WO1999004636A1 - Evaluation of, delivery of, and use of agents to treat heart disorders - Google Patents

Evaluation of, delivery of, and use of agents to treat heart disorders Download PDF

Info

Publication number
WO1999004636A1
WO1999004636A1 PCT/US1998/014968 US9814968W WO9904636A1 WO 1999004636 A1 WO1999004636 A1 WO 1999004636A1 US 9814968 W US9814968 W US 9814968W WO 9904636 A1 WO9904636 A1 WO 9904636A1
Authority
WO
WIPO (PCT)
Prior art keywords
heart
treatment
phospholamban
evaluating
cell
Prior art date
Application number
PCT/US1998/014968
Other languages
French (fr)
Other versions
WO1999004636A9 (en
Inventor
Roger J. Hajjar
Anthony Rosenzweig
Luis Guerrero
Original Assignee
The General Hospital Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The General Hospital Corporation filed Critical The General Hospital Corporation
Priority to CA002297328A priority Critical patent/CA2297328A1/en
Priority to EP98934656A priority patent/EP1009236A4/en
Publication of WO1999004636A1 publication Critical patent/WO1999004636A1/en
Publication of WO1999004636A9 publication Critical patent/WO1999004636A9/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1138Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against receptors or cell surface proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • 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
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid
    • C12N2799/022Uses of viruses as vector for the expression of a heterologous nucleic acid where the vector is derived from an adenovirus
    • 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
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid
    • C12N2799/027Uses of viruses as vector for the expression of a heterologous nucleic acid where the vector is derived from a retrovirus

Definitions

  • This invention relates to methods for evaluating treatments suitable for heart disorders and to methods for delivering compounds to the heart of a subject.
  • SR sarcoplasmic reticulum
  • SERCA2a SR Ca--- "1" -ATPase activity
  • Phospholamban is a small phosphoprotein, about 6,080 daltons in size, which is an integral element of the cardiac SR membrane. Phospholamban is phosphorylated in vivo in response to ⁇ -adrenergic agonist stimulation. In the dephosphorylated state, phospholamban inhibits SR Ca2 + -ATPase activity by decreasing the affinity of the enzyme for Ca2 + .
  • Heart failure is characterized by a number of abnormalities at the cellular level in the various steps of excitation-contraction coupling of the cardiac cells.
  • One of the key abnormalities in both human and experimental heart failure is a defect in SR function which is associated with abnormal intracellular Ca ⁇ "1" handling.
  • Deficient SR Ca ⁇ + uptake during relaxation has been identified in failing hearts from both humans and animal models and has been associated with a decrease in the activity of SR Ca-*- "1" - ATPase activity and altered Ca ⁇ + kinetics.
  • the invention features, a method of evaluating a treatment for a heart disorder.
  • the method includes: providing a heart cell, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; administering the treatment to the heart cell; and evaluating the effect of the treatment on the heart cell, thereby evaluating the treatment for a heart disorder.
  • the method includes evaluating the effect of the treatment on a parameter related to heart function.
  • the parameter can include an assessment of contractility, Ca2 + metabolism, e.g., intracellular Cs? + concentration, SR Ca2 + ATPase activity, force generation, a force frequency relationship, cardiocyte survival or apoptosis or ion channel activity, e.g., sodium calcium exchange, sodium channel activity, calcium channel activity, or sodium potassium ATPase pump activity.
  • the treatment is administered in vivo, e.g., to an experimental animal.
  • the experimental animal can be an animal in which a gene related to cardiac structure or function is misexpressed. Misexpression can be achieved by methods known in the art, for example, by transgenesis, including the creation of knockout animals, or by classic breeding experiments or manipulation.
  • the misexpressed gene can be a gene encoding a sarcomeric protein, a gene encoding a protein which conditions cardiocyte survival or apoptosis, or a gene encoding a calcium regulatory protein.
  • Sarcomeric proteins include myosin heavy chain, troponin I, troponin C, troponin T, tropomyosin, actin, myosin light chain kinase, myosin light chain 1, myosin light chain 2 or myosin light chain 3.
  • Proteins which modify cardiocyte survival or apoptosis include IGF-1 receptor, PI3 kinase, AKT kinase or members of the caspase family of proteins.
  • Calcium regulatory proteins include phospholamban, SR Ca2 + ATPase, sodium-calcium exchanger, calcium channel (L and T), calsequestrin or calreticulin.
  • the experimental animal can be an animal model for a disorder, e.g., a heart disorder.
  • the treatment is administered in vitro.
  • the cell is derived from an experimental animal or a human.
  • the cell can be cultured and/or immortalized.
  • the nucleic acid encodes a phospholamban protein.
  • the phospholamban can be from the same species that the heart cell is from or it can be from a different species.
  • a mouse phospholamban can be expressed in a mouse cell or a human phospholamban can be expressed in a cell from an experimental animal.
  • the nucleic acid is introduced into the heart cell by way of a vector suitable for somatic gene transfer, e.g., a viral vector, e.g., an adenoviral vector.
  • a vector suitable for somatic gene transfer e.g., a viral vector, e.g., an adenoviral vector.
  • the invention features, a method of evaluating a treatment for a heart disorder. The method includes: providing a heart, into some or all the cells of which has been introduced, by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; administering the treatment to the heart; and evaluating the effect of the treatment on the heart, thereby evaluating the treatment for a heart disorder.
  • the method includes evaluating the effect of the treatment on a parameter related to heart function.
  • the parameter can include an assessment of contractility, Ca2+ metabolism, e.g., intracellular C ⁇ + concentration, SR Ca-"- "1" ATPase activity, force generation, a force frequency relationship, cardiocyte survival or apoptosis or ion channel activity, e.g., sodium calcium exchange, sodium channel activity, calcium channel activity, or sodium potassium ATPase pump activity.
  • an assessment of contractility, Ca2+ metabolism e.g., intracellular C ⁇ + concentration
  • SR Ca-"- "1" ATPase activity e.g., force generation, a force frequency relationship
  • cardiocyte survival or apoptosis or ion channel activity e.g., sodium calcium exchange, sodium channel activity, calcium channel activity, or sodium potassium ATPase pump activity.
  • the treatment is administered in vivo, e.g., to an experimental animal.
  • the experimental animal can be an animal in which a gene related to cardiac structure or function is misexpressed. Misexpression can be achieved by methods known in the art, for example, by transgenesis, including the creation of knockout animals, or by classic breeding experiments or manipulation.
  • the misexpressed gene can be a gene encoding a sarcomeric protein, a gene encoding a protein which conditions cardiocyte survival or apoptosis, or a gene encoding a calcium regulatory protein.
  • Sarcomeric proteins include myosin heavy chain, troponin I, troponin C, troponin T, tropomyosin, actin, myosin light chain kinase, myosin light chain 1, myosin light chain 2 or myosin light chain 3.
  • Proteins which modify cardiocyte survival or apoptosis include IGF-1 receptor, PI3 kinase, AKT kinase or members of the caspase family of proteins.
  • Calcium regulatory proteins include phospholamban, SR Ca2 + ATPase, sodium-calcium exchanger, calcium channel (L and T), calsequestrin or calreticulin.
  • the experimental animal can be an animal model for a disorder, e.g., a heart disorder.
  • the treatment is administered in vitro.
  • the heart is derived from an experimental animal or a human.
  • the nucleic acid encodes a phospholamban protein.
  • the phospholamban can be from the same species that the heart is from or it can be from a different species.
  • a mouse phospholamban can be expressed in a mouse heart or a human phospholamban can be expressed in the heart of an experimental animal.
  • the phospholamban can be delivered to the heart using methods described herein.
  • the nucleic acid is introduced into the heart by way of a vector suitable for somatic gene transfer, e.g., a viral vector, e.g., an adenoviral vector.
  • a vector suitable for somatic gene transfer e.g., a viral vector, e.g., an adenoviral vector.
  • the invention features, a method of evaluating a treatment for a heart disorder. The method includes: providing heart tissue into some or all of the cells of which has been introduced, by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; administering the treatment to the heart tissue; and evaluating the effect of the treatment on the heart tissue, thereby evaluating the treatment for a heart disorder.
  • the method includes evaluating the effect of the treatment on a parameter related to heart function.
  • the parameter can include an assessment of contractility, Ca--- 4" metabolism, e.g., intracellular Ca ⁇ "1" concentration, SR Ca- ⁇ "1" ATPase activity, force generation, a force frequency relationship, cardiocyte survival or apoptosis or ion channel activity, e.g., sodium calcium exchange, sodium channel activity, calcium channel activity, or sodium potassium ATPase pump activity.
  • the treatment is administered in vivo, e.g., to an experimental animal.
  • the experimental animal can be an animal in which a gene related to cardiac structure or function is misexpressed.
  • Misexpression can be achieved by methods known in the art, for example, by transgenesis, including the creation of knockout animals, or by classic breeding experiments or manipulation.
  • the misexpressed gene can be a gene encoding a sarcomeric protein, a gene encoding a protein which conditions cardiocyte survival or apoptosis, or a gene encoding a calcium regulatory protein.
  • Sarcomeric proteins include myosin heavy chain, troponin I, troponin C, troponin T, tropomyosin, actin, myosin light chain kinase, myosin light chain 1, myosin light chain 2 or myosin light chain 3.
  • Proteins which modify cardiocyte survival or apoptosis include IGF-1 receptor, PI3 kinase, AKT kinase or members of the caspase family of proteins.
  • Calcium regulatory proteins include phospholamban, SR Ca2 + ATPase, sodium-calcium exchanger, calcium channel (L and T), calsequestrin or calreticulin.
  • the experimental animal can be an animal model for a disorder, e.g., a heart disorder.
  • the treatment is administered in vitro.
  • the heart tissue is derived from an experimental animal or a human.
  • the nucleic acid encodes a phospholamban protein.
  • the phospholamban can be from the same species that the heart tissue is from or it can be from a different species.
  • a mouse phospholamban can be expressed in a mouse heart tissue or a human phospholamban can be expressed in heart tissue from an experimental animal.
  • the nucleic acid is introduced into the heart tissue by way of a vector suitable for somatic gene transfer, e.g., a viral vector, e.g., an adenoviral vector.
  • a vector suitable for somatic gene transfer e.g., a viral vector, e.g., an adenoviral vector.
  • the invention features, a method of evaluating a treatment for a heart disorder.
  • the method includes: providing a first and a second heart cell, into each of which has been introduced, by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; administering the treatment to a first heart cell, preferably in vitro; evaluating the effect of the treatment on the first heart cell; administering the treatment to a second heart cell, preferably in vivo; and evaluating the effect of the treatment on the second heart cell, thereby evaluating the treatment for a heart disorder.
  • the method includes evaluating the effect of the treatment on a parameter related to heart function.
  • the parameter can include an assessment of contractility, Ca ⁇ "1" metabolism, e.g., intracellular Ca ⁇ 4" concentration, SR Ca2 + ATPase activity, force generation, a force frequency relationship, cardiocyte survival or apoptosis or ion channel activity, e.g., sodium calcium exchange, sodium channel activity, calcium channel activity, or sodium potassium ATPase pump activity.
  • the treatment is administered in vivo, e.g., to an experimental animal.
  • the experimental animal can be an animal in which a gene related to cardiac structure or function is misexpressed. Misexpression can be achieved by methods known in the art, for example, by transgenesis, including the creation of knockout animals, or by classic breeding experiments or manipulation.
  • the misexpressed gene can be a gene encoding a sarcomeric protein, a gene encoding a protein which conditions cardiocyte survival or apoptosis, or a gene encoding a calcium regulatory protein.
  • Sarcomeric proteins include myosin heavy chain, troponin I, troponin C, troponin T, tropomyosin, actin, myosin light chain kinase, myosin light chain 1, myosin light chain 2 or myosin light chain 3.
  • Proteins which modify cardiocyte survival or apoptosis include IGF-1 receptor, PI3 kinase, AKT kinase or members of the caspase family of proteins.
  • Calcium regulatory proteins include phospholamban, SR Ca2 + ATPase, sodium-calcium exchanger, calcium channel (L and T), calsequestrin or calreticulin.
  • the experimental animal can be an animal model for a disorder, e.g., a heart disorder.
  • the nucleic acid encodes a phospholamban protein.
  • the phospholamban can be from the same species that the heart cell is from or it can be from a different species.
  • a mouse phospholamban can be expressed in a mouse cell or a human phospholamban can be expressed in a cell from an experimental animal.
  • the first and second cell can be from the same or different animals, can be from the same or different species, e.g., the first cell can be from a mouse and the second cell can be from a human or both cells can be human.
  • the first and second cell can have the same or different genotypes.
  • the evaluation of the treatment in the first cell can be the same or different from the evaluation of the treatment in the second cell, e.g., the intracellular Ca- ⁇ 4" concentration can be measured in the first cell and the SR Ca ⁇ " -ATPase activity can be measured in the second cell or the intracellular Ca- ⁇ 4" concentration can be measured in both cells.
  • the invention features, a method of evaluating a treatment for a heart disorder.
  • the method includes: providing a first administration of a treatment to a heart cell, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; evaluating the effect of the first administration on the heart cell; providing a second administration of a treatment to a heart cell, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; and evaluating the effect of the second administration on the heart cell, thereby evaluating a treatment for a heart disorder.
  • the method includes evaluating the effect of the treatment on a parameter related to heart function.
  • the parameter can include an assessment of contractility, Ca- ⁇ 4" metabolism, e.g., intracellular Ca ⁇ 4" concentration, SR Ca--- 4" ATPase activity, force generation, a force frequency relationship, cardiocyte survival or apoptosis or ion channel activity, e.g., sodium calcium exchange, sodium channel activity, calcium channel activity, or sodium potassium ATPase pump activity.
  • an assessment of contractility, Ca- ⁇ 4" metabolism e.g., intracellular Ca ⁇ 4" concentration, SR Ca--- 4" ATPase activity, force generation, a force frequency relationship, cardiocyte survival or apoptosis or ion channel activity, e.g., sodium calcium exchange, sodium channel activity, calcium channel activity, or sodium potassium ATPase pump activity.
  • the treatment is administered in vivo, e.g., to an experimental animal.
  • the experimental animal can be an animal in which a gene related to cardiac structure or function is misexpressed. Misexpression can be achieved by methods known in the art, for example, by transgenesis, including the creation of knockout animals, or by classic breeding experiments or manipulation.
  • the misexpressed gene can be a gene encoding a sarcomeric protein, a gene encoding a protein which conditions cardiocyte survival or apoptosis, or a gene encoding a calcium regulatory protein.
  • Sarcomeric proteins include myosin heavy chain, troponin I, troponin C, troponin T, tropomyosin, actin, myosin light chain kinase, myosin light chain 1, myosin light chain 2 or myosin light chain 3.
  • Proteins which modify cardiocyte survival or apoptosis include IGF-1 receptor, PI3 kinase, AKT kinase or members of the caspase family of proteins.
  • Calcium regulatory proteins include phospholamban, SR Ca2 + ATPase, sodium-calcium exchanger, calcium channel (L and T), calsequestrin or calreticulin.
  • the experimental animal can be an animal model for a disorder, e.g., a heart disorder.
  • the nucleic acid encodes a phospholamban protein.
  • the phospholamban can be from the same species that the heart cell is from or it can be from a different species.
  • a mouse phospholamban can be expressed in a mouse cell or a human phospholamban can be expressed in a cell from an experimental animal.
  • the first and second administration can be administered to the same or to different cells.
  • the first and second administration can be administered under the same or different conditions, e.g., the first administration can consist of a relatively low level treatment, e.g., a lower concentration of a substance, and the second administration can consist of a relatively high level treatment, e.g., a higher concentration of a substance or both administrations can consist of the same level treatment.
  • the invention features, a method of evaluating a treatment for a heart disorder.
  • the method includes: providing a heart cell, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; administering the treatment to the heart cell; evaluating the effect of the treatment on the heart cell; providing a heart, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; administering the treatment to the heart ; and evaluating the effect of the treatment on the heart, thereby evaluating the treatment for a heart disorder.
  • the method includes evaluating the effect of the treatment on a parameter related to heart function.
  • the parameter can include an assessment of contractility, Ca ⁇ " metabolism, e.g., intracellular Ca- ⁇ 4" concentration, SR Ca-"- 4" ATPase activity, force generation, a force frequency relationship, cardiocyte survival or apoptosis or ion channel activity, e.g., sodium calcium exchange, sodium channel activity, calcium channel activity, or sodium potassium ATPase pump activity.
  • the treatment is administered in vivo, e.g., to an experimental animal.
  • the experimental animal can be an animal in which a gene related to cardiac structure or function is misexpressed. Misexpression can be achieved by methods known in the art, for example, by transgenesis, including the creation of knockout animals, or by classic breeding experiments or manipulation.
  • the misexpressed gene can be a gene encoding a sarcomeric protein, a gene encoding a protein which conditions cardiocyte survival or apoptosis, or a gene encoding a calcium regulatory protein.
  • Sarcomeric proteins include myosin heavy chain, troponin I, troponin C, troponin T, tropomyosin, actin, myosin light chain kinase, myosin light chain 1, myosin light chain 2 or myosin light chain 3.
  • Proteins which modify cardiocyte survival or apoptosis include IGF-1 receptor, PI3 kinase, AKT kinase or members of the caspase family of proteins.
  • Calcium regulatory proteins include phospholamban, SR Ca2 + ATPase, sodium-calcium exchanger, calcium channel (L and T), calsequestrin or calreticulin.
  • the experimental animal can be an animal model for a disorder, e.g., a heart disorder.
  • the nucleic acid encodes a phospholamban protein.
  • the phospholamban can be from the same species that the heart cell and/or the heart is from or it can be from a different species.
  • a mouse phospholamban can be expressed in a mouse heart cell and/or heart or a human phospholamban can be expressed in a heart cell and/or heart from an experimental animal.
  • the treatment can be administered to the heart cell in vitro and to the heart in vivo or the treatment can be administered to the heart cell and to the heart in vitro.
  • the invention features, a method of delivering a compound to the heart of a subject. The method includes: restricting the aortic flow of blood out of the heart, such that blood flow is re-directed to the coronary arteries; introducing the compound into the lumen of the circulatory system such that it flows into the coronary arteries; allowing the heart to pump while the aortic outflow of blood is restricted, thereby allowing the compound to flow into and be delivered to the heart; and reestablishing the flow of blood to the heart .
  • the compound includes: a nucleic acid which directs the expression of a peptide, e.g., a phospholamban or a SR Ca ⁇ 4" -ATPase and a viral vector suitable for somatic gene delivery, e.g., an adenoviral vector.
  • a heart disorder e.g., heart failure, ischemia, arrhythmia, myocardial infarction, congestive heart failure, transplant rejection.
  • the subject can be a human or an experimental animal.
  • the experimental animal can be an animal in which a gene related to cardiac structure or function is misexpressed. Misexpression can be achieved by methods known in the art, for example, by transgenesis, including the creation of knockout animals, or by classic breeding experiments or manipulation.
  • the misexpressed gene can be a gene encoding a sarcomeric protein, a gene encoding a protein which conditions cardiocyte survival or apoptosis, or a gene encoding a calcium regulatory protein.
  • Sarcomeric proteins include myosin heavy chain, troponin I, troponin C, troponin T, tropomyosin, actin, myosin light chain kinase, myosin light chain 1, myosin light chain 2 or myosin light chain 3.
  • Proteins which modify cardiocyte survival or apoptosis include IGF-1 receptor, PI3 kinase, AKT kinase or members of the caspase family of proteins.
  • Calcium regulatory proteins include phospholamban, SR Ca2 + ATPase, sodium-calcium exchanger, calcium channel (L and T), calsequestrin or calreticulin.
  • the experimental animal can be an animal model for a disorder, e.g., a heart disorder.
  • the method further includes restricting blood flow into the left side of the heart, e.g., by restricting the pulmonary circulation through obstraction of the pulmonary artery, so as to lessen dilution of the compound.
  • the method further includes opening the pericardium and introducing the compound, e.g., using a catheter.
  • the compound is: introduced into the lumen of the aorta, e.g., the aortic root, introduced into the coronary ostia or introduced into the lumen of the heart.
  • the nucleic acid which directs the expression of the peptide, is homogeneously overexpressed in the heart of the subject.
  • the invention features, a heart cell, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban.
  • the heart cell can be provided as a purified preparation.
  • the invention features, a heart tissue, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban.
  • the heart tissue can be provided as a tissue preparation.
  • the invention features, a heart, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban.
  • the heart can be provided in a subject or ex vivo, i.e. removed from a subject.
  • the invention features, a method for treating a subject at risk for a heart disorder.
  • the method includes introducing into somatic heart tissue of the subject, a nucleic acid which encodes phospholamban.
  • the nucleic acid is introduced using the methods described herein.
  • the phospholamban can be from the same species as the subject or it can be from a different species.
  • a human phospholamban can be introduced into a human heart or a human phospholamban can be can be introduced into the heart of an experimental animal.
  • the nucleic acid is introduced into the heart by way of a vector suitable for somatic gene transfer, e.g., a viral vector, e.g., an adenoviral vector.
  • the subject can be a human, an experimental animal, e.g., a rat or a mouse, a domestic animal, e.g., a dog, cow, sheep, pig or horse, or a non- human primate, e.g., a monkey.
  • the subject can be suffering from a cardiac disorder, such as heart failure, ischemia, myocardial infarction, congestive heart failure, arrhythmia, transplant rejection and the like.
  • the term "treatment” refers to a procedure (e.g., a surgical method) or the administration of a substance, e.g., a compound which is being evaluated for use in the alleviation or prevention of a heart disorder or symptoms thereof.
  • a procedure e.g., a surgical method
  • a therapeutic agent such as a drug, a peptide, an antibody, an ionophore and the like.
  • the term "heart disorder” refers to a structural or functional abnormality of the heart, that impairs its normal functioning.
  • the heart disorder can be heart failure, ischemia, myocardial infarction, congestive heart failure, arrhythmia, transplant rejection and the like.
  • the term includes disorders characterized by abnormalities of contraction, abnormalities in Ca ⁇ 4" metabolism, and disorders characterized by arrhytmia.
  • heart cell refers to a cell which can be: (a) part of a heart present in a subject, (b) part of a heart which is maintained in vitro, (c) part of a heart tissue, or (d) a cell which is isolated from the heart of a subject.
  • the cell can be a cardiac myocyte.
  • heart refers to a heart present in a subject or to a heart which is maintained outside a subject.
  • heart tissue refers to tissue which is derived from the heart of a subject.
  • simatic gene transfer refers to the transfer of genes into a somatic cell as opposed to transferring genes into the germ line.
  • the term "compound” refers to a compound, which can be delivered effectively to the heart of a subject using the methods of the invention. Such compounds can include, for example, a gene, a drug, an antibiotic, an enzyme, a chemical compound, a mixture of chemical compounds or a biological macromolecule.
  • the term “subject” refers to an experimental animal, e.g., a rat or a mouse, a domestic animal, e.g., a dog, cow, sheep, pig or horse, a non-human primate, e.g., a monkey and in the case of therapeutic methods, humans. However, it is noted that human cells, tissue or hearts can be used in vitro evaluations.
  • a subject can suffer from a heart disorder, such as heart failure, ischemia, myocardial infarction, congestive heart failure, arrhythmia, transplant rejection and the like.
  • the experimental animal can be an animal in which a gene related to cardiac structure or function is misexpressed. Misexpression can be achieved by methods known in the art, for example, by transgenesis, including the creation of knockout animals, or by classic breeding experiments or manipulation.
  • the misexpressed gene can be a gene encoding a sarcomeric protein, a gene encoding a protein which conditions cardiocyte survival or apoptosis, or a gene encoding a calcium regulatory protein.
  • Sarcomeric proteins include myosin heavy chain, troponin I, troponin C, troponin T, tropomyosin, actin, myosin light chain kinase, myosin light chain 1, myosin light chain 2 or myosin light chain 3.
  • Proteins which modify cardiocyte survival or apoptosis include IGF-1 receptor, PI3 kinase, AKT kinase or members of the caspase family of proteins.
  • Calcium regulatory proteins include phospholamban, SR Ca2 + ATPase, sodium-calcium exchanger, calcium channel (L and T), calsequestrin or calreticulin.
  • the experimental animal can be an animal model for a heart disorder, such as a hypertensive mouse or rat.
  • misexpression refers to a non-wild type pattern of gene expression. It includes: expression at non-wild type levels, i.e., over or under expression; a pattern of expression that differs from wild type in terms of the time or stage at which the gene is expressed, e.g., increased or decreased expression (as compared with wild type) at a predetermined developmental period or stage; a pattern of expression that differs from wild type in terms of decreased expression (as compared with wild type) in a predetermined cell type or tissue type; a pattern of expression that differs from wild type in terms of the splicing size, amino acid sequence, post- transitional modification, or biological activity of the expressed polypeptide; a pattern of expression that differs from wild type in terms of the effect of an environmental stimulus or extracellular stimulus on expression of the gene, e.g., a pattern of increased or decreased expression (as compared with wild type) in the presence of an increase or decrease in the strength of the stimulus. Misexpression includes any expression from a
  • the term "restricting the aortic flow of blood out of the heart” refers to substantially blocking the flow of blood into the distal aorta and its branches. For example, at least 50 % of the blood flowing out of the heart is restricted, preferably 75 % and more preferably 80, 90, or 100 % of the blood is restricted from flowing out of the heart.
  • the blood flow can be restricted by obstructing the aorta and the pulmonary artery, e.g., with clamps.
  • the term "introducing” refers to a process by which a compound can be placed into a chamber or the lumen of the heart of a subject.
  • the pericardium can be opened and the compound can be injected into the heart, e.g., using a syringe and a catheter.
  • the compound can be: introduced into the lumen of the aorta, e.g., the aortic root, introduced into the coronary ostia or introduced into the lumen of the heart.
  • the terms “homogeneous fashion” and “homogeneously overexpressing” are satisfied if one or more of the following requirements are met: (a) the compound contacts at least 10 %, preferably 20, 20, 40, 50, 60, 70, 80, 90 or 100 % of the cells of the heart and (b) at least 10 %, preferably 20, 20, 40, 50, 60, 70, 80, 90 or 100 % of the heart cells take up the compound.
  • purified preparation refers to a preparation in which at least 50, preferably 60, 70, 80 , 90 or 100 % of the cells are heart cells into which phospholamban has been introduced by somatic gene transfer.
  • the methods of the invention allow rapid and low cost development of cardiac overexpression models.
  • the methods of the invention also provide ways of examining multiple genes interacting in transgenic models, testing gene therapy approaches and evaluating treatments of cardiac disorders.
  • Ad.RSV.PL Ad.RSV.PL
  • Ad.RSV.SERCA2a Ad.RSV.SERCA2a
  • CPA cyclopiazonic acid
  • Figure 4A shows intracellular Ca2 + transients and shortening in an uninfected cardiomyocyte and in a cardiomyocyte infected for 48 hours with 10 pfu/cell of Ad.RSV. ⁇ gal and stimulated at 1 Hz.
  • Figure 4B shows intracellular Ca- ⁇ 4" transients and shortening in an uninfected cardiomyocyte and in a cardiomyocyte infected for 48 hours with 1, 10, and 100 pfu/cell of Ad.RSV.PL stimulated at 1 Hz.
  • Figure 4C shows intracellular Ca--- 4" transients and shortening in an uninfected cardiomyocyte, a cardiomyocyte infected with 1- pfu/cell of Ad.RSV.PL, and a cardiomyocyte infected with 10 pfu/cell of Ad.RSV.PL and 10 pfu/cell of Ad.RSV.SERCA2a for 48 hours, stimulated at 1 Hz.
  • Figure 7 A is a graph showing the response of intracellular Ca2 + transients to increasing frequency of stimulation in an uninfected cardiomyocyte.
  • Figure 7B is a graph showing the response of intracellular Ca ⁇ " transients to increasing frequency of stimulation in a cardiomyocyte infected with 10 pfu/cell of Ad.RSV.PL.
  • Figure 7C is a graph showing the response of intracellular Ca- ⁇ 4" transients to increasing frequency of stimulation in a cardiomyocyte infected with 10 pfu/cell of Ad.RSV.PL and 10 pfu/cell of Ad.RS V. SERC A2a for 48 hours.
  • Figure 8 shows intracavitary pressure tracings from rats 48 hours after cardiac gene transfer with either Ad.EGFP (left) or Ad.PL (right).
  • the pressure tracing of the Ad.PL transduced hearts displays a markedly prolonged relaxation and reduced pressure development.
  • Figure 9 is a drawing showing the somatic gene delivery method.
  • Figure 10A is a graph demonstrating that infection of neonatal cardiac myocytes with the construct Ad.asPL increased the contraction amplitude and significantly shortened the time course of the contraction.
  • Figure 1 OB is a graph demonstrating that adenovirus-mediated gene transfer of the antisense cDNA for phospholamban results in a modification of intracellular calcium handling. Phospholamban and Heart Disorder
  • somatic gene transfer e.g., adenoviral gene transfer
  • SERCA2a is both dose dependent and time dependent in rat neonatal cardiomyocytes.
  • the smaller size of phospholamban compared with SERCA2a (6 kD in its monomer form compared with 110 kD) may explain, at least in part, the more effective protein expression by Ad.RSV.PL than by Ad.RSV.SERCA2a under similar conditions. Nevertheless, using these recombinant adenoviruses, significant overexpression of phospholamban and SERCA2a was achieved, individually and in combination. Co- infection with both Ad.RSV.PL and Ad.RSV.SERCA2a mediated overexpression of both SERCA2a and phospholamban that was the same as the expression from infection with either Ad.RSV.PL or Ad.RSV.SERCA2a alone.
  • the ability to simultaneously manipulate expression of multiple proteins in the context of primary myocytes is an advantage of somatic gene transfer for the study of interacting components of complex systems.
  • phospholamban relative to SERCA2a is altered in a number of disease states. In hypothyroidism phospholamban levels are increased, whereas in hyperthyroidism phospholamban levels are decreased. An increased ratio of phospholamban to SERCA2a is an important characteristic of both human and experimental heart failure. Both experimental and human heart failure are characterized by a prolonged Ca + transient and impaired relaxation. Increasing levels of phospholamban relative to SERCA2a significantly altered intracellular Ca 2+ handling in the isolated cardiomyocytes by prolonging the relaxation phase of the Ca 2+ transient, decreasing Ca 2+ release, and increasing resting Ca 2+ .
  • a treatment can be evaluated by assessing the effect of the treatment on a parameter related to contractility. For example, SR Ca- ⁇ 4" ATPase activity or intracellular Ca- ⁇ 4" concentration can be measured, using the methods described above. Furthermore, force generation by hearts or heart tissue can be measured using methods described in Strauss et al., Am. J. Physiol., 262:1437-45, 1992, the contents of which are incorporated herein by reference. In many drug screening programs which test libraries of therapeutic agents and natural extracts, high throughput assays are desirable in order to maximize the number of therapeutic agents surveyed in a given period of time.
  • Assays which are performed in cell-free systems are preferred as "primary" screens in that they can be generated to permit rapid development and relatively easy detection of the parameter being measured, e.g., the intracellular levels of Ca--- 4" , which is mediated by a test therapeutic agent.
  • the effects of cellular toxicity and/or bioavailability of the test therapeutic agent can be generally ignored in the in vitro system, the assay instead being focused primarily on the effect of the therapeutic agent on the parameter being measured, e.g., the intracellular levels of Ca--- 4" . It is often desirable to screen candidate treatments in two stages, wherein the first stage is performed in vitro, and the second stage is performed in vivo.
  • the efficacy of a test therapeutic agent can be assessed by generating dose response curves from data obtained using various concentrations of the test therapeutic agent.
  • a control assay can also be performed to provide a baseline for comparison. In the control assay, the heart cell is incubated in the absence of a test agent. Propagation of Heart Cells
  • a heart cell culture can be obtained by allowing heart cells to migrate out of fragments of heart tissue adhering to a suitable substrate (e.g., a culture dish) or by disaggregating the tissue, e.g., mechanically or enzymatically to produce a suspension of heart cells.
  • a suitable substrate e.g., a culture dish
  • the enzymes trypsin, collagenase, elastase, hyaluronidase, DNase, pronase, dispase, or various combinations thereof can be used. Trypsin and pronase give the most complete disaggregation but may damage the cells. Collagenase and dispase give a less complete dissagregation but are less harmful.
  • tissue e.g., heart tissue
  • cells e.g., heart cells
  • Expression vectors suitable for somatic gene transfer, can be used to express the compound, e.g., the phospholamban gene.
  • examples of such vectors include replication defective retroviral vectors, adenoviral vectors and adeno-associated viral vectors.
  • Adenoviral vectors suitable for use by the methods of the invention include (Ad.RSV.lacZ), which includes the Rous sarcoma virus promoter and the lacZ reporter gene as well as (Ad.CMV.lacZ), which includes the cytomegalovirus promoter and the lacZ reporter gene.
  • Methods for the preparation and use of viral vectors are described in WO 96/13597, WO 96/33281, WO 97/15679, and Trapnell et al., Curr.
  • the nucleic acid which results in the overexpression of phospholamban can be derived from the natural phospholamban gene including all the introns and exons, it can be a cDNA molecule derived from the natural gene (Fujji et al., J. Biol. Chem. 266:11669-11675, 1991, the contents of which are incorporated herein by reference) or a chemically synthesized cDNA molecule.
  • the nucleic acid encoding the phospholamban protein can be under the control of the naturally occurring promoter or any other promoter that drives a high level expression of the phospholamban gene.
  • Ad.RSV.SERCA2a has been described in detail by Hajjar et al., Circulation, 95: 423-429, 1997, the contents of which are incorporated herein by reference.
  • Ad.RSV. ⁇ gal which carries a nuclear localizing form of ⁇ -galactosidase, is described in Dong et al., J. Biol. Chem. 27:29969-77, 1996, the contents of which are incorporated herein by reference.
  • the rabbit phospholamban cDNA is described in Lylton J. MacLennan D.H., J. Biol. Chem., 1988, 263:15024-15031, the contents of which are incorporated herein by reference.
  • the phospholamban cDNA was subcloned into the bacterial plasmid vector pAdRSV4, which uses the RS V long terminal repeat as a promoter and the SV40 polyadenylation signal and contains map units with adenovirus sequences from 0 to 1 and from 9 to 16.
  • the position and orientation of the phospholamban cDNA were confirmed by restriction enzyme digestion and by polymerase chain reaction.
  • the plasmid vector containing phospholamban (pAd.RSV-PL) was then cotransfected into 293 cells with PJM17.
  • the homologous recombinants between pAd.RSV.PL and pJM17 contain the phospholamban cDNA substituted for El .
  • Ad.RSV.PL This adenovirus is structurally similar to Ad.RSV. ⁇ gal and to Ad.RSV.SERCA2a- described in Dong O. et al., J. Biol. Chem, 1996, 271 :29969-29977, 1976.
  • the recombinant viruses were prepared as high-titer stocks by propagation in 293 cells as described in Graham, F.L.
  • Neonatal Cardiomyocytes Spontaneously beating cardiomyocytes were prepared from 1 to 2 day old rats and cultured in P-10 medium (GIBCO,BRL) in the presence of 5% fetal calf serum and 10% horse serum for 3 days as described previously in Kang J.X. et al., Proc. Natl. Acad. Sci. U.S.A., 1995, 92:3097-4001 and Kang J.X. and Leaf A., Euro. J. Pharmacol, 1996, 297:97-106, the contents of which are incorporated herein by reference.
  • cardiomyocytes infected with Ad.RSV.PL (multiplicity of infection of 10 pfu/cell) exhibited a significant increase in resting Ca + not evident in uninfected cells.
  • coinfection with Ad.RSV.SERCA2a (multiplicity of infection of 10 pfu/cell) restored the frequency response to normal.
  • the response to increasing stimulation frequencies in mammalian cardiomyocytes is governed by the SR.
  • the coverslip was superfused with a HEPES-buffered solution at a rate of 20 mL/h.
  • Cells were stimulated at different frequencies (0.1 to 2.0 Hz) using an external stimulator (Grass Instruments).
  • a dual excitation spectrofluorometer (IONOPTIX) was used to record fluorescence emissions (505 nm) elicited from exciting wavelengths of 360 and 380 nm.
  • Adenoviral gene transfer of phospholamban provides an attractive system for further elucidation of the effects of inhibiting SR Ca 2+ -ATPase on intracellular Ca 2+ handling.
  • a decrease in SRCa 2+ uptake rates is expected to lead to a smaller amount of Ca 2+ sequestered by the SR, resulting in a smaller amount of Ca 2+ release.
  • a significantly prolonged Ca 2+ transient and a higher resting (Ca 2+ ) was observed reflecting the decreased Ca 24 uptake and a decrease in peak (Ca + ) levels reflecting less Ca + available for release.
  • Phospholamban has been shown to play a key role in modulating the response of agents that increase cAMP levels in cardiomyocytes. Since phosphorylation of phospholamban reduces the inhibition to the SR Ca 2+ pump, thereby enhancing the SR Ca 2+ - ATPase, we were specifically interested in evaluating the effects of ⁇ -agonism on the relaxation phase of the Ca 2+ transient. In the basal state, the overexpression of phospholamban significantly prolongs the Ca 2+ transient. As shown in Figure 6, at maximal isoproterenol stimulation, the time course of the Ca 2+ transients in the uninfected cardiomyocytes and the cardiomyocytes infected with Ad. RSV.PL were decreased to the same level.
  • SR Membranes from Isolated Rat Cardiomyocytes To isolate SR membrane from cultured cardiomyocyes, a procedure modified from Harigaya et al., Circ. Res., 1969, 25:781-794, as well as, Wienzek et al., 1992, 23:1149-1163, the contents of which are incorporated herein by reference, was used. Briefly, isolated neonatal cardiomyocytes were suspended in a buffer containing (mmol/L) sucrose 500, phenylmerhyisulfonyl fluoride 1 and PIPES 20, at pH 7.4. The cardiomyocytes were then disrupted with a homogenizer. The homogenates were centrifuged at 500g for 20 minutes.
  • the resultant supernatant was centrifuged at 25,000g for 60 minutes to pellet the SR-enriched membrane.
  • the pellet was resuspended in a buffer containing (mmol/L) KC1 600, sucrose 30, and PIPES 20, frozen in liquid nitrogen, and stored at -70°C. Protein concentration was determined in these preparations by a modified Bradford procedure, described in Bradford et al., Anal.
  • the blots were incubated in a solution containing peroxidase-labeled goat anti-mouse IgG (dilution, 1 :1000) for 90 minutes at room temperature. The blot was then incubated in a chemiluminescence system and exposed to an X-OMAT x-ray film (Fuji Films) for 1 minute. The densities of the bands were evaluated using NIH Image. Normalization was performed by dividing densitometric units of each membrane preparation by the protein amounts in each of these preparations. Serial dilution of the membrane preparations revealed a linear relationship between amounts of protein and the densities of the SERCA2a immunoreactive hands (data not shown). 6. SR Ca 2+ -ATPase Activity
  • SR Ca2 + -ATPase activity assays were carried out according to Chu A. et al., Methods Enzymol, 1988, 157:36-46, the contents of which are incorporated herein by reference, on the basis of pyruvate NADH-coupled reactions.
  • a photomotor Beckman DU 640 adjusted at a wavelength of 540 nm, oxidation of NADH (which is coupled to the SR Ca--- 4" - ATPase) was assessed at 37°C in the membrane preparations by the difference of the total absorbance and basal absorbance.
  • the reaction was carried out in a volume of 1 mL. All experiments were carried out in triplicate.
  • the activity of the Ca-"- + -ATPase was calculated as follows: ⁇ absorbance/6.22 x protein x time (in nmol ATP/mg protein x min). The measurements were repeated at different [Ca--- 4" ] levels.
  • the effect of the specific Ca-"- + -ATPase inhibitor CPA at a concentration range of 0.001 to 10 ⁇ mol/L was also studied in those preparations, as described in Schwinger et al., Circulation, 1995, 92:3220-3228 and Baudet et al., Circ. Res., 1993, 73:813-819, the contents of which are incorporated herein by reference.
  • CPA inhibited the SR Ca-"- + -ATPase activity in a dose-dependent fashion in all three membrane preparations (uninfected, Ad.RSV.PL, and d.RSV.PL+Ad.RSV.SERCA2a).
  • the SR Ca 2+ - ATPase plays a key role in excitation-contraction coupling, lowering Ca + during relaxation in cardiomyocytes, and "loading" the SR with Ca 2+ for the subsequent release and contractile activation.
  • the Ca + -pumping activity of this enzyme is influenced by phospholamban. In the unphosphorylated state, phospholamban inhibits the Ca 2+ - ATPase, whereas phosphorylation of phospholamban by cAMP-dependent protein kinase and by Ca 2+ calmodulin-dependent protein kinase reverses this inhibition. Therefore, an increase in phospholamban content should decrease the affinity of the SR Ca 2+ pump for Ca 2+ .
  • Rats and mice were anesthetized with intraperitoneal pentobarbital and placed on a ventilator.
  • the chest was entered form the left side through the third intercostal space.
  • the pericardium was opened and a 7-0 suture placed at the apex of the left ventricle.
  • the aorta and pulmonary artery were identified.
  • a 22 G catheter containing 200 ⁇ l of adenovirus was advanced from the apex of the left ventricle to the aortic root.
  • the aorta and pulmonary artery were clamped distal to the site of the catheter and the adenovirus solution was injected as shown in Figure 9.
  • the clamp was maintained for 10 seconds while the heart was pumping against a closed system (isovolumically).
  • Delayed cardiac relaxation in failing hearts is attributed to a reduced activity of the Sarcoplasmic Reticulum Calcium ATPase.
  • Phospholamban inhibits SERCA2a activity and is, therefore, a potential target to improve cardiac function.
  • Ad.asPL adenovirus carrying the full length antisense cDNA of phospholamban
  • adenovirus-mediated gene transfer of the antisense cDNA for phospholamban also resulted in a modification of intracellular calcium handling and shortening in myocardial cells (see Figure 10B) indicating that such vectors can be used for increasing the contractility of myocardial cells in heart failure.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Cardiology (AREA)
  • Biotechnology (AREA)
  • Medicinal Chemistry (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Toxicology (AREA)
  • Physics & Mathematics (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Hospice & Palliative Care (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The invention provides methods of evaluating a tratement for a heart disorder. The invention also provides methods of delivering a compound to the heart of a subject. A heart cell, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban, a heart tissue, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban and a heart, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban are also provided by the invention. Finally, the invention provides a method for treating a subject at risk for a heart disorder.

Description

EVALUATION OF, DELIVERY OF, AND USE OF AGENTS TO TREAT
HEART DISORDERS
Field of the Invention This invention relates to methods for evaluating treatments suitable for heart disorders and to methods for delivering compounds to the heart of a subject.
Background of the Invention The sarcoplasmic reticulum (SR) is an internal membrane system, which plays a critical role in the regulation of cytosolic Ca---"1" concentrations and thus, excitation- contraction coupling in muscle. Contraction is mediated through the release of Ca*--4" from the SR, while relaxation involves the active re-uptake of Ca2+ into the SR lumen by a Ca2+ -ATPase. In cardiac muscle, the SR Ca---"1" -ATPase activity (SERCA2a) is under reversible regulation by phospholamban.
Phospholamban is a small phosphoprotein, about 6,080 daltons in size, which is an integral element of the cardiac SR membrane. Phospholamban is phosphorylated in vivo in response to β-adrenergic agonist stimulation. In the dephosphorylated state, phospholamban inhibits SR Ca2+ -ATPase activity by decreasing the affinity of the enzyme for Ca2+.
Heart failure is characterized by a number of abnormalities at the cellular level in the various steps of excitation-contraction coupling of the cardiac cells. One of the key abnormalities in both human and experimental heart failure is a defect in SR function which is associated with abnormal intracellular Ca^"1" handling. Deficient SR Ca^+ uptake during relaxation has been identified in failing hearts from both humans and animal models and has been associated with a decrease in the activity of SR Ca-*-"1" - ATPase activity and altered Ca^+ kinetics.
Summary of the Invention In general, the invention features, a method of evaluating a treatment for a heart disorder. The method includes: providing a heart cell, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; administering the treatment to the heart cell; and evaluating the effect of the treatment on the heart cell, thereby evaluating the treatment for a heart disorder.
In preferred embodiments, the method includes evaluating the effect of the treatment on a parameter related to heart function. The parameter, by way of example, can include an assessment of contractility, Ca2+ metabolism, e.g., intracellular Cs?+ concentration, SR Ca2+ ATPase activity, force generation, a force frequency relationship, cardiocyte survival or apoptosis or ion channel activity, e.g., sodium calcium exchange, sodium channel activity, calcium channel activity, or sodium potassium ATPase pump activity.
In preferred embodiments, the treatment is administered in vivo, e.g., to an experimental animal. The experimental animal can be an animal in which a gene related to cardiac structure or function is misexpressed. Misexpression can be achieved by methods known in the art, for example, by transgenesis, including the creation of knockout animals, or by classic breeding experiments or manipulation. The misexpressed gene can be a gene encoding a sarcomeric protein, a gene encoding a protein which conditions cardiocyte survival or apoptosis, or a gene encoding a calcium regulatory protein. Sarcomeric proteins include myosin heavy chain, troponin I, troponin C, troponin T, tropomyosin, actin, myosin light chain kinase, myosin light chain 1, myosin light chain 2 or myosin light chain 3. Proteins which modify cardiocyte survival or apoptosis include IGF-1 receptor, PI3 kinase, AKT kinase or members of the caspase family of proteins. Calcium regulatory proteins include phospholamban, SR Ca2+ ATPase, sodium-calcium exchanger, calcium channel (L and T), calsequestrin or calreticulin. The experimental animal can be an animal model for a disorder, e.g., a heart disorder.
In preferred embodiments, the treatment is administered in vitro. In preferred embodiments the cell is derived from an experimental animal or a human. In preferred embodiments the cell can be cultured and/or immortalized.
In preferred embodiments, the nucleic acid encodes a phospholamban protein. The phospholamban can be from the same species that the heart cell is from or it can be from a different species. For example, a mouse phospholamban can be expressed in a mouse cell or a human phospholamban can be expressed in a cell from an experimental animal.
In preferred embodiments, the nucleic acid is introduced into the heart cell by way of a vector suitable for somatic gene transfer, e.g., a viral vector, e.g., an adenoviral vector. In another aspect, the invention features, a method of evaluating a treatment for a heart disorder. The method includes: providing a heart, into some or all the cells of which has been introduced, by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; administering the treatment to the heart; and evaluating the effect of the treatment on the heart, thereby evaluating the treatment for a heart disorder. In preferred embodiments, the method includes evaluating the effect of the treatment on a parameter related to heart function. The parameter, by way of example, can include an assessment of contractility, Ca2+ metabolism, e.g., intracellular C ^+ concentration, SR Ca-"-"1" ATPase activity, force generation, a force frequency relationship, cardiocyte survival or apoptosis or ion channel activity, e.g., sodium calcium exchange, sodium channel activity, calcium channel activity, or sodium potassium ATPase pump activity.
In preferred embodiments, the treatment is administered in vivo, e.g., to an experimental animal. The experimental animal can be an animal in which a gene related to cardiac structure or function is misexpressed. Misexpression can be achieved by methods known in the art, for example, by transgenesis, including the creation of knockout animals, or by classic breeding experiments or manipulation. The misexpressed gene can be a gene encoding a sarcomeric protein, a gene encoding a protein which conditions cardiocyte survival or apoptosis, or a gene encoding a calcium regulatory protein. Sarcomeric proteins include myosin heavy chain, troponin I, troponin C, troponin T, tropomyosin, actin, myosin light chain kinase, myosin light chain 1, myosin light chain 2 or myosin light chain 3. Proteins which modify cardiocyte survival or apoptosis include IGF-1 receptor, PI3 kinase, AKT kinase or members of the caspase family of proteins. Calcium regulatory proteins include phospholamban, SR Ca2+ ATPase, sodium-calcium exchanger, calcium channel (L and T), calsequestrin or calreticulin. The experimental animal can be an animal model for a disorder, e.g., a heart disorder.
In preferred embodiments, the treatment is administered in vitro. In preferred embodiments the heart is derived from an experimental animal or a human. In preferred embodiments, the nucleic acid encodes a phospholamban protein.
The phospholamban can be from the same species that the heart is from or it can be from a different species. For example, a mouse phospholamban can be expressed in a mouse heart or a human phospholamban can be expressed in the heart of an experimental animal. The phospholamban can be delivered to the heart using methods described herein.
In preferred embodiments, the nucleic acid is introduced into the heart by way of a vector suitable for somatic gene transfer, e.g., a viral vector, e.g., an adenoviral vector. In another aspect, the invention features, a method of evaluating a treatment for a heart disorder. The method includes: providing heart tissue into some or all of the cells of which has been introduced, by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; administering the treatment to the heart tissue; and evaluating the effect of the treatment on the heart tissue, thereby evaluating the treatment for a heart disorder.
In preferred embodiments, the method includes evaluating the effect of the treatment on a parameter related to heart function. The parameter, by way of example, can include an assessment of contractility, Ca---4" metabolism, e.g., intracellular Ca^"1" concentration, SR Ca-^"1" ATPase activity, force generation, a force frequency relationship, cardiocyte survival or apoptosis or ion channel activity, e.g., sodium calcium exchange, sodium channel activity, calcium channel activity, or sodium potassium ATPase pump activity. In preferred embodiments, the treatment is administered in vivo, e.g., to an experimental animal. The experimental animal can be an animal in which a gene related to cardiac structure or function is misexpressed. Misexpression can be achieved by methods known in the art, for example, by transgenesis, including the creation of knockout animals, or by classic breeding experiments or manipulation. The misexpressed gene can be a gene encoding a sarcomeric protein, a gene encoding a protein which conditions cardiocyte survival or apoptosis, or a gene encoding a calcium regulatory protein. Sarcomeric proteins include myosin heavy chain, troponin I, troponin C, troponin T, tropomyosin, actin, myosin light chain kinase, myosin light chain 1, myosin light chain 2 or myosin light chain 3. Proteins which modify cardiocyte survival or apoptosis include IGF-1 receptor, PI3 kinase, AKT kinase or members of the caspase family of proteins. Calcium regulatory proteins include phospholamban, SR Ca2+ ATPase, sodium-calcium exchanger, calcium channel (L and T), calsequestrin or calreticulin. The experimental animal can be an animal model for a disorder, e.g., a heart disorder. In preferred embodiments, the treatment is administered in vitro. In preferred embodiments the heart tissue is derived from an experimental animal or a human.
In preferred embodiments, the nucleic acid encodes a phospholamban protein. The phospholamban can be from the same species that the heart tissue is from or it can be from a different species. For example, a mouse phospholamban can be expressed in a mouse heart tissue or a human phospholamban can be expressed in heart tissue from an experimental animal.
In preferred embodiments, the nucleic acid is introduced into the heart tissue by way of a vector suitable for somatic gene transfer, e.g., a viral vector, e.g., an adenoviral vector. In another aspect, the invention features, a method of evaluating a treatment for a heart disorder. The method includes: providing a first and a second heart cell, into each of which has been introduced, by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; administering the treatment to a first heart cell, preferably in vitro; evaluating the effect of the treatment on the first heart cell; administering the treatment to a second heart cell, preferably in vivo; and evaluating the effect of the treatment on the second heart cell, thereby evaluating the treatment for a heart disorder.
In preferred embodiments, the method includes evaluating the effect of the treatment on a parameter related to heart function. The parameter, by way of example, can include an assessment of contractility, Ca^"1" metabolism, e.g., intracellular Ca^4" concentration, SR Ca2+ ATPase activity, force generation, a force frequency relationship, cardiocyte survival or apoptosis or ion channel activity, e.g., sodium calcium exchange, sodium channel activity, calcium channel activity, or sodium potassium ATPase pump activity.
In preferred embodiments, the treatment is administered in vivo, e.g., to an experimental animal. The experimental animal can be an animal in which a gene related to cardiac structure or function is misexpressed. Misexpression can be achieved by methods known in the art, for example, by transgenesis, including the creation of knockout animals, or by classic breeding experiments or manipulation. The misexpressed gene can be a gene encoding a sarcomeric protein, a gene encoding a protein which conditions cardiocyte survival or apoptosis, or a gene encoding a calcium regulatory protein. Sarcomeric proteins include myosin heavy chain, troponin I, troponin C, troponin T, tropomyosin, actin, myosin light chain kinase, myosin light chain 1, myosin light chain 2 or myosin light chain 3. Proteins which modify cardiocyte survival or apoptosis include IGF-1 receptor, PI3 kinase, AKT kinase or members of the caspase family of proteins. Calcium regulatory proteins include phospholamban, SR Ca2+ ATPase, sodium-calcium exchanger, calcium channel (L and T), calsequestrin or calreticulin. The experimental animal can be an animal model for a disorder, e.g., a heart disorder.
In preferred embodiments, the nucleic acid encodes a phospholamban protein. The phospholamban can be from the same species that the heart cell is from or it can be from a different species. For example, a mouse phospholamban can be expressed in a mouse cell or a human phospholamban can be expressed in a cell from an experimental animal.
In preferred embodiments, the first and second cell can be from the same or different animals, can be from the same or different species, e.g., the first cell can be from a mouse and the second cell can be from a human or both cells can be human. The first and second cell can have the same or different genotypes. In further preferred embodiments the evaluation of the treatment in the first cell can be the same or different from the evaluation of the treatment in the second cell, e.g., the intracellular Ca-^4" concentration can be measured in the first cell and the SR Ca^ " -ATPase activity can be measured in the second cell or the intracellular Ca-^4" concentration can be measured in both cells.
In another aspect, the invention features, a method of evaluating a treatment for a heart disorder. The method includes: providing a first administration of a treatment to a heart cell, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; evaluating the effect of the first administration on the heart cell; providing a second administration of a treatment to a heart cell, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; and evaluating the effect of the second administration on the heart cell, thereby evaluating a treatment for a heart disorder. In preferred embodiments, the method includes evaluating the effect of the treatment on a parameter related to heart function. The parameter, by way of example, can include an assessment of contractility, Ca-^4" metabolism, e.g., intracellular Ca^4" concentration, SR Ca---4" ATPase activity, force generation, a force frequency relationship, cardiocyte survival or apoptosis or ion channel activity, e.g., sodium calcium exchange, sodium channel activity, calcium channel activity, or sodium potassium ATPase pump activity.
In preferred embodiments, the treatment is administered in vivo, e.g., to an experimental animal. The experimental animal can be an animal in which a gene related to cardiac structure or function is misexpressed. Misexpression can be achieved by methods known in the art, for example, by transgenesis, including the creation of knockout animals, or by classic breeding experiments or manipulation. The misexpressed gene can be a gene encoding a sarcomeric protein, a gene encoding a protein which conditions cardiocyte survival or apoptosis, or a gene encoding a calcium regulatory protein. Sarcomeric proteins include myosin heavy chain, troponin I, troponin C, troponin T, tropomyosin, actin, myosin light chain kinase, myosin light chain 1, myosin light chain 2 or myosin light chain 3. Proteins which modify cardiocyte survival or apoptosis include IGF-1 receptor, PI3 kinase, AKT kinase or members of the caspase family of proteins. Calcium regulatory proteins include phospholamban, SR Ca2+ ATPase, sodium-calcium exchanger, calcium channel (L and T), calsequestrin or calreticulin. The experimental animal can be an animal model for a disorder, e.g., a heart disorder. In preferred embodiments, the nucleic acid encodes a phospholamban protein. The phospholamban can be from the same species that the heart cell is from or it can be from a different species. For example, a mouse phospholamban can be expressed in a mouse cell or a human phospholamban can be expressed in a cell from an experimental animal.
In preferred embodiments the first and second administration can be administered to the same or to different cells. The first and second administration can be administered under the same or different conditions, e.g., the first administration can consist of a relatively low level treatment, e.g., a lower concentration of a substance, and the second administration can consist of a relatively high level treatment, e.g., a higher concentration of a substance or both administrations can consist of the same level treatment.
In another aspect, the invention features, a method of evaluating a treatment for a heart disorder. The method includes: providing a heart cell, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; administering the treatment to the heart cell; evaluating the effect of the treatment on the heart cell; providing a heart, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; administering the treatment to the heart ; and evaluating the effect of the treatment on the heart, thereby evaluating the treatment for a heart disorder.
In preferred embodiments, the method includes evaluating the effect of the treatment on a parameter related to heart function. The parameter, by way of example, can include an assessment of contractility, Ca^ " metabolism, e.g., intracellular Ca-^4" concentration, SR Ca-"-4" ATPase activity, force generation, a force frequency relationship, cardiocyte survival or apoptosis or ion channel activity, e.g., sodium calcium exchange, sodium channel activity, calcium channel activity, or sodium potassium ATPase pump activity.
In preferred embodiments, the treatment is administered in vivo, e.g., to an experimental animal. The experimental animal can be an animal in which a gene related to cardiac structure or function is misexpressed. Misexpression can be achieved by methods known in the art, for example, by transgenesis, including the creation of knockout animals, or by classic breeding experiments or manipulation. The misexpressed gene can be a gene encoding a sarcomeric protein, a gene encoding a protein which conditions cardiocyte survival or apoptosis, or a gene encoding a calcium regulatory protein. Sarcomeric proteins include myosin heavy chain, troponin I, troponin C, troponin T, tropomyosin, actin, myosin light chain kinase, myosin light chain 1, myosin light chain 2 or myosin light chain 3. Proteins which modify cardiocyte survival or apoptosis include IGF-1 receptor, PI3 kinase, AKT kinase or members of the caspase family of proteins. Calcium regulatory proteins include phospholamban, SR Ca2+ ATPase, sodium-calcium exchanger, calcium channel (L and T), calsequestrin or calreticulin. The experimental animal can be an animal model for a disorder, e.g., a heart disorder.
In preferred embodiments, the nucleic acid encodes a phospholamban protein. The phospholamban can be from the same species that the heart cell and/or the heart is from or it can be from a different species. For example, a mouse phospholamban can be expressed in a mouse heart cell and/or heart or a human phospholamban can be expressed in a heart cell and/or heart from an experimental animal.
In preferred embodiments the treatment can be administered to the heart cell in vitro and to the heart in vivo or the treatment can be administered to the heart cell and to the heart in vitro. In another aspect, the invention features, a method of delivering a compound to the heart of a subject. The method includes: restricting the aortic flow of blood out of the heart, such that blood flow is re-directed to the coronary arteries; introducing the compound into the lumen of the circulatory system such that it flows into the coronary arteries; allowing the heart to pump while the aortic outflow of blood is restricted, thereby allowing the compound to flow into and be delivered to the heart; and reestablishing the flow of blood to the heart .
In preferred embodiments, the compound includes: a nucleic acid which directs the expression of a peptide, e.g., a phospholamban or a SR Ca^4" -ATPase and a viral vector suitable for somatic gene delivery, e.g., an adenoviral vector. In preferred embodiments, the subject is at risk for a heart disorder, e.g., heart failure, ischemia, arrhythmia, myocardial infarction, congestive heart failure, transplant rejection.
In preferred embodiments, the subject can be a human or an experimental animal. The experimental animal can be an animal in which a gene related to cardiac structure or function is misexpressed. Misexpression can be achieved by methods known in the art, for example, by transgenesis, including the creation of knockout animals, or by classic breeding experiments or manipulation. The misexpressed gene can be a gene encoding a sarcomeric protein, a gene encoding a protein which conditions cardiocyte survival or apoptosis, or a gene encoding a calcium regulatory protein. Sarcomeric proteins include myosin heavy chain, troponin I, troponin C, troponin T, tropomyosin, actin, myosin light chain kinase, myosin light chain 1, myosin light chain 2 or myosin light chain 3. Proteins which modify cardiocyte survival or apoptosis include IGF-1 receptor, PI3 kinase, AKT kinase or members of the caspase family of proteins. Calcium regulatory proteins include phospholamban, SR Ca2+ ATPase, sodium-calcium exchanger, calcium channel (L and T), calsequestrin or calreticulin. The experimental animal can be an animal model for a disorder, e.g., a heart disorder.
In preferred embodiments, the method further includes restricting blood flow into the left side of the heart, e.g., by restricting the pulmonary circulation through obstraction of the pulmonary artery, so as to lessen dilution of the compound.
In preferred embodiments the method further includes opening the pericardium and introducing the compound, e.g., using a catheter.
In preferred embodiments, the compound is: introduced into the lumen of the aorta, e.g., the aortic root, introduced into the coronary ostia or introduced into the lumen of the heart.
In preferred embodiments, the nucleic acid, which directs the expression of the peptide, is homogeneously overexpressed in the heart of the subject.
In another aspect, the invention features, a heart cell, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban. The heart cell can be provided as a purified preparation.
In another aspect, the invention features, a heart tissue, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban. The heart tissue can be provided as a tissue preparation.
In another aspect, the invention features, a heart, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban. The heart can be provided in a subject or ex vivo, i.e. removed from a subject.
In another aspect, the invention features, a method for treating a subject at risk for a heart disorder. The method includes introducing into somatic heart tissue of the subject, a nucleic acid which encodes phospholamban.
In preferred embodiments, the nucleic acid is introduced using the methods described herein.
In preferred embodiments, the phospholamban can be from the same species as the subject or it can be from a different species. For example, a human phospholamban can be introduced into a human heart or a human phospholamban can be can be introduced into the heart of an experimental animal. In preferred embodiments, the nucleic acid is introduced into the heart by way of a vector suitable for somatic gene transfer, e.g., a viral vector, e.g., an adenoviral vector. In preferred embodiments, the subject can be a human, an experimental animal, e.g., a rat or a mouse, a domestic animal, e.g., a dog, cow, sheep, pig or horse, or a non- human primate, e.g., a monkey. The subject can be suffering from a cardiac disorder, such as heart failure, ischemia, myocardial infarction, congestive heart failure, arrhythmia, transplant rejection and the like.
As used herein, the term "treatment" refers to a procedure (e.g., a surgical method) or the administration of a substance, e.g., a compound which is being evaluated for use in the alleviation or prevention of a heart disorder or symptoms thereof. For example, such treatment can be a surgical procedure, or the administration of a therapeutic agent such as a drug, a peptide, an antibody, an ionophore and the like. As used herein, the term "heart disorder" refers to a structural or functional abnormality of the heart, that impairs its normal functioning. For example, the heart disorder can be heart failure, ischemia, myocardial infarction, congestive heart failure, arrhythmia, transplant rejection and the like. The term includes disorders characterized by abnormalities of contraction, abnormalities in Ca^4" metabolism, and disorders characterized by arrhytmia.
As used herein, the term "heart cell" refers to a cell which can be: (a) part of a heart present in a subject, (b) part of a heart which is maintained in vitro, (c) part of a heart tissue, or (d) a cell which is isolated from the heart of a subject. For example, the cell can be a cardiac myocyte.
As used herein, the term "heart" refers to a heart present in a subject or to a heart which is maintained outside a subject.
As used herein, the term "heart tissue" refers to tissue which is derived from the heart of a subject. As used herein, the term "somatic gene transfer" refers to the transfer of genes into a somatic cell as opposed to transferring genes into the germ line.
As used herein , the term "compound" refers to a compound, which can be delivered effectively to the heart of a subject using the methods of the invention. Such compounds can include, for example, a gene, a drug, an antibiotic, an enzyme, a chemical compound, a mixture of chemical compounds or a biological macromolecule. As used herein, the term "subject" refers to an experimental animal, e.g., a rat or a mouse, a domestic animal, e.g., a dog, cow, sheep, pig or horse, a non-human primate, e.g., a monkey and in the case of therapeutic methods, humans. However, it is noted that human cells, tissue or hearts can be used in vitro evaluations. A subject can suffer from a heart disorder, such as heart failure, ischemia, myocardial infarction, congestive heart failure, arrhythmia, transplant rejection and the like. The experimental animal can be an animal in which a gene related to cardiac structure or function is misexpressed. Misexpression can be achieved by methods known in the art, for example, by transgenesis, including the creation of knockout animals, or by classic breeding experiments or manipulation. The misexpressed gene can be a gene encoding a sarcomeric protein, a gene encoding a protein which conditions cardiocyte survival or apoptosis, or a gene encoding a calcium regulatory protein. Sarcomeric proteins include myosin heavy chain, troponin I, troponin C, troponin T, tropomyosin, actin, myosin light chain kinase, myosin light chain 1, myosin light chain 2 or myosin light chain 3. Proteins which modify cardiocyte survival or apoptosis include IGF-1 receptor, PI3 kinase, AKT kinase or members of the caspase family of proteins. Calcium regulatory proteins include phospholamban, SR Ca2+ ATPase, sodium-calcium exchanger, calcium channel (L and T), calsequestrin or calreticulin. The experimental animal can be an animal model for a heart disorder, such as a hypertensive mouse or rat.
As used herein, the term "misexpression" refers to a non-wild type pattern of gene expression. It includes: expression at non-wild type levels, i.e., over or under expression; a pattern of expression that differs from wild type in terms of the time or stage at which the gene is expressed, e.g., increased or decreased expression (as compared with wild type) at a predetermined developmental period or stage; a pattern of expression that differs from wild type in terms of decreased expression (as compared with wild type) in a predetermined cell type or tissue type; a pattern of expression that differs from wild type in terms of the splicing size, amino acid sequence, post- transitional modification, or biological activity of the expressed polypeptide; a pattern of expression that differs from wild type in terms of the effect of an environmental stimulus or extracellular stimulus on expression of the gene, e.g., a pattern of increased or decreased expression (as compared with wild type) in the presence of an increase or decrease in the strength of the stimulus. Misexpression includes any expression from a transgenic nucleic acid.
As used herein, the term "restricting the aortic flow of blood out of the heart" refers to substantially blocking the flow of blood into the distal aorta and its branches. For example, at least 50 % of the blood flowing out of the heart is restricted, preferably 75 % and more preferably 80, 90, or 100 % of the blood is restricted from flowing out of the heart. The blood flow can be restricted by obstructing the aorta and the pulmonary artery, e.g., with clamps.
As used, herein, the term "introducing" refers to a process by which a compound can be placed into a chamber or the lumen of the heart of a subject. For example, the pericardium can be opened and the compound can be injected into the heart, e.g., using a syringe and a catheter. The compound can be: introduced into the lumen of the aorta, e.g., the aortic root, introduced into the coronary ostia or introduced into the lumen of the heart.
As used herein, the terms "homogeneous fashion" and "homogeneously overexpressing" are satisfied if one or more of the following requirements are met: (a) the compound contacts at least 10 %, preferably 20, 20, 40, 50, 60, 70, 80, 90 or 100 % of the cells of the heart and (b) at least 10 %, preferably 20, 20, 40, 50, 60, 70, 80, 90 or 100 % of the heart cells take up the compound.
As used herein, the term "purified preparation" refers to a preparation in which at least 50, preferably 60, 70, 80 , 90 or 100 % of the cells are heart cells into which phospholamban has been introduced by somatic gene transfer.
The methods of the invention allow rapid and low cost development of cardiac overexpression models. The methods of the invention also provide ways of examining multiple genes interacting in transgenic models, testing gene therapy approaches and evaluating treatments of cardiac disorders.
Other features and advantages of the invention will be apparent from the following detailed description, and from the claims.
Brief Description of the Drawings The drawings are first briefly described. Figure 1 is a graph depicting protein levels of SR Ca^4"- ATPase in uninfected cardiomyocytes (n=8) and in cardiomyocytes infected for 48 hours with 1, 10, and 100 pfu/cell of Ad.RSV.PL.
Figure 2 is a graph depicting protein levels of phospholamban and SERCA2a in uninfected cardiomyocytes (n=8) and in cardiomyocytes infected for 48 hours with 10 pfu/cell with either Ad.RSV.PL and/or Ad.RSV.SERCA2a. There were no significant differences between the phospholamban protein levels in the group of myocytes infected with Ad.RSV.PL alone at a multiplicity of infection of 10 pfu/cell and the group of myocytes infected with Ad.RSV.PL at a multiplicity of infection of 10 pfu/cell and Ad.RSV.SERCA2a at a multiplicity of infection of 10 pfu/cell (P>2). Similarly, there were no significant differences between the SERCA2a protein levels in the group of myocytes infected with Ad.RSV.SERCA2a alone at a multiplicity of infection of 10 pfu/cell and the group of myocytes infected with Ad.RSV.PL at a multiplicity of infection of 10 pfu/cell and Ad.RSV.SERCA2a at a multiplicity of infection of 10 pfu/cell (P>2). Figure 3 A is a graph depicting SERCA2a activity as a function of Ca---4" in membrane preparations of uninfected cardiomyocytes (■, n=6), cardiomyocytes infected with 10 pfu/cell of Ad.RSV.PL (•, n=6), and cardiomyocytes infected with 10 pfu/cell of Ad.RSV.PL and 10 pfu/cell of Ad.RSV.SERCA2a (}, n=6). Figure 3B is a graph depicting the effect of increasing concentrations of cyclopiazonic acid (CPA) on SRECA2 activity at a [Ca-^4-] of 10 μmol/L in membrane preparations of uninfected cardiomyocytes (■, n=6), cardiomyocytes infected with 10 pfu/cell of Ad.RSV.PL (•, n=6), and cardiomyocytes infected with 10 pfu/cell of Ad.RSV.PL and 10 pfu/cell of Ad.RSV.SERCA2a (}, n=6).
Figure 4A shows intracellular Ca2+ transients and shortening in an uninfected cardiomyocyte and in a cardiomyocyte infected for 48 hours with 10 pfu/cell of Ad.RSV.βgal and stimulated at 1 Hz. Figure 4B shows intracellular Ca-^4" transients and shortening in an uninfected cardiomyocyte and in a cardiomyocyte infected for 48 hours with 1, 10, and 100 pfu/cell of Ad.RSV.PL stimulated at 1 Hz. Figure 4C shows intracellular Ca---4" transients and shortening in an uninfected cardiomyocyte, a cardiomyocyte infected with 1- pfu/cell of Ad.RSV.PL, and a cardiomyocyte infected with 10 pfu/cell of Ad.RSV.PL and 10 pfu/cell of Ad.RSV.SERCA2a for 48 hours, stimulated at 1 Hz.
Figure 5 A is a graph showing the mean of the peak of the intracellular Ca---4" transients in uninfected cardiomyocytes (n=10), cardiomyocytes infected with 10 pfu/cell of Ad.RSV.PL (n=12), and cardiomyocytes infected with 10 pfu/cell of Ad.RSV.PL and 10 pfu/cell of Ad.RSV.SERCA2a (n=10) for 48 hours and stimulated at 1 Hz. Figure 5B is a graph showing the mean of the resting levels of [Ca---4"] in uninfected cardiomyocytes (n=10), cardiomyocytes infected with 10 pfu/cell of ad.RSV.PL (n=12), and cardiomyocytes infected with 10 pfu/cell of Ad.RSV.PL and 10 pfu/cell of Ad.RSV.SERCA2a (n=10) for 48 hours, stimulated at 1 Hz. Figure 5C is a graph showing the mean of the time to 80% relaxation of the intracellular Ca-^4" transients in uninfected cardiomyocytes (n=10), cardiomyocytes infected with 10 pfu/cell of Ad.RSV.PL (n=12), and cardiomyocytes infected with 10 pfu/cell of Ad.RSV.PL and 10 pfu/cell of Ad.RSV.SERCA2a (n=10) for 48 hours, stimulated at 1 Hz. P<.05 compared with uninfected cells. P<.05 compared with Ad.RSV.PL (multiplicity of infection of 10 pfu/cell) .
Figure 6 is a graph showing the effect of increasing concentrations of Isoprotenerol on the time course of the intracellular Ca---" transients in uninfected cardiomyocytes (n=5) and cardiomyocytes infected with 10 pfu/cell of Ad.RSV.PL (n=5), stimulated at 1 Hz. Figure 7 A is a graph showing the response of intracellular Ca2+ transients to increasing frequency of stimulation in an uninfected cardiomyocyte. Figure 7B is a graph showing the response of intracellular Ca^ " transients to increasing frequency of stimulation in a cardiomyocyte infected with 10 pfu/cell of Ad.RSV.PL. Figure 7C is a graph showing the response of intracellular Ca-^4" transients to increasing frequency of stimulation in a cardiomyocyte infected with 10 pfu/cell of Ad.RSV.PL and 10 pfu/cell of Ad.RS V. SERC A2a for 48 hours.
Figure 8 shows intracavitary pressure tracings from rats 48 hours after cardiac gene transfer with either Ad.EGFP (left) or Ad.PL (right). The pressure tracing of the Ad.PL transduced hearts displays a markedly prolonged relaxation and reduced pressure development. Figure 9 is a drawing showing the somatic gene delivery method.
Figure 10A is a graph demonstrating that infection of neonatal cardiac myocytes with the construct Ad.asPL increased the contraction amplitude and significantly shortened the time course of the contraction. Figure 1 OB is a graph demonstrating that adenovirus-mediated gene transfer of the antisense cDNA for phospholamban results in a modification of intracellular calcium handling. Phospholamban and Heart Disorder
The inventors have discovered that somatic gene transfer, e.g., adenoviral gene transfer, is particularly effective in mammalian myocardium both in vivo and in vitro and particularly useful for screening treatments suitable for heart disorders. Adenoviral gene transfer of SERCA2a is both dose dependent and time dependent in rat neonatal cardiomyocytes. An adenovirus encoding phospholamban under the RSV promoter, provided a 4-fold increase in phospholamban, which was also dose dependent. The smaller size of phospholamban compared with SERCA2a (6 kD in its monomer form compared with 110 kD) may explain, at least in part, the more effective protein expression by Ad.RSV.PL than by Ad.RSV.SERCA2a under similar conditions. Nevertheless, using these recombinant adenoviruses, significant overexpression of phospholamban and SERCA2a was achieved, individually and in combination. Co- infection with both Ad.RSV.PL and Ad.RSV.SERCA2a mediated overexpression of both SERCA2a and phospholamban that was the same as the expression from infection with either Ad.RSV.PL or Ad.RSV.SERCA2a alone. The ability to simultaneously manipulate expression of multiple proteins in the context of primary myocytes is an advantage of somatic gene transfer for the study of interacting components of complex systems.
The expression of phospholamban relative to SERCA2a is altered in a number of disease states. In hypothyroidism phospholamban levels are increased, whereas in hyperthyroidism phospholamban levels are decreased. An increased ratio of phospholamban to SERCA2a is an important characteristic of both human and experimental heart failure. Both experimental and human heart failure are characterized by a prolonged Ca + transient and impaired relaxation. Increasing levels of phospholamban relative to SERCA2a significantly altered intracellular Ca2+ handling in the isolated cardiomyocytes by prolonging the relaxation phase of the Ca2+ transient, decreasing Ca2+ release, and increasing resting Ca2+. These results show that altering the relative ratio of phospholamban to SERVA2a can account for the abnormalities in Ca + handling observed in failing ventricular myocardium. In addition, overexpressing SERCA2a can largely "rescue" the phenotype created by increasing the phospholamban- to-SERCA2a ratio. Restoring the normal phospholamban-to-SERCA2a ratio through somatic gene transfer can correct the abnormalities of Ca2+ handling and contraction seen in failing hearts. Evaluation of Treatment
A treatment can be evaluated by assessing the effect of the treatment on a parameter related to contractility. For example, SR Ca-^4" ATPase activity or intracellular Ca-^4" concentration can be measured, using the methods described above. Furthermore, force generation by hearts or heart tissue can be measured using methods described in Strauss et al., Am. J. Physiol., 262:1437-45, 1992, the contents of which are incorporated herein by reference. In many drug screening programs which test libraries of therapeutic agents and natural extracts, high throughput assays are desirable in order to maximize the number of therapeutic agents surveyed in a given period of time. Assays which are performed in cell-free systems, such as may be derived with cardiac muscle cell extracts, are preferred as "primary" screens in that they can be generated to permit rapid development and relatively easy detection of the parameter being measured, e.g., the intracellular levels of Ca---4", which is mediated by a test therapeutic agent. Moreover, the effects of cellular toxicity and/or bioavailability of the test therapeutic agent can be generally ignored in the in vitro system, the assay instead being focused primarily on the effect of the therapeutic agent on the parameter being measured, e.g., the intracellular levels of Ca---4". It is often desirable to screen candidate treatments in two stages, wherein the first stage is performed in vitro, and the second stage is performed in vivo.
The efficacy of a test therapeutic agent can be assessed by generating dose response curves from data obtained using various concentrations of the test therapeutic agent. Moreover, a control assay can also be performed to provide a baseline for comparison. In the control assay, the heart cell is incubated in the absence of a test agent. Propagation of Heart Cells
A heart cell culture can be obtained by allowing heart cells to migrate out of fragments of heart tissue adhering to a suitable substrate (e.g., a culture dish) or by disaggregating the tissue, e.g., mechanically or enzymatically to produce a suspension of heart cells. For example, the enzymes trypsin, collagenase, elastase, hyaluronidase, DNase, pronase, dispase, or various combinations thereof can be used. Trypsin and pronase give the most complete disaggregation but may damage the cells. Collagenase and dispase give a less complete dissagregation but are less harmful. Methods for isolating tissue (e.g., heart tissue) and the disaggregation of tissue to obtain cells (e.g., heart cells) are described in Freshney R. I., Culture of Animal Cells, A Manual of Basic Technique, Third Edition, 1994, the contents of which are incorporated herein by reference. Viral Vectors Suitable for Somatic Gene Transfer
Expression vectors, suitable for somatic gene transfer, can be used to express the compound, e.g., the phospholamban gene. Examples of such vectors include replication defective retroviral vectors, adenoviral vectors and adeno-associated viral vectors. Adenoviral vectors suitable for use by the methods of the invention include (Ad.RSV.lacZ), which includes the Rous sarcoma virus promoter and the lacZ reporter gene as well as (Ad.CMV.lacZ), which includes the cytomegalovirus promoter and the lacZ reporter gene. Methods for the preparation and use of viral vectors are described in WO 96/13597, WO 96/33281, WO 97/15679, and Trapnell et al., Curr. Opin. Biotechnol. 5(6):617-625, 1994, the contents of which are incorporated herein by reference. Expression of Phospholamban The nucleic acid which results in the overexpression of phospholamban can be derived from the natural phospholamban gene including all the introns and exons, it can be a cDNA molecule derived from the natural gene (Fujji et al., J. Biol. Chem. 266:11669-11675, 1991, the contents of which are incorporated herein by reference) or a chemically synthesized cDNA molecule. The nucleic acid encoding the phospholamban protein can be under the control of the naturally occurring promoter or any other promoter that drives a high level expression of the phospholamban gene.
The following examples which further illustrate the invention should not be construed as limiting. Examples
1. Construction of El -Deleted Recombinant Adenovirus Vectors
The construction of Ad.RSV.SERCA2a has been described in detail by Hajjar et al., Circulation, 95: 423-429, 1997, the contents of which are incorporated herein by reference. Ad.RSV.βgal, which carries a nuclear localizing form of β-galactosidase, is described in Dong et al., J. Biol. Chem. 27:29969-77, 1996, the contents of which are incorporated herein by reference. The rabbit phospholamban cDNA is described in Lylton J. MacLennan D.H., J. Biol. Chem., 1988, 263:15024-15031, the contents of which are incorporated herein by reference. Briefly, the phospholamban cDNA was subcloned into the bacterial plasmid vector pAdRSV4, which uses the RS V long terminal repeat as a promoter and the SV40 polyadenylation signal and contains map units with adenovirus sequences from 0 to 1 and from 9 to 16. The position and orientation of the phospholamban cDNA were confirmed by restriction enzyme digestion and by polymerase chain reaction. The plasmid vector containing phospholamban (pAd.RSV-PL) was then cotransfected into 293 cells with PJM17. The homologous recombinants between pAd.RSV.PL and pJM17 contain the phospholamban cDNA substituted for El . By use of this strategy, independent plaques were isolated, and expression of phospholamban protein was verified by immunostaining. A positive plaque was further plaque-purified, and protein expression was reconfirmed to yield the recombinant adenovirus Ad.RSV.PL. This adenovirus is structurally similar to Ad.RSV.βgal and to Ad.RSV.SERCA2a- described in Dong O. et al., J. Biol. Chem, 1996, 271 :29969-29977, 1976. The recombinant viruses were prepared as high-titer stocks by propagation in 293 cells as described in Graham, F.L. et al., Methods in Molecular Biology: Gene Transfer and Expression Protocols, 1991, 109-128, the contents of which are incorporated herein by reference. The titers of stocks used for these studies were as follows: 3.1x10^ pfu/mL for Ad.RSV.PL 2.6x10 0 pfu/m: for Ad.RSV.SERCA2a, and 2.7xl014 pfu/mL for Ad.RSV.βgal, with a particle- to-pfu ratio of 40:1, 42:1, and 37:1, respectively.
2. Preparation of Neonatal Cardiomyocytes Spontaneously beating cardiomyocytes were prepared from 1 to 2 day old rats and cultured in P-10 medium (GIBCO,BRL) in the presence of 5% fetal calf serum and 10% horse serum for 3 days as described previously in Kang J.X. et al., Proc. Natl. Acad. Sci. U.S.A., 1995, 92:3097-4001 and Kang J.X. and Leaf A., Euro. J. Pharmacol, 1996, 297:97-106, the contents of which are incorporated herein by reference. Measurements of cell shortening and cytosolic Ca---4" were performed on neonatal cardiomyocytes cultured on round, coated, glass coverslips (0.1 mm thickness, 31 mm diameter) in 35 mm culture dishes. Cells were counted using a hemocytometer. Approximately 5x10*^ cells were plated in each coverslip. 3. Adenoviral Infection of Isolated Cells
In three different infection experiments with increasing concentrations of Ad.RSV.βgal, the percentages of cells expressing βgal after 48 hours, by histochemical staining in 10 different high-power fields were 98.2% (multiplicity of infection, 1 pfu/cell), 99.1%) (multiplicity of infection, 10 pfu/cell), and 100% (multiplicity of infection, 100 pfu/cell). In a similar manner, myocardial cells were infected with three concentrations of Ad.RSV.PL 1.0, 10. and 100 pfu/cell for 48 hours. Infection with either Ad.RSV.βgal, Ad.RSV.PL, or Ad.RSV.SERCA2a did not change the morphology of the cells. For each infection experiment with the adenovirus, one myocyte was used to measure functional parameters. As shown in Figure 1 , there was a 4-fold increase in phospholamban protein levels in a dose-dependent increase in the protein expression of phospholamban between 1 and 10 pfu/cell but no further increases between 10 and 100 pfu/cell. Coinfection of Ad.RSV.SERCA2a with Ad.RSV.PL produced an increase in protein expression of both SERCA2a and phospholamban, as shown by the immunoblot in Figure 2. There were no significant differences between the phospholamban protein levels in the group of myocytes infected with Ad.RSV.PL alone at a multiplicity of infection of 10 pfu/cell and the group of myocytes infected with Ad.RSV.PL at an multiplicity of infection of 10 pfu/cell and Ad.RSV.SERCA2a at a multiplicity of infection of 10 pfu/cell (P>2). Similarly, there were no significant differences between the SERCA2a protein levels in the group of myocytes infected with Ad.RSV.SERCA2a alone at a multiplicity of infection of 10 pfu/cell and the group of myocytes infected with Ad.RSV.PL at a multiplicity of infection of 10 pfu/cell and Ad.RSV.SERCA2a at an multiplicity of infection of 10 pfu/cell (P>2).
As shown in Figure 7, cardiomyocytes infected with Ad.RSV.PL (multiplicity of infection of 10 pfu/cell) exhibited a significant increase in resting Ca + not evident in uninfected cells. Furthermore, coinfection with Ad.RSV.SERCA2a (multiplicity of infection of 10 pfu/cell) restored the frequency response to normal. The response to increasing stimulation frequencies in mammalian cardiomyocytes is governed by the SR. We have shown that in the uninfected cardiomyocytes, an increase in stimulation frequency did not significantly alter either peak or resting (Ca2+). This response is typical of rat cardiomyocytes that have either a flat response to increasing frequency of stimulation or a decrease in contractile force. However, in cardiomyocytes infected with Ad.RSV.PL, there was a significantly greater increase in resting (Ca2+) and a decrease in peak (Ca +). These results would suggest that diminished SR Ca2+ uptake leads to a diminished CA2+ release, which becomes even more accentuated at higher frequencies of stimulation.
3. Intracellular Ca-^4" Measurements and Cell Shortening Detection
Measurements of intracellular Ca---4" and cell shortening were performed as described earlier in Hajjar et al. (1997), Kang et al. (1995) and Kang et al (1996), the contents of which are incorporated herein by reference. Briefly, myocardial cells were loaded with the Ca2+ indicator fura 2 by incubating the cells in medium containing 2 μ mol/L fura 2-AM (Molecular Probex) for 30 minutes. The cells were then washed with PBS and allowed to equilibrate for 10 minutes in a light-sealed temperature-controlled chamber (32°C) mounted on a Zeiss Axlovers 10 inverted microscope (Zeiss). The coverslip was superfused with a HEPES-buffered solution at a rate of 20 mL/h. Cells were stimulated at different frequencies (0.1 to 2.0 Hz) using an external stimulator (Grass Instruments). A dual excitation spectrofluorometer (IONOPTIX) was used to record fluorescence emissions (505 nm) elicited from exciting wavelengths of 360 and 380 nm. [Ca^4"] was calculated according to the following formula: [Ca-*-+]=.^/ (R- Rr V/ R-max -R)D, where R is the ratio of fluorescence of the cell at 360 and 380 nm: Rmin anc- R-max represent the ratios of fura 2 fluorescence in the presence of saturating amounts of Ca^4" and effectively "zero Ca^4" respectively, K^ is the dissociation constant of Ca^ " from fura 2; and D is the ratio of fluorescence of fura 2 at 380 nm in zero Ca-^4" and saturating amounts of Ca^4". Unless otherwise stated, measurements of peak [Ca2+] were made at the end of diastole. High-contrast microspheres attached to the cell surface of the cardiomyocytes were imaged using a charge-coupled device video camera attached to the microscope, and motion along a selected raster line segment who quantified by a video motion detector system (IONOPTIX). As shown in Figure 4A, cardiomyocytes infected with Ad.RSV.βgal did not affect the Ca^4" transient or shortening compared with control uninfected cardiomyocytes. As depicted in Figure 4B, the Ca^4" transient and shortening were significantly altered with increasing concentrations of Ad.RSV.PL (multiplicity of infection of 1, 10, and 100 pfu/cell): observed changes included prolongation of the Ca^ " transient and shortening and a decrease in the peak Ca-^4". These results, summarized in the following Table, show that there was a dose-dependent prolongation of the Ca^ " transient and mechanical shortening up to 10 pfu/cell, with no further significant prolongation at 100 pfu/cell, with no further significant prolongation at 100 pfu/cell. Physiological Parameters of Cardiomyocytes Overexpressing Phospholamban
Figure imgf000022_0001
Similarly, peak [Ca---4"] decreased up to 10 pfu/cell, with no further decrease at 100 pfu cell. Coinfection with Ad.RSV.SERCA2a (multiplicity of infection of 10 pfu/cell) restored both the Ca^4" transient and the shortening to near normal levels, as shown in Figure 4C. Fig. 5 shows a significant decrease in mean peak [Ca---4"], a significant increase in mean resting [Ca---4"], and a significant prolongation of the Ca---4" transient in the group of cardiomyocytes infected with Ad.RSV.PL (multiplicity of infection of 10 pfu/cell) compared with uninfected cells (panels a through c, respectively). These effects were partially restored by the addition of Ad.RSV.SERCA2a (multiplicity of infection of 10 pfu/cell) (Figure 5). Similarly, the time course of shortening was significantly prolonged in cardiomyocytes infected with Ad.RSV.PL at a multiplicity of infection of 10 pfu/cell (time to 80% relaxation, from 387±22 to 780±44 milliseconds; P <0.5; n=12), whereas coinfection with Ad.RSV.SERCA2a restored the time course to normal (405±25 milliseconds, n=10, >.1 compared with uninfected cells).
Adenoviral gene transfer of phospholamban provides an attractive system for further elucidation of the effects of inhibiting SR Ca2+-ATPase on intracellular Ca2+ handling. A decrease in SRCa2+ uptake rates is expected to lead to a smaller amount of Ca2+ sequestered by the SR, resulting in a smaller amount of Ca2+ release. In neonatal cardiomyocytes, a significantly prolonged Ca2+ transient and a higher resting (Ca2+) was observed reflecting the decreased Ca24 uptake and a decrease in peak (Ca +) levels reflecting less Ca + available for release. These results show that the SR Ca24- ATPase is important during relaxation by controlling the rate and amount of C A2+ sequestered and during contraction by releasing the Ca + that is taken up by the SR. Overexpression of both phospholamban and SERCA2a partially restored the Ca2+ transient; however, the time course of the Ca2+ transient was still prolonged in cardiomyocytes infected with both Ad.RSV.SERCA2a and Ad.RSV.PL. This finding was somewhat surprising, since the SR Ca2+-ATPase activity was restored to normal and even enhanced in cardiomyocytes infected with both Ad.RSV.SERCA2a and Ad.RSV.PL. Phospholamban has been shown to play a key role in modulating the response of agents that increase cAMP levels in cardiomyocytes. Since phosphorylation of phospholamban reduces the inhibition to the SR Ca2+ pump, thereby enhancing the SR Ca2+- ATPase, we were specifically interested in evaluating the effects of β-agonism on the relaxation phase of the Ca2+ transient. In the basal state, the overexpression of phospholamban significantly prolongs the Ca2+ transient. As shown in Figure 6, at maximal isoproterenol stimulation, the time course of the Ca2+ transients in the uninfected cardiomyocytes and the cardiomyocytes infected with Ad. RSV.PL were decreased to the same level. These findings show that phospholamban plays a major role in the enhanced relaxation of the heart to β-agonism. In addition, it corroborates these findings that phospholamban decreases the affinity of the SR Ca2+ pump for Ca + but does not decrease the maximal Ca2+ uptake rate.
4. Preparation of SR Membranes From Isolated Rat Cardiomyocytes To isolate SR membrane from cultured cardiomyocyes, a procedure modified from Harigaya et al., Circ. Res., 1969, 25:781-794, as well as, Wienzek et al., 1992, 23:1149-1163, the contents of which are incorporated herein by reference, was used. Briefly, isolated neonatal cardiomyocytes were suspended in a buffer containing (mmol/L) sucrose 500, phenylmerhyisulfonyl fluoride 1 and PIPES 20, at pH 7.4. The cardiomyocytes were then disrupted with a homogenizer. The homogenates were centrifuged at 500g for 20 minutes. The resultant supernatant was centrifuged at 25,000g for 60 minutes to pellet the SR-enriched membrane. The pellet was resuspended in a buffer containing (mmol/L) KC1 600, sucrose 30, and PIPES 20, frozen in liquid nitrogen, and stored at -70°C. Protein concentration was determined in these preparations by a modified Bradford procedure, described in Bradford et al., Anal.
Biochem., 1976, 72:248-260, the contents of which are incorporated herein by reference, using bovine scrum albumin for the standard curve (Bio-Rad).
5. Western Blot Analysis of Phospholamban and SERCA2a in SR Preparations
SDS-PAGE was performed on the isolated membranes from cell cultures under reducing conditions on a 7.5%> separation gel with a 4% stacking gel in a Miniprotean II cell (Bio-Rad). Proteins were then transferred to a Hybond-ECL nitrocellulose for 2 hours. The blots were blocked in 5% nonfat milk in Tris-buffered saline for 3 hours at room temperature. For immunoreaction, the blot was incubated with 1 :2500 diluted monoclonal anti-SERCA2 antibody (Affinity BioReagents) or 1 :2500 diluted anti- cardiac phospholamban monoclonal IgG (UBI) for 90 minutes at room temperature.
After washing, the blots were incubated in a solution containing peroxidase-labeled goat anti-mouse IgG (dilution, 1 :1000) for 90 minutes at room temperature. The blot was then incubated in a chemiluminescence system and exposed to an X-OMAT x-ray film (Fuji Films) for 1 minute. The densities of the bands were evaluated using NIH Image. Normalization was performed by dividing densitometric units of each membrane preparation by the protein amounts in each of these preparations. Serial dilution of the membrane preparations revealed a linear relationship between amounts of protein and the densities of the SERCA2a immunoreactive hands (data not shown). 6. SR Ca2+-ATPase Activity
SR Ca2+-ATPase activity assays were carried out according to Chu A. et al., Methods Enzymol, 1988, 157:36-46, the contents of which are incorporated herein by reference, on the basis of pyruvate NADH-coupled reactions. By use of a photomotor (Beckman DU 640) adjusted at a wavelength of 540 nm, oxidation of NADH (which is coupled to the SR Ca---4"- ATPase) was assessed at 37°C in the membrane preparations by the difference of the total absorbance and basal absorbance. The reaction was carried out in a volume of 1 mL. All experiments were carried out in triplicate. The activity of the Ca-"-+-ATPase was calculated as follows: Δabsorbance/6.22 x protein x time (in nmol ATP/mg protein x min). The measurements were repeated at different [Ca---4"] levels. The effect of the specific Ca-"-+-ATPase inhibitor CPA at a concentration range of 0.001 to 10 μmol/L was also studied in those preparations, as described in Schwinger et al., Circulation, 1995, 92:3220-3228 and Baudet et al., Circ. Res., 1993, 73:813-819, the contents of which are incorporated herein by reference. As shown in Figure 3 A, the relationship between ATPase activity and Ca^ " was shifted to the right in the preparations from cardiomyocytes overexpressing phospholamban compared with the uninfected preparations without changing maximal Ca-"-+-ATPase activity. Coinfection with Ad.RSV.SERCA2a restored the CA2+-ATPase activity and also increased the maximal Ca2+-ATPase activity. To verify that the ATPase activity measured from the membrane preparations was SR-related, the specific inhibitor CPA was used after maximally activating the SR Ca-^4"- ATPase with 10 μmol/L of Ca---4". As shown in Figure 3B, CPA inhibited the SR Ca-"-+-ATPase activity in a dose-dependent fashion in all three membrane preparations (uninfected, Ad.RSV.PL, and d.RSV.PL+Ad.RSV.SERCA2a).
The SR Ca2+- ATPase plays a key role in excitation-contraction coupling, lowering Ca + during relaxation in cardiomyocytes, and "loading" the SR with Ca2+ for the subsequent release and contractile activation. The Ca +-pumping activity of this enzyme is influenced by phospholamban. In the unphosphorylated state, phospholamban inhibits the Ca2+- ATPase, whereas phosphorylation of phospholamban by cAMP-dependent protein kinase and by Ca2+ calmodulin-dependent protein kinase reverses this inhibition. Therefore, an increase in phospholamban content should decrease the affinity of the SR Ca2+ pump for Ca2+. As shown in Figure 4, overexpression of phospholamban shifted the relationship between SR Ca +- ATPase activity and Ca2+ to the right, indicating a decrease of the sensitivity of the SR Ca2+ to pump to Ca +. However, there was no change in the maximal Ca2+-ATPase activity in the Ad.RSV. PL-infected cardiomyocytes. This shows that the Vmax of the Ca2+- ATPase of cardiac SR is not altered by interaction with phospholamban and phosphorylation, and that in mice overexpressing phospholamban, the affinity of the SR Ca + pump for Ca2+ was decreased but that the maximal velocity of the SR Ca2+ uptake was not changed. From the present experiment, it can also be concluded that phospholamban affects the affinity of the SR Ca2+ pump for CA2+ without changing the maximal ATPase activity. The concomitant overexpression of SERCA2a and phospholamban restored the ATPase activity and also increased the maximal Ca2+-ATPase activity. This brings further evidence that the expression of additional SR Ca2+-ATPase pumps can overcome the inhibitory effects of phospholamban.
7. Statistical Analyses
Data were represented as mean ± SEM for continuous variables. Stutent's test was used to compare the means of normally distributed continuous variables. Parametric one-way ANOVA techniques were used to compare normally distributed contiguous variables among uninfected groups of cells, Ad.RSV. βgal-infected cells, Ad.RSV.PL- infected cells, and Ad.RSV.SERCA2a-infected cells.
8. Retroviral Somatic Gene Transfer
Rats and mice were anesthetized with intraperitoneal pentobarbital and placed on a ventilator. The chest was entered form the left side through the third intercostal space. The pericardium was opened and a 7-0 suture placed at the apex of the left ventricle. The aorta and pulmonary artery were identified. A 22 G catheter containing 200 μl of adenovirus was advanced from the apex of the left ventricle to the aortic root. The aorta and pulmonary artery were clamped distal to the site of the catheter and the adenovirus solution was injected as shown in Figure 9. The clamp was maintained for 10 seconds while the heart was pumping against a closed system (isovolumically). This allowed the adenovirus solution to circulate down the coronary arteries and perfuse the whole heart without direct manipulation of the coronaries. After the 10 seconds, the clamp on the aorta and the pulmonary artery was released, the chest was evacuated from air and blood and closed. Finally, the animals were taken off the ventilator.
The expression pattern seen after direct injection is localized whereas the catheter-based technique is essentially homogeneous. The pressure tracing of the Ad.PL transduced hearts displayed a markedly prolonged relaxation and reduced pressure development as shown in Figure 8. 9. Gene Transfer of the Sarcoplasmic Reticulum Calcium ATPase Improves Left Ventricular Function in Aortic-Banded Rats in Transition to Failure In human and experimental models of heart failure, sarcoplasmic reticulum Ca-^4" ATPase (SERCA2a) activity has been shown to be significantly decreased. In this example the ability of SERCA2a expression to improve ventricular function in heart failure was investigated by creating an ascending aortic constriction in 10 rats. After 20- 24 weeks, during the transition from left ventricular hypertrophy to failure, 200 μl of a solution containing 5x10^ plaque forming units of replication-deficient adenovirus carrying SERCA2a (Ad.SERCA) (n*=4) or the reporter gene β-galctosidase (Ad.βgal) (n=6) were injected intracoronary via the catheter-based technique described supra. Two days after the procedure, the rats underwent open chest measurement of left ventricular pressure. Heart rate (HR), left ventricular end-diastolic pressure (LVEDP), and left ventricular systolic pressure (LVSP) were measured. Peak +dP/dt and -dP/dt were calculated. As shown in Table 1, the magnitudes of peak +dP/dt and -dP/dt which are indices of systolic and diastolic function were markedly increased in hearts transduced with the SERCA2a carrying adenovirus. Therefore, this example indicates that overexpression of SERCA2a in a rat model of pressure-overload hypertrophy in transition to failure improved left ventricular systolic and diastolic function.
TABLE 1
Figure imgf000026_0001
# p<0.05 compared to Ad.βgal
10. Gene Transfer of Antisense of Phospholamban Improves Contractility in Isolated Cardiomyocytes
Delayed cardiac relaxation in failing hearts is attributed to a reduced activity of the Sarcoplasmic Reticulum Calcium ATPase. Phospholamban inhibits SERCA2a activity and is, therefore, a potential target to improve cardiac function. In this Example, an adenovirus carrying the full length antisense cDNA of phospholamban (Ad.asPL) was constructed using the methods described above. This construct was then used to infect neonatal cardiac myocytes as described in Example 3. As indicated in Figure 10A, infection of neonatal cardiac myocytes with the Ad.asPL construct increased the contraction amplitude and significantly shortened the time course of the contraction. The adenovirus-mediated gene transfer of the antisense cDNA for phospholamban also resulted in a modification of intracellular calcium handling and shortening in myocardial cells (see Figure 10B) indicating that such vectors can be used for increasing the contractility of myocardial cells in heart failure.
Equivalents Those skilled in the art will recognize, or be able to ascertain, using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims:

Claims

CLAIMS:
1. A method of evaluating a treatment for a heart disorder, comprising: providing a heart cell, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; administering the treatment to the heart cell; and evaluating the effect of the treatment on the heart cell, thereby evaluating the treatment for a heart disorder.
2. The method of claim 1, wherein the evaluation includes evaluating the effect of the treatment on a parameter related to contractility.
3. The method of claim 2, wherein the parameter related to contractility is intracellular Ca---4" concentration.
4. The method of claim 2, wherein the parameter related to contractility is SR Ca24" ATPase activity.
5. The method of claim 1 , wherein the treatment is administered in vivo.
6. The method of claim 5, wherein the treatment is administered to an experimental animal.
7. The method of claim 6, wherein the experimental animal is a transgenic animal.
8. The method of claim 7, wherein the transgenic animal expresses a transgene encoding a protein in the phospholamban pathway.
9. The method of claim 1 , wherein the treatment is administered in vitro.
10. The method of claim 1 , wherein the nucleic acid encodes a phospholamban protein.
11. A method of evaluating a treatment for a heart disorder, comprising: providing a heart, into some or all the cells of which has been introduced, by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; administering the treatment to the heart; and evaluating the effect of the treatment on the heart, thereby evaluating the treatment for a heart disorder.
12. The method of claim 11 , wherein the evaluation includes evaluating the effect of the treatment on a parameter related to contractility.
13. The method of claim 12, wherein the parameter related to contractility is intracellular Ca24" concentration.
14. The method of claim 12, wherein the parameter related to contractility is SR Ca2+ ATPase activity.
15. The method of claim 12, wherein the parameter related to contractility is force generation.
16. The method of claim 11, wherein the treatment is administered in vivo.
17. The method of claim 11 , wherein the treatment is administered to an experimental animal.
18. The method of claim 17, wherein the experimental animal is a transgenic animal.
19. The method of claim 18, wherein the transgenic animal expresses a transgene encoding a protein in the phospholamban pathway.
20. The method of claim 11, wherein the treatment is administered in vitro.
21. The method of claim 11 , wherein the nucleic acid encodes a phospholamban protein.
22. A method of evaluating a treatment for a heart disorder, comprising: providing heart tissue into some or all of the cells of which has been introduced by somatic gene transfer a nucleic acid which results in the expression of phospholamban; administering the treatment to the heart tissue; and evaluating the effect of the treatment on the heart tissue, thereby evaluating the treatment for a heart disorder.
23. The method of claim 22, wherein the evaluation includes evaluating the effect of the treatment on a parameter related to contractility.
24. The method of claim 23, wherein the parameter related to contractility is intracellular Ca2+ concentration.
25. The method of claim 23, wherein the parameter related to contractility is SR Ca24" ATPase activity.
26. The method of claim 23, wherein the parameter related to contractility is force generation.
27. The method of claim 22, wherein the treatment is administered in vivo.
28. The method of claim 22, wherein the treatment is administered to an experimental animal.
29. The method of claim 28, wherein the experimental animal is a transgenic animal.
30. The method of claim 29, wherein the transgenic animal expresses a transgene encoding a protein in the phospholamban pathway.
31. The method of claim 22, wherein the treatment is administered in vitro.
32. The method of claim 22, wherein the nucleic acid encodes a phospholamban protein.
33. A method of evaluating a treatment for a heart disorder, comprising: providing a first and a second heart cell, into each of which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; administering the treatment to a first heart cell in vitro; evaluating the effect of the in vitro treatment on the first heart cell; administering the treatment to a second heart cell in vivo; and evaluating the effect of the in vivo treatment on the second heart cell, thereby evaluating the treatment for a heart disorder.
34. A method of evaluating a treatment for a heart disorder, comprising: providing a first administration of the treatment to a heart cell, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; evaluating the effect of the first administration on the heart cell; providing a second administration of the treatment to a heart cell, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; and evaluating the effect of the second administration on the heart cell, thereby evaluating the treatment for a heart disorder.
35. The method of claim 34, wherein the first and second administration are administered to the same cell.
36. The method of claim 34, wherein the first and second administration are administered to different cells.
37. The method of claim 34, wherein the first and second administration are administered under the same conditions.
38. The method of claim 34, wherein the first and second administration are administered under different conditions.
39. A method of evaluating a treatment for a heart disorder, comprising: providing a heart cell, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; administering the treatment to the heart cell; evaluating the effect of the treatment on the heart cell; providing a heart, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban; administering the treatment to the heart ; and evaluating the effect of the treatment on the heart, thereby evaluating the treatment for a heart disorder.
40. The method of claim 39, wherein the treatment is administered to the heart cell in vitro and to the heart in vivo.
41. The method of claim 39, wherein the treatment is administered to the heart cell and to the heart in vitro.
42. A method of delivering a compound to the heart of a subject, comprising: restricting the aortic flow of blood out of the heart, such that blood flow is re-directed to the coronary arteries; introducing said compound into the lumen of the circulatory system, such that said compound flows into the coronary arteries; allowing the heart to pump while the aortic outflow of blood is restricted; and reestablishing the flow of blood, thereby allowing said compound to flow into and be delivered to the heart.
43. The method of claim 42, wherein the compound comprises a nucleic acid which directs the expression of a peptide.
44. The method of claim 42, wherein the compound comprises a virus vector suitable for somatic gene delivery.
45. The method of claim 40, wherein the vector is an adenovirus vector.
46. The method of claim 43, wherein the peptide is phospholamban.
47. The method of claim 42, wherein the subject is a human.
48. The method of claim 47, wherein the human is suffering from a cardiac disorder.
49. The method of claim 48, wherein the cardiac disorder is heart failure.
50. The method of claim 48, wherein the cardiac disorder is ischemia.
51. The method of claim 48, wherein the cardiac disorder is transplant rejection.
52. The method of claim 42, wherein the subject is an experimental animal.
53. The method of claim 52, wherein the experimental animal is a transgenic animal.
54. The method of claim 53, wherein the transgenic animal expresses a transgene encoding a protein inthe phospholamban pathway.
55. The method of claim 42, wherein the blood flow restriction is achieved by obstructing the aorta.
56. The method of claim 55, further comprising obstructing the pulmonary artery.
57. The method of claim 42, further comprising opening the pericardium.
58. The method of claim 42, wherein the compound is introduced by a catheter.
59. The method of claim 43, wherein the nucleic acid, which directs the expression of the peptide, is homogeneously overexpressed in the heart of the subject.
60. The method of claim 42, wherein the compound is introduced into the aortic root.
61. The method of claim 42, wherein the compound is introduced into the lumen of the heart.
62. A heart cell, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban.
63. A heart tissue, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban.
64. A heart, into which has been introduced by somatic gene transfer, a nucleic acid which results in the expression of phospholamban.
65. A method for treating a subject at risk for a heart disorder, comprising: introducing into somatic heart tissue of the subject, a nucleic acid which encodes phospholamban.
PCT/US1998/014968 1997-07-22 1998-07-20 Evaluation of, delivery of, and use of agents to treat heart disorders WO1999004636A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CA002297328A CA2297328A1 (en) 1997-07-22 1998-07-20 Evaluation of, delivery of, and use of agents to treat heart disorders
EP98934656A EP1009236A4 (en) 1997-07-22 1998-07-20 Evaluation of, delivery of, and use of agents to treat heart disorders

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US5335697P 1997-07-22 1997-07-22
US60/053,356 1997-07-22

Publications (2)

Publication Number Publication Date
WO1999004636A1 true WO1999004636A1 (en) 1999-02-04
WO1999004636A9 WO1999004636A9 (en) 1999-04-29

Family

ID=21983650

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1998/014968 WO1999004636A1 (en) 1997-07-22 1998-07-20 Evaluation of, delivery of, and use of agents to treat heart disorders

Country Status (3)

Country Link
EP (1) EP1009236A4 (en)
CA (1) CA2297328A1 (en)
WO (1) WO1999004636A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000063426A2 (en) * 1999-04-15 2000-10-26 Devgen Nv Compound screening methods
WO2002097128A2 (en) * 2001-05-30 2002-12-05 Medigene Ag Method for identifying substances directed against myocardial cell hypertrophy
US7291604B2 (en) 2003-09-03 2007-11-06 The General Hospital Corporation Methods of treating restenosis

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
FUJII J., ET AL.: "STRUCTURE OF THE RABBIT PHOSPHOLAMBAN GENE, CLONING OF THE HUMAN CDNA, AND ASSIGNMENT OF THE GENE TO HUMAN CHROMOSOME 6.", JOURNAL OF BIOLOGICAL CHEMISTRY, AMERICAN SOCIETY FOR BIOCHEMISTRY AND MOLECULAR BIOLOGY, US, vol. 266., no. 18., 25 June 1991 (1991-06-25), US, pages 11669 - 11675., XP002914767, ISSN: 0021-9258 *
HAJJAR R. J., ET AL.: "EFFECTS OF ADENOVIRAL GENE TRANSFER OF PHOSPHOLAMBAN ON LATRACELLULAR CALCIUM HOMEOSTATIS IN ISOLATED MYOCYTES.", CIRCULATION, LIPPINCOTT WILLIAMS & WILKINS, US, vol. 94., no. 08., 15 October 1996 (1996-10-15), US, pages I - 159., XP002914769, ISSN: 0009-7322 *
KOCH W. J., ET AL.: "TRANSGENIC MANIPULATION OF MYOCARDIAL G PROTEIN-COUPLED RECEPTORS AND RECEPTOR KINASES.", CIRCULATION RESEARCH., GRUNE AND STRATTON, BALTIMORE, US, vol. 78., no. 04., 1 April 1996 (1996-04-01), US, pages 511 - 516., XP002914768, ISSN: 0009-7330 *
See also references of EP1009236A4 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000063426A2 (en) * 1999-04-15 2000-10-26 Devgen Nv Compound screening methods
WO2000063426A3 (en) * 1999-04-15 2001-02-08 Devgen Nv Compound screening methods
WO2002097128A2 (en) * 2001-05-30 2002-12-05 Medigene Ag Method for identifying substances directed against myocardial cell hypertrophy
WO2002097128A3 (en) * 2001-05-30 2003-09-12 Medigene Ag Method for identifying substances directed against myocardial cell hypertrophy
US7291604B2 (en) 2003-09-03 2007-11-06 The General Hospital Corporation Methods of treating restenosis
US8133878B1 (en) 2003-09-03 2012-03-13 The General Hospital Corporation Methods for treating restenosis

Also Published As

Publication number Publication date
WO1999004636A9 (en) 1999-04-29
EP1009236A4 (en) 2000-10-25
EP1009236A1 (en) 2000-06-21
CA2297328A1 (en) 1999-02-04

Similar Documents

Publication Publication Date Title
US20200171059A1 (en) Modulating phosphatase activity in cardiac cells
US20050095227A1 (en) Treating heart failure
Hajjar et al. Adenoviral gene transfer of phospholamban in isolated rat cardiomyocytes: rescue effects by concomitant gene transfer of sarcoplasmic reticulum Ca2+-ATPase
US20090239940A1 (en) Treating heart failure and ventricular arrhythmias
Hajjar et al. Physiological effects of adenoviral gene transfer of sarcoplasmic reticulum calcium ATPase in isolated rat myocytes
US6436907B1 (en) Adenovirus-medicated gene transfer to cardiac and vascular smooth muscle
CA2837322C (en) Cell and gene based methods to improve cardiac function
Kypson et al. Adenovirus-mediated gene transfer of the β2-adrenergic receptor to donor hearts enhances cardiac function
US6605274B1 (en) Method for in vivo regulation of cardiac muscle contractility
EP0934422B1 (en) Gene therapy for congestive heart failure
US20020040010A1 (en) Use of agents to treat heart disorders
EP1009236A1 (en) Evaluation of, delivery of, and use of agents to treat heart disorders
EP1374909A2 (en) Evaluation of, delivery of, and use of agents to treat heart disorders
US20050250721A1 (en) Gene therapy for congestive heart failure
US8007778B2 (en) Use of calcium binding proteins to improve cardiac contractile function
Berman et al. Metabolic mechanisms and diabetes candidate genes: insights gained from genetic engineering with recombinant adenoviruses
KR20010029483A (en) Gene therapy for congestive heart failure

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): CA JP

AL Designated countries for regional patents

Kind code of ref document: A1

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

AK Designated states

Kind code of ref document: C2

Designated state(s): CA JP

AL Designated countries for regional patents

Kind code of ref document: C2

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

COP Corrected version of pamphlet

Free format text: PAGES 1-25, DESCRIPTION, REPLACED BY NEW PAGES 1-25; PAGES 26-30, CLAIMS, REPLACED BY NEW PAGES 26-30; PAGES 1/17-17/17, DRAWINGS, REPLACED BY NEW PAGES 1/15-15/15; DUE TO LATE TRANSMITTAL BY THE RECEIVING OFFICE

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

Ref document number: 2297328

Country of ref document: CA

Ref country code: CA

Ref document number: 2297328

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1998934656

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1998934656

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1998934656

Country of ref document: EP