WO1998055618A1 - Mitofusin genes and their uses - Google Patents

Mitofusin genes and their uses Download PDF

Info

Publication number
WO1998055618A1
WO1998055618A1 PCT/US1998/012064 US9812064W WO9855618A1 WO 1998055618 A1 WO1998055618 A1 WO 1998055618A1 US 9812064 W US9812064 W US 9812064W WO 9855618 A1 WO9855618 A1 WO 9855618A1
Authority
WO
WIPO (PCT)
Prior art keywords
fzo
mitofusin
protein
leu
ser
Prior art date
Application number
PCT/US1998/012064
Other languages
French (fr)
Other versions
WO1998055618A9 (en
Inventor
Margaret T. Fuller
Karen G. Hales
Original Assignee
The Board Of Trustees Of The Leland Stanford Junior University
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 The Board Of Trustees Of The Leland Stanford Junior University filed Critical The Board Of Trustees Of The Leland Stanford Junior University
Priority to AU80660/98A priority Critical patent/AU736643B2/en
Priority to EP98928991A priority patent/EP1012279A4/en
Priority to JP50317299A priority patent/JP2002503964A/en
Priority to CA002292840A priority patent/CA2292840A1/en
Publication of WO1998055618A1 publication Critical patent/WO1998055618A1/en
Publication of WO1998055618A9 publication Critical patent/WO1998055618A9/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y301/00Hydrolases acting on ester bonds (3.1)
    • C12Y301/05Triphosphoric monoester hydrolases (3.1.5)
    • C12Y301/05001Triphosphoric monoester hydrolases (3.1.5) dGTPase (3.1.5.1)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies

Definitions

  • Mitochondria are dynamic organelles that undergo regulated fusion in many cell types Analysis of serial sections from rodent skeletal muscle, lymphocytes, liver, spinal ganglion cells, and from the yeast Saccharomyces cerevisiae has shown that all the mitochondrial material of a cell can exist as a giant branched reticulum
  • HA influenza virus hemagglutinin
  • Mitofusin genes and proteins are provided as used herein, the term "mitofusin” indicates the Drosophila Fzo protein or any of its homologues from insects, other invertebrates, yeast, and vertebrates including mouse and humans
  • Motifusms are large predicted GTPases with a predicted trans-membrane domain, coiled-coil regions, and a C-terminal region showing a high pi characteristic of mitochondrial matrix proteins
  • the mitofusin Fzo is the first known protein mediator of mitochondrial fusion, and mediates developmentally regulated post-meiotic fusion of mitochondria in Drosophila spermatids
  • Figure 1 A is a genetic, cytological, and molecular map of the fzo region on chromosome 3R Stippled numbered boxes represent polytene chromosome bands, open bars are deficiencies, with + indicating complementation of fzo and - indicating failure to complement
  • the distal breakpoint of Df(3R)EB6 is at +2 kb on a genomic phage walk that extends 90 kb distal from hh
  • Figure 1 B is a molecular map of the fzo region of the genomic walk, with coordinates 0 to 20 indicated in kb Deficiencies represented as in (A) Restriction sites R, EcoRI, X, Xbal, Z, Xhol Thick line on molecular map represents genomic DNA used to screen testis cDNA library This region indicates a genomic fragment that has all sequences required to express fzo in vivo, and to restore fzo function in mutants
  • Figure 2A shows the predicted ammo acid sequence (single letter ammo acid code) of the fzo gene product, with regions matching GTPase motifs in boldface and numbered G1-G4 Underlined region is the predicted transmembrane domain Italics show the hydrophobic region including the predicted transmembrane domain and 13 adjacent uncharged residues Asterisks indicate predicted coiled coil regions with scores above 0 4 Letters above ammo acid sequence indicate introduced missense mutations
  • Figures 2B and 2C show alignments of predicted G1 and G2 motifs (B) and G3 and G4 motifs (C) in Fzo and homologs Boxes indicate identities with Fzo
  • Figure 2D is a schematic diagram of Fzo and human, C elegans, and S cerevisiae homologs, showing predicted GTPase domain (stippled box) and predicted transmembrane domain (dark bar) Spirals indicate predicted coiled coil regions with probabilities between 0 4 and 1 0 (large spirals) and between 0 2 and 0 4 (small spirals), respectively Percentage identities to Fzo overall and within the predicted GTPase domains are indicated
  • Figure 2E shows overlaid plots of coiled coil probabilities (open) and hydrophobicity (filled- in) for Fzo and homologs Sequences were analyzed with the COILS program (MDITK matrix, weighted second and fifth heptad positions) and with the TMpred program using default parameters, a TMpred output of 0 5x10 3 or above is considered significant Stars represent GTPase motifs
  • Figure 2F shows alignment of predicted transmembrane domain and surrounding region in Fzo and homologs Charged residues are in boldface Asterisks indicate 13- and 20-res ⁇ due uncharged regions in Fzo, note similar distribution of charged and uncharged residues in homologs
  • Figure 2G is alpha helical projections of homologous regions near the N termini of Fzo and the C elegans homolog showing hydrophobic, acidic, and basic/polar faces
  • Figure 2H is predicted isoelect ⁇ c points for regions of Fzo and homologs after conceptual division at the transmembrane region Gray bars, N terminus to transmembrane region, black bars, transmembrane region to C terminus
  • mitofusin Fzo is the first known protein mediator of mitochondrial fusion
  • mitofusin indicates the Drosophila Fzo protein or any of its homologs from insects, other invertebrates, yeast, and vertebrates, including mouse and humans
  • the provided mitofusin genes and fragments thereof and genomic regulatory regions are useful in the production of mitofusin protein, as a probe for detection of mitofusin specific sequences and related genes, and for the modulation of gene activity
  • the encoded mitofusin protein is useful in enhancing the fusion of mitochondria, as an immunogen to raise specific antibodies, in drug screening for compositions that mimic or modulate mitofusin activity or expression, including altered forms of mitofusin protein
  • the mitofusin genes and encoded proteins may be used to drive or mediate mitochondrial fusion for the purposes of introducing intact or partial mitochondrial genomes into mitochondria in diseased host cells or tissues, either in culture or in the intact animal or individual
  • the use of mitofusin proteins to drive fusion of membrane-bound entities, e g mitochondria, in vitro enables investigation of the mechanism of mitochondrial fusion, recombination or complementation between mitochondrial DNA molecules and maintenance of mitochondrial genomes
  • Mitofusins are herein identified in D melanogaster, S cerevisiae, C elegans, and humans Specific examples of mitofusin genes are provided in SEQ ID NO 1 (Drosophila) and SEQ ID NO 3 (human) The respective encoded proteins are provided as SEQ ID NO 2 and SEQ ID NO 4, respectively Two distinct forms have been identified in humans
  • the yeast, worm and human homologs share signature conserved features with Drosophila Fzo protein All are large GTPases with a predicted trans-membrane domain flanked by predicted coiled-coil regions near the C-termmus The region of the protein C-terminal to the predicted trans- membrane domain has a high pi, characteristic of proteins found in the mitochondrial matrix, while the region of the protein N-terminal to the predicted trans-membrane domain has a lower pi, characteristic of cytoplasmic proteins
  • nucleic Acids encoding Mitofusins The nucleic acid sequence encoding a mitofusin may be cDNA or genomic DNA or a fragment thereof
  • mitofusin gene shall be intended to mean the open reading frame encoding specific mitofusin polypeptides, introns, as well as adjacent 5' and 3' non-coding nucleotide sequences involved in the regulation of expression, up to about 1 kb beyond the coding region, but possibly further in either direction
  • the gene may be introduced into an appropriate vector for extrachromosomal maintenance or for integration into the host
  • cDNA as used herein is intended to include all nucleic acids that share the arrangement of sequence elements found in native mature mRNA species, where sequence elements are exons and 3' and 5' non-coding regions Normally mRNA species have contiguous exons, with the intervening introns, when present, removed by nuclear RNA splicing, to create a continuous open reading frame encoding a mitofusin protein
  • a genomic sequence of interest comprises the nucleic acid present between the initiation codon and the stop codon, as defined in the listed sequences, including all of the introns that are normally present in a native chromosome It may further include the 3' and 5' untranslated regions found in the mature mRNA It may further include specific transc ⁇ ptional and translational regulatory sequences, such as promoters, enhancers, etc , including about 1 kb, but possibly more, of flanking genomic DNA at either the 5' or 3' end of the transcribed region
  • the genomic DNA may be isolated as a fragment of 100 kbp or smaller, and substantially free of flanking chromosomal sequence
  • the genomic DNA flanking the coding region, either 3' or 5', or internal regulatory sequences as sometimes found in introns, contains sequences required for proper tissue and stage specific expression
  • Genomic regions of interest include the non-transcribed sequences 5' or 3' to a mitofusin gene, for example as shown in Figure 3B
  • This region of DNA contains the native promoter elements that direct expression of the linked fzo gene
  • a promoter region will usually have at least about 100 nt of sequence located 5' to a mitofusin gene and will often extend 5' to include other regulatory elements
  • sequence of this 5' region may be utilized for promoter elements, including enhancer binding sites, that provide for developmental regulation in tissues where mitofusins are expressed
  • tissue specific expression is useful for determining the pattern of expression, and for providing promoters that mimic the native pattern of expression
  • Naturally occurring polymorphisms in the promoter region are useful for determining natural variations in expression, particularly those that may be associated with disease
  • mutations may be introduced into the promoter region to determine the effect of altering expression in experimentally defined systems
  • Methods for the identification of specific 5 DNA motifs involved in the binding of transc ⁇ ptional factors are known in the art, e g sequence similarity to known binding motifs, gel retardation studies, etc For examples, see Blackwell et al (1995. Mol Med 1 194-205. Mortlock et al (1996 ) Genome Res 6 327-33, and Joulin and Richard-Foy (1995) Eur J Biochem 232 620-626
  • the regulatory sequences may be used to identify cis acting sequences required for
  • transc ⁇ ptional or translational regulation of mitofusin expression especially in different tissues or stages of development, and to identify cis acting sequences and trans acting factors that regulate or mediate mitofusin expression
  • Such transcription or translational control regions may be operably linked to a mitofusin gene in order to promote expression of wild type or altered human or Drosophila mitofusins or other proteins of interest in cultured ceils, or in embryonic, fetal or adult
  • coding sequences may be used to identify cis acting sequences and possible trans-acting factors that regulate or mediate import of mitofusins into or on to mitochondria
  • Such mitochondrial import signals may be operably linked to a mitofusin or other gene to promote import of wild-type or altered mitofusins or other proteins of interest into or onto mitochondria in vivo or in
  • nucleic acid compositions of the subject invention may encode all or a part of the subject polypeptides Double or single stranded fragments may be obtained of the DNA sequence by chemically synthesizing oligonucleotides in accordance with conventional methods, by restriction enzyme digestion, by PCR amplification, etc For the most part, DNA fragments will be of at least
  • primers that will generate an amplification product of at least about 50 nt, preferably at least about 100 nt
  • Algorithms for the selection of primer sequences are generally known, and are available in commercial software packages
  • Amplification primers hybridize to complementary strands of DNA, and will prime towards each other
  • the mitofusin genes are isolated and obtained in substantial purity, generally as other than an intact chromosome Usually, the DNA will be obtained substantially free of other nucleic acid sequences that do not include a mitofusin sequence or fragment thereof, generally being at least about 50%, usually at least about 90% pure and are typically ' recombinant", ; e flanked by one or more nucleotides with which it is not normally associated on a naturally occurring chromosome
  • DNA sequences are used in a variety of ways They may be used as probes for identifying mitofusin related genes Between mammalian species, e g human and mouse, homologs have substantial sequence similarity, / e at least 75% sequence identity between nucleotide sequences Sequence similarity is calculated based on a reference sequence, which may be a subset of a larger sequence, such as a conserved motif, coding region, flanking region, etc A reference sequence will usually be at least about 18 nt long, more usually at least about 30 nt long, and may extend to the complete sequence that is being compared Algorithms for sequence analysis are known in the art, such as BLAST, described in Altschul et al (1990) J Mol.
  • the ammo acid sequence similarity between invertebrates and vertebrates is sufficient to identify homologous genes Regions of more highly conserved sequence are identified through a comparison of the provided sequences, for examples as shown in Figure 4B and 4C Such conserved regions are used to design degenerate oligonucleotide primers, as known in the art Conveniently, the primers are used in an polymerase chain reaction amplification with cDNA or genomic DNA from the target organism as a substrate
  • the resulting amplified nucleic acid product comprises a fragment of the mitofusin from the target organism, and can be used to isolate the complete gene by various methods known in the art, including rapid amplification of cloned ends (RACE), hybridization to cDNA libraries, etc
  • Nucleic acids having sequence similarity are detected by hybridization under low stringency conditions, for example, at 50°C and 10XSSC (0 9 M sal ⁇ ne/0 09 M sodium citrate) and remain bound when subjected to washing at 55 C C in 1XSSC Sequence identity may be determined by hybridization under stringent conditions, for example, at 50°C or higher and 0 1XSSC (9 mM sal ⁇ ne/0 9 mM sodium citrate)
  • probes, particularly labeled probes of DNA sequences one can isolate homologous or related genes
  • the source of homologous genes may be any species, e g primate species, particularly human, rodents, such as rats and mice, canines, felines, bovines, ov es, equines, yeast, Drosophila, Caenhorabditis, etc
  • the DNA may also be used to identify expression of the gene in a biological specimen
  • a biological specimen is used as a source of mRNA
  • the mRNA may be amplified by RT-PCR, using reverse transcnptase to form a complementary DNA strand, followed by polymerase chain reaction amplification using primers specific for the subject DNA sequences
  • the mRNA sample is separated by gel electrophoresis, transferred to a suitable support, e g nitrocellulose, nylon, etc , and then probed with a fragment of the subject DNA as a probe
  • Other techniques such as oligonucleotide ligation assays, in situ hybridizations, and hybridization to DNA probes arrayed on a solid chip may also find use Detection of mRNA hybridizing to the subject sequence is indicative of mitofusin gene expression
  • sequence of a mitofusin gene may be mutated in various ways known in the art to generate targeted changes in promoter 5 strength, sequence of the encoded protein, efc
  • the DNA sequence or product product of such a mutation will be substantially similar to the sequences provided herein, / e will differ by at least one nucleotide or ammo acid, respectively, and may differ by at least two but not more than about ten nucleotides or ammo acids
  • sequence changes may be substitutions, insertions or deletions
  • Deletions may further include larger changes, such as deletions of a domain or exon Other
  • modifications of interest include epitope tagging, e g with the FLAg system, HA, efc
  • epitope tagging e g with the FLAg system
  • HA efc
  • fusion proteins with green fluorescent proteins GFP
  • Such mutated genes may be used to study structure-function relationships of mitofusins, or to alter properties of the protein that affect its function or regulation For example, constitutively active fusogens, or a dominant negatively active protein to block fusion, may be created in this manner
  • the subject gene may be employed for producing all or portions of the mitofusin protein
  • an expression cassette may be employed The expression vector will provide a transc ⁇ ptional and translational initiation region, which may be mducible or constitutive, where the coding region is operably linked under the transc ⁇ ptional control of the transcnptional initiation region, and a transcnptional and translational termination region These control regions may be
  • the peptide may be expressed in prokaryotes or eukaryotes in accordance with conventional ways, depending upon the purpose for expression
  • a unicellular organism such as E coli, B subtilis, S cerevisiae, insect cells in combination with baculovirus vectors, or cells of a higher organism such as vertebrates, particularly mammals,
  • 35 e g COS 7 cells may be used as the expression host cells
  • Small peptides can also be synthesized in the laboratory Peptides that are subsets of a complete mitofusin sequence, usually at least about 8 ammo acids in length, more usually at least about 12 ammo may be used to identify and investigate parts of the protein important for function, such as the GTPase domain, mitochondrial import signals, or the coiled-coil regions, or to raise antibodies directed against these regions
  • the protein may be isolated and purified in accordance with conventional ways
  • a lysate may be prepared of the expression host and the lysate purified using HPLC, exclusion chromatography, gel electrophoresis, affinity chromatography, or other purification technique
  • the purified protein will generally be at least about
  • the use of the expressed protein for in vitro models of mitochondrial fusion is of particular interest
  • the protein may be used to explore conditions and sequences that are required for association of the protein with mitochondria in cell extracts, and for setting up in vitro systems to assay mitochondrial fusion
  • the expressed mitofusin polypeptides are used for the production of antibodies, where short fragments provide for antibodies specific for the particular polypeptide, and larger fragments or the entire protein allow for the production of antibodies over the surface of the polypeptide
  • Antibodies may be raised to the wild-type or variant forms of mitofusin
  • Antibodies may be raised to isolated peptides corresponding to these domains, or to the native protein, e g by immunization with cells expressing mitofusin, immunization with hposomes having mitofusin inserted in the membrane, efc
  • Antibodies are prepared in accordance with conventional ways, where the expressed polypeptide or protein is used as an immunogen, by itself or conjugated to known immunogenic carriers, e g KLH, pre-S HBsAg, other viral or eukaryotic proteins, or the like Various adjuvants may be employed, with a series of injections, as appropriate For monoclonal antibodies, after one or more booster injections, the spleen is isolated, the lymphocytes immortalized by cell fusion, and then screened for high affinity antibody binding The immortalized cells, ; e hyb ⁇ domas, producing the desired antibodies may then be expanded For further description, see Monoclonal Antibodies A Laboratory Manual.
  • the mRNA encoding the heavy and light chains may be isolated and mutagenized by cloning in E coli, and the heavy and light chains mixed to further enhance the affinity of the antibody
  • Alternatives to in vivo immunization as a method of raising antibodies include binding to phage "display" libraries, usually in conjunction with in vitro affinity maturation
  • Biochemical studies may be performed to determine whether a sequence polymorphism in a mitofusin coding region or control regions is associated with disease, particularly degenerative diseases associated with mitochondrial defects, e g amyotrophic lateral sclerosis, Huntmgton's disease, Parkinson's disease, and Alzheimer's disease Disease associated polymorphisms may include mutations that alter expression level, that affect the fusogenic activity of the protein, that alter the subcellular localization of the mitofusin, efc
  • Changes in the promoter or enhancer sequence that may affect expression levels of mitofusin can be compared to expression levels of the normal allele by various methods known in the art
  • Methods for determining promoter or enhancer strength include quantitation of the expressed natural protein, insertion of the variant control element into a vector with a reporter gene such as ⁇ -galactosidase, luciferase, chloramphenicol acetyltransferase, etc that provides for convenient quantitation, and the like
  • a reporter gene such as ⁇ -galactosidase, luciferase, chloramphenicol acetyltransferase, etc that provides for convenient quantitation, and the like
  • a number of methods are available for analyzing nucleic acids for the presence of a specific sequence Where large amounts of DNA are available, genomic DNA is used directly Alternatively, the region of interest is cloned into a suitable vector and grown in sufficient quantity for analysis
  • Cells that express a mitofusin may
  • Amplification may also be used to determine whether a polymorphism is present, by using a primer that is specific for the polymorphism
  • various methods are known in the art that utilize oligonucleotide ligation as a means of detecting polymorphisms, for examples see Riley ef al (1990) N A R 18 2887-2890, and Delahunty ef al (1996. Am J Hum Genet 58 1239-1246
  • a detectable label may be included in an amplification reaction
  • Suitable labels include fluorochromes, e g fluorescem isothiocyanate (FITC), rhodamme, Texas Red, phycoeryth ⁇ n, allophycocyanm, 6-carboxyfluoresce ⁇ n (6-FAM), 2',7'-d ⁇ methoxy-4',5'-d ⁇ chloro-6-carboxyfluoresce ⁇ n (JOE), 6-carboxy-X-rhodam ⁇ ne (ROX), 6-carboxy-2',4',7',4,7-hexachlorofluoresce ⁇ n (HEX), 5-carboxyfluoresce ⁇ n (5-FAM) or N,N,N',N'-tetramethyl-6-carboxyrhodam ⁇ ne (TAMRA), radioactive labels, e g 32 P, 35 S, 3 H, etc
  • the label may be a two stage system, where the amplified DNA is conjugated to biot
  • the amplified or cloned fragment may be sequenced by dideoxy or other methods, and the sequence of bases compared to a wild-type mitofusin sequence Hybridization with the variant sequence may also be used to determine its presence, by Southern blots, dot blots, efc Single strand conformational polymorphism (SSCP) analysis, denaturing gradient gel electrophoresis (DGGE), and heteroduplex analysis in gel matrices are used to detect conformational changes created by DNA sequence variation as alterations in electrophoretic mobility
  • the hybridization pattern of a control and variant sequence to an array of oligonucleotide probes immobilised on a solid support, as described in WO 95/11995, may also be used as a means of detecting the presence of variant sequences Alternatively, where a polymorphism creates or destroys a recognition site for a restriction endonuclease, the fragment is digested with that endonuclease, and the products size fractionated
  • Hybridization with a polymorphism specific probe may also be used to determine its presence, by Southern blots, dot blots, efc
  • the hybridization pattern of a control and variant sequence to an array of oligonucleotide probes immobilised on a solid support, as described in US 5,445,934, or in WO95/35505, may also be used as a means of detecting the presence of variant sequences
  • Screening for mutations in mitofusin may be based on the functional or antigenic characteristics of the protein
  • Various immunoassays designed to detect polymorphisms in mitofusin proteins may be used in screening Where many diverse genetic mutations lead to a particular disease phenotype, functional protein assays have proven to be effective screening tools
  • the activity of the encoded mitofusin protein may be determined by comparison with the wild-type protein
  • Antibodies specific for a mitofusin may be used in staining or in immunoassays
  • Samples include biological fluids such as semen, blood, cerebrospmal fluid, tears, saliva, lymph, dialysis fluid and the like, organ or tissue culture derived fluids, and fluids extracted from physiological tissues Also included in the term are derivatives and fractions of such fluids
  • the cells may be dissociated, in the case of solid tissues, or tissue sections may be analyzed Alternatively a lysate of the cells may be prepared
  • Diagnosis may be performed by a number of methods to determine the absence or presence or altered amounts of normal or abnormal mitofusin in patient cells
  • detection may utilize staining of cells or histological sections, performed in accordance with conventional methods
  • Cells are permeabilized to stain intra-mitochondnal and/or cytoplasmic molecules
  • the antibodies of interest are added to the cell sample, and incubated for a period of time sufficient to allow binding to the epitope, usually at least about 10 minutes
  • the antibody may be labeled with radioisotopes, enzymes, fluorescers, chemilummescers, or other labels for direct detection Alternatively, a second stage antibody or reagent is used to amplify the signal Such reagents are well known in the art
  • the primary antibody may be conjugated to biotm, with horseradish peroxidase-conjugated avid added as a second stage reagent
  • the secondary antibody conjugated to a flourescent compound e g flourescem,
  • a functional assay may be based on the membrane changes mediated by mitofusin gene products
  • Other assays may, for example, detect conformational changes, or changes in the subcellular localization of mitofusin proteins
  • the mitofusin genes, gene fragments, or the encoded protein or protein fragments are useful in gene therapy to treat degenerative and other disorders involving mitochondria, including myopathies and Alzheimer's disease
  • Expression vectors may be used to introduce the mitofusin gene into a cell
  • Such vectors generally have convenient restriction sites located near the promoter sequence to provide for the insertion of nucleic acid sequences
  • Transcription cassettes may be prepared comprising a transcription initiation region, the target gene or fragment thereof, and a transcnptional termination region
  • the transcription cassettes may be introduced into a variety of vectors, e g plasmid, retrovirus, e g lentivirus, adenovirus, and the like, where the vectors are able to transiently or stably be maintained in the cells, usually for a period of at least about one day, more usually for a period of at least about several days to several weeks
  • the gene or mitofusin protein may be introduced into tissues or host cells by any number of routes, including viral infection, microinjection, or fusion of vesicles or mitochondria Jet injection may also be used for intramuscular administration, as described by Furth ef al (1992) Anal Biochem 205 365-368
  • the DNA may be coated onto gold microparticles, and delivered intradermally by a particle bombardment device, or "gene gun” as described in the literature (see, for example, Tang ef al (1992) Nature 356 152-154), where gold microprojectiles are coated with the mitofusin or DNA, then bombarded into skin cells
  • Antisense molecules can be used to down-regulate expression of mitofusin in cells
  • the anti-sense reagent may be antisense oligonucleotides (ODN), particularly synthetic ODN having chemical modifications from native nucleic acids, or nucleic acid constructs that express such anti- sense molecules as RNA
  • ODN antisense oligonucleotides
  • a specific region or regions of the endogenous sense strand mRNA sequence is chosen to be complemented by the antisense sequence
  • Selection of a specific sequence for the oligonucleotide may use an empirical method, where several candidate sequences are assayed for inhibition of expression of the target gene in an in vitro or animal model
  • a combination of sequences may also be used, where several regions of the mRNA sequence are selected for antisense complementation
  • Antisense oligonucleotides may be chemically synthesized by methods known in the art (see Wagner ef al (1993) supra and Milligan ef al , supra ) Preferred oligonucleotides are chemically modified from the native phosphodiester structure, in order to increase their mtracellular stability and binding affinity A number of such modifications have been described in the literature, which alter the chemistry of the backbone, sugars or heterocyclic bases
  • phosphorothioates phosphorodithioates, where both of the non-bridging oxygens are substituted with sulfur, phosphoroamidites, alkyl phosphotnesters and boranophosphates
  • Achiral phosphate derivatives include 3'-0'-5'-S-phosphoroth ⁇ oate, 3'-S-5'-0-phosphoroth ⁇ oate, 3'-CH2-5'-0-phosphonate and 3'- NH-5'-0-phosphoroam ⁇ date
  • Peptide nucleic acids replace the entire ribose phosphodiester backbone with a peptide linkage
  • Sugar modifications are also used to enhance stability and affinity
  • the ⁇ -anomer of deoxy ⁇ bose may be used, where the base is inverted with respect to the natural ⁇ -anomer
  • the 2'-OH of the ribose sugar may be altered to form 2'-0-methyl or 2'-0-allyl sugars, which provides resistance to degradation without comprising affinity Modification of
  • catalytic nucleic acid compounds e g ⁇ bozymes, anti-sense conjugates, etc may be used to inhibit gene expression
  • Ribozymes may be synthesized in vitro and administered to the patient, or may be encoded on an expression vector, from which the nbozyme is synthesized in the targeted cell (for example, see International patent application WO 9523225, and Beigelman et al (1995) Nucl Acids Res 234434-42)
  • Examples of oligonucleotides with catalytic activity are described in WO 9506764 Conjugates of anti-sense ODN with a metal complex, e g terpyr ⁇ dylCu(ll), capable of mediating mRNA hydrolysis are described in Bashkin ef al (1995) Appl Biochem Biotechnol 54 43-56
  • the subject nucleic acids can be used to generate genetically modified non-human animals or site specific gene modifications in cell lines, for the study of mitofusin function or regulation, or to create animal models of diseases, including mitochondrial diseases, muscle myopathies, neurodegenerative disorders, and aging
  • transgenic is intended to encompass genetically modified animals having a deletion or other knock-out of mitofusin gene activity, having an exogenous mitofusin gene that is stably transmitted in the host cells where the gene may be altered in sequence to produce a modified protein, or having an exogenous mitofusin promoter operably linked to a reporter gene
  • Transgenic animals may be made through homologous recombination, where the mitofusin locus is altered Alternatively, a nucleic acid construct is randomly integrated into the genome Vectors for stable integration include plasmids, retroviruses and other animal viruses, YACs, and the like Of interest are transgenic mammals, e g cows, pigs, goats, horses, efc
  • Investigation of genetic function may also utilize non-mammalian models, particularly using those organisms that are biologically and genetically well-characterized, such as C elegans, D melanogaster and S cerevisiae
  • the subject gene sequences may be used to knock-out corresponding gene function or to complement defined genetic lesions in order to determine the physiological and biochemical pathways involved in mitofusin function
  • Drug screening may be performed in combination with complementation or knock-out studies, e g to study progression of degenerative disease, to test therapies, or for drug discovery
  • the modified cells or animals are useful in the study of mitofusin function and regulation
  • a series of small deletions and/or substitutions may be made in the mitofusin gene to determine the role of different domains in GTPase activity, membrane fusion, efc
  • Specific constructs of interest include, but are not limited to, anti-sense mitofusin constructs to block mitofusin expression, expression of dominant negative mitofusin mutations, and over-expression of a mitofusin gene
  • One may also provide for expression of the mitofusin gene or variants thereof in cells or tissues where it is not normally expressed or at abnormal times of development
  • DNA constructs for homologous recombination will comprise at least a portion of the mitofusin gene with the desired genetic modification, and will include regions of homology to the target locus DNA constructs for random integration need not include regions of homology to mediate recombination Conveniently
  • the chime ⁇ c animals are screened for the presence of the modified gene and males and females having the modification are mated to produce homozygous progeny If the gene alterations cause lethality at some point in development, tissues or organs can be maintained as allogeneic or congenic grafts or transplants, or in in vitro culture
  • the transgenic animals may be any non-human mammal, such as laboratory animals, domestic animals, etc
  • the transgenic animals may be used in functional studies, drug screening, etc , e g to determine the effect of a candidate drug on mitochondrial fusion, to test potential therapeutics or treatment regimens, efc
  • Drug Screening Assays By providing for the production of large amounts of mitofusin protein, one can identify ligands or substrates that bind to, modulate or mimic the action of mitofusin Drug screening identifies agents that provide a replacement or enhancement for mitofusin function in affected cells
  • assays may be used for this purpose, including labeled in vitro protein-protein binding assays, prote -DNA binding assays, protein-mitochondria or protein mitochindna fraction, electrophoretic mobility shift assays, immunoassays for protein binding, and the like All or a fragment of the purified protein may also be used for determination of three-dimensional crystal structure, which can be used for modeling intermolecular interactions, membrane fusion, efc Altered mitofusin molecules, either as an isolated recombinant protein, or as a genetically modified cell or animal model may be assayed to investigate structure function parameters, including but not limited to potential mitochondrial import sequences, protein interaction domains, GTPase
  • agent as used herein describes any molecule, e g protein or pharmaceutical, with the capability of altering or mimicking the physiological function of a subject mitofusin Generally a plurality of assay mixtures are run in parallel with different agent concentrations to obtain a differential response to the various concentrations Typically, one of these concentrations serves as a negative control, / e at zero concentration or below the level of detection
  • Candidate agents encompass numerous chemical classes, though typically they are organic molecules, preferably small organic compounds having a molecular weight of more than 50 and less than about 2,500 daltons
  • Candidate agents comprise functional groups necessary for structural interaction with proteins, particularly hydrogen bonding, and typically include at least an amme, carbonyl, hydroxyl or carboxyl group, preferably at least two of the functional chemical groups
  • the candidate agents often comprise cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more of the above functional groups
  • Candidate agents are also found among biomolecules including, but not limited to peptides, sacchandes, fatty acids, steroids, pu ⁇ nes, pyrimidines, derivatives, structural analogs or combinations thereof
  • Candidate agents are obtained from a wide variety of sources including libraries of synthetic or natural compounds For example, numerous means are available for random and directed synthesis of a wide variety of organic compounds and biomolecules, including expression of randomized oligonucleotides and oligopeptides Alternatively
  • the screening assay is a binding assay
  • the label can directly or indirectly provide a detectable signal
  • Various labels include radioisotopes, fluorescers, chemilummescers, enzymes, specific binding molecules, particles, e g magnetic particles, and the like
  • Specific binding molecules include pairs, such as biotm and streptavidm, digoxm and antidigoxm etc
  • the complementary member would normally be labeled with a molecule that provides for detection, in accordance with known procedures
  • reagents may be included in the screening assay These include reagents like salts, neutral proteins, e g albumin, detergents, etc that are used to facilitate optimal protein- protein binding and/or reduce non-specific or background interactions Reagents that improve the efficiency of the assay, such as protease inhibitors, nuclease inhibitors, anti-microbial agents, etc may be used.
  • the mixture of components are added in any order that provides for the requisite binding Incubations are performed at any suitable temperature, typically between 4 and 40 °C Incubation periods are selected for optimum activity, but may also be optimized to facilitate rapid high-throughput screening
  • the fzo 1 allele was isolated in a screen for ethyl methanesulfonate (EMS) induced recessive male sterile mutations
  • the fzo 2 allele was isolated in an EMS screen essentially as described in Lin ef al (1996) Devel 122 1331-1341, except that mutagenized chromosomes were tested for failure to complement fzo 1
  • One new allele, fzo 2 was identified from 1799 mutagenized third chromosomes screened
  • Df(3R)M95A (94D, 95A3) and Df(3R)EB6 (94C2-5, 94E3, Mohler ef al (1995) Devel 121, 237-247) were gifts from G Reuter and J Mohler, respectively
  • Fertility was determined by placing individual flies in yeasted vials with three males or virgin females as appropriate and scoring the presence or absence of larvae after seven days at 25°C Progeny of tested females were counted At least ten individual males and four individual females for each of the allelic combinations fzo 1 2fzo fzo 1 /fzo 2 , fzo 1 /Df(3R)P20, and fzo 2 /Df(3R)P20, as well as heterozygous sibling classes fzo/TM3 or Df(3R)P20/TM3 were tested (The fzo 2 chromosome carried a secondary lethal and could not be made homozygous )
  • testes from the above genotypes as well as from wild type Oregon R flies were dissected in TB1 buffer (7 mM K 2 HPO 7 mM KH 2 PO place [pH 6 7], 80 mM KCI, 16mM NaCI, 5mM MgCI 2 , 1% PEG-6000), opened with forceps to allow
  • testes were dissected in
  • RFLP Mapping The fzo 1 mutation was further localized by recombination with respect to 35 restriction fragment length polymorphisms (RFLPs) in the interval between hh fiaf3 and a ry * marked P element insertion in the pointed gene associated with the pnt 07 ⁇ 25 allele
  • RFLPs restriction fragment length polymorphisms
  • the parental genomic DNAs were digested separately with
  • the pKH3fzo + plasmid and a plasmid encoding the ⁇ 2-3 constitutively active transposase were mixed in a 3 1 molar ratio to a final DNA concentration of 0 4 mg/ml and injected into y w 6 or w 1118 embryos
  • An additional insertion on the second chromosome was obtained by mobilizing one of the third chromosome insertions with the TM3, Sb ⁇ 2-3 transposase source
  • all four independent fzo nsertions were crossed into w, fzo mutant backgrounds by independent assortment for the second chromosome insertions and by recombination onto a hh bar3 fzo 1 chromosome and then independent assortment for the third chromosome insertions, which were tested for rescue only of allelic combinations with f
  • RNA in situ Hybridization For in situ hybridization to testes, digoxygenm-labeled RNA probes representing both the fzo cDNA sense and anti-sense strands (primed with T7 or T3 primers on a linearized cDNA construct) were made and submitted to alkaline hydrolysis (1 hour) as described in the Genius 4 RNA Labeling Kit user's guide (Boehnnger Mannheim) Paraformaldehyde fixation of freshly dissected testes was as in Tautz (1989) Chromosoma 98, 81-
  • Antibodies were raised to a fusion protein containing a 6-h ⁇ st ⁇ d ⁇ ne tag and the C-terminal 115 ammo acids of Fzo
  • a 0 5 kb BamHI/Hindlll restriction fragment from fzo cDNA1 (the Hmdlll site is from the Bluescript SK- multiple cloning site) was subcloned into vector pQE30 (QIAGEN)
  • the fusion protein was induced, 35 harvested under denaturing conditions, and purified on a Ni-NTA column with imidazole elution as described in the QIAexpressionist manual (QIAGEN) Aliquots from each column fraction were electrophoresed on an SDS-polyacrylamide gel and stained with Coomassie Blue Fractions with greatest purity and highest concentration of the fusion protein were dialyzed in 1 5 M urea, 0 1 M Na phosphate, 0 01 M T ⁇ s-HCI, and
  • fzo encodes a novel conserved predicted transmembrane GTPase
  • the fzo ' mutation was mapped by recombination to 0 5 cM distal of hh bar3 and 0 5 cM proximal of pnt 07825 , in agreement with complementation of fzo 1 by Df(3R)EB6 but not by Df(3R)M95A ( Figure 3A)
  • the fzo 1 mutation was further localized by RFLP mapping to an 11 kb region defined by polymorphic Xbal and EcoRI sites on an existing genomic phage walk ( Figures 2A, B)
  • a 12 kb deficiency (Df(3R)P20) generated by imprecise excision of a nearby P element (cnc ° 3871 ) failed to complement fzo mutations, consistent with the RFLP mapping data cDNA clones corresponding to transcripts from the fzo region were isolated from a
  • the representative fzo cDNA was sequenced fully on both strands and shown to have a complete open reading frame (ORF) with flanking AT-nch sequences and a single consensus translational start site
  • the fzo transcription unit encodes a predicted protein of 718 ammo acids Database searches with the ammo acid sequence identified ORFs that encode related predicted proteins in mammals, C elegans, and S cerevisiae Human expressed sequence tags (ESTs) from brain (T06373, R20140, T37724), heart (AA248162, AA248083), fibroblasts (W49736), liver/spleen (H58349, AA010217), and pancreas (AA155601) encode predicted peptides homologous to Fzo
  • the cDNA clone from which a human fetal brain EST (T06373) was derived was sequenced and shown to encode an incomplete predicted protein (lacking its N terminus) with 35% identity to Fzo
  • the region of highest homology between the Fzo, human fetal brain (50% identity to Fzo), C elegans (49%), and S cerevisiae (20%) predicted proteins is a 186 ammo acid region containing four completely conserved signature motifs found in virtually all GTPases (G1-G4 in Figure 2A-C)
  • the C saccharolyticum predicted protein also contains these motifs Outside the individual motifs there is no significant similarity to any known GTPase
  • the spacing between the GTPase motifs, their N-termmal placement in Fzo and homologs, and the overall predicted protein size are reminiscent of the dynamm family (Warnock and Schmid, 1996 BioEssays 18, 885-893)
  • the G2 motif (Bourne ef al , 1991 Nature 349, 117-127) consists only of a conserved threonme and has not been defined in dynamms Both the Fzo and dynamin families have a conserved threonme exactly 20
  • fzo mRNA is expressed in primary spermatocytes
  • the fzo message was highly expressed in pre-meiotic primary spermatocytes during their growth period but was not detectable at earlier stages in germlme stem cells or mitotically dividing spermatogonia
  • the transcript persisted in meiotic cells but was not detectable in differentiating spermatids
  • the transcript was present in both fzo '/fzo 1 and fzo 2 /Df(3R)P20 testes
  • a control sense-strand fzo RNA probe did not hybridize in cells of any stage
  • Anti-Fzo 604 71 ⁇ antibodies detect greatly reduced levels of Fzo in fzo 1 and fzo 2 mutant fesfes
  • Antibodies raised against a fusion protein containing the C-terminal 115 residues of Fzo detected a wide doublet in Western blots of wild type testis extracts
  • the Fzo protein was partially resistant to extraction and denaturation and under standard denaturing conditions ran predominantly in streaky globs at an apparent molecular weight of 67 kD
  • some of the protein shifted up to a smoother band at an apparent molecular weight of 82 kD, the predicted size for Fzo
  • the wide Fzo doublet was greatly reduced in fzo* and fzo 2 /Df(3R)P20 mutant testes and increased in testes from wild type flies carrying six copies of the fzo + transgene
  • the Fzo protein is associated with mitochondria in early spermatids during a short time period that spans mitochondrial fusion
  • the Fzo protein was associated with mitochondria in wild type spermatids during a narrow developmental window corresponding to the time that Fzo function is required
  • Fzo is a novel predicted GTPase
  • the Drosophila fzo gene encodes a conserved predicted GTPase required for mitochondrial fusion during spermatogenesis and associated with mitochondria only during a short time spanning the fusion event
  • Fzo contains four motifs common to virtually all known GTPases and conserved among Fzo homologs from mammals to yeast
  • Fzo is the first protein to be assigned a function in this new family of large predicted transmembrane GTPases Within this family, the GTPase domain placement, motif spacing, and protein size (but not overall sequence) suggest a possible distant relationship with dynamms, which play a central biomechanical role in endocytic membrane trafficking
  • the fzo K99T and fzo R249L mutations are recessive Analogous mutations in mammalian dynamms cause dominant negative phenotypes when expressed in tissue culture cells, formation of macromoiecular dynamm ring-shaped complexes appears to require GTP binding by all subunits In contrast to dynamms, Fzo molecules may act individually or form complexes in which only some subunits must bind GTP for proper assembly or function
  • mitofusins span both membranes with its C terminus in the matrix, then the predicted GTPase domain would be oriented toward the cytoplasm, where it could mediate interactions between mitochondria or regulate assembly of additional proteins
  • the mitofusin protein could act as part of a ligand/receptor pair between separate mitochondria, binding to copies of itself or to other molecules displayed on the outer mitochondrial membrane Alternatively, the mitofusin protein could recruit other molecules to form a complex that links adjacent mitochondria Several regions of mitofusin could potentially participate in such protein-protein interactions, including the predicted coiled coil region at residues 377-400 as well as the N-termmal senne-rich region with its adjacent predicted "t ⁇ -amphipathic" ⁇ helix
  • mitofusin binding and hydrolysis by mitofusins may regulate the specificity of these protein-protein interactions, as Rab GTPases seem to regulate formation of the SNARE complex prior to membrane fusion in the secretory and endocytic pathways
  • the mitofusin predicted GTPase may have a biomechanical role, as may dynamin GTPases in the formation of endocytic vesicles
  • GTP hydrolysis could cause a conformational change in the mitofusin itself or in other recruited proteins, bringing membranes close together and/or exposing residues that could act as a hydrophobic bridge between the fusing mitochondria
  • Candidate regions in mitofusin to serve as such a bridge would be the hydrophobic faces of either the t ⁇ -amphipathic ⁇ helix or the other predicted coiled coil regions This scenario would be analogous to influenza virus fusion, during which low pH triggers a conformational change in HA, or to a proposed conformational change in SNARE proteins that may be
  • the Mitofusin family of predicted GTPases may mediate mitochondrial fusion and/or other functions in diverse cell types ESTs from genes encoding the human homologs were derived from brain, heart, pancreas, liver/spleen, and fibroblast cDNA libraries Mitochondrial fusion is known to occur in liver and is suggested to occur in certain rat neuronal cells and mouse 3T6 fibroblasts Mammalian spermatid mitochondria do not undergo massive fusion but become connected in a helical sheath around the sperm midpiece End-to- end contacts between mitochondria in this sheath contain structurally distinct "stud-like bridging elements", which seem to allow transmission of membrane potential through the helix Formation of these connections, which are also seen in rat cardiac tissue, may require the same protein mediators that are needed for full mitochondrial fusion in other cells Analysis of the role of mitofusins in diverse organisms will allow assignment of a general function for this new family of large multidomain GTPases
  • GenBank Accession Numbers The GenBank accession number for the fzo cDNA is U95821 and for the partial cDNA encoding the fzo human fetal brain homolog is U95822
  • GCAACTAATC TACGGCCTGG ATTGCCAATC CTATATGAGT
  • GACTTTCAGC CAGATCTTAG 1620
  • MOLECULE TYPE protein
  • SEQUENCE DESCRIPTION SEQ ID NO : 2 :
  • Val Ser Arg Leu lie Asp Glu Asn Lys Glu Arg Arg Ala Asn Leu Asn
  • Gin Leu lie Tyr Gly Leu Asp Cys Gin Ser Tyr Met Ser Asp Phe Gin
  • GCGTCCGTTA CATCTAGAAC TTCTATGGGC ATCATTATTG TTGGAGGAGT GATTTGGAAA 1080

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Mitofusin genes and encoded polypeptides are provided, including the Drosophila Fzo protein and its homologs from insects, other invertebrates, yeast, and vertebrates including mouse and humans. Mitofusins are large predicted GTPases with a predicted trans-membrane domain, coiled-coil regions, and a C-terminal region showing a high pl characteristic of mitochondrial matrix proteins. The mitofusins are the first known protein mediator of mitochondrial fusion, and mediate developmentally regulated post-meiotic fusion of mitochondria. Missense mutations that alter conserved residues required for GTP binding in other GTPases inhibit the in vivo fusogenic activity of Fzo but do not affect its localization.

Description

MITOFUSIN GENES AND THEIR USES
INTRODUCTION Accumulation of mutations in the mitochondrial genome has been proposed as an important contributor to aging and degenerative diseases In several cases human mitochondrial disorders have been shown to be caused by mutations or deletions of mitochondrial DNA
There is evidence for defects in energy metabolism, excitotoxicity, and for oxidative damage in the etiology of neurodegenerative diseases, including amyotrophic lateral sclerosis, Huntington's disease, Parkinson's disease, and Alzheimer's disease It is likely that there is a complex interplay between these mechanisms Mitochondrial DNA is particularly susceptible to oxidative stress, and there is evidence of age-dependent damage and deterioration of respiratory enzyme activities with normal aging This may contribute to the delayed onset and age dependence of neurodegenerative diseases
Mitochondria are dynamic organelles that undergo regulated fusion in many cell types Analysis of serial sections from rodent skeletal muscle, lymphocytes, liver, spinal ganglion cells, and from the yeast Saccharomyces cerevisiae has shown that all the mitochondrial material of a cell can exist as a giant branched reticulum
Specific protein mediators that act as biomechanical triggers and/or regulate specificity and timing of membrane fusion events have been identified in a wide variety of other cellular and subcellular contexts The best characterized biomechanically acting fusogen is influenza virus hemagglutinin (HA) that mediates fusion of endocytosed viruses to cells Regions resembling the HA fusion peptide exist in the ADAM family proteins, which are implicated in sperm/egg and myoblast fusion
The identification of protein mediators of mitochondrial fusion and their possible role in maintenance of mitochondrial function and genomic integrity is of great interest for diagnosis, drug screening and potential therapeutic strategies, including targeted delivery of genes, proteins and molecules to existing mitochondria If recombination between differently mutated mitochondrial DNA molecules allows restoration of a functional copy, the ability of mitochondria to fuse may play an important role in maintenance of mitochondrial genomes Alternatively, fusion of mitochondria may allow complementation between two mutations in different genes in the mitochondrial genome, allowing restoration of mitochondrial function even in the absence of recombination
Relevant literature
Larsson, N -G , and D A Clayton 1995 Molecular genetic aspects of human mitochondrial disorders Annual Review of Genetics 29:151-178 Kawano, S , H Takano, and T Kuroiwa 1995 Sexuality of mitochondria fusion, recombination, and plasmids Int Rev Cytol 161:49-110
SUMMARY OF THE INVENTION Mitofusin genes and proteins are provided As used herein, the term "mitofusin" indicates the Drosophila Fzo protein or any of its homologues from insects, other invertebrates, yeast, and vertebrates including mouse and humans Motifusms are large predicted GTPases with a predicted trans-membrane domain, coiled-coil regions, and a C-terminal region showing a high pi characteristic of mitochondrial matrix proteins The mitofusin Fzo is the first known protein mediator of mitochondrial fusion, and mediates developmentally regulated post-meiotic fusion of mitochondria in Drosophila spermatids
BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 A is a genetic, cytological, and molecular map of the fzo region on chromosome 3R Stippled numbered boxes represent polytene chromosome bands, open bars are deficiencies, with + indicating complementation of fzo and - indicating failure to complement The distal breakpoint of Df(3R)EB6 is at +2 kb on a genomic phage walk that extends 90 kb distal from hh Figure 1 B is a molecular map of the fzo region of the genomic walk, with coordinates 0 to 20 indicated in kb Deficiencies represented as in (A) Restriction sites R, EcoRI, X, Xbal, Z, Xhol Thick line on molecular map represents genomic DNA used to screen testis cDNA library This region indicates a genomic fragment that has all sequences required to express fzo in vivo, and to restore fzo function in mutants
Figure 2A shows the predicted ammo acid sequence (single letter ammo acid code) of the fzo gene product, with regions matching GTPase motifs in boldface and numbered G1-G4 Underlined region is the predicted transmembrane domain Italics show the hydrophobic region including the predicted transmembrane domain and 13 adjacent uncharged residues Asterisks indicate predicted coiled coil regions with scores above 0 4 Letters above ammo acid sequence indicate introduced missense mutations
Figures 2B and 2C show alignments of predicted G1 and G2 motifs (B) and G3 and G4 motifs (C) in Fzo and homologs Boxes indicate identities with Fzo
Figure 2D is a schematic diagram of Fzo and human, C elegans, and S cerevisiae homologs, showing predicted GTPase domain (stippled box) and predicted transmembrane domain (dark bar) Spirals indicate predicted coiled coil regions with probabilities between 0 4 and 1 0 (large spirals) and between 0 2 and 0 4 (small spirals), respectively Percentage identities to Fzo overall and within the predicted GTPase domains are indicated
Figure 2E shows overlaid plots of coiled coil probabilities (open) and hydrophobicity (filled- in) for Fzo and homologs Sequences were analyzed with the COILS program (MDITK matrix, weighted second and fifth heptad positions) and with the TMpred program using default parameters, a TMpred output of 0 5x103 or above is considered significant Stars represent GTPase motifs
Figure 2F shows alignment of predicted transmembrane domain and surrounding region in Fzo and homologs Charged residues are in boldface Asterisks indicate 13- and 20-resιdue uncharged regions in Fzo, note similar distribution of charged and uncharged residues in homologs
Figure 2G is alpha helical projections of homologous regions near the N termini of Fzo and the C elegans homolog showing hydrophobic, acidic, and basic/polar faces
Figure 2H is predicted isoelectπc points for regions of Fzo and homologs after conceptual division at the transmembrane region Gray bars, N terminus to transmembrane region, black bars, transmembrane region to C terminus
DESCRIPTION OF THE SPECIFIC EMBODIMENTS Mitofusin genes and proteins are provided The mitofusin Fzo is the first known protein mediator of mitochondrial fusion As used herein, the term "mitofusin" indicates the Drosophila Fzo protein or any of its homologs from insects, other invertebrates, yeast, and vertebrates, including mouse and humans
The provided mitofusin genes and fragments thereof and genomic regulatory regions are useful in the production of mitofusin protein, as a probe for detection of mitofusin specific sequences and related genes, and for the modulation of gene activity The encoded mitofusin protein is useful in enhancing the fusion of mitochondria, as an immunogen to raise specific antibodies, in drug screening for compositions that mimic or modulate mitofusin activity or expression, including altered forms of mitofusin protein
The mitofusin genes and encoded proteins may be used to drive or mediate mitochondrial fusion for the purposes of introducing intact or partial mitochondrial genomes into mitochondria in diseased host cells or tissues, either in culture or in the intact animal or individual The use of mitofusin proteins to drive fusion of membrane-bound entities, e g mitochondria, in vitro enables investigation of the mechanism of mitochondrial fusion, recombination or complementation between mitochondrial DNA molecules and maintenance of mitochondrial genomes
Mitofusins are herein identified in D melanogaster, S cerevisiae, C elegans, and humans Specific examples of mitofusin genes are provided in SEQ ID NO 1 (Drosophila) and SEQ ID NO 3 (human) The respective encoded proteins are provided as SEQ ID NO 2 and SEQ ID NO 4, respectively Two distinct forms have been identified in humans
The yeast, worm and human homologs share signature conserved features with Drosophila Fzo protein All are large GTPases with a predicted trans-membrane domain flanked by predicted coiled-coil regions near the C-termmus The region of the protein C-terminal to the predicted trans- membrane domain has a high pi, characteristic of proteins found in the mitochondrial matrix, while the region of the protein N-terminal to the predicted trans-membrane domain has a lower pi, characteristic of cytoplasmic proteins These observations, coupled with the association of mitofusin protein with mitochondria at the time of fusion, indicate that mitofusins are inserted into mitochondria with the C-terminal tail in the mitochondrial matrix and the GTPase domain either between the inner and outer mitochondrial membranes or exposed on the cytoplasmic face of mitochondria The predicted GTPase activity is required for mitochondrial fusion in vivo, but not for import of the protein into or association with mitochondria
Nucleic Acids encoding Mitofusins The nucleic acid sequence encoding a mitofusin may be cDNA or genomic DNA or a fragment thereof The term "mitofusin gene" shall be intended to mean the open reading frame encoding specific mitofusin polypeptides, introns, as well as adjacent 5' and 3' non-coding nucleotide sequences involved in the regulation of expression, up to about 1 kb beyond the coding region, but possibly further in either direction The gene may be introduced into an appropriate vector for extrachromosomal maintenance or for integration into the host
The term "cDNA" as used herein is intended to include all nucleic acids that share the arrangement of sequence elements found in native mature mRNA species, where sequence elements are exons and 3' and 5' non-coding regions Normally mRNA species have contiguous exons, with the intervening introns, when present, removed by nuclear RNA splicing, to create a continuous open reading frame encoding a mitofusin protein
A genomic sequence of interest comprises the nucleic acid present between the initiation codon and the stop codon, as defined in the listed sequences, including all of the introns that are normally present in a native chromosome It may further include the 3' and 5' untranslated regions found in the mature mRNA It may further include specific transcπptional and translational regulatory sequences, such as promoters, enhancers, etc , including about 1 kb, but possibly more, of flanking genomic DNA at either the 5' or 3' end of the transcribed region The genomic DNA may be isolated as a fragment of 100 kbp or smaller, and substantially free of flanking chromosomal sequence The genomic DNA flanking the coding region, either 3' or 5', or internal regulatory sequences as sometimes found in introns, contains sequences required for proper tissue and stage specific expression
Genomic regions of interest include the non-transcribed sequences 5' or 3' to a mitofusin gene, for example as shown in Figure 3B This region of DNA contains the native promoter elements that direct expression of the linked fzo gene A promoter region will usually have at least about 100 nt of sequence located 5' to a mitofusin gene and will often extend 5' to include other regulatory elements
The sequence of this 5' region may be utilized for promoter elements, including enhancer binding sites, that provide for developmental regulation in tissues where mitofusins are expressed The tissue specific expression is useful for determining the pattern of expression, and for providing promoters that mimic the native pattern of expression Naturally occurring polymorphisms in the promoter region are useful for determining natural variations in expression, particularly those that may be associated with disease
Alternatively, mutations may be introduced into the promoter region to determine the effect of altering expression in experimentally defined systems Methods for the identification of specific 5 DNA motifs involved in the binding of transcπptional factors are known in the art, e g sequence similarity to known binding motifs, gel retardation studies, etc For examples, see Blackwell et al (1995. Mol Med 1 194-205. Mortlock et al (1996 ) Genome Res 6 327-33, and Joulin and Richard-Foy (1995) Eur J Biochem 232 620-626
The regulatory sequences may be used to identify cis acting sequences required for
10 transcπptional or translational regulation of mitofusin expression, especially in different tissues or stages of development, and to identify cis acting sequences and trans acting factors that regulate or mediate mitofusin expression Such transcription or translational control regions may be operably linked to a mitofusin gene in order to promote expression of wild type or altered human or Drosophila mitofusins or other proteins of interest in cultured ceils, or in embryonic, fetal or adult
15 tissues, and for gene therapy
In addition coding sequences may be used to identify cis acting sequences and possible trans-acting factors that regulate or mediate import of mitofusins into or on to mitochondria Such mitochondrial import signals may be operably linked to a mitofusin or other gene to promote import of wild-type or altered mitofusins or other proteins of interest into or onto mitochondria in vivo or in
20 vitro
The nucleic acid compositions of the subject invention may encode all or a part of the subject polypeptides Double or single stranded fragments may be obtained of the DNA sequence by chemically synthesizing oligonucleotides in accordance with conventional methods, by restriction enzyme digestion, by PCR amplification, etc For the most part, DNA fragments will be of at least
25 15 nt, usually at least 18 nt, more usually at least about 50 nt Such small DNA fragments are useful as primers for PCR, hybridization screening, etc Larger DNA fragments, / e greater than 100 nt are useful for production of the encoded polypeptide For use in amplification reactions, such as PCR, a pair of primers will be used The exact composition of the primer sequences is not critical to the invention, but for most applications the primers will hybridize to the subject sequence
30 under stringent conditions, as known in the art It is preferable to choose a pair of primers that will generate an amplification product of at least about 50 nt, preferably at least about 100 nt Algorithms for the selection of primer sequences are generally known, and are available in commercial software packages Amplification primers hybridize to complementary strands of DNA, and will prime towards each other
35 The mitofusin genes are isolated and obtained in substantial purity, generally as other than an intact chromosome Usually, the DNA will be obtained substantially free of other nucleic acid sequences that do not include a mitofusin sequence or fragment thereof, generally being at least about 50%, usually at least about 90% pure and are typically ' recombinant", ; e flanked by one or more nucleotides with which it is not normally associated on a naturally occurring chromosome
The DNA sequences are used in a variety of ways They may be used as probes for identifying mitofusin related genes Between mammalian species, e g human and mouse, homologs have substantial sequence similarity, / e at least 75% sequence identity between nucleotide sequences Sequence similarity is calculated based on a reference sequence, which may be a subset of a larger sequence, such as a conserved motif, coding region, flanking region, etc A reference sequence will usually be at least about 18 nt long, more usually at least about 30 nt long, and may extend to the complete sequence that is being compared Algorithms for sequence analysis are known in the art, such as BLAST, described in Altschul et al (1990) J Mol
Figure imgf000008_0001
The ammo acid sequence similarity between invertebrates and vertebrates is sufficient to identify homologous genes Regions of more highly conserved sequence are identified through a comparison of the provided sequences, for examples as shown in Figure 4B and 4C Such conserved regions are used to design degenerate oligonucleotide primers, as known in the art Conveniently, the primers are used in an polymerase chain reaction amplification with cDNA or genomic DNA from the target organism as a substrate The resulting amplified nucleic acid product comprises a fragment of the mitofusin from the target organism, and can be used to isolate the complete gene by various methods known in the art, including rapid amplification of cloned ends (RACE), hybridization to cDNA libraries, etc
Nucleic acids having sequence similarity are detected by hybridization under low stringency conditions, for example, at 50°C and 10XSSC (0 9 M salιne/0 09 M sodium citrate) and remain bound when subjected to washing at 55CC in 1XSSC Sequence identity may be determined by hybridization under stringent conditions, for example, at 50°C or higher and 0 1XSSC (9 mM salιne/0 9 mM sodium citrate) By using probes, particularly labeled probes of DNA sequences, one can isolate homologous or related genes The source of homologous genes may be any species, e g primate species, particularly human, rodents, such as rats and mice, canines, felines, bovines, ov es, equines, yeast, Drosophila, Caenhorabditis, etc
The DNA may also be used to identify expression of the gene in a biological specimen The manner in which one probes cells for the presence of particular nucleotide sequences, as genomic DNA or RNA, is well established in the literature and does not require elaboration here Conveniently, a biological specimen is used as a source of mRNA The mRNA may be amplified by RT-PCR, using reverse transcnptase to form a complementary DNA strand, followed by polymerase chain reaction amplification using primers specific for the subject DNA sequences Alternatively, the mRNA sample is separated by gel electrophoresis, transferred to a suitable support, e g nitrocellulose, nylon, etc , and then probed with a fragment of the subject DNA as a probe Other techniques, such as oligonucleotide ligation assays, in situ hybridizations, and hybridization to DNA probes arrayed on a solid chip may also find use Detection of mRNA hybridizing to the subject sequence is indicative of mitofusin gene expression in the sample
The sequence of a mitofusin gene, including flanking promoter regions and coding regions, may be mutated in various ways known in the art to generate targeted changes in promoter 5 strength, sequence of the encoded protein, efc The DNA sequence or product product of such a mutation will be substantially similar to the sequences provided herein, / e will differ by at least one nucleotide or ammo acid, respectively, and may differ by at least two but not more than about ten nucleotides or ammo acids The sequence changes may be substitutions, insertions or deletions Deletions may further include larger changes, such as deletions of a domain or exon Other
10 modifications of interest include epitope tagging, e g with the FLAg system, HA, efc For studies of subcellular localization, fusion proteins with green fluorescent proteins (GFP) may be used
Techniques for in vitro mutagenesis of cloned genes are known Examples of protocols for site specific mutagenesis may be found in Gustm et al , Biotechniques 14 22 (1993), Barany, Gene 37 111-23 (1985), Colicelli ef al Mol Gen Genet 199 537-9 (1985), and Prentki et al Gene 29 303-
15 13 (1984) Methods for site specific mutagenesis can be found in Sambrook ef al , Molecular Cloning A Laboratory Manual, CSH Press 1989, pp 15 3-15 108, Werner et al , Gene 126 35-41 (1993), Sayers ef al , Biotechniques 13 592-6 (1992), Jones and Winistorfer, Biotechniques 12 528- 30 (1992), Barton ef al , Nucleic Acids Res 18 7349-55 (1990), Marotti and Tomich, Gene Anal Tech 6 67-70 (1989), and Zhu, Anal Biochem 177 120-4 (1989)
20 Such mutated genes may be used to study structure-function relationships of mitofusins, or to alter properties of the protein that affect its function or regulation For example, constitutively active fusogens, or a dominant negatively active protein to block fusion, may be created in this manner
Mitofusin Polvpeptides
25 The subject gene may be employed for producing all or portions of the mitofusin protein
For expression, an expression cassette may be employed The expression vector will provide a transcπptional and translational initiation region, which may be mducible or constitutive, where the coding region is operably linked under the transcπptional control of the transcnptional initiation region, and a transcnptional and translational termination region These control regions may be
30 native to a mitofusin gene, or may be derived from exogenous sources
The peptide may be expressed in prokaryotes or eukaryotes in accordance with conventional ways, depending upon the purpose for expression For large scale production of the protein, a unicellular organism, such as E coli, B subtilis, S cerevisiae, insect cells in combination with baculovirus vectors, or cells of a higher organism such as vertebrates, particularly mammals,
35 e g COS 7 cells, may be used as the expression host cells In many situations, it may be desirable to express the mitofusin gene in eukaryotic cells, where the mitofusin gene will benefit from native folding and post-translational modifications Small peptides can also be synthesized in the laboratory Peptides that are subsets of a complete mitofusin sequence, usually at least about 8 ammo acids in length, more usually at least about 12 ammo may be used to identify and investigate parts of the protein important for function, such as the GTPase domain, mitochondrial import signals, or the coiled-coil regions, or to raise antibodies directed against these regions With the availability of the protein or fragments thereof in large amounts, by employing an expression host, the protein may be isolated and purified in accordance with conventional ways A lysate may be prepared of the expression host and the lysate purified using HPLC, exclusion chromatography, gel electrophoresis, affinity chromatography, or other purification technique The purified protein will generally be at least about 80% pure, preferably at least about 90% pure, and may be up to and including 100% pure Pure is intended to mean free of other proteins, as well as cellular debris
The use of the expressed protein for in vitro models of mitochondrial fusion is of particular interest The protein may be used to explore conditions and sequences that are required for association of the protein with mitochondria in cell extracts, and for setting up in vitro systems to assay mitochondrial fusion
The expressed mitofusin polypeptides are used for the production of antibodies, where short fragments provide for antibodies specific for the particular polypeptide, and larger fragments or the entire protein allow for the production of antibodies over the surface of the polypeptide Antibodies may be raised to the wild-type or variant forms of mitofusin Antibodies may be raised to isolated peptides corresponding to these domains, or to the native protein, e g by immunization with cells expressing mitofusin, immunization with hposomes having mitofusin inserted in the membrane, efc
Antibodies are prepared in accordance with conventional ways, where the expressed polypeptide or protein is used as an immunogen, by itself or conjugated to known immunogenic carriers, e g KLH, pre-S HBsAg, other viral or eukaryotic proteins, or the like Various adjuvants may be employed, with a series of injections, as appropriate For monoclonal antibodies, after one or more booster injections, the spleen is isolated, the lymphocytes immortalized by cell fusion, and then screened for high affinity antibody binding The immortalized cells, ; e hybπdomas, producing the desired antibodies may then be expanded For further description, see Monoclonal Antibodies A Laboratory Manual. Harlow and Lane eds , Cold Spring Harbor Laboratories, Cold Spring Harbor, New York, 1988 If desired, the mRNA encoding the heavy and light chains may be isolated and mutagenized by cloning in E coli, and the heavy and light chains mixed to further enhance the affinity of the antibody Alternatives to in vivo immunization as a method of raising antibodies include binding to phage "display" libraries, usually in conjunction with in vitro affinity maturation
Diagnostic Uses Biochemical studies may be performed to determine whether a sequence polymorphism in a mitofusin coding region or control regions is associated with disease, particularly degenerative diseases associated with mitochondrial defects, e g amyotrophic lateral sclerosis, Huntmgton's disease, Parkinson's disease, and Alzheimer's disease Disease associated polymorphisms may include mutations that alter expression level, that affect the fusogenic activity of the protein, that alter the subcellular localization of the mitofusin, efc
Changes in the promoter or enhancer sequence that may affect expression levels of mitofusin can be compared to expression levels of the normal allele by various methods known in the art Methods for determining promoter or enhancer strength include quantitation of the expressed natural protein, insertion of the variant control element into a vector with a reporter gene such as β-galactosidase, luciferase, chloramphenicol acetyltransferase, etc that provides for convenient quantitation, and the like A number of methods are available for analyzing nucleic acids for the presence of a specific sequence Where large amounts of DNA are available, genomic DNA is used directly Alternatively, the region of interest is cloned into a suitable vector and grown in sufficient quantity for analysis Cells that express a mitofusin may be used as a source of mRNA, which may be assayed directly or reverse transcribed into cDNA for analysis The nucleic acid may be amplified by conventional techniques, such as the polymerase chain reaction (PCR), to provide sufficient amounts for analysis The use of the polymerase chain reaction is described in Saiki, ef al (1985) Science 239 487, and a review of current techniques may be found in Sambrook, ef al Molecular Cloning A Laboratory Manual. CSH Press 1989, pp 14 2-14 33 Amplification may also be used to determine whether a polymorphism is present, by using a primer that is specific for the polymorphism Alternatively, various methods are known in the art that utilize oligonucleotide ligation as a means of detecting polymorphisms, for examples see Riley ef al (1990) N A R 18 2887-2890, and Delahunty ef al (1996. Am J Hum Genet 58 1239-1246
A detectable label may be included in an amplification reaction Suitable labels include fluorochromes, e g fluorescem isothiocyanate (FITC), rhodamme, Texas Red, phycoerythπn, allophycocyanm, 6-carboxyfluoresceιn (6-FAM), 2',7'-dιmethoxy-4',5'-dιchloro-6-carboxyfluoresceιn (JOE), 6-carboxy-X-rhodamιne (ROX), 6-carboxy-2',4',7',4,7-hexachlorofluoresceιn (HEX), 5-carboxyfluoresceιn (5-FAM) or N,N,N',N'-tetramethyl-6-carboxyrhodamιne (TAMRA), radioactive labels, e g 32P, 35S, 3H, etc The label may be a two stage system, where the amplified DNA is conjugated to biotm, haptens, efc having a high affinity binding partner, e g avidm, specific antibodies, efc , where the binding partner is conjugated to a detectable label The label may be conjugated to one or both of the primers Alternatively, the pool of nucleotides used in the amplification is labeled, so as to incorporate the label into the amplification product
The amplified or cloned fragment may be sequenced by dideoxy or other methods, and the sequence of bases compared to a wild-type mitofusin sequence Hybridization with the variant sequence may also be used to determine its presence, by Southern blots, dot blots, efc Single strand conformational polymorphism (SSCP) analysis, denaturing gradient gel electrophoresis (DGGE), and heteroduplex analysis in gel matrices are used to detect conformational changes created by DNA sequence variation as alterations in electrophoretic mobility The hybridization pattern of a control and variant sequence to an array of oligonucleotide probes immobilised on a solid support, as described in WO 95/11995, may also be used as a means of detecting the presence of variant sequences Alternatively, where a polymorphism creates or destroys a recognition site for a restriction endonuclease, the fragment is digested with that endonuclease, and the products size fractionated to determine whether the fragment was digested Fractionation is performed by gel or capillary electrophoresis, particularly acrylamide or agarose gels
Hybridization with a polymorphism specific probe may also be used to determine its presence, by Southern blots, dot blots, efc The hybridization pattern of a control and variant sequence to an array of oligonucleotide probes immobilised on a solid support, as described in US 5,445,934, or in WO95/35505, may also be used as a means of detecting the presence of variant sequences
Screening for mutations in mitofusin may be based on the functional or antigenic characteristics of the protein Various immunoassays designed to detect polymorphisms in mitofusin proteins may be used in screening Where many diverse genetic mutations lead to a particular disease phenotype, functional protein assays have proven to be effective screening tools The activity of the encoded mitofusin protein may be determined by comparison with the wild-type protein
Antibodies specific for a mitofusin may be used in staining or in immunoassays Samples, as used herein, include biological fluids such as semen, blood, cerebrospmal fluid, tears, saliva, lymph, dialysis fluid and the like, organ or tissue culture derived fluids, and fluids extracted from physiological tissues Also included in the term are derivatives and fractions of such fluids The cells may be dissociated, in the case of solid tissues, or tissue sections may be analyzed Alternatively a lysate of the cells may be prepared
Diagnosis may be performed by a number of methods to determine the absence or presence or altered amounts of normal or abnormal mitofusin in patient cells For example detection may utilize staining of cells or histological sections, performed in accordance with conventional methods Cells are permeabilized to stain intra-mitochondnal and/or cytoplasmic molecules The antibodies of interest are added to the cell sample, and incubated for a period of time sufficient to allow binding to the epitope, usually at least about 10 minutes The antibody may be labeled with radioisotopes, enzymes, fluorescers, chemilummescers, or other labels for direct detection Alternatively, a second stage antibody or reagent is used to amplify the signal Such reagents are well known in the art For example, the primary antibody may be conjugated to biotm, with horseradish peroxidase-conjugated avid added as a second stage reagent Alternatively, the secondary antibody conjugated to a flourescent compound, e g flourescem, rhodamme, Texas red, etc Final detection uses a substrate that undergoes a color change in the presence of the peroxidase The absence or presence of antibody binding may be determined by various methods, including flow cytometry of dissociated cells, microscopy, radiography, scintillation counting, efc Determination of the subcellular localization of mitofusin through antibody binding may be used in mutation analysis to detect mutations that cause failure to express or properly localize mitofusin proteins The assay can be performed by immunohistochemistry or immunofluorescence, where the cell sample is stained with a mitofusin specific antibody followed by labeled secondary antibodies as described above to determine whether mitofusin is properly localized in the mitochondria Alternatively, cell lysates may be fractionated and the level of mitofusin in the mitochondrial fraction quantitated
Other diagnostic assays of interest are based on the functional properties of mitofusin proteins For example, a functional assay may be based on the membrane changes mediated by mitofusin gene products Other assays may, for example, detect conformational changes, or changes in the subcellular localization of mitofusin proteins
Modulation of Gene Expression The mitofusin genes, gene fragments, or the encoded protein or protein fragments are useful in gene therapy to treat degenerative and other disorders involving mitochondria, including myopathies and Alzheimer's disease Expression vectors may be used to introduce the mitofusin gene into a cell Such vectors generally have convenient restriction sites located near the promoter sequence to provide for the insertion of nucleic acid sequences Transcription cassettes may be prepared comprising a transcription initiation region, the target gene or fragment thereof, and a transcnptional termination region The transcription cassettes may be introduced into a variety of vectors, e g plasmid, retrovirus, e g lentivirus, adenovirus, and the like, where the vectors are able to transiently or stably be maintained in the cells, usually for a period of at least about one day, more usually for a period of at least about several days to several weeks
The gene or mitofusin protein may be introduced into tissues or host cells by any number of routes, including viral infection, microinjection, or fusion of vesicles or mitochondria Jet injection may also be used for intramuscular administration, as described by Furth ef al (1992) Anal Biochem 205 365-368 The DNA may be coated onto gold microparticles, and delivered intradermally by a particle bombardment device, or "gene gun" as described in the literature (see, for example, Tang ef al (1992) Nature 356 152-154), where gold microprojectiles are coated with the mitofusin or DNA, then bombarded into skin cells Antisense molecules can be used to down-regulate expression of mitofusin in cells The anti-sense reagent may be antisense oligonucleotides (ODN), particularly synthetic ODN having chemical modifications from native nucleic acids, or nucleic acid constructs that express such anti- sense molecules as RNA The antisense sequence is complementary to the mRNA of the targeted gene, and inhibits expression of the targeted gene products Antisense molecules inhibit gene expression through various mechanisms, e g by reducing the amount of mRNA available for translation, through activation of RNAse H, or stenc hindrance One or a combination of antisense molecules may be administered, where a combination may comprise multiple different sequences Antisense molecules may be produced by expression of all or a part of the target gene sequence in an appropriate vector, where the transcnptional initiation is oriented such that an antisense strand is produced as an RNA molecule Alternatively, the antisense molecule is a synthetic oligonucleotide Antisense oligonucleotides will generally be at least about 7, usually at least about 12, more usually at least about 20 nucleotides in length, and not more than about 500, usually not more than about 50, more usually not more than about 35 nucleotides in length, where the length is governed by efficiency of inhibition, specificity, including absence of cross-reactivity, and the like It has been found that short oligonucleotides, of from 7 to 8 bases in length, can be strong and selective inhibitors of gene expression (see Wagner ef al (1996) Nature Biotechnology 14 840-844)
A specific region or regions of the endogenous sense strand mRNA sequence is chosen to be complemented by the antisense sequence Selection of a specific sequence for the oligonucleotide may use an empirical method, where several candidate sequences are assayed for inhibition of expression of the target gene in an in vitro or animal model A combination of sequences may also be used, where several regions of the mRNA sequence are selected for antisense complementation
Antisense oligonucleotides may be chemically synthesized by methods known in the art (see Wagner ef al (1993) supra and Milligan ef al , supra ) Preferred oligonucleotides are chemically modified from the native phosphodiester structure, in order to increase their mtracellular stability and binding affinity A number of such modifications have been described in the literature, which alter the chemistry of the backbone, sugars or heterocyclic bases
Among useful changes in the backbone chemistry are phosphorothioates, phosphorodithioates, where both of the non-bridging oxygens are substituted with sulfur, phosphoroamidites, alkyl phosphotnesters and boranophosphates Achiral phosphate derivatives include 3'-0'-5'-S-phosphorothιoate, 3'-S-5'-0-phosphorothιoate, 3'-CH2-5'-0-phosphonate and 3'- NH-5'-0-phosphoroamιdate Peptide nucleic acids replace the entire ribose phosphodiester backbone with a peptide linkage Sugar modifications are also used to enhance stability and affinity The α-anomer of deoxyπbose may be used, where the base is inverted with respect to the natural β-anomer The 2'-OH of the ribose sugar may be altered to form 2'-0-methyl or 2'-0-allyl sugars, which provides resistance to degradation without comprising affinity Modification of the heterocyclic bases must maintain proper base pairing Some useful substitutions include deoxyundine for deoxythymidme, 5-methyl-2'-deoxycytιdιne and 5-bromo-2'-deoxycytιdιne for deoxycytidine 5- propynyl-2'-deoxyurιdιne and 5-propynyl-2'-deoxycytιdme have been shown to increase affinity and biological activity when substituted for deoxythymidme and deoxycytidine, respectively
As an alternative to anti-sense inhibitors, catalytic nucleic acid compounds, e g πbozymes, anti-sense conjugates, etc may be used to inhibit gene expression Ribozymes may be synthesized in vitro and administered to the patient, or may be encoded on an expression vector, from which the nbozyme is synthesized in the targeted cell (for example, see International patent application WO 9523225, and Beigelman et al (1995) Nucl Acids Res 234434-42) Examples of oligonucleotides with catalytic activity are described in WO 9506764 Conjugates of anti-sense ODN with a metal complex, e g terpyrιdylCu(ll), capable of mediating mRNA hydrolysis are described in Bashkin ef al (1995) Appl Biochem Biotechnol 54 43-56
Cell and Animal Models The subject nucleic acids can be used to generate genetically modified non-human animals or site specific gene modifications in cell lines, for the study of mitofusin function or regulation, or to create animal models of diseases, including mitochondrial diseases, muscle myopathies, neurodegenerative disorders, and aging The term "transgenic" is intended to encompass genetically modified animals having a deletion or other knock-out of mitofusin gene activity, having an exogenous mitofusin gene that is stably transmitted in the host cells where the gene may be altered in sequence to produce a modified protein, or having an exogenous mitofusin promoter operably linked to a reporter gene Transgenic animals may be made through homologous recombination, where the mitofusin locus is altered Alternatively, a nucleic acid construct is randomly integrated into the genome Vectors for stable integration include plasmids, retroviruses and other animal viruses, YACs, and the like Of interest are transgenic mammals, e g cows, pigs, goats, horses, efc , and particularly rodents, e g rats, mice, efc
Investigation of genetic function may also utilize non-mammalian models, particularly using those organisms that are biologically and genetically well-characterized, such as C elegans, D melanogaster and S cerevisiae The subject gene sequences may be used to knock-out corresponding gene function or to complement defined genetic lesions in order to determine the physiological and biochemical pathways involved in mitofusin function Drug screening may be performed in combination with complementation or knock-out studies, e g to study progression of degenerative disease, to test therapies, or for drug discovery
The modified cells or animals are useful in the study of mitofusin function and regulation For example, a series of small deletions and/or substitutions may be made in the mitofusin gene to determine the role of different domains in GTPase activity, membrane fusion, efc Specific constructs of interest include, but are not limited to, anti-sense mitofusin constructs to block mitofusin expression, expression of dominant negative mitofusin mutations, and over-expression of a mitofusin gene One may also provide for expression of the mitofusin gene or variants thereof in cells or tissues where it is not normally expressed or at abnormal times of development In addition, by providing expression of mitofusin protein in cells in which it is otherwise not normally produced, one can induce changes in mitochondrial behavior DNA constructs for homologous recombination will comprise at least a portion of the mitofusin gene with the desired genetic modification, and will include regions of homology to the target locus DNA constructs for random integration need not include regions of homology to mediate recombination Conveniently, markers for positive and negative selection are included Methods for generating cells having targeted gene modifications through homologous recombination are known in the art For various techniques for transfectmg mammalian cells, see Keown ef al (1990) Methods in Enzvmoloov 185:527-537 For embryonic stem (ES) cells, an ES cell line may be employed, or embryonic cells may be obtained freshly from a host, e g mouse, rat, guinea pig, efc Such cells are grown on an appropriate fibroblast-feeder layer or grown in the presence of appropriate growth factors, such as leukemia inhibiting factor (LIF) When ES cells have been transformed, they may be used to produce transgenic animals After transformation, the cells are plated onto a feeder layer in an appropriate medium Cells containing the construct may be detected by employing a selective medium After sufficient time for colonies to grow, they are picked and analyzed for the occurrence of homologous recombination or integration of the construct Those colonies that are positive may then be used for embryo manipulation and blastocyst injection Blastocysts are obtained from 4 to 6 week old superovulated females The ES cells are trypsmized, and the modified cells are injected into the blastocoel of the blastocyst After injection, the blastocysts are returned to each uterine horn of pseudopregnant females Females are then allowed to go to term and the resulting chimeπc animals screened for cells bearing the construct By providing for a different phenotype of the blastocyst and the ES cells, chimeπc progeny can be readily detected
The chimeπc animals are screened for the presence of the modified gene and males and females having the modification are mated to produce homozygous progeny If the gene alterations cause lethality at some point in development, tissues or organs can be maintained as allogeneic or congenic grafts or transplants, or in in vitro culture The transgenic animals may be any non-human mammal, such as laboratory animals, domestic animals, etc The transgenic animals may be used in functional studies, drug screening, etc , e g to determine the effect of a candidate drug on mitochondrial fusion, to test potential therapeutics or treatment regimens, efc
Drug Screening Assays By providing for the production of large amounts of mitofusin protein, one can identify ligands or substrates that bind to, modulate or mimic the action of mitofusin Drug screening identifies agents that provide a replacement or enhancement for mitofusin function in affected cells A wide variety of assays may be used for this purpose, including labeled in vitro protein-protein binding assays, prote -DNA binding assays, protein-mitochondria or protein mitochindna fraction, electrophoretic mobility shift assays, immunoassays for protein binding, and the like All or a fragment of the purified protein may also be used for determination of three-dimensional crystal structure, which can be used for modeling intermolecular interactions, membrane fusion, efc Altered mitofusin molecules, either as an isolated recombinant protein, or as a genetically modified cell or animal model may be assayed to investigate structure function parameters, including but not limited to potential mitochondrial import sequences, protein interaction domains, GTPase motifs, and dominant negative acting forms of the protein
The term "agent" as used herein describes any molecule, e g protein or pharmaceutical, with the capability of altering or mimicking the physiological function of a subject mitofusin Generally a plurality of assay mixtures are run in parallel with different agent concentrations to obtain a differential response to the various concentrations Typically, one of these concentrations serves as a negative control, / e at zero concentration or below the level of detection
Candidate agents encompass numerous chemical classes, though typically they are organic molecules, preferably small organic compounds having a molecular weight of more than 50 and less than about 2,500 daltons Candidate agents comprise functional groups necessary for structural interaction with proteins, particularly hydrogen bonding, and typically include at least an amme, carbonyl, hydroxyl or carboxyl group, preferably at least two of the functional chemical groups The candidate agents often comprise cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more of the above functional groups Candidate agents are also found among biomolecules including, but not limited to peptides, sacchandes, fatty acids, steroids, puπnes, pyrimidines, derivatives, structural analogs or combinations thereof Candidate agents are obtained from a wide variety of sources including libraries of synthetic or natural compounds For example, numerous means are available for random and directed synthesis of a wide variety of organic compounds and biomolecules, including expression of randomized oligonucleotides and oligopeptides Alternatively, libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available or readily produced Additionally, natural or synthetically produced libraries and compounds are readily modified through conventional chemical, physical and biochemical means, and may be used to produce combinatorial libraries Known pharmacological agents may be subjected to directed or random chemical modifications, such as acylation, alkylation, estenfication, amidification, efc to produce structural analogs
Where the screening assay is a binding assay, one or more of the molecules may be joined to a label, where the label can directly or indirectly provide a detectable signal Various labels include radioisotopes, fluorescers, chemilummescers, enzymes, specific binding molecules, particles, e g magnetic particles, and the like Specific binding molecules include pairs, such as biotm and streptavidm, digoxm and antidigoxm etc For the specific binding members, the complementary member would normally be labeled with a molecule that provides for detection, in accordance with known procedures
A variety of other reagents may be included in the screening assay These include reagents like salts, neutral proteins, e g albumin, detergents, etc that are used to facilitate optimal protein- protein binding and/or reduce non-specific or background interactions Reagents that improve the efficiency of the assay, such as protease inhibitors, nuclease inhibitors, anti-microbial agents, etc may be used The mixture of components are added in any order that provides for the requisite binding Incubations are performed at any suitable temperature, typically between 4 and 40 °C Incubation periods are selected for optimum activity, but may also be optimized to facilitate rapid high-throughput screening
EXPERIMENTAL The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the subject invention, and are not intended to limit the scope of what is regarded as the invention Efforts have been made to ensure accuracy with respect to the numbers used (e g amounts, temperature, concentrations, etc ) but some experimental errors and deviations should be allowed for Unless otherwise indicated, parts are parts by weight, molecular weight is average molecular weight, temperature is in degrees centigrade, and pressure is at or near atmospheric
Experimental Procedures
Fly Strains and Culture Flies were grown on standard cornmeal molasses agar media at 25°C All visible markers and balancer chromosomes were as described in Flybase (1996) Nucleic Acids Res 24, 53-56 (flybase bio Indiana edu) unless otherwise noted
The fzo1 allele was isolated in a screen for ethyl methanesulfonate (EMS) induced recessive male sterile mutations The fzo 2 allele was isolated in an EMS screen essentially as described in Lin ef al (1996) Devel 122 1331-1341, except that mutagenized chromosomes were tested for failure to complement fzo 1 One new allele, fzo2, was identified from 1799 mutagenized third chromosomes screened
The deletions Df(3R)M95A (94D, 95A3) and Df(3R)EB6 (94C2-5, 94E3, Mohler ef al (1995) Devel 121, 237-247) were gifts from G Reuter and J Mohler, respectively The Df(3R)P20 deletion was generated by mobilizing a lethal ry + marked P element in 94E3-5 associated with the cnco3B7i a||e|e as f0||ows plies transheterozygous for cnc 037B1 and a TM3, Sb chromosome carrying the Δ2-3 transposase insert (Reuter ef al (1993) Dros Inf Serv 72, 78) were crossed to ry e bar3 tx2TM3, Sb flies F1 cnc03781'2ry e bar3 tx males which had lost the P(ry+) and therefore had ry eyes were crossed to e fzo V TM3, Ser females F2 cnc03781'2e fzo1 males were tested for the fzo phenotype, and stocks of new alleles were recovered by crossing together male and virgin female cnc03781'2TM3, Ser F2 siblings From among approximately 200 ry chromosomes tested, two deletions failing to complement fzo ' were identified Molecular breakpoints of the smaller deletion, Df(3R)P20 were mapped by Southern hybridization analysis using standard methods Df(3R)P20 failed to complement mutations in cnc and was therefore not homozygous viable
Fertility was determined by placing individual flies in yeasted vials with three males or virgin females as appropriate and scoring the presence or absence of larvae after seven days at 25°C Progeny of tested females were counted At least ten individual males and four individual females for each of the allelic combinations fzo 12fzo fzo1 /fzo2, fzo1/Df(3R)P20, and fzo2/Df(3R)P20, as well as heterozygous sibling classes fzo/TM3 or Df(3R)P20/TM3 were tested (The fzo 2 chromosome carried a secondary lethal and could not be made homozygous )
The effects of fzo1 and fzo2 mutations on viability were tested by crossing five individual females of each of the above allelic combinations to Df(3R)P202 TM3 males at 25°C and comparing 5 numbers of fzo/Df progeny to total progeny Progeny were collected for nine days after eclosion began
Light and Electron Microscopy For light microscopy, testes from the above genotypes as well as from wild type Oregon R flies were dissected in TB1 buffer (7 mM K 2HPO 7 mM KH2PO„ [pH 6 7], 80 mM KCI, 16mM NaCI, 5mM MgCI 2, 1% PEG-6000), opened with forceps to allow
10 spillage of contents, placed under cover slips, and examined immediately by phase contrast microscopy with a Zeiss Axiophot microscope To check for a mitochondrial membrane potential, 10 μg/ml rhodamme 123 was included in the dissection buffer and samples examined under epifluorescence
For ultrastructural analysis by transmission electron microscopy, testes were dissected in
15 TB1 and immediately placed in fixative (2% glutaraldehyde, 1% paraformaldehyde, 0 1M sodium phosphate or sodium cacodylate buffer [pH 7]) After overnight fixation, samples were washed in 0 1% phosphate or cacodylate buffer for 15 minutes and stained with 1% osmium tetroxide in the same buffer for two hours Testes were washed three times in water, stained for 1 hour in 1% uranyl acetate, washed three times in water and dehydrated through an ethanol series (30%, 50%
20 70%, 95%, 100%) After five minutes in 1 1 ethanol propylene oxide and five minutes in propylene oxide, samples were embedded in Spurr's resin and polymerized overnight at 60°C Thin sections (80-90 nm) were cut with a Reichert-Jung microtome, placed on Formvar-coated slot grids and examined on a Phillips 410 transmission electron microscope
Recombination Mapping For initial meiotic mapping, females heterozygous for an e fzo1 25 chromosome and a ru h th stp" cu sr ca chromosome were crossed to ru h th st " cu sr Bsb/TM3 or ru h th st cu e Pn ca/TM3 males Male progeny with markers indicating single recombination events were selected for each interval and individually crossed to three e fzo 1/TM3 females to score fzo and make stocks of the recombinant chromosomes with TM3 The fzo 1 mutation mapped between e and ca, roughly 9 cM distal to e 30 To localize fzo 1 further, p" e fzo1/hhbar3 tx females were crossed to e hh bar3 tx/TM3 males and recombinant male progeny crossed individually to e fzo 1/TM3 females to score fzo and make stocks with TM3 fzo1 mapped 0 58±0 17 cM distal to hh bar3 (Figure 4), based on 227 recombmants between hhbar3 and fx
RFLP Mapping The fzo 1 mutation was further localized by recombination with respect to 35 restriction fragment length polymorphisms (RFLPs) in the interval between hh fiaf3and a ry* marked P element insertion in the pointed gene associated with the pnt 07β25 allele Briefly, females transheterozygous for ry506 hhbar3 fzo1 (parental chromosome 1) and ry^pnf7825 (parental chromosome 2) were crossed to ry506e hhbar3tx/TM3 males Recombinant F1 Sb + progeny that were phenotypically either hh bar3 ry* or hhbar3* ryappeared at a frequency of 31 per 2995 flies, indicating 5 that hhba'3 and pnt07825 were 1 04+/-0 18 cM apart Males representing each of these two reciprocal recombinant classes were crossed individually to e fzo 1/TM3 females to score fzo on the recombinant chromosomes and to construct stocks with TM3 The fzo 1 mutation mapped in the center of the interval, 0 5 cM distal to hh b Λ3and 0 5 cM proximal to pnt07825
RFLPs between the two parental chromosomes were identified using Southern blot
10 analysis Standard molecular biology techniques were used throughout this work, and DNA fragments were purified with a QIAquick kit (QIAGEN) as per the manufacturer's instructions Genomic DNA was isolated from flies homozygous for parental chromosome 1 as well as from flies transheterozygous for parental chromosomes 1 and 2 (the P element insertion on parental chromosome 2 was a recessive lethal) The parental genomic DNAs were digested separately with
15 34 standard four-, five-, and six-cutter restriction enzymes, electrophoresed, blotted, and hybridized with several different radiolabeled DNA fragments from the distal third of a 90 kb genomic walk (Mohler et al (1991) Mech Devel 34, 3-10) At least one RFLP between the two parental chromosomes was detected for each probe Genomic DNA was isolated from 35 recombinant stocks (either from homozygotes or from flies transheterozygous for the recombinant chromosome
20 and parental chromosome 1) These DNAs were digested with the restriction enzymes previously shown to have polymorphic sites in the region, blotted, and probed with the appropriate fragments Analysis of linkage between these molecular markers and fzo 1 defined the region of the fzo 1 mutation to be between a polymorphic Xbal site at +11 and a polymorphic EcoRI site at +22 on the genomic walk (Mohler et al , 1991 ) The Xbal and EcoRI RFLPs segregated from fzo 1 in one and
25 two of the 35 recombinants, respectively
Isolation of cDNAs and Sequence Analysis Radiolabeled EcoRI restriction fragments corresponding to +5 to +22 on the genomic walk, were used to probe a λ-ZAP (Stratagene) testis cDNA library Eighteen positive cDNA clones of various lengths were shown by Southern hybridization analysis to be from the same transcription unit The seven longest cDNAs were 2 4 kb
30 and had identical restriction maps One of these was mapped to genomic DNA by Southern hybridization and, consistent with restriction mapping analysis, seemed to have no detectable introns This cDNA was sequenced on both strands by the dideoxy chain termination method using the ABI PRISM dye terminator cycle sequencing system (Perkin-Elmer) T3 or T7 primers were used on the intact cDNA or dropout subclones, and 19- or 20-mer oligonucleotides were
35 synthesized to prime sequencing runs through any remaining gaps Representative members of each shorter cDNA class were restriction mapped, partially sequenced, and shown to be truncated versions of the longer class The cDNA (2399 bp) was conceptually translated with DNA Stnder software and the Fzo predicted protein sequence compared using the BLAST program to nucleotide sequences in GenBank and dbEST translated in all reading frames Significant homology was detected to predicted proteins from H sapiens, M musculus, C elegans, and S cerevisiae The human cDNA clone from which a human fetal brain EST (GenBank T06373) was derived was obtained from American Type Culture Collection and sequenced on one strand Predicted proteins encoded by Fzo homologs were themselves subjected to homology searches as above Alignments were done with the help of the CLUSTALW program using DNAstar software
Analysis of the Fzo predicted protein with the BLOCKS program indicated similarity to the dynamm family in the vicinity of the P-loop, a motif found in nucleotide-bindmg proteins and designated as the G1 GTPase motif (Bourne ef al . (1991 ) Nature 349. 117-127) The sequences of Fzo and homologs were analyzed for predicted transmembrane domains and regions likely to form coiled coils with the TMpredict program and the COILS program, respectively The Compute pl/Mw program on the ExPASy Molecular Biology Server was used to calculate predicted isoelectnc points
P element Mediated Germlme Transformation A 3 kb genomic fragment was isolated from an Xhol/Xbal digestion of phage D14 DNA (which corresponds to +3 to +16 on the genomic walk) and subcloned into the w +-marked P element transformation vector pCaSpeR4 (Thummel and Pirrotta (1992) Dros Inf Serv 71, 150) to make plasmid pKH2 The 2 4 kb EcoRI restriction fragment from phage D14 had previously been subcloned into pUC9 to make construct D5-19, a 1 kb Xbal/EcoRI fragment was isolated from a digestion of D5-19 and subcloned into pKH2 to make pKH3fzo+ The resulting 4 kb insert in the pKH3fzo + rescue construct contained the genomic region of the full length fzo cDNA plus approximately 1 kb 5' and 500 base pairs 3'
The pKH3fzo+ plasmid and a plasmid encoding the Δ 2-3 constitutively active transposase were mixed in a 3 1 molar ratio to a final DNA concentration of 0 4 mg/ml and injected into y w 6 or w1118 embryos Three independent fzo insertions, one on the second chromosome and two on the third, were isolated from the progeny of 32 fertile injectees An additional insertion on the second chromosome was obtained by mobilizing one of the third chromosome insertions with the TM3, Sb Δ 2-3 transposase source To test for rescue, all four independent fzo nsertions were crossed into w, fzo mutant backgrounds by independent assortment for the second chromosome insertions and by recombination onto a hh bar3 fzo1 chromosome and then independent assortment for the third chromosome insertions, which were tested for rescue only of allelic combinations with fzo '
Site-directed Oligomutagenesis and Retransformation with Mutated fzo Transgenes To introduce mutations into the Fzo predicted GTP-bindmg domain, a 1 8 kb Xba l/BamHI restriction fragment (representing the first two thirds of the fzo coding region) from the pKH3fzo + germlme transformation construct (see above) was subcloned into Bluescnpt SK- Using standard methods, mutagenesis was performed with oligonucleotides of the sequences [SEQ ID NO 5] 5' ACCTCAAATGGAACTAGTGCCGTGATC 3' and [SEQ ID NO 6] 5' TACTCAACAATCTATGGGATAAG 3' The former exchanged AA for CT at nucleotides corresponding to those at positions 369-370 in the fzo cDNA, introducing a Spel restriction site and changing the encoded ammo acid at position 99 from a lysine to a threonme The latter replaced a 5 G with a T at nucleotide 819, eliminating a Clal restriction site and changing the encoded ammo acid at position 249 from an argmine to a leucine Mutagenized constructs were selected by virtue of the above mentioned altered restriction sites For each mutagenized construct, a 1 8 kb Xba l/BamHI restriction fragment was subcloned back into the 10 kb Xba l/BamHI vector fragment from plasmid pKH3fzo+ to create two new germlme transformation constructs, pKH3fzo K99T and pKH3fzoR249L,
10 which were injected into fly embryos as described above For both these constructs, the Xbal and BamHI restriction sites used for the final subclonmg were regenerated by the hgations In addition, the reading frame remained unchanged at the mutagenesis and subclonmg sites, as shown by detection of mutant proteins with anti-Fzo 605"718 antibodies, which recognize only epitopes encoded by regions 3' to the sites of ligation and mutagenesis
15 To test for rescue of the mutant phenotype by the fzo K99T and fzoR249L mutant transgenes, respectively, appropriate crosses were made to introduce separately the eight and five independent second chromosome transgene insertions into fzo mutant backgrounds To test for any dominant effect, appropriate crosses were made to obtain males with one wild type copy of fzo and as many as four (fzoK99T) or six (fzoR249L) different copies of the mutated transgenes
20 RNA in situ Hybridization For in situ hybridization to testes, digoxygenm-labeled RNA probes representing both the fzo cDNA sense and anti-sense strands (primed with T7 or T3 primers on a linearized cDNA construct) were made and submitted to alkaline hydrolysis (1 hour) as described in the Genius 4 RNA Labeling Kit user's guide (Boehnnger Mannheim) Paraformaldehyde fixation of freshly dissected testes was as in Tautz (1989) Chromosoma 98, 81-
25 85 except no heptane or methanol was used Subsequent treatment was as in Gonzalez (1994) Techniques for studying mitosis in Drosophila In The cell cycle A practical approach, R Brookes and P Fantes, eds IRL, Oxford University Press), pp 143-175, except 50 μg/ml hepaπn was included in the hybridization solution (HS), hybridization and early washes were at 65 °C, and the secondary antibody was preadsorbed to Drosophila embryos and diluted 1 2000 Mounted
30 preparations were examined under Nomarski optics with a Zeiss Axiophot microscope
Generation ofanti-Fzo antibodies Antibodies were raised to a fusion protein containing a 6-hιstιdιne tag and the C-terminal 115 ammo acids of Fzo To make the expression construct, a 0 5 kb BamHI/Hindlll restriction fragment from fzo cDNA1 (the Hmdlll site is from the Bluescript SK- multiple cloning site) was subcloned into vector pQE30 (QIAGEN) The fusion protein was induced, 35 harvested under denaturing conditions, and purified on a Ni-NTA column with imidazole elution as described in the QIAexpressionist manual (QIAGEN) Aliquots from each column fraction were electrophoresed on an SDS-polyacrylamide gel and stained with Coomassie Blue Fractions with greatest purity and highest concentration of the fusion protein were dialyzed in 1 5 M urea, 0 1 M Na phosphate, 0 01 M Tπs-HCI, and 500 mM NaCI, pH 7 The fusion protein formed a precipitate at urea concentrations below 4 M After dialysis, precipitated protein was emulsified in complete 5 Freund's adjuvant and injected into rabbits, standard schedules were followed for booster injections and serum collections (Berkeley Antibody Company, Richmond CA) Approximately 500 mg and 250 mg of the fusion protein were used for initial and booster injections, respectively
Western Blots Protein extracts were electrophoresed in 10% SDS-polyacrylamide gels and transferred to nitrocellulose filters in methanol buffer using standard methods Testis extracts were
10 prepared by placing freshly dissected testes into a solution containing 8M urea, 0 1M sodium phosphate and 0 01 M Tns-CI, pH 7 and mixing thoroughly until all proteins had dissolved One volume of SDS-PAGE sample buffer was added, and the samples were then boiled for 15 minutes and spun at top speed in a microcentπfuge for ten minutes Eight testes' worth of extract supernatant was loaded in each lane Whole fly extracts were prepared by homogenizing flies in
15 the above urea solution, with further treatment as above One fifth of a fly's worth of extract was loaded per lane
Filters were blocked in Blotto with 0 1% Tween 20 and incubated for two hours at room temperature in either preimmune or anti-Fzo 60 71B serum (fourth bleed) diluted 1 1200, or mouse monoclonal anti-actm antibody (Amersham) diluted 1 100, in the blocking solution Subsequent
20 washes, incubation with horseradish peroxidase-conjugated secondary antibodies, and detection were performed as described in the ECL Western Blotting manual (Amersham) The secondary antibodies were diluted 1 7000 (anti-rabbit IgG) or 1 2000 (anti-mouse IgG)
Immunofluorescence Fly testes were prepared for immunofluorescence staining as described in Hime ( 1996) J Cell Sci 709, 2779-2788 The resulting slides were incubated
25 overnight at 4°C in preimmune or anti-Fzo 6tM 718 serum (fourth bleed) diluted 1 150 in PBTB (phosphate-buffered saline with 0 1% Triton X-100 and 3% bovine serum albumin), washed four times at room temperature in PBTB, and incubated for one hour at 37°C in FITC-conjugated anti- rabbit IgG (Jackson Labs) diluted 1 200 in PBTB (0 5 mg/ml RNAse was included for samples to be later stained with propidium iodide) Slides were washed 4x10 minutes in PBTB and mounted in
30 85% glycerol, 2 5% N-propyl gallate For some samples, 1 μg/ml DAPI (Sigma) was included in the second PBTB wash after incubation in secondary antibody, for others, 1 μg/ml propidium iodide was included in the mounting medium Samples were examined using epifluorescence on a Zeiss Axiophot microscope, images were collected with a Photometries cooled CCD camera (courtesy of Bruce Baker) Emissions from different fluorochromes on the same sample were collected
35 separately and overlaid using Adobe Photoshop Wild type function of fzo is required for developmental^ regulated fusion of mitochondria during Drosophila spermatogenesis Mutations in fzo cause male sterility associated with defects in mitochondrial fusion during Nebenkern formation in post-meiotic early round spermatids In wild type testes, mitochondria in each haploid spermatid aggregate beside the nucleus and fuse into a Nebenkern, a phase-dark spherical structure consisting of two interwrapped mitochondrial derivatives as viewed by TEM In fzo mutant males, mitochondria aggregate in early haploid spermatids, forming somewhat misshapen Nebenkerns as viewed by phase contrast microscopy, but fail to fuse into two giant mitochondria Instead, many smaller mitochondria appear to wrap around each other at the onion stage as viewed by TEM Despite the prior defects in fusion, mitochondria unfurl and elongate in fzo mutants At the early elongation stage, unfurling mitochondria in fzo mutants appear fragmented compared to wild type as seen by phase contrast microscopy At the late elongation stage, two elongating mitochondrial derivatives per spermatid are seen in wild type In contrast, many elongating mitochondria are associated with each haploid nucleus in fzo Cross sections through wild type elongating spermatids viewed by TEM reveal two mitochondrial derivatives associated with each axoneme, the major derivative contains electron dense paracrystalline material while the minor does not Each axoneme in fzo mutant males is associated with a large number of mitochondrial derivatives, with roughly half containing the paracrystalline material characteristic of major derivatives Thus the defects in fzo mutant spermatids appear specific for mitochondrial fusion, as mitochondrial aggregation, membrane wrapping, and elongation all occur (although the unfused mitochondria do not elongate the full axonemal length because they lack sufficient membrane material) In addition, spermatid mitochondria in fzo mutants take up the dye rhodamme 123 in amounts similar to wild type, indicating presence of a membrane potential and suggesting that fzo mutations probably do not grossly affect respiration Two EMS-mduced alleles, fzo 1 and fzo2, were characterized as described above fzo1/fzo1, fzo '/fzo2, fzo1/Df(3R)P20, and fzo2/Df(3R)P20 flies showed identical phenotypes, suggesting that both mutations are strong loss of function alleles The severity of the phenotype was consistent among all spermatids in all testes observed The fzo 1 and fzo2 mutations did not noticeably affect female fertility or overall viability of the animal
fzo encodes a novel conserved predicted transmembrane GTPase The fzo ' mutation was mapped by recombination to 0 5 cM distal of hh bar3 and 0 5 cM proximal of pnt07825, in agreement with complementation of fzo 1 by Df(3R)EB6 but not by Df(3R)M95A (Figure 3A) The fzo1 mutation was further localized by RFLP mapping to an 11 kb region defined by polymorphic Xbal and EcoRI sites on an existing genomic phage walk (Figures 2A, B) In addition, a 12 kb deficiency (Df(3R)P20) generated by imprecise excision of a nearby P element (cnc °3871) failed to complement fzo mutations, consistent with the RFLP mapping data cDNA clones corresponding to transcripts from the fzo region were isolated from a testis cDNA library All eighteen positive clones from the region represented the same transcription unit, based on Southern blot hybridization The largest cDNA clones were 2 4 kilobases (kb) long and had identical restriction maps One of these was chosen for further analysis and mapped by Southern blot hybridization to cloned genomic DNA in the fzo region
To confirm that this transcription unit corresponded to fzo, we introduced a 4 kb genomic fragment containing the candidate locus plus 1 kb of sequence upstream and approximately 500 bp of sequence downstream of the cDNA into flies by P element mediated germlme transformation A single copy of any of four independent transgene insertions fully restored fertility and normal mitochondrial morphogenesis to fzo 1/fzo 1, fzo1/Df(3R)P20, and fzo '/fzo2 males (and also fzo2/Df(3R)P20 males, for the second chromosome insertions)
The representative fzo cDNA was sequenced fully on both strands and shown to have a complete open reading frame (ORF) with flanking AT-nch sequences and a single consensus translational start site The fzo transcription unit encodes a predicted protein of 718 ammo acids Database searches with the ammo acid sequence identified ORFs that encode related predicted proteins in mammals, C elegans, and S cerevisiae Human expressed sequence tags (ESTs) from brain (T06373, R20140, T37724), heart (AA248162, AA248083), fibroblasts (W49736), liver/spleen (H58349, AA010217), and pancreas (AA155601) encode predicted peptides homologous to Fzo The cDNA clone from which a human fetal brain EST (T06373) was derived was sequenced and shown to encode an incomplete predicted protein (lacking its N terminus) with 35% identity to Fzo The heart, fibroblast, and other brain ESTs are virtually identical to regions of this cDNA, while one of the liver/spleen clones (H58349) and the pancreas EST (AA155601) seem to originate from a different gene and encode a 125 residue peptide 56% identical to the C terminus of the human fetal brain gene product The two human isoforms are both 28% identical to Fzo in this C-terminal region Four mouse ESTs (W41601 , AA199015, AA212845, AA052806) together encode 211 ammo acids with 22% identity to the Fzo C terminus and 84% identity to the analogous region in the sequenced human brain homolog ORFs from C elegans (U29244, ORF 14) and S cerevisiae (Z36048) encode complete predicted proteins with 28% and 19% identity to Fzo, respectively (Figure 4B-D) The S cerevisiae predicted protein is itself 24% identical to a partial predicted protein from the thermophilic bacterium Caldocellum saccharolyticum (L18965 ORF 6), which is 11% identical to Fzo
The region of highest homology between the Fzo, human fetal brain (50% identity to Fzo), C elegans (49%), and S cerevisiae (20%) predicted proteins is a 186 ammo acid region containing four completely conserved signature motifs found in virtually all GTPases (G1-G4 in Figure 2A-C) The C saccharolyticum predicted protein also contains these motifs Outside the individual motifs there is no significant similarity to any known GTPase However, the spacing between the GTPase motifs, their N-termmal placement in Fzo and homologs, and the overall predicted protein size are reminiscent of the dynamm family (Warnock and Schmid, 1996 BioEssays 18, 885-893) The G2 motif (Bourne ef al , 1991 Nature 349, 117-127) consists only of a conserved threonme and has not been defined in dynamms Both the Fzo and dynamin families have a conserved threonme exactly 20 residues beyond the G1 motif that we propose represents G2 Outside the GTPase domain, Fzo and its homologs have moderate homology at the ammo acid level (30%, 21%, and 19% identity beween Fzo and the human fetal brain, C elegans, and S cerevisiae homologs, respectively) and share several predicted structural features All have a predicted transmembrane domain near the C terminus embedded in a large (-35 ammo acids) uncharged region interrupted by one to three basic residues All the homologs have predicted coiled coil regions flanking the predicted transmembrane domain, though the C terminal coiled coil probability in the S cerevisiae homolog is lower than in other homologs The C elegans and S cerevisiae homologs have strongly predicted coiled coil regions near their N termini The analogous region in Fzo has a lower coiled coil probability (023) but is strikingly similar to the C elegans homolog in its α helical projection, showing clear acidic, hydrophobic, and basic/polar faces All four homologs are acidic overall between the N terminus and the transmembrane domain, with predicted isoelectπc points (pi) near 5, and are basic in the C terminal tail, with predicted pis near 9 (Figure 4H)
fzo mRNA is expressed in primary spermatocytes The fzo message was highly expressed in pre-meiotic primary spermatocytes during their growth period but was not detectable at earlier stages in germlme stem cells or mitotically dividing spermatogonia The transcript persisted in meiotic cells but was not detectable in differentiating spermatids The transcript was present in both fzo '/fzo1 and fzo2/Df(3R)P20 testes A control sense-strand fzo RNA probe did not hybridize in cells of any stage
Anti-Fzo604 71β antibodies detect greatly reduced levels of Fzo in fzo 1 and fzo2 mutant fesfes Antibodies raised against a fusion protein containing the C-terminal 115 residues of Fzo detected a wide doublet in Western blots of wild type testis extracts The Fzo protein was partially resistant to extraction and denaturation and under standard denaturing conditions ran predominantly in streaky globs at an apparent molecular weight of 67 kD When subjected to additional boiling and denaturants, some of the protein shifted up to a smoother band at an apparent molecular weight of 82 kD, the predicted size for Fzo The wide Fzo doublet was greatly reduced in fzo* and fzo2/Df(3R)P20 mutant testes and increased in testes from wild type flies carrying six copies of the fzo + transgene
Very low levels of Fzo or a cross-reacting protein with the same mobility pattern were apparent in extracts from whole adult agametic males, indicating expression in somatic tissues Similar low levels of this protein were seen in agametic females, ovaries, and embryos of all stages and were unaffected in fzo 2/Df(3R)P20 whole male extracts However, in fzo 1/fzo1 males, the Fzo protein is highly expressed in somatic tissues despite its downregulation in fzo 1/fzo1 testes The fzo1 mutation therefore appears to affect tissue specific expression of Fzo Extracts from whole wild type males carrying six copies of the fzo + transgene showed more Fzo protein than agametic males but considerably less than fzo 1/fzo1 males Fzo may normally be expressed weakly in somatic tissues, it is therefore possible that Fzo is required elsewhere in addition to the testis and that the fzo1 and fzo2 alleles cause loss of function in the male germlme only Alternatively, the weakly-expressed somatic protein could be a Fzo isoform or an unrelated cross- reacting protein
The Fzo protein is associated with mitochondria in early spermatids during a short time period that spans mitochondrial fusion The anti-Fzo 604 718 antiserum stained onion stage
Nebenkems brightly in wild type spermatids but was undetectable or present at greatly reduced levels in fzo '/fzo1 or fzo2/Df(3R)P20 testes, respectively A wild type fzo transgene in a fzo 1/fzo1 mutant background restored detectable Fzo protein to the Nebenkern
The Fzo protein was associated with mitochondria in wild type spermatids during a narrow developmental window corresponding to the time that Fzo function is required
Mitochondria align on the spindle equator throughout meiotic divisions, however, the Fzo protein was not detected associated with mitochondria until the end of meiosis, during telophase II In post-meiotic haploid spermatids, Fzo was associated with aggregating mitochondria and was detected at highest levels associated with onion stage Nebenkems The Fzo protein was detected at lower levels associated with early elongation-stage mitochondrial derivatives and was not detected associated with more elongated mitochondria In testes from flies carrying multiple different copies of the fzo Transgene, the anti-Fzo antibody stained mitochondria much more brightly but with a similar time course to wild type Preimmune serum did not stain wild type testes
Conserved residues in the GTPase domain are required for Fzo function but not for targeting of the protein to mitochondria Mediation of mitochondrial fusion by Fzo appears to require the predicted GTPase activity Missense mutations in the fzo transgene that alter conserved residues (lysιne-99 and argιnιne-249) required in other GTPases for guanme nucleotide binding were introduced into the fzo transgene construct For the fzo K99T and fzoR249L transgenes, respectively, none of the eight and five independent insertions on the second chromosome (in one copy or two, for those that were viable as homozygotes) was able to restore fertility or sperm motility to fzo 1/fzo1, fzo1/Df(3R)P20, or fzo2/Df(3R)P20 mutant males In two copies, the fzoK99T transgene did not detectably improve the subcellular mutant phenotype, while the fzoR249L transgene appeared to allow some mitochondrial fusion In a wild type background, neither mutant transgene impaired fertility or mitochondrial fusion Fzo K99T and Fzo"*2491- mutant proteins were properly localized to spermatid mitochondria with wild type timing of appearance and disappearance
Fzo is a novel predicted GTPase The Drosophila fzo gene encodes a conserved predicted GTPase required for mitochondrial fusion during spermatogenesis and associated with mitochondria only during a short time spanning the fusion event Fzo contains four motifs common to virtually all known GTPases and conserved among Fzo homologs from mammals to yeast Fzo is the first protein to be assigned a function in this new family of large predicted transmembrane GTPases Within this family, the GTPase domain placement, motif spacing, and protein size (but not overall sequence) suggest a possible distant relationship with dynamms, which play a central biomechanical role in endocytic membrane trafficking
Mutations predicted by analogy to dimmish guanme nucleotide binding did not affect localization of Fzo to mitochondria but eliminated or reduced its ability to mediate mitochondrial fusion The fzoK99T mutation, predicted to disallow key hydrogen bonds with the GTP β and y phosphates, caused a severe loss of function phenotype In contrast, the fzo R249L mutation appeared to allow some mitochondrial fusion to occur, though not enough to restore normal sperm morphology or motility The Fzo argιnιne-249 is part of the G4 motif and is predicted to contact the ribose moiety of GTP Nearly all known GTPases have a lysme at positions analogous to FzoR249, perhaps the conservative change to argmine in Fzo and its higher eukaryotic homologs reflects somewhat decreased importance of this residue for stable nucleotide binding Mutations analogous to fzo R249L in H-ras reduce but do not eliminate GTP binding It is possible that Fzo R249L may have residual GTP affinity, explaining its partial function
The fzoK99T and fzoR249L mutations are recessive Analogous mutations in mammalian dynamms cause dominant negative phenotypes when expressed in tissue culture cells, formation of macromoiecular dynamm ring-shaped complexes appears to require GTP binding by all subunits In contrast to dynamms, Fzo molecules may act individually or form complexes in which only some subunits must bind GTP for proper assembly or function
Models for mitofusin orientation and function The conserved overall charge distribution and predicted transmembrane domain of mitofusins are consistent with a possible N out-Cιn orientation on mitochondria Typically, mitochondrial matrix proteins are more basic (with a higher predicted isoelectnc point (pi)) than their cytoplasmic isoforms, similarly, inner mitochondrial membrane proteins are generally basic in the matrix-residing region and more acidic in outside regions Mitofusins, when conceptually divided at their transmembrane domains, have N termini with predicted pis of 5 3 to 6 and C termini with pis of 8 1 to 9 3, suggesting that the C terminus may reside in the matrix The Fzo protein has eight acidic residues and only one argmine in its first 50 ammo acids, making import via a traditional N terminal targeting signal unlikely It is possible that the seπne- rich nature of this region allows mitochondrial targeting despite the acidic residues Alternatively, Fzo could be targeted to mitochondria by an internal basic region just C terminal to the predicted transmembrane domain (e g residues 621-636), leading to an N 0Ut-Cιn orientation, as for the S cerevisiae Bcslp protein The predicted transmembrane region of the Fzo protein consists of 13- and 20-resιdue uncharged regions separated by an argmine, the organization of hydrophobic and charged residues in this region is structurally conserved among Fzo homologs This region could potentially span one membrane twice Alternatively, as overall charge distribution in the protein suggests compartmental separation of the N and C terminal portions, it is plausible although unprecedented that Fzo could span both the inner and outer mitochondrial membranes at a contact site between these two membranes With the 20-resιdue region in the inner membrane and the argmine (perhaps with adjacent residues) in the mtermembrane space, the 13-resιdue hydrophobic region could traverse the outer membrane in a β sheet conformation, which requires fewer hydrophobic residues to span the membrane than an α helix This putative β sheet region could form lateral hydrogen bonds with other copies of itself or with β sheet transmembrane regions from other outer membrane proteins like porm In mammalian tissue culture cells, mitochondrial fusion appears to initiate where inner/outer membrane contact sites on each of two separate mitochondria are apposed Stable inner/outer membrane contact sites that are independent of protein import channels have been observed, perhaps the primary function of these sites is for mitochondrial fusion Mitofusins are good candidates to act at such putative fusion-mediating contact sites between inner and outer mitochondrial membranes
If mitofusins span both membranes with its C terminus in the matrix, then the predicted GTPase domain would be oriented toward the cytoplasm, where it could mediate interactions between mitochondria or regulate assembly of additional proteins The mitofusin protein could act as part of a ligand/receptor pair between separate mitochondria, binding to copies of itself or to other molecules displayed on the outer mitochondrial membrane Alternatively, the mitofusin protein could recruit other molecules to form a complex that links adjacent mitochondria Several regions of mitofusin could potentially participate in such protein-protein interactions, including the predicted coiled coil region at residues 377-400 as well as the N-termmal senne-rich region with its adjacent predicted "tπ-amphipathic" α helix
In either case, GTP binding and hydrolysis by mitofusins may regulate the specificity of these protein-protein interactions, as Rab GTPases seem to regulate formation of the SNARE complex prior to membrane fusion in the secretory and endocytic pathways Alternatively, the mitofusin predicted GTPase may have a biomechanical role, as may dynamin GTPases in the formation of endocytic vesicles GTP hydrolysis could cause a conformational change in the mitofusin itself or in other recruited proteins, bringing membranes close together and/or exposing residues that could act as a hydrophobic bridge between the fusing mitochondria Candidate regions in mitofusin to serve as such a bridge would be the hydrophobic faces of either the tπ-amphipathic α helix or the other predicted coiled coil regions This scenario would be analogous to influenza virus fusion, during which low pH triggers a conformational change in HA, or to a proposed conformational change in SNARE proteins that may be induced either by NSF ATP hydrolysis or by a yet unidentified GTPase during secretory membrane fusion events The orientation of the mitofusin protein may be different from that postulated in the above model, inserting in only one of the two mitochondrial membranes and/or taking on an N 0Ut-Cιn conformation It is possible that the transmembrane region traverses one membrane twice (or loops partially into a membrane from one side), causing both ends of the mitofusin to be in the same compartment mitofusins could mediate fusion of only one membrane, with other fusogens acting on the other Alternatively, the mitofusin protein may be only indirectly required for fusion, playing a role in modification or mitochondrial import of the molecules that participate directly
The Mitofusin family of predicted GTPases The mammalian mitofusins may mediate mitochondrial fusion and/or other functions in diverse cell types ESTs from genes encoding the human homologs were derived from brain, heart, pancreas, liver/spleen, and fibroblast cDNA libraries Mitochondrial fusion is known to occur in liver and is suggested to occur in certain rat neuronal cells and mouse 3T6 fibroblasts Mammalian spermatid mitochondria do not undergo massive fusion but become connected in a helical sheath around the sperm midpiece End-to- end contacts between mitochondria in this sheath contain structurally distinct "stud-like bridging elements", which seem to allow transmission of membrane potential through the helix Formation of these connections, which are also seen in rat cardiac tissue, may require the same protein mediators that are needed for full mitochondrial fusion in other cells Analysis of the role of mitofusins in diverse organisms will allow assignment of a general function for this new family of large multidomain GTPases, for which the name "mitofusins" is proposed
GenBank Accession Numbers The GenBank accession number for the fzo cDNA is U95821 and for the partial cDNA encoding the fzo human fetal brain homolog is U95822
All publications and patent applications cited in this specification are herein incorporated by reference as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference The citation of any publication is for its disclosure prior to the filing date and should not be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention
Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to those of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims
SEQUENCE LISTING
(1) GENERAL INFORMATION
(i) APPLICANT: Fuller, Margaret Hales , Karen
(ii) TITLE OF THE INVENTION: Mitofusin Genes and their Uses
(iii) NUMBER OF SEQUENCES: 6
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Bozicevic & Reed, LLP
(B) STREET: 285 Hamilton Avenue, Suite 200
(C) CITY: Palo Alto
(D) STATE: CA
(E) COUNTRY: USA
(F) ZIP: 94301
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Diskette
(B) COMPUTER: IBM Compatible
(C) OPERATING SYSTEM: DOS
(D) SOFTWARE: FastSEQ for Windows Version 2.0
(vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER:
(B) FILING DATE:
(C) CLASSIFICATION:
(vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: 06/048,961
(B) FILING DATE: 06-JUN-1997
(viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: Sherwood, Pamela J
(B) REGISTRATION NUMBER: 36,677
(C) REFERENCE/DOCKET NUMBER: SUN-63P
(ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: 650-327-3400
(B) TELEFAX: 650 327-3231
(C) TELEX:
(2) INFORMATION FOR SEQ ID NO : 1 :
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 2399 base pairs (B) TYPE: nucleic acid
(C) STRANDEDNESS : double
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: CDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 1 :
GAGCAAAACA ACATCTACAG TTGCCTATAT TTCATAAATA AATTTGTTAA CATTTTTGTA 60
ATATCTAATC ATAATGGCGG AATCTGACTC CGGAGAAAGT ACGTCGTCGG TGTCCTCGTT 120
TATATCCTCA TCGTCGTCTT CGCGATTAAG TGAGTTTGTG GACGCAAAGA CAGAACTGCA 180
GGATATATAT CACGATTTGA GTAATTACCT GTCCAATTTC CTAACCATTT TGGAGGAGAC 240
TGTCCTCTTA AAAGATCGAC AAATGCTGGA GCACCTGTGC GCCTTCTCCA GCAGGGTGGA 300
GGCCATTGCA AAGGTTCTTT CACGTGATCG AATGAAGGTG GCATTTTTTG GACGCACCTC 360
AAATGGAAAA AGTGCCGTGA TCAATGCACT TCTGCATGAA AAAATCCTGC CCAGCGCCAT 420
GGGCCATACC ACCAGCTGTT TTTGTCAAGT GCAAGCTAAT GGCTCGAATG AAACCGAGCA 480
CGTAAAGGTC GAGCAGGAGG ATGAGCATAT GGAACTGAGT GCCCTAAGCC AACTGGCCAG 540
TGCACATTCT CCTGGGGCCC TAAAACCCTC AACTCTGCTG CAGGTCAATA TGGCCAAGAA 600
CCGTTGCTCG ATATTGGATT ACGATGTGGT TTTGATGGAT ACACCTGGAG TGGATGTAAC 660
AGCGCAACTG GACGATTGCC TAGATAGCTA CTGCATGGAT GCGGATGTTT TCATTCTAGT 720
TCTCAACGCC GAGTCCACTG TTTCGCGCGT GGAAAGGCAG TTCTTCAAGG ACGTGGCATC 780
CAAACTCTCG CGTCCAAATC TCTTTATACT CAACAATCGA TGGGATAAGG CCAGCAGTCT 840
GGAGCCGGAA ATGGAGCAGA AGGTAAAGGA TCAGCATATG GAACGTTGCG TTAACCTGCT 900
CGTGGATGAA TTAGGTGTTT ATTCAACTGC ACAGGAAGCG TGGGAAAGGA TCTATCATGT 960
TTCAGCACTG GAGGCATTGC ATATAAGGAA TGGTCAGATT ACGAATCCCT CGGGACAAAC 1020
CCAACAGCGA TATCAGGAGT TTCTGCGTTT CGAAAATGAT TTTTCGAATT GCCTCGCGGT 1080
GTCAGCGTTA AAAACCAAAT TTGGTCCACA CTTGCTAAGT GCGCAGAAGA TTTTAAACCA 1140
GTTAAAATCA ACTCTGATAT GCCCTTTCAT AGAGAAAGTA AGTCGTCTTA TCGATGAGAA 1200
TAAGGAGAGA AGAGCTAACT TGAATGCCGA AATAGAGGAC TGGTTAATAC TAATGCAAGA 1260
GGATAGAGAA GCGCTTCAAT ATTGTTTCGA AGAACTGACT GAAATGACAC AAAGAGTAGG 1320
TCGGTGCGTT TTGAGCGACC AGATAAAAAC GTTAATACCC TCGTCTGTGC TATCATTCTC 1380
GCAACCATTT CACCCGGAAT TCCCAGCACA AATAGGCCAG TACCAACGCT CGTTATGTGC 1440
CCATTTGGAT AAACTTCTTG AAGATCGTGT CCTTCAATGT CTCTCCATAC CCCTACAAAG 1500
AGAAATATTA GATATAGAGA AAGAAATTGG GCTTCCGATC GCCGAGAACT CTTGCGATTG 1560
GCAACTAATC TACGGCCTGG ATTGCCAATC CTATATGAGT GACTTTCAGC CAGATCTTAG 1620
GTTTCGATTT TCTTTGGGTT TTACTGCCCT GTGGCATCGT CTTGAAGGCA ACCTACCGTT 1680
GCACGCAAGT CCATTTCGAA TTCAAAAGTT ACAAAATGGT CACAAGAAAT GTTCGCCCCT 1740
GCCACCTTTA GTTAACGGAA ACCATTGGCA GATGCTGGAA TCTTTGGTGA AGTCTAAAGG 1800
TAGCTTGGGC ACCGTTTTAC TGAGCGCCAT GGCCATCCGT TCGTTCAACT GGCCAATTGT 1860
ATTGATCCTT GGTGGGCTCG TCGGATCCTT TTACATCTAC GAGTACGCCG CTTGGACAAC 1920
TGCCGCCCAA GAGCGAAGTT TCAAGAGCCA GTACGCCAGG CTCTTGCAAC AACGTCTGCG 1980
GTCGGATGTG CAGCAAACTG TTAGCGGTTT TGAGCTCCAG TTGCGACAGC ACCTGGCAAC 2040
GGTCCGAAAT TGCTGGGAAG CCCAGTCCAA TGAGACACTG AATGACCTGA ACGTAAGGAC 2100
CGCGGAGCTG ACCAAACAAA TACAATCGAT GGAGGTGTTG CAGCTCAGCC TGAAGAAGTT 2160
TCGGGACAAG GGACAGCTGC TGGCCAGTCG GTTGGGAGAC TTTCAAGAGA CCTACTTGAC 2220
CAAGAGCTGA CAATTATGAG GGGGGTTACA ACAAATTCTA AATGTTCTTA ATAGTTTTAA 2280
TTTATTTTTG GTTACCTAAT TAAGTATTGT AATTCCGTTA TGTTACTTAG AAATTTTGTA 2340
TGTATTTGGT TGAATATTTT AAAATATTAA ACGATTGGTC TTCACTACTT TAAAGTTAA 2399
(2) INFORMATION FOR SEQ ID NO : 2 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 718 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO : 2 :
Met Ala Glu Ser Asp Ser Gly Glu Ser Thr Ser Ser Val Ser Ser Phe
1 5 10 15 lie Ser Ser Ser Ser Ser Ser Arg Leu Ser Glu Phe Val Asp Ala Lys
20 25 30
Thr Glu Leu Gin Asp lie Tyr His Asp Leu Ser Asn Tyr Leu Ser Asn
35 40 45
Phe Leu Thr lie Leu Glu Glu Thr Val Leu Leu Lys Asp Arg Gin Met
50 55 60
Leu Glu His Leu Cys Ala Phe Ser Ser Arg Val Glu Ala lie Ala Lys 65 70 75 80
Val Leu Ser Arg Asp Arg Met Lys Val Ala Phe Phe Gly Arg Thr Ser
85 90 95
Asn Gly Lys Ser Ala Val lie Asn Ala Leu Leu His Glu Lys lie Leu
100 105 110
Pro Ser Ala Met Gly His Thr Thr Ser Cys Phe Cys Gin Val Gin Ala
115 120 125
Asn Gly Ser Asn Glu Thr Glu His Val Lys Val Glu Gin Glu Asp Glu
130 135 140
His Met Glu Leu Ser Ala Leu Ser Gin Leu Ala Ser Ala His Ser Pro 145 150 155 160
Gly Ala Leu Lys Pro Ser Thr Leu Leu Gin Val Asn Met Ala Lys Asn
165 170 175
Arg Cys Ser lie Leu Asp Tyr Asp Val Val Leu Met Asp Thr Pro Gly
180 185 190
Val Asp Val Thr Ala Gin Leu Asp Asp Cys Leu Asp Ser Tyr Cys Met
195 200 205
Asp Ala Asp Val Phe lie Leu Val Leu Asn Ala Glu Ser Thr Val Ser
210 215 220
Arg Val Glu Arg Gin Phe Phe Lys Asp Val Ala Ser Lys Leu Ser Arg 225 230 235 240
Pro Asn Leu Phe lie Leu Asn Asn Arg Trp Asp Lys Ala Ser Ser Leu
245 250 255
Glu Pro Glu Met Glu Gin Lys Val Lys Asp Gin His Met Glu Arg Cys
260 265 270
Val Asn Leu Leu Val Asp Glu Leu Gly Val Tyr Ser Thr Ala Gin Glu
275 280 285
Ala Trp Glu Arg lie Tyr His Val Ser Ala Leu Glu Ala Leu His lie
290 295 300
Arg Asn Gly Gin lie Thr Asn Pro Ser Gly Gin Thr Gin Gin Arg Tyr 305 310 315 320
Gin Glu Phe Leu Arg Phe Glu Asn Asp Phe Ser Asn Cys Leu Ala Val
325 330 335
Ser Ala Leu Lys Thr Lys Phe Gly Pro His Leu Leu Ser Ala Gin Lys
340 345 350 lie Leu Asn Gin Leu Lys Ser Thr Leu lie Cys Pro Phe lie Glu Lys
355 360 365
Val Ser Arg Leu lie Asp Glu Asn Lys Glu Arg Arg Ala Asn Leu Asn
370 375 380
Ala Glu lie Glu Asp Trp Leu lie Leu Met Gin Glu Asp Arg Glu Ala 385 390 395 400
Leu Gin Tyr Cys Phe Glu Glu Leu Thr Glu Met Thr Gin Arg Val Gly
405 410 415
Arg Cys Val Leu Ser Asp Gin lie Lys Thr Leu lie Pro Ser Ser Val 420 425 430 Leu Ser Phe Ser Gin Pro Phe His Pro Glu Phe Pro Ala Gin lie Gly
435 440 445
Gin Tyr Gin Arg Ser Leu Cys Ala His Leu Asp Lys Leu Leu Glu Asp
450 455 460
Arg Val Leu Gin Cys Leu Ser lie Pro Leu Gin Arg Glu lie Leu Asp 465 470 475 480 lie Glu Lys Glu lie Gly Leu Pro lie Ala Glu Asn Ser Cys Asp Trp
485 490 495
Gin Leu lie Tyr Gly Leu Asp Cys Gin Ser Tyr Met Ser Asp Phe Gin
500 505 510
Pro Asp Leu Arg Phe Arg Phe Ser Leu Gly Phe Thr Ala Leu Trp His
515 520 525
Arg Leu Glu Gly Asn Leu Pro Leu His Ala Ser Pro Phe Arg lie Gin
530 535 540
Lys Leu Gin Asn Gly His Lys Lys Cys Ser Pro Leu Pro Pro Leu Val 545 550 555 560
Asn Gly Asn His Trp Gin Met Leu Glu Ser Leu Val Lys Ser Lys Gly
565 570 575
Ser Leu Gly Thr Val Leu Leu Ser Ala Met Ala lie Arg Ser Phe Asn
580 585 590
Trp Pro He Val Leu He Leu Gly Gly Leu Val Gly Ser Phe Tyr He
595 600 605
Tyr Glu Tyr Ala Ala Trp Thr Thr Ala Ala Gin Glu Arg Ser Phe Lys
610 615 620
Ser Gin Tyr Ala Arg Leu Leu Gin Gin Arg Leu Arg Ser Asp Val Gin 625 630 635 640
Gin Thr Val Ser Gly Phe Glu Leu Gin Leu Arg Gin His Leu Ala Thr
645 650 655
Val Arg Asn Cys Trp Glu Ala Gin Ser Asn Glu Thr Leu Asn Asp Leu
660 665 670
Asn Val Arg Thr Ala Glu Leu Thr Lys Gin He Gin Ser Met Glu Val
675 680 685
Leu Gin Leu Ser Leu Lys Lys Phe Arg Asp Lys Gly Gin Leu Leu Ala
690 695 700
Ser Arg Leu Gly Asp Phe Gin Glu Thr Tyr Leu Thr Lys Ser 705 710 715
(2) INFORMATION FOR SEQ ID NO : 3 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 2176 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 3:
ACAGTTAATC AACTGGCCCA TGCCCTTCAC ATGGACAAAG ATTTGAAAGC TGGCTGTCTT 60
GTACGTGTGT TTTGCCCAAA AGCAAAATGT GCCCTCTTGA GAGATGACCT GGTGTTAGTA 120
GACAGTCCAG GCACAGATGT CACTACAGAG CTGGATAGCT GGATTGATAA GTTTTGCCTA 180
GATGCTGATG TCTTTGTTTT GGTCGCAAAC TCTGAATCAA CACTAATGAA TACGGAAAAA 240
CACTTTTTTC ACAAGGTGAA TGAGCGGCTT TCCAAGCCTA ATATTTTCAT TCTCAATAAT 300
CGTTGGGATG CCTCTGCATC AGAGCCAGAA TATATGGAAG ACGTACGCAG ACAGCACATG 360
GAAAGATGCC TGCATTTCTT GGTGGAGGAG CTCAAAGTTG TAAATGCTTT AGAAGCACAG 420
AATCGTATCT TCTTTGTTTC AGCAAAGGAA GTTCTTAGTG CTAGAAAGCA AAAAGCACAG 480 GGGATGCCAG AAAGTGGTGT GGCACTTGCT GAAGGATTTC ATGCAAGATT ACAGGAATTT 540
CAGAATTTTG AACAAATCTT TGAGGAGTGT ATCTCGCAGT CAGCAGTGAA AACAAAGTTC 600
GAACAGCACA CTATCAGAGC TAAACAGATA CTAGCTACTG TGAAAAACAT AATGGATTCA 660
GTAAACCTGG CAGCTGAAGA TAAAAGGCAT TATTCAGTGG AAGAGAGGGA AGACCAAATT 720
GATAGACTGG ACTTTATTCG AAACCAGATG AACCTTTTAA CACTGGATGT TAAGAAAAAA 780
ATCAAGGAGG TTACCGAGGA GGTGCCAAAC AAAGTTTCAT GTGCAATGAC AGATGAAATT 840
TGTCGACTGT CTGTTTTGGT TGATGAATTT TGTTCAGAGT TTCATCCTAA TCCAGATGTA 900
TTAAAAATAT ATAAAAGTCT CCCTAGATCT TTAGCTTCTA CTCCCACTGC TCCTACCACT 960
CCAGCAACGC CAGATAATGC ATCACAGGAA GAACTCATGA TTACATTAGT AACAGGATTG 1020
GCGTCCGTTA CATCTAGAAC TTCTATGGGC ATCATTATTG TTGGAGGAGT GATTTGGAAA 1080
ACTATAGGCT GGAAACTCCT ATCTGTTTCA TTAACTATGT ATGGAGCTTT GTATCTTTAT 1140
GAAAGACTGA GCTGGACCAC CCATGCCAAG GAGCGAGCCT TTAAACAGCA GTTTGTAAAC 1200
TATGCAACTG AAAAACTGAG GATGATTGTT AGCTCCACGA GTGCAAACTG CAGTCACCAA 1260
GTAAAACAAC AAATAGCTAC CACTTTTGCT CGCCTGTGCC AACAAGTTGA TATTACCCAC 1320
AAACAGCTGG AAGAAGAAAT TGCTAGATTA CCCAAAGAAA TAGATCAGTT GGAGAAAATC 1380
CAAAACAATT CAAAGCTCTT AAGAAATAAA GCTGTTCAAC TTGAAAATGA GCTGGAGAAT 1440
TTTACTAAGC AGTTTCTACC TTCAAGCAAT GAAGAATCCT AACAATAGAG ATTGCTTTGG 1500
TGACCATGAT AGGAGGAAAC GAAACTTGTA AGATTGGAAC AGTTGTTATT TTTATGAAAT 1560
TACTTTAAAT ATGAATTGTA CTATCTGTAC CTAAATAGCA AAGCCCTGTG TAGATTCTGG 1620
TAATGATCTG TCTCAGGGTA TGTGTATTTT TGAAGAGTGT TATGTCCTTA GTTTTAATTT 1680
TGAGTAAAGA AAAGGCTAAA ATCATGAATT AGTTACAAGC AACAGTACCA ACTTATGTGA 1740
CCCCTGAGGG GTGGGGCTGT GAGCTCTTAA TTTGTTTTTG ATTCTGAAAA ACTCTGCTTC 1800
CTGGCATCCA GGAGTTAGAG ATTGAGCCTT TCATCTTCTT TCTCAACACT AGTTTTTGAT 1860
GCTTTCTTTC ATGGGAATAG TCACTTTTTT ATTTAGTAAA ATCGCATTGC TGGAACCACC 1920
CGCCGCATCT CTTCTTCACG GCGCCTGCGC TCTTCCTCCT GCCTGAGCTC CAGTTGCTTT 1980
CGTTTTTGCA CCTCTTGGTT GTGCGGCTCT TCCATCCTCC GAAGTTCTTC TTGGCGCCTC 2040
ATCAAATCCT GTCTCATTAG CATGACCTGG TGCTCATGGC GTGCAGCTTC CATCTCCATC 2100
TCCAGCTTCT CACGAGCCTA GCAGAAGGGG GTGGTGTCTC CAACCCCCCA GCACTGAGGA 2160
AGAGCGGGGC TCTTCT 2176
(2) INFORMATION FOR SEQ ID NO: 4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 493 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:4:
Thr Val Asn Gin Leu Ala His Ala Leu His Met Asp Lys Asp Leu Lys
1 5 10 15
Ala Gly Cys Leu Val Arg Val Phe Cys Pro Lys Ala Lys Cys Ala Leu
20 25 30
Leu Arg Asp Asp Leu Val Leu Val Asp Ser Pro Gly Thr Asp Val Thr
35 40 45
Thr Glu Leu Asp Ser Trp He Asp Lys Phe Cys Leu Asp Ala Asp Val
50 55 60
Phe Val Leu Val Ala Asn Ser Glu Ser Thr Leu Met Asn Thr Glu Lys 65 70 75 80
His Phe Phe His Lys Val Asn Glu Arg Leu Ser Lys Pro Asn He Phe
85 90 95
He Leu Asn Asn Arg Trp Asp Ala Ser Ala Ser Glu Pro Glu Tyr Met 100 105 110 Glu Asp Val Arg Arg Gin His Met Glu Arg Cys Leu His Phe Leu Val
115 120 125
Glu Glu Leu Lys Val Val Asn Ala Leu Glu Ala Gin Asn Arg He Phe
130 135 140
Phe Val Ser Ala Lys Glu Val Leu Ser Ala Arg Lys Gin Lys Ala Gin 145 150 155 160
Gly Met Pro Glu Ser Gly Val Ala Leu Ala Glu Gly Phe His Ala Arg
165 170 175
Leu Gin Glu Phe Gin Asn Phe Glu Gin He Phe Glu Glu Cys He Ser
180 185 190
Gin Ser Ala Val Lys Thr Lys Phe Glu Gin His Thr He Arg Ala Lys
195 200 205
Gin He Leu Ala Thr Val Lys Asn He Met Asp Ser Val Asn Leu Ala
210 215 220
Ala Glu Asp Lys Arg His Tyr Ser Val Glu Glu Arg Glu Asp Gin He 225 230 235 240
Asp Arg Leu Asp Phe He Arg Asn Gin Met Asn Leu Leu Thr Leu Asp
245 250 255
Val Lys Lys Lys He Lys Glu Val Thr Glu Glu Val Pro Asn Lys Val
260 265 270
Ser Cys Ala Met Thr Asp Glu He Cys Arg Leu Ser Val Leu Val Asp
275 280 285
Glu Phe Cys Ser Glu Phe His Pro Asn Pro Asp Val Leu Lys He Tyr
290 295 300
Lys Ser Leu Pro Arg Ser Leu Ala Ser Thr Pro Thr Ala Pro Thr Thr 305 310 315 320
Pro Ala Thr Pro Asp Asn Ala Ser Gin Glu Glu Leu Met He Thr Leu
325 330 335
Val Thr Gly Leu Ala Ser Val Thr Ser Arg Thr Ser Met Gly He He
340 345 350
He Val Gly Gly Val He Trp Lys Thr He Gly Trp Lys Leu Leu Ser
355 360 365
Val Ser Leu Thr Met Tyr Gly Ala Leu Tyr Leu Tyr Glu Arg Leu Ser
370 375 380
Trp Thr Thr His Ala Lys Glu Arg Ala Phe Lys Gin Gin Phe Val Asn 385 390 395 400
Tyr Ala Thr Glu Lys Leu Arg Met He Val Ser Ser Thr Ser Ala Asn
405 410 415
Cys Ser His Gin Val Lys Gin Gin He Ala Thr Thr Phe Ala Arg Leu
420 425 430
Cys Gin Gin Val Asp He Thr His Lys Gin Leu Glu Glu Glu He Ala
435 440 445
Arg Leu Pro Lys Glu He Asp Gin Leu Glu Lys He Gin Asn Asn Ser
450 455 460
Lys Leu Leu Arg Asn Lys Ala Val Gin Leu Glu Asn Glu Leu Glu Asn 465 470 475 480
Phe Thr Lys Gin Phe Leu Pro Ser Ser Asn Glu Glu Ser 485 490
(2) INFORMATION FOR SEQ ID NO: 5:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 27 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: Other
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 5 :
ACCTCAAATG GAACTAGTGC CGTGATC 27
(2) INFORMATION FOR SEQ ID NO : 6 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: Other
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 6 :
TACTCAACAA TCTATGGGAT AAG 23

Claims

WHAT IS CLAIMED IS
1 A purified polypeptide composition comprising at least 50 weight % of the protein present as a mitofusin protein or a fragment thereof
2 A purified polypeptide according to Claim 1 , wherein said polypeptide comprises a loss of function mutation
3 A purified polypeptide according to Claim 1 , wherein said mitofusin protein comprises the ammo acid sequence as set forth in SEQ ID NO 2 or SEQ ID NO 4
4 A DNA molecule or fragment thereof of at least about 18 nucleotides as part of other than a naturally occurring chromosome, comprising a sequence encoding a protein according to any of claims 1 to 3, or a complement thereof
5 A DNA molecule according to Claim 4, comprising a DNA sequence as set forth in SEQ ID NO 1 or SEQ ID NO 3
6 An isolated DNA molecule according to any of claims 4 to 5, wherein said DNA molecule comprises a transcnptional initiation region 5' to said sequence encoding a mitofusin protein
7 A cell comprising a DNA composition according to any of claims 4 to 6
8 An array of oligonucleotides comprising one or more sequences according to any of claims 4 to 6
9 An antibody specific for the polypeptide of any one of claims 1 to 3
10 A method of screening for biologically active agents that modulate mitofusin function, the method comprising combining a candidate biologically active agent with any one of
(a) a polypeptide according to any of claims 1 to 3, or
(b) a cell according to claim 7, and determining the effect of said agent on mitofusin function
11 A method of enhancing membrane fusion in a population of membrane-bound entities, the method comprising introducing a mitofusin polypeptide into said membrane under conditions that permit fusion of said entities
12 The method according to Claim 11 , wherein said membrane-bound entities are mitochondria
PCT/US1998/012064 1997-06-06 1998-06-05 Mitofusin genes and their uses WO1998055618A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
AU80660/98A AU736643B2 (en) 1997-06-06 1998-06-05 Mitofusin genes and their uses
EP98928991A EP1012279A4 (en) 1997-06-06 1998-06-05 Mitofusin genes and their uses
JP50317299A JP2002503964A (en) 1997-06-06 1998-06-05 Mitofusin gene and its use
CA002292840A CA2292840A1 (en) 1997-06-06 1998-06-05 Mitofusin genes and their uses

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US4896197P 1997-06-06 1997-06-06
US60/048,961 1997-06-06

Publications (2)

Publication Number Publication Date
WO1998055618A1 true WO1998055618A1 (en) 1998-12-10
WO1998055618A9 WO1998055618A9 (en) 1999-04-08

Family

ID=21957363

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1998/012064 WO1998055618A1 (en) 1997-06-06 1998-06-05 Mitofusin genes and their uses

Country Status (6)

Country Link
US (2) US6127159A (en)
EP (1) EP1012279A4 (en)
JP (1) JP2002503964A (en)
AU (1) AU736643B2 (en)
CA (1) CA2292840A1 (en)
WO (1) WO1998055618A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001025274A1 (en) * 1999-10-06 2001-04-12 The Board Of Trustees Of The Leland Stanford Junior University Mitofusins, fzo homologs and functional derivatives thereof
WO2004074482A1 (en) * 2003-02-21 2004-09-02 Universidad De Barcelona Diabetes and obesity diagnosis method
WO2015121461A1 (en) * 2014-02-17 2015-08-20 Nestec S.A. Methods and uses of mitofusins
WO2018057648A1 (en) * 2016-09-20 2018-03-29 The Board Of Trustees Of The Leland Stanford Junior University Peptide regulators of mitochondrial fusion and methods of use
US10167156B2 (en) 2015-07-24 2019-01-01 Curt G. Joa, Inc. Vacuum commutation apparatus and methods
US10724038B1 (en) * 2017-12-10 2020-07-28 Persona Biomed, Inc. Target-oriented therapeutic drug and in-vitro method of discovery thereof for modulating onset or progression of liver metastasis

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2002223768A (en) 2001-01-31 2002-08-13 Japan Science & Technology Corp Human mitochondoria protein and polynucleotide encoding the protein
IN2012DN00605A (en) * 2002-12-11 2015-06-12 Bristol Mayers Squibb Company
US8062891B2 (en) * 2003-10-24 2011-11-22 Gencia Corporation Nonviral vectors for delivering polynucleotides to plants
US8133733B2 (en) * 2003-10-24 2012-03-13 Gencia Corporation Nonviral vectors for delivering polynucleotides to target tissues
ES2411962T3 (en) 2003-10-24 2013-07-09 Gencia Corporation Methods and compositions for delivering polynucleotides
US20090123468A1 (en) * 2003-10-24 2009-05-14 Gencia Corporation Transducible polypeptides for modifying metabolism
US8507277B2 (en) * 2003-10-24 2013-08-13 Gencia Corporation Nonviral vectors for delivering polynucleotides
JP4675330B2 (en) 2003-11-14 2011-04-20 デューク ユニバーシティ Charcot-Marie-Tooth disease type 2A detection method
JP6253596B2 (en) * 2012-02-16 2017-12-27 ザ ペン ステイト リサーチ ファンデーション Acyl coenzyme A: a method for identifying an inhibitor of expression, function or activity of lysocardiolipin acyltransferase 1 (ALCAT1)
JP6644538B2 (en) * 2015-12-11 2020-02-12 ローランドディー.ジー.株式会社 Liquid ejection device and ink jet recording device provided with the same
AU2020215510A1 (en) 2019-01-28 2021-07-22 Mitochondria Emotion, Inc. Trans-4-hydroxycyclohexyl phenyl amide mitofusin activators and methods of use thereof
EP3917508A4 (en) 2019-01-28 2022-12-21 Mitochondria Emotion, Inc. Mitofusin activators and methods of use thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4898814A (en) * 1986-10-06 1990-02-06 Donald Guthrie Foundation For Medical Research, Inc. A cDNA clone for human tyrosinase
AU7623194A (en) * 1993-09-03 1995-03-22 Vpi Holdings Ltd. Oligonucleotides with rna cleavage activity

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
DATABASE MPSRCH GENBANK 1 January 1900 (1900-01-01), MATOBA R: "The Addition of 5'-Coding Information to a 3'-Directed cDNA Library Improves Analysis of Gene Expression", XP002910496, Database accession no. D16943 *
HALES K. G., ET AL.: "DEVELOPMENTALLY REGULATED MITOCHONDRIAL FUSION MEDIATED BY A CONSERVED, NOVEL PREDICTED GTPASE.", CELL, CELL PRESS, US, vol. 90., 11 July 1997 (1997-07-11), US, pages 121 - 129., XP002910495, ISSN: 0092-8674, DOI: 10.1016/S0092-8674(00)80319-0 *
See also references of EP1012279A4 *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001025274A1 (en) * 1999-10-06 2001-04-12 The Board Of Trustees Of The Leland Stanford Junior University Mitofusins, fzo homologs and functional derivatives thereof
WO2004074482A1 (en) * 2003-02-21 2004-09-02 Universidad De Barcelona Diabetes and obesity diagnosis method
WO2015121461A1 (en) * 2014-02-17 2015-08-20 Nestec S.A. Methods and uses of mitofusins
CN106030311A (en) * 2014-02-17 2016-10-12 雀巢产品技术援助有限公司 Methods and uses of mitofusins
US10167156B2 (en) 2015-07-24 2019-01-01 Curt G. Joa, Inc. Vacuum commutation apparatus and methods
WO2018057648A1 (en) * 2016-09-20 2018-03-29 The Board Of Trustees Of The Leland Stanford Junior University Peptide regulators of mitochondrial fusion and methods of use
US10578610B2 (en) 2016-09-20 2020-03-03 Washington University Peptide regulators of mitochondrial fusion and methods of use
US10724038B1 (en) * 2017-12-10 2020-07-28 Persona Biomed, Inc. Target-oriented therapeutic drug and in-vitro method of discovery thereof for modulating onset or progression of liver metastasis

Also Published As

Publication number Publication date
US6284507B1 (en) 2001-09-04
CA2292840A1 (en) 1998-12-10
WO1998055618A9 (en) 1999-04-08
AU736643B2 (en) 2001-08-02
AU8066098A (en) 1998-12-21
EP1012279A1 (en) 2000-06-28
US6127159A (en) 2000-10-03
JP2002503964A (en) 2002-02-05
EP1012279A4 (en) 2003-08-06

Similar Documents

Publication Publication Date Title
US6284507B1 (en) Mitofusin genes and their uses
US6359193B1 (en) Nucleotide sequences of lats genes
US6627405B1 (en) 53BP2 complexes
PT2045322E (en) Double-muscling in mammals
Suh et al. Cloning, genomic organization, alternative transcripts and expression analysis of CD99L2, a novel paralog of human CD99, and identification of evolutionary conserved motifs
JP2001523947A (en) Telomerase reverse transcriptase
JPH08509504A (en) Protein kinase
JP2000502570A (en) FHIT proteins and nucleic acids and methods based thereon
JPH09509851A (en) Human phosphodiesterase type IVC, and its production and use
Mann et al. Drosophila protein phosphatase V functionally complements a SIT4 mutant in Saccharomyces cerevisiae and its amino‐terminal region can confer this complementation to a heterologous phosphatase catalytic domain.
US6953680B2 (en) Mitofusins, Fzo Homologs and functional derivatives thereof
JP2003501026A (en) Lipid metabolism transcription factor
DE69936973T2 (en) phosphodiesterase
DE10004815B4 (en) Human intestinal sodium phosphate cotransporters
WO2001055410A9 (en) Ceramidase compositions and methods based thereon
WO1999000403A1 (en) Costal2 genes and their uses
JP2002503466A (en) Retinoblastoma protein complex and retinoblastoma interacting protein
JPWO2002064770A1 (en) Novel scavenger receptor class A protein
KR19990075173A (en) Epilepsy-induced Mutant Mice and Methods for Preparing the Same
FR2813612A1 (en) USE OF IP3 KINASES FOR THE PREPARATION OF NEW DRUGS IP3 KINASES AND CORRESPONDING DNA SEQUENCES
da Silva Molecular Characterization of a Gene Encoding a Mucin-Like Protein From Caenorhabditis elegans
ITTO20010138A1 (en) NEW METALLOPROTEASIS HAVING TROMBOSPONDINE DOMAINS AND NUCLEIC ACID COMPOSITIONS THAT CODE THEM.
CN1309135A (en) Novel human protein able to suppress cancer cell growth and its coding sequence
WO2003048196A2 (en) Means and methods for diagnosing and treating craniofacial malformations
MXPA00007131A (en) Asthma related genes

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI 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)
AK Designated states

Kind code of ref document: C2

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: C2

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

COP Corrected version of pamphlet

Free format text: PAGES 1/5-5/5, DRAWINGS, REPLACED BY NEW PAGES 1/5-5/5; DUE TO LATE TRANSMITTAL BY THE RECEIVING OFFICE

121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2292840

Country of ref document: CA

Ref country code: CA

Ref document number: 2292840

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 80660/98

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 1998928991

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1998928991

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 80660/98

Country of ref document: AU

WWW Wipo information: withdrawn in national office

Ref document number: 1998928991

Country of ref document: EP