenesis: prevalence in normal children and effect on thyroid function", J CLIN ENDOCRINOL METAB, vol. 88, 2003, pages 1534 - 6
MORGENTHALER STHILLY WG: "A strategy to discover genes that carry multi-allelic or mono-allelic risk for common diseases: a cohort allelic sums test (CAST", MUTAT RES, vol. 615, 2007, pages 28 - 56, XP005826818, DOI: doi:10.1016/j.mrfmmm.2006.09.003
NETTORE I C ET AL: "The molecular causes of thyroid dysgenesis: A systematic review", JOURNAL OF ENDOCRINOLOGICAL INVESTIGATION 2013 EDITRICE KURTIS S.R.L. ITA, vol. 36, no. 8, September 2013 (2013-09-01), pages 654 - 664, XP009508610, ISSN: 0391-4097 *
NEVE PKETELBANT-BALASSE PWILLEMS CDUMONT JE: "Effect of inhibitors of microtubules and microfilaments on dog thyroid slices in vitro", EXP CELL RES, vol. 74, 1972, pages 227 - 44, XP024852108, DOI: doi:10.1016/0014-4827(72)90501-0
NILSSON MFAGMAN H.: "Mechanisms of thyroid development and dysgenesis: an analysis based on developmental stages and concurrent embryonic anatomy", CURR TOP DEV BIOL., vol. 106, 2013, pages 123 - 170
NILSSON MFAGMAN H: "Development of the thyroid gland", DEVELOPMENT, vol. 144, 2017, pages 2123 - 2140
NIRSCHL JJMAGIERA MMLAZARUS JEJANKE CHOLZBAUR EL: "a-Tubulin Tyrosination and CLIP-170 Phosphorylation Regulate the Initiation of Dynein-Driven Transport in Neurons", CELL REP, vol. 14, 2016, pages 2637 - 52
NOGALES EWOLF SGDOWNING KH: "Structure of the alpha beta tubulin dimer by electron crystallography", NATURE, vol. 391, 1998, pages 199 - 203
OPITZ RHITZ MPVANDERNOOT ITRUBIROHA AABU-KHUDIR RSAMUELS MDESILETS VCOSTAGLIOLA SANDELFINGER GDELADOEY J: "Functional zebrafish studies based on human genotyping point to netrin-1 as a link between aberrant cardiovascular development and thyroid dysgenesis", ENDOCRINOLOGY, vol. 156, 2015, pages 377 - 88
PATEL SRRICHARDSON JLSCHULZE HKAHLE EGALJART NDRABEK KSHIVDASANI RAHARTWIG JHITALIANO JE JR.: "Differential roles of microtubule assembly and sliding in proplatelet formation by megakaryocytes", BLOOD, vol. 106, 2015, pages 4076 - 85
POHLENZ JMAQUEEM ACUA KWEISS REVAN SANDE JREFETOFF S: "Improved radioimmunoassay for measurement of mouse thyrotro- pin in serum: strain differences in thyrotropin concentration and thyrotroph sensitivity to thyroid hormone", THYROID J AM THYROID ASSOC, vol. 9, 1999, pages 1265 - 1271
PROSSER SLPELLETIER L: "Mitotic spindle assembly in animal cells: a fine balancing act", NAT REV MOL CELL BIOL, vol. 18, 2017, pages 187 - 201
PROTA, A. E.BARGSTEN, K.ZURWERRA, D.FIELD, J. J.DIAZ, J. F.ALTMANN, K. H.STEINMETZ, M. O.: "Molecular mechanism of action of microtubule-stabilizing anticancer agents", SCIENCE, vol. 339, 2013, pages 587 - 590, XP055394386, DOI: doi:10.1126/science.1230582
RAIBORG CWENZEL EMSTENMARK H: "ER-endosome contact sites: molecular compositions and functions", EMBO J, vol. 34, 2015, pages 1848 - 58
RAMOS HECARRE ACHEVRIER LSZINNAI GTRON ECERQUEIRA TLLEGER JCABROL SPUEL OQUEINNEC C: "Extreme phenotypic variability of thyroid dysgenesis in six new cases of congenital hypothyroidism due to PAX8 gene loss-of-function mutations", EUR J ENDOCRINOL, vol. 171, 2014, pages 499 - 507
RIBAULT ET AL.: "French Fetal Goiter Study Group. Experience with intraamniotic thyroxine treatment in nonimmune fetal goitrous hypothyroidism in 12 cases", J CLIN ENDOCRINOL METAB., vol. 94, no. 10, October 2009 (2009-10-01), pages 3731 - 9
ROUSSET BDUPUY CMIOT FDUMONT J: "Chapter 2 Thyroid Hormone Synthesis And Secretion", ENDOTEXT, 2015
SALI, A.BLUNDELL, T. L: "Comparative protein modelling by satisfaction of spatial restraints", J. MOL. BIOL., vol. 234, 1993, pages 779 - 815, XP024008717, DOI: doi:10.1006/jmbi.1993.1626
SCHWER HDLECINE PTIWARI SITALIANO JE JRHARTWIG JHSHIVDASANI RA: "A lineage-restricted and divergent beta-tubulin isoform is essential for the biogenesis, structure and function of blood platelets", CURR BIOL, vol. 11, 2001, pages 579 - 86
SENEE VCHELALA CDUCHATELET SFENG DBLANC HCOSSEC JCCHARON CNICOLINO MBOILEAU PCAVENER DR: "Mutations in GLIS3 are responsible for a rare syndrome with neonatal diabetes mellitus and congenital hypothyroidism", NAT GENET, vol. 38, 2006, pages 682 - 687, XP007904110, DOI: doi:10.1038/ng1802
SHENDURE ET AL., NATURE BIOTECH., vol. 26, no. 10, 2008, pages 1135 - 45
SQUIZZATOROMUALDI EBULLER HRGERDES VE, CLINICAL REVIEW: THYROID DYSFUNCTION AND EFFECTS ON COAGULATION AND FIBRINOLYSIS: A SYSTEMATIC REVIEW, vol. 92, 2007, pages 2415 - 20
STOUPA AKARIYAWASAM DCARRE APOLAK M: "Update of Thyroid Developmental Genes", ENDOCRINOL METAB CLIN NORTH AM, vol. 45, 2016, pages 243 - 54
SURA-TRUEBA SAUMAS CCARRE ADURIF SLEGER JPOLAK MDE ROUX N: "An inactivating mutation within the first extracellular loop of the thyrotropin receptor impedes normal posttranslational maturation of the extracellular domain", ENDOCRINOLOGY, vol. 150, 2009, pages 1043 - 50
SZINNAI GLACROIX LCARRE AGUIMIOT FTALBOT MMARTINOVIC JDELEZOIDE ALVEKEMANS MMICHIELS SCAILLOU B: "Sodium/iodide symporter (NIS) gene expression is the limiting step for the onset of thyroid function in the human fetus", J CLIN ENDOCRINOL METAB, vol. 92, 2007, pages 70 - 6, XP055344322, DOI: doi:10.1210/jc.2006-1450
TRUEBA SSAUGE JMATTEI GETCHEVERS HMARTINOVIC JCZERNICHOW PVEKEMANS MPOLAK MATTIE-BITACH T: "PAX8, TITF1, and FOXE1 gene expression patterns during human development: new insights into human thyroid development and thyroid dysgenesis-associated malformations", J CLIN ENDOCRINOL METAB, vol. 90, 2005, pages 455 - 62
WANG DVILLASANTE ALEWIS SACOWAN NJ: "The mammalian beta-tubulin repertoire: hematopoietic expression of a novel, heterologous beta-tubulin isotype", J CELL BIOL, vol. 103, 1986, pages 1903 - 10, XP001095924, DOI: doi:10.1083/jcb.103.5.1903
WOLFF JBHATTACHARYYA B: "Microtubules and thyroid hormone mobilization", ANN N ACAD SCI, vol. 253, 1975, pages 763 - 70, XP000579492, DOI: doi:10.1111/j.1749-6632.1975.tb19244.x

Similar Documents

Publication Publication Date Title
Stoupa et al. TUBB 1 mutations cause thyroid dysgenesis associated with abnormal platelet physiology
US11505788B2 (en) Fusion proteins and methods thereof
US10334829B2 (en) IMiD screening methods IMiD-sensitive cells with mutant CRBN
Carré et al. Mutations in BOREALIN cause thyroid dysgenesis
Stamou et al. Discovering genes essential to the hypothalamic regulation of human reproduction using a human disease model: adjusting to life in the “-omics” era
Rao et al. Advillin acts upstream of phospholipase C ϵ1 in steroid-resistant nephrotic syndrome
Pelletier et al. Absence of a differentiation defect in muscle satellite cells from DM2 patients
Baetens et al. Biallelic and monoallelic ESR2 variants associated with 46, XY disorders of sex development
KR20100015883A (en) Method of diagnosing, classifying and treating endometrial cancer and precancer
Joshi et al. Functional consequences of transferrin receptor‐2 mutations causing hereditary hemochromatosis type 3
Frints et al. Pathogenic variants in E3 ubiquitin ligase RLIM/RNF12 lead to a syndromic X-linked intellectual disability and behavior disorder
JP2011528894A (en) New oncogene NRF2
Wu et al. Somatic mutations of CADM1 in aldosterone-producing adenomas and gap junction-dependent regulation of aldosterone production
US20070249518A1 (en) Compositions and Methods for Treating Mental Disorders
Vishnopolska et al. Comprehensive identification of pathogenic gene variants in patients with neuroendocrine disorders
Nierhoff et al. New cell surface markers for murine fetal hepatic stem cells identified through high density complementary DNA microarrays
Romero et al. Novel role of the synaptic scaffold protein Dlgap4 in ventricular surface integrity and neuronal migration during cortical development
WO2019229113A1 (en) Detection of tubb1 mutations for diagnosing thyroid dysgenesis
Pang et al. Cloning, characterization, and expression analysis of the novel acetyltransferase retrogene Ard1b in the mouse
Du et al. Functional characterization of novel NPRL3 mutations identified in three families with focal epilepsy
WO2018002147A1 (en) Detection of borealin mutations for diagnosing thyroid dysgenesis
WO2013018866A1 (en) Method for assessing myelodysplastic syndrome or myeloid tumor predisposition, polypeptide and antibody therefor, and candidate screening method for therapeutic drug or prophylactic drug therefor
US20120003244A1 (en) Methods for apcdd1 mediated regulation of hair growth and pigmentation and mutants thereof
Noh et al. Defective phagocytic function of induced microglia-like cells is correlated with rapid progression of sporadic ALS
Xie et al. SRSF2 in Sertoli cells is essential for testicular development and spermatogenesis in mice

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: 19726430

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: 19726430

Country of ref document: EP

Kind code of ref document: A1