US20150010647A1 - Methods for Identifying Fragile Histidine Triad (FHIT) Interaction and Uses Thereof - Google Patents

Methods for Identifying Fragile Histidine Triad (FHIT) Interaction and Uses Thereof Download PDF

Info

Publication number
US20150010647A1
US20150010647A1 US14/483,228 US201414483228A US2015010647A1 US 20150010647 A1 US20150010647 A1 US 20150010647A1 US 201414483228 A US201414483228 A US 201414483228A US 2015010647 A1 US2015010647 A1 US 2015010647A1
Authority
US
United States
Prior art keywords
fhit
cells
cell
fdxr
amount
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US14/483,228
Inventor
Carlo M. Croce
Francesco Trapasso
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ohio State University
Original Assignee
Ohio State 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 Ohio State University filed Critical Ohio State University
Priority to US14/483,228 priority Critical patent/US20150010647A1/en
Assigned to THE OHIO STATE UNIVERSITY reassignment THE OHIO STATE UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CROCE, CARLO M., TRAPASSO, FRANCESCO
Publication of US20150010647A1 publication Critical patent/US20150010647A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: OHIO STATE UNIVERSITY
Priority to US15/264,325 priority patent/US20160375107A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH reassignment NATIONAL INSTITUTES OF HEALTH CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: THE OHIO UNIVERSITY
Assigned to NATIONAL INSTITUTES OF HEALTH reassignment NATIONAL INSTITUTES OF HEALTH CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: THE OHIO STATE UNIVERSITY
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • 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
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y306/00Hydrolases acting on acid anhydrides (3.6)
    • C12Y306/01Hydrolases acting on acid anhydrides (3.6) in phosphorus-containing anhydrides (3.6.1)
    • C12Y306/01029Bis(5'-adenosyl)-triphosphatase (3.6.1.29)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/577Immunoassay; Biospecific binding assay; Materials therefor involving monoclonal antibodies binding reaction mechanisms characterised by the use of monoclonal antibodies; monoclonal antibodies per se are classified with their corresponding antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10041Use of virus, viral particle or viral elements as a vector
    • C12N2710/10043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10071Demonstrated in vivo effect
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10343Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/136Screening for pharmacological compounds
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/902Oxidoreductases (1.)
    • G01N2333/90293Oxidoreductases (1.) acting on reduced ferredoxin as donor (1.18)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)

Definitions

  • the FHIT gene encompasses the most active common fragile site at chromosome 3p14.2. Fhit expression is lost or reduced in a large fraction of most types of human tumors due to allelic loss, genomic rearrangement, promoter hypermethylation, or combinations thereof. Fhit knock-out mice show increased susceptibility to cancer development and FHIT gene therapy prevents tumors in carcinogen-exposed Fhit-deficient mice.
  • Fhit restoration by stable transfection in cancer cells has little effect in vitro, unless cells are exposed to stress, including the stress of the nude mouse environment in vivo; viral-mediated Fhit restoration, a process that simultaneously supplies stress and Fhit expression, suppresses tumorigenesis in vivo and triggers apoptosis of many types of malignant cells in vitro, including lung cancer cells.
  • Fhit expression is down-regulated by exposure to DNA damaging agents and Fhit plays a role in response to such agents), with Fhit-deficient cells escaping apoptosis and accumulating mutations.
  • proteins within cells were chemically cross-linked after infection of lung cancer cells with AdFHIT-His6 virus.
  • the proteins linked to Fhit and pathways affected by them were identified and characterized.
  • a method of diagnosing whether a subject has, or is at risk for developing, a cancer associated disorder comprising measuring the level of at least fragile histidine triad (Fhit) gene in a test sample from the subject, wherein an alteration in the level of the Fhit gene product in the test sample, relative to the level of a corresponding Fhit gene product in a control sample, is indicative of the subject either having, or being at risk for developing, a cancer associated disorder.
  • Fhit fragile histidine triad
  • FIGS. 1 A- 1 H Subcellular localization of Fhit protein in cytosol and mitochondria:
  • FIG. 1A immunofluorescence microscopy was performed with anti-Fhit serum on H1299 cells (D1) treated with PonA for 48 h; Fhit staining was detected using fluorescein isothiocyanate (green)-conjugated anti-rabbit immunoglobulin (IgG); Mito-Tracker Red staining, which identifies mitochondria, shows partial colocalization with Fhit. The yellow color on the fourth panel (lower right) shows the co-localizations points.
  • FIG. 1B immunoelectron microscopy of A549 AdFHIT (left) or AdFHIT-His 6 -infected cells (right) performed with a penta-His antibody shows Fhit mitochondrial localization (right); A549 cells infected with AdFHIT served as control and show only a few scattered grains (left panel).
  • FIG. 1C immunoblot analysis of AdFHIT-infected A549 subcellular fractions using anti-Fhit indicates Fhit protein distribution in the cytosol, membranes, cytoskeleton, and mitochondria.
  • FIG. 1D immunoblot analysis of proteins from mitochondria of A549 cells infected with AdFHIT-His 6 after treatment with sodium carbonate ( FIG. 1E ) and increasing concentrations of digitonin ( FIG. 1F ) indicates that Fhit is mainly distributed in mitochondrial matrix; filters were probed with Fhit and CoxIV antisera; lanes in FIG. 1F represent supernatants after treatment with 0, 0.10, 0.15, and 0.20% digitonin.
  • FIG. 1G immunoblot analyses of subcellular fractions from MKN74/E4 and MKN74/A116 cells (stably expressing exogenous Fhit), and FIG. 1H , HCT116 (an endogenous Fhit-positive colon cancer cell line) using anti-Fhit, confirms Fhit mitochondrial localization; GAPDH and CoxIV antisera served as controls.
  • FIGS. 2 A-F Exogenous and endogenous Fhit forms a complex with endogenous Hsp60, Hsp10, and Fdxr proteins:
  • FIGS. 2A , 2 B, 2 C Protein complexes, isolated with recombinant Fhit-His 6 protein, were separated on polyacrylamide gels and probed with antisera against Hsp60 ( FIG. 2A ), Hsp10 ( FIG. 2B ), and Fdxr ( FIG. 2C ); in the latter panel, prepared after mitochondria isolation, it is shown that Fhit recruits Fdxr in the mitochondria in a time-dependent manner. Filters were loaded with protein isolated after infection of A549 cells with AdFHIT-His 6 with or without DSP.
  • FIG. 2D coimmunoprecipitation with anti-Hsp60 after infection of A549 cells with AdFHIT; filters were probed with Hsp60, Fhit, and Hsp10 antisera.
  • FIG. 2E A549 cells were co-transfected with the V5-tagged FDXR gene and FHIT plasmids; immunoprecipitation was with anti-V5 and detection with Fdxr and Fhit antisera.
  • FIG. 2F immunoprecipitation and immunoblot detection of endogenous interactor proteins (Fdxr and Hsp10) from DSP-treated Fhit-positive HCT116 cells. Filters were probed with antisera against each target protein. Endogenous Fhit co-precipitated with Hsp10 and Fdxr.
  • FIGS. 3 A-D Knockdown of Hsp60/Hsp10 reduces the level of Fhit in the mitochondria:
  • FIG. 3A nickel-H6 pull down experiment of A549 cells AdFHIT-His 6 infected on subcellular fractions (Cytosol and Mitochondria) using H6 antibody; lysates were incubated with nickel beads to isolate the DSP cross-linked Fhit-His 6 protein complex and loaded on a 4-20% polyacrylamide gel. 24 h after infection, the Hsp60-Fhit complex was present in both compartments; 48 h after infection, the complex was detectable again in both compartments and the increase of Fhit complex proteins appears related to the increase of Fhit protein at 48 h after AdFHIT-His 6 infection, with a slight increase in the mitochondria (densitometry analysis on input samples was performed).
  • FIG. 3B immunoblot analysis of Hsp60, Hsp10, Fhit, and GAPDH in Fhit-positive D1 cells after 72 h of Hsp60/Hsp10 silencing showing Fhit, Hsp60, and Hsp10 levels after a CHX chase (30 ⁇ g/ml) for 1-12 h.
  • FIG. 3C immunoblot analysis of cytosol/mitochondrial protein fractions of A549 cells 72 h after transfection with Hsp60 and Hsp10 siRNAs and 24 h after AdFHIT infection at m.o.i. 1, with Hsp60, Hsp10, Fhit, GAPDH, and CoxIV antisera.
  • Hsp60/10 silencing does not appear to affect the Fhit cytosolic level, but is associated with a decrease of Fhit in the mitochondrial fraction.
  • Scrambled (Scr) siRNAs were used as controls.
  • FIG. 3D subcellular fractionation and immunoprecipitation of “endogenous” Fhit complex proteins.
  • Endogenous Hsp60 co-precipitated Fhit and Fdxr.
  • FIGS. 4 A-F Fhit expression induces intracellular ROS generation after treatment of cells with peroxide:
  • FIG. 4A fluorescence-activated cell sorter (FACS) analysis for ROS assessment in A549 cells 48 h after transfection with FHIT plasmid, with and without a 5-h H 2 O 2 treatment. Empty vector-transfected cells served as control. Intracellular superoxide was determined according to the fluorescence of ethidium as a result of oxidation of hydroethidine by O 2 . M2 refers to the fraction of ROS positive cells.
  • FACS fluorescence-activated cell sorter
  • FIG. 4B FACS analysis for ROS assessment by the fluorescence produced from the oxidation of hydroethidine in D1 and E1 cells; 48 h after PonA treatment, cells were treated for 5 h with 0.5 and 1.0 mM H 2 O 2 and oxidative stress was measured; % positive refers to the fraction of fluorescent cells, indicating ROS.
  • FIG. 4C increased green fluorescent DCF signal in H1299 Fhit-expressing cells (D1) under stress conditions.
  • Cells were incubated with 2′,7′-dichlorodihydrofluorescein diacetate, a ROS indicator that can be oxidized in the presence of ROS to the highly green fluorescent dye DCF, at 48 h after Fhit induction and after a 5-h H 2 O 2 treatment of E1 and D1 cells (magnification ⁇ 40).
  • FIG. 4D MTS cell viability assays were performed on E1 and D1 cells.
  • Cells were treated with PonA for 48 h and then with increasing concentrations of H 2 O 2 (0.125, 0.25, and 0.5 mM) for 4 h.
  • Analysis was at 24 h after H 2 O 2 treatment.
  • Columns report the average of four experiments ⁇ S.E. Each point was measured in quadruplicate and standard deviation calculated; p ⁇ 0.05 was considered significant.
  • FIG. 4E FACS analysis of D1 and E1 cell cycle kinetics at 48 h after oxidative stress treatment.
  • Cells were treated with PonA for 48 h and then with increasing concentrations of H 2 O 2 (0.25 and 0.5 mM) for 4 h. Analysis was at 48 h after H 2 O 2 treatment. All experiments were performed twice in triplicate.
  • FIG. 4F colony formation assay of H1299/D1 and H1299/E1 cells after 5 mM PonA stimulation and a 5-h H 2 O 2 treatment at the indicated concentrations.
  • FIG. 5A immunoblot analysis with antisera against Fdxr, Fhit, and GAPDH. Proteins were extracted from E1 (control) and D1 cells 48 h after treatment with PonA.
  • FIG. 5B immunoblot analysis of Fdxr expression in D1 and E1 cells after a 4-h treatment with 25 ⁇ M MG132, a proteasome inhibitor. GAPDH detection shows equal protein loading.
  • FIG. 5C immunoblot analysis of Fdxr, Fhit, and GAPDH in D1, expressing Fhit, and E1 cells, showing Fdxr level after CHX chase (30 ⁇ g/ml) for 4-12 h. Densitometry based on GAPDH levels shows enhanced stability of Fdxr in the presence of Fhit.
  • FIG. 5D FACS analysis of FDXR +/+/+ and FDXR +/ ⁇ / ⁇ cell cycle kinetics after infection with AdFHIT m.o.i. 50 and 100. The experiment was performed 48 h after infection and was repeated three times with similar results. Profiles of AdGFP-infected cells were similar to those of non-infected cells (not shown).
  • FIG. 5E immunoblot analysis showing expression of Fdxr, Fhit, and GAPDH after infection of FDXR +/+/+ and FDXR +/ ⁇ / ⁇ with AdFHIT m.o.i. 50 and 100. Proteins were extracted 48 h after infection.
  • FIG. 5F real-time RT-PCR analysis for FDXR expression at 48 h after AdFHIT m.o.i. 50.
  • the PCR product was normalized to GADPH and Actin expression and each point was repeated in quadruplicate; differences between control and Fhit positive samples were not significant.
  • FIG. 5G caspase 3 and Parp1 activation.
  • Immunoblot analysis using Fhit, caspase 3, Parp1 antisera, of total cell lysates from HCT116 FDXR +/+/+ cells 48, 72, and 96 h after infection with AdFHIT and AdGFP at m.o.i. 50.
  • GAPDH and CoxIV served as internal protein markers.
  • FIG. 5H immunoblot analysis, using Fhit and cytochrome c antisera, of cytosol/mitochondria fractions from HCT116 FDXR +/+/+ cells 48, 72, and 96 h after infection with AdFHIT and AdGFP at m.o.i. 50.
  • GAPDH and ⁇ -actin served as internal protein markers.
  • FIGS. 6 A-E Fhit enhances the sensitivity of cancer cells to paclitaxel and cisplatin:
  • FIGS. 6A , 6 B MTS assays performed on E1 and D1 cells.
  • Cells were treated with PonA for 48 h and then treated with paclitaxel (50-500 ng/ml) ( FIG. 6A ) or cisplatin (0.05-0.2 mM) ( FIG. 6B ) for 24 or 48 h.
  • Bars report the average of four experiments ⁇ S.E. Each point was measured in quadruplicate and standard deviation calculated; asterisks next to brackets in FIG. 6A and FIG. 6B indicate statistically significant differences in drug response of D1 and E1 cells, p ⁇ 0.05.
  • FIG. 6C , FIG. 6D the graphs show representative results of flow cytometry analyses of E1 and D1 cells.
  • Cells were treated with PonA for 48 h and then with paclitaxel (50-500 ng/ml) ( FIG. 6C ) or cisplatin (0.05-0.2 mM) ( FIG. 6D ).
  • paclitaxel 50-500 ng/ml
  • cisplatin 0.05-0.2 mM
  • FIG. 6E caspase 3 and Parp1 cleavage: immunoblot analyses, using Fhit, caspase 3, and Parp1 antisera, of total cell lysates from PonA-induced D1 cells after 48 h of treatment with paclitaxel (50 and 100 ng/ml) or cisplatin (0.05 and 0.1 mM). GAPDH served as loading control.
  • FIG. 7 TABLE 1 Candidate Fhit protein partners isolated through mass spectrometry. Proteins selectively captured in the A549 AdFHIT-H 6 -infected cells sample. Amino acid sequence of identified peptides, Mascot scores, and protein sequence coverage are listed.
  • FIGS. 8 A- 8 C Ad-His6 biological activity is comparable to AdFHIT:
  • FIG. 8A Western blot analysis of A549 cells infected with Ad-His6, MOI 20. Fhit-His protein was detected by antipentaHis and antiFhit serum. Both Ad FHIT and Ad-His6 carry a GFP cDNA regulated by a CMV5 promoter through an internal ribosome entry sequence downstream of FHIT. ⁇ -tubulin was used to normalize sample loading.
  • FIG. 8B How cytometry analysis of A549 cells 96 hr after infection with Ad-His6, MOI 15.
  • Upper panel indicates the subG1 DNA content of infected cells (experiment repeated thrice; average values of subG1 fractions 22%+/ ⁇ 4.3 for Ad FHIT, 29%+/ ⁇ 5 for Ad-His6; the difference is not statistically significant; lower panel shows percentages of cells with mature caspase-3, an indication of apoptosis.
  • the extent of cell death in A549 cells infected with Ad-His6 is comparable to the result obtained after infection with Ad FHIT.
  • FIG. 8C In vivo cross-linking of Fhit-His6. Silver staining of gel with cell lysates after His6 pull down and cross-link reversal conditions, separated by 4-20% gradient SDS-PAGE. Internal negative controls included His6 pull down of Ad FHIT infected cells (cross-linked, CL) and Ad-His6 infected cells (not cross-linked, NT).
  • FIGS. 9A-9F Initial validation of candidate Fhit protein partners identified through nanobore LC-MS/MS. Selected ion chromatograms (SIC) for AdFHIT-His6 and control samples are shown. The six SICs pairs report ion currents of the six following m/z values: 1) 672.8 (peak at retention time 30 min. was identified as tryptic peptide TVIIEQSWGSPK [SEQ ID NO: 5] belonging to Hsp60), 2) 685.4 (peak at retention time 32 min. identified as tryptic peptide LGPALATGNVVVMK [SEQ ID NO: 22] belonging to Aldh2), 3) 617.3 (peak at retention time 39 min.
  • SIC selected ion chromatograms
  • FIG. 10 Fhit induces generation of ROS in MKN74 gastric cancer cells.
  • ROS assessment was performed with MKN74A116, a human gastric cancer cell line carrying a p53 mutant allele and expressing exogenous Fhit; Fhit-negative MKN74E4 cells were used as a control.
  • MKN74A116 a human gastric cancer cell line carrying a p53 mutant allele and expressing exogenous Fhit
  • Fhit-negative MKN74E4 cells were used as a control.
  • MKN74 cells for 5 hr with 0.5, 1.0 and 2.0 mM H 2 O 2
  • Results indicate a significantly higher rate of ROS generation in cells expressing exogenous Fhit compared to controls; toxicity was observed in Fhit-expressing cells after 2 mM H 2 O 2 treatment. Numbers report the average of four experiments ⁇ S.E.
  • A, B, C, or combinations thereof refers to all permutations and combinations of the listed items preceding the term.
  • A, B, C, or combinations thereof is intended to include at least one of: A, B, C, AB, AC, BC, or ABC, and if order is important in a particular context, also BA, CA, CB, ACB, CBA, BCA, BAC, or CAB.
  • GAPDH glyceraldehyde-3-phosphate dehydrogenase
  • DSP dithiobis(succinimidyl propionate)
  • LC-MS/MS liquid-chromatography tandem mass spectrometry
  • Fdxr ferredoxin reductase
  • PonA ponasterone A
  • m.o.i. multiplicity of infection
  • ROS reactive oxygen species
  • FU 5-fluorouracil
  • DCFH-DA dichlorofluorescein-diacetate
  • DCF 2′,7′-dichlorofluorescein
  • CHX cycloheximide
  • siRNA small interfering RNA
  • RT reverse transcriptase
  • MTS 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium.
  • Fhit protein is lost in most cancers, its restoration suppresses tumorigenicity, and virus-mediated FHIT gene therapy induces apoptosis and suppresses tumors in preclinical models.
  • Protein cross-linking and proteomics methods are used to characterize a Fhit protein complex involved in triggering Fhit-mediated apoptosis.
  • the complex includes Hsp60 and Hsp10 that mediate Fhit stability and may affect import into mitochondria, where it interacts with ferredoxin reductase, responsible for transferring electrons from NADPH to cytochrome P450 via ferredoxin.
  • Viral-mediated Fhit restoration increases production of intracellular reactive oxygen species, followed by increased apoptosis of lung cancer cells under oxidative stress conditions; conversely, Fhit-negative cells escape apoptosis, carrying serious oxidative DNA damage that may contribute to an increased mutation rate.
  • Characterization of Fhit interacting proteins has identified direct effectors of the Fhit-mediated apoptotic pathway that is lost in most cancers through loss of Fhit.
  • Fhit protein interactors adenovirus-transduced Fhit-His 6 for Fhit complex purification after cross-linking, and Fhit-linked proteins, Hsp60, Hsp10, and Fdxr, were identified; subcellular location of these proteins suggested that mitochondria might be foci of Fhit activity.
  • Hsp “stress proteins” as molecular chaperones perform functions such as protein translocation, folding, and assembly.
  • Targeted disruption of the FDXR gene in HCT116 colon cancer cells showed that it was essential for viability; reduction of the gene copy number resulted in decreased sensitivity to 5-fluorouracil-induced apoptosis and FDXR is a target gene of the p53 family.
  • activated p53 induces apoptosis in response to cellular stresses in part through ROS, and simultaneously p53 increases transcription of the FDXR gene, which in turn enhances p53 function by increasing ROS-induced apoptosis.
  • Fhit does not affect the FDXR transcriptional level but may affect stability of the protein.
  • H1299 cells missing both Fhit and p53, Fdxr overexpression increases sensitivity to ROS-induced cell death, and H1299 cells expressing inducible Fhit or p53 are sensitive to ROS-induced cell death; cancer cells missing Fhit, p53, or both would lack ways to increase Fdxr expression, and would be less sensitive to oxidative damage and would survive.
  • the inventors herein cross-linked proteins within cells after viral-mediated Fhit overexpression in lung cancer cells, and characterized proteins associated with Fhit and the pathways affected by them.
  • A549 lung cancer-derived cells which are susceptible to Fhit-induced apoptosis (10), were infected with AdFHIT or AdFHIT-His 6 and treated with DSP, a cross-linker that crosses membranes and fixes proteins in complex in vivo.
  • Cells were lysed and proteins isolated with nickel beads avid for the His 6 epitope tag. Purified proteins were treated with dithiothreitol to cleave DSP and dissociate the complex, and digested by trypsin; protein constituents were identified by LC-MS/MS (FIG. 7 —Table 1 and FIG. 9 ).
  • Hsp60 and Hsp10 are also distributed in the cytosol.
  • Fhit Subcellular Localization Because candidate Fhit interactors are mitochondrial proteins, the inventors herein determined if Fhit, which lacks a mitochondrial localization signal, localized in these organelles.
  • Fhit negative H1299 lung cancer cells carrying an inducible FHIT cDNA (D1 cells) were treated with the inducer, PonA for 48 h and indirect immunofluorescence detection of Fhit subcellular location was assessed using anti-Fhit serum and MitoTracker Red 580, a marker of mitochondria;
  • Fhit fluorescent signal green staining, FIG. 1A
  • FIG. 1A was cytoplasmic and partly co-localized (yellow staining, FIG.
  • mitochondria were purified from A549 cells infected with AdFHIT m.o.i. 1, as described above.
  • the sodium carbonate procedure is a nondestructive approach that allows effective release in the supernatant of both soluble proteins and peripheral membrane proteins from intracellular membranes after inducing the generation of open sheets of membranes; furthermore, it allows recovery of integral proteins with the membranes (pellet)
  • FIG. 1E shows that Fhit was only detectable in the soluble fraction.
  • mitochondria were treated with 0.10 and 0.15% digitonin to selectively disrupt mitochondrial outer membrane, releasing proteins contained in the intermembrane space and the matrix; as shown in FIG. 1F , gradual disruption of outer and inner membranes releases increasing amounts of Fhit protein, suggesting that Fhit is mainly distributed either at the luminal side of the inner membrane or in the matrix of mitochondria.
  • Mitochondrial localization was confirmed in gastric cancer-derived MKN74A116 cells stably expressing exogenous Fhit and in HCT116 colon cancer cells expressing endogenous Fhit ( FIG. 1G and FIG. 1H ).
  • Hsp60 and Hsp10 are candidate interactor proteins that the inventors focused on Hsp60 and Hsp10 as possible chaperonins and on Fdxr, a mitochondrial respiratory chain protein transactivated by p53 and involved in responses to therapeutic drugs.
  • AdFHIT or AdFHIT-His6 were infected with AdFHIT or AdFHIT-His6 at m.o.i. 20, with or without DSP.
  • Fhit complexes were purified through the His 6 tag and co-purified proteins were detected with antisera against Hsp60, Hsp10, and Fdxr; Hsp60 and Fdxr were detected only in lysates of cells exposed to DSP ( FIG. 2A and FIG. 2C ), whereas Hsp10 was also detectable without cross-linking ( FIG. 2B ).
  • FIG. 2C A time course experiment after infection showed recruitment of Fdxr by Fhit ( FIG. 2C ); also, endogenous Hsp60 co-immunoprecipitated Fhit and Hsp10 in the absence of DSP ( FIG. 2D ).
  • each endogenous candidate interactor protein from DSP-treated Fhit-positive HCT116 cells and looked for co-precipitation of endogenous Fhit ( FIG. 2F ).
  • Endogenous Fhit co-precipitated with Hsp10 and Fdxr, confirming the presence of endogenous Fhit in mitochondria and its interaction with endogenous chaperones and respiratory chain protein in the absence of stress.
  • A549 cells were infected with AdFHIT-His 6 m.o.i. 5 and protein lysates were collected from cytosol and mitochondrial fractions after cross-linking. Complexes were isolated by Fhit-H6-nickel pull down, separated on a polyacrylamide gel, and filters probed with Hsp60 and Fhit antisera. At 24 and 48 h after infection interaction with Hsp60 is observed in the cytosol and mitochondria ( FIG. 3A ) commensurate with the increase in Fhit expression at these times (Input), as shown in FIG. 3A .
  • H1299 with inducible Fhit expression were transfected with Hsp60 and Hsp10 siRNAs and 72 h after transfection a CHX chase was performed at 1, 6, and 12 h and Fhit protein expression was assessed and compared with cells transfected with the scrambled sequence.
  • Hsp60 was immunoprecipitated from total cell lysates of these cells at 48 h after PonA induction, with or without H 2 O 2 , and coprecipitated Fhit and Fdxr ( FIG. 3C , right panel). Induction of Fhit expression in D1 cells does not cause biological changes in vitro; thus the Fhit complex does not form as a consequence of apoptosis.
  • a time course experiment was performed in D1 cells after PonA-induced Fhit expression, with and without stress conditions, to determine whether there were biological changes in Fhit protein interactors. The co-inventors did not detect changes in localization after Fhit expression.
  • Fdxr Fhit Induces Generation of Reactive Oxygen Species (ROS)—Fdxr, a 54-kDa flavoprotein, is located on the matrix side of the inner mitochondrial membrane, and is responsible for transferring electrons from NADPH, via the single electron shuttle ferredoxin-cytochrome P450, to substrates. Under substrate-limiting conditions, electrons leak from this shuttling system and generate ROS. Fdxr mediates p53-dependent, 5-fluorouracil-induced apoptosis in colorectal cancer cells, through generation of ROS, potent intracellular oxidants, and regulators of apoptosis.
  • ROS Reactive Oxygen Species
  • the inventors herein investigated determine whether ROS production could be involved in Fhit-mediated apoptosis.
  • Overexpression of Fdxr increases sensitivity of tumor cells to apoptosis on H 2 O 2 treatment, through ROS production.
  • the inventors examined ROS production in A549 cells, with and without H 2 O 2 treatment, after transient transfection with the FHIT expression plasmid. Intracellular superoxide was assessed by measuring ethidium fluorescence, as a result of oxidation of hydroethydine by superoxide. Intracellular superoxide was measured 5 h after stimulation with increasing concentrations of H 2 O 2 .
  • ROS generation was ⁇ 3 times higher (16.7 versus 5.4% at 0.5 mM H 2 O 2 and 18.8 versus 7.7% at 1.0 mM H 2 O 2 ) in FHIT-transfected cells. 2 mM H 2 O 2 was toxic to Fhit-expressing but not to non-expressing cells ( FIG. 4A ).
  • DCFH-DA was used to measure the redox state of Fhit-overexpressing cells.
  • Peroxidases, cytochrome c, and Fe 2+ can oxidize DCFH-DA to fluorescent 2′,7′-dichlorofluorescein (DCF) in the presence of H 2 O 2 ; thus, DCF indicates H 2 O 2 levels and peroxidase activity.
  • DCF 2′,7′-dichlorofluorescein
  • Increased DCF fluorescence was detected in D1 cells compared with E1 cells under stress conditions ( FIG. 4C ).
  • H 2 O 2 treatment caused reduced cell viability or growth arrest in both E1 and D1 cells, but this phenotype was more pronounced in D1 cells ( FIG. 4D ).
  • FIG. 4F To assess if the G 2 /M arrest could affect long-term viability of the cells, a colony assay was performed ( FIG. 4F ). No colonies were detected in Fhit-expressing cells after exposure to 0.25 mM or higher concentrations of H2O2.
  • the inventors next measured the Fdxr level, with or without Fhit expression, in the presence of MG132, an inhibitor of proteasome degradation; 4 h after MG132 treatment a significant increase of Fdxr protein was observed in D1 cells compared with E1 cells ( FIG. 5B ), showing that Fhit protects Fdxr from proteasome degradation.
  • the rate of Fdxr degradation in the presence or absence of Fhit protein was evaluated by the 4-12-h CHX chase ( FIG. 5C ); the rate of Fdxr degradation was higher in Fhit-negative E1 cells (declining from 1 to 0.3) compared with D1 cells, with no significant decrease.
  • Fhit prevents destabilization of the Fdxr protein by protecting it from proteasome degradation.
  • HCT116 colon cancer cells which express endogenous wild-type p53 and Fhit and carry three FDXR alleles (FDXR +/+/+ ), and HCT116 FDXR +/ ⁇ / ⁇ cells with two alleles knocked-out (28), were used to determine whether AdFHIT-induced apoptosis is influenced by the Fdxr expression level; the FDXR null condition was not compatible with viability.
  • Wild-type HCT116 cells (FDXR +/+/+ ) were susceptible to exogenous Fhit-mediated apoptosis in a dose-dependent manner, as the fraction of sub-G 1 cells was 12.1 and 18.8% at m.o.i. 50 and 100, respectively; FDXR +/ ⁇ / ⁇ cells were refractory at 48 and 72 h (data not shown) to Fhit-induced cell death, with a sub-G 1 population of 4.7 and 4.3% at m.o.i. 50 and 100 ( FIG. 5D ).
  • the Fhit-mediated increase of Fdxr expression was not at the transcriptional level, as determined by real time RT-PCR ( FIG. 5F ) and thus not related to the p53 transcriptional activation.
  • Fhit Enhances ROS-related Effects of Chemotherapeutic Agents Genetics of intracellular ROS is an early event in the apoptosis of lung cancer cells induced by treatment with paclitaxel.
  • Cisplatin induces Fdxr expression and the cisplatin-induced apoptotic pathway is associated with ROS generation.
  • Fhit expressing D1 cells were more sensitive than E1 cells to cisplatin, measured by MTS assay at 24 and 48 h ( FIG. 6B ).
  • H1299, MKN74-E4, and A116, and HCT116 cells were maintained in RPMI 1640 medium plus 10% fetal bovine serum and penicillin/streptomycin (Sigma).
  • HEK293 cells (Microbix) used for preparation of recombinant adenoviruses were cultured in Dulbecco's modified Eagle's medium plus 10% fetal bovine serum and penicillin/streptomycin.
  • AdFHIT-His6 virus was prepared as described in Example II herein. [His6—SEQ ID NO: 32] [penta-His—SEQ ID NO: 33].
  • Full-length FDXR was amplified from human brain cDNAs (Clontech), subcloned into the pcDNA3.1/V5-HisTOPO TA vector (Invitrogen) and sequenced; details are as described under supplemental Methods. Cells were transfected using LipofectamineTM (Invitrogen) following the manufacturer's directions.
  • Protein Interaction Analyses Protein Interaction Analyses: Proteins were extracted in 15 mM Tris-Cl, pH 7.5, 120 mM NaCl, 25 mM KCl, 2 mM EGTA, 0.1 mM dithiothreitol, 0.5% Triton X-100, 10 mg/ml leupeptin, 0.5 mM phenylmethylsulfonyl fluoride.
  • Fhit was sublocalized in ponasterone A (PonA)-induced, Fhit-expressing H1299 D1 cells by indirect immunofluorescence detection using anti-Fhit serum and by detection of FhitHis6 in A549 AdFHIT-His 6 -infected cells in immunoelectron micrographs using anti-pentaHis.
  • mitochondria were isolated with the Mitochondria/Cytosol Fractionation kit and the FractionPREPTM Cell Fractionation System was used to extract proteins from cytosol, membranes, nuclei, and cytoskeleton (Biovision Research Products).
  • mitochondria were resuspended in 0.1 M sodium carbonate, pH 11.5, on ice for 30 min with periodic vortexing and fractionated as described herein.
  • HCT116 FDXR +/+/+ and FDXR +/ ⁇ / ⁇ cells were infected with AdFHIT or AdGFP at m.o.i. 50 and 100 and assessed at 48 h postinfection.
  • PonA-induced H1299 D1 and E1 cells were treated with 0.25 and 0.5 mM H 2 O 2 or with chemotherapeutic drugs and incubated for varying times, as indicated in the text and figures. For both experiments the cells were collected, washed with phosphate-buffered saline, and resuspended in cold 70% ethanol.
  • cells were spun down, washed in phosphate-buffered saline, and suspended in 0.1 mg/ml propidium iodide/Triton X-100 staining solution (0.1% Triton X-100, 0.2 mg/ml DNase-free RNase A) for 30 min at room temperature and analyzed by flow cytometry.
  • ROS Intracellular Reactive Oxygen Species
  • DCFH-DA Dichlorofluorescein diacetate
  • H 2 O 2 0.1 to 1.0 mM
  • DCF fluorescence was measured by flow cytometry on a FAC-Scan flow cytometer and fluorescence microscopy.
  • Protein loading was normalized with GAPDH and CoxIV. 1 ⁇ 10 6 H1299 D1 and E1 lung cancer cells were transfected as described above and at 24 h after transfection the cells were PonA-induced; 48 h after induction a cycloheximide (CHX) (10 ⁇ g/ml) chase at 1, 4, 6, and 12 h was performed and the protein lysates were analyzed as described herein.
  • CHX cycloheximide
  • RNA isolated with TRIzol reagent was processed after DNase treatment (Ambion) directly to cDNA by reverse transcription using SuperScript First-Strand (Invitrogen).
  • Target sequences were amplified by qPCR using Power SYBR Green PCR Master Mix (Applied Biosystems).
  • FDXR primers were: forward, 3′-TCGACCCAAGCGTGCCCTTTG-5′ [SEQ ID No. 24]; reverse, 3′-GTGGCCCAGGAGGCGCAGCATC-5′ [SEQ ID No. 25]. Samples were normalized using Actin and GADPH genes.
  • Chemotherapeutic Drug Treatment Paclitaxel (Sigma) was dissolved in DMSO as a 10 mmol/liter stock solution and stored at ⁇ 80° C. Cisplatin (Sigma) was dissolved in water and freshly prepared before use. H1299 D1 and E1 cells were seeded (1 ⁇ 10 4 cells/well) in 96-well culture plates, PonA-induced, and after 24 h treated with paclitaxel (50, 100, and 500 ng/ml) or cisplatin (0.05, 0.1, and 0.2 mM).
  • H1299 D1 and E1 cells PonA-induced were incubated for 24, 48, and 72 h and assessed for viability with an MTS kit (Cell Titer 96® Aqueous MTS kit, Upstate Biotechnology, Lake Placid, N.Y.), as recommended by the manufacturer.
  • MTS kit Cell Titer 96® Aqueous MTS kit, Upstate Biotechnology, Lake Placid, N.Y.
  • the recombinant adenovirus carrying the wild-type FHIT cDNA was prepared as previously described (Ishii et al., 2001 Cancer Res 61:1578-1584).
  • a His-tagged FHIT cDNA was generated by PCR with the following oligonucleotides: 5′-ACgTggATCCCTgTgAggACATgTCgTTCAgATTTggC-3′ (forward) [SEQ ID NO: 26] and 5′-TTgTggATCCTTATCAgTgATggTgATggTgATgCgATCCTCTCTgAAAgTAgCCCgCAg-3′ [SEQ ID NO: 27].
  • Ad-His6 was generated with the AdenoVatorTM kit (Qbiogene, Carlsbad, Calif.), following the manufacturer's procedure.
  • Ad GFP used as control, was purchased from Qbiogene (Carlsbad, Calif.).
  • V5-tagged ferredoxin reductase cDNA For the preparation of a V5-tagged ferredoxin reductase cDNA, PCR amplification was performed with the same primer sequences with the exclusion of the FDXR physiological stop codon in the reverse primer. That is, both wild-type and V5-tagged ferredoxin reductase (FDXR) cDNAs were prepared by using as a template the wild-type coding sequence of the human ferredoxin reductase gene (GenBank Accession #NM — 024417). The ferredoxin reductase coding sequence was amplified from human brain cDNAs (Clontech).
  • the primers used to generate the V5-tagged FDXR cDNA were: Forward: 5′-CTgTTCCCAgCCATggCTTCgCgCTg-3′ [SEQ ID NO: 30]; and Reverse: 5′-gTggCCCAggAggCgCAgCATC-3′ [SEQ ID NO: 31]. It is to be noted that the oligonucleotide sequences are identical except for the reverse primer used for the generation of the V5-tagged cDNA, where the physiological stop codon of the FDXR was omitted in order to fuse in frame the FDXR coding sequence with a V5-tag.
  • the amplification products were subcloned into the pcDNA3.1/V5-HisTOPO TA vector (Invitrogen, Carlsbad, Calif.). Sequencing excluded mutations in the amplified products.
  • the adenovirus being capable of isolating Fhit-His6 comprises an adenovirus carrying a FHIT-His6 cDNA.
  • the FHIT-His6 cDNA can be prepared by using as a template the wild-type coding sequence of the human FHIT gene (GenBank Accession #NM — 002012).
  • a PCR amplification of the wild-type FHIT coding sequence was carried out with a reverse primer designed to abolish the physiological stop codon and to add to the endogenous FHIT sequence a stretch of 18 bp coding for six hystidines followed by an artificial stop codon.
  • both forward and reverse primers carried a BamHI restriction site for an easy subcloning.
  • the oligonucleotide sequences used for this amplification were the following: Forward: 5′-ACgTggATCCCTgTgAggACATgTCgTTCAgATTTggC-3′ [SEQ ID NO: 26]; and Reverse: 5′-TTgTggATCCTTATCAgTgATggTgATggTgATgCgATCCTCTCTgAAAgTAgACCCgCAg-3′ [SEQ ID NO: 27].
  • the PCR amplification product was sequenced to exclude random mutations before to be cloned in an Ad5 recombinant genome (AdenoVatorTM, a vector purchased by Qbiogene).
  • a method of isolating exogenously over-expressed Fhit-His6 includes using an adenovirus carrying a FHIT-His6 cDNA wherein the Fhit-His6 is isolated through the His tag.
  • Fhit-His6 represents the recombinant protein whose expression is driven into a mammalian cell through the Ad FHIT-His6 vector.
  • the His6 epitope allows for the recovery of the recombinant Fhit-His6 protein plus the protein complex interacting with the recombinant protein itself by taking advantage of the Ni-NTA system. This system is commercially available from Qiagen.
  • human A549 cancer cells were infected with Ad FHIT-His6; forty-eight hours after infection, photo-cross-linking of intracellular protein complexes was performed with the cross-linker dithiobis(succinimidyl propionate) [DSP] purchased from Pierce in order to stabilize protein complexes in living cells.
  • DSP cross-linker dithiobis(succinimidyl propionate)
  • Cells were disrupted in a protein extraction buffer and Fhit-His6 protein complex was isolated with the Ni-NTA magnetic-bead technology by taking advantage of the great affinity of the His6 tag for such beads. The isolated Fhit-His6 protein complex was then investigated through mass spectrometry to identify all proteins present in the complex.
  • a recombinant adenovirus carrying fragile histidine triad (Fhit) FHIT cDNA can be modified at its 3′ with a sequence encoding a histidine-six epitope tag (AdFHIT-His6).
  • a method for mediating an apoptotic process in at least one cell comprises exposing the cell to a fragile histidine triad (Fhit) gene product in an amount sufficient to mediate the apoptotic process in the cell.
  • Fhit fragile histidine triad
  • a method for inducing an apoptosis process in a cell comprises exposing the cell to a fragile histidine triad (Fhit) gene product in an amount sufficient to cause generation of reactive oxygen species (ROS) in the cell.
  • Fhit fragile histidine triad
  • a method for mediating an apoptotic process in at least one cell comprising: exposing the cell to a sufficient amount of fragile histidine triad (Fhit) gene product to allow the Fhit to enter mitochondria in the cell and to interact with Fdxr protein in the cell and to cause an increase in Fdxr protein level that is associated with generation of ROS, and causing a change in the apoptotic process in the cell.
  • Fhit fragile histidine triad
  • Fhit protein overexpression in Fhit-negative cancer cells is able to trigger programmed cell death (or apoptosis).
  • the inventors provide a rationale about the role of Fhit protein in the process of apoptosis.
  • FHIT gene therapy of cancer cells performed with Ad FHIT (at the multiplicity of infection 50 or MOI50, i.e., 50 viral particles per cell) is responsible of Fhit overexpression; then, the newly synthesized recombinant protein is taken by its interactors Hsp60/Hsp10 from the cytosol to the mitochondria where Fhit interacts with FDXR (ferredoxin reductase) a protein belonging to the respiratory chain. This interaction leads to the mitochondrial generation of ROS (Reactive Oxygen Species).
  • FDXR ferrredoxin reductase
  • ROS represent the early step for the initiation of the intrinsic (or mitochondrial) pathway of the apoptotic process; in fact, they induce a damage in the mitochondrial membranes that, in turn, release cytochrome c into the cytosol. This step is crucial for the execution of apoptosis, as cytochrome c contributes with other cytosolic molecules (i.e., Apaf-1 and pro-caspase 3) to the generation of the apoptosome, a multiprotein complex able to drive the cell, in a non-reversible fashion, to apoptosis.
  • cytochrome c contributes with other cytosolic molecules (i.e., Apaf-1 and pro-caspase 3) to the generation of the apoptosome, a multiprotein complex able to drive the cell, in a non-reversible fashion, to apoptosis.
  • a method commonly used to study apoptosis consists in the detection of mature caspase-3 (an indicator of incipient apoptosis) by flow cytometric analysis (Becton Dickinson) [for reference, see Trapasso et al., 2003, PNAS, 100, 1592-1597].
  • a method for preparing a V5-tagged ferredoxin reductase cDNA comprises PCR amplifying with primer sequences: 5′-CTgTTCCCAgCCATggCTTCgCgCTg-3′ (forward) [SEQ ID NO: 28], and 5′-TCAgTggCCCAggAggCgCAgCATC-3′ [SEQ ID NO: 29] and subcloning the amplification products pcDNA3.1/V5-HisTOPO TA vector.
  • Both wild-type and V5-tagged ferredoxin reductase (FDXR) cDNAs were prepared by using as a template the wild-type coding sequence of the human ferredoxin reductase gene (GenBank Accession #NM — 024417).
  • the ferredoxin reductase coding sequence was amplified from human brain cDNAs (Clontech). That is, the primers used to amplify the wild-type FDXR cDNA were: Forward: 5′-CTgTTCCCAgCCATggCTTCgCgCTg-3′ [SEQ ID NO: 28]; Reverse: 5′-TCAgTggCCCAggAggCgCAgCATC-3′ [SEQ ID NO: 29].
  • the primers used to generate the V5-tagged FDXR cDNA were: Forward: 5′-CTgTTCCCAgCCATggCTTCgCgCTg-3′ [SEQ ID NO: 30], and Reverse: 5′-gTggCCCAggAggCgCAgCATC-3′ [SEQ ID NO: 31]. Note that the oligonucleotide sequences are identical except for the reverse primer used for the generation of the V5-tagged cDNA, where the physiological stop codon of the FDXR was omitted in order to fuse in frame the FDXR coding sequence with a V5-tag.
  • a method for generating a recombinant adenovirus comprises preparing a recombinant adenovirus carrying the wild-type FHIT cDNA (AdFHIT); and generating a His-tagged FHIT cDNA using PCR with the following oligonucleotides: 5′-ACgTggATCCCTgTgAggACATgTCgTTCAgATTTggC-3′ (forward) [SEQ ID NO: 26], and 5′-TTgTggATCCTTATCAgTgATggTgATggTgATgCgATCCTCTCTgAAAgTAgACCCgCAg-3′ [SEQ ID NO: 27].
  • the FHIT-His6 cDNA was prepared as described herein.
  • the recombinant adenoviral vector Ad FHIT-His6 was prepared according to the manufacturer's suggestions (Qbiogene). Briefly, the amplified FHIT-His6 PCR fragment was digested with BamHI and subcloned into the BamHI linearized transfer vector pAdenoVator-CMV5-IRES-GFP. The pAdenoVator-CMV5-IRES-GFP/FHIT-His6 was co-transfected with the E1/E3 deleted Ad5 backbone viral DNA into 293 cells.
  • Viral plaques were screened for the presence of the Fhit-His6 protein by Western blot with specific penta-His antibodies (Qiagen).
  • One positive clone was plaque-purified and amplified on 293 cells. After freeze/thaw cycles, the adenoviruses in the supernatant were purified on two successive cesium chloride gradients. The recombinant adenovirus was titered by the TCID50 method and aliquoted. Virus stocks were stored at ⁇ 80° C. Finally, the recombinant adenovirus carrying the wild-type FHIT cDNA (Ad FHIT) was previously generated by Trapasso et al. 2003, PNAS, 100, 1592-1597.
  • Mitochondrial localization studies Confocal microscopy was used to assess Fhit protein distribution by immunofluorescence; H1299 D1 cells, with inducible FHIT cDNA, and E1 cells, with empty vector, were treated with PonA for 48 hr, and living cells were stained with Mitotracker Red 580 (M-22425, Molecular Probes, Eugene, Oreg.) at a working concentration of 500 nM for 40 min under growth conditions. The cells were fixed and permeablilized by incubation in ice-cold acetone for 5 min and then washed in PBS.
  • Mitotracker Red 580 M-22425, Molecular Probes, Eugene, Oreg.
  • A549 cells infected with AdHis6 or AdFHIT, MOI 5 were fixed in 4% paraformaldehyde in PBS pH7.2 for 30 min at 4° C., washed 3 times with PBS, and remaining free aldehyde groups were reduced using a 30 min incubation in 0.05% sodium borohydride in PBS. Following a PBS wash, samples were blocked with 50 mM glycine in PBS for 30 min, washed twice in PBS, and dehydrated with 25 and 50% ethanol for 15 min each, followed by 3 changes of 70% ethanol for 15 min each.
  • the samples were then infiltrated with 70% ethanol+LR White resin, hard grade (Electron Microscopy Sciences, Hatfield, Pa.) at 2:1 for 1 hr, 70% ethanol+LR White at 1:2 for 1 hr, 100% LR White for 1 hr and 100% LRW overnight at 4° C.
  • 900 nm thin section s were cut using a Reichert UCT Ultramicrotome and a diamond knife and placed on nickel grids.
  • the grids were floated section side down on drops of PBS for 5 min, 5% goat serum in PBS for 1 hr at room temperature (RT), and either Penta-His mouse monoclonal antibody at 20 mg/ml (Qiagen, Valencia, Calif.) diluted in PBS containing 0.1% BSA and 0.05% Tween-20 (BSA/Tw) or BSA/Tw alone overnight at 4° C. in a humidified chamber.
  • BSA/Tw 0.1% BSA and 0.05% Tween-20
  • BSA/Tw BSA/Tw alone overnight at 4° C. in a humidified chamber.
  • the following day the grids were washed 6 ⁇ for 5 min each using PBS and then incubated with h goat anti-mouse 10 nm colloidal gold conjugate (Ted Pella, Redding, Calif.) diluted 1:10 in BSA/Tw for 2 hrs at RT.
  • the grids were washed 6 ⁇ each for 5 min with PBS, rinsed with DI H2O and post-stained with 2.5% aqueous uranyl acetate for 3 min. Images were collected on a Tecnai 12 electron microscope equipped with a US1000 Gatan 2K digital camera.
  • Mobile phase A was H2O/acetonitrile/formic acid (FA)/TFA 97.9:2:0.08:0.02 (v/v/v/v); mobile phase B was H2O/acetonitrile/FA/TFA 4.9:95:0.08:0.02 (v/v/v/v).
  • MS detection was performed on an Applied Biosystems (Framingham, Mass.) QSTAR XL hybrid LC-MS/MS operating in positive ion mode, with nanoelectrospray potential at 1800 V, curtain gas at 15 units, CAD gas at 3 units.
  • Information-dependent acquisition was performed by selecting the two most abundant peaks for MS/MS analysis after a full TOF-MS scan from 400 to 1200 m/z lasting 2 seconds. Both MS/MS analyses were performed in enhanced mode (2 seconds/scan).
  • MS/MS spectra were searched by interrogating the Swiss Prot database on the Mascot search engine (matrixscience.com) (accessed on June 2006). The following search parameters were used. MS tolerance: 50 ppm; MS/MS tolerance: 1 Da; methionine oxidized (variable modification); cysteine carbamidomethylated (fixed modification); enzyme: trypsin; max. missed cleavages: 1.
  • Protein lists obtained from, respectively, both A549 infected with Ad FHIT-His6 and control were compared, and proteins exclusively present in the A549-Ad FHIT-His6 list were kept for further validation.
  • LC-MS/MS raw data were inspected using selected ion chromatogram (SIC) displaying mode.
  • SIC comparison it could be assessed the exclusive presence of the peptides of interest, identified as belonging to the six candidate proteins under examination, in the Ad FRIT-His6 sample.
  • Such verification step already provided rather strong evidence for the specific capture of the six candidate protein.
  • these findings were further validated by biochemical and functional assays.

Abstract

Provided herein are methods and compositions for the diagnosis, prognosis and treatment of a cancer associated disorders using the Fhit gene.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • The present invention is a divisional application of U.S. Ser. No. 12/739,541, filed Feb. 25, 2011, now allowed, which claims the benefit of PCT application No. PCT/US/2008/081294 filed Oct. 27, 2008, which claims priority to claims the benefit of the provisional patent application Ser. No. 61/000,480 filed Oct. 26, 2007.
  • AND STATEMENT REGARDING SPONSORED RESEARCH
  • This invention was made with government support under NCI Grant Nos. CA77738 and CA78890. The government has certain rights in this invention.
  • BACKGROUND OF THE INVENTION
  • The FHIT gene encompasses the most active common fragile site at chromosome 3p14.2. Fhit expression is lost or reduced in a large fraction of most types of human tumors due to allelic loss, genomic rearrangement, promoter hypermethylation, or combinations thereof. Fhit knock-out mice show increased susceptibility to cancer development and FHIT gene therapy prevents tumors in carcinogen-exposed Fhit-deficient mice. Fhit restoration by stable transfection in cancer cells has little effect in vitro, unless cells are exposed to stress, including the stress of the nude mouse environment in vivo; viral-mediated Fhit restoration, a process that simultaneously supplies stress and Fhit expression, suppresses tumorigenesis in vivo and triggers apoptosis of many types of malignant cells in vitro, including lung cancer cells.
  • In lung hyperplastic lesions, DNA damage checkpoint genes are already activated, leading to selection for mutations in checkpoint proteins and neoplastic progression. Evidence of DNA alteration at FRA3B within FHIT accompanied the hyperplasia and checkpoint activation. Loss of FHIT alleles occurs in normal appearing bronchial epithelial cells of smokers, prior to pathologic changes or alterations in expression of other suppressor genes.
  • Fhit expression is down-regulated by exposure to DNA damaging agents and Fhit plays a role in response to such agents), with Fhit-deficient cells escaping apoptosis and accumulating mutations.
  • Although Fhit expression triggers apoptosis in several experimental models through caspase-dependent mechanisms involving extrinsic and intrinsic apoptotic pathways, little is known about early events in this process and how Fhit loss is involved in tumor initiation.
  • Therefore, there is a need for methods for altering the expression of FHIT in subjects in need thereof. There is also a need for compositions that are useful to alter the expression of FHIT in subjects in need thereof.
  • SUMMARY OF THE INVENTION
  • In a broad aspect, there is provided methods which identify proteins that interact directly with Fhit to effect downstream signal pathways culminating in apoptosis. In one embodiment, proteins within cells were chemically cross-linked after infection of lung cancer cells with AdFHIT-His6 virus. The proteins linked to Fhit and pathways affected by them were identified and characterized.
  • In another broad aspect, there is provided herein a method of diagnosing whether a subject has, or is at risk for developing, a cancer associated disorder, comprising measuring the level of at least fragile histidine triad (Fhit) gene in a test sample from the subject, wherein an alteration in the level of the Fhit gene product in the test sample, relative to the level of a corresponding Fhit gene product in a control sample, is indicative of the subject either having, or being at risk for developing, a cancer associated disorder.
  • Various objects and advantages of this invention will become apparent to those skilled in the art from the following detailed description of the preferred embodiment, when read in light of the accompanying drawings.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawings will be provided by the Office upon request and payment of the necessary fee.
  • FIGS. 1A-1H—Subcellular localization of Fhit protein in cytosol and mitochondria:
  • FIG. 1A, immunofluorescence microscopy was performed with anti-Fhit serum on H1299 cells (D1) treated with PonA for 48 h; Fhit staining was detected using fluorescein isothiocyanate (green)-conjugated anti-rabbit immunoglobulin (IgG); Mito-Tracker Red staining, which identifies mitochondria, shows partial colocalization with Fhit. The yellow color on the fourth panel (lower right) shows the co-localizations points.
  • FIG. 1B, immunoelectron microscopy of A549 AdFHIT (left) or AdFHIT-His6-infected cells (right) performed with a penta-His antibody shows Fhit mitochondrial localization (right); A549 cells infected with AdFHIT served as control and show only a few scattered grains (left panel).
  • FIG. 1C, immunoblot analysis of AdFHIT-infected A549 subcellular fractions using anti-Fhit indicates Fhit protein distribution in the cytosol, membranes, cytoskeleton, and mitochondria.
  • FIG. 1D, immunoblot analysis of proteins from mitochondria of A549 cells infected with AdFHIT-His6 after treatment with sodium carbonate (FIG. 1E) and increasing concentrations of digitonin (FIG. 1F) indicates that Fhit is mainly distributed in mitochondrial matrix; filters were probed with Fhit and CoxIV antisera; lanes in FIG. 1F represent supernatants after treatment with 0, 0.10, 0.15, and 0.20% digitonin.
  • FIG. 1G, immunoblot analyses of subcellular fractions from MKN74/E4 and MKN74/A116 cells (stably expressing exogenous Fhit), and FIG. 1H, HCT116 (an endogenous Fhit-positive colon cancer cell line) using anti-Fhit, confirms Fhit mitochondrial localization; GAPDH and CoxIV antisera served as controls.
  • FIGS. 2A-F—Exogenous and endogenous Fhit forms a complex with endogenous Hsp60, Hsp10, and Fdxr proteins:
  • FIGS. 2A, 2B, 2C, Protein complexes, isolated with recombinant Fhit-His6 protein, were separated on polyacrylamide gels and probed with antisera against Hsp60 (FIG. 2A), Hsp10 (FIG. 2B), and Fdxr (FIG. 2C); in the latter panel, prepared after mitochondria isolation, it is shown that Fhit recruits Fdxr in the mitochondria in a time-dependent manner. Filters were loaded with protein isolated after infection of A549 cells with AdFHIT-His6 with or without DSP.
  • FIG. 2D, coimmunoprecipitation with anti-Hsp60 after infection of A549 cells with AdFHIT; filters were probed with Hsp60, Fhit, and Hsp10 antisera.
  • FIG. 2E, A549 cells were co-transfected with the V5-tagged FDXR gene and FHIT plasmids; immunoprecipitation was with anti-V5 and detection with Fdxr and Fhit antisera.
  • FIG. 2F, immunoprecipitation and immunoblot detection of endogenous interactor proteins (Fdxr and Hsp10) from DSP-treated Fhit-positive HCT116 cells. Filters were probed with antisera against each target protein. Endogenous Fhit co-precipitated with Hsp10 and Fdxr.
  • FIGS. 3A-D—Knockdown of Hsp60/Hsp10 reduces the level of Fhit in the mitochondria:
  • FIG. 3A, nickel-H6 pull down experiment of A549 cells AdFHIT-His6 infected on subcellular fractions (Cytosol and Mitochondria) using H6 antibody; lysates were incubated with nickel beads to isolate the DSP cross-linked Fhit-His6 protein complex and loaded on a 4-20% polyacrylamide gel. 24 h after infection, the Hsp60-Fhit complex was present in both compartments; 48 h after infection, the complex was detectable again in both compartments and the increase of Fhit complex proteins appears related to the increase of Fhit protein at 48 h after AdFHIT-His6 infection, with a slight increase in the mitochondria (densitometry analysis on input samples was performed).
  • FIG. 3B, immunoblot analysis of Hsp60, Hsp10, Fhit, and GAPDH in Fhit-positive D1 cells after 72 h of Hsp60/Hsp10 silencing showing Fhit, Hsp60, and Hsp10 levels after a CHX chase (30 μg/ml) for 1-12 h.
  • FIG. 3C, immunoblot analysis of cytosol/mitochondrial protein fractions of A549 cells 72 h after transfection with Hsp60 and Hsp10 siRNAs and 24 h after AdFHIT infection at m.o.i. 1, with Hsp60, Hsp10, Fhit, GAPDH, and CoxIV antisera. Hsp60/10 silencing does not appear to affect the Fhit cytosolic level, but is associated with a decrease of Fhit in the mitochondrial fraction. Scrambled (Scr) siRNAs were used as controls.
  • FIG. 3D, subcellular fractionation and immunoprecipitation of “endogenous” Fhit complex proteins. PonA-induced D1 and E1 cells, with and without peroxide treatment, were fractionated into cytosol and mitochondria and subcellular fractions assessed for the presence of Fhit and interactors (left side) at 48 h after induction; 25 μg of proteins were loaded per lane. Endogenous Hsp60 co-precipitated Fhit and Fdxr.
  • FIGS. 4A-F—Fhit expression induces intracellular ROS generation after treatment of cells with peroxide:
  • FIG. 4A, fluorescence-activated cell sorter (FACS) analysis for ROS assessment in A549 cells 48 h after transfection with FHIT plasmid, with and without a 5-h H2O2 treatment. Empty vector-transfected cells served as control. Intracellular superoxide was determined according to the fluorescence of ethidium as a result of oxidation of hydroethidine by O2. M2 refers to the fraction of ROS positive cells.
  • FIG. 4B, FACS analysis for ROS assessment by the fluorescence produced from the oxidation of hydroethidine in D1 and E1 cells; 48 h after PonA treatment, cells were treated for 5 h with 0.5 and 1.0 mM H2O2 and oxidative stress was measured; % positive refers to the fraction of fluorescent cells, indicating ROS. These experiments were repeated three times with similar results.
  • FIG. 4C, increased green fluorescent DCF signal in H1299 Fhit-expressing cells (D1) under stress conditions. Cells were incubated with 2′,7′-dichlorodihydrofluorescein diacetate, a ROS indicator that can be oxidized in the presence of ROS to the highly green fluorescent dye DCF, at 48 h after Fhit induction and after a 5-h H2O2 treatment of E1 and D1 cells (magnification ×40).
  • FIG. 4D, MTS cell viability assays were performed on E1 and D1 cells. Cells were treated with PonA for 48 h and then with increasing concentrations of H2O2 (0.125, 0.25, and 0.5 mM) for 4 h. Analysis was at 24 h after H2O2 treatment. Columns report the average of four experiments±S.E. Each point was measured in quadruplicate and standard deviation calculated; p<0.05 was considered significant.
  • FIG. 4E, FACS analysis of D1 and E1 cell cycle kinetics at 48 h after oxidative stress treatment. Cells were treated with PonA for 48 h and then with increasing concentrations of H2O2 (0.25 and 0.5 mM) for 4 h. Analysis was at 48 h after H2O2 treatment. All experiments were performed twice in triplicate.
  • FIG. 4F, colony formation assay of H1299/D1 and H1299/E1 cells after 5 mM PonA stimulation and a 5-h H2O2 treatment at the indicated concentrations.
  • FIGS. 5A-H.—Apoptosis triggered by Fhit viral transduction can be mediated by its interaction with Fdxr:
  • FIG. 5A, immunoblot analysis with antisera against Fdxr, Fhit, and GAPDH. Proteins were extracted from E1 (control) and D1 cells 48 h after treatment with PonA.
  • FIG. 5B, immunoblot analysis of Fdxr expression in D1 and E1 cells after a 4-h treatment with 25 μM MG132, a proteasome inhibitor. GAPDH detection shows equal protein loading.
  • FIG. 5C, immunoblot analysis of Fdxr, Fhit, and GAPDH in D1, expressing Fhit, and E1 cells, showing Fdxr level after CHX chase (30 μg/ml) for 4-12 h. Densitometry based on GAPDH levels shows enhanced stability of Fdxr in the presence of Fhit.
  • FIG. 5D, FACS analysis of FDXR+/+/+ and FDXR+/−/− cell cycle kinetics after infection with AdFHIT m.o.i. 50 and 100. The experiment was performed 48 h after infection and was repeated three times with similar results. Profiles of AdGFP-infected cells were similar to those of non-infected cells (not shown).
  • FIG. 5E, immunoblot analysis showing expression of Fdxr, Fhit, and GAPDH after infection of FDXR+/+/+ and FDXR+/−/− with AdFHIT m.o.i. 50 and 100. Proteins were extracted 48 h after infection.
  • FIG. 5F, real-time RT-PCR analysis for FDXR expression at 48 h after AdFHIT m.o.i. 50. The PCR product was normalized to GADPH and Actin expression and each point was repeated in quadruplicate; differences between control and Fhit positive samples were not significant.
  • FIG. 5G, caspase 3 and Parp1 activation. Immunoblot analysis, using Fhit, caspase 3, Parp1 antisera, of total cell lysates from HCT116 FDXR+/+/+ cells 48, 72, and 96 h after infection with AdFHIT and AdGFP at m.o.i. 50. GAPDH and CoxIV served as internal protein markers.
  • FIG. 5H, immunoblot analysis, using Fhit and cytochrome c antisera, of cytosol/mitochondria fractions from HCT116 FDXR+/+/+ cells 48, 72, and 96 h after infection with AdFHIT and AdGFP at m.o.i. 50. GAPDH and β-actin served as internal protein markers.
  • FIGS. 6A-E—Fhit enhances the sensitivity of cancer cells to paclitaxel and cisplatin:
  • FIGS. 6A, 6B, MTS assays performed on E1 and D1 cells. Cells were treated with PonA for 48 h and then treated with paclitaxel (50-500 ng/ml) (FIG. 6A) or cisplatin (0.05-0.2 mM) (FIG. 6B) for 24 or 48 h. Bars report the average of four experiments±S.E. Each point was measured in quadruplicate and standard deviation calculated; asterisks next to brackets in FIG. 6A and FIG. 6B indicate statistically significant differences in drug response of D1 and E1 cells, p<0.05.
  • FIG. 6C, FIG. 6D, the graphs show representative results of flow cytometry analyses of E1 and D1 cells. Cells were treated with PonA for 48 h and then with paclitaxel (50-500 ng/ml) (FIG. 6C) or cisplatin (0.05-0.2 mM) (FIG. 6D). Each data point was measured in triplicate at 24, 48, and 72 h (data shown for 48 h).
  • FIG. 6E, caspase 3 and Parp1 cleavage: immunoblot analyses, using Fhit, caspase 3, and Parp1 antisera, of total cell lysates from PonA-induced D1 cells after 48 h of treatment with paclitaxel (50 and 100 ng/ml) or cisplatin (0.05 and 0.1 mM). GAPDH served as loading control.
  • FIG. 7—TABLE 1 Candidate Fhit protein partners isolated through mass spectrometry. Proteins selectively captured in the A549 AdFHIT-H6-infected cells sample. Amino acid sequence of identified peptides, Mascot scores, and protein sequence coverage are listed.
  • FIGS. 8A-8C—Ad-His6 biological activity is comparable to AdFHIT:
  • FIG. 8A, Western blot analysis of A549 cells infected with Ad-His6, MOI 20. Fhit-His protein was detected by antipentaHis and antiFhit serum. Both Ad FHIT and Ad-His6 carry a GFP cDNA regulated by a CMV5 promoter through an internal ribosome entry sequence downstream of FHIT. γ-tubulin was used to normalize sample loading.
  • FIG. 8B, How cytometry analysis of A549 cells 96 hr after infection with Ad-His6, MOI 15. Upper panel indicates the subG1 DNA content of infected cells (experiment repeated thrice; average values of subG1 fractions 22%+/−4.3 for Ad FHIT, 29%+/−5 for Ad-His6; the difference is not statistically significant; lower panel shows percentages of cells with mature caspase-3, an indication of apoptosis. The extent of cell death in A549 cells infected with Ad-His6 is comparable to the result obtained after infection with Ad FHIT.
  • FIG. 8C, In vivo cross-linking of Fhit-His6. Silver staining of gel with cell lysates after His6 pull down and cross-link reversal conditions, separated by 4-20% gradient SDS-PAGE. Internal negative controls included His6 pull down of Ad FHIT infected cells (cross-linked, CL) and Ad-His6 infected cells (not cross-linked, NT).
  • FIGS. 9A-9F. Initial validation of candidate Fhit protein partners identified through nanobore LC-MS/MS. Selected ion chromatograms (SIC) for AdFHIT-His6 and control samples are shown. The six SICs pairs report ion currents of the six following m/z values: 1) 672.8 (peak at retention time 30 min. was identified as tryptic peptide TVIIEQSWGSPK [SEQ ID NO: 5] belonging to Hsp60), 2) 685.4 (peak at retention time 32 min. identified as tryptic peptide LGPALATGNVVVMK [SEQ ID NO: 22] belonging to Aldh2), 3) 617.3 (peak at retention time 39 min. identified as tryptic peptide IFGVTTLDIVR [SEQ ID NO: 10] belonging to Mdh), 4) 658.4 (peak at retention time 26 min. identified as tryptic pep tide VLQATVVAVGSGSK [SEQ ID NO: 19] belonging to Hsp10), 5) 551.7 (peak at retention time 28 min. identified as tryptic peptide EIDGGLETLR [SEQ ID NO: 15] belonging to Etfb), 6) 598.3 (peak at retention time 23 min. identified as tryptic peptide FGVAPDHPEVK [SEQ ID NO: 23] belonging to Fdxr). Peptides of interest, indicated by red arrows, are exclusively present in Ad-His6 sample.
  • FIG. 10—TABLE 2, Fhit induces generation of ROS in MKN74 gastric cancer cells. ROS assessment was performed with MKN74A116, a human gastric cancer cell line carrying a p53 mutant allele and expressing exogenous Fhit; Fhit-negative MKN74E4 cells were used as a control. To induce ROS generation, we treated MKN74 cells for 5 hr with 0.5, 1.0 and 2.0 mM H2O2 Results indicate a significantly higher rate of ROS generation in cells expressing exogenous Fhit compared to controls; toxicity was observed in Fhit-expressing cells after 2 mM H2O2 treatment. Numbers report the average of four experiments±S.E.
  • DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT
  • It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not intended to limit the scope of the current teachings. In this application, the use of the singular includes the plural unless specifically stated otherwise.
  • The use of the word “a” or “an” when used in conjunction with the term “comprising” in the claims and/or the specification may mean “one,” but it is also consistent with the meaning of “one or more,” “at least one,” and “one or more than one.”
  • Also, the use of “comprise”, “contain”, and “include”, or modifications of those root words, for example but not limited to, “comprises”, “contained”, and “including”, are not intended to be limiting. The term “and/or” means that the terms before and after can be taken together or separately. For illustration purposes, but not as a limitation, “X and/or Y” can mean “X” or “Y” or “X and Y”.
  • The term “combinations thereof” as used herein refers to all permutations and combinations of the listed items preceding the term. For example, “A, B, C, or combinations thereof” is intended to include at least one of: A, B, C, AB, AC, BC, or ABC, and if order is important in a particular context, also BA, CA, CB, ACB, CBA, BCA, BAC, or CAB.
  • As used herein interchangeably, “gene product,” “DNA” and “gene,” are used herein interchangeably.
  • The following abbreviations may be used herein: GAPDH, glyceraldehyde-3-phosphate dehydrogenase; DSP, dithiobis(succinimidyl propionate); LC-MS/MS, liquid-chromatography tandem mass spectrometry; Fdxr, ferredoxin reductase; PonA, ponasterone A; m.o.i., multiplicity of infection; ROS, reactive oxygen species; FU, 5-fluorouracil; DCFH-DA, dichlorofluorescein-diacetate; DCF, 2′,7′-dichlorofluorescein; CHX, cycloheximide; siRNA, small interfering RNA; RT, reverse transcriptase; MTS, 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium.
  • The section headings used herein are for organizational purposes only and are not to be construed as limiting the described subject matter in any way. All literature and similar materials cited in this application, including patents, patent applications, articles, books, treatises, and internet web pages are expressly incorporated by reference in their entirety for any purpose. In the event that one or more of the incorporated literature and similar materials defines or uses a term in such a way that it contradicts that term's definition in this application, this application controls.
  • Fhit protein is lost in most cancers, its restoration suppresses tumorigenicity, and virus-mediated FHIT gene therapy induces apoptosis and suppresses tumors in preclinical models. Protein cross-linking and proteomics methods are used to characterize a Fhit protein complex involved in triggering Fhit-mediated apoptosis. The complex includes Hsp60 and Hsp10 that mediate Fhit stability and may affect import into mitochondria, where it interacts with ferredoxin reductase, responsible for transferring electrons from NADPH to cytochrome P450 via ferredoxin. Viral-mediated Fhit restoration increases production of intracellular reactive oxygen species, followed by increased apoptosis of lung cancer cells under oxidative stress conditions; conversely, Fhit-negative cells escape apoptosis, carrying serious oxidative DNA damage that may contribute to an increased mutation rate. Characterization of Fhit interacting proteins has identified direct effectors of the Fhit-mediated apoptotic pathway that is lost in most cancers through loss of Fhit.
  • Earlier searches for Fhit-interacting proteins pointed to several candidate proteins, none of which we could confirm as interactors by co-immunoprecipitation experiments, including Ubc9, α-tubulin, and Mdm2. To readdress the question of Fhit protein interactors, the following was used: adenovirus-transduced Fhit-His6 for Fhit complex purification after cross-linking, and Fhit-linked proteins, Hsp60, Hsp10, and Fdxr, were identified; subcellular location of these proteins suggested that mitochondria might be foci of Fhit activity. Hsp “stress proteins” as molecular chaperones perform functions such as protein translocation, folding, and assembly. The finding that Fhit interacts with Hsp60/Hsp10 after AdFHIT infection suggests that the Hsp complex may be important for Fhit stability, and possibly for its correct folding to import it into mitochondria, prior to activation of the apoptotic pathway, a suggestion we investigated by knocking down expression of Hsp60, Hsp10, or both in AdFHIT-infected lung cancer cells; Fhit stability was assessed after CHX chase in H1299 D1 cells, the lung cancer cell line expressing inducible Fhit. The level of Fhit protein in isolated mitochondria after knockdown of both Hsp60 and -10 was reduced, strengthening the proposal that Fhit-Hsp60/10 interaction is involved in Fhit stabilization and/or in correct folding for importation into mitochondria.
  • Targeted disruption of the FDXR gene in HCT116 colon cancer cells showed that it was essential for viability; reduction of the gene copy number resulted in decreased sensitivity to 5-fluorouracil-induced apoptosis and FDXR is a target gene of the p53 family. Overexpression of Fdxr-sensitized colon cancer cells to H2O2, 5-fluorouracil, and doxorubicin-induced cell death, indicating that Fdxr contributes to p53-mediated apoptosis through generation of oxidative stress in mitochondria. Thus, activated p53 induces apoptosis in response to cellular stresses in part through ROS, and simultaneously p53 increases transcription of the FDXR gene, which in turn enhances p53 function by increasing ROS-induced apoptosis.
  • Now shown herein is the presence of Fhit in the mitochondrial fraction; when Fhit is overexpressed or Fhit-expressing cells are stressed, Fhit can protect Fdxr from proteosomal degradation, leading to an increase in the Fdxr protein level, which is associated with generation of ROS and followed by apoptosis. Fhit does not affect the FDXR transcriptional level but may affect stability of the protein. In H1299 cells, missing both Fhit and p53, Fdxr overexpression increases sensitivity to ROS-induced cell death, and H1299 cells expressing inducible Fhit or p53 are sensitive to ROS-induced cell death; cancer cells missing Fhit, p53, or both would lack ways to increase Fdxr expression, and would be less sensitive to oxidative damage and would survive.
  • Discovery of the mitochondrial function of Fhit in apoptosis through interaction with Fdxr now extends functional parallels of the important tumor suppressors, Fhit and p53, lost sequentially in most cancers and involved in response to DNA damage, and illuminates their differences, with p53 acting as a transcriptional and Fhit a post-transcriptional Fdxr regulator. Delineation of direct downstream effectors of the Fhit suppressor pathway will lead to mechanistic studies of Fhit function that may influence preventive and therapeutic strategies to activate the Fhit pathway.
  • The finding that ROS generation is crucial for Fhit-mediated apoptosis emphasizes the importance of Fhit loss as a negative prognostic factor in various clinical settings; for example, assessment of Fhit status in preneoplastic or neoplastic conditions may be predictive of responses to antioxidant treatments.
  • To identify proteins that interact with Fhit to effect downstream apoptotic pathways, the inventors herein cross-linked proteins within cells after viral-mediated Fhit overexpression in lung cancer cells, and characterized proteins associated with Fhit and the pathways affected by them.
  • Results.
  • Isolation of a Fhit Protein Complex—To identify Fhit-interacting proteins, we generated an adenovirus carrying FHIT cDNA modified at its 3′ end with a sequence encoding a His6 epitope tag (AdFHIT-His6). The biological activity of this tagged Fhit protein expressed in A549 cells was comparable with wild-type Fhit activity (FIG. 8).
  • A549 lung cancer-derived cells, which are susceptible to Fhit-induced apoptosis (10), were infected with AdFHIT or AdFHIT-His6 and treated with DSP, a cross-linker that crosses membranes and fixes proteins in complex in vivo. Cells were lysed and proteins isolated with nickel beads avid for the His6 epitope tag. Purified proteins were treated with dithiothreitol to cleave DSP and dissociate the complex, and digested by trypsin; protein constituents were identified by LC-MS/MS (FIG. 7—Table 1 and FIG. 9).
  • Six proteins were identified, all with mitochondrial localization: Hsp60 and 10, ferredoxin reductase (Fdxr), malate dehydrogenase, electron-transfer flavoprotein, and mitochondrial aldehyde dehydrogenase 2; Hsp60 and Hsp10 are also distributed in the cytosol.
  • Fhit Subcellular Localization—Because candidate Fhit interactors are mitochondrial proteins, the inventors herein determined if Fhit, which lacks a mitochondrial localization signal, localized in these organelles. Fhit negative H1299 lung cancer cells carrying an inducible FHIT cDNA (D1 cells) were treated with the inducer, PonA for 48 h and indirect immunofluorescence detection of Fhit subcellular location was assessed using anti-Fhit serum and MitoTracker Red 580, a marker of mitochondria; Fhit fluorescent signal (green staining, FIG. 1A) was cytoplasmic and partly co-localized (yellow staining, FIG. 1A, lower right) with MitoTracker Red dye, indicating that exogenous Fhit localized to mitochondria and cytosol. Anti-Fhit specificity was confirmed by absence of green fluorescence in the Fhit negative H1299 clone E1 cells (not shown). To confirm mitochondrial localization, A549 cells infected with AdFHIT-His6 or AdFHIT at m.o.i. 20 were examined by immunoelectron microscopy 48 h later, by anti-pentaHis staining; FhitHis6-transduced cells demonstrated significant numbers of gold particles in mitochondria (FIG. 1B, right panel), whereas AdFHIT-transduced cells showed sparse reactivity (FIG. 1B, left panel).
  • To assess Fhit submitochondrial localization, mitochondria were purified from A549 cells infected with AdFHIT m.o.i. 1, as described above. The sodium carbonate procedure is a nondestructive approach that allows effective release in the supernatant of both soluble proteins and peripheral membrane proteins from intracellular membranes after inducing the generation of open sheets of membranes; furthermore, it allows recovery of integral proteins with the membranes (pellet)
  • FIG. 1E shows that Fhit was only detectable in the soluble fraction. To further define Fhit submitochondrial localization, mitochondria were treated with 0.10 and 0.15% digitonin to selectively disrupt mitochondrial outer membrane, releasing proteins contained in the intermembrane space and the matrix; as shown in FIG. 1F, gradual disruption of outer and inner membranes releases increasing amounts of Fhit protein, suggesting that Fhit is mainly distributed either at the luminal side of the inner membrane or in the matrix of mitochondria. Mitochondrial localization was confirmed in gastric cancer-derived MKN74A116 cells stably expressing exogenous Fhit and in HCT116 colon cancer cells expressing endogenous Fhit (FIG. 1G and FIG. 1H).
  • Fhit Interacts with Hsp60, Hsp10, and Fdxr—Among candidate interactor proteins, the inventors focused on Hsp60 and Hsp10 as possible chaperonins and on Fdxr, a mitochondrial respiratory chain protein transactivated by p53 and involved in responses to therapeutic drugs. To validate interactions, A549 cells were infected with AdFHIT or AdFHIT-His6 at m.o.i. 20, with or without DSP. Fhit complexes were purified through the His6 tag and co-purified proteins were detected with antisera against Hsp60, Hsp10, and Fdxr; Hsp60 and Fdxr were detected only in lysates of cells exposed to DSP (FIG. 2A and FIG. 2C), whereas Hsp10 was also detectable without cross-linking (FIG. 2B).
  • A time course experiment after infection showed recruitment of Fdxr by Fhit (FIG. 2C); also, endogenous Hsp60 co-immunoprecipitated Fhit and Hsp10 in the absence of DSP (FIG. 2D).
  • To verify specificity of interactions we generated an FDXR cDNA expression plasmid with a 3′ V5 epitope tag. A549 cells were co-transfected with FDXR-V5 and FHIT plasmids, and proteins were precipitated with monoclonal anti-V5; co-precipitated Fhit was detectable only after DSP cross-linking (FIG. 2E).
  • To determine whether these proteins also interact with endogenous Fhit, the inventors immunoprecipitated each endogenous candidate interactor protein from DSP-treated Fhit-positive HCT116 cells and looked for co-precipitation of endogenous Fhit (FIG. 2F).
  • Endogenous Fhit co-precipitated with Hsp10 and Fdxr, confirming the presence of endogenous Fhit in mitochondria and its interaction with endogenous chaperones and respiratory chain protein in the absence of stress.
  • Hsp60/10 Interaction Affects Fhit Stability and/or Mitochondrial Import—Hsp60 and -10 are molecular chaperones found in complex and may be important for folding and import of proteins into mitochondria. The inventors herein now believe that the Hsp60/10 complex was responsible for Fhit correct folding and mitochondrial addressing.
  • To investigate the location of these interactions, A549 cells were infected with AdFHIT-His6 m.o.i. 5 and protein lysates were collected from cytosol and mitochondrial fractions after cross-linking. Complexes were isolated by Fhit-H6-nickel pull down, separated on a polyacrylamide gel, and filters probed with Hsp60 and Fhit antisera. At 24 and 48 h after infection interaction with Hsp60 is observed in the cytosol and mitochondria (FIG. 3A) commensurate with the increase in Fhit expression at these times (Input), as shown in FIG. 3A.
  • To determine whether the Fhit-Hsp60/10 interaction is important for the stability of the Fhit protein, H1299 with inducible Fhit expression (D1 cells) were transfected with Hsp60 and Hsp10 siRNAs and 72 h after transfection a CHX chase was performed at 1, 6, and 12 h and Fhit protein expression was assessed and compared with cells transfected with the scrambled sequence.
  • As shown in FIG. 3B at 6 and 12 h of CHX after Hsp60 and Hsp10 silencing, there is a strong reduction of Fhit expression (from 1 to 0.4 at 1 and 12 h, respectively). Next, Hsp60 and 10 siRNAs were transfected into A549 cells individually or in combination; 24 h later, cells were infected with AdFHIT m.o.i. 1 and cytosol and mitochondria were fractionated 24 h later. After silencing both Hsps, the Fhit level was unaffected in the cytosol but reduced in mitochondria compared with control (FIG. 3B), showing that the Hsp60/10 complex may mediate virally transduced Fhit stabilization and mitochondrial localization. It is also true that if Hsp60 and Hsp10 are involved in Fhit stability after Fhit viral transduction, the cellular compartment with less Fhit would be affected by a decrease in Fhit stability. The inventors herein also examined the Fhit complex in H1299 D1 cells expressing inducible Fhit, with Fhit negative E1 cells as control; 48 h after Fhit induction in D1 cells (FIG. 3C, left panel), distribution of the Fhit complex proteins was similar in the cytosol and mitochondria of D1 and E1 cells, with and without H2O2.
  • Hsp60 was immunoprecipitated from total cell lysates of these cells at 48 h after PonA induction, with or without H2O2, and coprecipitated Fhit and Fdxr (FIG. 3C, right panel). Induction of Fhit expression in D1 cells does not cause biological changes in vitro; thus the Fhit complex does not form as a consequence of apoptosis. A time course experiment was performed in D1 cells after PonA-induced Fhit expression, with and without stress conditions, to determine whether there were biological changes in Fhit protein interactors. The co-inventors did not detect changes in localization after Fhit expression.
  • Fhit Induces Generation of Reactive Oxygen Species (ROS)—Fdxr, a 54-kDa flavoprotein, is located on the matrix side of the inner mitochondrial membrane, and is responsible for transferring electrons from NADPH, via the single electron shuttle ferredoxin-cytochrome P450, to substrates. Under substrate-limiting conditions, electrons leak from this shuttling system and generate ROS. Fdxr mediates p53-dependent, 5-fluorouracil-induced apoptosis in colorectal cancer cells, through generation of ROS, potent intracellular oxidants, and regulators of apoptosis.
  • The inventors herein then investigated determine whether ROS production could be involved in Fhit-mediated apoptosis. Overexpression of Fdxr increases sensitivity of tumor cells to apoptosis on H2O2 treatment, through ROS production. The inventors examined ROS production in A549 cells, with and without H2O2 treatment, after transient transfection with the FHIT expression plasmid. Intracellular superoxide was assessed by measuring ethidium fluorescence, as a result of oxidation of hydroethydine by superoxide. Intracellular superoxide was measured 5 h after stimulation with increasing concentrations of H2O2. ROS generation was ˜3 times higher (16.7 versus 5.4% at 0.5 mM H2O2 and 18.8 versus 7.7% at 1.0 mM H2O2) in FHIT-transfected cells. 2 mM H2O2 was toxic to Fhit-expressing but not to non-expressing cells (FIG. 4A).
  • A similar experiment was performed with p53 and Fhit negative lung cancer-derived H1299 D1 and E1 clones carrying PonA-inducible FHIT and empty vector expression plasmids, respectively; the cells were treated with 5 μM PonA and at 48 h treated with 0.5 and 1.0 mM H2O2; the % ROS-positive cells was higher in Fhit-positive D1 cells than in E1 control cells (20 versus 3.5% at 0.5 mM H2O2, and 78 versus 25% at 1.0 mM H2O2, respectively) (FIG. 4B).
  • These results were paralleled by experiments with human gastric cancer-derived cells, MKN74A116 (FIG. 10), which express mutant p53 and stably express exogenous Fhit.
  • To further study the generation of ROS after Fhit reconstitution during oxidative stress, DCFH-DA was used to measure the redox state of Fhit-overexpressing cells. Peroxidases, cytochrome c, and Fe2+ can oxidize DCFH-DA to fluorescent 2′,7′-dichlorofluorescein (DCF) in the presence of H2O2; thus, DCF indicates H2O2 levels and peroxidase activity. Increased DCF fluorescence was detected in D1 cells compared with E1 cells under stress conditions (FIG. 4C).
  • The decreased cell viability after H2O2 treatment in Fhit-expressing cells was also assessed by an MTS cytotoxicity assay 24 h after H2O2 treatment. H2O2 treatment caused reduced cell viability or growth arrest in both E1 and D1 cells, but this phenotype was more pronounced in D1 cells (FIG. 4D).
  • To determine whether H2O2 treatment with or without Fhit could affect cell viability or cell cycle kinetics we performed flow cytometry (FIG. 4E); when Fhit was present under stress conditions there was a consistent increase of G2/M arrest at 48 h after 0.25 and 0.5 mM H2O2 treatment, 45.5 and 49.5%, respectively, compared with 27.5 and 29% of E1 cells under the same conditions.
  • To assess if the G2/M arrest could affect long-term viability of the cells, a colony assay was performed (FIG. 4F). No colonies were detected in Fhit-expressing cells after exposure to 0.25 mM or higher concentrations of H2O2.
  • Fhit-induced ROS Generation Is Fdxr-dependent—To evaluate the role of Fdxr in Fhit-mediated ROS generation, the inventors examined the Fdxr level in D1 cells after Fhit induction and observed a 2.4-fold increase of its expression compared with E1 cells (FIG. 5A), an increase that was not due to increased transcription as determined by real time RT-PCR (FIG. 5F).
  • The inventors next measured the Fdxr level, with or without Fhit expression, in the presence of MG132, an inhibitor of proteasome degradation; 4 h after MG132 treatment a significant increase of Fdxr protein was observed in D1 cells compared with E1 cells (FIG. 5B), showing that Fhit protects Fdxr from proteasome degradation.
  • The rate of Fdxr degradation in the presence or absence of Fhit protein was evaluated by the 4-12-h CHX chase (FIG. 5C); the rate of Fdxr degradation was higher in Fhit-negative E1 cells (declining from 1 to 0.3) compared with D1 cells, with no significant decrease. Thus, the inventors herein now believe that Fhit prevents destabilization of the Fdxr protein by protecting it from proteasome degradation.
  • HCT116 colon cancer cells, which express endogenous wild-type p53 and Fhit and carry three FDXR alleles (FDXR+/+/+), and HCT116 FDXR+/−/− cells with two alleles knocked-out (28), were used to determine whether AdFHIT-induced apoptosis is influenced by the Fdxr expression level; the FDXR null condition was not compatible with viability.
  • These cells were infected with AdFHIT m.o.i. 50 or 100 and assessed for apoptosis at 48 and 72 h post-infection. Wild-type HCT116 cells (FDXR+/+/+) were susceptible to exogenous Fhit-mediated apoptosis in a dose-dependent manner, as the fraction of sub-G1 cells was 12.1 and 18.8% at m.o.i. 50 and 100, respectively; FDXR+/−/− cells were refractory at 48 and 72 h (data not shown) to Fhit-induced cell death, with a sub-G1 population of 4.7 and 4.3% at m.o.i. 50 and 100 (FIG. 5D).
  • Fhit overexpression led to increased Fdxr protein levels in both FDXR+/+/+ and FDXR+/−/− cells (FIG. 5E) and FDXR+/−/− cells were committed to Fhit-mediated apoptosis by 72 h after infection.
  • The Fhit-mediated increase of Fdxr expression was not at the transcriptional level, as determined by real time RT-PCR (FIG. 5F) and thus not related to the p53 transcriptional activation.
  • To better determine whether the sub-G1 peak detected in FDXR+/+/+ cells after AdFHIT infection was related to apoptosis induction, a time course experiment at 48, 72, and 96 h for caspase 3 and Parp1 cleavage was performed and compared with AdGFP-infected cells (FIG. 5G).
  • Caspase 3 cleavage and related Parp1 cleavage were observed at 48, 72, and 96 h after virus-mediated Fhit overexpression. The time course of cytochrome c release from mitochondria into cytosol was assessed after infection of HCT116 cells with AdFHIT m.o.i. 100 (FIG. 5H); progressive cytochrome c release was observed in HCT116 FDXR+/+/+ cells compared with GFP-infected cells, indicating initiation of the apoptotic cascade in Fhit overexpressing HCT116 FDXR+/+/+ cells.
  • Fhit Enhances ROS-related Effects of Chemotherapeutic Agents—Generation of intracellular ROS is an early event in the apoptosis of lung cancer cells induced by treatment with paclitaxel. The inventors tested paclitaxel on H1299 D1 and E1 cells with or without induced Fhit expression. After induction of Fhit expression, D1 cells were more sensitive to paclitaxel than E1 cells (FIG. 6A) as measured by the MTS cell viability test. Cisplatin induces Fdxr expression and the cisplatin-induced apoptotic pathway is associated with ROS generation.
  • Fhit expressing D1 cells were more sensitive than E1 cells to cisplatin, measured by MTS assay at 24 and 48 h (FIG. 6B).
  • To examine cell viability after drug treatment, we performed flow cytometry analysis (FIG. 6, FIG. 6C and FIG. 6D); PonA-induced D1 and E1 cells treated with increasing paclitaxel concentrations (50-500 ng/ml) showed increasing sub-G1 populations at 48 h: 9.6, 36, and 40%, respectively, for D1 cells compared with 4, 16.7, and 30% of E1 cells (FIG. 6C).
  • Similarly, increasing cisplatin concentrations (0.05-0.2 mM) led to increased sub-G1 populations at 48 h: 5, 16.2, and 30%, respectively, in D1 cells, compared with 2.3, 7, and 14.6% of E1 cells (FIG. 6C).
  • At 24 and 72 h (data not shown) increased sub-G1 populations in D1 compared with E1 cells were also detected. To determine whether the sub-G1 fractions of D1 cells represented apoptotic cells, lysates were prepared from drug-treated cells at 48 h and immunoblot analysis performed for caspase 3 and Parp1 cleavage (FIG. 6D).
  • Activated caspase 3 and related Parp1 cleavage was observed after paclitaxel (50 and 100 ng/ml) and cisplatin (0.05 and 0.1 mM) treatments compared with untreated cells (Ctrl). The inventors herein now believe that Fhit expression increases sensitivity to oxidative injury through participation with Fdxr in ROS generation.
  • EXAMPLE I
  • Materials and Methods
  • Cells, Vectors, and Antisera—A549, H1299, MKN74-E4, and A116, and HCT116 cells were maintained in RPMI 1640 medium plus 10% fetal bovine serum and penicillin/streptomycin (Sigma). HEK293 cells (Microbix) used for preparation of recombinant adenoviruses were cultured in Dulbecco's modified Eagle's medium plus 10% fetal bovine serum and penicillin/streptomycin. AdFHIT-His6 virus was prepared as described in Example II herein. [His6—SEQ ID NO: 32] [penta-His—SEQ ID NO: 33].
  • Full-length FDXR was amplified from human brain cDNAs (Clontech), subcloned into the pcDNA3.1/V5-HisTOPO TA vector (Invitrogen) and sequenced; details are as described under supplemental Methods. Cells were transfected using Lipofectamine™ (Invitrogen) following the manufacturer's directions.
  • Western Blot Analysis—Immunoblot analyses were performed as described in Trapasso, et al., (2003) Proc. Natl. Acad. Sci. U.S.A. 100, 1592-1597) using monoclonal anti-pentaHis (Qiagen); rabbit polyclonal anti-Fhit (Zymed Laboratories Inc.); rabbit polyclonal antisera against GFP, Hsp60, Hsp10, and cytochrome c (Santa Cruz Biotechnology); rabbit polyclonal anti-Fdxr (Abcam); monoclonal anti-CoxIV (Molecular Probes); anti-V5 (Sigma); anti-Parp1 (Santa Cruz Biotechnology); and anti-caspase 3 (Cell Signaling). Protein levels were normalized relative to β-actin or/and GAPDH3 level, detected with appropriate antisera (Santa Cruz Biotechnology).
  • Mass Spectrometry Studies—Protein pellets were solubilized and digested by trypsin as described herein. Peptide mixtures were injected for LC-MS/MS analysis. After protein identification by data base search, inspection of LC-MS/MS data was undertaken to assess the exclusive presence of mass peaks belonging to candidate partner proteins in samples from cells infected with AdFHIT-His6.
  • Protein Interaction Analyses—Proteins were extracted in 15 mM Tris-Cl, pH 7.5, 120 mM NaCl, 25 mM KCl, 2 mM EGTA, 0.1 mM dithiothreitol, 0.5% Triton X-100, 10 mg/ml leupeptin, 0.5 mM phenylmethylsulfonyl fluoride. Co-immunoprecipitation experiments, with or without dithiobis(succinimidyl propionate) (DSP), were performed by incubating 1 mg of total proteins with Hsp60, Hsp10, Fdxr, penta-His, and V5 antisera conjugated with Sepharose for 2 h at 4° C.; after washing, beads were boiled in 1× SDS sample buffer and proteins separated on 4-20% polyacrylamide gels (Bio-Rad), transferred to a poly(vinylidene difluoride) filter (Millipore), and probed with specific antisera.
  • Subcellular Localization of Fhit Protein—Fhit was sublocalized in ponasterone A (PonA)-induced, Fhit-expressing H1299 D1 cells by indirect immunofluorescence detection using anti-Fhit serum and by detection of FhitHis6 in A549 AdFHIT-His6-infected cells in immunoelectron micrographs using anti-pentaHis. In fractionation studies, mitochondria were isolated with the Mitochondria/Cytosol Fractionation kit and the FractionPREP™ Cell Fractionation System was used to extract proteins from cytosol, membranes, nuclei, and cytoskeleton (Biovision Research Products). For submitochondrial localization according to the method of Dahéron et al. (2001) Nucleic Acids Res. 29, 1308-1316, mitochondria were resuspended in 0.1 M sodium carbonate, pH 11.5, on ice for 30 min with periodic vortexing and fractionated as described herein.
  • Flow Cytometry—HCT116 FDXR+/+/+ and FDXR+/−/− cells were infected with AdFHIT or AdGFP at m.o.i. 50 and 100 and assessed at 48 h postinfection. PonA-induced H1299 D1 and E1 cells were treated with 0.25 and 0.5 mM H2O2 or with chemotherapeutic drugs and incubated for varying times, as indicated in the text and figures. For both experiments the cells were collected, washed with phosphate-buffered saline, and resuspended in cold 70% ethanol. For analysis, cells were spun down, washed in phosphate-buffered saline, and suspended in 0.1 mg/ml propidium iodide/Triton X-100 staining solution (0.1% Triton X-100, 0.2 mg/ml DNase-free RNase A) for 30 min at room temperature and analyzed by flow cytometry.
  • Assessment of Intracellular Reactive Oxygen Species (ROS)—Intracellular superoxide was measured through ethidium fluorescence as a result of oxidation by hydroethidine (dihydroethidium-HE; Molecular Probes). MNK74 stably Fhit expressing cells, A549 cells transiently expressing Fhit, and H1299 inducible Fhit expressing cells were treated with 0.5, 1.0, 2.0, and 4.0 mM H2O2 at 37° C.; 4 h later, hydroethidine (10 μM) was added to cells and incubated for 15 min at 37° C. Fluorescence was measured by flow cytometry. Dichlorofluorescein diacetate (DCFH-DA) (Molecular Probes) was used in D1 cells expressing induced Fhit, stressed with H2O2 (0.1 to 1.0 mM), treated with 10 μM DCFH-DA, and incubated for 1 h at 37° C. DCF fluorescence was measured by flow cytometry on a FAC-Scan flow cytometer and fluorescence microscopy.
  • Hsp60 and Hsp10 Silencing—A549 lung cancer cells at 8×105/well (6 wells plate) were transfected by Lipofectamine 2000 reagent (Invitrogen) and 6 μg of Hsp60 and/or Hsp10 siRNAs (Dharmacon catalog numbers NM002156 [GenBank] and NM002157 [GenBank], respectively); 48 h later cells were infected with AdFHIT at m.o.i. 1 and collected for cytosol/mitochondria protein fractionation 24 h later. Proteins were analyzed by SDS-PAGE and immunoblotting; filters were probed with Hsp60, Hsp10, and Fhit antisera. Protein loading was normalized with GAPDH and CoxIV. 1×106 H1299 D1 and E1 lung cancer cells were transfected as described above and at 24 h after transfection the cells were PonA-induced; 48 h after induction a cycloheximide (CHX) (10 μg/ml) chase at 1, 4, 6, and 12 h was performed and the protein lysates were analyzed as described herein.
  • Real-time RT-PCR—Total RNA isolated with TRIzol reagent (Invitrogen) was processed after DNase treatment (Ambion) directly to cDNA by reverse transcription using SuperScript First-Strand (Invitrogen). Target sequences were amplified by qPCR using Power SYBR Green PCR Master Mix (Applied Biosystems). FDXR primers were: forward, 3′-TCGACCCAAGCGTGCCCTTTG-5′ [SEQ ID No. 24]; reverse, 3′-GTGGCCCAGGAGGCGCAGCATC-5′ [SEQ ID No. 25]. Samples were normalized using Actin and GADPH genes.
  • Chemotherapeutic Drug Treatment—Paclitaxel (Sigma) was dissolved in DMSO as a 10 mmol/liter stock solution and stored at −80° C. Cisplatin (Sigma) was dissolved in water and freshly prepared before use. H1299 D1 and E1 cells were seeded (1×104 cells/well) in 96-well culture plates, PonA-induced, and after 24 h treated with paclitaxel (50, 100, and 500 ng/ml) or cisplatin (0.05, 0.1, and 0.2 mM). H1299 D1 and E1 cells PonA-induced were incubated for 24, 48, and 72 h and assessed for viability with an MTS kit (Cell Titer 96® Aqueous MTS kit, Upstate Biotechnology, Lake Placid, N.Y.), as recommended by the manufacturer.
  • EXAMPLE II
  • Generation of recombinant adenoviruses—The recombinant adenovirus carrying the wild-type FHIT cDNA (AdFHIT) was prepared as previously described (Ishii et al., 2001 Cancer Res 61:1578-1584). A His-tagged FHIT cDNA was generated by PCR with the following oligonucleotides: 5′-ACgTggATCCCTgTgAggACATgTCgTTCAgATTTggC-3′ (forward) [SEQ ID NO: 26] and 5′-TTgTggATCCTTATCAgTgATggTgATggTgATgCgATCCTCTCTgAAAgTAgCCCgCAg-3′ [SEQ ID NO: 27]. These primers were designed with a BamHI restriction site for subcloning into the transfer vector pAdenoVator-CMV5-IRES-GFP. The Ad-His6 was generated with the AdenoVator™ kit (Qbiogene, Carlsbad, Calif.), following the manufacturer's procedure. Ad GFP, used as control, was purchased from Qbiogene (Carlsbad, Calif.).
  • Generation of a recombinant expression vector carrying FDXR cDNA—Wild-type ferredoxin reductase full-length was amplified from human brain cDNAs (Clontech, Palo Alto, Calif.) with primers: 5′-CTgTTCCCAgCCATggCTTCgCgCTg-3′ (forward) [SEQ ID NO: 28] and 5′-TCAgTggCCCAggAggCgCAgCATC-3′ [SEQ ID NO: 29]. The amplification products were subcloned into the pcDNA3.1/V5-HisTOPO TA vector (Invitrogen, Carlsbad, Calif.). Sequencing excluded mutations in the amplified products.
  • For the preparation of a V5-tagged ferredoxin reductase cDNA, PCR amplification was performed with the same primer sequences with the exclusion of the FDXR physiological stop codon in the reverse primer. That is, both wild-type and V5-tagged ferredoxin reductase (FDXR) cDNAs were prepared by using as a template the wild-type coding sequence of the human ferredoxin reductase gene (GenBank Accession #NM024417). The ferredoxin reductase coding sequence was amplified from human brain cDNAs (Clontech).
  • The primers used to generate the V5-tagged FDXR cDNA were: Forward: 5′-CTgTTCCCAgCCATggCTTCgCgCTg-3′ [SEQ ID NO: 30]; and Reverse: 5′-gTggCCCAggAggCgCAgCATC-3′ [SEQ ID NO: 31]. It is to be noted that the oligonucleotide sequences are identical except for the reverse primer used for the generation of the V5-tagged cDNA, where the physiological stop codon of the FDXR was omitted in order to fuse in frame the FDXR coding sequence with a V5-tag.
  • The amplification products were subcloned into the pcDNA3.1/V5-HisTOPO TA vector (Invitrogen, Carlsbad, Calif.). Sequencing excluded mutations in the amplified products.
  • In certain embodiments, the adenovirus being capable of isolating Fhit-His6, comprises an adenovirus carrying a FHIT-His6 cDNA. The FHIT-His6 cDNA can be prepared by using as a template the wild-type coding sequence of the human FHIT gene (GenBank Accession #NM002012). In order to introduce a polyhistidine-tag at the C-terminus of final Fhit product, a PCR amplification of the wild-type FHIT coding sequence was carried out with a reverse primer designed to abolish the physiological stop codon and to add to the endogenous FHIT sequence a stretch of 18 bp coding for six hystidines followed by an artificial stop codon. Furthermore, both forward and reverse primers carried a BamHI restriction site for an easy subcloning. The oligonucleotide sequences used for this amplification were the following: Forward: 5′-ACgTggATCCCTgTgAggACATgTCgTTCAgATTTggC-3′ [SEQ ID NO: 26]; and Reverse: 5′-TTgTggATCCTTATCAgTgATggTgATggTgATgCgATCCTCTCTgAAAgTAgACCCgCAg-3′ [SEQ ID NO: 27]. The PCR amplification product was sequenced to exclude random mutations before to be cloned in an Ad5 recombinant genome (AdenoVator™, a vector purchased by Qbiogene).
  • In certain embodiments, a method of isolating exogenously over-expressed Fhit-His6 includes using an adenovirus carrying a FHIT-His6 cDNA wherein the Fhit-His6 is isolated through the His tag. Fhit-His6 represents the recombinant protein whose expression is driven into a mammalian cell through the Ad FHIT-His6 vector. The His6 epitope allows for the recovery of the recombinant Fhit-His6 protein plus the protein complex interacting with the recombinant protein itself by taking advantage of the Ni-NTA system. This system is commercially available from Qiagen. Briefly, human A549 cancer cells were infected with Ad FHIT-His6; forty-eight hours after infection, photo-cross-linking of intracellular protein complexes was performed with the cross-linker dithiobis(succinimidyl propionate) [DSP] purchased from Pierce in order to stabilize protein complexes in living cells. Cells were disrupted in a protein extraction buffer and Fhit-His6 protein complex was isolated with the Ni-NTA magnetic-bead technology by taking advantage of the great affinity of the His6 tag for such beads. The isolated Fhit-His6 protein complex was then investigated through mass spectrometry to identify all proteins present in the complex.
  • In certain embodiments, a recombinant adenovirus carrying fragile histidine triad (Fhit) FHIT cDNA can be modified at its 3′ with a sequence encoding a histidine-six epitope tag (AdFHIT-His6).
  • In certain embodiments, a method for mediating an apoptotic process in at least one cell, comprises exposing the cell to a fragile histidine triad (Fhit) gene product in an amount sufficient to mediate the apoptotic process in the cell.
  • In certain embodiments, a method for inducing an apoptosis process in a cell, comprises exposing the cell to a fragile histidine triad (Fhit) gene product in an amount sufficient to cause generation of reactive oxygen species (ROS) in the cell.
  • In certain embodiments, a method for mediating an apoptotic process in at least one cell, comprising: exposing the cell to a sufficient amount of fragile histidine triad (Fhit) gene product to allow the Fhit to enter mitochondria in the cell and to interact with Fdxr protein in the cell and to cause an increase in Fdxr protein level that is associated with generation of ROS, and causing a change in the apoptotic process in the cell.
  • It is to be noted that in previous studies, it was extensively proved that Fhit protein overexpression in Fhit-negative cancer cells is able to trigger programmed cell death (or apoptosis). In the instant invention, the inventors provide a rationale about the role of Fhit protein in the process of apoptosis. In fact, FHIT gene therapy of cancer cells performed with Ad FHIT (at the multiplicity of infection 50 or MOI50, i.e., 50 viral particles per cell) is responsible of Fhit overexpression; then, the newly synthesized recombinant protein is taken by its interactors Hsp60/Hsp10 from the cytosol to the mitochondria where Fhit interacts with FDXR (ferredoxin reductase) a protein belonging to the respiratory chain. This interaction leads to the mitochondrial generation of ROS (Reactive Oxygen Species). ROS represent the early step for the initiation of the intrinsic (or mitochondrial) pathway of the apoptotic process; in fact, they induce a damage in the mitochondrial membranes that, in turn, release cytochrome c into the cytosol. This step is crucial for the execution of apoptosis, as cytochrome c contributes with other cytosolic molecules (i.e., Apaf-1 and pro-caspase 3) to the generation of the apoptosome, a multiprotein complex able to drive the cell, in a non-reversible fashion, to apoptosis.
  • A method commonly used to study apoptosis consists in the detection of mature caspase-3 (an indicator of incipient apoptosis) by flow cytometric analysis (Becton Dickinson) [for reference, see Trapasso et al., 2003, PNAS, 100, 1592-1597].
  • In certain embodiments, a method for preparing a V5-tagged ferredoxin reductase cDNA, comprises PCR amplifying with primer sequences: 5′-CTgTTCCCAgCCATggCTTCgCgCTg-3′ (forward) [SEQ ID NO: 28], and 5′-TCAgTggCCCAggAggCgCAgCATC-3′ [SEQ ID NO: 29] and subcloning the amplification products pcDNA3.1/V5-HisTOPO TA vector.
  • Both wild-type and V5-tagged ferredoxin reductase (FDXR) cDNAs were prepared by using as a template the wild-type coding sequence of the human ferredoxin reductase gene (GenBank Accession #NM024417). The ferredoxin reductase coding sequence was amplified from human brain cDNAs (Clontech). That is, the primers used to amplify the wild-type FDXR cDNA were: Forward: 5′-CTgTTCCCAgCCATggCTTCgCgCTg-3′ [SEQ ID NO: 28]; Reverse: 5′-TCAgTggCCCAggAggCgCAgCATC-3′ [SEQ ID NO: 29].
  • The primers used to generate the V5-tagged FDXR cDNA were: Forward: 5′-CTgTTCCCAgCCATggCTTCgCgCTg-3′ [SEQ ID NO: 30], and Reverse: 5′-gTggCCCAggAggCgCAgCATC-3′ [SEQ ID NO: 31]. Note that the oligonucleotide sequences are identical except for the reverse primer used for the generation of the V5-tagged cDNA, where the physiological stop codon of the FDXR was omitted in order to fuse in frame the FDXR coding sequence with a V5-tag.
  • Finally, the two products were subcloned in the pcDNA3.1/V5-HisTOPO TA expression vector (purchased from Invitrogen) and then sequenced to assess that both products had no random mutations.
  • In certain embodiments, a method for generating a recombinant adenovirus, comprises preparing a recombinant adenovirus carrying the wild-type FHIT cDNA (AdFHIT); and generating a His-tagged FHIT cDNA using PCR with the following oligonucleotides: 5′-ACgTggATCCCTgTgAggACATgTCgTTCAgATTTggC-3′ (forward) [SEQ ID NO: 26], and 5′-TTgTggATCCTTATCAgTgATggTgATggTgATgCgATCCTCTCTgAAAgTAgACCCgCAg-3′ [SEQ ID NO: 27]. The FHIT-His6 cDNA was prepared as described herein. The recombinant adenoviral vector Ad FHIT-His6 was prepared according to the manufacturer's suggestions (Qbiogene). Briefly, the amplified FHIT-His6 PCR fragment was digested with BamHI and subcloned into the BamHI linearized transfer vector pAdenoVator-CMV5-IRES-GFP. The pAdenoVator-CMV5-IRES-GFP/FHIT-His6 was co-transfected with the E1/E3 deleted Ad5 backbone viral DNA into 293 cells. Viral plaques were screened for the presence of the Fhit-His6 protein by Western blot with specific penta-His antibodies (Qiagen). One positive clone was plaque-purified and amplified on 293 cells. After freeze/thaw cycles, the adenoviruses in the supernatant were purified on two successive cesium chloride gradients. The recombinant adenovirus was titered by the TCID50 method and aliquoted. Virus stocks were stored at −80° C. Finally, the recombinant adenovirus carrying the wild-type FHIT cDNA (Ad FHIT) was previously generated by Trapasso et al. 2003, PNAS, 100, 1592-1597.
  • Mitochondrial localization studies—Confocal microscopy was used to assess Fhit protein distribution by immunofluorescence; H1299 D1 cells, with inducible FHIT cDNA, and E1 cells, with empty vector, were treated with PonA for 48 hr, and living cells were stained with Mitotracker Red 580 (M-22425, Molecular Probes, Eugene, Oreg.) at a working concentration of 500 nM for 40 min under growth conditions. The cells were fixed and permeablilized by incubation in ice-cold acetone for 5 min and then washed in PBS. Cells were incubated for 1 hr with 5% BSA to block non-specific interactions and than incubated overnight with Fhit antiserum (Zymed, S. San Francisco, Calif.) at a working concentration of 1.6 μg/ml, washed with PBS and incubated with Alexa Fluor 488 donkey anti-rabbit IgG (Molecular Probes). The slides were mounted in mounting medium for fluorescence with DAPI (Vector, Burlingane, Calif.) and visualized. For immunoelectron microscopy localization of Fhit, A549 cells infected with AdHis6 or AdFHIT, MOI 5, were fixed in 4% paraformaldehyde in PBS pH7.2 for 30 min at 4° C., washed 3 times with PBS, and remaining free aldehyde groups were reduced using a 30 min incubation in 0.05% sodium borohydride in PBS. Following a PBS wash, samples were blocked with 50 mM glycine in PBS for 30 min, washed twice in PBS, and dehydrated with 25 and 50% ethanol for 15 min each, followed by 3 changes of 70% ethanol for 15 min each. The samples were then infiltrated with 70% ethanol+LR White resin, hard grade (Electron Microscopy Sciences, Hatfield, Pa.) at 2:1 for 1 hr, 70% ethanol+LR White at 1:2 for 1 hr, 100% LR White for 1 hr and 100% LRW overnight at 4° C. The following day the cells received 2 more changes of 100% LR White and were polymerized in gelatin capsules at 58° C. for 20-24 hr. 900 nm thin section s were cut using a Reichert UCT Ultramicrotome and a diamond knife and placed on nickel grids. The grids were floated section side down on drops of PBS for 5 min, 5% goat serum in PBS for 1 hr at room temperature (RT), and either Penta-His mouse monoclonal antibody at 20 mg/ml (Qiagen, Valencia, Calif.) diluted in PBS containing 0.1% BSA and 0.05% Tween-20 (BSA/Tw) or BSA/Tw alone overnight at 4° C. in a humidified chamber. The following day the grids were washed 6× for 5 min each using PBS and then incubated with h goat anti-mouse 10 nm colloidal gold conjugate (Ted Pella, Redding, Calif.) diluted 1:10 in BSA/Tw for 2 hrs at RT. The grids were washed 6× each for 5 min with PBS, rinsed with DI H2O and post-stained with 2.5% aqueous uranyl acetate for 3 min. Images were collected on a Tecnai 12 electron microscope equipped with a US1000 Gatan 2K digital camera.
  • Sample digestion and LC-MS/MS analysis—Proteins isolated with Ni-NTA beads were precipitated with cold acetone and resuspended in 6 M urea buffered at pH 8 with 0.1 M Tris-HCl. Protein reduction and alkylation were achieved, respectively, by the addition of DTT (final concentration 10 mM, 1 hr incubation at 37° C.) and iodoacetamide (final concentration 25 mM, 1 hr incubation at 37° C.). After neutralizing excess iodoacetamide with DTT (additional 5 mM), urea concentration was lowered to 1.5 M by dilution with 1 mM CaCl2. Overnight digestion was carried out using 50 ng of TPCK-treated trypsin (Sigma). Total digestion solution volume was 100 μl.
  • Chromatography was performed on an Ultimate nano LC system from Dionex (Sunnyvale, Calif.). Digest mixtures (30 μl) were directly injected onto a Pepmap C18 RP cartridge (0.3 mm ID×5 mm length) and washed for 10 minutes with H2O/trifluoroacetic acid (TFA)/acetonitrile 97.9:0.1:2 (v/v/v) before the RP trap was switched on-line to a 75 μm×150 mm Pepmap C18 nano LC column. Gradient elution of peptides was achieved at 300 nl/min using a 45-min linear gradient going from 5% B to 50% B. Mobile phase A was H2O/acetonitrile/formic acid (FA)/TFA 97.9:2:0.08:0.02 (v/v/v/v); mobile phase B was H2O/acetonitrile/FA/TFA 4.9:95:0.08:0.02 (v/v/v/v).
  • MS detection was performed on an Applied Biosystems (Framingham, Mass.) QSTAR XL hybrid LC-MS/MS operating in positive ion mode, with nanoelectrospray potential at 1800 V, curtain gas at 15 units, CAD gas at 3 units. Information-dependent acquisition (IDA) was performed by selecting the two most abundant peaks for MS/MS analysis after a full TOF-MS scan from 400 to 1200 m/z lasting 2 seconds. Both MS/MS analyses were performed in enhanced mode (2 seconds/scan).
  • LC-MS/MS Data Analysis—MS/MS spectra were searched by interrogating the Swiss Prot database on the Mascot search engine (matrixscience.com) (accessed on June 2006). The following search parameters were used. MS tolerance: 50 ppm; MS/MS tolerance: 1 Da; methionine oxidized (variable modification); cysteine carbamidomethylated (fixed modification); enzyme: trypsin; max. missed cleavages: 1.
  • Protein lists obtained from, respectively, both A549 infected with Ad FHIT-His6 and control were compared, and proteins exclusively present in the A549-Ad FHIT-His6 list were kept for further validation. As a first validation procedure, LC-MS/MS raw data were inspected using selected ion chromatogram (SIC) displaying mode. By SIC comparison, it could be assessed the exclusive presence of the peptides of interest, identified as belonging to the six candidate proteins under examination, in the Ad FRIT-His6 sample. Such verification step already provided rather strong evidence for the specific capture of the six candidate protein. Also, these findings were further validated by biochemical and functional assays.
  • In accordance with the provisions of the patent statutes, the principle and mode of operation of this invention have been explained and illustrated in its preferred embodiment. However, it must be understood that this invention may be practiced otherwise than as specifically explained and illustrated without departing from its spirit or scope.
  • The references discussed herein, to the extent that they provide exemplary procedural or other details supplementary to those set forth herein, are specifically incorporated herein by reference.

Claims (19)

What is claimed is:
1. A method of sensitizing at least one cell to a chemotherapeutic agent comprising:
exposing the at least one cell to an exogenous fragile histidine triad (Fhit) gene product in an amount sufficient to sensitize the at least one cell; and thereafter,
administering a chemotherapeutic agent to the at least one sensitized cell.
2. The method of claim 1, wherein the cell is a cancer cell.
3. The method of claim 2, wherein the cancer cell comprises one or more of: a lung cancer cell, a colon cancer cell, or a gastric cancer cell.
4. The method of claim 1, wherein the cell is a human cell.
5. The method of claim 1, wherein the chemotherapeutic agent comprises one or more of: paclitaxel or cisplatin.
6. The method of claim 1, wherein the Fhit gene product comprises Fhit-His6.
7. The method of claim 3, wherein the Fhit-His6, comprises an adenovirus carrying a FHIT-His6 cDNA.
8. The method of claim 1, wherein the Fhit gene product comprises a recombinant adenovirus carrying fragile histidine triad (Fhit) FHIT cDNA modified at its 3′ with a sequence encoding a histidine-six epitope tag (AdFHIT-His6).
9. The method of claim 1, wherein the amount of Fhit gene product is administered in an amount sufficient to mediate an apoptotic process in the cell.
10. The method of claim 1, wherein the amount of Fhit gene product is administered in an amount sufficient to cause generation of reactive oxygen species (ROS) in the cell.
11. The method of claim 1, wherein the amount of Fhit gene product is administered to a sufficient amount to:
allow the Fhit gene product to enter mitochondria in the cell;
interact with Fdxr protein in the cell;
cause an increase in Fdxr protein level in the cell that is associated with generation of reactive oxygen species (ROS); and/or,
cause a change in an apoptotic process in the cell.
12. A pharmaceutical composition for treating a cancer associated disorder, comprising at least one isolated Fhit gene product, and a pharmaceutically-acceptable carrier.
13. The pharmaceutical composition of claim 12, further comprising at least one chemotherapeutic composition present in a dosage form that contacts at least one cell at a time subsequent to the at least one isolated Fhit gene product.
14. The pharmaceutical composition of claim 13, wherein the chemotherapeutic composition comprises one or more of: paclitaxel or cisplatin.
15. The pharmaceutical composition of claim 14, the paclitaxel is present in an amount between about 50 ng/ml to about 500 ng/ml.
16. The pharmaceutical composition of claim 14, the paclitaxel is present in an amount of: 50 ng/ml, 100 ng/ml, or 500 ng/ml.
17. The pharmaceutical composition of claim 14, wherein the cisplatin is present in an amount between about 0.05 mM to about 0.2 mM.
18. The pharmaceutical composition of claim 14, the cisplatin is present in an amount of: 0.05 mM, 0.1 mM, or 0.2 mM.
19. The pharmaceutical composition of claim 14, wherein the cancer comprises one or more of: a lung cancer, a colon cancer, or a gastric cancer.
US14/483,228 2007-10-26 2014-09-11 Methods for Identifying Fragile Histidine Triad (FHIT) Interaction and Uses Thereof Abandoned US20150010647A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US14/483,228 US20150010647A1 (en) 2007-10-26 2014-09-11 Methods for Identifying Fragile Histidine Triad (FHIT) Interaction and Uses Thereof
US15/264,325 US20160375107A1 (en) 2007-10-26 2016-09-13 Fragile histidine triad (fhit) compositions and uses thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US48007P 2007-10-26 2007-10-26
PCT/US2008/081294 WO2009055773A2 (en) 2007-10-26 2008-10-27 Methods for identifying fragile histidine triad (fhit) interaction and uses thereof
US73954111A 2011-02-25 2011-02-25
US14/483,228 US20150010647A1 (en) 2007-10-26 2014-09-11 Methods for Identifying Fragile Histidine Triad (FHIT) Interaction and Uses Thereof

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2008/081294 Division WO2009055773A2 (en) 2007-10-26 2008-10-27 Methods for identifying fragile histidine triad (fhit) interaction and uses thereof
US12/739,541 Division US8911998B2 (en) 2007-10-26 2008-10-27 Methods for identifying fragile histidine triad (FHIT) interaction and uses thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/264,325 Division US20160375107A1 (en) 2007-10-26 2016-09-13 Fragile histidine triad (fhit) compositions and uses thereof

Publications (1)

Publication Number Publication Date
US20150010647A1 true US20150010647A1 (en) 2015-01-08

Family

ID=40580439

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/739,541 Active 2032-02-25 US8911998B2 (en) 2007-10-26 2008-10-27 Methods for identifying fragile histidine triad (FHIT) interaction and uses thereof
US14/483,228 Abandoned US20150010647A1 (en) 2007-10-26 2014-09-11 Methods for Identifying Fragile Histidine Triad (FHIT) Interaction and Uses Thereof
US15/264,325 Abandoned US20160375107A1 (en) 2007-10-26 2016-09-13 Fragile histidine triad (fhit) compositions and uses thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US12/739,541 Active 2032-02-25 US8911998B2 (en) 2007-10-26 2008-10-27 Methods for identifying fragile histidine triad (FHIT) interaction and uses thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/264,325 Abandoned US20160375107A1 (en) 2007-10-26 2016-09-13 Fragile histidine triad (fhit) compositions and uses thereof

Country Status (7)

Country Link
US (3) US8911998B2 (en)
EP (1) EP2201112A4 (en)
JP (1) JP2011504093A (en)
CN (2) CN103898069A (en)
AU (1) AU2008316577B2 (en)
CA (1) CA2703707A1 (en)
WO (1) WO2009055773A2 (en)

Families Citing this family (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103882124B (en) 2005-08-01 2015-11-18 俄亥俄州立大学研究基金会 For the method and composition based on MicroRNA of the diagnosis of mammary cancer, prognosis and treatment
WO2007081720A2 (en) 2006-01-05 2007-07-19 The Ohio State University Research Foundation Microrna-based methods and compositions for the diagnosis, prognosis and treatment of lung cancer
EP2591794A1 (en) 2006-01-05 2013-05-15 The Ohio State University Research Foundation MicroRNA expressions abnormalities in pancreatic endocrine and acinar tumors
CA2633754C (en) 2006-01-05 2013-06-18 The Ohio State University Research Foundation Microrna-based methods and compositions for the diagnosis and treatment of solid cancers
EP2371971B1 (en) 2006-03-20 2013-11-27 The Ohio State University Research Foundation Microrna fingerprints during human megakaryocytopoiesis
EP2455493B1 (en) 2006-07-13 2014-01-08 The Ohio State University Research Foundation Micro-RNA-based methods and compositions for the diagnosis and treatment of colon related diseases
EP2061907B1 (en) 2006-09-19 2011-11-23 The Ohio State University Research Foundation Tcl1 expression in chronic lymphocytic leukemia (cll) regulated by mir-29 and mir-181
WO2008054828A2 (en) 2006-11-01 2008-05-08 The Ohio State University Research Foundation Microrna expression signature for predicting survival and metastases in hepatocellular carcinoma
CN103555825B (en) 2007-01-31 2015-09-30 俄亥俄州立大学研究基金会 For the method and composition based on microRNA of the diagnosis of acute myelocytic leukemia (AML), prognosis and treatment
US8465917B2 (en) 2007-06-08 2013-06-18 The Ohio State University Research Foundation Methods for determining heptocellular carcinoma subtype and detecting hepatic cancer stem cells
CN101918424A (en) 2007-06-15 2010-12-15 俄亥俄州立大学研究基金会 Oncogenic ALL-1 fusion proteins for targeting Drosha-mediated microRNA processing
US8367632B2 (en) 2007-07-31 2013-02-05 Ohio State University Research Foundation Methods for reverting methylation by targeting methyltransferases
EP2173908B1 (en) 2007-08-03 2016-01-06 The Ohio State University Research Foundation Ultraconserved regions encoding ncrnas
WO2009026487A1 (en) 2007-08-22 2009-02-26 The Ohio State University Research Foundation Methods and compositions for inducing deregulation of epha7 and erk phosphorylation in human acute leukemias
CN103898069A (en) 2007-10-26 2014-07-02 俄亥俄州立大学研究基金会 Methods for identifying fragile histidine triad (fhit) interaction and uses thereof
CN102149827B (en) 2008-06-11 2014-08-20 由卫生与公众服务部代表的美利坚合众国政府 Use of MiR-26 family as a predictive marker of hepatocellular carcinoma and responsiveness to therapy
US8916533B2 (en) 2009-11-23 2014-12-23 The Ohio State University Materials and methods useful for affecting tumor cell growth, migration and invasion
WO2012065049A1 (en) 2010-11-12 2012-05-18 The Ohio State University Research Foundation Materials and methods related to microrna-21, mismatch repair, and colorectal cancer
CN103313706A (en) 2010-11-15 2013-09-18 俄亥俄州立大学研究基金会 Controlled release mucoadhesive systems
EP2683387A4 (en) 2011-03-07 2014-09-03 Univ Ohio State MUTATOR ACTIVITY INDUCED BY MICRORNA-155 (miR-155) LINKS INFLAMMATION AND CANCER
EP2766500A4 (en) 2011-10-14 2015-10-14 Univ Ohio State Methods and materials related to ovarian cancer
US9481885B2 (en) 2011-12-13 2016-11-01 Ohio State Innovation Foundation Methods and compositions related to miR-21 and miR-29a, exosome inhibition, and cancer metastasis
EP2804960A4 (en) 2012-01-20 2015-08-19 Univ Ohio State Breast cancer biomarker signatures for invasiveness and prognosis

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070178105A1 (en) * 1996-02-09 2007-08-02 Croce Carlo M FHIT proteins and nucleic acids and methods based thereon

Family Cites Families (168)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4196265A (en) 1977-06-15 1980-04-01 The Wistar Institute Method of producing antibodies
US4172124A (en) 1978-04-28 1979-10-23 The Wistar Institute Method of producing tumor antibodies
US4608337A (en) 1980-11-07 1986-08-26 The Wistar Institute Human hybridomas and the production of human monoclonal antibodies by human hybridomas
US4701409A (en) 1984-11-15 1987-10-20 The Wistar Institute Detection of B-cell neoplasms
US4693975A (en) 1984-11-20 1987-09-15 The Wistar Institute Human hybridroma fusion partner for production of human monoclonal antibodies
US5015568A (en) 1986-07-09 1991-05-14 The Wistar Institute Diagnostic methods for detecting lymphomas in humans
US5202429A (en) 1986-07-09 1993-04-13 The Wistar Institute DNA molecules having human BCL-2 gene sequences
US5198338A (en) 1989-05-31 1993-03-30 Temple University Molecular probing for human t-cell leukemia and lymphoma
EP0480970A1 (en) 1989-07-03 1992-04-22 Temple University - Of The Commonwealth System Higher Education Tcl-5 gene rearrangement involved in t-cell leukemia and melanoma
US5149628A (en) 1989-11-15 1992-09-22 Temple University Methods for detecting bcl-3 gene in human leukemias
WO1993012136A1 (en) 1991-12-11 1993-06-24 Thomas Jefferson University Detection and treatment of acute leukemias resulting from chromosome abnormalities in the all-1 region
US6040140A (en) 1991-12-11 2000-03-21 Thomas Jefferson University Methods for screening and treating leukemias resulting from all-1 region chromosome abnormalities
US5633135A (en) 1991-12-11 1997-05-27 Thomas Jefferson University Chimeric nucleic acids and proteins resulting from ALL-1 region chromosome abnormalities
CA2148350A1 (en) 1992-10-29 1994-05-11 Carlo Croce Methods of detecting micrometastasis of prostate cancer
US5674682A (en) 1992-10-29 1997-10-07 Thomas Jefferson University Nucleic acid primers for detecting micrometastasis of prostate cancer
GB9307754D0 (en) 1993-04-15 1993-06-02 Perry Robert E Diagnostic probes and therapeutic products
WO1994026930A1 (en) 1993-05-14 1994-11-24 Thomas Jefferson University Methods for screening and treating leukemias resulting from all-1 region chromosome abnormalities
US7175995B1 (en) 1994-10-27 2007-02-13 Thomas Jefferson University TCL-1 protein and related methods
US5985598A (en) 1994-10-27 1999-11-16 Thomas Jefferson University TCL-1 gene and protein and related methods and compositions
US5695944A (en) 1995-05-05 1997-12-09 Thomas Jefferson University Modulation of bcl-2 phosphorylation
US5567586A (en) 1995-05-18 1996-10-22 Thomas Jefferson University Methods of indentifying solid tumors with chromosome abnormalities in the ALL-1 region
US5928884A (en) 1996-02-09 1999-07-27 Croce; Carlo M. FHIT proteins and nucleic acids and methods based thereon
AU4003597A (en) 1996-09-04 1998-03-26 Howard Florey Institute Of Experimental Physiology And Medicine Methods of diagnosing and treating cancer
US6187536B1 (en) 1997-02-18 2001-02-13 Thomas Jefferson University Methods of identifying and detecting pancreatic cancer
EP0972083A1 (en) 1997-04-04 2000-01-19 THE TEXAS A&amp;M UNIVERSITY SYSTEM Noninvasive detection of colonic biomarkers using fecal messenger rna
CA2335315A1 (en) 1998-07-20 2000-01-27 Thomas Jefferson University Nitrilase homologs
AU5128999A (en) 1998-07-24 2000-02-14 Yeda Research And Development Co. Ltd. Prevention of metastasis with 5-aza-2'-deoxycytidine
AU2869300A (en) * 1999-02-05 2000-08-25 Uab Research Foundation, The Fiber receptor-independent system for the propagation of adenoviral vectors
CA2361396A1 (en) 1999-02-25 2000-08-31 Carlo M. Croce Compositions, kits, and methods relating to the human fez1 gene, a novel tumor suppressor gene
US7141417B1 (en) 1999-02-25 2006-11-28 Thomas Jefferson University Compositions, kits, and methods relating to the human FEZ1 gene, a novel tumor suppressor gene
EP1165586A4 (en) 1999-03-15 2003-05-28 Univ Jefferson TCL-1b GENE AND PROTEIN AND RELATED METHODS AND COMPOSITIONS
US6579857B1 (en) 1999-06-11 2003-06-17 Evanston Northwestern Healthcare Research Institute Combination cancer therapy comprising adenosine and deaminase enzyme inhibitors
US7163801B2 (en) 1999-09-01 2007-01-16 The Burnham Institute Methods for determining the prognosis for cancer patients using tucan
US20040132017A1 (en) 1999-12-16 2004-07-08 Robert Richards Oxidoreductase gene associated with the fra16d fragile site
US6891031B2 (en) 2000-02-18 2005-05-10 The Regents Of The University Of California Coordinate cytokine regulatory sequences
US20010026796A1 (en) 2000-03-14 2001-10-04 Croce Carlo M. TCL1 enhances Akt kinase activity and mediates its nuclear translocation
US6924414B2 (en) 2000-04-11 2005-08-02 Thomas Jefferson University Muir-torre-like syndrome in Fhit deficient mice
WO2001087958A2 (en) 2000-05-16 2001-11-22 Thomas Jefferson University CRYSTAL STRUCTURE OF WORM NitFhit REVEALS THAT A Nit TETRAMER BINDS TWO Fhit DIMERS
US7060811B2 (en) 2000-10-13 2006-06-13 Board Of Regents, The University Of Texas System WWOX: a tumor suppressor gene mutated in multiple cancers
WO2002064172A2 (en) 2001-02-12 2002-08-22 Thomas Jefferson University Fhit gene therapy prevents tumor development in fhit-deficient mice
WO2002064171A1 (en) 2001-02-12 2002-08-22 Thomas Jefferson University Adenoviral transduction of fragile histidine triad (fhit) into cancer cells
US20040033502A1 (en) 2001-03-28 2004-02-19 Amanda Williams Gene expression profiles in esophageal tissue
US20050176025A1 (en) 2001-05-18 2005-08-11 Sirna Therapeutics, Inc. RNA interference mediated inhibition of B-cell CLL/Lymphoma-2 (BCL-2) gene expression using short interfering nucleic acid (siNA)
US8021658B2 (en) * 2001-05-25 2011-09-20 Thomas Jefferson University Alternative splice forms of proteins as basis for multiple therapeutic modalities
EP2428571B1 (en) 2001-09-28 2018-07-18 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. MicroRNA molecules
US7371736B2 (en) 2001-11-07 2008-05-13 The Board Of Trustees Of The University Of Arkansas Gene expression profiling based identification of DKK1 as a potential therapeutic targets for controlling bone loss
GB0128898D0 (en) 2001-12-03 2002-01-23 Biotech Res Ventures Pte Ltd Materials and methods relating to the stabilization and activation of a tumour suppressor protein
KR101210018B1 (en) 2002-03-08 2012-12-07 에자이 알앤드디 매니지먼트 가부시키가이샤 Macrocyclic compounds useful as pharmaceutical
EP2258872B1 (en) 2002-03-13 2013-08-14 Genomic Health, Inc. Gene expression profiling in biopsied tumor tissues
AU2003226279A1 (en) 2002-04-08 2003-10-27 Ciphergen Biosystems, Inc. Serum biomarkers in hepatocellular carcinoma
CA2484920A1 (en) 2002-04-29 2003-11-13 Thomas Jefferson University Human chronic lymphocytic leukemia modeled in mouse by targeted tcl1 expression
AU2003273542A1 (en) 2002-05-31 2003-12-19 The Board Of Trustees Of The Leland Stanford Junior University Methods of identifying and isolating stem cells and cancer stem cells
US20050260639A1 (en) 2002-09-30 2005-11-24 Oncotherapy Science, Inc. Method for diagnosing pancreatic cancer
US20050266443A1 (en) 2002-10-11 2005-12-01 Thomas Jefferson University Novel tumor suppressor gene and compositions and methods for making and using the same
JP2006512908A (en) 2002-10-11 2006-04-20 トーマス ジェファーソン ユニバーシティー Tumor suppressor genes and compositions and methods for their preparation and use
CN102304570B (en) 2002-11-13 2015-01-21 托马斯杰斐逊大学 Compositions and methods for cancer diagnosis and therapy
US7250496B2 (en) 2002-11-14 2007-07-31 Rosetta Genomics Ltd. Bioinformatically detectable group of novel regulatory genes and uses thereof
WO2004071464A2 (en) 2003-02-12 2004-08-26 Johns Hopkins University School Of Medicine Diagnostic application of differentially-expressed genes in lympho-hematopoietic stem cells
WO2004079013A1 (en) 2003-03-03 2004-09-16 Arizona Board Of Regents On Behalf Of The University Of Arizona Ecto-5’-nucleotidase (cd73) used in the diagnosis and the treatment of pancreatic cancer
US7183384B2 (en) 2003-03-06 2007-02-27 A & G Pharmaceutical, Inc. Monoclonal antibody 7H11 reactive with human cancer
AU2003286741A1 (en) 2003-05-02 2004-11-26 Thomas Jefferson University Methods and compositions for diagnosis and therapy of parkin-associated disorders
WO2006031210A1 (en) 2003-05-29 2006-03-23 Board Of Regents, The University Of Texas Systems Jabi as a prognostic marker and a therapeutic target for human cancer
EP1644492B1 (en) 2003-06-18 2009-01-07 Genelux Corporation Modified recombinant vaccinia viruses, uses thereof
CA2533701A1 (en) 2003-07-31 2005-02-17 Isis Pharmaceuticals, Inc. Oligomeric compounds and compositions for use in modulation of small non-coding rnas
US8106180B2 (en) 2003-08-07 2012-01-31 Whitehead Institute For Biomedical Research Methods and products for expression of micro RNAs
US20050037362A1 (en) 2003-08-11 2005-02-17 Eppendorf Array Technologies, S.A. Detection and quantification of siRNA on microarrays
US8412541B2 (en) 2003-08-14 2013-04-02 Edda Technology, Inc. Method and system for intelligent qualitative and quantitative analysis for medical diagnosis
US20050084883A1 (en) 2003-08-25 2005-04-21 The Johns Hopkins University School Of Medicine Method of diagnosis and treatment of pancreatic endocrine neoplasms based on differential gene expression analysis
EP1670955A2 (en) 2003-09-22 2006-06-21 Rosetta Inpharmatics LLC. Synthetic lethal screen using rna interference
EP1668155A2 (en) 2003-09-24 2006-06-14 Oncotherapy Science, Inc. Methods for detecting, diagnosing and treating hepatocellular carcinomas (hcc)
US20050186589A1 (en) 2003-11-07 2005-08-25 University Of Massachusetts Interspersed repetitive element RNAs as substrates, inhibitors and delivery vehicles for RNAi
US20050164252A1 (en) 2003-12-04 2005-07-28 Yeung Wah Hin A. Methods using non-genic sequences for the detection, modification and treatment of any disease or improvement of functions of a cell
DE602004031881D1 (en) 2003-12-19 2011-04-28 Univ California METHOD AND MATERIALS FOR ASSESSING PROSTATE ACID THERAPIES
WO2005078139A2 (en) 2004-02-09 2005-08-25 Thomas Jefferson University DIAGNOSIS AND TREATMENT OF CANCERS WITH MicroRNA LOCATED IN OR NEAR CANCER-ASSOCIATED CHROMOSOMAL FEATURES
US20050256072A1 (en) 2004-02-09 2005-11-17 University Of Massachusetts Dual functional oligonucleotides for use in repressing mutant gene expression
CA2556435C (en) 2004-02-13 2014-08-12 The Rockefeller University Anti-microrna oligonucleotide molecules
AU2005214286A1 (en) 2004-02-23 2005-09-01 Erasmus University Medical Center Rotterdam Classification, diagnosis and prognosis of acute myeloid leukemia by gene expression profiling
US7365058B2 (en) 2004-04-13 2008-04-29 The Rockefeller University MicroRNA and methods for inhibiting same
KR20070004957A (en) 2004-04-20 2007-01-09 제나코 바이오메디컬 프로덕츠, 인코포레이티드 Method for detecting ncrna
EP1784501B1 (en) 2004-05-14 2015-11-18 Rosetta Genomics Ltd VIRAL AND VIRUS ASSOCIATED MicroRNAS AND USES THEREOF
EP2290071B1 (en) 2004-05-28 2014-12-31 Asuragen, Inc. Methods and compositions involving microRNA
US7635563B2 (en) 2004-06-30 2009-12-22 Massachusetts Institute Of Technology High throughput methods relating to microRNA expression analysis
US20060037088A1 (en) 2004-08-13 2006-02-16 Shulin Li Gene expression levels as predictors of chemoradiation response of cancer
EP2338994B1 (en) 2004-09-02 2014-03-19 Yale University Regulation of oncogenes by microRNAs
US7592441B2 (en) 2004-10-04 2009-09-22 Rosetta Genomics Ltd Liver cancer-related nucleic acids
US7642348B2 (en) 2004-10-04 2010-01-05 Rosetta Genomics Ltd Prostate cancer-related nucleic acids
FR2877350B1 (en) 2004-11-03 2010-08-27 Centre Nat Rech Scient IDENTIFICATION AND USE OF miRNAs INVOLVED IN THE DIFFERENTIATION OF CELLS FROM MYELOID LEUKEMIA
DK2302055T3 (en) 2004-11-12 2014-10-13 Asuragen Inc Methods and compositions involving miRNA and miRNA inhibitor molecules
WO2006066158A2 (en) 2004-12-14 2006-06-22 Alnylam Pharmaceuticals, Inc. Rnai modulation of mll-af4 and uses thereof
US20060185027A1 (en) 2004-12-23 2006-08-17 David Bartel Systems and methods for identifying miRNA targets and for altering miRNA and target expression
EP1959012A3 (en) 2004-12-29 2009-12-30 Exiqon A/S Novel oligonucleotide compositions and probe sequences useful for detection and analysis of microRNAs and their target mRNAs
WO2006081284A2 (en) 2005-01-25 2006-08-03 Rosetta Inpharmatics Llc Methods for quantitating small rna molecules
US8071306B2 (en) 2005-01-25 2011-12-06 Merck Sharp & Dohme Corp. Methods for quantitating small RNA molecules
US20070065840A1 (en) 2005-03-23 2007-03-22 Irena Naguibneva Novel oligonucleotide compositions and probe sequences useful for detection and analysis of microRNAS and their target mRNAS
GB2425311A (en) 2005-04-15 2006-10-25 Ist Superiore Sanita Micro RNA against kit protein
US8247543B2 (en) 2005-04-29 2012-08-21 The Rockefeller University Human microRNAs and methods for inhibiting same
WO2006133022A2 (en) 2005-06-03 2006-12-14 The Johns Hopkins University Compositions and methods for decreasing microrna expression for the treatment of neoplasia
US20070065844A1 (en) 2005-06-08 2007-03-22 Massachusetts Institute Of Technology Solution-based methods for RNA expression profiling
US20060292616A1 (en) 2005-06-23 2006-12-28 U.S. Genomics, Inc. Single molecule miRNA-based disease diagnostic methods
CN103882124B (en) 2005-08-01 2015-11-18 俄亥俄州立大学研究基金会 For the method and composition based on MicroRNA of the diagnosis of mammary cancer, prognosis and treatment
US20070213292A1 (en) 2005-08-10 2007-09-13 The Rockefeller University Chemically modified oligonucleotides for use in modulating micro RNA and uses thereof
CA2618995A1 (en) 2005-08-10 2007-02-22 The Rockefeller University Antagomirs for use in inhibiting mir-122
AU2006291165B2 (en) 2005-09-12 2013-03-14 The Ohio State University Research Foundation Compositions and methods for the diagnosis and therapy of BCL2-associated cancers
WO2007044413A2 (en) 2005-10-05 2007-04-19 The Ohio State University Research Foundation Wwox gene, vectors containing the same, and uses in treatment of cancer
US20070092882A1 (en) 2005-10-21 2007-04-26 Hui Wang Analysis of microRNA
US7390792B2 (en) 2005-12-15 2008-06-24 Board Of Regents, The University Of Texas System MicroRNA1 therapies
CA2633754C (en) 2006-01-05 2013-06-18 The Ohio State University Research Foundation Microrna-based methods and compositions for the diagnosis and treatment of solid cancers
EP2591794A1 (en) 2006-01-05 2013-05-15 The Ohio State University Research Foundation MicroRNA expressions abnormalities in pancreatic endocrine and acinar tumors
WO2007081720A2 (en) 2006-01-05 2007-07-19 The Ohio State University Research Foundation Microrna-based methods and compositions for the diagnosis, prognosis and treatment of lung cancer
US20090007281A1 (en) 2006-01-13 2009-01-01 Battelle Memorial Institute Animal Model for Assessing Copd-Related Diseases
EP2371971B1 (en) 2006-03-20 2013-11-27 The Ohio State University Research Foundation Microrna fingerprints during human megakaryocytopoiesis
WO2007112097A2 (en) 2006-03-24 2007-10-04 Children's Medical Center Corporation Novel signature self renewal gene expression programs
DK2666859T3 (en) 2006-04-03 2019-04-08 Roche Innovation Ct Copenhagen As PHARMACEUTICAL COMPOSITION CONTAINING ANTI-miRNA ANTISENSE OLIGONUCLEOTIDES
WO2007127190A2 (en) 2006-04-24 2007-11-08 The Ohio State University Research Foundation Pre-b cell proliferation and lymphoblastic leukemia/high-grade lymphoma in mir155 transgenic mice
EP2455493B1 (en) 2006-07-13 2014-01-08 The Ohio State University Research Foundation Micro-RNA-based methods and compositions for the diagnosis and treatment of colon related diseases
AU2007297823B2 (en) 2006-08-30 2012-04-19 The Regents Of The University Of Michigan New small molecule inhibitors of MDM2 and the uses thereof
US20080193943A1 (en) 2006-09-05 2008-08-14 Abbott Laboratories Companion diagnostic assays for cancer therapy
EP2115138A2 (en) 2006-09-19 2009-11-11 Asuragen, Inc. Micrornas differentially expressed in pancreatic diseases and uses thereof
CA2663962A1 (en) 2006-09-19 2008-03-27 Asuragen, Inc. Mir-15, mir-26, mir-31,mir-145, mir-147, mir-188, mir-215, mir-216, mir-331, mmu-mir-292-3p regulated genes and pathways as targets for therapeutic intervention
EP2061907B1 (en) 2006-09-19 2011-11-23 The Ohio State University Research Foundation Tcl1 expression in chronic lymphocytic leukemia (cll) regulated by mir-29 and mir-181
CA2663878A1 (en) 2006-09-19 2008-03-27 Asuragen, Inc. Mir-200 regulated genes and pathways as targets for therapeutic intervention
WO2008054828A2 (en) 2006-11-01 2008-05-08 The Ohio State University Research Foundation Microrna expression signature for predicting survival and metastases in hepatocellular carcinoma
US8293684B2 (en) 2006-11-29 2012-10-23 Exiqon Locked nucleic acid reagents for labelling nucleic acids
WO2008070082A2 (en) 2006-12-04 2008-06-12 The Johns Hopkins University Stem-progenitor cell specific micro-ribonucleic acids and uses thereof
CA2671294A1 (en) 2006-12-08 2008-06-19 Asuragen, Inc. Mir-21 regulated genes and pathways as targets for therapeutic intervention
CA2671299A1 (en) 2006-12-08 2008-06-19 Asuragen, Inc. Functions and targets of let-7 micro rnas
EP2104734A2 (en) 2006-12-08 2009-09-30 Asuragen, INC. Mir-20 regulated genes and pathways as targets for therapeutic intervention
CN101622348A (en) 2006-12-08 2010-01-06 奥斯瑞根公司 Gene and the approach regulated as the miR-20 of targets for therapeutic intervention
CA2671270A1 (en) 2006-12-29 2008-07-17 Asuragen, Inc. Mir-16 regulated genes and pathways as targets for therapeutic intervention
CN103555825B (en) 2007-01-31 2015-09-30 俄亥俄州立大学研究基金会 For the method and composition based on microRNA of the diagnosis of acute myelocytic leukemia (AML), prognosis and treatment
WO2008104974A2 (en) 2007-02-27 2008-09-04 Rosetta Genomics Ltd. Composition and methods for modulating cell proliferation and cell death
EP2126584B1 (en) 2007-03-16 2012-12-19 CovalX AG Direct mass spectrometric analysis of drug candidates targeting protein complexes
WO2008124777A1 (en) 2007-04-10 2008-10-16 National Taiwan University Predicting post-treatment survival in cancer patients with micrornas
US20100144850A1 (en) 2007-04-30 2010-06-10 The Ohio State University Research Foundation Methods for Differentiating Pancreatic Cancer from Normal Pancreatic Function and/or Chronic Pancreatitis
US20090005336A1 (en) 2007-05-08 2009-01-01 Zhiguo Wang Use of the microRNA miR-1 for the treatment, prevention, and diagnosis of cardiac conditions
US20090131354A1 (en) 2007-05-22 2009-05-21 Bader Andreas G miR-126 REGULATED GENES AND PATHWAYS AS TARGETS FOR THERAPEUTIC INTERVENTION
US20090232893A1 (en) 2007-05-22 2009-09-17 Bader Andreas G miR-143 REGULATED GENES AND PATHWAYS AS TARGETS FOR THERAPEUTIC INTERVENTION
WO2008154098A2 (en) 2007-06-07 2008-12-18 Wisconsin Alumni Research Foundation Reagents and methods for mirna expression analysis and identification of cancer biomarkers
US8465917B2 (en) 2007-06-08 2013-06-18 The Ohio State University Research Foundation Methods for determining heptocellular carcinoma subtype and detecting hepatic cancer stem cells
AU2008261951A1 (en) 2007-06-08 2008-12-18 Asuragen, Inc. miR-34 regulated genes and pathways as targets for therapeutic intervention
CN101918424A (en) 2007-06-15 2010-12-15 俄亥俄州立大学研究基金会 Oncogenic ALL-1 fusion proteins for targeting Drosha-mediated microRNA processing
US8367632B2 (en) 2007-07-31 2013-02-05 Ohio State University Research Foundation Methods for reverting methylation by targeting methyltransferases
EP2173908B1 (en) 2007-08-03 2016-01-06 The Ohio State University Research Foundation Ultraconserved regions encoding ncrnas
WO2009026487A1 (en) 2007-08-22 2009-02-26 The Ohio State University Research Foundation Methods and compositions for inducing deregulation of epha7 and erk phosphorylation in human acute leukemias
US20090061424A1 (en) 2007-08-30 2009-03-05 Sigma-Aldrich Company Universal ligation array for analyzing gene expression or genomic variations
EP2775001B1 (en) 2007-09-06 2016-03-09 The Ohio State University Research Foundation MicroRNA signatures in human ovarian cancer
CA2702241A1 (en) 2007-10-11 2009-04-16 The Ohio State University Research Foundation Methods and compositions for the diagnosis and treatment of esophageal adenocarcinomas
CN103898069A (en) 2007-10-26 2014-07-02 俄亥俄州立大学研究基金会 Methods for identifying fragile histidine triad (fhit) interaction and uses thereof
AU2008321253B2 (en) 2007-11-12 2014-01-16 Masaryk Memorial Cancer Institute Therapeutic applications of p53 isoforms in regenerative medicine, aging and cancer
US20090123933A1 (en) 2007-11-12 2009-05-14 Wake Forest University Health Sciences Microrna biomarkers in lupus
JP2011505143A (en) 2007-11-30 2011-02-24 ジ・オハイオ・ステイト・ユニバーシティ・リサーチ・ファウンデイション MicroRNA expression profiling and peripheral blood targeting in lung cancer
WO2009070805A2 (en) 2007-12-01 2009-06-04 Asuragen, Inc. Mir-124 regulated genes and pathways as targets for therapeutic intervention
WO2009086156A2 (en) 2007-12-21 2009-07-09 Asuragen, Inc. Mir-10 regulated genes and pathways as targets for therapeutic intervention
ES2936256T3 (en) 2008-02-01 2023-03-15 Massachusetts Gen Hospital Use of microvesicles in the diagnosis, and prognosis of diseases and medical conditions
WO2009100430A2 (en) 2008-02-08 2009-08-13 Asuragen, Inc miRNAs DIFFERENTIALLY EXPRESSED IN LYMPH NODES FROM CANCER PATIENTS
CA2716906A1 (en) 2008-02-28 2009-09-03 The Ohio State University Research Foundation Microrna-based methods and compositions for the diagnosis, prognosis and treatment of gastric cancer
EP3112477A1 (en) 2008-02-28 2017-01-04 The Ohio State University Research Foundation Microrna-based methods and compositions for the diagnosis, prognosis and treatment of prostate related disorders
CN102007408A (en) 2008-02-28 2011-04-06 俄亥俄州立大学研究基金会 Microrna signatures associated with cytogenetics and prognosis in acute myeloid leukemia (aml) and uses thereof
US20110052502A1 (en) 2008-02-28 2011-03-03 The Ohio State University Research Foundation MicroRNA Signatures Associated with Human Chronic Lymphocytic Leukemia (CCL) and Uses Thereof
EP2268832A2 (en) 2008-03-06 2011-01-05 Asuragen, INC. Microrna markers for recurrence of colorectal cancer
WO2009154835A2 (en) 2008-03-26 2009-12-23 Asuragen, Inc. Compositions and methods related to mir-16 and therapy of prostate cancer
WO2009137807A2 (en) 2008-05-08 2009-11-12 Asuragen, Inc. Compositions and methods related to mirna modulation of neovascularization or angiogenesis
CN102112110A (en) 2008-06-06 2011-06-29 米尔纳医疗股份有限公司 Novel compositions for the in vivo delivery of RNAi agents
CN102149827B (en) 2008-06-11 2014-08-20 由卫生与公众服务部代表的美利坚合众国政府 Use of MiR-26 family as a predictive marker of hepatocellular carcinoma and responsiveness to therapy
WO2010019694A1 (en) 2008-08-12 2010-02-18 The Ohio State University Research Foundation Micro-rna-based compositions and methods for the diagnosis, prognosis and treatment of multiple myeloma
JP2012509886A (en) 2008-11-21 2012-04-26 ジ・オハイオ・ステイト・ユニバーシティ・リサーチ・ファウンデイション Tcl1 as a transcriptional regulator
WO2010065156A1 (en) 2008-12-05 2010-06-10 The Ohio State University Research Foundation Microrna-based methods and compositions for the diagnosis and treatment of ovarian cancer
CN102549166A (en) 2009-02-26 2012-07-04 俄亥俄州立大学研究基金会 Micrornas in never-smokers and related materials and methods

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070178105A1 (en) * 1996-02-09 2007-08-02 Croce Carlo M FHIT proteins and nucleic acids and methods based thereon

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Kim et al.; FHIT protein enhances paclitaxel-induced apoptosis in lung cancer cells; Int. J. Cancer: 118, 1692–1698; published online October 17, 2005 *
Zawacka-Pankau et al. Expression and simple, one-step purification of fragile histidine triad (Fhit) tumor suppressor mutant forms in Escherichia coli and their interaction with protoporphyrin IX. Biotechnol Lett. 2007 Jun;29(6):877-83. Epub 2007 Feb 20. *

Also Published As

Publication number Publication date
CN103898069A (en) 2014-07-02
EP2201112A4 (en) 2011-02-02
US20120010092A1 (en) 2012-01-12
CN102137927A (en) 2011-07-27
AU2008316577A1 (en) 2009-04-30
WO2009055773A2 (en) 2009-04-30
JP2011504093A (en) 2011-02-03
US20160375107A1 (en) 2016-12-29
US8911998B2 (en) 2014-12-16
WO2009055773A3 (en) 2009-06-18
EP2201112A2 (en) 2010-06-30
CN102137927B (en) 2014-03-12
CA2703707A1 (en) 2009-04-30
AU2008316577B2 (en) 2014-04-10

Similar Documents

Publication Publication Date Title
US20160375107A1 (en) Fragile histidine triad (fhit) compositions and uses thereof
Trapasso et al. Fhit interaction with ferredoxin reductase triggers generation of reactive oxygen species and apoptosis of cancer cells
Li Survivin study: what is the next wave?
US20160313349A1 (en) Method of diagnosis and treatment
US20150252436A1 (en) Methods and compositions for the diagnosis, prognosis and treatment of cancer
US20230279496A1 (en) Clear Cell Renal Cell Carcinoma Biomarkers
Zhu et al. Integrin alpha 7 interacts with high temperature requirement A2 (HtrA2) to induce prostate cancer cell death
JP2015521475A (en) BAG3 as a biochemical serum marker and tissue marker
WO2016152352A1 (en) Melanoma-specific biomarker and use thereof
US20100098715A1 (en) Annexin ii compositions for treating or monitoring inflammation or immune-mediated disorders
Cai et al. COLEC10 induces endoplasmic reticulum stress by occupying GRP78 and inhibits hepatocellular carcinoma
AU2014203456B2 (en) Methods for identifying fragile histidine triad (FHIT) interaction and uses thereof
JP2005518222A (en) Agents and methods for identifying and modulating the expression of genes regulated by CDK inhibitors
JP4871630B2 (en) A method for inhibiting cell growth, comprising inhibiting phosphatase
US20230375528A1 (en) Screening method for the identification of novel therapeutic compounds
JP2007267660A (en) Method for screening anti-cancer agent
JP2010117358A (en) Protein involved in cancer
US20030083249A1 (en) Methods of inducing apoptosis in hyperproliferative cells
Zhang et al. TRIM47-CDO1 axis dictates hepatocellular carcinoma progression by modulating ferroptotic cell death through the ubiquitin‒proteasome system
Sojka et al. The human testis-enriched HSPA2 interacts with HIF-1α in epidermal keratinocytes, yet HIF-1α stability and HIF-1-dependent gene expression rely on the HSPA (HSP70) activity
US9079938B2 (en) ASPP2 splicing variant
WO2012156928A1 (en) Vdac1-s as a cell marker
EP1451218A2 (en) Methods for the treatment and prognosis of leukemia and other cancer types
AU2002326111A1 (en) Methods for the treatment and prognosis of leukemia and other cancer types

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE OHIO STATE UNIVERSITY, OHIO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CROCE, CARLO M.;TRAPASSO, FRANCESCO;SIGNING DATES FROM 20110214 TO 20110221;REEL/FRAME:033718/0816

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:OHIO STATE UNIVERSITY;REEL/FRAME:037119/0725

Effective date: 20151112

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH, MARYLAND

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:THE OHIO UNIVERSITY;REEL/FRAME:053125/0200

Effective date: 20200706

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH, MARYLAND

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:THE OHIO STATE UNIVERSITY;REEL/FRAME:053185/0336

Effective date: 20200713