WO1996021728A2 - Transgenic animals having a defective thyroid hormone receptor gene - Google Patents

Transgenic animals having a defective thyroid hormone receptor gene Download PDF

Info

Publication number
WO1996021728A2
WO1996021728A2 PCT/EP1996/000161 EP9600161W WO9621728A2 WO 1996021728 A2 WO1996021728 A2 WO 1996021728A2 EP 9600161 W EP9600161 W EP 9600161W WO 9621728 A2 WO9621728 A2 WO 9621728A2
Authority
WO
WIPO (PCT)
Prior art keywords
gene
thyroid hormone
hormone receptor
trα
thyroid
Prior art date
Application number
PCT/EP1996/000161
Other languages
French (fr)
Other versions
WO1996021728A3 (en
Inventor
Bjorn Vennstrom
Original Assignee
Karo Bio Ab
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 Karo Bio Ab filed Critical Karo Bio Ab
Priority to AU47851/96A priority Critical patent/AU4785196A/en
Publication of WO1996021728A2 publication Critical patent/WO1996021728A2/en
Publication of WO1996021728A3 publication Critical patent/WO1996021728A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0276Knockout animals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/72Receptors; Cell surface antigens; Cell surface determinants for hormones
    • C07K14/721Steroid/thyroid hormone superfamily, e.g. GR, EcR, androgen receptor, oestrogen receptor
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases

Definitions

  • This invention relates to the production and use of transgenic mice and tissues and cell lines
  • the invention also relates to the testing of thyroid hormones such as T3, T4, rT3 as well as antagonists and agonist thereof identification and exploitation of components of other thyroid hormones such as T3, T4, rT3 as well as antagonists and agonist thereof identification and exploitation of components of other thyroid hormones such as T3, T4, rT3 as well as antagonists and agonist thereof identification and exploitation of components of other thyroid hormones such as T3, T4, rT3 as well as antagonists and agonist thereof identification and exploitation of components of other thyroid hormones such as T3, T4, rT3 as well as antagonists and agonist thereof identification and exploitation of components of other thyroid hormones such as T3, T4, rT3 as well as antagonists and agonist thereof identification and exploitation of components of other thyroid hormones such as T3, T4, rT3 as well as antagonists and agonist thereof identification and exploitation of other thyroid hormones such as T3, T4, rT3 as well as antagonists and agonist thereof identification and exploitation of other thyroid hormones such as T3, T4,
  • thyroid hormones influence every organ and cell type adult mammals, alter the metabolism of nutrients, and take part in regulating the expendit of energy and oxygen consumption (Larsen and Ingbar, 1992). They influence virtually every aspect of lipid metabolism as they have been shown to stimulate synthesis,
  • thyroid hormones are produced by the thyroid gland and distributed in a tightly regulated fashion. Except in lo
  • TSH Ihyroid Stimulating Hormone
  • TRH Ihyrotropin Releasing Hormone
  • TRH is secreted from the median emine where it reaches the anterior pituitary gland via a specialized vascular bed, the hypophysea
  • TRH portal venous system. TRH is delivered to the anterior pituitary gland where it stimulates
  • thyroid hormones influence both the production of TRH and TSH. It has been demonstrated that T3 is able to antagonize TRH mediated TSH release by the anterior pituitary gland. In this manner, even in the presence of TRH, high levels of circulating thyroid hormones can block the production of TSH.
  • TRH Tragerson et al., 1987
  • thyroid hormone production is therefore an intricate process aimed at providing neither an
  • TSH is the primary regulator of this pathway, as it dictates the production of thyroid
  • This gland is one of the largest endocrine organs, weighing
  • the follicles are filled with colloid which consists mainly of a clear proteinaceous substance called thyroglobulin.
  • Thyroglobulin is a thyroid protein which forms a peptide linkage with thyroid hormone precursors and thyroid hormones.
  • ATP adenosine triphosphate
  • tissue specific monodeiodinases enters the cell, it is deiodinated by tissue specific monodeiodinases to the more active for T3.
  • tissue specific monodeiodinases There are three general classes of deiodinases, type I (3, 3', 5' monodeiodinase 1 or t I 5 * D), type II (3, 3 * , 5' monodeiodinase 2 or type 2 5'D) and type III (3, 3 ⁇ 5
  • thyroid hormone a class of transcription factors referred to as thyroid hormone
  • the steroid/thyroid hormone receptor superfamily is one group of ligand dependent
  • the N-terminal domain is the most
  • variable between family members and in many receptors facilitates DNA binding and ligand
  • the hinge region contains sequences important for receptor
  • the ligand binding domain encompasses almost the entire carboxyl terminus (C-terminus) of the proteins and has
  • binding domain may participate in repression of transactivation (Baniamad et al., 1992).
  • HRE's Hormone Response Elements
  • TRE Ihyroid Response Element
  • TR ⁇ was the founding member of the subfamily of nuclear hormone receptors which
  • TR thyroid
  • VD3R vitamin D3
  • RAR retinoic acid
  • retinoic X, RXR retinoic X receptor receptors
  • proteins referred to as orphan receptors are proteins referred to as orphan receptors, as they show
  • TR binds specific DNA sequences, TRE's, and influence gene expression. As with other HRE's, these sequences are generally found in the 5" flanking region of the gene adjacent
  • Liganded TR bound to a TRE can either activate or repress gene expression.
  • TRE's which act as silencers are referred to as negative TRE's, nTRE's (for a review see Williams and Brent, 1995).
  • TR's appear to be nuclea
  • TR's are able to bind to TRE's in the absence of hormone.
  • this cont unliganded TR has been shown to act as a constitutive repressor of basal transcription on genes containing TRE's (Graupner et al., 1989).
  • the basic TRE's are a rather degenerate group of sequences which occur in a variety of
  • TRE's include a direct repeat hexamer separated by 4 base pairs (bp) (Dir4), an
  • the Dir 4 element is found in
  • T3 three genes physiologically regulated by T3, the rat growth hormone (rGH), alpha myosin
  • ⁇ MHC heavy chain
  • ME malic enzyme
  • hepatic ME mRNA shows a 15-20 fold fluctuation in response to hypothyroidism
  • hypothyroidism hyperthyroidism (Towle et al., 1980, 1981) and hypothyroidism is associated with a decrease
  • TR a very important but poorly characterized property of TR is its ability to mediate
  • thyroid hormones repress TSH transcription in vitro and in mammals thyroid
  • v-erbA receptors, as v-erbA was found to have homology to these receptors.
  • the cellular homologue of v-erbA was identified as the chicken thyroid hormone receptor alpha, c-erb
  • the isoforms encoded by the ⁇ gene are designated ⁇ -1 and ⁇ -2 and those
  • TR ⁇ -1 is encoded by exons 1 through 9, whereas TR ⁇ -2 results from alternative splicing of exon 9 to exon 10. This alternative splicing event replaces the 40 final amino
  • TR ⁇ -2 isoform The precise role of the TR ⁇ -2 isoform is
  • TR ⁇ receptor isoforms bind thyroid hormone
  • TR ⁇ -2 isoform is expressed mai in the pituitary gland (Hodin et al., 1989) although recent evidence suggests that it is also
  • TR ⁇ -2 is expressed in some tissues at levels up to tenfold hig than the hormone binding variants and in the adult testes it is the only TR expressed (Ko
  • TR ⁇ -1 isoforms prior to fetal hormone production and decreased expression of receptor is associated with the production of hormone by the fetus. This is in contrast to
  • TR as a transactivator and a repressor and its implication for basal
  • hypothyroidism would result in the loss of ligand-dependent activ
  • hypothyroidism is used to describe a clinical state of thyroid hormone deficiency.
  • hypothyroidism results in abnormalities in all organ systems and the extent and permanence of these abnormalities are
  • the CNS pathology consists of hypoplasia of cortical neurons
  • cerebellar ataxia in which the patient has a wide-based and unstable gait. Histopathology the brain shows edema, mucinous deposits in and around nerve fibers, foci of degeneratio with reactive gliosis and in general, severe atherosclerosis of the cerebral vessels is prese
  • the cardiac system is also affected in hypothyroidism.
  • Severe anemia can also dev due to decreased production of erythropoeitin and abnormalities in red blood cell production.
  • Muscle stiffness and aching fluid in the lungs (pleural effusions), mucinous edema of the skin,
  • hair loss and hypercholesterolemia are also frequently associated with hypothyroidism.
  • hypothyroidism in rats results in decreased brain size, a reduction in dendritic arborization of
  • cerebellar Purkinje cells and in delays in myelin formation in the CNS and PNS. Additionally,
  • hyperthyroidism is used to describe symptoms associated with thyroid hormone
  • thyroid hormone excess As with hypothyroidism, there are many causes of hormone excess, and these are also generally classified as primary, secondary and tertiary. In general, thyroid hormone excess is
  • thyroid hormones act
  • GRTH Generalized Resistance to Thyroid Hormone
  • GRTH is an inherited syndrome of reduced tissue responsivene thyroid hormone (Refetoff et al., 1967). The syndrome is associated with two different T
  • the patient had a chromosomal deletion
  • the abnormality is due to mutations in the TR ⁇ locus which render it unable to bind thyroid hormones effectively but still able to bind DNA and interfere with the function of
  • v-erbA has served as a paradigm transdominant negative thyroid hormone receptor for thi syndrome.
  • Patients with GRTH are generally identified on the basis of extremely high (two to five times normal) levels of thyroid hormone in the presence of inappropriately normal levels of TSH and
  • GRTH GRTH exhibit variable but mild phenotypes and generally have bone abnormalities
  • ADHD hyperactivity disorder
  • tissues such as the heart, exhibit characteristics of hyperthyroidism, i.e. tachycardia, whereas tissues such as bone display characteristics of hypothyroidism.
  • hypothyroidism and are more pronounced in this disease than in those with the dominantly
  • a transgenic animal which is heterozygous for an at least partially defective thyroid hormone receptor gene.
  • gene may be inactivated for example by an insertion, deletion, substitution or inversion or
  • the animal is a rodent , more preferably a mouse.
  • the thyroid hormone receptor gene is the TR ⁇ gene. More preferably, the gene the TR ⁇ l gene. In such a case the mammal may include a functional TR ⁇ 2 gene.
  • One heterozygous transgenic animal in accordance with the invention may be bred with
  • the invention also provides cells derived from the animal of the invention which are heterozygous or homozygous for a defective thyroid hormone receptor.
  • the thyroid hormone receptor gene is the TR ⁇ gene. More preferably the gene is
  • the method may involve
  • the transgenic animal of the invention is suitable for testing the effects of agonists and antagonists of thyroid hormone action, in particular those that discriminate between TR ⁇ and
  • the TRa gene encodes for two different receptor isoforms, the ligand binding receptor T and the non ligand binding variant TR ⁇ -2.
  • the two receptors arise from alternative splici
  • TR ⁇ -1 is encoded by exons 1 thr
  • TR ⁇ -2 results from alternative splicing using a donor site present 128 bp after start of exon 9, here referred to as exon 9a, to splice to the acceptor site of exon 10.
  • Thi alternative splicing event replaces the 40 final amino acids encoded by TR ⁇ -1 with 120 a
  • TR ⁇ -2 the TR ⁇ -2 variant is unable to bind thyroid
  • exon 9b deletion of nucleotides in exon 9 which are after the splice site, (here ref to as exon 9b) would selectively disrupt TR ⁇ -1.
  • exon 9b deletion of nucleotides in exon 9 which are after the splice site
  • erbA is encoded on the opposite strand of the TR ⁇ locus ( Figure 2) and the last exon of t rev-erbA locus overlaps exon 10 of the TR ⁇ locus (Laudet, et. al., 1991).
  • the targeting construct had to be made so that i) the coding sequence specific t TR ⁇ -1 messenger RNA (mRNA) was removed, ii) the production of a TR ⁇ -2 mRNA with a normal coding sequence was possible, iii) the region of TR ⁇ exon 10 which overlapped the rev-erbA locus was undisturbed, and iv) a selectable marker was added.
  • mRNA coding sequence specific messenger RNA
  • exon 9 TR ⁇ -1 specific region (exon 9b) and replace it with TR ⁇ -2 specific sequences. Then,
  • poly (A) poly adenylation signal
  • TR ⁇ transcript This required that the selectable marker had its own promoter and poly (A)
  • RT RT
  • PCR polymerase chain reaction
  • the pmTR ⁇ ex. 9a + 10 intermediate vector was made as follows.
  • TR ⁇ -1 specific sequen was cleaved with Stu I and Sal I to remove the region encoding for TR ⁇ -1 specific sequen including much of the 3' UT region of exon 9, and replaced by the TR ⁇ -2 specific sequence
  • neomycin as expressed by the pMClneo poly A vector (Stratagene) which consists of the neomycin gene driven by the
  • tk thymidine kinase (tk) promoter followed by a poly (A) signal.
  • the intermediate vector, p/7.TR ⁇ ex.9a+ 10 was then cleaved with Sal I. This allows for the ligation of the fragmen
  • the exon 10 probe and the 3' probe corresponded to restriction fragments which gave normal strength signal, indicating binding to a single copy sequence, were cho as specific for binding to the mouse TR ⁇ locus and met the criteria for probes described above
  • the targeting vector was cesium chloride purified and the Bgl II fragment was removed,
  • DNA was digested with Bam HI which generates a 23 kb fragment from the
  • the recombined locus has two Bam HI sites 226 bp apart which
  • TR ⁇ -1 +/- mice were then interbred to obtain homozygotes and offspring scr by PCR to identify TR ⁇ -1 -/- mice.
  • FVB mouse strain were obtained (gift of F. Baas). Restriction mapping of the locus was
  • the targeting vector used for homologous recombination was constructed as follows usin
  • RT-PCR was performed on poly (A) selected RNA from mouse brain using poly d(T) as the
  • fibroblasts that were derived from day 13-14 embryos of neomycin resistant transgenic mice
  • DMEM Dulbecco's modified Eagle's medium
  • 2-mercaptoethanol 1 n_M sodium pyruvate, 1 x non-essential amino acids (Seromed), a 1000 U/ml leukemia inhibitory factor (LIF) was used.
  • Neomycin-resistant clones were picked 8 days after
  • the 5' probe was isolated by restriction digestion using Bam HI site in the plasmid
  • the 3' probe corresponds to a Bam HI / Hi fragment distal to exon 10.
  • the exon 10 probe (internal probe) was generated by PCR us
  • the 568 bp internal probe was generated using a 5' primer specific to exon 8 980 nt of mouse cDNA) and a 3' primer corresponding to the beginning of exon 9 (1002-1024 nt of mouse cDNA). PCR generated probes were labeled using [ ⁇ - 32 P]dCTP in the reaction
  • Blastocysts from C57bl females were collected 3.5 days after conception, and 10-25 ES cells were injected into each blastocoel. Re-expanded blastocysts were implanted into the uterine
  • chimerism were bred with BalbC females, and germline transmission was scored by coat color.
  • TRal +/- mice were identified by PCR and were then interbred to obtain homozygotes.
  • DNA was prepared from tail clips according to Laird et al. (1991), and analyzed by PCR analysis using the 5 'SAL primers described above. The 3" primers were different for each paired reaction. In the first reaction the 3' primer (3' rvTR ⁇ l) corresponded to nucleotides 1349
  • PCR produ were subjected to gel electrophoresis in ethidium bromide (EtBr) containing agarose gels a
  • genotype was determined by Sothern blotting. 1 cm of the tails of 3 week old pups was cut off, dissolved in lysis buffer and
  • Phenotype of mice lacking a functional TR ⁇ l receptor Phenotype of mice lacking a functional TR ⁇ l receptor
  • homozygous for the wt TR ⁇ gene 3 homozygous for the targeted allele, and 5 heterozygous.
  • RNA and the +/- mice all three RNAs. This was verified by a RT-RCR assay as shown figure 5, which utilized oligonucleotide primers specific for the 3 RNAs (see experimenta).
  • Figure 6 shows that the striatum-specific gene SE6C is expressed at much lower levels in /- mice as compared to the wt animals, whereas RC3 levels were unaffected. This indicates that the brain development in the -/- mice is affected by the deletion of the TR ⁇ l gene.
  • Thyroid hormone assays The free T3 and T4 levels of 3 months old homozygous TR ⁇ l -/- mice was determined as described. Figure 7 shows that the 12 -/- and 11 +/ + animals tested have thyroid hormo
  • mice lacking the gene for the thyroid hormone receptor a were anaesthetized and implanted with a telemetric device (DataScience model TA10ETA-F20) inside the peritoneum
  • Fig. 8 shows basal heart rate in 2 knockout mice and 2 hetrozygote control mice. Mice 1, 2
  • mice 3 were hetrozygote control mice (dotted lines) and mice 3, 4 were homozygote knockout mice
  • Fig. 9 shows corresponding data for temperature changes. Body temperature measured in 4
  • mice via implanted telemetric equipment. Mice 1, 2 were hetrozygote control mice (dotted
  • mice 3, 4 were homozygote knockout mice lacking the Th ⁇ -receptor. There was no obvious change in temperature between the knockout mice and the control mice. The change in ECG parameters are shown in table 1. It can be seen that the homozygote lacking the Tr ⁇ -1 had ECG changes with shorter time between Q-wave to peak of the T- (QT- ⁇ ) and to end of the T-wave (QT end ).
  • mice 1 and 2 were control mice. All values are time in msec. All data were obtained an
  • mice lacking the Tr ⁇ -1 had lower heart rate an moderate prolongation of QT end and QT peak . This might indicate disturbances in cardiac
  • thyroid hormones are mediated b two different receptors that are coexpressed in some tissues, whereas other tissues express
  • mice and their tissues and cells described above are suitable for testing the specifity of the effects of agonists and antagonists of thyroid hormone action.
  • mice are also suitable for providing
  • mice or cells derived from them may be used to study:
  • tissues include the hypophysis (producing growth hormone, prolacting, thyroid
  • Basal metabolic rate, gluconeogenesis, lipogenesis, lipolysis and thermogenesis are increased in hyperthyroidism and decreased during hypothyroidism. The effect of
  • receptor-specific thyroid hormone antagonists or agonists on such metabolic proces can be determined with the mice described above.
  • the role of compone extrareceptor effects can be studied and exploited.
  • the human TR ⁇ gene is located close to the c-erbB2 gene, and is therefore usually co-amplified. It could
  • TRs extrareceptor effects
  • genes and their encoded proteins that participate in such pathways can be isolated, characterized, and
  • mice used for subsequent screening for novel agonists and -antagonists. 13.
  • the mice will be suitable for providing "proof of action” for agonists and antagoni TR action and extrareceptor effects.
  • a transferable silencing domain is present in the thyroid hormone receptor in the v-erba oncogene product and in the retinoic acid
  • thyroid stimulating hormone ⁇ gene regulation of the thyroid stimulating hormone ⁇ gene by thyroid hormone: receptor intera
  • thyroid hormone its possible pathophysiological significance. J. Endocrinol. Invest., 6, 59-66.
  • Neonatal hypothyroidism affects the timely expression of myelin-associated glycoprotein in the rat brain.
  • the c-erb-A protein is a high-affinity receptor for thyroid hormone.
  • TR ⁇ 2 immunoreactive thyroid hormone ⁇ 2 receptor
  • Thyroid hormone regulates TRH biosynthesis in the para ventricular nucleu the rat hypothalamus. Science, 238, 78-80.
  • a nuclear protein is required for thyroid hormone receptor binding to an inhibitory half-site in the epidermal growth factor receptor promoter. Mol. Endocrinol., 6, 627-635.
  • Thyroid hormone regulates the mouse thyrotropin P-subunit gene promoter in transfected primary thyrotropes. J.

Abstract

According to one aspect of the invention there is provided a transgenic animal which is heterozygous for an at least partially defective thyroid hormone receptor gene. The defective gene may be inactivated for example by an insertion, deletion, substitution or inversion or other suitable genetic manipulation. According to other aspects of the invention there are provided cells derived from the transgenic mammal of the invention and methods of testing the agonist/antagonist properties of a compound using the transgenic mammal of the invention or such cells.

Description

TRANSGENIC ANIMALS HAVING A __-r_-CTTV_; THYROID HORMONE RECEPTOR GENE
This invention relates to the production and use of transgenic mice and tissues and cell lines
thereof that in a homozygous form lack the gene for thyroid hormone receptor alpha 1
(TRαl). The invention also relates to the testing of thyroid hormones such as T3, T4, rT3 as well as antagonists and agonist thereof identification and exploitation of components of other
physiological pathways that are regulated by thyroid hormones or their derivatives.
INTRODUCTION
Thyroid hormones
Thyroid hormones, thyroxine (T4) and 3, 5, S'-triiodothyronine (T3) exert profound effects
on the growth, development, and homeostasis of organisms (Schwartz, 1983). In the
amphibian, thyroid hormone is crucial for metamorphosis (Gilbert and Frieden, 1981;
Galton, 1983). The remarkable transformation of tadpole to terrestrial frog is absolutely
dependent on the presence of thyroid hormone. Furthermore, when exogenous thyroid
hormones are given, the tadpole tail is resorbed prematurely and precocious development ensues (Beckingham-Smith and Tata, 1976). These early studies showed that thyroid hormones play a crucial role in normal development and suggested that the findings may be
applied to higher organisms. Indeed, in the developing fetus and perinatal animal, they are
fundamental for the normal maturation of the skeleton and central nervous system (CNS)
(Schwartz, 1983). In addition to developmental effects, thyroid hormones influence every organ and cell type adult mammals, alter the metabolism of nutrients, and take part in regulating the expendit of energy and oxygen consumption (Larsen and Ingbar, 1992). They influence virtually every aspect of lipid metabolism as they have been shown to stimulate synthesis,
mobilization, and degradation of lipids (Sestoft, 1980; Chait et al, 1979). Finally, the
hormones exert pleiotropic effects on the heart, ranging from altering the contractile properties of cardiac muscle to influencing heart rate (Klein, 1990).
Because thyroid hormones have important and diverse influences on the organism, their
appropriate production and distribution are essential. In mammals, thyroid hormones are produced by the thyroid gland and distributed in a tightly regulated fashion. Except in lo
vertebrates, control of thyroid hormone production is mediated by the anterior pituitary peptide thyrotropin, also known as Ihyroid Stimulating Hormone (TSH). In higher
vertebrates, the control of TSH secretion is in turn influenced by Ihyrotropin Releasing Hormone (TRH). TRH is a modified tripeptide produced in the supraoptic and
paraventricular nuclei of the hypothalamus. Once TRH is produced, it is transported to an
stored in a structure called the median eminence. TRH is secreted from the median emine where it reaches the anterior pituitary gland via a specialized vascular bed, the hypophysea
portal venous system. TRH is delivered to the anterior pituitary gland where it stimulates
both the production and release of TSH. Subsequently, TSH passes into the blood stream
is transported to the thyroid gland where it regulates thyroid hormone production.
Importantly, through a negative feedback regulatory loop, thyroid hormones influence both the production of TRH and TSH. It has been demonstrated that T3 is able to antagonize TRH mediated TSH release by the anterior pituitary gland. In this manner, even in the presence of TRH, high levels of circulating thyroid hormones can block the production of TSH. Thyroid
hormones may also inhibit the production or release of TRH (Segerson et al., 1987). Finally,
it has been shown that high amounts of thyroid hormone reduce the responsiveness of the
thyroid gland to TSH (Larsen and Ingbar, 1992), suggesting that autoregulation provides an
additional sensitive control mechanism for thyroid hormone production. The regulation of thyroid hormone production is therefore an intricate process aimed at providing neither an
excess nor a deficiency of hormone.
TSH is the primary regulator of this pathway, as it dictates the production of thyroid
hormone by the thyroid gland. This gland is one of the largest endocrine organs, weighing
20 grams, and is located closely affixed to the anterior and lateral aspects of the trachea. It
has an enormous capacity to respond to TSH and with hyperstimulation can enlarge to many
hundreds of grams, a condition referred to as goiter. By light microscopy, the normal gland
can be shown to consist of closely packed sacs or follicles lined by a single layer of cuboidal
cells. The follicles are filled with colloid which consists mainly of a clear proteinaceous substance called thyroglobulin. Thyroglobulin is a thyroid protein which forms a peptide linkage with thyroid hormone precursors and thyroid hormones.
Synthesis of T4 and T3 occurs as follows: i) active transport of iodide into the thyroid
gland, ii) oxidation of iodide and iodination of tyrosyl residues within thyroglobulin to form the inactive thyroid hormone precursors MIT and DIT, and finally, iii) the coupling of iodotyrosines to form the hormones T4 and T3. Release of the hormone is achieved by
hydrolysis of thyroglobulin by thyroid peptidases and proteases to produce free iodinated
amino acids which are transferred to the blood. Upon entering the blood, thyroid hormon
are bound reversibly to proteins synthesized in the liver and transported to various tissues.
The cellular uptake of thyroid hormones is poorly understood. There is evidence for a car
transport system for each hormone independently, and this carrier system is dependent on cellular adenosine triphosphate (ATP) for energy and sodium (Mendel et al. , 1988 and
Kreening et al., 1983). In addition to the aforementioned carrier system, there is also a
saturable, stereospecific, sodium independent system (Blondeau et al., 1988). Once T4
enters the cell, it is deiodinated by tissue specific monodeiodinases to the more active for T3. There are three general classes of deiodinases, type I (3, 3', 5' monodeiodinase 1 or t I 5*D), type II (3, 3*, 5' monodeiodinase 2 or type 2 5'D) and type III (3, 3\ 5
monodeiodinase, 5D) (Kohrle et al., 1991). Once thyroid hormones have entered the cell,
they bind to and activate a class of transcription factors referred to as thyroid hormone
receptors alpha and beta.
Nuclear Hormone Receptors
The steroid/thyroid hormone receptor superfamily is one group of ligand dependent
transcription factors which function as sequence specific DNA binding proteins. This famil of receptors is divided into two subfamilies based on sequence homology, the steroid recep
family and the thyroid hormone/retinoic acid receptor family. The basic structure of this family of receptors and the function of various domains is shown in the accompanying Figure
1 which indicates percentage amino acid homology.
These nuclear receptors appear to have arisen from a single ancestral gene, and multiple
members of subfamilies probably arose by gene duplication events followed by dispersal to
multiple chromosomes (Laudet et al., 1992). The basic structure of the superfamily proteins
is similar and composed of several functional domains. The N-terminal domain is the most
variable between family members and in many receptors facilitates DNA binding and ligand
independent transactivation (Tasset et al., 1990). The DNA binding domain has the highest
sequence homology in the superfamily, and is critical for recognition and binding to a
specific sequence in DNA. The hinge region contains sequences important for receptor
localization (LaCasse et al., 1993) and possibly for DNA binding. The ligand binding domain encompasses almost the entire carboxyl terminus (C-terminus) of the proteins and has
additional functions such as hormone-dependent dimerization, ligand binding and the
activation of transcription (transactivation). Recent evidence also suggests that the ligand
binding domain may participate in repression of transactivation (Baniamad et al., 1992).
The DNA sequences specifically recognized and bound by steroid hormone receptors are
referred to as Hormone Response Elements (HRE's). HRE's which specifically interact with thyroid hormone receptors are referred to as Ihyroid Response Element (TRE). These have
been defined functionally by the ability of the specific receptor to bind to the sequence and
influence transcription using the experimental techniques of transient transfection, gel- retardation assays and DNA footprinting.
TR, TRE's and Control of Gene Expression
TRα was the founding member of the subfamily of nuclear hormone receptors which
includes, among others, the thyroid (TR), vitamin D3 (VD3R), retinoic acid (RAR) and
cis-retinoic acid (retinoic X, RXR) receptors (Evans, 1988; Glass and Holloway, 1990).
Also included in this family are proteins referred to as orphan receptors, as they show
conserved homology with the aforementioned receptors. However, no specific ligand has
been identified and their physiologic roles are not yet elucidated. The basic structure of
is similar to that of other members of the steroid hormone superfamily.
TR binds specific DNA sequences, TRE's, and influence gene expression. As with other HRE's, these sequences are generally found in the 5" flanking region of the gene adjacent
the coding sequence. However, there are examples of TRE's found in introns and 3' UT regions. Liganded TR bound to a TRE can either activate or repress gene expression.
TRE's which act as silencers are referred to as negative TRE's, nTRE's (for a review see Williams and Brent, 1995).
In contrast to many other ligand-dependent transcription factors, TR's appear to be nuclea
and bound to chromatin, even in the absence of ligand. Therefore, unlike other classical
steroid receptors, TR's are able to bind to TRE's in the absence of hormone. In this cont unliganded TR has been shown to act as a constitutive repressor of basal transcription on genes containing TRE's (Graupner et al., 1989).
The basic TRE's are a rather degenerate group of sequences which occur in a variety of
contexts but generally consist of the hexameric sequence AGGTCA arranged as two identifiable half sites. The optimal binding and transactivation sites for TR, based on studies
of natural TRE's, include a direct repeat hexamer separated by 4 base pairs (bp) (Dir4), an
inverted hexamer with a 6 bp spacer (TREinvό) and two hexameric sequences oriented as a
palindrome (TREpal).
In most cases, the experiments used to define TRE's are consistent with the effects of thyroid
hormone in vivo, although there is limited knowledge regarding the specific hormone-
dependent regulation of genes in an organism. For example, the Dir 4 element is found in
three genes physiologically regulated by T3, the rat growth hormone (rGH), alpha myosin
heavy chain (αMHC) and malic enzyme (ME) promoters.
The physiologic correlate of the TR-mediated response in experimental systems are suggested
by the findings that GH secretion is blunted in hypothyroidism (Yaffe and Samuels, 1984), hepatic ME mRNA shows a 15-20 fold fluctuation in response to hypothyroidism and
hyperthyroidism (Towle et al., 1980, 1981) and hypothyroidism is associated with a decrease
in the level of cardiac αMHC (Lompre et al., 1984). This suggests that the in vitro data
demonstrating hormone dependent activation of transcription using the promoter elements of
these genes is consistent with the findings of thyroid hormone induced gene expression in a physiologic context.
Finally, a very important but poorly characterized property of TR is its ability to mediate
ligand-dependent repression of gene expression. The promoters for the α and β subunit
genes of TSH have been characterized extensively (Chatterjee et al. , 1989; Burnside et al.
1989; Wood et al. ; 1989 and Wondisford et al., 1993) although a limited number of other genes have also been identified which have nTRE's in their 5* flanking regions. There is
clear consensus sequence and the mechanism of T3-induced repression of transcription is
active area of research. However, it is postulated that negative regulation by TR may be
conferred by binding of a receptor monomer to a single half site (Crone et al., 1990 and
Thompson et al., 1992). Importantly, this regulation has physiologic implications as it is
known that thyroid hormones repress TSH transcription in vitro and in mammals thyroid
hormone excess results in decreased TSH, whereas hypothyroidism results in increased TS
Thyroid Hormone Receptors During the early 1980's the v-erbA oncogene was cloned and sequenced. The role of v-er and the characterization of its cellular homologue were facilitated by the cloning and
characterization of the sequence of the first steroid receptors, the estrogen and glucocortic
receptors, as v-erbA was found to have homology to these receptors. In 1986, the cellular homologue of v-erbA was identified as the chicken thyroid hormone receptor alpha, c-erb
or cTRα-1 (Sap et al., 1986) and immediately thereafter a second distinctive nuclear recep for T3, human TRβ, was identified (Weinberger et al., 1986). Characterization of the two
genes show they are similar in their intron exon boundaries, their overall structure and are
most related in the cysteine-rich DNA binding and C-terminal hormone binding domains.
Further analysis demonstrated that in addition to the two different receptors, alternate
splicing of the initial transcripts of each of the genes yield different mRNAs (Mitsuhashi et
al. , 1988; Izumo and Mahdavi, 1988). The various forms and isoforms are shown in Figure
1.
In mammals, the isoforms encoded by the α gene are designated α-1 and α-2 and those
encoded by the β gene are designated β-1 and β-2 (Sjόberg, 1991 and for a review see Forrest, 1994). There is an additional isoform in chicken designated β-0. The TRα gene
encodes the ligand binding isoform TRα-1 and the non ligand binding variant TRα-2. The
two receptors arise from alternative splicing which generates proteins with different C-
termini. TRα-1 is encoded by exons 1 through 9, whereas TRα-2 results from alternative splicing of exon 9 to exon 10. This alternative splicing event replaces the 40 final amino
acids encoded by TRα-1 with 120 amino acids specific for TRα-2 which abolishes the
hormone binding function and alters the DNA binding properties, RXR heterodimerization and the transactivation function of the protein. The precise role of the TRα-2 isoform is
unclear at present. In contrast, both TRβ receptor isoforms bind thyroid hormone and
activate transcription.
The various TR's show tissue specific and developmental specific patterns of expression (Sjόberg, 1994 and references therein). For example, the TRβ-2 isoform is expressed mai in the pituitary gland (Hodin et al., 1989) although recent evidence suggests that it is also
expressed at very low levels in some other tissues (Schwartz et al. , 1994). In addition, th
non-ligand binding variant, TRα-2 is expressed in some tissues at levels up to tenfold hig than the hormone binding variants and in the adult testes it is the only TR expressed (Ko
et al., 1989 and Jannini et al., 1994). Studies comparing the isoforms suggest that fetal
hormone production may influence TR expression patterns. For example, the early CNS
expresses TRα-1 isoforms prior to fetal hormone production and decreased expression of receptor is associated with the production of hormone by the fetus. This is in contrast to
1 which is abruptly increased in the CNS during the perinatal period in association with th
production of endogenous thyroid hormone. Other findings suggest that the expression of various receptors in the adult are also influenced by thyroid hormones (Hodin et al., 1990
Strait et al., 1990). These data suggest that not only do various TR's have different effects
gene expression, but they may also be differentially expressed in response to thyroid horm
The precise roles of the various receptors and isoforms remained to be defined.
Diseases associated with thyroid hormone disorders
The function of TR as a transactivator and a repressor and its implication for basal
transcription are poorly understood, but may be important in disease associated with both hypothyroidism and dominant negative interference by mutated receptors and receptor
isoforms. For example, hypothyroidism would result in the loss of ligand-dependent activ
of transcription, but in addition, unliganded TR would act as a constitutive repressor of ba transcription. Therefore, in order to understand the implications of receptor abnormalities in contrast to hormonal disturbance, it is important to describe the symptoms and abnormalities
associated with thyroid hormone excess and thyroid hormone deficiency. These abnormalities will then be compared to those associated with mutations in the TRβ gene in humans and the v-
erbA oncogene in experimental model systems.
Hypothyroidism
The term hypothyroidism is used to describe a clinical state of thyroid hormone deficiency.
There are many causes of thyroid hormone deficiency which are classified as primary if the
defect is at the level of the thyroid gland, secondary if there is a lack of TSH production, and
tertiary if it is due to lack of TRH. Regardless of the cause, hypothyroidism results in abnormalities in all organ systems and the extent and permanence of these abnormalities are
influenced by the timing and the severity of the deficiency.
Untreated hypothyroidism in the developing human is associated with permanent and profound
defects. This is the impetus which led to the widespread use of universal screening programs
to detect and treat congenital hypothyroidism. The major organ systems affected in the
developing human are the CNS and the skeleton. Untreated congenital hypothyroidism results in mental retardation. The associated CNS pathology consists of hypoplasia of cortical neurons
with poor development of cellular processes, retarded myelination and reduced vascularity.
Skeletal maturation is delayed and specific abnormalities in bone formation result in a speckled appearance of epiphyseal centers of ossification (epiphyseal dysgenesis or stippling). Linear growth is severely impaired leading to dwarfism in which the limbs are disproportionatel short in relation to the trunk. Finally, sensori-neural hearing loss is often present. These
abnormalities can be prevented if exogenous thyroid hormone is given beginning shortly
birth, but only partially reversed if treatment begins after three to six months of age.
In the adult, most organ systems are affected during hypothyroidism but return to normal
treatment with hormone. Life-threatening disorders are associated with abnormalities of
CNS and heart. Neurologic symptoms consist of a general slowing of intellectual functio
lethargy and somnolence, and in severe hypothyroidism epileptic seizures, stupor and co can develop. In addition, a particular abnormality can develop in some patients referred
cerebellar ataxia in which the patient has a wide-based and unstable gait. Histopathology the brain shows edema, mucinous deposits in and around nerve fibers, foci of degeneratio with reactive gliosis and in general, severe atherosclerosis of the cerebral vessels is prese
The cardiac system is also affected in hypothyroidism. The so called "myxedema heart"
associated with a decreased heart rate, "flabby" myocardium and decreased cardiac outpu which results in contraction of the blood volume and hypoperfusion of vital organs. This
manifest as symptoms of cold intolerance due to decreased perfusion and in some cases le
to kidney failure and congestive heart failure.
Other symptoms include decreased fertility in both men and women. In women this is du
failure to ovulate and in men to decreased sperm production. Severe anemia can also dev due to decreased production of erythropoeitin and abnormalities in red blood cell production.
Muscle stiffness and aching, fluid in the lungs (pleural effusions), mucinous edema of the skin,
hair loss and hypercholesterolemia are also frequently associated with hypothyroidism.
The molecular mechanism of these defects is largely unknown although experimentally induced
hypothyroidism in rats results in decreased brain size, a reduction in dendritic arborization of
cerebellar Purkinje cells and in delays in myelin formation in the CNS and PNS. Additionally,
studies have shown that the expression of genes which are important for normal CNS
development are decreased in hypothyroidism (Figueiredo et al., 1993 and Rodriguez-Pena et
al., 1993).
Hyperthyroidism
The term hyperthyroidism is used to describe symptoms associated with thyroid hormone
excess. As with hypothyroidism, there are many causes of hormone excess, and these are also generally classified as primary, secondary and tertiary. In general, thyroid hormone excess is
not as severe as that of thyroid hormone deficiency. The effects of hyperthyroidism are most pronounced on the cardiovascular system. Excess thyroid hormone results in an overall hypermetabolic state and the need to dissipate excess heat. This leads to compensatory changes which include dilatation of the peripheral vessels. In addition, thyroid hormones act
directly on the myocardium to stimulate the heart rate and increase the force of contraction.
Therefore, the cardiac output and workload on the heart are increased considerably. In addition, abnormalities in the normal electrical conduction of the heart often leads to cardiac arrhythmias. The stress on the heart associated with these abnormalities can lead to cardi decompensation if oxygen delivery to the heart can not exceed oxygen consumption. This the main life-threatening complication of hyperthyroidism.
Other symptoms include shortness of breath, increased appetite, muscle weakness and
fatigability, and in the CNS symptoms of excess excitation are manifest as hyperkinesia,
tremors, seizures, emotional lability and nervousness. The mechanism by which thyroid
hormone exerts the effects on the CNS and heart are unknown. However, no clear alterat in cerebral metabolism or vascular abnormalities have been noted.
Generalized Resistance to Thyroid Hormone
Importantly, no human disease has been associated with a dominant negative mutation or
deletion of the TRα-1 gene in humans. In contrast, disorders of the TRβ locus in humans
associated with the syndrome of Generalized Resistance to Thyroid Hormone (GRTH) (fo
review see Usala, 1994). GRTH is an inherited syndrome of reduced tissue responsivene thyroid hormone (Refetoff et al., 1967). The syndrome is associated with two different T
abnormalities. In the first case described, the patient had a chromosomal deletion
encompassing the TRβ locus which resulted in no receptor protein being made. In all othe
cases, the abnormality is due to mutations in the TRβ locus which render it unable to bind thyroid hormones effectively but still able to bind DNA and interfere with the function of
normal receptor. This mutant receptor inhibits the function of TR in experimental system
v-erbA has served as a paradigm transdominant negative thyroid hormone receptor for thi syndrome.
Patients with GRTH are generally identified on the basis of extremely high (two to five times normal) levels of thyroid hormone in the presence of inappropriately normal levels of TSH and
the absence of clinical evidence of hyperthyroidism. Patients with the dominantly inherited
form of GRTH exhibit variable but mild phenotypes and generally have bone abnormalities
characteristic of juvenile hypothyroidism ranging from delayed bone development to short stature, varying levels of mild mental retardation, tachycardia and attention deficit
hyperactivity disorder (ADHD) (Hauser et al. , 1993). More careful examination of these
patients has demonstrated that tissues show varying levels of resistance to thyroid hormone.
Some tissues, such as the heart, exhibit characteristics of hyperthyroidism, i.e. tachycardia, whereas tissues such as bone display characteristics of hypothyroidism.
Patients homozygous for a deletion of the TRβ locus also have high circulating levels of thyroid hormone with inappropriate TSH secretion, but also have hearing loss and a particular
bone abnormality referred to as stippled epiphyses (Refetoff et al., 1967 and Takeda et. al.,
1992). These patients were the result of consanguineous marriage and the phenotype associated with this or other recessive mutations remain to be defined. Although the majority of symptoms are similar, deafness and stippled epiphyses are more characteristic of juvenile
hypothyroidism and are more pronounced in this disease than in those with the dominantly
inherited form of the disease. These findings suggest that although the molecular defect underlying GRTH has been established, the role of TRα in modulating the influence of TRβ in an organism remains t
elucidated.
According to one aspect of the invention there is provided a transgenic animal which is heterozygous for an at least partially defective thyroid hormone receptor gene. The defec
gene may be inactivated for example by an insertion, deletion, substitution or inversion or
other suitable genetic mannipulation.
Preferably, the animal is a rodent , more preferably a mouse.
Preferably, the thyroid hormone receptor gene is the TRα gene. More preferably, the gene the TRαl gene. In such a case the mammal may include a functional TRα2 gene.
One heterozygous transgenic animal in accordance with the invention may be bred with
another such heterozygous transgenic mammal to produce a mammal which is homozygou the defective thyroid hormone receptor gene.
The invention also provides cells derived from the animal of the invention which are heterozygous or homozygous for a defective thyroid hormone receptor.
According to another aspect of the invention there is provided a method of producing a transgenic animal in accordance with the invention the method comprising :
1) preparing a gene encoding an at least partially defective thyroid hormone receptor as
described above;
2) introducing that gene into suitable carrier cells;
3) inserting those carrier cells into an embryo; and
4) replacing the embryo into a mother, and allowing the embryo to develop to full term.
Preferably, the thyroid hormone receptor gene is the TRα gene. More preferably the gene is
the TRαl gene.
According to a further aspect of the invention there is provided a method of testing the
agonist/antagonist properties of a compound in relation to the thyroid hormone receptor, the
method comprising:
contacting a transgenic animal in accordance with the invention with the compound and
monitoring subsequent development of the animal. Alternatively, the method may involve
using cells or tissues derived from the transgenic animal.
The transgenic animal of the invention is suitable for testing the effects of agonists and antagonists of thyroid hormone action, in particular those that discriminate between TRα and
TRβ .
The production of transgenic animals in accordance with the invention and their uses are now described, by way of example only, with reference to the accompanying drawings Figure
9.
Experimental outline for disruption of the mouse Trα-1 gene by homologous recombination
Genomic mapping and strategy for targeting construct
The TRa gene encodes for two different receptor isoforms, the ligand binding receptor T and the non ligand binding variant TRα-2. The two receptors arise from alternative splici
which generates proteins with different carboxy termini. TRα-1 is encoded by exons 1 thr
9 whereas TRα-2 results from alternative splicing using a donor site present 128 bp after start of exon 9, here referred to as exon 9a, to splice to the acceptor site of exon 10. Thi alternative splicing event replaces the 40 final amino acids encoded by TRα-1 with 120 a
acids specific for TRα-2. As mentioned, the TRα-2 variant is unable to bind thyroid
hormone and its physiologic function is unknown. Therefore, a targeted disruption of any the exons 1 through 9a would ablate both the hormone binding and non hormone binding
variants whereas deletion of nucleotides in exon 9 which are after the splice site, (here ref to as exon 9b) would selectively disrupt TRα-1. In addition, the orphan hormone recepto
erbA is encoded on the opposite strand of the TRα locus (Figure 2) and the last exon of t rev-erbA locus overlaps exon 10 of the TRα locus (Laudet, et. al., 1991).
Therefore, the targeting construct had to be made so that i) the coding sequence specific t TRα-1 messenger RNA (mRNA) was removed, ii) the production of a TRα-2 mRNA with a normal coding sequence was possible, iii) the region of TRα exon 10 which overlapped the rev-erbA locus was undisturbed, and iv) a selectable marker was added.
To accomplish this, the following cloning strategy was used. We planned to eliminate the
exon 9 TRα-1 specific region (exon 9b) and replace it with TRα-2 specific sequences. Then,
by the addition of a poly adenylation signal (poly (A)) to the end of the TRα-2 specific sequence, we would ensure appropriate processing of the transcript. This would also allow us
to leave the original exon 10 unaltered and thereby eliminate any chance of disturbing the rev-
erbA locus. In order to select for ES cells which had incorporated the vector, a selectable
marker was also required. To prevent the possibility of a fusion protein being made, we chose
to orient the selectable marker gene in the opposite transcriptional orientation to that of the
TRα transcript. This required that the selectable marker had its own promoter and poly (A)
signal.
To construct a targeting vector which fulfilled the criteria described above, we chose the
following approach:
i) Mapping of the 7__α locus
We first made a preliminary map of the TRα genomic locus using a 13 kb fragment of
genomic DNA derived from the FVB mouse strain (gift of F. Baas) which was believed to
contain exon 9 and exon 10 sequences (Figure 3A). Preliminary mapping showed that two Bgl II sites flanked a region of the locus which included exon 8 to exon 10. This fragment w subcloned and more detailed restriction mapping was done. Restriction mapping was
performed in an attempt to find a unique site in the 3' untranslated region (3' UT) of exo
which could be used for inserting a selectable marker and to replace exon 9b specific
sequences with exon 10 and a poly (A) signal. As depicted in Figure 3 A, the only appro
unique site was a Sea I site in exon 9b. However, most cloning vectors have Sea I sites i
ampicillin resistance gene which makes the use of the Sea I site impractical. In addition,
strategy could more easily be accomplished using a Sal I site. Therefore, we converted th
I site into a Sal I site.
ii) Production of the exon 9a +10 fragment
Simultaneously, a fragment (ex. 9a + 10) extending from the Stu I site in exon 9 (upstrea the splice donor) to just after the stop codon in exon 10 was cloned by reverse transcriptio
(RT) followed by polymerase chain reaction (PCR) using mouse brain as a template. This
generated a 522 bp fragment which was specific for the 3' terminus of the TRα-2 transcri which included the desired coding region and 47 bp of 3' UT but no poly (A) signal. In addition, a Sal I site was added using PCR directed mutagenesis at the 3' end of the produ
This allowed the fragment to be purified using a Stu I/Sal I cleavage.
iii) Generation of the pmTR ex. 9a +10 intermediate vector
The pmTRαex. 9a + 10 intermediate vector was made as follows. The pmTRαBgl II vect
was cleaved with Stu I and Sal I to remove the region encoding for TRα-1 specific sequen including much of the 3' UT region of exon 9, and replaced by the TRα-2 specific sequence
9a+ 10 described above. The resulting construct consists of exon 9a fused to exon 10 with
exon 9b regions necessary for the production of a TRα-1 transcript deleted. It also leaves the
original exon 10 region unaltered and rev-erbA unaffected.
iv) Preparation of a SV 40 poly (A) signal and selectable marker
Appropriate processing of the modified transcript, consisting of exons 9a fused to 10, still required the addition of a poly (A) signal. In addition, a selectable marker was required to
screen for cells which had taken up the vector. To ensure appropriate processing, we chose to use a SV 40 poly (A) signal and place it after the stop codon in exon 10 using the artificially
introduced Sal I site. As a selectable marker we chose to use neomycin as expressed by the pMClneo poly A vector (Stratagene) which consists of the neomycin gene driven by the
thymidine kinase (tk) promoter followed by a poly (A) signal.
In order to accomplish the addition of the SV 40 poly (A) signal, concomitantly with the
addition of the selectable marker, we proceeded as follows. A 224 bp SV 40 poly (A) signal flanked by Bam HI site was isolated. This fragment was then ligated into the Bam HI digested
pMClneo poly A plasmid so that its transcriptional orientation was opposite to that of the
neomycin resistance gene transcription. Once this was accomplished a Sal I/Xho I cleavage was used to release a fragment consisting of the SV 40 poly (A) followed by the neomycin selectable marker transcriptional unit in the opposite orientation. v) The targeting vector
In order to insert the Sal I/Xho I fragment described above, the intermediate vector, p/7.TRαex.9a+ 10 was then cleaved with Sal I. This allows for the ligation of the fragmen
consisting of the SV 40 poly (A) followed by the neomycin selectable marker into the Sal
site. The resulting vector, when used for recombination at the original locus, would gene
recombined locus as shown in Figure 3B. Of note, 2 new Bam HI sites are present in the recombined locus which were used in screening of recombinant clones as described below.
Probe design
In order to screen for homologous recombination, several probes were required as follows a probe specific to sequences 5' of the targeting construct, ii) a probe specific to sequences
of the targeting construct, iii) a probe corresponding to sequences in the targeting vector a
the normal allele which could be used to assess both the integrity of the 3' integration site for the presence of nonhomologous recombination, and finally, iv) a probe designed to tes
integrity of the 5' recombination site. To identify appropriate probes, various restriction
enzyme digestions of approximately 19 kb of the genomic locus were performed and subje
to gel electrophoresis followed by Southern Blotting. Thereafter the filter was hybridized high molecular weight DNA from 129 ES cells which was labeled by nick translation. Ba
which gave strong signals were presumed to be due to hybridization with repetitive sequen
and these areas were not used for generating probes (data not shown). The 5' probe, the i
8 internal probe, the exon 10 probe and the 3' probe corresponded to restriction fragments which gave normal strength signal, indicating binding to a single copy sequence, were cho as specific for binding to the mouse TRα locus and met the criteria for probes described above
(Figure 3A, B).
Electroporation and Screening for recombinant clones The targeting vector was cesium chloride purified and the Bgl II fragment was removed,
purified and used for electroporation into embryonic stem (ES) cells. DNA from neomycin resistant clones was extracted and screened by Southern blotting for homologous
recombination. DNA was digested with Bam HI which generates a 23 kb fragment from the
normal locus. In contrast, the recombined locus has two Bam HI sites 226 bp apart which
divides the 23 kb fragment into an approximately 13 kb 5' fragment and a 9.94 kb 3' fragment. The Bam HI digested DNA was then subject to Southern analyses and the filter hybridized
consecutively with three different probes: i) an internal probe specific for exon 10 sequences
ii) a 5" probe which recognizes sequences 5' of the targeting vector and iii) a 3* probe
recognizing sequences 3' of the targeting vector. Exon 10 probe gave only one band for the normal locus whereas the homologous recombination on one allele gave three fragments
corresponding to the 23 kb normal allele and the 13 kb and 9.9 kb fragments from the
recombined allele. Probing with the 5' and 3" probes demonstrate the 23 kb normal allele and the 13 kb and 9.9 kb recombined alleles, respectively. In addition, the internal probe gave no
additional bands, indicating that no additional nonhomologous recombination had occurred.
Finally, to verify that the 5' recombination site remained intact and that no other recombination
events had occurred, the DNA was digested with Stu I and probed with an intron 8 probe
(Figure 4). The appropriate recombination gave only one band as both the normal and recombined alleles are similar in this area. The frequency of homologous recombination approximately 1 :100 despite using non-isogenic DNA.
Generation and Screening of Mutant Mice
Targeted ES cells from three independent clones were injected into blastocysts and impla
into pseudopregnant females according to standard protocols. Male coat color chimeras bred and offspring screened by coat color followed by PCR of tail DNA for the targeted
disruption. TRα-1 +/- mice were then interbred to obtain homozygotes and offspring scr by PCR to identify TRα-1 -/- mice.
Experimental details
Genomic mapping and targeting vector
Two independent plasmid clones which contained a total of 19 kb of 3* mTRα locus from
FVB mouse strain were obtained (gift of F. Baas). Restriction mapping of the locus was
performed according to standard protocols (Sambrook et al., 1989 and RESULTS section
The targeting vector used for homologous recombination was constructed as follows usin
standard experimental procedures (Sambrook et al., 1989): A 7.2 kb Bgl II fragment including the region of mTRα from intron 7 to exon 10 was isolated and subcloned into t
SP70 vector (Promega) which had been modified so that only Cla I and EcoRV restrictio
remained in the polylinker. The Sea I site in the 3" UT region of exon 9 was converted in Sal I site by Sea I partial digestion followed by Sal I linker addition .
RT-PCR was performed on poly (A) selected RNA from mouse brain using poly d(T) as the
reverse primer. The cDNA was used as a template for PCR. Both primers had sequences
encoding a Sal I restriction enzyme site (underlined regions) followed by sequence
corresponding to either the beginning of exon 9 (5'SAL) (5*
GGAGTCGACCGAGAAGAGTCAGGAG-3' . or the 3' UT region of exon 10, 47 nucleotides
after the stop codon (3'SAL) (5'GCTACTCCTTCTCCCGTCGACAAC-3'V The resulting 570
bp PCR product (9a -I- 10) was purified and cloned into pGEM 3Zf(+) vector at a Sal I site
(pGEM 9a + 10) and sequenced. Thereafter the pSP70Bgl II vector was cleaved with Stu I and
Sal I to release a 1100 bp fragment consisting of sequence just upstream of the splice donor site in exon 9, and pan of the 3' UT region of exon 9. The pGEM 9a+ 10 vector was simultaneously digested with Stu I and Sal I to release a 522 nucleotide fragment which
included exon 9a downstream of the Stu I site and exon 10, 47 bp after the stop codon. The
fragments were purified on low melting agarose TAE. The Stu I/Sal I fragment from pGEM 9a -I- 10 was then ligated into the Stu 1/ Sal I digested intermediate cloning vector to produce
the pmTRαex.9a+ 10 vector. The remainder of the vector was cloned as described in results.
Maintenance and Electroporation ofES Cells
Cells (Handyside et al. , 1989) were grown on mitomycin C-treated primary mouse embryo
fibroblasts that were derived from day 13-14 embryos of neomycin resistant transgenic mice
(Muller et. al., 1991). As growth medium, Dulbecco's modified Eagle's medium (DMEM) supplemented with 15% fetal bovine serum, 20 U penicillin, 20mg/ml streptomycin, 10
2-mercaptoethanol, 1 n_M sodium pyruvate, 1 x non-essential amino acids (Seromed), a 1000 U/ml leukemia inhibitory factor (LIF) was used.
Prior to electroporation into embryonic stem (ES) cells, cesium chloride purified plasmi
digested with Bgl II and purified using Promega Magic DNA clean up system. Electrop
of 1 x 107 ES cells in 800 ml of Optimem with 10-50 μg Bgl II digested targeting vector
carried out using a BioRad Gene Pulser and parameters 250 V, 960 mF. Cells were plat
dishes with feeder cells, and 24 hours after electroporation the growth medium was supplemented with 375 μg/ml G418. Neomycin-resistant clones were picked 8 days after
electroporation and expanded for DNA analysis. We screened for homologous recombin events by Southern blotting.
Probe preparation and Southern Blot Analysis
The 5' probe was isolated by restriction digestion using Bam HI site in the plasmid
intermediate and the Kpn I site in the clone. The 3' probe corresponds to a Bam HI / Hi fragment distal to exon 10. The exon 10 probe (internal probe) was generated by PCR us
5' primer with the sequence GCGGAATTCAAGCTTTGGGGAAGACGACAGCAGTG
includes an EcoRI and Hind III site (the underlined portion represents 1377-1401 of mT cDNA) and the 3" SAL primer used to generate the PCR fragment used in the targeting construct. The 568 bp internal probe was generated using a 5' primer specific to exon 8 980 nt of mouse cDNA) and a 3' primer corresponding to the beginning of exon 9 (1002-1024 nt of mouse cDNA). PCR generated probes were labeled using [α-32P]dCTP in the reaction
mix and the remaining probes were labeled with [α -32P]dCTP using the random priming
method and a commercially available megaprime kit (Pharmacia, LKB).
DNA from neomycin resistant clones was prepared using a lysis buffer consisting of lOmM Tris Hcl pH = 7.8, lmM EDTA, 0.2M NaCl, 1 %SDS and 0.5mg/ml proteinase K followed
by phenol-chloroform extracted and ethanol precipitation. The DNA was then screened by
Southern blotting for homologous recombination. DNA was digested with Bam HI and
separated on 0.8% TBE agarose gels. Southern blotting was then performed using Hybond N nylon filters (Amersham), and the filters hybridized according to the manufacturers
recommendation, then stripped and rehybridized with probes as described above. Positive
clones were confirmed by digestion with Stu I followed by Southern analysis and filter
hybridization with the intron 8 probe.
Generation and Genotyping of mice
Blastocysts from C57bl females were collected 3.5 days after conception, and 10-25 ES cells were injected into each blastocoel. Re-expanded blastocysts were implanted into the uterine
horn of 2.5 days pseudopregnant F1(B6CBA) females. Male offspring with coat color
chimerism were bred with BalbC females, and germline transmission was scored by coat color.
TRal +/- mice were identified by PCR and were then interbred to obtain homozygotes. DNA was prepared from tail clips according to Laird et al. (1991), and analyzed by PCR analysis using the 5 'SAL primers described above. The 3" primers were different for each paired reaction. In the first reaction the 3' primer (3' rvTRαl) corresponded to nucleotides 1349
1362 of mTRa-1 cDNA and in the second reaction the 3' primer (3' SV40) corresponded t
sequence 5'-ACCACAACTCGAATGCAGTG-3' of the SV 40 poly (A) tail. PCR produ were subjected to gel electrophoresis in ethidium bromide (EtBr) containing agarose gels a
visualized using UV light. In some instances, the genotype was determined by Sothern blotting. 1 cm of the tails of 3 week old pups was cut off, dissolved in lysis buffer and
extracted with phenol (Barlow et al 1994). Southern blot analyses were done with probes a described above for identification of ES cell clones that had undergone homologous
recombination.
RNA analysis of mice To verify that no TRα-1 message was made and that TRα-2 message was normally produc
in the -/- mice, 5μg of poly (A) RNA (Vennstrom and Bishop, 1982) from various tissues
used for northern blotting. Northern filters were hybridized with a full length mouse TR - cDNA probe which recognizes both isoforms. RT-PCR was done using poly d(T) as a pri for cDNA synthesis the 5' primer, 5'SAL, and 3' primer, 3' rvTRαl described above. T
PCR products were then electrophoresed through agarose gels containing EtBr and viewed
under UV light.
Analysis of hormones
Levels of free T3 And T4 in the blood of the mice was done as described previously (Barl et al 1994)
Phenotype of mice lacking a functional TRαl receptor
Breeding properties
To date, 3 founders have given rise to £100 heterozygous animals. The heterozygous animals
exhibit normal behaviour as compared to control mice in terms of viability, fertility, and
growth. When heterozygotes are bred, the resulting litter size is normal (7-10 pups), and the sex ratio male: female of the offsspring 1 : 1. An example of a cross between heterozygotes is
shown in Figure 4.
DNA prepared from the tails of 10 pups of one litter was cut with BamHI, electrophoresed in
an agarose gel,, and subjected to Southern analysis. The figure shows that 2 animals were
homozygous for the wt TRα gene, 3 homozygous for the targeted allele, and 5 heterozygous.
Homozygous animals (£50 to date) are fertile, giving normal sized litters. No goiter or any other abnormalities have been observed in animals up to 18 months of age.
Analysis of thyroid hormone receptor mRNA
To determine if the wt +/+, heterozygous +/- and homozygous -/- mice expressed the expected TRαl, TRα2/wt and TRα2/mutant mRNA, poly A + RNA from the brains of 8 week
old animals was electrophoresed in agarose gels and subjected to Northern analysis by probing
with a radiolabelled TRα cDNA probe. The results (not shown) indicated that the +/+ animals expressed only the TRαl and TRα2/wt RNAs, the -/- animals only a TRα2/muta
RNA, and the +/- mice all three RNAs. This was verified by a RT-RCR assay as shown figure 5, which utilized oligonucleotide primers specific for the 3 RNAs (see experimenta
details).
Analysis of brain gene expression
To determine if the lack of TRαl receptor affected brain gene expression, polyA+ RNA
prepared from TRαl mice, and subjected to a Northern analysis with cDNA probes speci for 2 brain-specific genes, RC3 and SE6C. The same amount of RNA was loaded in all la
Figure 6 shows that the striatum-specific gene SE6C is expressed at much lower levels in /- mice as compared to the wt animals, whereas RC3 levels were unaffected. This indicat that the brain development in the -/- mice is affected by the deletion of the TRαl gene.
Thyroid hormone assays The free T3 and T4 levels of 3 months old homozygous TRαl -/- mice was determined as described. Figure 7 shows that the 12 -/- and 11 +/ + animals tested have thyroid hormo
levels that range from euthyroid to hypothyroid, with an average that is subnormal.
EXAMELE In this example of the uses of the mice in accordance with the invention, heart rate, temperature and ECG-changes in mice lacking the gene for thyroid hormone receptor α were studied.
Methods:
Homozygote mice lacking the gene for the thyroid hormone receptor a, were anaesthetized and implanted with a telemetric device (DataScience model TA10ETA-F20) inside the peritoneum
in the upper abdomen. This allows us to measure ECG, heart rate and body temperature in the
unrestrained mice living in the animal quarter. We started the data collection two days after
the implantation of the telemetric device and continued for 47 hours. All the data were
collected using a computer system and the data were analyzed using a spread sheet program.
(Excel).
Fig. 8 shows basal heart rate in 2 knockout mice and 2 hetrozygote control mice. Mice 1, 2
were hetrozygote control mice (dotted lines) and mice 3, 4 were homozygote knockout mice
lacking the Trα-1. It will be seen that heart rate was around 620 in control mice and around
520 in the mice lacking the Trα-1.
Fig. 9 shows corresponding data for temperature changes. Body temperature measured in 4
mice via implanted telemetric equipment. Mice 1, 2 were hetrozygote control mice (dotted
lines) and mice 3, 4 were homozygote knockout mice lacking the Thβ-receptor. There was no obvious change in temperature between the knockout mice and the control mice. The change in ECG parameters are shown in table 1. It can be seen that the homozygote lacking the Trα-1 had ECG changes with shorter time between Q-wave to peak of the T- (QT-^) and to end of the T-wave (QTend).
Mice QRS PQ QTpc , QTend
1 11.6 28.9 13.2 20.1
2 10.6 30.9 12.8 21.8
3 12.7 28.6 15.6 24.5
4 12.3 33.3 15.6 25.4
Table 1 : ECG changes in mice lacking the Trα-1. Mice 3 and 4 were lacking the Trα-1
mice 1 and 2 were control mice. All values are time in msec. All data were obtained an
averaged during day 2 after starting the data collection.
The results of these experiments show that mice lacking the Trα-1 had lower heart rate an moderate prolongation of QTend and QTpeak. This might indicate disturbances in cardiac
function in these animals.
Treatment of many diseases associated with thyroid hormone function can today not be do since administration of increased doses of the hormone, to achive a desired effect in a giv
tissue, leads to adverse effects in another. The effects of thyroid hormones are mediated b two different receptors that are coexpressed in some tissues, whereas other tissues express
one of them. It should therefore be possible to design agonists and antagonists that are spe for each of the receptors and that can mediate a desired activation or repression of recepto
O:\SP\JPD\KB55PCT.WPD 32 function. The mice and their tissues and cells described above are suitable for testing the specifity of the effects of agonists and antagonists of thyroid hormone action. In addition, effects of thyroid hormones that are mediated via pathways other than those that employ the
TRs ("extrareceptor effects") can be characterized. The mice are also suitable for providing
"proof of action" for agonists and antagonists TR action.
In particular, the mice or cells derived from them may be used to study:
1. Effect on hypercholesterolemia. Administration of excess thyroid hormones decreases
high serum cholesterol levels. However, an adverse side effect is that cardiac output also increases which can lead to arrythmia. If these 2 functions of thyroid hormones are
mediated by distinct receptors, a proper administration of receptor specific agonists or
antagonists would lead to the desired decrease in serum cholesterol while leaving cardiac
function normal. In addition, the role of components of extrareceptor effects (defined below) can be studied and exploited.
2. Hypo- and hyperthyroidism adversely affect bone structure. The use of receptor specific thyroid hormone agonist or antagonists for treatment of e.g. hypercholesterolemia or other diseases must therefore include a test for their influence on bone synthesis and
turnover.
3. Regulation of heart functions such as pulse, arrythmia, or myocardiac muscle can be targeted by the use of receptor specific thyroid hormone agonist or antagonists. In addition, the role of components of extrareceptor effects can be studied and exploite
4. Many organs or tissues produce hormones in a thyroid hormone dependent manner. tissues include the hypophysis (producing growth hormone, prolacting, thyroid
stimulating hormone, luteinizing hormone), the hypothalamus (thyrotropin releasing hormone, oxytocin), periferal tissues (insulin growth factor I). The effect of recepto
specific thyroid hormone ago- or agonists on such endocrine systems can be determi with the mice described above.
5. Basal metabolic rate, gluconeogenesis, lipogenesis, lipolysis and thermogenesis are increased in hyperthyroidism and decreased during hypothyroidism. The effect of
receptor- specific thyroid hormone antagonists or agonists on such metabolic proces can be determined with the mice described above. In addition, the role of compone extrareceptor effects can be studied and exploited.
6. Toxic effects of agonists and antagonists on normal and abnormal physiological
metabolic processes. In addition, the role of components of extrareceptor effects ca studied and exploited.
7. Effects on brain or other neuronal function (hearing, peripheral nervous system), as
as effects on embryonal and fetal development of receptor specific thyroid hormone antagonists or agonists on such endocrine systems can be determined with the mice described above.
8. Effects on increasing or decreasing body growth with growth disorders.
9. A large number of genes or gene products are known to be regulated by thyroid hormones. The effects of agonists and antagonists of such systems can be tested before
clinical trials can commence
10. Effect on hemopoesis. Hypothyroid patients are usually anemic.
11. Effects in cancer chemotherapy. Human breast cancers ofthen have an amplified gene for
the cell surface receptor c-erbB2, located on chromosome 17. The human TRα gene is located close to the c-erbB2 gene, and is therefore usually co-amplified. It could
therefore contribute to the emergence of cancer. Although the contribution of the TRα
gene to tumor growth is unclear, the TR deficient mice offer an opportunity to test
antagonists and agonists in tumor chemotherapy
12. The effects of thyroid hormones that are mediated via pathways other than those that
employ the TRs ("extrareceptor effects") can be characterized, and the genes and their encoded proteins that participate in such pathways can be isolated, characterized, and
used for subsequent screening for novel agonists and -antagonists. 13. The mice will be suitable for providing "proof of action" for agonists and antagoni TR action and extrareceptor effects.
REFERENCES
Baniamad, A., Koehne, A. C. and Renkawitz, R. (1992). A transferable silencing domain is present in the thyroid hormone receptor in the v-erba oncogene product and in the retinoic acid
receptor. EMBO J., 11 , 1015-1023.
Barlow, C , Meister, B., Lardelli, M., Lendahl, U. and Vennstrom, B. (1994). Thyroid abnormalities and hepatocellular carcinoma in mice transgenic for v-erbA. EMBO J., 13, 4241-
4250.
Beckingham-Smith H. and Tata, J (1976). The hormonal control of amphibian metamorphosis.
Blackwell, Oxford.
Blondeau, J. P., Osty, J. and Francon, J. (1988). Characterization of the thyroid hormone
transport system of isolated hepatocytes. J. Biol. Chem., 263, 2685-2692.
Brinster, R. L. , Chen, H. Y., Trumbauer, M. W. and Avarbock, M. R. (1980). Translation of globin MRNA by the mouse ovum. Nature, 283, 499-501.
Burnside, J., Darling, D. S., Carr, F. E. and Chin, W. W. (1989). 'Thyroid hormone
regulation of the rat glycoprotein hormone (x-subunit gene promoter activity. J. Biol. Chem. ,
264, 6886-6991. Chait, A. , Bierman, E. L., and Albers, J. J. (1979). Regulatory role of triiodothyronine i degradation of low density lipoprotein by cultured human skin fibroblasts. J. Clin. Endoc
Metab., 48, 887-889.
Chatterjee, V. K. K. , Lee, J-K., Rentoumis, A. and Jameson, J. L. (1989). Negative
regulation of the thyroid stimulating hormone α gene by thyroid hormone: receptor intera
adjacent to the TATA box. Flroc. Natl. Acad. Sci. U.S.A., 86, 1994-1998.
Crone, D. E. , Kim, H-S. and Spindler, S. R. (1990). α and β thyroid hormone receptors
inunediately adjacent to the rat growth hormone gene TATA box in a negatively hormone
responsive promoter region. J. Biol. Chem., 265, 10851-10856.
Evans, R. M. (1988). 'The steroid and thyroid hormone receptor superfamily. Science, 2
889-895.
Figueiredo, B. , Almazan, G. M., Tetzlaff, W., Miller, F. D. and Cuello, A. C. (1993). expression in the developing cerebellum during perinatal hypo- and hyperthyroidism. Brai
Res. Mol. Brain Res. , 17, 258-68.
Foger, K. R., Wong, E. A. , Wahl, G. and Capecchi, M. R. (1982). Patterns of integratio DNA microinjected into cultured mammalian cells: evidence for homologous recombinati
between injected plasmid DNA molecules. Mol. Cell. Biol. 2, 1372-1387. Forrest, D. (1994). The erb A/ thyroid hormone receptor genes in development of the central nervous system. Sem. Cancer Biol., 5, 167-176.
Galton, V. H. (1983). Thyroid hormone action in amphibian metamorphosis. In:
Oppenheimer, J. H. and Samuels, H. H. (eds.) Molecular basis of thyroid hormone action.
Academic Press, New York, New York, 445-483.
Gilbert, L. I. and Frieden, E. (1991). Metamorphosis: a problem in developmental biology,
2nd ed. Plenum Press, New York, New York.
Glass, C. K. and Holloway, J. M. (1990). Regulation of gene expression by the thyroid hormone receptor. BBA reviews, 1032, 157.
Gordon, J. W., Scangos, G. A., Plotkin, D. J., Barbosa, J. A. and Ruddle, F. H. (1980). Genetic transformation of mouse embryos by microinjection of purified DNA. Proc. Natl.
Acad. Sci. U.S.A., 77, 7380-7384.
Graupner, G., Wills, K. N., Tzukerman, M., Zhang, K-K and Pfahl, M. (1989). Dual regulatory role for thyroid hormone receptors allows control of rednoic acid receptor activity.
Nature, 340, 653-656.
Hanahan, D. (1989). Transgenic mice as probes into complex systems. Science, 246, 1265- 1275.
Handyside, A.H., O'Neill, G.T. , Jones, M. , and Hooper, M.L. (1989). Roux Arch Dev
198, 48-55.
Hauser, P., Zametkin, A. J., Mardnez, P., Vitiello, B., Matochik, J. A., Nfixson, A. J.
Weintraub, B. D. (1993). Attention deficit hyperactivity disorder in patients with generali
resistance to thyroid hormone. NEJM. , 328, 997-1001.
Hodin, R. A., Lazar, M. A., Wintman, B. I., Darling, D. S., Koenig, R. J. , Larsen, P.
Moore, D. D. and Chin, W. W. (1989). Identification of a thyroid hormone receptor that pituitary specific. Science, 244, 76-79.
Hodin, R. A., Lazar, M. A. and Chin, W. W. (1990). Differential and tissue-specific
regulation of multiple rat c-erba messenger RNA species by thyroid hormone. J. Clin. Inv
85, 101-105.
Hogan, B., Beddington, R. , Costantini, F. and Lacy, E. (1994) 2nd ed. Manipulating the Mouse Embryo. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York.
Izumo, S. and Mahdavi, V. (1988). Thyroid hormone receptor α isoforins generated by
alternative splicing differentially activate myosin HC gene transcription. Namre, 334, 539 Jannini, E. A., Dolci, S., Ulisse, S. and Nikodem, V. M. (1994). Developmental regulation of
the thyroid hormone receptor alpha- 1 MRNA expression in the rat testis. Mol. Endocrinol. , 8, 89-96.
Joyner, A. L. (1993). Gene targeting: A practical approach. IRL press at Oxford University
Press, England.
Klein, I. (1990) 'Thyroid hormone and the cardiovascular system. Am. J. Med., 88, 631-637.
Koenig, R. J. , Lazar, M. A., Hodin, R. A., et. al., (1989). Inhibition of thyroid hormone
action by a non-hormone binding c-erba protein generated by alternate niRNA splicing.
Nature, 337, 659-661.
Kohrle, J., Hexch, R. D., Leonard, J. L. (1991) Intracellular pathways of thyroid hormone
metabolism. In: Braverinen, L. E. and Utiger, R. D. (ed.) Werner and Ingbar's The thyroid
gland. J.B. Lippincott Co., 5th ed., Philadelphia, Pennsylvania.
Krenning, E. P., Docter., R. Visser, T. J., et. al., (1983). Plasma membrane transport of
thyroid hormone: its possible pathophysiological significance. J. Endocrinol. Invest., 6, 59-66.
LaCasse, E. C, Lochnan, H. A., Walker, P. and Lefebvre, Y. A. (1993).
Identification of binding proteins for nuclear localisation signals of the glucocorticoid and thyroid hormone receptors. Endocrinology, 132, 1017-1025
Larsen, P. R. , and Ingbar, S. J. (1992) The thyroid gland. In: Wilson, J. D. and Foster, D.W. (eds.) Williams Textbook of Endocidnology, Sth ed., W. B. Saunders
Company, Philadelphia, Pennsylvania.
Laudet, V., Begue, A. , Henry-Duthoit, C, Joubel, A., Martin, P. , Stehelin, D. and Saule, S. (1991) Genomic organization of the human thyroid hormone receptor α (c-e 1) gene. Nucl. Acids Res., 19, 1105-1112
Laudet, V. Hanni, C, Coil, J., Catzefiis, F. and Stehelin, D. (1992) Evolution of the nuclear receptor gene superfamily. EMBO J. , 11, 1003-1013.
Lompre, A. M., Nadal-Ginard, B. and Mahdavi, V. (1984). Expression of the cardiac ventricular α- and β-myosin heavy chain genes is developmentally and
hormonally regulated. J. Biol. Chem., 259, 6437-6446.
McCIaren, A. and Biggers, J. D. (1958). Successful development and birth of n-dee cultivated in vitro as early embryos. Namre, 182, 877-878.
Mendel, C, Weiseger, R. A. and Cavalieri, R. R. (1988). Uptake of 3, 5,
3'-iodothyronine by the perfused rat liver, a return to the free hormone hypothesis. Endocrinol., 123, 1817- 1824.
Mitsuhashi, T., Tennyson, G. E. and Nikodem, V. M. (1989). Alternative splicing generates
messages encoding rat c-erba proteins that do not bind thyroid hormone. Flroc. Natl. Acad.
Sci. U.S.A., 85, 5804-5808.
Muller, W., Kuhn, R. , and Rajewsky, K. (1991). Eur. J. Immunol., 21, 921-95.
Refetoff, S., DeWind, L. T. and DeGroot, L. (1967). Familial syndrome combining deaf mutism, stippled epiphyses, goiter, and abnormally high PBI: possible target organ
refractoriness to thyroid hormone. J Clin. Endc)crinol. Metab., 27, 279-294.
Rodriguez-Pena, A., Ibarrola, N., Iniguez, M. A., Munoz, A. and Bemal J. (1993). Neonatal hypothyroidism affects the timely expression of myelin-associated glycoprotein in the rat brain.
J. Clin. Invest., 91, 812-818.
Sambrook, J., Frisch, E.F. and Maniatis, T. (1989). Molecular Cloning: A Laboratory
Manual. Cold Spring Harbor Laboratory Press, Cold Spring Harbor.
Sap, J., Munoz, A., Damm, K., Goldberg, Y., Ghysdael, J., Leutz, A., Beug, H. and
Vennstrom, B. (1986). The c-erb-A protein is a high-affinity receptor for thyroid hormone.
Namre, 324, 635-640. Schwartz, H. L. (1983). Effect of thyroid hormone in growth and development. In:
Oppenheimer, J. H. and Samuels, H. H. (eds.), Molecular basis of thyroid hormone acti New York: Academic Press, 413-444.
Schwartz, H. L., Lazar, M. A. and Oppenheimer, J. H. (1994). Widespread distribution
immunoreactive thyroid hormone β2 receptor (TRβ2) in the nuclei of extrapimitary rat tis
J. Biol. Chem., 269, 24777-24782.
Segerson, T. P., Kauer, J., Wolfe, H. C, Mostake, H. , Wu, P. , Jackson, M. D. and Le
R. M. (1987). Thyroid hormone regulates TRH biosynthesis in the para ventricular nucleu the rat hypothalamus. Science, 238, 78-80.
Sestoft, L. (1980). Metabolic aspects of the calorigenic effect of thyroid hormone in mam
Clin. Endocrinol., 13, 489-506.
Sjoberg, M., Vennstrom, B., and Forrest, D. (1992). Thyroid hormone receptors in chic
retinal development: differential expression of mRNAs for α and β N-temiinal variant
receptors. Develop. , 114, 39-47.
Sjoberg, M. (1994). Expression and function of chicken thyroid hormone receptors. (The
Karolinska Institute, Stockholm, Sweden. Strait, K. A., Schwartz, H. L., Perez-Castillo, A. and Oppenheimer, J. H. (1990).
Quantitation of erba (x and P expression in developing and adult rat tissues under the influence of thyroidal stams and comparison with nuclear binding capacity. J. Biol. Chem., 265, 10514-
10521.
Takeda, K., Sakurai, A., DeGroot, L. J. and Refetoff, S. (1992). Recessive inheritance of thyroid hormone resistance caused by complete deletion of the protein-coding region of the
thyroid hormone receptor-beta gene. J. Clin. Endociinol. Metab., 74, 49-55.
Tasset, D., Tora, L., Fromental, C, Scheer, E. and Chambon, P. (1990). Distinct classes of
transcriptional activating domains function by different mechanisms. Cell, 62, 1177-1187.
Thompson, K. L., Santon, J. B. , Shephard, L. B., Walton, G. M. and Gill, G. N. (1992). A nuclear protein is required for thyroid hormone receptor binding to an inhibitory half-site in the epidermal growth factor receptor promoter. Mol. Endocrinol., 6, 627-635.
Towle, H. C, Mariash, C. N. Oppenheimer, J. H. (1980). Changes in hepatic levels of messenger ribonucleic acid for make enzyme during induction by thyroid hormone or diet.
Biochemistry, 19, 579-585.
Towle, H. C, Mariash, C. N. Schwartz, H. L., Oppenheimer, J. H. (1981). Quantitation of rat liver messenger ribonucleic acid for malic enzyme during induction by thyroid hormone. Biochemistry, 20, 3486-3492.
Usala, S. (1994). llyroid hormone resistance. In: Weintraub, B. D. (ed.), Molecular Endocrinology. Raven Press, New York, 393-409.
Vennstrom, B. and Bishop, J. M. (1982). Isolation and characterization of chicken DNA
homologous to the two putative oncogenes of avian erythroblastosis virus. Cell, 1, 135-1
Weinberger, C, Thompson, C. C , Ong, E. S. , Lebo, R., Gruol, D. J. and Evans, R. M
(1986). 'The c-erba gene encodes a thyroid hormone receptor. Namre, 324, 641-646.
Whitten, W. K. (1956). Culture of tubal mouse ova. Namre, 177, 96.
Whitten, W. K. and Biggers, J. D. (1968). Complete development in vitro of the
preimplantation stages of the mouse in a simple chemically defined medium. J. Reprod. Fertil., 17, 399^.01.
Williams, G. R. and Brent, G. A. (1995). 'Ryroid Hormone Response Elements, In: Weintraub, B., (ed.), Molecular Endocrinology. Raven Press, New York, New York, 21
248.
Wondisford, F. E., Steinfelder, H. J., Nations, M. and Radovick, S. (1993). AP- 1 antagonizes thyroid hormone receptor action on the thyrotropin P-subunit gene. J. Biol.
Chem., 268, 2749-2754.
Wood, W. M., Kao, M. Y., Gordon, D. F. and Ridgeway, E. C. (1989). Thyroid hormone regulates the mouse thyrotropin P-subunit gene promoter in transfected primary thyrotropes. J.
Biol. Chem., 264, 14840-14847.
Yaffe, B. M. and Samuels, H. H. (1984). Hon-nonal regulation of the growth hormone gene. J. Biol. Chem., 259, 6284-6291.

Claims

1. A transgenic animal which is heterozygous for an at least partially defective thyroid hormone receptor gene.
2. A transgenic animal according to claim 1 in which the defective gene has been
inactivated by an insertion, deletion, substimtion or inversion or other suitable gene
manipulation.
3. A transgenic animal according to claim 1 or 2 which is a rodent.
4. A transgenic animal according to claim 3 which is a mouse.
5. A transgenic animal according to claim 1, 2, 3 or 4 in which the defective thyroid hormone receptor gene is the TRα gene.
6. A transgenic animal according to claim 5 in which the defective thyroid hormone receptor gene is the TRα l gene.
7. A transgenic animal according to claim 6 which has a functional TRα2 gene.
8. Cells derived from a transgenic animal according to any preceding claim which are
heterozygous or homozygous for a defective thyroid hormone receptor gene.
9. Cells according to claim 8 in which the thyroid hormone receptor gene is the TRα l gene.
10. Cells according to claim 9 which have a functional TRα2 gene.
11. A method of producing a transgenic animal in accordance with any one of claims 1 to 8
the method comprising :
1) preparing a gene encoding an at least partially defective thyroid hormone receptor
as described above;
2) introducing that gene into suitable carrier cells;
3) inserting those carrier cells into an embryo; and
4) replacing the embryo into a mother animal, and allowing the embryo to develop
to full term.
12. A method according to claim 11 in which the thyroid hormone receptor gene is the TRα gene.
13. A method according to claim 12 in which the defective thyroid hormone receptor gene is the TRαl gene.
14. A method of testing the agonist/antagonist properties of a compound in relation to the
thyroid hormone receptor, the method comprising contacting a transgenic animal in accordance with any one of claims 1 to 7 with the compound and monitoring subse development of the animal.
15. A method of testing the agonist/antagonist properties of a compound in relation to t
thyroid hormone receptor, the method comprising contacting cells or tissues deriv
from a transgenic animal in accordance with any one of claims 1 to 10.
16. A method according to claim 14 or 15 in which the animal or cells have a defective
gene.
17. A method according to claim 16 in which the animal has a functional Reα2 gene.
PCT/EP1996/000161 1995-01-13 1996-01-15 Transgenic animals having a defective thyroid hormone receptor gene WO1996021728A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU47851/96A AU4785196A (en) 1995-01-13 1996-01-15 Transgenic animals having a defective thyroid hormone receptor gene

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB9500683.9 1995-01-13
GBGB9500683.9A GB9500683D0 (en) 1995-01-13 1995-01-13 Transgenic animals

Publications (2)

Publication Number Publication Date
WO1996021728A2 true WO1996021728A2 (en) 1996-07-18
WO1996021728A3 WO1996021728A3 (en) 1996-09-06

Family

ID=10768005

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP1996/000161 WO1996021728A2 (en) 1995-01-13 1996-01-15 Transgenic animals having a defective thyroid hormone receptor gene

Country Status (3)

Country Link
AU (1) AU4785196A (en)
GB (1) GB9500683D0 (en)
WO (1) WO1996021728A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998046748A1 (en) * 1997-04-11 1998-10-22 Millennium Pharmaceuticals Inc. Therapeutic compositions and diagnostic assays for diseases involving trbp

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995030741A1 (en) * 1994-05-10 1995-11-16 The Salk Institute For Biological Studies Receptor-deficient animals and cell lines derived therefrom, and uses thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995030741A1 (en) * 1994-05-10 1995-11-16 The Salk Institute For Biological Studies Receptor-deficient animals and cell lines derived therefrom, and uses thereof

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
CLINICAL RESEARCH, vol. 42, no. 2, April 1994, page pa209 XP002007168 SAFER J. ET AL.: "Thyroid hormone resistance in transgenic animals containing mutant human thyroid hormone receptors" *
NATURE, vol. 374, 9 March 1995, pages 159-162, XP002007174 SAITOU, M. ET AL.: "Inhibition of skin development by targeted expression of a dominant-negative retinoic acid receptor" *
NUCLEIC ACIDS RESEARCH, vol. 19, no. 5, 1991, pages 1105-1112, XP002007172 LAUDET, V. ET AL.: "Genomic organization of the human thyroid hormone receptor alpha (c-erbA-1) gene" *
PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 90, 15 February 1993, pages 1590-1594, XP002007169 LI, E. ET AL.: "Normal development and growth of mice carrying a targeted disruption of the alpha1 retinoic acid receptor gene" *
PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 90, August 1993, pages 7225-7229, XP002007170 LUFKIN, T. ET AL.: "High postnatal lethality and testis degeneration in retinoic acid receptor alpha mutant mice" *
SCIENCE, vol. 240, 1988, pages 889-895, XP002007173 EVANS, R. J.: "The steroid and thyroid hormone receptor superfamily" cited in the application *
SEMINARS IN CANCER BIOLOGY, vol. 5, 1994, pages 167-176, XP002007171 FORREST, D.: "The erbA/thyroid hormone receptor genes in development of the central nervous system" cited in the application *
THE EMBO JOURNAL, vol. 13, no. 18, 15 September 1994, pages 4241-4250, XP002007167 BARLOW, C. ET AL : "Thyroid abnormalities and hepatocellular carcinoma in mice transgenic for v-erbA" *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998046748A1 (en) * 1997-04-11 1998-10-22 Millennium Pharmaceuticals Inc. Therapeutic compositions and diagnostic assays for diseases involving trbp
US6037173A (en) * 1997-04-11 2000-03-14 Millennium Pharmaceuticals, Inc. Isolated nucleic acid encoding TRBP

Also Published As

Publication number Publication date
WO1996021728A3 (en) 1996-09-06
GB9500683D0 (en) 1995-03-08
AU4785196A (en) 1996-07-31

Similar Documents

Publication Publication Date Title
Zhang et al. Inactivation of the myogenic bHLH gene MRF4 results in up-regulation of myogenin and rib anomalies.
Charles et al. Pituitary-specific Gata2 knockout: effects on gonadotrope and thyrotrope function
Matzuk et al. Functional analysis of activins during mammalian development
Hoser et al. Sox12 deletion in the mouse reveals nonreciprocal redundancy with the related Sox4 and Sox11 transcription factors
Li et al. Gsh‐4 encodes a LIM‐type homeodomain, is expressed in the developing central nervous system and is required for early postnatal survival.
Sadovsky et al. Mice deficient in the orphan receptor steroidogenic factor 1 lack adrenal glands and gonads but express P450 side-chain-cleavage enzyme in the placenta and have normal embryonic serum levels of corticosteroids.
Cretekos et al. Regulatory divergence modifies limb length between mammals
Supp et al. Sp4, a member of the Sp1-family of zinc finger transcription factors, is required for normal murine growth, viability, and male fertility
Geiman et al. Lsh, a SNF2 family member, is required for normal murine development
US5698766A (en) Transgenic animal model for testing drugs for treating eating disorders and epilepsy
Baur et al. Specificity protein 2 (Sp2) is essential for mouse development and autonomous proliferation of mouse embryonic fibroblasts
Löf et al. Genetically modified mouse models to investigate thyroid development, function and growth
US5858774A (en) Antisense DNA constructs for expression of hybrid MRNAs driven by inducible, tissue-specific promoters
JP2000516212A (en) Galanin
JP2003513645A (en) Melanocortin-3 receptor deficient cells, non-human transgenic animals and methods for selecting compounds that regulate body weight
JP2006506968A (en) Agouti-related protein-deficient cells, non-human transgenic animals, and methods for selecting compounds that modulate energy metabolism
US20020104107A1 (en) Point mutant mice with hypersensitive alpha 4 nicotinic receptors: dompaminergic pathology and increased anxiety
WO1996021728A2 (en) Transgenic animals having a defective thyroid hormone receptor gene
US6080911A (en) Mice models of growth hormone insensitivity
Mysliwiec et al. Generation of a conditional null allele of jumonji
WO2007058986A2 (en) Methods for diagnosing and identifying effective therapeutics for major depressive disorder
US20060212953A1 (en) Tr2, tr4, tr2/tr4 double knockouts and uses thereof
US8835711B2 (en) ANF-RGC in-gene knock-out animal
JP2000510001A (en) Transgenic animal having a disrupted NPY Y1 receptor gene
US20220386574A1 (en) P21-cre animal models

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AU CA JP KR US

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE

AK Designated states

Kind code of ref document: A3

Designated state(s): AU CA JP KR US

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase