WO2004020666A2 - Diagnostic and therapeutic use of thyroid hormone binding protein for neurodegenerative diseases - Google Patents

Diagnostic and therapeutic use of thyroid hormone binding protein for neurodegenerative diseases Download PDF

Info

Publication number
WO2004020666A2
WO2004020666A2 PCT/EP2003/009536 EP0309536W WO2004020666A2 WO 2004020666 A2 WO2004020666 A2 WO 2004020666A2 EP 0309536 W EP0309536 W EP 0309536W WO 2004020666 A2 WO2004020666 A2 WO 2004020666A2
Authority
WO
WIPO (PCT)
Prior art keywords
thbp
disease
gene coding
activity
level
Prior art date
Application number
PCT/EP2003/009536
Other languages
French (fr)
Other versions
WO2004020666A3 (en
Inventor
Heinz Von Der Kammer
Johannes Pohlner
Original Assignee
Evotec Neurosciences Gmbh
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 Evotec Neurosciences Gmbh filed Critical Evotec Neurosciences Gmbh
Priority to AU2003271568A priority Critical patent/AU2003271568A1/en
Priority to EP03753364A priority patent/EP1534856A2/en
Publication of WO2004020666A2 publication Critical patent/WO2004020666A2/en
Publication of WO2004020666A3 publication Critical patent/WO2004020666A3/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • G01N33/6896Neurological disorders, e.g. Alzheimer's disease
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/74Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving hormones or other non-cytokine intercellular protein regulatory factors such as growth factors, including receptors to hormones and growth factors
    • G01N33/78Thyroid gland hormones, e.g. T3, T4, TBH, TBG or their receptors
    • 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
    • A01K2217/072Animals genetically altered by homologous recombination maintaining or altering function, i.e. knock in
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/02Screening involving studying the effect of compounds C on the interaction between interacting molecules A and B (e.g. A = enzyme and B = substrate for A, or A = receptor and B = ligand for the receptor)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/28Neurological disorders
    • G01N2800/2814Dementia; Cognitive disorders
    • G01N2800/2821Alzheimer

Definitions

  • the present invention relates to methods of diagnosing, prognosticating and monitoring the progression of neurodegenerative diseases in a subject. Furthermore, methods of therapy control and screening for modulating agents of neurodegenerative diseases are provided. The invention also discloses pharmaceutical compositions, kits, and recombinant animal models.
  • AD Alzheimer's disease
  • AD Alzheimer's disease
  • these diseases constitute an enormous health, social, and economic burden.
  • AD is the most common neurodegenerative disease, accounting for about 70% of all dementia cases, and it is probably the most devastating age-related neurodegenerative condition affecting about 10% of the population over 65 years of age and up to 45% over age 85 (for a recent review see Vickers et al., Progress in Neurobiology 2000, 60: 139-165).
  • AD Alzheimer's disease
  • amyloid- ⁇ (A ⁇ ) protein evolves from the cleavage of the amyloid precursor protein (APP) by different kinds of proteases.
  • the cleavage by the ⁇ / ⁇ -secretase leads to the formation of A ⁇ peptides of different lengths, typically a short more soluble and slow aggregating peptide consisting of 40 amino acids and a longer 42 amino acid peptide, which rapidly aggregates outside the cells, forming the characteristic amyloid plaques (Selkoe, Physiological Rev 2001 , 81 : 741-66; Greenfield et al., Frontiers Bioscience 2000, 5: D72-83).
  • AD patients Two types of plaques, diffuse plaques and neuritic plaques, can be detected in the brain of AD patients, the latter ones being the classical, most prevalent type. They are primarily found in the cerebral cortex and hippocampus.
  • the neuritic plaques have a diameter of 50 ⁇ m to 200 ⁇ m and are composed of insoluble fibrillar amyloids, fragments of dead neurons, of microglia and astrocytes, and other components such as neurotransmitters, apolipoprotein E, glycosaminoglycans, ⁇ 1-antichymotrypsin and others.
  • the generation of toxic A ⁇ deposits in the brain starts very early in the course of AD, and it is discussed to be a key player for the subsequent destructive processes leading to AD pathology.
  • NFTs neurofibrillary tangles
  • abnormal neurites described as neuropil threads
  • a loss of neurons can be observed. It is discussed that said neuron loss may be due to a damaged microtubule-associated transport system (Johnson and Jenkins, J Alzheimers Dis 1996, 1 : 38-58; Johnson and Hartigan, J Alzheimers Dis 1999, 1 : 329-351 ).
  • AD neurofibrillary tangles and their increasing number correlates well with the clinical severity of AD (Schmitt et al., Neurology 2000, 55: 370-376).
  • AD is a progressive disease that is associated with early deficits in memory formation and ultimately leads to the complete erosion of higher cognitive function.
  • the cognitive disturbances include among other things memory impairment, aphasia, agnosia and the loss of executive functioning.
  • a characteristic feature of the pathogenesis of AD is the selective vulnerability of particular brain regions and subpopulations of nerve cells to the degenerative process. Specifically, the temporal lobe region and the hippocampus are affected early and more severely during the progression of the disease.
  • AD Alzheimer's disease
  • AD apolipoprotein E gene
  • Thyroid hormones mediate their pleiotropic effects on tissues and cells via nuclear thyroid hormone receptors (TRalpha and TRbeta) that act as transcription factors. These receptors belong to a large superfamily of nuclear hormone receptors including the receptors for steroid hormones, retinoids, and vitamin D. Steroid hormone receptors shuttle between the nucleus and the cytoplasm, where they interact with their appropriate ligand/agonist (for review: Chawla et al., Science 2001 , 294: 1866-1870). In contrast, the retinoid and thyroid hormone receptors remain permanently bound to their genomic target sequence elements. The function of intracellular shuttling and homeostasis of retinoids and thyroid hormones is exerted by specialized cytoplasmic binding proteins.
  • the thyroid hormone receptors form obligate heterodimers with the retinoid X receptor (Chawla et al., Science 2001 , 294: 1866-1870; Wu et al., J. Biol. Chem. 2002, 277: 8898-8905).
  • T3 thyroid hormone triiodothyronine
  • a 58 kDa monomer of the ubiquitous enzyme pyruvate kinase has been described as a T3-binding protein in human cells (Kato et al., Proc. Natl. Acad. Sci. U.S.A. 1989, 86: 7861-7865).
  • T3 binding to this protein is allosterically effected by the glycolytic intermediate fructose-1 ,6- bisphosphate and impacts on overall transcriptional regulation by the thyroid hormone (Ashizawa et al., Biochemistry 1991 , 30: 7105-7111).
  • T3-binding protein In Xenopus species, a 59 kDa subunit of the aldehyde dehydrogenease has been described as a T3- binding protein (Yamauchi et al., J. Biol. Chem. 1999, 274: 8460-8469). Yet other T3-binding proteins were isolated from rat kidney and liver cytoplasm: a 58 kDa protein that regulates the access of T3 to its nuclear receptor (Hashizume et al., J. Biol. Chem.
  • THBP NADPH-dependent thyroid hormone binding protein
  • THBP is abundantly expressed in neural tissue including brain, muscle, heart and kidney, to lower levels in lung and liver, and there is none detectable in placenta and pancreas (Kim et al., Proc. Natl. Acad. Sci. U. S.A. 1992, 89: 9292- 9296).
  • THBP is responsible for most of the intracellular high-affinity (nano olar) T3 and T4 (thyroxine) binding in kidney from rat (Vie et al., Mol. Endocrinol. 1997, 1 1 : 1728-1736).
  • mu-crystallin/THBP was detected in photoreceptors of the mammalian retina (Wistow and Segovia, Invest. Ophthalmol. Visual Sci. 1994, 35:2073; Sperbeck et al., Mol. Biol. Cell 1997, 8:312A; Segovia et al., Mol. Vis. 1997, 3:9).
  • a number of differential gene expression screens have identified mu- crystallin/THBP, including screens in the field of mouse hair follicle development (Aoki et al., J. Invest. Dermatol. 2000, 1 15: 402-405), screens of a hormone- refractory prostate cancer xenograft model (Mousses et al., Cancer Res.
  • the present invention discloses the detection and differential regulation of thyroid hormone binding protein (THBP) gene expression in the temporal cortex versus frontal cortex of Alzheimer's disease brain in comparison to age-matched control tissues. This finding can be put in context with a number of pathophysiologic roles for THBP. Firstly, like its bacterial homologs lysine and ornithine cyclodeaminase, THBP may be involved in synthesis or breakdown of amino acids (Kim et al., Proc. Natl. Acad. Sci. U.S.A. 1992, 89: 9292-9296). For instance, levels of the amino acid glutamate, which possesses a general excitatory role in the central nervous system, may be affected.
  • THBP thyroid hormone binding protein
  • thyroid hormone signaling may affect the plasticity of the adult brain including the particularly vulnerable hippocampus formation (for review, Calza et al., Brain Res. Bull. 1997, 44: 549-557). As such, a differential regulation of THBP gene expression may well be a cause of neurodegenerative diseases.
  • thyroid hormones were shown to negatively regulate the transcriptional activity of the amyloid precursor protein (APP) in recombinant neuroblastoma cells (Belandia et al., J. Biol. Chem.
  • level as used herein is meant to comprise a gage of, or a measure of the amount of, or a concentration of a transcription product, for instance an mRNA, or a translation product, for instance a protein or polypeptide.
  • activity shall be understood as a measure for the ability of a transcription product or a translation product to produce a biological effect or a measure for a level of biologically active molecules.
  • activity also refers to enzymatic activity.
  • level and/or “activity” as used herein further refer to gene expression levels or gene activity. Gene expression can be defined as the utilization of the information contained in a gene by transcription and translation leading to the production of a gene product.
  • “Dysregulation” shall mean an upregulation or downregulation of gene expression.
  • a gene product comprises either RNA or protein and is the result of expression of a gene. The amount of a gene product can be used to measure how active a gene is.
  • the term "gene” as used in the present specification and in the claims comprises both coding regions (exons) as well as non-coding regions (e.g. non-coding regulatory elements such as promoters or enhancers, introns, leader and trailer sequences).
  • the term “ORF” is an acronym for "open reading frame” and refers to a nucleic acid sequence that does not possess a stop codon in at least one reading frame and therefore can potentially be translated into a sequence of amino acids.
  • regulatory elements shall comprise inducible and non-inducible promoters, enhancers, operators, and other elements that drive and regulate gene expression.
  • fragment as used herein is meant to comprise e.g. an alternatively spliced, or truncated, or otherwise cleaved transcription product or translation product.
  • derivative as used herein refers to a mutant, or an RNA-edited, or a chemically modified, or otherwise altered transcription product, or to a mutant, or chemically modified, or otherwise altered translation product.
  • a “derivative” may be generated by processes such as altered phosphorylation, or glycosylation, or acetylation, or lipidation, or by altered signal peptide cleavage or other types of maturation cleavage. These processes may occur post-translationally.
  • the term "modulator” as used in the present invention and in the claims refers to a molecule capable of changing or altering the level and/or the activity of a gene, or a transcription product of a gene, or a translation product of a gene.
  • a “modulator” is capable of changing or altering the biological activity of a transcription product or a translation product of a gene.
  • Said modulation may be an increase or a decrease in enzyme activity, a change in binding characteristics, or any other change or alteration in the biological, functional, or immunological properties of said translation product of a gene.
  • agent refers to any substance, chemical, composition, or extract that have a positive or negative biological effect on a cell, tissue, body fluid, or within the context of any biological system, or any assay system examined. They can be agonists, antagonists, partial agonists or inverse agonists of a target.
  • agents, reagents, or compounds may be nucleic acids, natural or synthetic peptides or protein complexes, or fusion proteins.
  • oligonucleotide primer or “primer” refer to short nucleic acid sequences which can anneal to a given target polynucleotide by hybridization of the complementary base pairs and can be extended by a polymerase. They may be chosen to be specific to a particular sequence or they may be randomly selected, e.g. they will prime all possible sequences in a mix. The length of primers used herein may vary from 10 nucleotides to 80 nucleotides.
  • Probes are short nucleic acid sequences of the nucleic acid sequences described and disclosed herein or sequences complementary therewith. They may comprise full length sequences, or fragments, derivatives, isoforms, or variants of a given sequence. The identification of hybridization complexes between a "probe” and an assayed sample allows the detection of the presence of other similar sequences within that sample.
  • homolog or homology is a term used in the art to describe the relatedness of a nucieotide or peptide sequence to another nucieotide or peptide sequence, which is determined by the degree of identity and/or similarity between said sequences compared.
  • variant refers to any polypeptide or protein, in reference to polypeptides and proteins disclosed in the present invention, in which one or more amino acids are added and/or substituted and/or deleted and/or inserted at the N-terminus, and/or the C-terminus, and/or within the native amino acid sequences of the native polypeptides or proteins of the present invention.
  • variant shall include any shorter or longer version of a polypeptide or protein.
  • “Variants” shall also comprise a sequence that has at least about 80% sequence identity, more preferably at least about 90% sequence identity, and most preferably at least about 95% sequence identity with the amino acid sequence of thyroid hormone binding protein, SEQ ID NO. 2.
  • Proteins and polypeptides of the present invention include variants, fragments and chemical derivatives of the protein comprising the amino acid sequences of thyroid hormone binding protein, SEQ ID NO. 2. They can include proteins and polypeptides which can be isolated from nature or be produced by recombinant and/or synthetic means. Native proteins or polypeptides refer to naturally-occurring truncated or secreted forms, naturally occurring variant forms (e.g. splice-variants) and naturally occurring allelic variants.
  • isolated as used herein is considered to refer to molecules that are removed from their natural environment, i.e.
  • sequences encoding such molecules can be linked by the hand of man to polynucleotides, to which they are not linked in their natural state, and that such molecules can be produced by recombinant and/or synthetic means. Even if for said purposes those sequences may be introduced into living or non-living organisms by methods known to those skilled in the art, and even if those sequences are still present in said organisms, they are still considered to be isolated.
  • the terms "risk”, “susceptibility”, and “predisposition” are tantamount and are used with respect to the probability of developing a neurodegenerative disease, preferably Alzheimer's disease.
  • the term 'AD' shall mean Alzheimer's disease.
  • AD-type neuropathology refers to neuropathological, neurophysiological, histopathological and clinical hallmarks as described in the instant invention and as commonly known from state- of-the-art literature (see: Iqbal, Swaab, Winblad and Wisniewski, Alzheimer ' s Disease and Related Disorders (Etiology, Pathogenesis and Therapeutics), Wiley & Sons, New York, Weinheim, Toronto, 1999; Scinto and Daffner, Early Diagnosis of Alzheimer ' s Disease, Humana Press, Totowa, New Jersey, 2000; Mayeux and Christen, Epidemiology of Alzheimer ' s Disease: From Gene to Prevention, Springer Press, Berlin, Heidelberg, New York, 1999; Younkin, Tanzi and Christen, Presenilins and Alzheimer ' s Disease, Springer Press, Berlin, Heidelberg, New York, 1998).
  • Neurodegenerative diseases or disorders according to the present invention comprise Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, Pick's disease, fronto-temporal dementia, progressive nuclear palsy, corticobasal degeneration, cerebro-vascular dementia, multiple system atrophy, argyrophilic grain dementia and other tauopathies, and mild- cognitive impairment.
  • Further conditions involving neurodegenerative processes are, for instance, ischemic stroke, age-related macular degeneration, narcolepsy, motor neuron diseases, prion diseases, traumatic nerve injury and repair, and multiple sclerosis.
  • the invention features a method of diagnosing or prognosticating a neurodegenerative disease in a subject, or determining whether a subject is at increased risk of developing said disease.
  • the method comprises: determining a level, or an activity, or both said level and said activity of (i) a transcription product of the gene coding for thyroid hormone binding protein (THBP), and/or of (ii) a translation product of the gene coding for thyroid hormone binding protein (THBP), and/or of (iii) a fragment, or derivative, or variant of said transcription or translation product in a sample from said subject and comparing said level, and/or said activity to a reference value representing a known disease or health status, thereby diagnosing or prognosticating said neurodegenerative disease in said subject, or determining whether said subject is at increased risk of developing said neurodegenerative disease.
  • THBP thyroid hormone binding protein
  • THBP a translation product of the gene coding for thyroid hormone binding protein
  • a fragment, or derivative, or variant of said transcription or translation product in a
  • the invention also relates to the construction and the use of primers and probes which are unique to the nucleic acid sequences, or fragments, or variants thereof, as disclosed in the present invention.
  • the oligonucleotide primers and/or probes can be labeled specifically with fluorescent, bioluminescent, magnetic, or radioactive substances.
  • the invention further relates to the detection and the production of said nucleic acid sequences, or fragments and variants thereof, using said specific oligonucleotide primers in appropriate combinations.
  • PCR-analysis a method well known to those skilled in the art, can be performed with said primer combinations to amplify said gene specific nucleic acid sequences from a sample containing nucleic acids. Such sample may be derived either from healthy or diseased subjects.
  • the invention provides nucleic acid sequences, oligonucleotide primers, and probes of at least 10 bases in length up to the entire coding and gene sequences, useful for the detection of gene mutations and single nucieotide polymorphisms in a given sample comprising nucleic acid sequences to be examined, which may be associated with neurodegenerative diseases, in particular Alzheimer's disease.
  • This feature has utility for developing rapid DNA-based diagnostic tests, preferably also in the format of a kit.
  • the invention features a method of monitoring the progression of a neurodegenerative disease in a subject.
  • a level, or an activity, or both said level and said activity, of (i) a transcription product of the gene coding for thyroid hormone binding protein (THBP), and/or of (ii) a translation product of the gene coding for thyroid hormone binding protein (THBP), and/or of, (iii) a fragment, or derivative, or variant of said transcription or translation product in a sample from said subject is determined.
  • Said level and/or said activity is compared to a reference value representing a known disease or health status. Thereby, the progression of said neurodegenerative disease in said subject is monitored.
  • the invention features a method of evaluating a treatment for a neurodegenerative disease, comprising determining a level, or an activity, or both said level and said activity of (i) a transcription product of the gene coding for thyroid hormone binding protein (THBP), and/or of (ii) a translation product of the gene coding for thyroid hormone binding protein (THBP), and/or of (iii) a fragment, or derivative, or variant of said transcription or translation product in a sample obtained from a subject being treated for said disease. Said level, or said activity, or both said level and said activity are compared to a reference value representing a known disease or health status, thereby evaluating the treatment for said neurodegenerative disease.
  • THBP thyroid hormone binding protein
  • THBP thyroid hormone binding protein
  • said gene coding for thyroid hormone binding protein is the gene coding for NADPH-dependent thyroid hormone binding protein, THBP, also termed mu-crystallin or CRYM (NCBI Genbank accession number: L02950 and M90841 ), represented by SEQ ID NO. 2 and SEQ ID NO. 3 (GenBank accession numbers: Q14894 and U85772).
  • the gene coding for said thyroid hormone binding protein is also generally referred to as the thyroid hormone binding protein gene, or THBP gene, or THBP, and further, said thyroid hormone binding protein is also generally referred to as THBP.
  • said neurodegenerative disease or disorder is Alzheimer's disease, and said subjects suffer from Alzheimer's disease.
  • the present invention discloses the detection, differential expression and regulation of the gene coding for thyroid hormone binding protein (THBP) in specific brain regions of AD patients. Consequently, the thyroid hormone binding protein gene (THBP) and its corresponding transcription and translation products may have a causative role in the regional selective neuronal degeneration typically observed in AD. Alternatively, the gene coding for THBP and its products may confer a neuroprotective function to the remaining surviving nerve cells. Based on these disclosures, the present invention has utility for the diagnostic evaluation and prognosis as well as for the identification of a predisposition to a neurodegenerative disease, in particular AD- Furthermore, the present invention provides methods for the diagnostic monitoring of patients undergoing treatment for such a disease.
  • THBP thyroid hormone binding protein
  • said sample to be analyzed and determined is selected from the group comprising brain tissue or other tissues, or body cells.
  • the sample can also comprise cerebrospinal fluid or other body fluids including saliva, urine, blood, serum plasma, mucus.
  • the methods of diagnosis, prognosis, monitoring the progression or evaluating a treatment for a neurodegenerative disease, according to the instant invention can be practiced ex corpore, and such methods preferably relate to samples, for instance, body fluids or cells, removed, collected, or isolated from a subject or patient.
  • said reference value is that of a level, or an activity, or both said level and said activity of (i) a transcription product of the gene coding for THBP, and/or of (ii) a translation product of the gene coding for THBP, and/or of (iii) a fragment, or derivative, or variant of said transcription or translation product in a sample from a subject not suffering from said neurodegenerative disease.
  • an alteration in the level and/or activity of a transcription product of the gene coding for THBP and/or of a translation product of the gene coding for THBP and/or of a fragment, or derivative, or variant thereof in a sample cell, or tissue, or body fluid from said subject relative to a reference value representing a known health status indicates a diagnosis, or prognosis, or increased risk of becoming diseased with a neurodegenerative disease, particularly AD.
  • measurement of the level of transcription products of the gene coding for THBP is performed in a sample from a subject using a quantitative PCR-analysis with primer combinations to amplify said gene specific sequences from cDNA obtained by reverse transcription of RNA extracted from a sample of a subject.
  • a Northern blot with probes specific for said gene can also be applied. It might further be preferred to measure transcription products by means of chip-based micro-array technologies. These techniques are known to those of ordinary skill in the art (see Sambrook and Russell, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 2001 ; Schena M., Microarray Biochip Technology, Eaton Publishing, Natick, MA, 2000).
  • An example of an immunoassay is the detection and measurement of enzyme activity as disclosed and described in the patent application WO 02/14543.
  • a level and/or an activity of a translation product of the gene coding for THBP and/or of a fragment, or derivative, or variant of said translation product, and/or a level of activity of said translation product and/or of a fragment, or derivative, or variant thereof can be detected using an immunoassay, an activity assay, and/or a binding assay.
  • assays can measure the amount of binding between said protein molecule and an anti-protein antibody by the use of enzymatic, chromodynamic, radioactive, magnetic, or luminescent labels which are attached to either the anti-protein antibody or a secondary antibody which binds the anti-protein antibody.
  • other high affinity ligands may be used.
  • Immunoassays which can be used include e.g. ELISAs, Western blots and other techniques known to those of ordinary skill in the art (see Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1999 and Edwards R, Immunodiagnostics: A Practical Approach, Oxford University Press, Oxford; England, 1999). All these detection techniques may also be employed in the format of microarrays, protein-arrays, antibody microarrays, tissue microarrays, electronic biochip or protein-chip based technologies (see Schena M., Microarray Biochip Technology, Eaton Publishing, Natick, MA, 2000).
  • the level, or the activity, or both said level and said activity of (i) a transcription product of the gene coding for THBP, and/ ⁇ r of (ii) a translation product of the gene coding for THBP, and/or of (iii) a fragment, or derivative, or variant of said transcription or translation product in a series of samples taken from said subject over a period of time is compared, in order to monitor the progression of said disease.
  • said subject receives a treatment prior to one or more of said sample gatherings.
  • said level and/or activity is determined before and after said treatment of said subject.
  • the invention features a kit for diagnosing or prognosticating neurodegenerative diseases, in particular AD, in a subject, or determining the propensity or predisposition of a subject to develop a neurodegenerative disease, in particular AD, said kit comprising:
  • reagents which is selected from the group consisting of (i) reagents that selectively detect a transcription product of the gene coding for THBP (ii) reagents that selectively detect a translation product of the gene coding for THBP; and
  • a neurodegenerative disease in particular AD
  • determining the propensity or predisposition of said subject to develop such a disease wherein a varied level, or activity, or both said level and said activity, of said transcription product and/or said translation product compared to a reference value representing a known health status; or a level, or activity, or both said level and said activity, of said transcription product and/or said translation product similar or equal to a reference value representing a known disease status, indicates a diagnosis or prognosis of a neurodegenerative disease, in particular AD, or an increased propensity or predisposition of developing such a disease.
  • the kit, according to the present invention may be particularly useful for the identification of individuals that are at risk of developing a neurodegenerative disease, in particular AD. Consequently, the kit, according to the present invention, may serve as a means for targeting identified individuals for early preventive measures or therapeutic intervention prior to disease onset, before irreversible damage in the course of the disease has been inflicted. Furthermore, in preferred embodiments, the kit featured in the invention is useful for monitoring a progression of a neurodegenerative disease, in particular AD in a subject, as well as monitoring success or failure of therapeutic treatment for such a disease of said subject.
  • the invention features a method of treating or preventing a neurodegenerative disease, in particular AD, in a subject comprising the administration to said subject in a therapeutically or prophylactically effective amount of an agent or agents which directly or indirectly affect a level, or an activity, or both said level and said activity, of (i) the gene coding for THBP, and/or (ii) a transcription product of the gene coding for THBP, and/or (iii) a translation product of the gene coding for THBP, and/or (iv) a fragment, or derivative, or variant of (i) to (iii).
  • an agent or agents which directly or indirectly affect a level, or an activity, or both said level and said activity, of (i) the gene coding for THBP, and/or (ii) a transcription product of the gene coding for THBP, and/or (iii) a translation product of the gene coding for THBP, and/or (iv) a fragment, or derivative, or variant of (
  • Said agent may comprise a small molecule, or it may also comprise a peptide, an oligopeptide, or a polypeptide.
  • Said peptide, oligopeptide, or polypeptide may comprise an amino acid sequence of a translation product of the gene coding for THBP, or a fragment, or derivative, or a variant thereof.
  • An agent for treating or preventing a neurodegenerative disease, in particular AD, according to the instant invention may also consist of a nucieotide, an oligonucleotide, or a polynucleotide.
  • Said oligonucleotide or polynucleotide may comprise a nucieotide sequence of the gene coding for THBP, either in sense orientation or in antisense orientation.
  • the method comprises the application of per se known methods of gene therapy and/or antisense nucleic acid technology to administer said agent or agents.
  • gene therapy includes several approaches: molecular replacement of a mutated gene, addition of a new gene resulting in the synthesis of a therapeutic protein, and modulation of endogenous cellular gene expression by recombinant expression methods or by drugs. Gene-transfer techniques are described in detail (see e.g.
  • the invention features a method of treating or preventing a neurodegenerative disease by means of antisense nucleic acid therapy, i.e. the down-regulation of an inappropriately expressed or defective gene by the introduction of antisense nucleic acids or derivatives thereof into certain critical cells (see e.g. Gillespie, DN&P 1992, 5: 389-395; Agrawal and Akhtar, Trends Biotechnol 1995, 13: 197-199; Crooke, Biotechnology 1992, 10: 882-6).
  • ribozymes i.e. RNA molecules that act as enzymes, destroying RNA that carries the message of disease has also been described (see e.g.
  • the subject to be treated is a human, and therapeutic antisense nucleic acids or derivatives thereof are directed against transcription products of the gene coding for thyroid hormone binding protein. It is preferred that cells of the central nervous system, preferably the brain, of a subject are treated in such a way. Cell penetration can be performed by known strategies such as coupling of antisense nucleic acids and derivatives thereof to carrier particles, or the above described techniques. Strategies for administering targeted therapeutic oligo- deoxynucleotides are known to those of skill in the art (see e.g. Wickstrom, Trends Biotechnol 1992, 10: 281-287). In some cases, delivery can be performed by mere topical application.
  • RNAi RNA interference
  • the method comprises grafting donor cells into the central nervous system, preferably the brain, of said subject, or donor cells preferably treated so as to minimize or reduce graft rejection, wherein said donor cells are genetically modified by insertion of at least one transgene encoding said agent or agents.
  • Said transgene might be carried by a viral vector, in particular a retroviral vector.
  • the transgene can be inserted into the donor cells by a nonviral physical transfection of DNA encoding a transgene, in particular by microinjection.
  • said agent for treating and preventing a neurodegenerative disease is a therapeutic protein which can be administered to said subject, preferably a human, by a process comprising introducing subject cells into said subject, said subject cells having been treated in vitro to insert a DNA segment encoding said therapeutic protein, said subject cells expressing in vivo in said subject a therapeutically effective amount of said therapeutic protein.
  • Said DNA segment can be inserted into said cells in vitro by a viral vector, in particular a retroviral vector.
  • Methods of treatment comprise the application of therapeutic cloning, transplantation, and stem cell therapy using embryonic stem cells or embryonic germ cells and neuronal adult stem cells, combined with any of the previously described cell- and gene therapeutic methods.
  • Stem cells may be totipotent or pluripotent. They may also be organ-specific.
  • Strategies for repairing diseased and/or damaged brain cells or tissue comprise (i) taking donor cells from an adult tissue. Nuclei of those cells are transplanted into unfertilized egg cells from which the genetic material has been removed. Embryonic stem cells are isolated from the blastocyst stage of the cells which underwent somatic cell nuclear transfer.
  • stem cells preferably neuronal cells (Lanza et al., Nature Medicine 1999, 9: 975-977), or (ii) purifying adult stem cells, isolated from the central nervous system, or from bone marrow (mesenchymal stem cells), for in vitro expansion and subsequent grafting and transplantation, or (iii) directly inducing endogenous neural stem cells to proliferate, migrate, and differentiate into functional neurons (Peterson DA, Curr. Opin. Pharmacol. 2002, 2: 34-42).
  • Adult neural stem cells are of great potential for repairing damaged or diseased brain tissues, as the germinal centers of the adult brain are free of neuronal damage or dysfunction (Colman A, Drug Discovery World 2001 , 7: 66-71 ).
  • the subject for treatment or prevention can be a human, an experimental animal, e.g. a mouse or a rat, a domestic animal, or a non-human primate.
  • the experimental animal can be an animal model for a neurodegenerative disorder, e.g. a transgenic mouse and/or a knock-out mouse with an AD-type neuropathology.
  • the invention features a modulator of an activity, or a level, or both said activity and said level of at least one substance which is selected from the group consisting of (i) the gene coding for THBP, and/or (ii) a transcription product of the gene coding for THBP, and/or (iii) a translation product of the gene coding for THBP, and/or (iv) a fragment, or derivative, or variant of (i) to (iii).
  • the invention features a pharmaceutical composition
  • a pharmaceutical composition comprising said modulator and preferably a pharmaceutical carrier.
  • Said carrier refers to a diluent, adjuvant, excipient, or vehicle with which the modulator is administered.
  • the invention features a modulator of an activity, or a level, or both said activity and said level of at least one substance which is selected from the group consisting of (i) the gene coding for THBP, and/or (ii) a transcription product of the gene coding for THBP, and/or (iii) a translation product of the gene coding. for THBP, and/or (iv) a fragment, or derivative, or variant of (i) to (iii) for use in a pharmaceutical composition.
  • the invention provides for the use of a modulator of an activity, or a level, or both said activity and said level of at least one substance which is selected from the group consisting of (i) the gene coding for THBP, and/or (ii) a transcription product of the gene coding for THBP, and/or (iii) a translation product of the gene coding for THBP, and/or (iv) a fragment, or derivative, or variant of (i) to (iii) for a preparation of a medicament for treating or preventing a neurodegenerative disease, in particular AD.
  • a modulator of an activity or a level, or both said activity and said level of at least one substance which is selected from the group consisting of (i) the gene coding for THBP, and/or (ii) a transcription product of the gene coding for THBP, and/or (iii) a translation product of the gene coding for THBP, and/or (iv) a fragment, or derivative, or variant of (
  • the present invention also provides a kit comprising one or more containers filled with a therapeutically or prophylactically effective amount of said pharmaceutical composition.
  • the invention features a recombinant, non-human animal comprising a non-native THBP gene sequence, or a fragment, or a derivative, or variant thereof.
  • the generation of said recombinant, non-human animal comprises (i) providing a gene targeting construct containing said gene sequence and a selectable marker sequence, and (ii) introducing said targeting construct into a stem cell of a non-human animal, and (iii) introducing said non-human animal stem cell into a non-human embryo, and (iv) transplanting said embryo into a pseudopregnant non-human animal, and (v) allowing said embryo to develop to term, and (vi) identifying a genetically altered non-human animal whose genome comprises a modification of said gene sequence in both alleles, and (vii) breeding the genetically altered non-human animal of step (vi) to obtain a genetically altered non-human animal whose genome comprises a modification of said endogenous gene, wherein said gene is mis-expressed, or under-
  • the invention features an assay for screening for a modulator of neurodegenerative diseases, in particular AD, or related diseases and disorders of one or more substances selected from the group consisting of (i) the gene coding for THBP, and/or (ii) a transcription product of the gene coding for THBP, and/or (iii) a translation product of the gene coding for THBP, and/or (iv) a fragment, or derivative, or variant of (i) to (iii).
  • This screening method comprises (a) contacting a cell with a test compound, and (b) measuring the activity, or the level, or both the activity and the level of one or more substances recited in (i) to (iv), and (c) measuring the activity, or the level, or both the activity and the level of said substances in a control cell not contacted with said test compound, and (d) comparing the levels of the substance in the cells of step (b) and (c), wherein an alteration in the activity and/or level of said substances in the contacte ⁇ cells indicates that the test compound is a modulator of said diseases and disorders.
  • the invention features a screening assay for a modulator of neurodegenerative diseases, in particular AD, or related diseases and disorders of one or more substances selected from the group consisting of (i) the gene coding for THBP, and/or (ii) a transcription product of the gene coding for THBP, and/or (iii) a translation product of the gene coding for THBP, and/or (iv) a fragment, or derivative, or variant of (i) to (iii), comprising (a) administering a test compound to a test animal which is predisposed to developing or has already developed symptoms of a neurodegenerative disease or related diseases or disorders, and (b) measuring the activity and/or level of one or more substances recited in (i) to (iv), and (c) measuring the activity and/or level of said substances in a matched control animal which is equally predisposed to developing or has already developed said symptoms of a neurodegenerative disease, and to which animal no such test compound has been administered, and (d) comparing the group consisting of
  • test animal and/or said control animal is a recombinant, non-human animal which expresses the gene coding for THBP, or a fragment, or a derivative thereof, under the control of a transcriptional regulatory element which is not the native THBP gene transcriptional control regulatory element.
  • the present invention provides a method for producing a medicament comprising the steps of (i) identifying a modulator of neurodegenerative diseases by a method of the aforementioned screening assays and (ii) admixing the modulator with a pharmaceutical carrier.
  • said modulator may also be identifiable by other types of screening assays.
  • the present invention provides for an assay for testing a compound, preferably for screening a plurality of compounds, for inhibition of binding between a ligand and THBP, or a fragment, or derivative, or variant thereof.
  • Said screening assay comprises the steps of (i) adding a liquid suspension of said THBP, or a fragment, or derivative, or variant thereof, to a plurality of containers, and (ii) adding a compound or a plurality of compounds to be screened for said inhibition to said plurality of containers, and (iii) adding detectable, preferably fluorescently labelled ligand to said containers, and (iv) incubating said THBP, or said fragment, or derivative or variant thereof, and said compound or plurality of compounds, and said detectable, preferably fluorescently labelled ligand, and (v) measuring the amounts of preferably fluorescence associated with said THBP, or with said fragment, or derivative, or variant thereof, and (vi) determining the degree of inhibition by one or more of said compounds of binding of said ligand
  • THBP translation product or fragment, or derivative, or variant thereof it might be preferred to reconstitute said THBP translation product or fragment, or derivative, or variant thereof into artificial liposomes to generate the corresponding proteoliposomes to determine the inhibition of binding between a ligand and said THBP translation product.
  • Methods of reconstitution of membrane proteins from detergent into liposomes are known to those skilled in the art (Banerjee and Datta, Mol. Cell Biochem. 1983, 50: 3-15; Schwarz et al., Biochemistry 1999, 38: 9456-9464; Krivosheev and Usanov, Biochemistry-Moscow 1997, 62: 1064-1073).
  • any other detectable label known to the person skilled in the art e.g. radioactive labels, and detect it accordingly.
  • Said method may be useful for the identification of novel compounds as well as for evaluating compounds which have been improved or otherwise optimized in their ability to inhibit the binding of a ligand to a gene product of the gene coding for THBP, or a fragment, or derivative, or variant thereof.
  • a fluorescent binding assay in this case based on the use of carrier particles, is disclosed and described in patent application WO 00/52451.
  • a further example is the competitive assay method as described in patent WO 02/01226.
  • the present invention provides a method for producing a medicament comprising the steps of (i) identifying a compound as an inhibitor of binding between a ligand and a gene product of the gene coding for THBP by the aforementioned inhibitory binding assay and (ii) admixing the compound with a pharmaceutical carrier.
  • said compound may also be identifiable by other types of screening assays.
  • the invention features an assay for testing a compound, preferably for screening a plurality of compounds to determine the degree of binding of said compounds to THBP, or to a fragment, or derivative, or variant thereof.
  • Said screening assay comprises, (i) adding a liquid suspension of said THBP, or a fragment, or derivative, or variant thereof, to a plurality of containers, and (ii) adding a detectable, preferably a fluorescently labelled compound or a plurality of detectable, preferably fluorescently labelled compounds to be screened for said binding to said plurality of containers, and (iii) incubating said THBP, or said fragment, or derivative, or variant thereof, and said detectable, preferably fluorescently labelled compound or fluorescently labelled compounds, and (iv) measuring the amounts of preferably fluorescence associated with said THBP, or with said fragment, or derivative, or variant thereof, and (v) determining the degree of binding by one or more of said compounds to said THBP, or said fragment, or derivative, or variant thereof.
  • a fluorescent label In this type of assay it might be preferred to use a fluorescent label. However, any other type of detectable label might also be employed. Also in this type of assay it might be preferred to reconstitute a THBP translation product or fragment, or derivative, or variant thereof into artificial liposomes as described in the present invention. Said assay methods may be useful for the identification of novel compounds as well as for evaluating compounds which have been improved or otherwise optimized in their ability to bind to THBP, or a fragment, or derivative, or variant thereof.
  • the present invention provides a method for producing a medicament comprising the steps of (i) identifying a compound as a binder to a gene product of the gene coding for THBP by the aforementioned binding assays and (ii) admixing the compound with a pharmaceutical carrier.
  • said compound may also be identifiable by other types of screening assays.
  • the present invention provides for a medicament obtainable by any of the methods according to the herein claimed screening assays.
  • the instant invention provides for a medicament obtained by any of the methods according to the herein claimed screening assays.
  • the present invention features a protein molecule, said protein molecule being a translation product of the gene coding for THBP, SEQ ID NO. 2, or a fragment, or derivative, or variant thereof, for use as a diagnostic target for detecting a neurodegenerative disease, in particular Alzheimer's disease.
  • the present invention further features a protein molecule, said protein molecule being a translation product of the gene coding for THBP, SEQ ID NO. 2, or a fragment, or derivative, or variant thereof, for use as a screening target for reagents or compounds preventing, or treating, or ameliorating a neurodegenerative disease, in particular Alzheimer's disease.
  • the present invention features an antibody which is specifically immunoreactive with an immunogen, wherein said immunogen is a translation product of the gene coding for THBP, SEQ ID NO. 2, or a fragment, or derivative, or variant thereof.
  • the immunogen may comprise immunogenic or antigenic epitopes or portions Of a translation product of said gene, wherein said immunogenic or antigenic portion of a translation product is a polypeptide, and wherein said polypeptide elicits an antibody response in an animal, and wherein said polypeptide is immunospecifically bound by said antibody.
  • Methods for generating antibodies are well known in the art (see Harlow et al., Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1988).
  • antibody encompasses all forms of antibodies known in the art, such as polyclonal, monoclonal, chimeric, recombinatorial, anti-idiotypic, humanized, or single chain antibodies, as well as fragments thereof (see Dubel and Breitli ⁇ g, Recombinant Antibodies, Wiley-Liss, New York, NY, 1999).
  • Antibodies of the present invention are useful, for instance, in a variety of diagnostic and therapeutic methods, based on state-in-the-art techniques (see Harlow and Lane, Using Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1999 and Edwards R., Immunodiagnostics: A Practical Approach, Oxford University Press, Oxford, England, 1999) such as enzyme-immuno assays (e.g. enzyme-linked immunosorbent assay, ELISA), radioimmuno assays, chemoluminescence-immuno assays, Western-blot, immunoprecipitation and antibody microarrays. These methods involve the detection of translation products of the THBP gene, or fragments, or derivatives, or variants thereof.
  • enzyme-immuno assays e.g. enzyme-linked immunosorbent assay, ELISA
  • radioimmuno assays e.g. enzyme-linked immunosorbent assay, ELISA
  • radioimmuno assays
  • said antibodies can be used for detecting the pathological state of a cell in a sample from a subject, comprising immunocytochemical staining of said cell with said antibody, wherein an altered degree of staining, or an altered staining pattern in said cell compared to a cell representing a known health status indicates a pathological state of said cell.
  • the pathological state relates to a neurodegenerative disease, in particular to AD.
  • Immunocytochemical staining of a cell can be carried out by a number of different experimental methods well known in the art.
  • Figure 1 depicts the brain regions with selective vulnerability to neuronal loss and degeneration in AD.
  • neurons within the inferior temporal lobe, the entorhinal cortex, the hippocampus, and the amygdala are subject to degenerative processes in AD (Terry et al., Annals of Neurology 1981 , 10: 184-192). These brain regions are mostly involved in the processing of learning and memory functions.
  • neurons within the frontal cortex, the occipital cortex, and the cerebellum remain largely intact and preserved from neurodegenerative processes in AD.
  • Brain tissues from the frontal cortex (F), the temporal cortex (T) and the hippocampus (H) of AD patients and healthy, age-matched control individuals were used for the herein disclosed examples.
  • the image of a normal healthy brain was taken from a publication by Strange (Brain Biochemistry and Brain Disorders, Oxford University Press, Oxford, 1992, p.4).
  • Figure 2 discloses the initial identification of the differential expression of the gene coding for THBP in a fluorescence differential display screen.
  • the figure shows a clipping of a large preparative fluorescent differential display gel.
  • PCR products from the frontal cortex (F) and the temporal cortex (T) of two healthy control subjects and six AD patients were loaded in duplicate onto a denaturing polyacrylamide gel (from left to right).
  • PCR products were obtained by amplification of the individual cDNAs with the corresponding one-base-anchor oligonucleotide and the specific Cy3 labelled random primers.
  • the arrow indicates the migration position where significant differences in intensity of the signals for a transcription product of the gene coding for THBP derived from frontal cortex as compared to the signals derived from the temporal cortex of AD patients exist.
  • the differential expression reflects a down-regulation of THBP gene transcription in the temporal cortex compared to the frontal cortex of AD patients. Comparing the signals derived from temporal cortex and frontal cortex of healthy non-AD control subjects with each other, no such difference in signal intensity, i.e. no altered expression level can be detected.
  • Figures 3 and 4 illustrate the verification of the differential expression of the THBP gene in AD brain tissues by quantitative RT-PCR analysis. Quantification of RT-PCR products from RNA samples collected from the frontal cortex (F) and the temporal cortex (T) of AD patients ( Figure 3a) and samples from the frontal cortex (F) and the hippocampus (H) of AD patients ( Figure 4a) was performed by the LightCycler rapid thermal cycling technique. Likewise, samples of healthy, age-matched control individuals were compared ( Figure 3b for frontal cortex and temporal cortex, Figure 4b for frontal cortex and hippocampus). The data were normalized to the combined average values of a set of standard genes which showed no significant differences in their gene expression levels.
  • Said set of standard genes consisted of genes for cyclophilin B, the ribosomal protein S9, the transferrin receptor, GAPDH, and beta- actin.
  • the figures depict the kinetics of amplification by plotting the cycle number against the amount of amplified material as measured by its fluorescence.
  • FIG. 5 depicts SEQ ID NO. 1 , the nucieotide sequence of the 395 bp thyroid hormone binding protein (THBP) cDNA fragment, identified and obtained by fluorescence differential display and subsequent cloning.
  • Figure 6 charts the schematic alignment of SEQ ID NO. 1 to the nucieotide sequence of thyroid hormone binding protein (THBP) cDNA (GenBank accession number U85772). The open rectangle represents the open reading frame of the gene coding for THBP, thin bars represent the 5' and 3' untranslated regions (UTRs).
  • THBP thyroid hormone binding protein
  • Figure 7 outlines the sequence alignment of SEQ ID NO. 1 to the nucieotide sequence of thyroid hormone binding protein (THBP) cDNA (GenBank accession number U85772).
  • THBP thyroid hormone binding protein
  • Figure 8 discloses SEQ ID NO. 2, the polypeptide sequence of human THBP, comprising 314 amino acids (GenBank accession number Q14894).
  • Figure 9 shows SEQ ID NO. 3, the nucieotide sequence of the human THBP cDNA.
  • the length of the THBP cDNA according to NCBI GenBank entry U85772 is 1228 base pairs.
  • Figure 10 depicts human pre-central sections labeled with an affinity-purified rabbit anti-THBP antiserum (green signals) raised against a peptide corresponding to amino acids 169 to 183 of THBP.
  • Immunoreactivity of THBP was observed in both the cortex (CT) and the white matter (WM) ( Figure 10a, low magnification).
  • CT cortex
  • WM white matter
  • Figure 10b high magnification
  • cytoplasm of the glial cells were found to be immunopositive
  • Figure 10c high magnification
  • a similar immunostaining pattern was obtained by using an antiserum raised against a peptide mapping to amino acids 246 to 260 of THBP. Blue signals indicate nuclei stained with DAPI.
  • Table 1 lists the gene expression levels in the frontal cortex relative to the temporal cortex for the THBP gene in seven Alzheimer's disease patients, herein identified by internal reference numbers P010, P011 , P012, P014, P016, P017, P019 (1.71 to 4.19 fold) and five healthy, age-matched control individuals, herein identified by internal reference numbers C005, C008, C011 , C012, C014 (0.87 to 1.30 fold).
  • the scatter plot diagram visualizes individual values of the frontal to temporal cortex regulation ratios in control samples (dots) and in AD patient samples (triangles), respectively. The values shown are reciprocal values according to the formula described herein (see below).
  • Table 2 lists THBP gene expression levels in the frontal cortex relative to the hippocampus in six Alzheimer's disease patients, herein identified by internal reference numbers P010, P01 1 , P012, P014, P016, P019 (0.93 to 13.90 fold) and three healthy, age-matched control individuals, herein identified by internal reference numbers C004, C005, C008 (1.27 and 2.18 fold).
  • the scatter plot diagram visualizes individual values of the frontal cortex to hippocampus regulation ratios in control samples (dots) and in AD patient samples (triangles), respectively. The values shown are reciprocal values according to the formula described herein (see below).
  • Brain tissues from AD patients and age-matched control subjects were collected, on average, within 6 hours post-mortem and immediately frozen on dry ice. Sample sections from each tissue were fixed in paraformaldehyde for histopathological confirmation of the diagnosis. Brain areas for differential expression analysis were identified (see Figure 1 ) and stored at -80 °C until RNA extractions were performed.
  • DD screening method In order to identify changes in gene expression in different tissue, a modified and improved differential display (DD) screening method was employed.
  • the original DD screening method is known to those skilled in the art (Liang and Pardee, Science 1995, 267: 1186-7). This technique compares two populations of RNA and provides clones of genes that are expressed in one population but not in the other. Several samples can be analyzed simultaneously and both up- and down-regulated genes can be identified in the same experiment. By adjusting and refining several steps in the DD method as well as modifying technical parameters, e.g.
  • DD polymerase chain reactions
  • RNA extracted as described above (ii). Equal amounts of 0.05 ⁇ g RNA each were transcribed into cDNA in 20 ⁇ l reactions containing 0.5 mM each dNTP, 1 ⁇ l Sensiscript Reverse Transcriptase and 1x RT buffer (Qiagen), 10 U RNase inhibitor (Qiagen) and 1 ⁇ M of either one-base- anchor oligonucleotides HT-
  • PCR polymerase chain reaction
  • a polymerase chain reaction employing the corresponding one-base-anchor oligonucleotide (1 ⁇ M) along with either one of the Cy3 labelled random DD primers (1 ⁇ M), 1x GeneAmp PCR buffer (Applied Biosystems), 1 .5 mM MgCl2 (Applied Biosystems), 2 ⁇ M dNTP-Mix (dATP, dGTP, dCTP, dTTP Amersham Pharmacia Biotech), 5 % DMSO (Sigma), 1 U AmpliTaq DNA Polymerase (Applied Biosystems) in a 20 ⁇ l final volume.
  • PCR polymerase chain reaction
  • PCR conditions were set as follows: one round at 94°C for 30 sec for denaturing, cooling 1 °C/sec down to 40°C, 40°C for 4 min for low- stringency annealing of primer, heating 1 °C/sec up to 72°C, 72°C for 1 min for extension. This round was followed by 39 high-stringency cycles: 94°C for 30 sec, cooling 1 °C/sec down to 60°C, 60°C for 2 min, heating 1 °C/sec up to 72°C, 72°C for 1 min. One final step at 72°C for 5 min was added to the last cycle (PCR cycler: Multi Cycler PTC 200, MJ Research).
  • a full-scale picture was printed, differentially expressed bands marked, excised from the gel, transferred into 1.5 ml containers, overlayed with 200 ⁇ l sterile water and kept at -20°C until extraction.
  • Elution and reamplification of DD products The differential bands were extracted from the gel by boiling in 200 ⁇ l H2O for 10 min, cooling down on ice and precipitation from the supernatant fluids by using ethanol (Merck) and glycogen/sodium acetate (Merck) at -20°C over night, and subsequent centrifugation at 13.000 rpm for 25 min at 4°C.
  • the obtained preparations were used as templates for reamplification by 15 high-stringency cycles in 25- ⁇ l PCR mixtures containing the corresponding primer pairs as used for the DD PCR (see above) under identical conditions, with the exception of the initial round at 94°C for 5 min, followed by 15 cycles of: 94°C for 45 sec, 60°C for 45 sec, ramp 1 °C/sec to 70°C for 45 sec, and one final step at 72°C for 5 min.
  • THBP thyroid hormone binding protein
  • PCR amplification (95°C and 1 sec, 56 ⁇ C and 5 sec, and 72°C and 5 sec) was performed in a volume of 20 ⁇ l containing LightCycler-FastStart DNA Master SYBR Green I mix (contains FastStart Taq DNA polymerase, reaction buffer, dNTP mix with dUTP instead of dTTP, SYBR Green I dye, and 1 mM MgCI 2 ; Roche), 0.5 ⁇ M primers, 2 ⁇ l of a cDNA dilution series (final concentration of 40, 20, 10, 5, 1 and 0.5 ng human total brain cDNA; Clontech) and, depending on the primers used, additional 3 M MgCI 2 .
  • LightCycler-FastStart DNA Master SYBR Green I mix contains FastStart Taq DNA polymerase, reaction buffer, dNTP mix with dUTP instead of dTTP, SYBR Green I dye, and 1 mM MgCI 2
  • the PCR protocol was applied to determine the PCR efficiency of a set of reference genes which were selected as a reference standard for quantification.
  • the mean value of five such reference genes was determined: (1 ) cyclophilin B, using the specific primers 5'- ACTGAAGCACTACGGGCCTG-3' and 5'-AGCCGTTGGTGTCTTTGCC-3' except for MgCl 2 (an additional 1 mM was added instead of 3 mM).
  • Melting curve analysis revealed a single peak at approximately 87°C with no visible primer dimers.
  • Agarose gel analysis of the PCR product showed one single band of the expected size (62 bp).
  • the logarithm of the cDNA concentration was plotted against the threshold cycle number C t for the gene coding for thyroid hormone binding protein and the five reference standard genes. The slopes and the intercepts of the standard curves (i.e. linear regressions) were calculated for all genes.
  • cDNAs from frontal cortex and temporal cortex, and from frontal cortex and hippocampus, respectively were analyzed in parallel and normalized to cyclophilin B. The C t values were measured and converted to ng total brain cDNA using the corresponding standard curves:
  • THBP frontal [ng] / cyclophilin B frontal [ng] the set of reference standard genes was analyzed in parallel to determine the mean average value of the temporal to frontal ratios, and of the hippocampal to frontal ratios, respectively, of expression levels of the reference standard genes for each individual brain sample.
  • cyclophilin B was analyzed in step 2 and step 3, and the ratio from one gene to another gene remained constant in different runs, it was possible to normalize the values for the THBP gene to the mean average value of the set of reference standard genes instead of normalizing to one single gene alone.
  • the calculation was performed by dividing the respective ratio shown above by the deviation of cyclophilin B from the mean value of all housekeeping genes. The results of such quantitative RT-PCR analysis for the gene coding for THBP are shown in Figure 3 and 4.
  • THBP immunofluorescence staining of THBP in human brain
  • frozen sections were prepared with a cryostat (Leica CM3050S) from post-mortem pre-central gyrus of a donor person and fixed in 4% PFA for 20 min. After washing in PBS, the sections were pre-incubated with blocking buffer (10% normal goat serum, 0.2% Triton X-100 in PBS) for 30 min and then incubated with affinity-purified rabbit anti-THBP antisera (1 :40 diluted in blocking buffer; custom-made, Biogenes, Berlin, Germany) overnight at 4°C.
  • blocking buffer 10% normal goat serum, 0.2% Triton X-100 in PBS
  • the sections were incubated with FITC-conjugated goat anti-rabbit IgG (1 :150 diluted in 1 % BSA/PBS) for 2 hours at room temperature and then again washed in PBS. Staining of the nuclei was performed by incubation of the sections with 5 ⁇ M DAPI in PBS for 3 min (blue signal).
  • the sections were treated with 1 % Sudan Black B in 70% ethanol for 2-10 min at room temperature and then sequentially dipped in 70% ethanol, destilled water and PBS.
  • the sections were coverslipped with 'Vectrashield' mounting medium (Vector Laboratories, Burlingame, CA) and observed under an inverted microscope (1X81 , Olympus Optical).
  • the digital images were captured with the appropriate software (AnalySiS, Olympus Optical).

Abstract

The present invention discloses the differential expression of the gene coding for THBP, thyroid hormone binding protein, in specific brain regions of Alzheimer's disease patients. Based on this finding, the invention provides a method for diagnosing or prognosticating Alzheimer's disease in a subject, or for determining whether a subject is at increased risk of developing Alzheimer's disease. Furthermore, this invention provides therapeutic and prophylactic methods for treating or preventing Alzheimer's disease and related neurodegenerative disorders using the gene coding for THBP. A method of screening for modulating agents of neurodegenerative diseases is also disclosed.

Description

DIAGNOSTIC AND THERAPEUTIC USE OF THYROID HORMONE BINDING PROTEIN FOR NEURODEGENERATIVE DISEASES
The present invention relates to methods of diagnosing, prognosticating and monitoring the progression of neurodegenerative diseases in a subject. Furthermore, methods of therapy control and screening for modulating agents of neurodegenerative diseases are provided. The invention also discloses pharmaceutical compositions, kits, and recombinant animal models.
Neurodegenerative diseases, in particular Alzheimer's disease (AD), have a strongly debilitating impact on a patient's life. Furthermore, these diseases constitute an enormous health, social, and economic burden. AD is the most common neurodegenerative disease, accounting for about 70% of all dementia cases, and it is probably the most devastating age-related neurodegenerative condition affecting about 10% of the population over 65 years of age and up to 45% over age 85 (for a recent review see Vickers et al., Progress in Neurobiology 2000, 60: 139-165). Presently, this amounts to an estimated 12 million cases in the US, Europe, and Japan. This situation will inevitably worsen with the demographic increase in the number of old people ("aging of the baby boomers") in developed countries. The neuropathological hallmarks that occur in the brains of individuals with AD are senile plaques, composed of amyloid-β protein, and profound cytoskeletal changes coinciding with the appearance of abnormal filamentous structures and the formation of neurofibrillary tangles.
The amyloid-β (Aβ) protein evolves from the cleavage of the amyloid precursor protein (APP) by different kinds of proteases. The cleavage by the β/γ-secretase leads to the formation of Aβ peptides of different lengths, typically a short more soluble and slow aggregating peptide consisting of 40 amino acids and a longer 42 amino acid peptide, which rapidly aggregates outside the cells, forming the characteristic amyloid plaques (Selkoe, Physiological Rev 2001 , 81 : 741-66; Greenfield et al., Frontiers Bioscience 2000, 5: D72-83). Two types of plaques, diffuse plaques and neuritic plaques, can be detected in the brain of AD patients, the latter ones being the classical, most prevalent type. They are primarily found in the cerebral cortex and hippocampus. The neuritic plaques have a diameter of 50μm to 200μm and are composed of insoluble fibrillar amyloids, fragments of dead neurons, of microglia and astrocytes, and other components such as neurotransmitters, apolipoprotein E, glycosaminoglycans, α1-antichymotrypsin and others. The generation of toxic Aβ deposits in the brain starts very early in the course of AD, and it is discussed to be a key player for the subsequent destructive processes leading to AD pathology. The other pathological hallmarks of AD are neurofibrillary tangles (NFTs) and abnormal neurites, described as neuropil threads (Braak and Braak, Ada Neuropathol 1991 , 82: 239-259). NFTs emerge inside neurons and consist of chemically altered tau, which forms paired helical filaments twisted around each other. Along the formation of NFTs, a loss of neurons can be observed. It is discussed that said neuron loss may be due to a damaged microtubule-associated transport system (Johnson and Jenkins, J Alzheimers Dis 1996, 1 : 38-58; Johnson and Hartigan, J Alzheimers Dis 1999, 1 : 329-351 ). The appearance of neurofibrillary tangles and their increasing number correlates well with the clinical severity of AD (Schmitt et al., Neurology 2000, 55: 370-376). AD is a progressive disease that is associated with early deficits in memory formation and ultimately leads to the complete erosion of higher cognitive function. The cognitive disturbances include among other things memory impairment, aphasia, agnosia and the loss of executive functioning. A characteristic feature of the pathogenesis of AD is the selective vulnerability of particular brain regions and subpopulations of nerve cells to the degenerative process. Specifically, the temporal lobe region and the hippocampus are affected early and more severely during the progression of the disease. On the other hand, neurons within the frontal cortex, occipital cortex, and the cerebellum remain largely intact and are protected from neurodegeneration (Terry et al., Annals of Neurology 1981 , 10: 184-92). The age of onset of AD may vary within a range of 50 years, with early-onset AD occurring in people younger than 65 years of age, and late-onset of AD occurring in those older than 65 years. About 10% of all AD cases suffer from early-onset AD, with only 1-2% being familial, inherited cases.
Currently, there is no cure for AD, nor is there an effective treatment to halt the progression of AD or even to diagnose AD ante-mortem with high probability. Several risk factors have been identified that predispose an individual to develop AD, among them most prominently the epsilon 4 allele of the three different existing alleles (epsilon 2, 3, and 4) of the apolipoprotein E gene (ApoE) (Strittmatter et al., Proc Nat! Acad Sci USA 1993, 90: 1977-81 ; Roses, Ann NY Acad Sci 1998, 855: 738-43). Efforts to detect further susceptibility genes and disease-linked polymorphisms lead to the assumption that specific regions and genes on human chromosomes 10 and 12 may be associated with late-onset AD (Myers et al., Science 2000, 290: 2304-5; Bertram et al., Science 2000, 290: 2303; Scott et al., Am J Hum Genet 2000, 66: 922-32).
Although there are rare examples of early-onset AD which have been attributed to genetic defects in the genes for amyloid precursor protein (APP) on chromosome 21 , presenilin-1 on chromosome 14, and presenilin-2 on chromosome 1 , the prevalent form of late-onset sporadic AD is of hitherto unknown etiologic origin. The mutations found to date account for only half of the familial AD cases, which is less than 2% of all AD patients. The late onset and complex pathogenesis of neurodegenerative disorders pose a formidable challenge to the development of therapeutic and diagnostic agents. It is crucial to expand the pool of potential drug targets and diagnostic markers. It is therefore an object of the present invention to provide insight into the pathogenesis of neurological diseases and to provide methods, materials, agents, compositions, and animal models which are suited inter alia for the diagnosis and development of a treatment of these diseases. This object has been solved by the features of the independent claims. The subclaims define preferred embodiments of the present invention.
Thyroid hormones mediate their pleiotropic effects on tissues and cells via nuclear thyroid hormone receptors (TRalpha and TRbeta) that act as transcription factors. These receptors belong to a large superfamily of nuclear hormone receptors including the receptors for steroid hormones, retinoids, and vitamin D. Steroid hormone receptors shuttle between the nucleus and the cytoplasm, where they interact with their appropriate ligand/agonist (for review: Chawla et al., Science 2001 , 294: 1866-1870). In contrast, the retinoid and thyroid hormone receptors remain permanently bound to their genomic target sequence elements. The function of intracellular shuttling and homeostasis of retinoids and thyroid hormones is exerted by specialized cytoplasmic binding proteins. Like many other nuclear hormone receptors, the thyroid hormone receptors form obligate heterodimers with the retinoid X receptor (Chawla et al., Science 2001 , 294: 1866-1870; Wu et al., J. Biol. Chem. 2002, 277: 8898-8905).
A number of different cytoplasmic binding proteins for the thyroid hormone triiodothyronine (T3) have been described. A 58 kDa monomer of the ubiquitous enzyme pyruvate kinase has been described as a T3-binding protein in human cells (Kato et al., Proc. Natl. Acad. Sci. U.S.A. 1989, 86: 7861-7865). T3 binding to this protein is allosterically effected by the glycolytic intermediate fructose-1 ,6- bisphosphate and impacts on overall transcriptional regulation by the thyroid hormone (Ashizawa et al., Biochemistry 1991 , 30: 7105-7111). In Xenopus species, a 59 kDa subunit of the aldehyde dehydrogenease has been described as a T3- binding protein (Yamauchi et al., J. Biol. Chem. 1999, 274: 8460-8469). Yet other T3-binding proteins were isolated from rat kidney and liver cytoplasm: a 58 kDa protein that regulates the access of T3 to its nuclear receptor (Hashizume et al., J. Biol. Chem. 1989, 264: 4864-4871 ) and an apparently homodimeric 38 kDa protein whose interaction with T3 was activated by NADPH and inhibited by NADP+ (Kobayashi et al., Endocrinology 1991 , 129: 1701-1708). This latter protein will hereafter' be considered as the NADPH-dependent thyroid hormone binding protein (THBP). The THBP gene was cloned from a human brain cDNA library and found to encode a 314 amino acids protein with a theoretical molecular weight of 33775 Da (Vie et al., Mol. Endocrinol. 1997, 1 1 : 1728-1736; NCBI GenBank accession numbers: U85772 and Q14894; Protein ID AAB81564.1 ). The deduced sequence is identical to a human protein termed mu-crystallin (Kim et al., Proc. Natl. Acad. Sci. U. S.A. 1992, 89: 9292-9296; Segovia et al., Mol. Vis. 1997, 3: 9; NCBI GenBank accession numbers: L02950 and M90841 ). A significant homology of 40-60% at the amino acid level was noted for two strictly bacterial enyzmes, namely lysine cyclodeaminase and ornithine cyclodeaminase (Kim et al., Proc. Natl. Acad. Sci. U. S.A. 1992, 89: 9292-9296; Vie et al., Mol. Endocrinol. 1997, 11 : 1728-1736). A southern blot showed that there is only one gene for human mu-crystallin and THBP (Kim et al., Proc. Natl. Acad. Sci. U. S.A. 1992, 89: 9292-9296) which is located on chromosome 16p (Chen et al., Genomics 1992, 14: 11 15-1 1 16).
In humans, THBP is abundantly expressed in neural tissue including brain, muscle, heart and kidney, to lower levels in lung and liver, and there is none detectable in placenta and pancreas (Kim et al., Proc. Natl. Acad. Sci. U. S.A. 1992, 89: 9292- 9296). THBP is responsible for most of the intracellular high-affinity (nano olar) T3 and T4 (thyroxine) binding in kidney from rat (Vie et al., Mol. Endocrinol. 1997, 1 1 : 1728-1736). Furthermore, mu-crystallin/THBP was detected in photoreceptors of the mammalian retina (Wistow and Segovia, Invest. Ophthalmol. Visual Sci. 1994, 35:2073; Sperbeck et al., Mol. Biol. Cell 1997, 8:312A; Segovia et al., Mol. Vis. 1997, 3:9). A number of differential gene expression screens have identified mu- crystallin/THBP, including screens in the field of mouse hair follicle development (Aoki et al., J. Invest. Dermatol. 2000, 1 15: 402-405), screens of a hormone- refractory prostate cancer xenograft model (Mousses et al., Cancer Res. 2002, 62: 1256-1260), a screen investigating end-stage ischemic and dilated cardiomyopathy (Yang et al., Circulation 2000, 102: 3046-3052), and screens in the field of cancer in general (WO 0194629).
The present invention discloses the detection and differential regulation of thyroid hormone binding protein (THBP) gene expression in the temporal cortex versus frontal cortex of Alzheimer's disease brain in comparison to age-matched control tissues. This finding can be put in context with a number of pathophysiologic roles for THBP. Firstly, like its bacterial homologs lysine and ornithine cyclodeaminase, THBP may be involved in synthesis or breakdown of amino acids (Kim et al., Proc. Natl. Acad. Sci. U.S.A. 1992, 89: 9292-9296). For instance, levels of the amino acid glutamate, which possesses a general excitatory role in the central nervous system, may be affected. In this scenario, the T3-binding property of THBP would be neglected. In a second scenario, thyroid hormone signaling may affect the plasticity of the adult brain including the particularly vulnerable hippocampus formation (for review, Calza et al., Brain Res. Bull. 1997, 44: 549-557). As such, a differential regulation of THBP gene expression may well be a cause of neurodegenerative diseases. Thirdly, thyroid hormones were shown to negatively regulate the transcriptional activity of the amyloid precursor protein (APP) in recombinant neuroblastoma cells (Belandia et al., J. Biol. Chem. 1998, 273: 30366-30371 ) and to have an impact on the distribution of intracellular and extracellular APP isoforms by regulating APP gene splicing (Latasa et al., Endocrinology 1998, 139:2692-8). To date, no experiments have been described that demonstrate a relationship between the dysregulation of thyroid hormone binding protein (mu-crystallin/THBP) gene expression and the pathology of neurodegenerative diseases, in particular AD. Such a link, as disclosed in the present invention, offers new ways, inter alia, for the diagnosis and treatment of said neurodegenerative diseases, in particular AD.
The singular forms "a", "an", and "the" as used herein and in the claims include plural reference unless the context dictates otherwise. For example, "a cell" means as well a plurality of cells, and so forth. The term "and/or" as used in the present specification and in the claims implies that the phrases before and after this term are to be considered either as alternatives or in combination. For instance, the wording "determination of a level and/or an activity" means that either only a level, or only an activity, or both a level and an activity are determined. The term "level" as used herein is meant to comprise a gage of, or a measure of the amount of, or a concentration of a transcription product, for instance an mRNA, or a translation product, for instance a protein or polypeptide. The term "activity" as used herein shall be understood as a measure for the ability of a transcription product or a translation product to produce a biological effect or a measure for a level of biologically active molecules. The term "activity" also refers to enzymatic activity. The terms "level" and/or "activity" as used herein further refer to gene expression levels or gene activity. Gene expression can be defined as the utilization of the information contained in a gene by transcription and translation leading to the production of a gene product. "Dysregulation" shall mean an upregulation or downregulation of gene expression. A gene product comprises either RNA or protein and is the result of expression of a gene. The amount of a gene product can be used to measure how active a gene is. The term "gene" as used in the present specification and in the claims comprises both coding regions (exons) as well as non-coding regions (e.g. non-coding regulatory elements such as promoters or enhancers, introns, leader and trailer sequences). The term "ORF" is an acronym for "open reading frame" and refers to a nucleic acid sequence that does not possess a stop codon in at least one reading frame and therefore can potentially be translated into a sequence of amino acids. "Regulatory elements" shall comprise inducible and non-inducible promoters, enhancers, operators, and other elements that drive and regulate gene expression. The term "fragment" as used herein is meant to comprise e.g. an alternatively spliced, or truncated, or otherwise cleaved transcription product or translation product. The term "derivative" as used herein refers to a mutant, or an RNA-edited, or a chemically modified, or otherwise altered transcription product, or to a mutant, or chemically modified, or otherwise altered translation product. For instance, a "derivative" may be generated by processes such as altered phosphorylation, or glycosylation, or acetylation, or lipidation, or by altered signal peptide cleavage or other types of maturation cleavage. These processes may occur post-translationally. The term "modulator" as used in the present invention and in the claims refers to a molecule capable of changing or altering the level and/or the activity of a gene, or a transcription product of a gene, or a translation product of a gene. Preferably, a "modulator" is capable of changing or altering the biological activity of a transcription product or a translation product of a gene. Said modulation, for instance, may be an increase or a decrease in enzyme activity, a change in binding characteristics, or any other change or alteration in the biological, functional, or immunological properties of said translation product of a gene. The terms "agent", "reagent", or "compound" refer to any substance, chemical, composition, or extract that have a positive or negative biological effect on a cell, tissue, body fluid, or within the context of any biological system, or any assay system examined. They can be agonists, antagonists, partial agonists or inverse agonists of a target. Such agents, reagents, or compounds may be nucleic acids, natural or synthetic peptides or protein complexes, or fusion proteins. They may also be antibodies, organic or anorganic molecules or compositions, small molecules, drugs and any combinations of any of said agents above. They may be used for testing, for diagnostic or for therapeutic purposes. The terms "oligonucleotide primer" or "primer" refer to short nucleic acid sequences which can anneal to a given target polynucleotide by hybridization of the complementary base pairs and can be extended by a polymerase. They may be chosen to be specific to a particular sequence or they may be randomly selected, e.g. they will prime all possible sequences in a mix. The length of primers used herein may vary from 10 nucleotides to 80 nucleotides. "Probes" are short nucleic acid sequences of the nucleic acid sequences described and disclosed herein or sequences complementary therewith. They may comprise full length sequences, or fragments, derivatives, isoforms, or variants of a given sequence. The identification of hybridization complexes between a "probe" and an assayed sample allows the detection of the presence of other similar sequences within that sample. As used herein, "homolog or homology" is a term used in the art to describe the relatedness of a nucieotide or peptide sequence to another nucieotide or peptide sequence, which is determined by the degree of identity and/or similarity between said sequences compared. The term "variant" as used herein refers to any polypeptide or protein, in reference to polypeptides and proteins disclosed in the present invention, in which one or more amino acids are added and/or substituted and/or deleted and/or inserted at the N-terminus, and/or the C-terminus, and/or within the native amino acid sequences of the native polypeptides or proteins of the present invention. Furthermore, the term "variant" shall include any shorter or longer version of a polypeptide or protein. "Variants" shall also comprise a sequence that has at least about 80% sequence identity, more preferably at least about 90% sequence identity, and most preferably at least about 95% sequence identity with the amino acid sequence of thyroid hormone binding protein, SEQ ID NO. 2. "Variants" include, for example, proteins with conservative amino acid substitutions in highly conservative regions. "Proteins and polypeptides" of the present invention include variants, fragments and chemical derivatives of the protein comprising the amino acid sequences of thyroid hormone binding protein, SEQ ID NO. 2. They can include proteins and polypeptides which can be isolated from nature or be produced by recombinant and/or synthetic means. Native proteins or polypeptides refer to naturally-occurring truncated or secreted forms, naturally occurring variant forms (e.g. splice-variants) and naturally occurring allelic variants. The term "isolated" as used herein is considered to refer to molecules that are removed from their natural environment, i.e. isolated from a cell or from a living organism in which they normally occur, and that are separated or essentially purified from the coexisting components with which they are found to be associated in nature. This notion further means that the sequences encoding such molecules can be linked by the hand of man to polynucleotides, to which they are not linked in their natural state, and that such molecules can be produced by recombinant and/or synthetic means. Even if for said purposes those sequences may be introduced into living or non-living organisms by methods known to those skilled in the art, and even if those sequences are still present in said organisms, they are still considered to be isolated. In the present invention, the terms "risk", "susceptibility", and "predisposition" are tantamount and are used with respect to the probability of developing a neurodegenerative disease, preferably Alzheimer's disease. The term 'AD' shall mean Alzheimer's disease. "AD-type neuropathology" as used herein refers to neuropathological, neurophysiological, histopathological and clinical hallmarks as described in the instant invention and as commonly known from state- of-the-art literature (see: Iqbal, Swaab, Winblad and Wisniewski, Alzheimer's Disease and Related Disorders (Etiology, Pathogenesis and Therapeutics), Wiley & Sons, New York, Weinheim, Toronto, 1999; Scinto and Daffner, Early Diagnosis of Alzheimer's Disease, Humana Press, Totowa, New Jersey, 2000; Mayeux and Christen, Epidemiology of Alzheimer's Disease: From Gene to Prevention, Springer Press, Berlin, Heidelberg, New York, 1999; Younkin, Tanzi and Christen, Presenilins and Alzheimer's Disease, Springer Press, Berlin, Heidelberg, New York, 1998). Neurodegenerative diseases or disorders according to the present invention comprise Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, Pick's disease, fronto-temporal dementia, progressive nuclear palsy, corticobasal degeneration, cerebro-vascular dementia, multiple system atrophy, argyrophilic grain dementia and other tauopathies, and mild- cognitive impairment. Further conditions involving neurodegenerative processes are, for instance, ischemic stroke, age-related macular degeneration, narcolepsy, motor neuron diseases, prion diseases, traumatic nerve injury and repair, and multiple sclerosis. In one aspect, the invention features a method of diagnosing or prognosticating a neurodegenerative disease in a subject, or determining whether a subject is at increased risk of developing said disease. The method comprises: determining a level, or an activity, or both said level and said activity of (i) a transcription product of the gene coding for thyroid hormone binding protein (THBP), and/or of (ii) a translation product of the gene coding for thyroid hormone binding protein (THBP), and/or of (iii) a fragment, or derivative, or variant of said transcription or translation product in a sample from said subject and comparing said level, and/or said activity to a reference value representing a known disease or health status, thereby diagnosing or prognosticating said neurodegenerative disease in said subject, or determining whether said subject is at increased risk of developing said neurodegenerative disease.
The invention also relates to the construction and the use of primers and probes which are unique to the nucleic acid sequences, or fragments, or variants thereof, as disclosed in the present invention. The oligonucleotide primers and/or probes can be labeled specifically with fluorescent, bioluminescent, magnetic, or radioactive substances. The invention further relates to the detection and the production of said nucleic acid sequences, or fragments and variants thereof, using said specific oligonucleotide primers in appropriate combinations. PCR-analysis, a method well known to those skilled in the art, can be performed with said primer combinations to amplify said gene specific nucleic acid sequences from a sample containing nucleic acids. Such sample may be derived either from healthy or diseased subjects. Whether an amplification results in a specific nucleic acid product or not, and whether a fragment of different length can be obtained or not, may be indicative for a neurodegenerative disease, in particular Alzheimer's disease. Thus, the invention provides nucleic acid sequences, oligonucleotide primers, and probes of at least 10 bases in length up to the entire coding and gene sequences, useful for the detection of gene mutations and single nucieotide polymorphisms in a given sample comprising nucleic acid sequences to be examined, which may be associated with neurodegenerative diseases, in particular Alzheimer's disease. This feature has utility for developing rapid DNA-based diagnostic tests, preferably also in the format of a kit.
In a further aspect, the invention features a method of monitoring the progression of a neurodegenerative disease in a subject. A level, or an activity, or both said level and said activity, of (i) a transcription product of the gene coding for thyroid hormone binding protein (THBP), and/or of (ii) a translation product of the gene coding for thyroid hormone binding protein (THBP), and/or of, (iii) a fragment, or derivative, or variant of said transcription or translation product in a sample from said subject is determined. Said level and/or said activity is compared to a reference value representing a known disease or health status. Thereby, the progression of said neurodegenerative disease in said subject is monitored.
In still a further aspect, the invention features a method of evaluating a treatment for a neurodegenerative disease, comprising determining a level, or an activity, or both said level and said activity of (i) a transcription product of the gene coding for thyroid hormone binding protein (THBP), and/or of (ii) a translation product of the gene coding for thyroid hormone binding protein (THBP), and/or of (iii) a fragment, or derivative, or variant of said transcription or translation product in a sample obtained from a subject being treated for said disease. Said level, or said activity, or both said level and said activity are compared to a reference value representing a known disease or health status, thereby evaluating the treatment for said neurodegenerative disease.
In a preferred embodiment of the herein claimed methods, kits, recombinant animals, molecules, assays, and uses of the instant invention, said gene coding for thyroid hormone binding protein is the gene coding for NADPH-dependent thyroid hormone binding protein, THBP, also termed mu-crystallin or CRYM (NCBI Genbank accession number: L02950 and M90841 ), represented by SEQ ID NO. 2 and SEQ ID NO. 3 (GenBank accession numbers: Q14894 and U85772). In the instant invention, the gene coding for said thyroid hormone binding protein is also generally referred to as the thyroid hormone binding protein gene, or THBP gene, or THBP, and further, said thyroid hormone binding protein is also generally referred to as THBP.
In a further preferred embodiment of the herein claimed methods, kits, recombinant animals, molecules, assays, and uses of the instant invention, said neurodegenerative disease or disorder is Alzheimer's disease, and said subjects suffer from Alzheimer's disease.
The present invention discloses the detection, differential expression and regulation of the gene coding for thyroid hormone binding protein (THBP) in specific brain regions of AD patients. Consequently, the thyroid hormone binding protein gene (THBP) and its corresponding transcription and translation products may have a causative role in the regional selective neuronal degeneration typically observed in AD. Alternatively, the gene coding for THBP and its products may confer a neuroprotective function to the remaining surviving nerve cells. Based on these disclosures, the present invention has utility for the diagnostic evaluation and prognosis as well as for the identification of a predisposition to a neurodegenerative disease, in particular AD- Furthermore, the present invention provides methods for the diagnostic monitoring of patients undergoing treatment for such a disease.
It is particularly preferred that said sample to be analyzed and determined is selected from the group comprising brain tissue or other tissues, or body cells. The sample can also comprise cerebrospinal fluid or other body fluids including saliva, urine, blood, serum plasma, mucus. Preferably, the methods of diagnosis, prognosis, monitoring the progression or evaluating a treatment for a neurodegenerative disease, according to the instant invention, can be practiced ex corpore, and such methods preferably relate to samples, for instance, body fluids or cells, removed, collected, or isolated from a subject or patient.
In further preferred embodiments, said reference value is that of a level, or an activity, or both said level and said activity of (i) a transcription product of the gene coding for THBP, and/or of (ii) a translation product of the gene coding for THBP, and/or of (iii) a fragment, or derivative, or variant of said transcription or translation product in a sample from a subject not suffering from said neurodegenerative disease.
In preferred embodiments, an alteration in the level and/or activity of a transcription product of the gene coding for THBP and/or of a translation product of the gene coding for THBP and/or of a fragment, or derivative, or variant thereof in a sample cell, or tissue, or body fluid from said subject relative to a reference value representing a known health status indicates a diagnosis, or prognosis, or increased risk of becoming diseased with a neurodegenerative disease, particularly AD.
In preferred embodiments, measurement of the level of transcription products of the gene coding for THBP is performed in a sample from a subject using a quantitative PCR-analysis with primer combinations to amplify said gene specific sequences from cDNA obtained by reverse transcription of RNA extracted from a sample of a subject. A Northern blot with probes specific for said gene can also be applied. It might further be preferred to measure transcription products by means of chip-based micro-array technologies. These techniques are known to those of ordinary skill in the art (see Sambrook and Russell, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 2001 ; Schena M., Microarray Biochip Technology, Eaton Publishing, Natick, MA, 2000). An example of an immunoassay is the detection and measurement of enzyme activity as disclosed and described in the patent application WO 02/14543.
Furthermore, a level and/or an activity of a translation product of the gene coding for THBP and/or of a fragment, or derivative, or variant of said translation product, and/or a level of activity of said translation product and/or of a fragment, or derivative, or variant thereof can be detected using an immunoassay, an activity assay, and/or a binding assay. These assays can measure the amount of binding between said protein molecule and an anti-protein antibody by the use of enzymatic, chromodynamic, radioactive, magnetic, or luminescent labels which are attached to either the anti-protein antibody or a secondary antibody which binds the anti-protein antibody. In addition, other high affinity ligands may be used. Immunoassays which can be used include e.g. ELISAs, Western blots and other techniques known to those of ordinary skill in the art (see Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1999 and Edwards R, Immunodiagnostics: A Practical Approach, Oxford University Press, Oxford; England, 1999). All these detection techniques may also be employed in the format of microarrays, protein-arrays, antibody microarrays, tissue microarrays, electronic biochip or protein-chip based technologies (see Schena M., Microarray Biochip Technology, Eaton Publishing, Natick, MA, 2000).
In a preferred embodiment, the level, or the activity, or both said level and said activity of (i) a transcription product of the gene coding for THBP, and/όr of (ii) a translation product of the gene coding for THBP, and/or of (iii) a fragment, or derivative, or variant of said transcription or translation product in a series of samples taken from said subject over a period of time is compared, in order to monitor the progression of said disease. In further preferred embodiments, said subject receives a treatment prior to one or more of said sample gatherings. In yet another preferred embodiment, said level and/or activity is determined before and after said treatment of said subject.
In another aspect, the invention features a kit for diagnosing or prognosticating neurodegenerative diseases, in particular AD, in a subject, or determining the propensity or predisposition of a subject to develop a neurodegenerative disease, in particular AD, said kit comprising:
(a) at least one reagent which is selected from the group consisting of (i) reagents that selectively detect a transcription product of the gene coding for THBP (ii) reagents that selectively detect a translation product of the gene coding for THBP; and
(b) instruction for diagnosing, or prognosticating a neurodegenerative disease, in particular AD, or determining the propensity or predisposition of a subject to develop such a disease by
- detecting a level, or an activity, or both said level and said activity, of said transcription product and/or said translation product of the gene coding for THBP, in a sample from said subject; and
- diagnosing or prognosticating a neurodegenerative disease, in particular AD, or determining the propensity or predisposition of said subject to develop such a disease, wherein a varied level, or activity, or both said level and said activity, of said transcription product and/or said translation product compared to a reference value representing a known health status; or a level, or activity, or both said level and said activity, of said transcription product and/or said translation product similar or equal to a reference value representing a known disease status, indicates a diagnosis or prognosis of a neurodegenerative disease, in particular AD, or an increased propensity or predisposition of developing such a disease. The kit, according to the present invention, may be particularly useful for the identification of individuals that are at risk of developing a neurodegenerative disease, in particular AD. Consequently, the kit, according to the present invention, may serve as a means for targeting identified individuals for early preventive measures or therapeutic intervention prior to disease onset, before irreversible damage in the course of the disease has been inflicted. Furthermore, in preferred embodiments, the kit featured in the invention is useful for monitoring a progression of a neurodegenerative disease, in particular AD in a subject, as well as monitoring success or failure of therapeutic treatment for such a disease of said subject. In another aspect, the invention features a method of treating or preventing a neurodegenerative disease, in particular AD, in a subject comprising the administration to said subject in a therapeutically or prophylactically effective amount of an agent or agents which directly or indirectly affect a level, or an activity, or both said level and said activity, of (i) the gene coding for THBP, and/or (ii) a transcription product of the gene coding for THBP, and/or (iii) a translation product of the gene coding for THBP, and/or (iv) a fragment, or derivative, or variant of (i) to (iii). Said agent may comprise a small molecule, or it may also comprise a peptide, an oligopeptide, or a polypeptide. Said peptide, oligopeptide, or polypeptide may comprise an amino acid sequence of a translation product of the gene coding for THBP, or a fragment, or derivative, or a variant thereof. An agent for treating or preventing a neurodegenerative disease, in particular AD, according to the instant invention, may also consist of a nucieotide, an oligonucleotide, or a polynucleotide. Said oligonucleotide or polynucleotide may comprise a nucieotide sequence of the gene coding for THBP, either in sense orientation or in antisense orientation.
In preferred embodiments, the method comprises the application of per se known methods of gene therapy and/or antisense nucleic acid technology to administer said agent or agents. In general, gene therapy includes several approaches: molecular replacement of a mutated gene, addition of a new gene resulting in the synthesis of a therapeutic protein, and modulation of endogenous cellular gene expression by recombinant expression methods or by drugs. Gene-transfer techniques are described in detail (see e.g. Behr, Ace Chem Res 1993, 26: 274-278 and Mulligan, Science 1993, 260: 926-931 ) and include direct gene-transfer techniques such as mechanical microinjection of DNA into a cell as well as indirect techniques employing biological vectors (like recombinant viruses, especially retroviruses) or model liposomes, or techniques based on transfection with DNA coprecipitation with polycations, cell membrane pertubation by chemical (solvents, detergents, polymers, enzymes) or physical means (mechanic, osmotic, thermic, electric shocks). The postnatal gene transfer into the central nervous system has been described in detail (see e.g. Wolff, Curr Opin Neurobiol 1993, 3: 743-748).
In particular, the invention features a method of treating or preventing a neurodegenerative disease by means of antisense nucleic acid therapy, i.e. the down-regulation of an inappropriately expressed or defective gene by the introduction of antisense nucleic acids or derivatives thereof into certain critical cells (see e.g. Gillespie, DN&P 1992, 5: 389-395; Agrawal and Akhtar, Trends Biotechnol 1995, 13: 197-199; Crooke, Biotechnology 1992, 10: 882-6). Apart from hybridization strategies, the application of ribozymes, i.e. RNA molecules that act as enzymes, destroying RNA that carries the message of disease has also been described (see e.g. Barinaga, Science 1993, 262: 1512-1514). In preferred embodiments, the subject to be treated is a human, and therapeutic antisense nucleic acids or derivatives thereof are directed against transcription products of the gene coding for thyroid hormone binding protein. It is preferred that cells of the central nervous system, preferably the brain, of a subject are treated in such a way. Cell penetration can be performed by known strategies such as coupling of antisense nucleic acids and derivatives thereof to carrier particles, or the above described techniques. Strategies for administering targeted therapeutic oligo- deoxynucleotides are known to those of skill in the art (see e.g. Wickstrom, Trends Biotechnol 1992, 10: 281-287). In some cases, delivery can be performed by mere topical application. Further approaches are directed to intracellular expression of antisense RNA. In this strategy, cells are transformed ex vivo with a recombinant gene that directs the synthesis of an RNA that is complementary to a region of target nucleic acid. Therapeutical use of intracellularly expressed antisense RNA is procedurally similar to gene therapy. A recently developed method of regulating the intracellular expression of genes by the use of double-stranded RNA, known variously as RNA interference (RNAi), can be another effective approach for nucleic acid therapy (Hannon, Nature 2002, 418:244-251 ).
In further preferred embodiments, the method comprises grafting donor cells into the central nervous system, preferably the brain, of said subject, or donor cells preferably treated so as to minimize or reduce graft rejection, wherein said donor cells are genetically modified by insertion of at least one transgene encoding said agent or agents. Said transgene might be carried by a viral vector, in particular a retroviral vector. The transgene can be inserted into the donor cells by a nonviral physical transfection of DNA encoding a transgene, in particular by microinjection. Insertion of the transgene can also be performed by electroporation, chemically mediated transfection, in particular calcium phosphate transfection or liposomal mediated transfection (see McCelland and Pardee, Expression Genetics: Accelerated and High-Throughput Methods, Eaton Publishing, Natick, MA, 1999). In preferred embodiments, said agent for treating and preventing a neurodegenerative disease, in particular AD, is a therapeutic protein which can be administered to said subject, preferably a human, by a process comprising introducing subject cells into said subject, said subject cells having been treated in vitro to insert a DNA segment encoding said therapeutic protein, said subject cells expressing in vivo in said subject a therapeutically effective amount of said therapeutic protein. Said DNA segment can be inserted into said cells in vitro by a viral vector, in particular a retroviral vector.
Methods of treatment, according to the present invention, comprise the application of therapeutic cloning, transplantation, and stem cell therapy using embryonic stem cells or embryonic germ cells and neuronal adult stem cells, combined with any of the previously described cell- and gene therapeutic methods. Stem cells may be totipotent or pluripotent. They may also be organ-specific. Strategies for repairing diseased and/or damaged brain cells or tissue comprise (i) taking donor cells from an adult tissue. Nuclei of those cells are transplanted into unfertilized egg cells from which the genetic material has been removed. Embryonic stem cells are isolated from the blastocyst stage of the cells which underwent somatic cell nuclear transfer. Use of differentiation factors then leads to a directed development of the stem cells to specialized cell types, preferably neuronal cells (Lanza et al., Nature Medicine 1999, 9: 975-977), or (ii) purifying adult stem cells, isolated from the central nervous system, or from bone marrow (mesenchymal stem cells), for in vitro expansion and subsequent grafting and transplantation, or (iii) directly inducing endogenous neural stem cells to proliferate, migrate, and differentiate into functional neurons (Peterson DA, Curr. Opin. Pharmacol. 2002, 2: 34-42). Adult neural stem cells are of great potential for repairing damaged or diseased brain tissues, as the germinal centers of the adult brain are free of neuronal damage or dysfunction (Colman A, Drug Discovery World 2001 , 7: 66-71 ).
In preferred embodiments, the subject for treatment or prevention, according to the present invention, can be a human, an experimental animal, e.g. a mouse or a rat, a domestic animal, or a non-human primate. The experimental animal can be an animal model for a neurodegenerative disorder, e.g. a transgenic mouse and/or a knock-out mouse with an AD-type neuropathology. In a further aspect, the invention features a modulator of an activity, or a level, or both said activity and said level of at least one substance which is selected from the group consisting of (i) the gene coding for THBP, and/or (ii) a transcription product of the gene coding for THBP, and/or (iii) a translation product of the gene coding for THBP, and/or (iv) a fragment, or derivative, or variant of (i) to (iii).
In an additional aspect, the invention features a pharmaceutical composition comprising said modulator and preferably a pharmaceutical carrier. Said carrier refers to a diluent, adjuvant, excipient, or vehicle with which the modulator is administered.
In a further aspect, the invention features a modulator of an activity, or a level, or both said activity and said level of at least one substance which is selected from the group consisting of (i) the gene coding for THBP, and/or (ii) a transcription product of the gene coding for THBP, and/or (iii) a translation product of the gene coding. for THBP, and/or (iv) a fragment, or derivative, or variant of (i) to (iii) for use in a pharmaceutical composition.
In another aspect, the invention provides for the use of a modulator of an activity, or a level, or both said activity and said level of at least one substance which is selected from the group consisting of (i) the gene coding for THBP, and/or (ii) a transcription product of the gene coding for THBP, and/or (iii) a translation product of the gene coding for THBP, and/or (iv) a fragment, or derivative, or variant of (i) to (iii) for a preparation of a medicament for treating or preventing a neurodegenerative disease, in particular AD.
In one aspect, the present invention also provides a kit comprising one or more containers filled with a therapeutically or prophylactically effective amount of said pharmaceutical composition.
In a further aspect, the invention features a recombinant, non-human animal comprising a non-native THBP gene sequence, or a fragment, or a derivative, or variant thereof. The generation of said recombinant, non-human animal comprises (i) providing a gene targeting construct containing said gene sequence and a selectable marker sequence, and (ii) introducing said targeting construct into a stem cell of a non-human animal, and (iii) introducing said non-human animal stem cell into a non-human embryo, and (iv) transplanting said embryo into a pseudopregnant non-human animal, and (v) allowing said embryo to develop to term, and (vi) identifying a genetically altered non-human animal whose genome comprises a modification of said gene sequence in both alleles, and (vii) breeding the genetically altered non-human animal of step (vi) to obtain a genetically altered non-human animal whose genome comprises a modification of said endogenous gene, wherein said gene is mis-expressed, or under-expressed, or over-expressed, and wherein said disruption or alteration results in said non-human animal exhibiting a predisposition to developing symptoms of a neurodegenerative disease, in particular AD. Strategies and techniques for the generation and construction of such an animal are known to those of ordinary skill in the art (see e.g. Capecchi, Science 1989, 244: 1288-1292 and Hogan et al., Manipulating the Mouse Embryo: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1994 and Jackson and Abbott, Mouse Genetics and Transgenics: A Practical Approach, Oxford University Press, Oxford, England, 1999). It is preferred to make use of such a recombinant non-human animal as an animal model for investigating neurodegenerative diseases, in particular Alzheimer's disease. Such an animal may be useful for screening, testing and validating compounds, agents and modulators in the development of diagnostics and therapeutics to treat neurodegenerative diseases, in particular Alzheimer's disease.
In another aspect, the invention features an assay for screening for a modulator of neurodegenerative diseases, in particular AD, or related diseases and disorders of one or more substances selected from the group consisting of (i) the gene coding for THBP, and/or (ii) a transcription product of the gene coding for THBP, and/or (iii) a translation product of the gene coding for THBP, and/or (iv) a fragment, or derivative, or variant of (i) to (iii). This screening method comprises (a) contacting a cell with a test compound, and (b) measuring the activity, or the level, or both the activity and the level of one or more substances recited in (i) to (iv), and (c) measuring the activity, or the level, or both the activity and the level of said substances in a control cell not contacted with said test compound, and (d) comparing the levels of the substance in the cells of step (b) and (c), wherein an alteration in the activity and/or level of said substances in the contacteά cells indicates that the test compound is a modulator of said diseases and disorders. In one further aspect, the invention features a screening assay for a modulator of neurodegenerative diseases, in particular AD, or related diseases and disorders of one or more substances selected from the group consisting of (i) the gene coding for THBP, and/or (ii) a transcription product of the gene coding for THBP, and/or (iii) a translation product of the gene coding for THBP, and/or (iv) a fragment, or derivative, or variant of (i) to (iii), comprising (a) administering a test compound to a test animal which is predisposed to developing or has already developed symptoms of a neurodegenerative disease or related diseases or disorders, and (b) measuring the activity and/or level of one or more substances recited in (i) to (iv), and (c) measuring the activity and/or level of said substances in a matched control animal which is equally predisposed to developing or has already developed said symptoms of a neurodegenerative disease, and to which animal no such test compound has been administered, and (d) comparing the activity and/or level of the substance in the animals of step (b) and (c), wherein an alteration in the activity and/or level of substances in the test animal indicates that the test compound is a modulator of said diseases and disorders.
In a preferred embodiment, said test animal and/or said control animal is a recombinant, non-human animal which expresses the gene coding for THBP, or a fragment, or a derivative thereof, under the control of a transcriptional regulatory element which is not the native THBP gene transcriptional control regulatory element.
In another embodiment, the present invention provides a method for producing a medicament comprising the steps of (i) identifying a modulator of neurodegenerative diseases by a method of the aforementioned screening assays and (ii) admixing the modulator with a pharmaceutical carrier. However, said modulator may also be identifiable by other types of screening assays.
In another aspect, the present invention provides for an assay for testing a compound, preferably for screening a plurality of compounds, for inhibition of binding between a ligand and THBP, or a fragment, or derivative, or variant thereof. Said screening assay comprises the steps of (i) adding a liquid suspension of said THBP, or a fragment, or derivative, or variant thereof, to a plurality of containers, and (ii) adding a compound or a plurality of compounds to be screened for said inhibition to said plurality of containers, and (iii) adding detectable, preferably fluorescently labelled ligand to said containers, and (iv) incubating said THBP, or said fragment, or derivative or variant thereof, and said compound or plurality of compounds, and said detectable, preferably fluorescently labelled ligand, and (v) measuring the amounts of preferably fluorescence associated with said THBP, or with said fragment, or derivative, or variant thereof, and (vi) determining the degree of inhibition by one or more of said compounds of binding of said ligand to said THBP, or said fragment, or derivative, or variant thereof. It might be preferred to reconstitute said THBP translation product or fragment, or derivative, or variant thereof into artificial liposomes to generate the corresponding proteoliposomes to determine the inhibition of binding between a ligand and said THBP translation product. Methods of reconstitution of membrane proteins from detergent into liposomes are known to those skilled in the art (Banerjee and Datta, Mol. Cell Biochem. 1983, 50: 3-15; Schwarz et al., Biochemistry 1999, 38: 9456-9464; Krivosheev and Usanov, Biochemistry-Moscow 1997, 62: 1064-1073). Instead of utilizing a fluorescently labelled ligand, it might in some aspects be preferred to use any other detectable label known to the person skilled in the art, e.g. radioactive labels, and detect it accordingly. Said method may be useful for the identification of novel compounds as well as for evaluating compounds which have been improved or otherwise optimized in their ability to inhibit the binding of a ligand to a gene product of the gene coding for THBP, or a fragment, or derivative, or variant thereof. One example of a fluorescent binding assay, in this case based on the use of carrier particles, is disclosed and described in patent application WO 00/52451. A further example is the competitive assay method as described in patent WO 02/01226. Preferred signal detection methods for screening assays of the instant invention are described in the following patent applications: WO 96/13744, WO 98/16814, WO 98/23942, WO 99/17086, WO 99/34195, WO 00/66985, WO 01/59436, WO 01/59416.
In one further embodiment, the present invention provides a method for producing a medicament comprising the steps of (i) identifying a compound as an inhibitor of binding between a ligand and a gene product of the gene coding for THBP by the aforementioned inhibitory binding assay and (ii) admixing the compound with a pharmaceutical carrier. However, said compound may also be identifiable by other types of screening assays. In another aspect, the invention features an assay for testing a compound, preferably for screening a plurality of compounds to determine the degree of binding of said compounds to THBP, or to a fragment, or derivative, or variant thereof. Said screening assay comprises, (i) adding a liquid suspension of said THBP, or a fragment, or derivative, or variant thereof, to a plurality of containers, and (ii) adding a detectable, preferably a fluorescently labelled compound or a plurality of detectable, preferably fluorescently labelled compounds to be screened for said binding to said plurality of containers, and (iii) incubating said THBP, or said fragment, or derivative, or variant thereof, and said detectable, preferably fluorescently labelled compound or fluorescently labelled compounds, and (iv) measuring the amounts of preferably fluorescence associated with said THBP, or with said fragment, or derivative, or variant thereof, and (v) determining the degree of binding by one or more of said compounds to said THBP, or said fragment, or derivative, or variant thereof. In this type of assay it might be preferred to use a fluorescent label. However, any other type of detectable label might also be employed. Also in this type of assay it might be preferred to reconstitute a THBP translation product or fragment, or derivative, or variant thereof into artificial liposomes as described in the present invention. Said assay methods may be useful for the identification of novel compounds as well as for evaluating compounds which have been improved or otherwise optimized in their ability to bind to THBP, or a fragment, or derivative, or variant thereof.
In one further embodiment, the present invention provides a method for producing a medicament comprising the steps of (i) identifying a compound as a binder to a gene product of the gene coding for THBP by the aforementioned binding assays and (ii) admixing the compound with a pharmaceutical carrier. However, said compound may also be identifiable by other types of screening assays.
In another embodiment, the present invention provides for a medicament obtainable by any of the methods according to the herein claimed screening assays. In one further embodiment, the instant invention provides for a medicament obtained by any of the methods according to the herein claimed screening assays.
The present invention features a protein molecule, said protein molecule being a translation product of the gene coding for THBP, SEQ ID NO. 2, or a fragment, or derivative, or variant thereof, for use as a diagnostic target for detecting a neurodegenerative disease, in particular Alzheimer's disease.
The present invention further features a protein molecule, said protein molecule being a translation product of the gene coding for THBP, SEQ ID NO. 2, or a fragment, or derivative, or variant thereof, for use as a screening target for reagents or compounds preventing, or treating, or ameliorating a neurodegenerative disease, in particular Alzheimer's disease.
The present invention features an antibody which is specifically immunoreactive with an immunogen, wherein said immunogen is a translation product of the gene coding for THBP, SEQ ID NO. 2, or a fragment, or derivative, or variant thereof. The immunogen may comprise immunogenic or antigenic epitopes or portions Of a translation product of said gene, wherein said immunogenic or antigenic portion of a translation product is a polypeptide, and wherein said polypeptide elicits an antibody response in an animal, and wherein said polypeptide is immunospecifically bound by said antibody. Methods for generating antibodies are well known in the art (see Harlow et al., Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1988). The term "antibody", as employed in the present invention, encompasses all forms of antibodies known in the art, such as polyclonal, monoclonal, chimeric, recombinatorial, anti-idiotypic, humanized, or single chain antibodies, as well as fragments thereof (see Dubel and Breitliπg, Recombinant Antibodies, Wiley-Liss, New York, NY, 1999). Antibodies of the present invention are useful, for instance, in a variety of diagnostic and therapeutic methods, based on state-in-the-art techniques (see Harlow and Lane, Using Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1999 and Edwards R., Immunodiagnostics: A Practical Approach, Oxford University Press, Oxford, England, 1999) such as enzyme-immuno assays (e.g. enzyme-linked immunosorbent assay, ELISA), radioimmuno assays, chemoluminescence-immuno assays, Western-blot, immunoprecipitation and antibody microarrays. These methods involve the detection of translation products of the THBP gene, or fragments, or derivatives, or variants thereof.
In a preferred embodiment of the present invention, said antibodies can be used for detecting the pathological state of a cell in a sample from a subject, comprising immunocytochemical staining of said cell with said antibody, wherein an altered degree of staining, or an altered staining pattern in said cell compared to a cell representing a known health status indicates a pathological state of said cell. Preferably, the pathological state relates to a neurodegenerative disease, in particular to AD. Immunocytochemical staining of a cell can be carried out by a number of different experimental methods well known in the art. It might be preferred, however, to apply an automated method for the detection of antibody binding, wherein the determination of the degree of staining of a cell, or the determination of the cellular or subcellular staining pattern of a cell, or the topological distribution of an antigen on the cell surface or among organelles and other subcellular structures within the cell, are carried out according to the method described in US patent 6150173.
Other features and advantages of the invention will be apparent from the following description of figures and examples which are illustrative only and not intended to limit the remainder of the disclosure in any way.
Figure 1 depicts the brain regions with selective vulnerability to neuronal loss and degeneration in AD. Primarily, neurons within the inferior temporal lobe, the entorhinal cortex, the hippocampus, and the amygdala are subject to degenerative processes in AD (Terry et al., Annals of Neurology 1981 , 10: 184-192). These brain regions are mostly involved in the processing of learning and memory functions. In contrast, neurons within the frontal cortex, the occipital cortex, and the cerebellum remain largely intact and preserved from neurodegenerative processes in AD. Brain tissues from the frontal cortex (F), the temporal cortex (T) and the hippocampus (H) of AD patients and healthy, age-matched control individuals were used for the herein disclosed examples. For illustrative purposes, the image of a normal healthy brain was taken from a publication by Strange (Brain Biochemistry and Brain Disorders, Oxford University Press, Oxford, 1992, p.4).
Figure 2 discloses the initial identification of the differential expression of the gene coding for THBP in a fluorescence differential display screen. The figure shows a clipping of a large preparative fluorescent differential display gel. PCR products from the frontal cortex (F) and the temporal cortex (T) of two healthy control subjects and six AD patients were loaded in duplicate onto a denaturing polyacrylamide gel (from left to right). PCR products were obtained by amplification of the individual cDNAs with the corresponding one-base-anchor oligonucleotide and the specific Cy3 labelled random primers. The arrow indicates the migration position where significant differences in intensity of the signals for a transcription product of the gene coding for THBP derived from frontal cortex as compared to the signals derived from the temporal cortex of AD patients exist. The differential expression reflects a down-regulation of THBP gene transcription in the temporal cortex compared to the frontal cortex of AD patients. Comparing the signals derived from temporal cortex and frontal cortex of healthy non-AD control subjects with each other, no such difference in signal intensity, i.e. no altered expression level can be detected.
Figures 3 and 4 illustrate the verification of the differential expression of the THBP gene in AD brain tissues by quantitative RT-PCR analysis. Quantification of RT-PCR products from RNA samples collected from the frontal cortex (F) and the temporal cortex (T) of AD patients (Figure 3a) and samples from the frontal cortex (F) and the hippocampus (H) of AD patients (Figure 4a) was performed by the LightCycler rapid thermal cycling technique. Likewise, samples of healthy, age-matched control individuals were compared (Figure 3b for frontal cortex and temporal cortex, Figure 4b for frontal cortex and hippocampus). The data were normalized to the combined average values of a set of standard genes which showed no significant differences in their gene expression levels. Said set of standard genes consisted of genes for cyclophilin B, the ribosomal protein S9, the transferrin receptor, GAPDH, and beta- actin. The figures depict the kinetics of amplification by plotting the cycle number against the amount of amplified material as measured by its fluorescence. Note that the amplification kinetics of the THBP cDNAs from both, the frontal and temporal cortices of a normal control individual, and from the frontal cortex and hippocampus of a normal control individual, respectively, during the exponential phase of the reaction are juxtaposed (Figures 3b and 4b, arrowheads), whereas in Alzheimer's disease (Figures 3a and 4a, arrowheads) there is a significant separation of the corresponding curves, indicating a differential expression of the THBP gene in the respective analyzed brain regions.
Figure 5 depicts SEQ ID NO. 1 , the nucieotide sequence of the 395 bp thyroid hormone binding protein (THBP) cDNA fragment, identified and obtained by fluorescence differential display and subsequent cloning. Figure 6 charts the schematic alignment of SEQ ID NO. 1 to the nucieotide sequence of thyroid hormone binding protein (THBP) cDNA (GenBank accession number U85772). The open rectangle represents the open reading frame of the gene coding for THBP, thin bars represent the 5' and 3' untranslated regions (UTRs).
Figure 7 outlines the sequence alignment of SEQ ID NO. 1 to the nucieotide sequence of thyroid hormone binding protein (THBP) cDNA (GenBank accession number U85772).
Figure 8 discloses SEQ ID NO. 2, the polypeptide sequence of human THBP, comprising 314 amino acids (GenBank accession number Q14894).
Figure 9 shows SEQ ID NO. 3, the nucieotide sequence of the human THBP cDNA. The length of the THBP cDNA according to NCBI GenBank entry U85772 is 1228 base pairs.
Figure 10 depicts human pre-central sections labeled with an affinity-purified rabbit anti-THBP antiserum (green signals) raised against a peptide corresponding to amino acids 169 to 183 of THBP. Immunoreactivity of THBP was observed in both the cortex (CT) and the white matter (WM) (Figure 10a, low magnification). In the cortex, THBP immunoreactivity appears in the nuclear membranes and the neuronal cytoplasm. Blood vessels were strongly immunostained (Figure 10b, high magnification). In the white matter, the cytoplasm of the glial cells were found to be immunopositive (Figure 10c, high magnification). A similar immunostaining pattern was obtained by using an antiserum raised against a peptide mapping to amino acids 246 to 260 of THBP. Blue signals indicate nuclei stained with DAPI.
Table 1 lists the gene expression levels in the frontal cortex relative to the temporal cortex for the THBP gene in seven Alzheimer's disease patients, herein identified by internal reference numbers P010, P011 , P012, P014, P016, P017, P019 (1.71 to 4.19 fold) and five healthy, age-matched control individuals, herein identified by internal reference numbers C005, C008, C011 , C012, C014 (0.87 to 1.30 fold). The scatter plot diagram visualizes individual values of the frontal to temporal cortex regulation ratios in control samples (dots) and in AD patient samples (triangles), respectively. The values shown are reciprocal values according to the formula described herein (see below). Table 2 lists THBP gene expression levels in the frontal cortex relative to the hippocampus in six Alzheimer's disease patients, herein identified by internal reference numbers P010, P01 1 , P012, P014, P016, P019 (0.93 to 13.90 fold) and three healthy, age-matched control individuals, herein identified by internal reference numbers C004, C005, C008 (1.27 and 2.18 fold). The scatter plot diagram visualizes individual values of the frontal cortex to hippocampus regulation ratios in control samples (dots) and in AD patient samples (triangles), respectively. The values shown are reciprocal values according to the formula described herein (see below).
EXAMPLE I:
(i) Brain tissue dissection from patients with AD:
Brain tissues from AD patients and age-matched control subjects were collected, on average, within 6 hours post-mortem and immediately frozen on dry ice. Sample sections from each tissue were fixed in paraformaldehyde for histopathological confirmation of the diagnosis. Brain areas for differential expression analysis were identified (see Figure 1 ) and stored at -80 °C until RNA extractions were performed.
(ii) Isolation of total mRNA:
Total RNA was extracted from post-mortem brain tissue by using the RNeasy kit (Qiagen) according to the manufacturer's protocol. The accurate RNA concentration and the RNA quality were determined with the DNA LabChip system using the Agilent 2100 Bioanalyzer (Agilent Technologies). For additional quality testing of the prepared RNA, i.e. exclusion of partial degradation and testing for DNA contamination, specifically designed intronic GAPDH oligonucleotides and genomic DNA as reference control were used to generate a melting curve with the LightCycler technology as described in the supplied protocol by the manufacturer (Roche).
(iii) cDNA synthesis and identification of differentially expressed genes by fluorescence differential display (FDD):
In order to identify changes in gene expression in different tissue, a modified and improved differential display (DD) screening method was employed. The original DD screening method is known to those skilled in the art (Liang and Pardee, Science 1995, 267: 1186-7). This technique compares two populations of RNA and provides clones of genes that are expressed in one population but not in the other. Several samples can be analyzed simultaneously and both up- and down-regulated genes can be identified in the same experiment. By adjusting and refining several steps in the DD method as well as modifying technical parameters, e.g. increasing redundancy, evaluating optimized reagents and conditions for reverse transcription of total RNA, optimizing polymerase chain reactions (PCR) and separation of the products thereof, a technique was developed which allows for highly reproducible and sensitive results. The applied and improved DD technique was described in detail by von der Kammer et al. (Nucleic Acids Research 1999, 27: 221 1-2218). A set of 64 specifically designed random primers were developed (standard set) to achieve a statistically comprehensive analysis of all possible RNA species. Further, the method was modified to generate a preparative DD slab-gel technique, based on the use of fluorescently labelled primers. In the present invention, RNA populations from carefully selected post-mortem brain tissues (frontal and temporal cortex) of AD patients and age-matched control subjects were compared.
As starting material for the DD analysis we used total RNA, extracted as described above (ii). Equal amounts of 0.05 μg RNA each were transcribed into cDNA in 20 μl reactions containing 0.5 mM each dNTP, 1 μl Sensiscript Reverse Transcriptase and 1x RT buffer (Qiagen), 10 U RNase inhibitor (Qiagen) and 1 μM of either one-base- anchor oligonucleotides HT-| -|A, HT-I -J G or HTn C (Liang et al., Nucleic Acids Research 1994, 22: 5763-5764; Zhao et al., Biotechniques 1995, 18: 842-850). Reverse transcription was performed for 60 min at 37°C with a final denaturation step at 93°C for 5 min. 2 μl of the obtained cDNA each was subjected to a polymerase chain reaction (PCR) employing the corresponding one-base-anchor oligonucleotide (1 μM) along with either one of the Cy3 labelled random DD primers (1 μM), 1x GeneAmp PCR buffer (Applied Biosystems), 1 .5 mM MgCl2 (Applied Biosystems), 2 μM dNTP-Mix (dATP, dGTP, dCTP, dTTP Amersham Pharmacia Biotech), 5 % DMSO (Sigma), 1 U AmpliTaq DNA Polymerase (Applied Biosystems) in a 20 μl final volume. PCR conditions were set as follows: one round at 94°C for 30 sec for denaturing, cooling 1 °C/sec down to 40°C, 40°C for 4 min for low- stringency annealing of primer, heating 1 °C/sec up to 72°C, 72°C for 1 min for extension. This round was followed by 39 high-stringency cycles: 94°C for 30 sec, cooling 1 °C/sec down to 60°C, 60°C for 2 min, heating 1 °C/sec up to 72°C, 72°C for 1 min. One final step at 72°C for 5 min was added to the last cycle (PCR cycler: Multi Cycler PTC 200, MJ Research). 8 μl DNA loading buffer were added to the 20 μl PCR product preparation, denatured for 5 min and kept on ice until loading onto a gel. 3.5 μl each were separated on 0.4 mm thick, 6% polyacrylamide (Long Ranger)/ 7 M urea sequencing gels in a slab-gel system (Hitachi Genetic Systems) at 2000 V, 60W, 30 mA, for 1 h 40 min. Following completion of the electrophoresis, gels were scanned with a FMBIO II fluorescence-scanner (Hitachi Genetic Systems), using the appropriate FMBIO II Analysis 8.0 software. A full-scale picture was printed, differentially expressed bands marked, excised from the gel, transferred into 1.5 ml containers, overlayed with 200 μl sterile water and kept at -20°C until extraction. Elution and reamplification of DD products: The differential bands were extracted from the gel by boiling in 200 μl H2O for 10 min, cooling down on ice and precipitation from the supernatant fluids by using ethanol (Merck) and glycogen/sodium acetate (Merck) at -20°C over night, and subsequent centrifugation at 13.000 rpm for 25 min at 4°C. Pellets were washed twice in ice-cold ethanol (80%), resuspended in 10 mM Tris pH 8.3 (Merck) and dialysed against 10 % glycerol (Merck) for 1 h at room temperature on a 0.025 μm VSWP membrane (Millipore). The obtained preparations were used as templates for reamplification by 15 high-stringency cycles in 25-μl PCR mixtures containing the corresponding primer pairs as used for the DD PCR (see above) under identical conditions, with the exception of the initial round at 94°C for 5 min, followed by 15 cycles of: 94°C for 45 sec, 60°C for 45 sec, ramp 1 °C/sec to 70°C for 45 sec, and one final step at 72°C for 5 min.
Cloning and sequencing of DD products: Re-amplified cDNAs were analyzed with the DNA LabChip system (Agilent 2100 Bioanalyzer, Agilent Technologies) and ligated into the pCR-Blunt ll-TOPO vector and transformed into E.coli Top10F' cells (Zero Blunt TOPO PCR Cloning Kit, Invitrogen) according to the manufacturer's instructions. Cloned cDNA fragments were sequenced by commercially available sequencing facilities. The result of one such FDD experiment for the gene coding for thyroid hormone binding protein is shown in Figure 2.
(iv) Confirmation of differential expression by quantitative RT-PCR: Positive corroboration of differential THBP gene expression was performed using the LightCycler technology (Roche). This technique features rapid thermal cyling for the polymerase chain reaction as well as real-time measurement of fluorescent signals during amplification and therefore allows for highly accurate quantification of RT-PCR products by using a kinetic, rather than an endpoint readout. The ratios of THBP cDNAs from the frontal cortex and temporal cortex, and from the frontal cortex and hippocampus, respectively, were determined (relative quantification).
First, a standard curve was generated to determine the efficiency of the PCR with specific primers for the thyroid hormone binding protein (THBP) gene: 5'-GCTGGGAGAAGTGATTAAGGGA-3' and 5'-CAAAGACTTGAACACGGTGGTC-3'.
PCR amplification (95°C and 1 sec, 56βC and 5 sec, and 72°C and 5 sec) was performed in a volume of 20 μl containing LightCycler-FastStart DNA Master SYBR Green I mix (contains FastStart Taq DNA polymerase, reaction buffer, dNTP mix with dUTP instead of dTTP, SYBR Green I dye, and 1 mM MgCI2; Roche), 0.5 μM primers, 2 μl of a cDNA dilution series (final concentration of 40, 20, 10, 5, 1 and 0.5 ng human total brain cDNA; Clontech) and, depending on the primers used, additional 3 M MgCI2. Melting curve analysis revealed a single peak at approximately 83°C with no visible primer di ers. Quality and size of the PCR product were determined with the DNA LabChip system (Agilent 2100 Bioanalyzer, Agilent Technologies). A single peak at the expected size of 67 bp for the gene coding for THBP was observed in the electropherogram of the sample.
In an analogous manner, the PCR protocol was applied to determine the PCR efficiency of a set of reference genes which were selected as a reference standard for quantification. In the present invention, the mean value of five such reference genes was determined: (1 ) cyclophilin B, using the specific primers 5'- ACTGAAGCACTACGGGCCTG-3' and 5'-AGCCGTTGGTGTCTTTGCC-3' except for MgCl2 (an additional 1 mM was added instead of 3 mM). Melting curve analysis revealed a single peak at approximately 87°C with no visible primer dimers. Agarose gel analysis of the PCR product showed one single band of the expected size (62 bp). (2) Ribosomal protein S9 (RPS9), using the specific primers 5'- GGTCAAATTTACCCTGGCCA-3" and 5'- TCTCATCAAGCGTCAGCAGTTC-3' (exception: additional 1 mM MgCI2 was added instead of 3 mM). Melting curve analysis revealed a single peak at approximately 85CC with no visible primer dimers. Agarose gel analysis of the PCR product showed one single band with the expected size (62 bp). (3) beta-actin, using the specific primers 5'- TGGAACGGTGAAGGTGACA-3' and 5'-GGCAAGGGACTTCCTGTAA-3'. Melting curve analysis revealed a single peak at approximately 87°C with no visible primer dimers. Agarose gel analysis of the PCR product showed one single band with the expected size (142 bp). (4) GAPDH, using the specific primers 5'- CGTCATGGGTGTGAACCATG-3' and 5'-GCTAAGCAGTTGGTGGTGCAG-3'. Melting curve analysis revealed a single peak at approximately 83°C with no visible primer dimers. Agarose gel analysis of the PCR product showed one single band with the expected size (81 bp). (5) Transferrin receptor TRR, using the specific primers 5'- GTCGCTGGTCAGTTCGTGATT-3' and 5'-AGCAGTTGGCTGTTGTACCTCTC-3'. Melting curve analysis revealed a single peak at approximately 83°C with no visible primer dimers. Agarose gel analysis of the PCR product showed one single band with the expected size (80 bp).
For calculation of the values, first the logarithm of the cDNA concentration was plotted against the threshold cycle number Ct for the gene coding for thyroid hormone binding protein and the five reference standard genes. The slopes and the intercepts of the standard curves (i.e. linear regressions) were calculated for all genes. In a second step, cDNAs from frontal cortex and temporal cortex, and from frontal cortex and hippocampus, respectively, were analyzed in parallel and normalized to cyclophilin B. The Ct values were measured and converted to ng total brain cDNA using the corresponding standard curves:
10 Λ ( (Ct value - intercept) / slope ) [ng total brain cDNA]
The values for frontal and temporal cortex THBP cDNAs, and the values for frontal cortex and hippocampus THBP cDNAs, respectively, were normalized to cyclophilin B, and the ratios were calculated according to formulas:
THBP temporal [ng] / cyclophilin B temporal [ng] Ratio =
THBP frontal [ng] / cyclophilin B frontal [ng].
THBP hippocampus [ng] / cyclophilin B hippocampus [ng] Ratio =
THBP frontal [ng] / cyclophilin B frontal [ng] In a third step, the set of reference standard genes was analyzed in parallel to determine the mean average value of the temporal to frontal ratios, and of the hippocampal to frontal ratios, respectively, of expression levels of the reference standard genes for each individual brain sample. As cyclophilin B was analyzed in step 2 and step 3, and the ratio from one gene to another gene remained constant in different runs, it was possible to normalize the values for the THBP gene to the mean average value of the set of reference standard genes instead of normalizing to one single gene alone. The calculation was performed by dividing the respective ratio shown above by the deviation of cyclophilin B from the mean value of all housekeeping genes. The results of such quantitative RT-PCR analysis for the gene coding for THBP are shown in Figure 3 and 4.
(v) Immunohistochemistry:
For immunofluorescence staining of THBP in human brain, frozen sections were prepared with a cryostat (Leica CM3050S) from post-mortem pre-central gyrus of a donor person and fixed in 4% PFA for 20 min. After washing in PBS, the sections were pre-incubated with blocking buffer (10% normal goat serum, 0.2% Triton X-100 in PBS) for 30 min and then incubated with affinity-purified rabbit anti-THBP antisera (1 :40 diluted in blocking buffer; custom-made, Biogenes, Berlin, Germany) overnight at 4°C. After rinsing three times in 0.1 % Triton X-100/PBS, the sections were incubated with FITC-conjugated goat anti-rabbit IgG (1 :150 diluted in 1 % BSA/PBS) for 2 hours at room temperature and then again washed in PBS. Staining of the nuclei was performed by incubation of the sections with 5μM DAPI in PBS for 3 min (blue signal). In order to block the autofluoresence of lipofuscin in human brain, the sections were treated with 1 % Sudan Black B in 70% ethanol for 2-10 min at room temperature and then sequentially dipped in 70% ethanol, destilled water and PBS. The sections were coverslipped with 'Vectrashield' mounting medium (Vector Laboratories, Burlingame, CA) and observed under an inverted microscope (1X81 , Olympus Optical). The digital images were captured with the appropriate software (AnalySiS, Olympus Optical).

Claims

1 . A method of diagnosing or prognosticating a neurodegenerative disease in a subject, or determining whether a subject is at increased risk of developing said disease, comprising: determining a level and/or an activity of
(i) a transcription product of the gene coding for THBP, and/or
(ii) a translation product of the gene coding for THBP, and/or
(iii) a fragment, or derivative, or variant of said transcription or translation product, in a sample from said subject and comparing said level and/or said activity to a reference value representing a known disease or health status, thereby diagnosing or prognosticating said neurodegenerative disease in said subject, or determining whether said subject is at increased risk of developing said neurodegenerative disease.
2. The method according to claim 1 wherein said gene coding for THBP, thyroid hormone binding protein, is the gene coding for SEQ ID NO. 2, NADPH-dependent thyroid hormone binding protein, NCBI Genbank accession number: Q14894.
3. The method according to claim 1 or 2 wherein said neurodegenerative disease is Alzheimer's disease.
4. A kit for diagnosing or prognosticating a neurodegenerative disease, in particular Alzheimer's disease, in a subject, or determining the propensity or predisposition of a subject to develop such a disease, said kit comprising:
(a) at least one reagent which is selected from the group consisting of (i) reagents that selectively detect a transcription product of the gene coding for THBP and (ii) reagents that selectively detect a translation product of the gene coding for THBP, and
(b) an instruction for diagnosing, or prognosticating a neurodegenerative disease, in particular Alzheimer's disease, or determining the propensity or predisposition of a subject to develop such a disease by (i) detecting a level, or an activity, or both said level and said activity, of said transcription product and/or said translation product of the gene coding for THBP in a sample from said subject; and (ii) diagnosing or prognosticating a neurodegenerative disease, in particular Alzheimer's disease, or determining the propensity or predisposition of said subject to develop such a disease, wherein a varied level, or activity, or both said level and said activity, of said transcription product and/or said translation product compared to a reference value representing a known health status; or a level, or activity, or both said level and said activity, of said transcription product and/or said translation product similar or equal to a reference value representing a known disease status indicates a diagnosis or prognosis of a neurodegenerative disease, in particular Alzheimer's disease, or an increased propensity or predisposition of developing such a disease.
5. A modulator of an activity and/or of a level of at least one substance which is selected from the group consisting of
(i) the gene coding for THBP, and/or
(ii) a transcription product of the gene coding for THBP, and/or
(iii) a translation product of the gene coding for THBP, and/or
(iv) a fragment, or derivative, or variant of (i) to (iii).
6. A recombinant, non-human animal comprising a non-native THBP gene sequence or a fragment, or a derivative, or a variant thereof, said animal being obtainable by: (i) providing a gene targeting construct comprising said gene sequence and a selectable marker sequence, and
(ii) introducing said targeting construct into a stem cell of a non-human animal, and
(iii) introducing said non-human animal stem cell into a non-human embryo, and
(iv) transplanting said embryo into a pseudopregnant non-human animal, and
(v) allowing said embryo to develop to term, and
(vi) identifying a genetically altered non-human animal whose genome comprises a modification of said gene sequence in both alleles, and
(vii) breeding the genetically altered non-human animal of step (vi) to obtain a genetically altered non-human animal whose genome comprises a modification of said endogenous gene, wherein said disruption results in said non-human animal exhibiting a predisposition to developing symptoms of a neurodegenerative disease or related diseases or disorders.
7. An assay for screening for a modulator of neurodegenerative diseases, in particular Alzheimer's disease, or related diseases or disorders of one or more substances selected from the group consisting of
(i) the gene coding for THBP, and/or (ii) a transcription product of the gene coding for THBP, and/or (iii) a translation product of the gene coding for THBP, and/or (iv) a fragment, or derivative, or variant of (i) to (iii), said method comprising:
(a) contacting a cell with a test compound;
(b) measuring the activity and/or level of one or more substances recited in (i) to (iv);
(c) measuring the activity and/or level of one or more substances recited in (i) to (iv) in a control cell not contacted with said test compound; and comparing the levels and/or activities of the substance in the cells of step (b) and (c), wherein an alteration in the activity and/or level of substances in the contacted cells indicates that the test compound is a modulator of said diseases or disorders.
8. A method of screening for a modulator of neurodegenerative diseases, in particular Alzheimer's disease, or related diseases or disorders of one or more substances selected from the group consisting of
(i) the gene coding for THBP, and/or
(ii) a transcription product of the gene coding for THBP, and/or
(iii) a translation product of the gene coding for THBP, and/or
(v) a fragment, or derivative, or variant of (i) to (iii), said method comprising:
(a) administering a test compound to a test animal which is predisposed to developing or has already developed symptoms of a neurodegenerative disease or related diseases or disorders in respect of the substances recited in (i) to (iv);
(b) measuring the activity and/or level of one or more substances recited in (i) to (iv);
(c) measuring the activity and/or level of one or more substances recited in (i) or (iv) in a matched control animal which is predisposed to developing or has already developed symptoms of a neurodegenerative disease or related diseases or disorders in respect to the substances recited in (i) to (iv) and to which animal no such test compound has been administered;
(d) comparing the activity and/or level of the substance in the animals of step (b) and (c), wherein an alteration in the activity and/or level of substances in the test animal indicates that the test compound is a modulator of said diseases or disorders.
9. The method according to claim 8 wherein said test animal and/or said control animal is a recombinant animal which expresses THBP, or a fragment, or a derivative, or a variant thereof, under the control of a transcriptional control element which is not the native THBP gene transcriptional control element.
10. An assay for testing a compound, preferably for screening a plurality of compounds to determine the degree of binding of said compounds to THBP, or to a fragment, or derivative, or variant thereof, said assay comprising the steps of:
(i) adding a liquid suspension of said THBP, or a fragment, or derivative, or variant thereof, to a plurality of containers; (ii) adding a detectable, in particular a fluorescently labelled compound or a plurality of detectable, in particular fluorescently labelled compounds to be screened for said binding to said plurality of containers; (iii) incubating said THBP, or said fragment, or derivative, or variant thereof, and said detectable, in particular fluorescently labelled compound or fluorescently labelled compounds; (iv) measuring amounts of preferably fluorescence associated with said THBP, or with said fragment, or derivative, or variant thereof; and (v) determining the degree of binding by one or more of said compounds to said
THBP, or said fragment, or derivative, or variant thereof.
1 1. A protein molecule, said protein molecule being a translation product of the gene coding for THBP, SEQ ID NO. 2, or a fragment, or derivative, or variant thereof, for use as a diagnostic target for detecting a neurodegenerative disease, preferably Alzheimer's disease.
12. A protein molecule, said protein molecule being a translation product of the gene coding for THBP, SEQ ID NO. 2, or a fragment, or derivative, or variant thereof, for use as a screening target for reagents or compounds preventing, or treating, or ameliorating a neurodegenerative disease, preferably Alzheimer's disease.
13. Use of an antibody specifically immunoreactive with an immunogen, wherein said immunogen is a translation product of the gene coding for THBP, SEQ ID NO. 2, or a fragment, or derivative, or variant thereof, for detecting the pathological state of a cell in a sample from a subject, comprising immunocytochemical staining of said cell with said antibody, wherein an altered degree of staining, or an altered staining pattern in said cell compared to a cell representing a known health status indicates a pathological state of said cell.
14. Use of an antibody according to claim 13, wherein said pathological state relates to a neurodegenerative disease.
PCT/EP2003/009536 2002-09-02 2003-08-28 Diagnostic and therapeutic use of thyroid hormone binding protein for neurodegenerative diseases WO2004020666A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU2003271568A AU2003271568A1 (en) 2002-09-02 2003-08-28 Diagnostic and therapeutic use of thyroid hormone binding protein for neurodegenerative diseases
EP03753364A EP1534856A2 (en) 2002-09-02 2003-08-28 Diagnostic and therapeutic use of thyroid hormone binding protein for neurodegenerative diseases

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP02019557.4 2002-09-02
EP02019557 2002-09-02
US40724902P 2002-09-03 2002-09-03
US60/407,249 2002-09-03

Publications (2)

Publication Number Publication Date
WO2004020666A2 true WO2004020666A2 (en) 2004-03-11
WO2004020666A3 WO2004020666A3 (en) 2004-09-02

Family

ID=56290477

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2003/009536 WO2004020666A2 (en) 2002-09-02 2003-08-28 Diagnostic and therapeutic use of thyroid hormone binding protein for neurodegenerative diseases

Country Status (3)

Country Link
EP (1) EP1534856A2 (en)
AU (1) AU2003271568A1 (en)
WO (1) WO2004020666A2 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4980281A (en) * 1988-02-10 1990-12-25 Housey Gerard M Method of screening for protein inhibitors and activators
WO2000067033A1 (en) * 1999-05-03 2000-11-09 Evotec Biosystems Ag Methods of diagnosing or treating alzheimer's disease
WO2002016636A2 (en) * 2000-08-24 2002-02-28 Evotec Neurosciences Gmbh Diagnostic and therapeutic use of a caveolae-associated integral membrane protein for alzheimer's disease and related neurodegenerative disorders
WO2002022867A2 (en) * 2000-09-13 2002-03-21 Evotec Neurosciences Gmbh Diagnostic and therapeutic use of a phosphoprotein enriched in astrocytes for alzheimer's disease and related neurodegenerative disorders
WO2002057496A2 (en) * 2001-01-18 2002-07-25 Socratech L.L.C. Gene expression profiling of endothelium in alzheimer's disease

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4980281A (en) * 1988-02-10 1990-12-25 Housey Gerard M Method of screening for protein inhibitors and activators
WO2000067033A1 (en) * 1999-05-03 2000-11-09 Evotec Biosystems Ag Methods of diagnosing or treating alzheimer's disease
WO2002016636A2 (en) * 2000-08-24 2002-02-28 Evotec Neurosciences Gmbh Diagnostic and therapeutic use of a caveolae-associated integral membrane protein for alzheimer's disease and related neurodegenerative disorders
WO2002022867A2 (en) * 2000-09-13 2002-03-21 Evotec Neurosciences Gmbh Diagnostic and therapeutic use of a phosphoprotein enriched in astrocytes for alzheimer's disease and related neurodegenerative disorders
WO2002057496A2 (en) * 2001-01-18 2002-07-25 Socratech L.L.C. Gene expression profiling of endothelium in alzheimer's disease

Non-Patent Citations (24)

* Cited by examiner, † Cited by third party
Title
"Consensus report of the Working Group on: "Molecular and Biochemical Markers of Alzheimer's Disease". The Ronald and Nancy Reagan Research Institute of the Alzheimer's Association and the National Institute on Aging Working Group." NEUROBIOLOGY OF AGING. UNITED STATES 1998 MAR-APR, vol. 19, no. 2, March 1998 (1998-03), pages 109-116, XP002243838 ISSN: 0197-4580 *
BROWN VANESSA M ET AL: "High-throughput imaging of brain gene expression." GENOME RESEARCH. UNITED STATES FEB 2002, vol. 12, no. 2, February 2002 (2002-02), pages 244-254, XP002231816 ISSN: 1088-9051 *
CALLAHAN L M ET AL: "Analysis of message expression in single neurons of Alzheimer's disease brain." NEUROBIOLOGY OF AGING. UNITED STATES 1998 JAN-FEB, vol. 19, no. 1 Suppl, January 1998 (1998-01), pages S99-105, XP002243837 ISSN: 0197-4580 *
CHOW N ET AL: "Expression profiles of multiple genes in single neurons of Alzheimer's disease." PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA. UNITED STATES 4 AUG 1998, vol. 95, no. 16, 4 August 1998 (1998-08-04), pages 9620-9625, XP002243836 ISSN: 0027-8424 *
DATABASE EMBL GENBANK [Online] XP002231826 Database accession no. BC018061 *
DATABASE EMBL GENBANK [Online] XP002231827 Database accession no. BC008398 *
DATABASE EMBL GENBANK [Online] XP002231828 Database accession no. L02950 *
DATABASE EMBL GENBANK [Online] XP002231830 Database accession no. U85772 cited in the application *
DATABASE NCBI GENBANK [Online] XP002275538 Database accession no. Q14894 cited in the application *
DATABASE WPI Section Ch, Week 199311 Derwent Publications Ltd., London, GB; Class B04, AN 1993-093573 XP002243840 *
DATABASE WPI Section Ch, Week 199319 Derwent Publications Ltd., London, GB; Class B04, AN 1993-159482 XP002243839 *
DOYU M ET AL: "Gene expression profile in Alzheimer's brain screened by molecular indexing." BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH. NETHERLANDS 19 FEB 2001, vol. 87, no. 1, 19 February 2001 (2001-02-19), pages 1-11, XP002231818 ISSN: 0169-328X *
GINSBERG S D ET AL: "Expression profile of transcripts in Alzheimer's disease tangle-bearing CA1 neurons." ANNALS OF NEUROLOGY. UNITED STATES JUL 2000, vol. 48, no. 1, July 2000 (2000-07), pages 77-87, XP008008699 ISSN: 0364-5134 *
HATA R ET AL: "Up-regulation of calcineurin Abeta mRNA in the Alzheimer's disease brain: assessment by cDNA microarray." BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS. UNITED STATES 8 JUN 2001, vol. 284, no. 2, 8 June 2001 (2001-06-08), pages 310-316, XP002231817 ISSN: 0006-291X *
KIM R Y ET AL: "mu-crystallin is a mammalian homologue of Agrobacterium ornithine cyclodeaminase and is expressed in human retina." PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA. UNITED STATES 1 OCT 1992, vol. 89, no. 19, 1 October 1992 (1992-10-01), pages 9292-9296, XP002231825 ISSN: 0027-8424 cited in the application *
LORING J F ET AL: "A gene expression profile of Alzheimer's disease." DNA AND CELL BIOLOGY. UNITED STATES NOV 2001, vol. 20, no. 11, November 2001 (2001-11), pages 683-695, XP001083747 ISSN: 1044-5498 *
PASINETTI G M ET AL: "FROM CDNA MICROARRAYS TO HIGH-THROUGHPUT PROTEOMICS. IMPLICATIONS IN THE SEARCH FOR PREVENTIVE INITIATIVES TO SLOW THE CLINICAL PROGRESSION OF ALZHEIMER'S DISEASE DEMENTIA" RESTORATIVE NEUROLOGY AND NEUROSCIENCE, XX, XX, vol. 18, no. 2/3, 2001, pages 137-142, XP009006184 *
PASINETTI G M: "Use of cDNA microarray in the search for molecular markers involved in the onset of Alzheimer's disease dementia." JOURNAL OF NEUROSCIENCE RESEARCH. UNITED STATES 15 SEP 2001, vol. 65, no. 6, 15 September 2001 (2001-09-15), pages 471-476, XP002231820 ISSN: 0360-4012 *
RYAN M ET AL: "Indexing-based differential display--studies on post-mortem Alzheimer's brains." BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH. NETHERLANDS 31 MAR 2001, vol. 88, no. 1-2, 31 March 2001 (2001-03-31), pages 199-202, XP002231819 ISSN: 0169-328X *
SCHONBERGER S J ET AL: "PROTEOMIC ANALYSIS OF THE BRAIN IN ALZHEIMER'S DISEASE: MOLECULAR PHENOTYPE OF A COMPLEX DISEASE PROCESS" PROTEOMICS, XX, XX, vol. 1, no. 12, December 2001 (2001-12), pages 1519-1528, XP009006180 *
TSUJI T ET AL: "ANALYSIS OF THE PROTEOMIC PROFILING OF BRAIN TISSUE IN ALZHEIMER'S DISEASE" DISEASE MARKERS, WILEY, CHICHESTER, GB, vol. 17, no. 4, 2001, pages 247-257, XP009006179 ISSN: 0278-0240 *
VAN LEUVEN F: "Single and multiple transgenic mice as models for Alzheimer's disease." PROGRESS IN NEUROBIOLOGY. ENGLAND JUN 2000, vol. 61, no. 3, June 2000 (2000-06), pages 305-312, XP001023899 ISSN: 0301-0082 *
VIÉ M P ET AL: "Purification, molecular cloning, and functional expression of the human nicodinamide-adenine dinucleotide phosphate-regulated thyroid hormone-binding protein." MOLECULAR ENDOCRINOLOGY (BALTIMORE, MD.) UNITED STATES OCT 1997, vol. 11, no. 11, October 1997 (1997-10), pages 1728-1736, XP002231823 ISSN: 0888-8809 *
WELLS J M: "FUTURE DIRECTIONS IN THE ANALYSIS OF GENE EXPRESSION IN ALZHEIMER DISEASE" ALZHEIMER DISEASE AND ASSOCIATED DISORDERS, RAVEN PRESS, NEW YORK, NY, US, vol. 13, no. SUPPL 1, April 1999 (1999-04), pages S78-S81, XP009006178 ISSN: 0893-0341 *

Also Published As

Publication number Publication date
WO2004020666A3 (en) 2004-09-02
AU2003271568A1 (en) 2004-03-19
EP1534856A2 (en) 2005-06-01

Similar Documents

Publication Publication Date Title
EP1776591B1 (en) Diagnostic and therapeutic use of a plasma membrane atpase
WO2004070388A1 (en) Diagnostic and therapeutic use of scn2b protein for neurodegenerative diseases
US20060259990A1 (en) Diagnostic and therapeutic use of tb2 gene and protein for neurodegenerative diseases
EP1514118A2 (en) Diagnostic and therapeutic use of ras-gtpase-activating sh3-domain-binding protein 2 (g3bp2) for neurodegenerative diseases
EP1497661A1 (en) Diagnostic and therapeutic use of ensadin-0477 gene and protein for neurodegenerative diseases
WO2004038411A2 (en) Diagnostic and therapeutic use of ensadin-0289 gene and protein for neurodegenerative diseases
US20060052280A1 (en) Diagnostic and therapeutic use of a golgi protein for neurodegenerative diseases
US20070166718A1 (en) Diagnostic and therapeutic use of mal2 gene and protein for neurodegenerative diseases
US20070269800A9 (en) Diagnostic and therapeutic use of foap-13 polynucleotides and polypeptides for neurodegenerative diseases
EP1565751A1 (en) Diagnostic and therapeutic use of h-rev107 protein for alzheimer disease
WO2004035823A2 (en) Diagnostic and therapeutic use of the nicotinamide mononucleotide adenylytransferase 2 (nmnat-2) gene and protein for neurodegenerative diseases
EP1490694B1 (en) Camp-regulated phosphorprotein for diagnostic and therapeutic use in neurodegenerative diseases
EP1534856A2 (en) Diagnostic and therapeutic use of thyroid hormone binding protein for neurodegenerative diseases
US20060073480A1 (en) Diagnostic and therapeutic use of vault polynucleotides and proteins for neurodegenerative diseases
EP1561117A1 (en) Diagnostic and therapeutic use of arl7 for alzheimer's disease
US20050153295A1 (en) Diagnostic and therapeutic use of human maguin proteins and nucleic acids for neurodegenerative diseases
WO2003104811A2 (en) Diagnostic and therapeutic use of steroidogenic acute regulatory protein for neurodegenerative diseases
US20060051757A1 (en) Diagnostic and therapeutic use of ensadin-0477 gene and protein for neurodegenerative diseases
US20060294602A1 (en) Diagnostic and therapeutic use of a rab family gtp-binding protein for neurodegenerative diseases
US20060160728A1 (en) Diagnostic and therapeutic use of ensandin-0138 gene and protein for neurodegenerative diseases
WO2003080661A1 (en) Diagnostic and therapeutic use of human maguin proteins and nucleic acids for neurodegenerative diseases
EP1771578A2 (en) Diagnostic and therapeutic use of slim3 for neurodegenerative diseases

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2003753364

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2003753364

Country of ref document: EP

NENP Non-entry into the national phase in:

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP