WO1995030693A1 - Proteines et genes de differentiation neurogene (neuro d) - Google Patents

Proteines et genes de differentiation neurogene (neuro d) Download PDF

Info

Publication number
WO1995030693A1
WO1995030693A1 PCT/US1995/005741 US9505741W WO9530693A1 WO 1995030693 A1 WO1995030693 A1 WO 1995030693A1 US 9505741 W US9505741 W US 9505741W WO 9530693 A1 WO9530693 A1 WO 9530693A1
Authority
WO
WIPO (PCT)
Prior art keywords
neurod
seq
ser
glu
leu
Prior art date
Application number
PCT/US1995/005741
Other languages
English (en)
Inventor
Jacqueline E. Lee
Stanley M. Hollenberg
Stephen J. Tapscott
Harold M. Weintraub
Original Assignee
Fred Hutchinson Cancer Research Center
Weintraub, Nancy
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 Fred Hutchinson Cancer Research Center, Weintraub, Nancy filed Critical Fred Hutchinson Cancer Research Center
Priority to EP95919759A priority Critical patent/EP0759938A4/fr
Priority to JP7529169A priority patent/JPH10503363A/ja
Priority to AU25446/95A priority patent/AU2544695A/en
Publication of WO1995030693A1 publication Critical patent/WO1995030693A1/fr
Priority to US08/910,973 priority patent/US5795723A/en
Priority to US09/499,227 priority patent/US6444463B1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy

Definitions

  • Neurogenic Differentiation Genes and Proteins
  • the invention relates to molecular biology and in particular to genes and proteins involved in vertebrate neural development. Background of the Invention
  • bHLH proteins form homodimeric and heterodimeric complexes binding DNA in the 5' regulatory regions of genes controlling expression.
  • mammalian MyoD and Drosophila AS-C are presently thought to play developmental roles in muscle development and in sensory organ development, respectively. Both proteins are thought to exert their effects by binding 5 1 regulatory nucleotide sequences in genes that seem specifically determinative of cellular differentiation and fate.
  • MyoD and AS-C remain largely unknown, as are the molecular details of the developmental pathways regulated by these genes.
  • the presently disclosed NeuroD represents a new sub-family of bHLH proteins and is implicated in vertebrate neuronal development.
  • NeuroD encodes a protein that is a distinctive member of the bHLH family.
  • the present invention provides a family of NeuroD proteins that share a highly conserved HLH region.
  • neuroD is highly expressed in the neurogenic derivatives of neural crest cells, the cranial and dorsal root ganglia, and postmitotic cells in the central nervous system (CNS).
  • CNS central nervous system
  • neuroD is expressed transiently and concomitant with neuronal differentiation in differentiating neurons in sensory organs such as in nasal epithelium and retina.
  • Xenopus embryos ectopic expression of neuroD in non-neuronal cells induced formation of neurons.
  • a representative nucleotide sequence of murine neuroD is shown in SEQ ID
  • the HLH coding domain of murine neuroD resides between nucleotides 577 and 696 in SEQ ID NO:l.
  • the deduced amino acid sequence of murine NeuroD is shown in SEQ ID NO:2.
  • a representative nucleotide sequence of Xenopus neuroD is shown in SEQ ID NO:3.
  • the HLH coding domain of Xenopus neuroD resides between nucleotides 376 and 495 in SEQ ID NO:3.
  • the deduced amino acid sequence of murine NeuroD is shown in SEQ ID NO:4.
  • FIGURE 1 schematically depicts the domain structure of the murine and Xenopus NeuroD bHLH proteins.
  • Tissue-specific bHLH proteins that regulate early neuroectodermal differentiation were discovered using expression cloning and screening assays designed to identify possible bHLH proteins capable of interacting with the protein product of the Drosophila daughterless gene. These proteins belong to a family of proteins that share conserved residues in the HLH region.
  • NeuroD is a member of a novel protein family and is found to be transiently expressed in differentiating neurons during embryogenesis. Its expression is also detected in adult brain, in the granule layer of hippocampus and cerebellum.
  • NeuroD contains the basic helix-loop-helix (bHLH) domain structure that has been implicated in the binding of bHLH proteins to upstream recognition sequences and activation of downstream target genes.
  • the present invention provides representative NeuroD proteins, which include the murine NeuroD protein of SEQ ID NO:2 and the amphibian NeuroD protein of SEQ ID NO:4. Based on homology with other bHLH proteins, the bHLH domain for the murine NeuroD protein is predicted to reside between amino acids 102 and 155 of SEQ ID NO:2, and between amino acids 101 and 157 of SEQ ID NO:4 for the amphibian NeuroD protein.
  • the present invention provides the identification of the human neuroD and, in addition, provides an unexpected homologous gene of the same family based on the almost identical sequence across the HLH domain shared between the two human genes at the amino acid level.
  • NeuroD proteins are transcriptional activators that control transcription of downstream target genes that cause neuronal progenitors to differentiate into mature neurons.
  • NeuroD proteins are expressed in differentiating neurons and are capable of causing the conversion of non-neuronal cells into neurons.
  • the present invention encompasses NeuroD variants that, for example, are modified in a manner that results in a NeuroD protein capable of binding to its recognition site, but unable to activate downstream genes.
  • NeuroD proteins encompass proteins retrieved from naturally occurring materials and closely related, functionally similar proteins retrieved by antisera specific to NeuroD, and recombinantly expressed proteins encoded by genetic materials (DNA, RNA, cDNA) retrieved on the basis of their similarity to the unique regions in the neuroD family of genes.
  • the present invention discloses representative isolated and purified polynucleotide molecules encoding proteins of the NeuroD family.
  • Representative polynucleotide molecules encoding NeuroD include the sequences presented in SEQ ID NOS:l, 3, 8, and 10.
  • Polynucleotide molecules encoding NeuroD include those sequences resulting in minor genetic polymorphisms, differences between species, those that contain amino acid substitutions, additions, and/or deletions.
  • Such changes may also • be directed towards endogenous neuroD sequences using, for example, gene therapy methods to alter the gene product.
  • NeuroD proteins of the present invention are capable of inducing the expression of neuronal-specific genes, such as N-CAM, ⁇ -tubulin, and Xen-1, neurofilament M (NF-M), Xen-2, tanabin-1, shaker-1, and frog HSCL, in a frog embryo.
  • neuronal-specific genes such as N-CAM, ⁇ -tubulin, and Xen-1, neurofilament M (NF-M), Xen-2, tanabin-1, shaker-1, and frog HSCL.
  • NeuroD activity may be detected when NeuroD is ectopically expressed in frog oocytes following, for example, injection of neuroD RNA into one of the two cells in a two-cell stage Xenopus embryo, and monitoring expression of neuronal-specific genes in the injected as compared to un-injected side of the embryo by immunochemistry or in situ hybridization.
  • “Over-expression” means an increased level of NeuroD protein or neuroD
  • the present invention provides isolated and purified polynucleotide molecules encoding NeuroD and other members of the NeuroD family.
  • the disclosed sequences may be used to identify and isolate neuroD polynucleotide molecules from suitable host cells such as canine, ovine, bovine, caprine, lagomorph, or avian.
  • the nucleotide sequences encoding the HLH region may be used to identify poylnycleotide molecules encoding other proteins of the NeuroD family.
  • Complementary DNA molecules encoding NeuroD family members may be obtained by constructing a cDNA library mRNA from, for example, fetal brain.
  • DNA molecules encoding NeuroD family members may be isolated from such a library using the disclosed sequences in standard hybridization techniques (e.g., Sambrook et al., ibid., and Bothwell, Yancopoulos and Alt, ibid.) or by amplification of sequences using polymerase chain reaction (PCR) amplification (e.g, Loh et al. Science 243: 217-222, 1989; Frohman et al., Proc. Natl. Acad. Sci. USA 85: 8998-9002, 1988; and Erlich (ed.), PCR Technology: Principles and Applications for DNA Amplification, Stockton Press, 1989; which are incorporated by reference herein in their entirety).
  • standard hybridization techniques e.g., Sambrook et al., ibid., and Bothwell, Yancopoulos and Alt, ibid.
  • PCR polymerase chain reaction
  • genomic DNA encoding NeuroD may be obtained using probes designed from the sequences disclosed herein. Suitable probes for use in identifying neuroD sequences may be obtained from nei/r ⁇ D-specific sequences that are highly conserved regions between mammalian and amphibian neuroD coding sequences. Primers, for example, from the region encoding the approximately 40 residues following the helix-2 domain are suitable for use in designing PCR primers. Alternatively, oligonucleotides containing specific DNA sequences from a human neuroD coding region may be used within the described methods to identify human neuroD genomic and cDNA clones. Upstream regulatory regions of neuroD may be obtained using the same methods.
  • Suitable PCR primers are between 7-50 nucleotides in length, more preferably between 15 and 25 nucleotides in length.
  • neuroD polynucleotide molecules may be isolated using standard hybridization techniques with probes of at least about 7 nucleotides in length and up to and including the full coding sequence. Southern analysis of mouse genomic DNA probed with the murine neuroD cDNA under stringent conditions showed the presence of only one gene, suggesting that under stringent conditions bHLH genes from other protein families will not be identified. Other members of the neuroD family can be identified using degenerate oligonucleotides based on the sequences disclosed herein for PCR amplification or by hybridization at moderate stringency.
  • a DNA molecule encoding NeuroD is inserted into a suitable expression vector, which is in turn used to transfect or transform a suitable host cell.
  • Suitable expression vectors for use in carrying out the present invention comprise a promoter capable of directing the transcription of a polynucleotide molecule of interest in a host cell.
  • Representative expression vectors may include both plasmid and/or viral vector sequences.
  • Suitable vectors include retroviral vectors, vaccinia viral vectors, CMV viral vectors, BlueScript® vectors, baculovirus vectors, and the like.
  • Promoters capable of directing the transcription of a cloned gene or cDNA may be inducible or constitutive promoters and include viral and cellular promoters.
  • suitable viral promoters include the immediate early cytomegalovirus promoter (Boshart et al., Cell 41: 521-530, 1985) and the SV40 promoter (Subramani et al., Mol. Cell. Biol. 1: 854-864, 1981).
  • Suitable cellular promoters for expression of proteins in mammalian host cells include the mouse metallothionien-1 promoter (Palmiter et al., U.S. Patent No. 4,579,821), a mouse Vk promoter (Bergman et al., Proc. Natl. Acad Sci. 81: 7041-7045, 1983; Grant et al. Nucleic Acid Res.
  • telomere sequence of interest typically, is a transcription termination signal located downstream of the coding sequence of interest. Suitable transcription termination signals include the early or late polyadenylation signals from SV40 (Kaufman and Sharp, Mol. Cell. Biol. 2:1304-1319, 1982), the polyadenylation signal from the Adenovirus 5 elB region, and the human growth hormone gene terminator (DeNoto et al., Nucleic Acid Res.
  • Mammalian cells may be transfected by a number of methods including calcium phosphate precipitation (Wigler et al., Cell 14: 725, 1978; Corsaro and Pearson, Somatic Cell Genetics 7: 603, 1981; Graham and Van der Eb, Virology 52: 456, 1973); lipofection, microinjection, and electroporation (Neumann et al., EMBO J. 1: 8410845, 1982).
  • Mammalian can be transduced with virus such as SV40, CMV, and the like.
  • virus such as SV40, CMV, and the like.
  • viral vectors cloned DNA molecules may be introduced by infection of susceptible cells with viral particles.
  • Retroviral vectors may be preferred for use in expressing NeuroD in mammalian cells particularly if NeuroD is used for gene therapy (for review, see, Miller et al. Methods in Enzymology 217: 581-599, 1994; which is incorporated herein by reference in its entirety). It may be preferable to use a selectable marker to identify cells that contain the cloned DNA. Selectable markers are generally introduced into the cells along with the cloned DNA molecules and include genes that confer resistance to drugs, such as neomycin, hygromycin, and methotrexate. Selectable markers may also complement auxotrophies in the host cell.
  • selectable markers provide detectable signals, such as beta-galactosidase to identify cells containing the cloned DNA molecules.
  • Selectable markers may be amplifiable. Such ampiifiable selectable markers may be used to amplify the number of sequences integrated into the host genome.
  • the polynucleotide molecules of the present invention may be expressed Saccharomyces cerevisiae, filamentous fiingi, and E. coli.
  • Methods for expressing cloned genes in Saccharomyces cerevisiae are generally known in the art (see, "Gene Expression Technology,” Methods in Enzymology, Vol. 185, Goeddel (ed.), Academic Press, San Diego, CA 1990, and "Guide to Yeast Genetics and Molecular Biology,” Methods in Enzymology, Guthrie and Fink (eds.), Academic Press, San Diego, CA, 1991; which are incorporated herein by reference).
  • Filamentous fungi may also be used to express the proteins of the present invention; for example, strains of the fiingi Aspergillus (McKnight et al., U.S. Patent No. 4,935,349, which is incorporated herein by reference). Methods for expressing genes and cDNAs in cultured mammalian cells and in E. coli is discussed in detail in Sambrook et al. (Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor, NY, 1989; which is incorporated herein by reference). As would be evident to one skilled in the art, one could express the protein of the instant invention in other host cells such as avian, insect, and plant cells using regulatory sequences, vectors and methods well established in the literature.
  • the term "capable of hybridizing under stringent conditions" as used herein means that the subject nucleic acid molecules (whether DNA or RNA) anneal to an oligonucleotide of 15 or more contiguous nucleotides of SEQ ID NO:l, SEQ ID NO:3, SEQ ID NO:8, or SEQ ID NO: 10.
  • NeuroD proteins produced according to the present invention may be purified using a number of established methods such as affinity chromatography using anti- NeuroD antibodies coupled to a solid support. Fusion proteins of antigenic tag and NeuroD can be purified using antibodies to the tag. Additional purification may be achieved using conventional purification means such as liquid chromatography, gradient centrifugation, and gel electrophoresis, among others. Methods of protein purification are known in the art (see generally, Scopes, R., Protein Purification, Springer-Verlag, NY, 1982, which is incorporated herein by reference) and may be applied to the purification of recombinant NeuroD described herein.
  • hybridization conditions will be evident to one skilled in the art and will generally be guided by the purpose of the hybridization, the type of hybridization (DNA-DNA or DNA-RNA), and the level of desired relatedness between the sequences.
  • Methods for hybridization are well established in the literature; See, for example: Sambrook, ibid.; Hames and Higgins, eds, Nucleic Acid Hybridization, A Practical Approach, IRL Press, Washington DC, 1985; Berger and Kimmel, eds, Methods in Enzymology, Vol.
  • Hybridization stringency can be altered by: adjusting the temperature of hybridization; adjusting the percentage of hel -destabilizing agents, such as formamide, in the hybridization mix; and adjusting the temperature and salt concentration of the wash solutions.
  • the stringency of hybridization is adjusted during the post-hybridization washes by varying the salt concentration and/or the temperature.
  • Stringency of hybridization may be reduced by reducing the percentage of formamide in the hybridization solution or by decreasing the temperature of the wash solution.
  • High stringency conditions may involve high temperature hybridization (e.g., 65-68°C in aqueous solution containing 4-6 X SSC, or 42°C in 50% formamide) combined with high temperature (e.g., 5-25°C below the T JU ) and a low salt concentration (e.g., 0.1 X SSC).
  • Reduced stringency conditions may involve lower hybridization temperatures (e.g., 35-42°C in 20-50% formamide) with intermediate temperature (e.g., 40-60°C) and washes in a higher salt concentration (e.g., 2-6 X SSC).
  • Moderate stringency conditions which may involve hybridization at a temperature between 50°C and 55°C and washes in 0.1 X SSC, 0.1% SDS at between 50°C and 55°C, may be used to identify clones encoding members of the NeuroD family.
  • the invention provides isolated and purified polynucleotide molecules encoding NeuroD capable of hybridizing under stringent conditions an oligonucleotide of 15 or more contiguous nucleotides of SEQ ID NO:l, SEQ ID NO:3, SEQ ID NO:8, SEQ ID NO: 10, and their complementary strands.
  • the subject isolated neuroD polynucleotide molecules preferably encode NeuroD proteins that trigger differentiation in ectodermal cells, particularly neuroectodermal stem cells, and in more committed cells of that lineage, for example, epidermal precursor cells.
  • Such neuroD expression products typically form heterodimeric bHLH protein complexes that bind in the 5'-regulatory regions of target genes and enhance or suppress transcription of the target gene.
  • cancer cells may contain non-functional NeuroD protein or may contain no NeuroD protein due to genetic mutation or somatic mutations such that these cells fail to differentiate.
  • the cancer cells may be treated in a manner to cause the over-expression of wild-type NeuroD protein to force differentiation of the cancer cells.
  • Antisense neuroD nucleotide sequences may be used to block expression of mutant neuroD expression in neuronal precursor cells to generate and harvest neuronal stem cells.
  • the use of antisense oligonucleotides and their applications have been reviewed in the literature (see, for example, Mol and Van der Krul, eds., Antisense Nucleic Acids and Proteins Fundamentals and Applications, New York, NY, 1992; which is incorporated by reference herein in its entirety).
  • Suitable antisense oligonucleotides are at least 11 nucleotide in length and may include untranslated (upstream or intron) and associated coding sequences.
  • the optimal length of antisense oligonucleotide is its on the strength of the interaction between the antisense oligonucleotide and its complement on the mRNA, the temperature and ionic environment translation takes place, the base sequence of the antisense oligonucleotide, and the presence of secondary and tertiary structure in the mRNA and/or in the antisense oligonucleotide.
  • Suitable target sequences for antisense oligonucleotides include intron-exon junctions (to prevent proper splicing), regions in which DNA RNA hybrids will prevent transport of mRNA from the nucleus to the cytoplasm, initiation factor binding sites, ribosome binding sites, and sites that interfere with ribosome progression.
  • a particularly preferred target region for antisense oligonucleotide is the 5' untranslated (promoter/enhancer) region of the gene of interest.
  • Antisense oligonucleotides may be prepared by the insertion of a DNA molecule containing the target DNA sequence into a suitable expression vector such that the DNA molecule is inserted downstream of a promoter in a reverse orientation as compared to the gene itself.
  • antisense oligonucleotides may be synthesized using standard manual or automated synthesis techniques. Synthesized oligonucleotides may be introduced into suitable cells by a variety of means including electroporation, calcium phosphate precipitation, or microinjection. The selection of a suitable antisense oligonucleotide administration method will be evident to one skilled in the art. With respect to synthesized oligonucleotides, the stability of antisense oligonucleotide-mRNA hybrids may be increased by the addition of stabilizing agents to the oligonucleotide.
  • Stabilizing agents include intercalating agents that are covalently attached to either or both ends of the oligonucleotide.
  • Oligonucleotides may be made resistant to nucleases by, for example, modifications to the phosphodiester backbone by the introduction of phosphotriesters, phosphonates, phosphorothioates, phosphoroselenoates, phosphoramidates, or phosphorodithioates. Oligonucleotides may also be made nuclease resistant by the synthesis of the oligonucleotides with alpha-anomers of the deoxyribonucleotides.
  • NeuroD binds to 5 1 regulatory regions of neurogenic genes that are involved in neuroectodermal differentiation, including development of neural and endocrine tissues.
  • the NeuroD protein alters expression of the subject gene by, for example, down-regulating or up-regulating transcription, or by inducing a change in transcription to an alternative open reading frame.
  • the subject polynucleotide molecules find a variety of uses, e.g., in preparing oligonucleotide probes, expression vectors, and transformed host cells, as disclosed below in the following Examples.
  • DNA sequences recognized by NeuroD may be determined using a number of methods known in the literature including immunoprecipitation (Biedenkapp et al, Nature 335: 835-837, 1988, Kinzler and Vorgelstein, Nuc. Acids Res. 17: 3645-3653, 1989; and Sompayrac and Danna, Proc. Natl. Acad. Sci. USA 87: 3274-3278, 1990; which are incorporated by reference herein), protein affinity columns (Oliphant et al., Mol. Cell. Biol.
  • hybrid NeuroD proteins may contain the replacement of one or more contiguous amino acids of the native NeuroD with the analogous amino acid(s) of NeuroD from another species.
  • Such interspecies hybrid proteins include hybrids having whole or partial domain replacements.
  • hybrid proteins may be obtained using recombinant DNA techniques. Briefly, DNA molecules encoding the hybrid NeuroD proteins of interest are prepared using generally available methods such as PCR mutagenesis, site-directed mutagenesis, and/or restriction digestion and ligation. The hybrid DNA is then inserted into expression vectors and transformed or transfected into suitable host cells. The biological activity may be assessed essentially as described in the assays set forth in more detail in the Examples that follow.
  • the invention also provides synthetic peptides, recombinantly derived peptides, fusion proteins, and the like.
  • the subject peptides have an amino acid sequence encoded by a nucleic acid which hybridizes under stringent conditions with an oligonucleotide of 15 or more contiguous nucleotides of SEQ ID NO:l, SEQ ID NO:3, SEQ ID NO:8, or SEQ ID NO: 10.
  • Representative amino acid sequences of the subject peptides are disclosed in SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:9, and SEQ ID NO: 11.
  • the subject peptides find a variety of uses, including preparation of specific antibodies. As noted above, the invention provides antibodies which bind to NeuroD.
  • non-human antisera or monoclonal antibodies e.g., murine, lagormorpha, porcine, equine
  • antibody producing cells are obtained from immunized animals, immortalized and screened, or screened first for the production of the antibody that binds to the NeuroD protein or peptides and then immortalized. It may be desirable to transfer the antigen binding regions (i.e., F(ab')2 or hypervariable regions) of non-human antibodies into the framework of a human antibody by recombinant DNA techniques to produce a substantially human molecule.
  • a human monoclonal antibody or portions thereof may be identified by first screening a human B-cell cDNA library for DNA molecules that encode antibodies that specifically bind to NeuroD according to the method generally set forth by Huse et al. (Science 246: 1275-1281, 1989, which is incorporated by reference herein in its entirety). The DNA molecule may then be cloned and amplified to obtain sequences that encode the antibody (or binding domain) of the desired specificity.
  • the invention also provides methods for inducing the expression of genes associated with neuronal phenotype in a cell that does not normally express those genes.
  • Examples of neuronal phenotypes that may be modulated by NeuroD expression include expression of neurotransmitters or neuromodulatory factors.
  • Cells that can be used for the purpose of modulation of gene expression by NeuroD include cells of the neuroectodermal lineage, glial cells, neural crest cells, and epidermal epithelial basal stem cells, and all types of both mesodermal and endodermal lineage cells.
  • NeuroD protein in stem cells causes redirection of epidermal cell differentiation and induces terminal differentiation into neurons, i.e., instead of epidermal cells.
  • Epithelial basal stem cells i.e., in skin and mucosal tissues are one of the few continuously regenerating cell types in an adult mammal.
  • Introduction of the subject nucleotide sequences into an epithelial basal stem cell may be accomplished in vitro or in vivo using a suitable gene therapy vector delivery system (e.g., a retroviral vector), a microinjection technique (see, for example, Tarn, Basic Life Sciences 37: 187-194, 1986, which is incorporated by reference herein in its entirety), or a transfection method (e.g., naked or liposome encapsulated DNA or RNA) (see, for example, Trends in Genetics 5: 138, 1989; Chen and Okayama, Biotechniques 6: 632-638, 1988; Mannino and Gould-Fogerite, Biotechniques 6: 682-690, 1988; Kojima et al., Biochem.
  • a suitable gene therapy vector delivery system e.g., a retroviral vector
  • a microinjection technique see, for example, Tarn, Basic Life Sciences 37: 187-194, 1986, which is incorporated by reference herein in its entirety
  • the introduction method may be chosen to achieve a transient expression of NeuroD in the host cell, or it may be preferable to achieve constitutive or regulated expression in a tissue specific manner.
  • Transformed host cells of the present invention find a variety of in vitro uses; for example: i) as convenient sources of neuronal growth factors, ii) in transient and continuous cultures for screening anti-cancer drugs capable of driving terminal differentiation in neural tumors, and iii) as sources of recombinantly expressed NeuroD protein for use as an antigen in preparing monoclonal and polyclonal antibodies useful in diagnostic assays.
  • Transformed host cells of the present invention also find a variety of in vivo uses, for example, for transplantation at sites of traumatic neural injury where motor or sensory neural activity has been lost.
  • Representative patient populations that may benefit from transplantation include: patients with hearing or vision loss due to optical or auditory nerve damage, patients with peripheral nerve damage and loss or motor or sensory neural activity, and patients with brain or spinal cord damage from traumatic injury.
  • donor cells from a patient such as epithelial basal stem cells are cultured in vitro and then transformed or transduced with a neuroD nucleotide sequence. The transformed cells are then returned to the patient by microinjection at the site of neural dysfunction.
  • nucleotide sequences of the invention include the following:
  • recombinant cell lines, ova, and transgenic embryos and animals including dominant-negative and "knock-out" recombinant cell lines in which the transcription regulatory activity of NeuroD protein is down-regulated or eUminated.
  • Such cells may contain altered neuroD coding sequences that result in the expression of a NeuroD protein that is not capable of enhancing, suppressing or activating transcription of the target gene.
  • the subject cell lines and animals find uses in screening for candidate therapeutic agents capable of either substituting for a function performed by NeuroD or correcting the cellular defect caused by a defective NeuroD.
  • neuroD polynucleotide molecules may be joined to reporter genes, such as ⁇ -galactosidase or luciferase, and inserted into the genome of a suitable embryonic host cell such as an mouse embryonic stem cell by, for example, homologous recombination (for review, see Capecchi, Trends in Genetics 5: 70-76, 1989; which is incorporated by reference).
  • reporter genes such as ⁇ -galactosidase or luciferase
  • NeuroD may then be obtained by subjecting the differentiating embryonic cells to cell sorting, leading to the purification of a population of neuroblasts.
  • Neuroblasts may be useful for studying neuroblast sensitivity to growth factors or chemotherapeutic agents.
  • the neuroblasts may also be used as a source from which to purify specific protein products or gene transcripts. These products may be used for the isolation of growth factors, or for the identification of cell surface markers that can be used to purify stem cell population from a donor for transplantation.
  • gene transfer vectors e.g., retroviral vectors, and the like
  • neuroD gene therapy may be used to correct traumatic neural injury that has resulted in loss of motor or sensory neural function.
  • gene transfer vectors may either be injected directly at the site of the traumatic injury, or the vectors may be used to construct transformed host cells that are then injected at the site of the traumatic injury.
  • the results disclosed in Example 10 indicate that introduction of neuroD induces a non-neuronal cell to become a neuron. This discovery raises for the first time the possibility of using transplantation and/or gene therapy to repair neural defects resulting from traumatic injury.
  • neuroD provides the possibility of providing specific gene therapy for the treatment of certain neurological disorders such as Alzheimer's disease, Huntington's disease, and Parkinson's disease, in which a population of neurons have been damaged.
  • Two basic methods of neuroD utilization can be envisioned in this regard.
  • neuroD is expressed in existing populations of neurons to modulate aspects of their neuronal phenotype (e.g., neurotransmitter expression or synapse targeting) to make the neurons express a factor or phenotype to overcome the deficiency that contributes to the disease.
  • recombinant neuroD sequences are introduced into existing neurons or endogenous neuroD expression is induced.
  • neuroD is expressed in non-neuronal cells (e.g., glial cells in the brain or another non-neuronal cell type such as basal epithelial cells) to induce expression of genes that confer a complete or partial neuronal phenotype that ameliorates aspects of the disease.
  • Parkinson's disease is caused, at least in part, by the death of neurons that supply the neurotransmitter dopamine to the basal ganglia. Increasing the levels of neurotransmitter ameliorates the symptoms of Parkinson's disease.
  • Expression of neuroD in basal ganglia neurons or glial cells may induce aspects of a neuronal phenotype such that the neurotransmitter dopamine is produced directly in these cells. It may also be possible to express neuroD in donor cells for transplantation into the affected region, either as syngeneic or allogeneic transplantations.
  • the isolated polynucleotide molecules encoding NeuroD of the present invention permit the establishment of primary (or continuous) cultures of proliferating embryonic neuronal stem cells under conditions mimicking those that are active in development and cancer.
  • the resultant cell lines find uses: i) as sources of novel neural growth factors, ii) in screening assays for anti-cancer compounds, and ⁇ i) in assays for identifying novel neuronal growth factors.
  • High level expression of neuroD in the embryonic optic tectum indicates that NeuroD protein may regulate expression of factors trophic for growing retinal cells.
  • Such cells may be useful sources of growth factors, and may be useful in screening assays for candidate therapeutic compounds.
  • the cell lines and transcription regulatory factors disclosed herein offer the unique advantage that since they are active very early in embryonic differentiation they represent potential switches, e.g., ON ⁇ OFF or OFF-»ON, controlling subsequent cell fate. If the switch can be shown to be reversible (i.e., ON ⁇ - OFF), the NeuroD transcription regulatory factor and neuroD nucleic acids disclosed herein provide exciting opportunities for restoring lost neural and/or endocrine functions in a subject.
  • the following examples are offered by way of illustration and not by way of limitation.
  • EXAMPLE 1 Construction of the embryonic stem cell "179" cDNA library.
  • a continuous murine embryonic stem cell line i.e., the ES cell line
  • E2A the putative binding partner of myoD
  • Recombinant ES stem cells were constructed (i.e., using homologous recombination) wherein both alleles of the putative myoD binding partner E2A were replaced with drug-selectable marker genes.
  • ES cells do not make functional E12 or E47 proteins, both of which are E2A gene products.
  • ES cells form subcutaneous tumors in congenic mice (i.e., 129J) that appear to contain representatives of many different embryonal cell types as judged histologically and through the use of RT-PCR gene expression assays.
  • 129J congenic mice
  • Individual embryonic stem cell tumors were induced in male 129J strain mice by subcutaneous injection of 1 x 10 ⁇ cells/site. Three weeks later each tumor was harvested and used to prepare an individual sample of RNAs. Following random priming and second strand synthesis the ds-cDNAs were selected based on their size on 0.7% agarose gels and those cDNAs in the range of 400-800 bp were ligated to either Bam HI or Bgl ⁇ linkers.
  • Linkers were used to mixiimize the possibility that an internal Bam HI site in a cDNA might inadvertently be cut during cloning, leading to an abnormally sized or out-of-frame expression product.
  • the resultant individual stem cell tumor DNAs were individually ligated into the Bam HI cloning site in the "fl-VP16" 2 ⁇ yeast expression vector.
  • This expression vector, fl-VP16 contains the VP16 activation domain of Herpes simplex virus (HSV) located between Hind HI (HTTP) and Eco RI (RI) sites and under the control of the Saccharomyces cereviseae alcohol dehydrogenase promoter; with LEU2 and Ampicillin-resistance selectable markers.
  • HSV Herpes simplex virus
  • the resultant cDNA library was termed the "179-library”.
  • a two-hybrid yeast screening assay was used essentially as described by Fields and Song (Nature 340:245, 1989) and modified as described herein was used to screen the 179-library described in Example 1.
  • Yeast two-hybrid screens are reviewed as disclosed in Fields and Sternglanz (Trends in Genetics 10: 286-292, 1994).
  • the library was screened for cDNAs that interacted with LexA-Da, a fusion protein between the Drosophila Da (Daughterless) bHLH domain and the prokaryotic Z,t?xA-DNA binding domain. Multimerized LexA binding sites were cloned upstream of two reporter genes, the HIS3 gene and the ⁇ -galactosidase gene. The S.
  • neuroD cereviseae strain L40 containing a plasmid encoding a LexA- Lamin fusion. Clones that scored positive in the interaction with lamin were discarded. Approximately 400 cDNA clones, which represented 60 different transcripts, were identified as positive in these assays. Twenty-five percent of the original clones were subsequently shown to be known bHLH genes on the basis of their reactivity with specific cDNA probes. One cDNA clone encoding a VP16- fusion protein that interacted with Da but not lamin was identified as unique by sequence analysis. This clone, initially termed tango, is now referred to as neuroD.
  • the neuroD cDNA insert was subcloned as a Bam HI-Not I insert into Bam HI-Not I linearized pBluescript SK + . The resulting plasmid was designated pSK+ 1-83.
  • the neuroD insert contained in the VPl 6-neuroD plasmid was used to re- probe a mouse cDNA library prepared from mouse embryos at developmental stage el ⁇ .5.
  • Candidate clones were isolated and sequenced essentially as described above. Several clones were isolated.
  • Plasmid pKS m7a RX contains 1646 bp of murine neuroD cDNA as an EcoRI-XhoI insert.
  • the amino acid sequence encoded by the insert begins at amino acid residue +73 and extends to the carboxy-terminus of the NeuroD protein.
  • the plasmid contains about 855 bp of NeuroD coding sequence, (encoding amino acids 73-536). None of the mouse cDNAs contained the complete 5 1 coding sequence.
  • a mouse strain 129/Sv genomic DNA library was screened with the YP ⁇ -neuroD plasmid insert (450 bp).
  • Genomic clones were isolated and sequenced and the sequences were aligned with the cDNA sequences. Alignment of the sequence and comparison of the genomic 5 1 coding sequences with the Xenopus neuroD clone (Example 8) confirmed the 5' neuroD coding sequence.
  • the complete neuroD coding sequence and deduced amino acid sequence are shown in SEQ ID NOS: l and 2.
  • EXAMPLE 3 NeuroD/wewro bHLH proteins share common structural similarities that include a basic region that binds DNA and an HLH region involved in protein-protein interactions required for the formation of homodimers and heterodimeric complexes.
  • a comparison of the amino acid sequence of the basic region of murine NeuroD (amino acids 102 to 113 of SEQ ID NO:2) with basic regions of other bHLH proteins revealed that murine NeuroD contained all of the conserved residues characteristic among this family of proteins. However, in addition, NeuroD contained several unique residues. These unique amino acid residues were not found in any other known HLH, making NeuroD a distinctive new member of the bHLH family.
  • the NARERNR basic region motif in NeuroD is also found in the Drosophila AS- C protein, a protein thought to be involved in neurogenesis. Similar, but not identical, NARERRR and NERERNR motifs (SEQ ID NOS:5 and 6, respectively) have been found in the Drosophila Atonal and MASH (mammalian achaete-scute homolog) proteins, respectively, which are also thought to be involved in neurogenesis.
  • the NARER motif (SEQ ID NO:7) of neuroD is shared by other bHLH proteins, and the Drosophila Daughterless (Da) and Mammalian E proteins.
  • bHLH proteins The basic region of bHLH proteins is important for DNA binding site recognition, and there is homology between NeuroD and other neuro-proteins in this functional region. Within the important dimer-determining HLH region of NeuroD, a low level of homology was recorded with mouse twist protein (i.e., 51% homology) and with MASH (i.e., 46% homology). NeuroD contains several regions of unique peptide sequence within the bHLH domain including the junction sequence (MHG). EXAMPLE 4 NeuroD is expressed in differentiating neurons during embryonic development. NeuroD expression was analyzed during embryonic development of mouse embryos using in situ hybridization with an antisense neuroD single-stranded riboprobe labeled with digoxigenin (Boehringer Mannheim).
  • a riboprobe was prepared from plasmid pSK+1-83 using T7 polymerase and digoxigenin- 11-UTP for labeling.
  • the hybridized probe was detected using anti-digoxigenin antibody conjugated with alkaline phosphatase.
  • Color development was carried out according to the manufacturer's instruction. Stages of development are commonly expressed as days following copulation and where formation of the vaginal plug is e ⁇ .5.
  • the results recorded in the in situ hybridization studies were as follows:
  • neuroD expression was observed in the developing trigerminal ganglia.
  • a distinctive pattern of neuroD expression was observed in all the cranial ganglia (i.e., V-XT) and in dorsal root ganglia (DRG) in the trunk region of the embryo.
  • neuroD expression was also observed in the central nervous system in post-mitotic cells in the brain and spinal cord that were undergoing neuronal differentiation.
  • the ventral portion of the cord from which the motor neurons arise and differentiate was observed to express neuroD at high levels; and expression in the posterior-ventral spinal cord was higher when compared to more mature anterior-ventral spinal cord.
  • neuroD In the el4.5 mouse embryo, expression of neuroD was observed in cranial ganglia and DRG, but expression of neuroD persisted in the neuronal regions of developing sensory organs and the central nervous system (CNS). Thus, neuroD expression was observed to be transient during neuronal development.
  • elO neurula stage of the embryo
  • elO the neurogenic derivatives of neural crest cells
  • the cranial and dorsal root ganglia the cranial and dorsal root ganglia
  • post mitotic cells in the CNS suggests an important possible link between expression and generation of sensory and motor nerves.
  • neuroD expression is transient, the results suggest that neuroD expression is operative as a switch controlling formation of sensory nervous tissue. It is noteworthy that in these studies neuroD expression was not observed in embryonic sympathetic and enteric ganglia (also derived from migrating neural crest cells). Overall, the results indicate that neuroD plays an important role in neuronal differentiation.
  • NeuroD is expressed in neural and brain tumor cells: murine probes identify human neuroD.
  • Example 4 Given the expression pattern in mouse embryo (Example 4), Northern blots of tumor cell line mRNAs were examined using murine neuroD cDNA (Example 2) as a molecular probe. As a first step, cell lines that have the potential for developing into neurons were screened. The D283 human medullablastoma cell line, which expressed many neuronal markers, expressed high levels of neuroD by Northern blot analysis. NeuroD was also transcribed at various levels by different human neuroblastoma cell lines and in certain rhabdomyosarcoma lines that are capable of converting to neurons.
  • Murine PC 12 pheochromacytoma cells and P19 embryocarcinoma cells differentiate into neurons in tissue culture in the presence of appropriate inducers, i.e., nerve growth factor and retinoic acid, respectively.
  • appropriate inducers i.e., nerve growth factor and retinoic acid
  • murine P19 but not PC12 cells expressed neuroD transcripts.
  • non-induced murine PC12 cells, P19 cells, and control 3T3 fibroblasts did not produce detectable levels of neuroD transcripts.
  • PC12 and P19 cells represent cell types that are potentially useful in screening assays for identifying inducers of neuroD expression that may stimulate nerve regeneration and differentiation of neural tumor cells.
  • Recombinant murine 3T3 fibroblast cells expressing either a myc-tagged murine NeuroD protein or myc-tagged Xenopus NeuroD protein were made. The recombinant cells were used as a test system for identifying antibody to NeuroD described below.
  • Xenopus NeuroD protein was tagged with the antigenic marker Myc to allow the determination of the specificity of anti-NeuroD antibodies to be determined.
  • Plasmid CS2+MT was used to produce the Myc fusion protein.
  • the CS2+MT vector contains the simian cytomegalovirus IE94 enhance/promoter (and an SP6 promoter in the 5' untranslated region of the IE94- driven transcript to allow in vitro RNA synthesis) operatively linked to a DNA sequence encoding six copies of the Myc epitope tag (Roth et al, J. Cell Biol. 115: 587-596, 1991; which is incorporated herein in its entirety), a polylinker for insertion of coding sequences, and an SV40 late polyadenylation site.
  • CS2-MT was digested with Xho I to linearize the plasmid at the polylinker site downstream of the DNA sequence encoding the myc tag.
  • the linearized plasmid was blunt-ended using Klenow and dNTPs.
  • a full length Xenopus cDNA clone was digested with Xho I and Eae I and blunt-ended using Klenow and dNTPs, and the 1.245 kb fragment of the Xenopus neuroD cDNA was isolated.
  • the neuroD fragment and the linearized vector were ligated to form plasmid CS2+MT xl-83.
  • CS2+MT was digested with Eco RI to linearize the plasmid at the polylinker site downstream of the DNA sequence encoding the myc tag.
  • the linearized plasmid was blunt-ended using Klenow and dNTPs and digested with Xho I to obtain a linearized plasmid having an Xho I adhesive end and a blunt end.
  • Plasmid pKS+m7a containing a partial murine NeuroD cDNA was digested with Xho I, and the NeuroD containing fragment was blunt-ended and digested with Xba I to obtain the approximately 1.6 kb fragment of the murine neuroD cDNA.
  • the neuroD fragment and the linearized vector were ligated to form plasmid CS2+MT Ml-83(m7a).
  • Plasmids CS2+MT xl-83 and CS2+MT Ml-83(m7a) were each transformed into murine 3T3 fibroblast cells and used as a test system for identifying antibody against NeuroD (Example 7).
  • a recombinant fusion protein of maltose binding protein (MBP) and amino acid residues 70-355 of murine NeuroD was used as an antigen to evoke antibodies in rabbits. Specificity of the resultant antisera was confirmed by ⁇ riinunostaming of the recombinant 3T3 cells described above. Double-i-mmunostaimng of the recombinant cells was observed with monoclonal antibodies to Myc (i.e., the control antigenic tag on the transfected DNA) and with rabbit anti-murine NeuroD in combination with anti-rabbit IgG.
  • MBP maltose binding protein
  • NeuroD is a highly evolutionarily conserved protein: sequence of Xenopus NeuroD. Approximately one million clones from a stage 17 Xenopus head library made by Kintner and Melton (Development 99: 311, 1987) were screened with the mouse cDNA insert as a probe at low stringency. The hybridization was performed with 50% formamide/4 X SSC at 33°C and washed with 2 X SSC/0.1% SDS at 40°C.
  • the domain structures of murine and Xenopus NeuroD are highly homologous with an "acidic" N-terminal domain (i.e., glutamic or aspartic acid rich); a basic region; helix 1, loop, helix 2; and a proline rich C-terminal region.
  • Xenopus NeuroD differ in amino acid sequence, both retain a glutamic or aspartic acid rich "acidic domain" (amino acids 102 to 113 of SEQ ID NO:2 and amino acids 56 to
  • activation domain for the NeuroD protein, in a manner analogous to the activation mechanisms currently understood for other known transcription regulatory factors.
  • EXAMPLE 9 Neuronal expression of Xenopus neuroD.
  • the expression pattern of neuroD in whole mount Xenopus embryos was determined using in situ hybridization with a single stranded digoxigenin-labeled Xenopus neuroD antisense cDNA riboprobe. Embryos were examined at several different stages.
  • cranial ganglia Consistent with the mouse expression pattern, by late stage, all cranial ganglia showed very strong staining patterns.
  • neural crest cells give rise to skeletal components of the head, all ganglia of the peripheral nervous system, and pigment cells.
  • the cranial sensory ganglia which are of mixed crest and placode origin, represent the only group of cells that express neuroD.
  • High levels of neuroD expression in the eye were also observed, correlating with active neuronal differentiation in the retina at this stage. Expression is observed in the developing olfactory placodes and otic vesicles, as was seen in mice.
  • the pineal gland also expressed neuroD. All of this expression in transient, suggesting that neuroD functions during the differentiation process but is not required for maintenance of these differentiated cell types.
  • stage 14 i.e., the mid-neurula stage
  • stage 14 i.e., the mid-neurula stage
  • Primary mechanosensory neurons in the spinal cord also referred to as Rohon-Beard cells and primary motor neurons, showed neuroD expression at this stage.
  • stage 24 all of the developing cranial ganglia, trigerminal, facio-acoustic, glosso-pharyngeal, and vagal nervous tissues showed a high level of neuroD expression. High levels of expression of neuroD was also observed in the eye at this stage. (Note that in Xenopus neuronal differentiation in the retina occurs at a much earlier stage than in mice, and neuroD expression was correspondingly earlier and stronger in this animal model.)
  • EXAMPLE 10 Ectopic expression of neuroD converts non-neuronal cells into neurons.
  • myc-tagged neuroD transcripts were synthesized in vitro using SP6 RNA polymerase.
  • the myc tagged-neuroD transcripts were microinjected into one of the two cells in & Xenopus 2-cell embryo, and the other cell of the embryo served as an internal control.
  • Antibodies to Xenopus N-CAM, a neural adhesion molecule, anti-Myc (to detect the exogenous protein), and ⁇ nmunostaining techniques were used to evaluate phenotypic expression of the neuronal marker (and control) gene during the subsequent developmental stages of the microinjected embryos.
  • an evaluation of over 130 embryos that were injected with neuroD RNA showed a striking increase in ectopic expression of N- CAM on the microinjected side of the embryo (i.e., Myc 4 ), as judged by increased immunostaining. The increased staining was observed in the region from which neural crest cells normally migrate.
  • ectopic expression or over- expression
  • neuroD caused neural crest stem cells to follow a neurogenic cell fate.
  • the ectopic immunostaining was observed in the facio-cranial region and epidermal layer, and in some cases the stained cells were in the ventral region of the embryo far from the neural tube.
  • the immunostained cells not only expressed N-CAM ectopically, but displayed a morphological phenotype of neuronal cells. At high magnification, the N-CAM expressing cells exhibited typical neuronal processes reminiscent of axonal processes.
  • neuroD RNA was injected into the top tier of 32-cell stage embryos, in order to target the injection into cells destined to become epidermis.
  • N-CAM staining was observed in the lateral and ventral epidermis without any noticeable effect on the endogenous nervous system, indicating that the staining of N-CAM in the epidermis represents the conversion of epidermal cell fate into neuronal cell fate.
  • the embryos were subjected to jinmunochemistry as described by Turner and Weintraub (Genes Dev. 8: 1434, 1994, which is incorporated by reference herein) using primary antibodies detected with alkaline phosphatase-conjugated goat anti-mouse or anti-rabbit antibodies diluted to 1:2000 (Boehringer-Mannheim).
  • Anti-acetylated alpha-tubulin was diluted 1:2000.
  • Anti-Xen-1 was diluted 1:1.
  • Anti-NF-M was diluted 1:2000.
  • Embryos stained for NF-M were fixed in Dent's fixative (20% dimethylsulfoxide/80% methanol) and cleared in 2:1 benzyl benzoate/benzyl alcohol as described by Dent et al.
  • the spinal cord on the microinjected side of the embryo failed to develop properly, and the tissues were strongly immunopositive when stained with anti-N-CAM.
  • many microinjected embryos exhibited an increase in cell mass in the cranial region of the embryo from which (in a normal embryo) the neural crest cells and their derivatives (i.e., cranial ganglionic cells) would migrate.
  • the observed cranial bulge exhibited strong immunostaining with antibodies specific for N-CAM.
  • NeuroD-injected embryos were also assayed for alteration in the expression of Xtwist, the Xenopus homolog of Drosophila twist, to determine whether neuroD converted non-neuronal components of neural crest cells into the neural lineage.
  • Xtwist is strongly expressed in the non-neuronal population cephalic neural crest cells that give rise to the connective tissue and skeleton of the head.
  • NewroD-injected embryos were completely missing Xtwist expression in the migrating cranial neural crest cells on the injected side.
  • the cranial region of the embryo is severely affected by ectopic neuroD, the injected side of the embryos displayed either small or no eyes in addition to poorly organized brains, otic vesicles, and olfactory organs.
  • the spinal cord showed retarded growth, remaining thinner and shorter on the wei roD-mjected side.
  • N-CAM staining in the normal embryo at early stages was not uniform throughout the entire neural plate, but rather was more prominent in the medial region of the neural plate.
  • Injected embryos analyzed for N-CAM expression show that the neural plate on the injected side of the early stage embryos was stained more intensely and more laterally.
  • the increase in N-CAM staining was not associated with any lateral expansion of the neural plate as assayed by visual inspection and staining with the epidermal marker EpA. This was in contrast to what has been observed with XASH-3 injection that causes neural plate expansion.
  • injected embryos were stained using two neuronal markers that are expressed in differentiated neurons, neural specific ⁇ -tubulin and tanabin.
  • In situ hybridization for ⁇ -tubulin and tanabin was carried out as described above.
  • Over- expression of neuroD dramatically increased the ⁇ -tubulin signals in the region of the neural plate containing both motor neurons and Rohon-Beard cells at stage 14.
  • the earliest ectopic ⁇ -tubulin positive cells on the injected side were observed at the end of gastrulation when the control side did not yet show any ⁇ -tubulin positive cells.
  • Tanabin was also expressed in more cells in the spinal cord in the neuroD injected side of the embryos at stage 14.
  • EXAMPLE 11 Human genomic NeuroD. Genomic clones encoding human NeuroD were obtained by probing a human fibroblast genomic library with the mouse neuroD cDNA Host E. coli strain LE392 (New England Biolabs) were grown in LB + 10 mM MgSO 4 0.2% maltose overnight at 37°C. The cells were harvested and resuspended in 10 mM MgSO to a final OD600 of 2. The resuspended cells were used as hosts for phage infection.
  • the optimal volume of phage stock for use in this screening was determined by using serial dilutions of the phage stock of a human fibroblast genomic library in lambda FLX II (Stratagene) to infect LE392 cells (New England Biolabs). To obtain approximately 50,000 plaques per plate, a 2.5 ⁇ l aliquot of the phage stock was used to infect 600 ⁇ l of the resuspended LE392 cells. The cells were incubated with the phage for 15 minutes at 37°C, after which the cells were mixed with 6.5 ml of top agar warmed to 50°C. The top agar was plated on solid LB, and incubated overnight at 37°C. A total of 22 15-cm plates were prepared in this manner.
  • Duplicate plaque lifts were prepared. A first set of Hybond membranes (Amersham) were placed onto the plates and allowed to sit for 2 minutes. The initial membranes were removed and the duplicate membranes were laid on the plates for 4 minutes. The membranes were allowed to air dry; then the phage were denatured in 0.5 M NaOH, 1.5 M NaCl for 7 minutes. The membranes were neutralized with two washes in neutralization buffer (1.5M NaCl, 0.5 M Tris, pH 7.2). After neutralization, the membranes were crosslinked by exposure to UV. A 1 kb Eco RI- Hind HI fragment containing murine neuroD coding sequences was random primed using the Random Priming Kit (Boehringer Mannheim) according to the manufacturer's instructions.
  • Membranes were prepared for hybridization by placing six membranes in 10 ml of FBI hybridization buffer [100 g polyethylene glycol 800, 350 ml 20% SDS, 75 ml 20X SSPE; add water to a final volume of one liter.] and incubating the membranes at 65°C for 10 minutes. After 10 minutes, denatured salmon sperm DNA was added to a final concentration of 10 ⁇ g/ml and denatured probe was added to a final concentration of 0.25-0.5 x 10 cpm/ml. The membranes were hybridized at 65°C for a period of 8 hours to overnight. After incubation, the excess probe was removed, and the membranes were washed first in 2 X SSC, 0.1% SDS for 30 minutes at 50°C.
  • FBI hybridization buffer 100 g polyethylene glycol 800, 350 ml 20% SDS, 75 ml 20X SSPE; add water to a final volume of one liter.
  • the first wash was followed by a final wash in 0.1 X SSC, 0.1% SDS for 30 minutes at 55°C. Autoradiographs of the membranes were prepared. The first screen identified 55 putative positive plaques. Thirty-one of the plaques were subjected to a secondary screen using the method essentially set forth above. Ten positive clones were identified and subjected to a tertiary screen as described above. Eight positive clones were identified after the tertiary screen.
  • Phage DNA was prepared from clones 14B1, 9F1, and 20A1.
  • the 14B1 and 20 Al phage DNA were digested with Pst I to isolate the 1.2 kb and 1.6 kb fragments, respectively, that hybridized to the mouse neuroD probe.
  • the 9F1 phage DNA was digested with Eco RI and Sad to obtain an approximately 2.2 kb fragment that hybridizes with the mouse neuroD probe.
  • the fragments were each subcloned into plasmid Bluescript SK (Stratagene) that had been linearized with the appropriate restriction enzyme(s).
  • the fragments were sequenced using Sequenase Version 2.0 from USB (US Biochemical) and the following primers: the universal primer M13-21, the T7 primer, and the T3 primer.
  • clone 9F1 has a high degree of homology throughout the sequence region that spans the translation start site to the end of the bHLH region.
  • the 9F1 clone has 100% identity to mouse NeuroD in the HLH region (i.e., residues 117-156 in SEQ ID O:9 and residues 117-156 in SEQ ID NO:2), and an overall identity of 94%.
  • the 14B1 clone also has 100% identity to the HLH region (i.e., residues 91- 130 in SEQ ID NO: 11 and residues 117-156 in SEQ ID NO:2), but only 40% identity to 9F1 and 39% identity to mouse NeuroD in the amino-terminal region.
  • 9F1 is the human homolog of mouse neuroD, whereas the strong conservation of the neuroD HLH identifies 14B1 as another member of the neuroD HLH subfamily.
  • EXAMPLE 12 Chromosome mapping of human neuroD clones. FISH karyotyping was performed on fixed metaphase spreads of the microcell hybrids essentially as described (Trask et al., Am. J. Hum. Genet. 48: 1-15, 1991; and Brandriff et al., Genomics 10: 75-82, 1991, which are incorporated by reference herein in their entirety). NeuroD sequences were detected using the 9F1 or 20A1 phage DNA as probes labeled using digoxigenin dUTP (Boehringer Mannheim) according to the manufacturer's instructions.
  • Phage DNA was biotinylated by random priming (Gibco/BRL BioNick Kit) and hybridized in situ to denatured metaphase chromosome spreads for 24-48 hours. Probes were detected with rhodamine-conjugated antibodies to digoxigenin and chromosomes were counterstained DAPI (Sigma). Signals were viewed through a fluorescence microscope and photographs were taken with color slide film. FISH analysis indicated clone 9F1 maps to human chromosome 2q, and clone 20 Al maps to human chromosome 5.
  • Chromosome mapping was also carried out on a human/rodent somatic cell hybrid panel (National Institute of General Medical Sciences; Camden NJ). This panel consists of DNA isolated from 24 human/rodent somatic cell hybrids retaining one human chromosomes. For one set of experiments, the panel of DNA's were digested with Eco RI and electrophoresed on an agarose gel. The DNA was transferred to Hybond-N membranes (Amersham). A random primed (Boehringer Mannheim) 4 kb Eco Rl-Sac I fragment of clone 9F1 was prepared. The filter was prehybridized in 10 ml of FBI hybridization buffer (see above) at 65 °C for 10 minutes.
  • denatured salmon sperm DNA was added to a final concentration of 10 ⁇ g/ml; denatured probe was added to a final concentration of one million cpm/ml.
  • the filter was hybridized at 65 °C for a period of 8 hours to overnight. After incubation, excess probe was removed, and the filter was washed first in 2 X SSC, 0.1% SDS for 30 minutes at 65°C. The first wash was followed by a final wash in 0.1 X SSC, 0.1% SDS for 30 minutes at 65°C. An autoradiograph of the filter was prepared. Autoradiographs confirmed the FISH mapping results.
  • the panel was digested with Pst I, electrophoresed and transferred essentially as described above.
  • a random-primed (Boehringer Mannheim) 1.6 kb Pst I fragment of clone 20A1 was prepared.
  • the membrane was prehybridized, hybridized with the 20A1 probe and washed as described above. Autoradiographs of the Southern showed that 20A1 mapped to human chromosome 5 and confirmed the FISH mapping results.
  • the 20Al-probed membrane was stripped by a wash in 0.5 M NaOH, 1.5 M NaCl.
  • the membrane was neutralized in 0.5 M Tris-HCl (pH 7.4), 1.5 M NaCl.
  • the filter was washed in 0.1 X SSC before prehybridization.
  • a random-primed (Boehringer Mannheim) 1.2 kb Pst I fragment of clone 14B1 was prepared.
  • the washed membrane was prehybridized and hybridized with the 14B1 probe as described above. After washing under the previously described conditions, the membrane was autoradiographed. Autoradiographs demonstrated that clone 14B1 mapped to chromosome 17.
  • a human neuroD cDNA To obtain a human neuroD cDNA, one million plaque forming units (pfu) were plated onto twenty LB + 10 mM MgSO (150 mm) plates using the bacterial strain XL-1 Blue (Stratagene). Plating and membrane lifts were performed using standard methods, as described in Example 11. After UV cross-linking, the membranes were pre-hybridized in an aqueous hybridization solution (1% bovine serum albumin, 1 mM EDTA, 0.5 M Na 2 HPO 4 (pH 7.4), 7% SDS) at 50°C for two hours.
  • an aqueous hybridization solution 1% bovine serum albumin, 1 mM EDTA, 0.5 M Na 2 HPO 4 (pH 7.4), 7% SDS
  • the neuroD cDNA insert was prepared by digesting the pKS+ m7a RX plasmid with Eco RI and Xho I, and isolating the fragment containing the cDNA by electroelution.
  • a probe was made with the cDNA containing fragment by random primed synthesis with random hexanucleotides, dGTP,dATP, dTTP, alpha- 3 P-labeled dCTP, and Klenow in a buffered solution (25 mM Tris (pH6.9), 50mM KC1, 5mM MgC- 2 , ImM DTT).
  • the probe was purified from the unincorporated nucleotides on a G-50 sepharose column. The purified probe was heat denatured at 90°C for 3 minutes.
  • the denatured probe was added to the membranes in hybridization solution.
  • the membranes were hybridized for 24 hours at 50°C. Excess probe was removed from the membranes, and the membranes were washed in 0.1 X SSC, 0.1% SDS for 20 minutes at 50°C. The wash solution was changed five times.
  • the membranes were blotted dry and covered with plastic film before being subjected to autoradiography. Autoradiography of the filters identified 68 positive clones. The clones are plaque-purified and rescreened to obtain pure, positive clones.
  • Leu Ser Pro Pro Leu Ser lie Asn Gly Asn Phe Ser Phe Lys His Glu 275 280 285
  • Leu Ser Lys lie Glu Thr Leu Arg Leu Ala Lys Asn Tyr lie Trp Ala 140 145 150
  • Leu Ser Glu lie Leu Arg Ser Gly Lys Ser Pro Asp Leu Val Ser Phe 155 160 165
  • GTA CAA ACT CTC TGC AAA GGT TTG TCG CAG CCC ACC ACC AAT CTA GTA
  • Gin Asp lie Gin Ser His Met Gin Thr Ala Ser Ser Ser Phe Pro Leu 205 210 215
  • GCCCCACTCT GGGGCTGTAA CAAACTTGAT CTTGTCCTGC CTTTAGATAT GGGGAAACCT 1230
  • MOLECULE TYPE DNA (genomic)
  • CAA GGT CCT CCA AGC TGG ACA GAC GAG TGT CTC AGT TCT CAG GAC GAG 152
  • MOLECULE TYPE DNA (genomic)
  • Ser Lys lie Glu Thr Leu Arg Leu Ala Lys Asn Tyr lie Trp Ala Leu 115 120 125
  • Lys lie Glu Thr Leu Arg Leu Ala Lys Asn Tyr lie Trp Ala Leu Ser 115 120 125

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Toxicology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)

Abstract

L'invention se rapporte à une molécule d'acide nucléique isolée qui comprend au moins 15 nucléotides et qui s'hybride dans des conditions rigoureuses avec un domaine hélice-boucle-hélice d'une protéine neuro D, résidant entre les nucléotides 577-696 de SEQ ID NO:1, les nucléotides 376-495 de SEQ ID NO:3, les nucléotides 405-524 de SEQ ID NO:8, ou les nucléotides 273-392 de SEQ ID NO:10, et à ses compléments.
PCT/US1995/005741 1994-05-06 1995-05-08 Proteines et genes de differentiation neurogene (neuro d) WO1995030693A1 (fr)

Priority Applications (5)

Application Number Priority Date Filing Date Title
EP95919759A EP0759938A4 (fr) 1994-05-06 1995-05-08 Proteines et genes de differentiation neurogene (neuro d)
JP7529169A JPH10503363A (ja) 1994-05-06 1995-05-08 神経分化(ニューロd)遺伝子及びタンパク質
AU25446/95A AU2544695A (en) 1994-05-06 1995-05-08 Neurogenic differentiation (neurod) genes and proteins
US08/910,973 US5795723A (en) 1994-05-06 1997-08-07 Expression of neurogenic bHLH genes in primitive neuroectodermal tumors
US09/499,227 US6444463B1 (en) 1994-05-06 2000-02-07 Neurogenic differentiation gene neurod3 and methods for inducing differentiation of cells

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US23922894A 1994-05-06 1994-05-06
US08/239,228 1994-05-06

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US23923894A Continuation-In-Part 1994-05-06 1994-05-06

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US08/552,142 Continuation-In-Part US5695995A (en) 1994-05-06 1995-11-02 Neurogenic differentiation (neurod) genes

Publications (1)

Publication Number Publication Date
WO1995030693A1 true WO1995030693A1 (fr) 1995-11-16

Family

ID=22901195

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1995/005741 WO1995030693A1 (fr) 1994-05-06 1995-05-08 Proteines et genes de differentiation neurogene (neuro d)

Country Status (5)

Country Link
EP (1) EP0759938A4 (fr)
JP (1) JPH10503363A (fr)
AU (1) AU2544695A (fr)
CA (1) CA2188450A1 (fr)
WO (1) WO1995030693A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998027206A2 (fr) * 1996-12-19 1998-06-25 Rhone-Poulenc Rorer S.A. Polypeptides de la famille 'basic helix-loop-helix' bhlh, sequences d'acides nucleiques correspondantes
EP0877802A1 (fr) * 1995-11-02 1998-11-18 Fred Hutchinson Cancer Research Center GENES ET PROTEINES OBTENUS PAR DIFFERENTIATION NEUROGENIQUE (NeuroD)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5322801A (en) * 1990-04-19 1994-06-21 The General Hospital Corporation Protein partner screening assays and uses thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5322801A (en) * 1990-04-19 1994-06-21 The General Hospital Corporation Protein partner screening assays and uses thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP0759938A4 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0877802A1 (fr) * 1995-11-02 1998-11-18 Fred Hutchinson Cancer Research Center GENES ET PROTEINES OBTENUS PAR DIFFERENTIATION NEUROGENIQUE (NeuroD)
EP0877802A4 (fr) * 1995-11-02 2001-12-05 Hutchinson Fred Cancer Res GENES ET PROTEINES OBTENUS PAR DIFFERENTIATION NEUROGENIQUE (NeuroD)
WO1998027206A2 (fr) * 1996-12-19 1998-06-25 Rhone-Poulenc Rorer S.A. Polypeptides de la famille 'basic helix-loop-helix' bhlh, sequences d'acides nucleiques correspondantes
FR2757524A1 (fr) * 1996-12-19 1998-06-26 Rhone Poulenc Rorer Sa Polypeptides de la famille bhlh, sequences d'acides nucleiques correspondantes
WO1998027206A3 (fr) * 1996-12-19 1998-10-01 Rhone Poulenc Rorer Sa Polypeptides de la famille 'basic helix-loop-helix' bhlh, sequences d'acides nucleiques correspondantes
US6998474B1 (en) 1996-12-19 2006-02-14 Centelion Sas Polypeptides of the “basic helix-loop-helix” bHLH family, corresponding nucleic acid sequences

Also Published As

Publication number Publication date
EP0759938A4 (fr) 1999-10-27
AU2544695A (en) 1995-11-29
EP0759938A1 (fr) 1997-03-05
CA2188450A1 (fr) 1995-11-16
JPH10503363A (ja) 1998-03-31

Similar Documents

Publication Publication Date Title
US5795723A (en) Expression of neurogenic bHLH genes in primitive neuroectodermal tumors
JP6722730B2 (ja) Nrg−2核酸分子、ポリペプチド、ならびに診断および治療法
Liu et al. Dlx genes encode DNA‐binding proteins that are expressed in an overlapping and sequential pattern during basal ganglia differentiation
Oishi et al. Spatio‐temporally regulated expression of receptor tyrosine kinases, mRor1, mRor2, during mouse development: implications in development and function of the nervous system
Xu et al. LH-2: a LIM/homeodomain gene expressed in developing lymphocytes and neural cells.
US5695995A (en) Neurogenic differentiation (neurod) genes
Chong et al. REST: a mammalian silencer protein that restricts sodium channel gene expression to neurons
Oulad‐Abdelghani et al. Meis2, a novel mouse Pbx‐related homeobox gene induced by retinoic acid during differentiation of P19 embryonal carcinoma cells
Benkhelifa et al. mafA, a novel member of the maf proto-oncogene family, displays developmental regulation and mitogenic capacity in avian neuroretina cells
Sasaki et al. The oncofetal gene Pem encodes a homeodomain and is regulated in primordial and pre-muscle stem cells
KR19990013797A (ko) 사람의 세마포린 L(H-SemaL) 및 다른 종의 상응하는 세마포린
Ohtoshi et al. Isolation and characterization of Vsx1, a novel mouse CVC paired-like homeobox gene expressed during embryogenesis and in the retina
US6162641A (en) Neuregulin response element and uses therefor
WO2002030976A1 (fr) Proteines et acides nucleiques de regulation cellulaire
US6444463B1 (en) Neurogenic differentiation gene neurod3 and methods for inducing differentiation of cells
WO1995030693A1 (fr) Proteines et genes de differentiation neurogene (neuro d)
WO1996031534A1 (fr) MOLECULES ISOLEES D'ACIDE NUCLEIQUE CODANT UN p57KIP2 ET LEURS UTILISATIONS
HU211976A9 (en) The ciliary neurotrophic factor receptor
CA2433501C (fr) Proteines bhlh, leurs genes et leurs utilisations
WO1998001755A1 (fr) Procede de criblage de proteines mena intervenant dans la dynamique des microfilaments
WO1998001755A9 (fr) Procede de criblage de proteines mena intervenant dans la dynamique des microfilaments
Kherrouche et al. Isolation and characterization of a chicken homologue of the Spi-1/PU. 1 transcription factor
Budhram-Mahadio-Head Regulation and Function of POU domain transcription factors, Brn-3a and Brn-3b
US6541251B1 (en) Pancreatic progenitor 1 gene and its uses
Carlock et al. Variable subcellular localization of a neuron-specific protein during NTera 2 differentiation into post-mitotic human neurons

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 08552142

Country of ref document: US

AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP US

AL Designated countries for regional patents

Kind code of ref document: A1

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

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

Ref document number: 2188450

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1995919759

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1995919759

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1995919759

Country of ref document: EP