Skip to main content

Gene therapy for lipid disorders

Abstract

Lipid disorders are associated with atherosclerotic vascular disease, and therapy is associated with a substantial reduction in cardiovascular events. Current approaches to the treatment of lipid disorders are ineffective in a substantial number of patients. New therapies for refractory hypercholesterolemia, severe hypertriglyceridemia, and low levels of high-density lipoprotein cholesterol are needed: somatic gene therapy is one viable approach. The molecular etiology and pathophysiology of most of the candidate diseases are well understood. Animal models exist for the diseases and in many cases preclinical proof-of-principle studies have already been performed. There has been progress in the development of vectors that provide long-term gene expression. New clinical gene therapy trials for lipid disorders are likely to be initiated within the next few years.

Introduction

A reduction in plasma cholesterol levels is associated with a substantial reduction in cardiovascular events [1]. Methods for reducing cholesterol levels and specifically low-density lipoprotein (LDL) cholesterol include lifestyle changes and drug therapy, including statins, niacin, bile acid sequestrants, and fibrates [2]. However, many patients cannot achieve optimal LDL cholesterol levels with existing cholesterol-lowering therapies [2]. The majority of these patients have genetic disorders that cause severe hypercholesterolemia that is partly refractory to existing therapies, whereas other patients do not tolerate existing therapies. Hypercholesterolemia therefore remains an important target for the development of novel therapies.

Plasma triglycerides are increasingly recognized as a risk factor for cardiovascular disease [3], and severe hypertriglyceridemia is associated with acute pancreatitis. Although triglyceride levels can frequently be decreased with a combination of lifestyle changes and drug therapy such as fibrates and niacin, there remain a substantial number of individuals in which triglycerides remain significantly elevated. Hypertriglyceridemia is therefore also a target for the development of novel therapies.

Finally, low plasma levels of high-density lipoprotein (HDL) cholesterol are associated with significantly increased cardiovascular risk [4]. Low-HDL cholesterol is one of the most common lipid disorders in patients with premature coronary artery disease [5]. There are a variety of genetic causes of low-HDL cholesterol [6]. A variety of data from animals and from humans suggest that increasing the HDL cholesterol concentration is beneficial. However, the majority of patients have difficulty raising the HDL cholesterol level despite lifestyle changes and drug therapy such as niacin and fibrates. Low-HDL cholesterol is therefore a very important target for the development of novel therapies.

There is much interest in the development of new small molecules that decrease LDL cholesterol, decrease triglycerides, or raise HDL cholesterol levels. In addition, gene therapy has been considered as a potential approach to some of these targets. The purpose of this article is to review the specific lipid disorders that are potential candidates for the development of clinical gene therapy, to discuss the specific issues involved for these disorders, to summarize the proof-of-principle studies in animals where such studies exist, and to comment on the potential progress toward clinical trials of gene therapy for these disorders.

Lipid disorders that are candidates for gene therapy

Disorders associated primarily with hypercholesterolemia or hypertriglyceridemia

Familial hypercholesterolemia

Familial hypercholesterolemia (FH) is caused by mutations in the LDL receptor [7]. Homozygous FH, owing to mutations in both LDL receptor alleles, occurs in about 1 in one million individuals worldwide and is associated with markedly elevated LDL cholesterol levels (about 500-1000 mg/dl) and markedly premature atherosclerotic cardiovascular disease. FH homozygotes are highly resistant to currently available drug therapy. The preferred treatment at present is LDL apheresis, a physical procedure in which LDL is selectively removed from the blood by passing plasma over columns that bind the LDL [8]. LDL apheresis must be performed every 1-2 weeks, and although patients on regular apheresis seem to have delayed onset and progression of atherosclerotic cardiovascular disease, they remain at a much higher than average risk. For these reasons, homozygous FH is a candidate for the development of gene therapy.

Although LDL receptors are expressed ubiquitously, the hepatic LDL receptor has the greatest quantitative effect in controlling plasma LDL levels. Importantly, a limited number of patients with homozygous FH have undergone liver transplantation and have experienced substantial reductions in LDL cholesterol levels. This indicates that gene therapy targeted toward the liver would be likely to be an effective therapy for this disease. The number of hepatocytes and the level of gene expression necessary to result in a therapeutic effect are uncertain. However, it is worth noting that patients with homozygous FH are frequently classified as either receptor-negative (no detectable functional LDL receptor activity) or receptor-defective (markedly reduced but detectable LDL receptor activity). Receptor-negative FH is associated with significantly higher cholesterol levels and earlier onset and severity of atherosclerotic cardiovascular disease than receptor-defective FH. A gene therapeutic approach that resulted in enough LDL receptor expression to convert a receptor-negative patient to 'receptor-defective' could therefore be considered therapeutically beneficial.

A considerable number of proof-of-principle studies have been performed in animal models of homozygous FH. The Watanabe heritable hyperlipidemic (WHHL) rabbit is homozygous for a natural mutation in the LDL receptor. A recombinant retrovirus was used to achieve ex vivo gene transfer of the rabbit LDL receptor to hepatocytes from WHHL rabbits, with subsequent reinfusion of the genetically modified hepatocytes. Plasma cholesterol levels were reduced by about 30% and persisted for 6 months [9]. Injection of a recombinant first-generation adenovirus encoding the LDL receptor into WHHL rabbits transiently reduced LDL cholesterol levels [10,11]. The LDL receptor knockout mouse has also been used as an FH model. The administration of a first-generation adenovirus encoding the human LDL receptor transiently reduced LDL cholesterol levels in chow-fed mice deficient in LDL receptor [12] and reduced cholesterol levels and atherosclerosis in mice deficient in LDL receptor that were fed with a diet high in fat (M Kawashiri, DJ Rader, unpublished data). Administration of antibody against CD40 ligand at the time of administration on recombinant LDL receptor adenovirus decreased immune-mediated promoter silencing and resulted in long-term transgene expression and cholesterol decrease in mice deficient in LDL receptor [13].

Although the LDL receptor is the logical gene to use for gene therapy of FH, there is concern that the LDL receptor expressed as a result of somatic gene transfer could be immunogenic in patients that lack the ability to synthesize the LDL receptor. Other genes have therefore been used in animals in attempts to ameliorate the hypercholesterolemia associated with a deficiency in LDL receptors. The very-low-density lipoprotein (VLDL) receptor is a lipoprotein receptor with homology to the LDL receptor, but it is not normally expressed in the liver. The murine VLDL receptor was expressed in the livers of mice deficient in LDL receptor by using a first-generation adenovirus; LDL cholesterol levels were reduced to a comparable degree and for a longer period compared with the expression of the LDL receptor [14]. A recombinant adeno-associated virus (AAV) was used to express the murine VLDL receptor in the livers of mice deficient in LDL receptor fed on a high-fat Western diet. Expression of the VLDL receptor decreased plasma cholesterol levels significantly for more than 7 months and reduced the development and progression of atherosclerosis [15]. Another novel approach is the use of chimeric molecules made by fusion of a soluble form of the LDL receptor with transferrin [16] that could potentially bind LDL in the circulation and mediate clearance through the transferrin receptor on hepatocytes.

Homozygous FH is the only lipid disorder that has been approached in a clinical gene therapy trial [17]. The trial used an ex vivo approach in which five homozygous FH patients underwent a surgical resection of the left lateral segment of the liver. Primary hepatocytes were isolated and after 2 days were infected with a recombinant retrovirus encoding the human LDL receptor gene. The genetically modified hepatocytes were harvested 24 h later and reinfused into the patients via a portal catheter that was placed in the inferior mesenteric vein at the time of the original surgery. Patients tolerated the infusions of autologous hepatocytes well without complications. Two of five patients had a decrease of about 20% in their LDL cholesterol levels. Kinetic studies of LDL metabolism demonstrated that LDL catabolism was increased in the same two patients, which was consistent with increased LDL receptor expression. Liver biopsies performed 4 months after treatment revealed LDL receptor transgene expression by in situ hybridization in all five subjects.

Homozygous FH will continue to be a model for the development of liver-directed somatic gene therapy. However, many heterozygous FH patients are also relatively refractory to existing drug therapy and remain at very high risk for development and progression of atherosclerotic vascular disease. Therefore, once a gene therapeutic strategy is found to be effective in homozygous FH, it might be extended to clinical trials in severe heterozygous FH as well.

Lipoprotein lipase deficiency (familial hyperchylomicronemia)

Lipoprotein lipase (LPL) deficiency is a rare autosomal recessive disorder characterized by markedly elevated plasma levels of triglycerides [18]. Normally, triglycerides in chylomicrons are hydrolyzed by LPL, and the genetic absence of LPL results in marked hyperchylomicronemia. LPL deficiency is associated with recurrent episodes of acute pancreatitis, which can be life-threatening and can also lead to chronic pancreatic insufficiency. LPL deficiency is often refractory to diet and drug therapy and is therefore a candidate for the development of gene therapy.

Several animal models of LPL deficiency exist. LPL knockout mice have severe hypertriglyceridemia and die within 24 h of birth [19]. Germline transgenic expression of LPL in skeletal muscle rescued the mice and normalized the triglyceride levels [20]. Cats and minks that are naturally deficient in LPL have been reported [21,22]. Liver-directed overexpression of human LPL with an adenoviral vector in LPL-deficient cats reduced triglyceride levels for up to 14 days [23].

Most LPL expression is in muscle and adipose tissue. Because skeletal muscle is an attractive target for somatic gene transfer, LPL deficiency is a candidate for the development of muscle-directed gene therapy. The intramuscular injection of recombinant AAV vectors in mice resulted in the stable expression of several secreted proteins [24,25,26]. Correction of hemophilia B was achieved by the intramuscular injection of an AAV vector in dogs [27] and in humans [28]. AAV might therefore be an attractive vector for the use of muscle-directed LPL gene transfer. The major issues are whether enough LPL expression could be achieved to normalize triglyceride levels and the duration of gene expression.

Gene transfer to increase LPL expression could potentially have therapeutic value in conditions other than homozygous LPL deficiency. Transgenic mice overexpressing LPL have less diet-induced hyperlipidemia, less hyperlipidemia associated with diabetes, and reduced diet-induced atherosclerosis. Liver-directed gene transfer of LPL with recombinant adenovirus decreased fasting and post-prandial levels of triglycerides in heterozygous LPL-deficient mice [29] and decreased levels of triglycerides and cholesterol in mice deficient in apolipoprotein E (apoE) and in LDL receptor [30].

ApoE deficiency and familial dysbetalipoproteinemia

ApoE serves as a ligand that mediates the clearance of chylomicron and VLDL remnant lipoproteins by binding to the LDL receptor and related members of the same gene family [31]. Most apoE in plasma is derived from the liver. A genetic deficiency of apoE results in substantially elevated levels of lipoprotein remnants and is associated with an increased risk for atherosclerotic vascular disease [32]. ApoE knockout mice develop high levels of remnant lipoproteins and severe atherosclerosis on a chow diet [33]. Liver-directed gene transfer of human apoE markedly decreased plasma cholesterol levels for about 2 weeks with a first-generation adenovirus [34,35] and for at least 8 weeks with a second-generation adenovirus [36]. Gene transfer of apoE to the liver in apoE-deficient mice also induced substantial regression of atherosclerosis [37,38]. Although plasma apoE is mostly derived from the liver, apoE produced in any tissue could theoretically be effective if secreted into the blood. ApoE derived from macrophages after transplantation of wild-type bone marrow into apoE-deficient mice also normalized plasma cholesterol levels and reduced the development of atherosclerosis [39]. ApoE gene therapy in patients with apoE deficiency would correct the hyperlipidemia but this condition is extremely rare and is therefore not an optimal model for the development of this type of gene therapy.

ApoE gene therapy could have therapeutic value in another apoE-related lipid disorder sometimes called familial dysbetalipoproteinemia or Type III hyperlipoproteinemia [40]. In this disorder, a mutant apoE has decreased binding to receptors, thus resulting in a reduced clearance of remnant lipoproteins and hyperlipidemia. The most common cause is homozygosity for a common polymorphism in apoE called apoE2 (the wild-type form is known as apoE3). Rarer causes are other apoE mutations that often cause hyperlipidemia in a dominant fashion. Gene therapy to increase the hepatic production of wild-type apoE3 could be a therapeutic strategy for this condition.

Familial combined hyperlipidemia and diabetic dyslipidemia

Familial combined hyperlipidemia is an inherited phenotype characterized by elevated VLDL and LDL; it is believed to be normally due to overproduction of VLDL by the liver [41]. The specific genetic basis of this common disorder is currently unknown. Type II diabetes mellitus is also characterized by the hepatic overproduction of VLDL and increased VLDL and LDL cholesterol [42]. Both of these conditions are associated with a significantly increased risk for atherosclerotic vascular disease. Although many patients with these disorders can be adequately treated with lifestyle and drug therapy, a substantial number cannot achieve adequate control of their hyperlipidemia. Gene therapy might therefore be one therapeutic approach to this class of patients with refractory combined hyperlipidemia.

One approach could be expression of the VLDL receptor in the liver. As noted above, although the VLDL receptor is not normally expressed in the liver, it could theoretically bind VLDL and reduce the levels of both VLDL and LDL. Proof-of-principle studies in relevant animal models are needed to address this question. Another approach could be increasing the hepatic expression of apoE. Because apoE serves as a ligand for the LDL receptor, other related receptors, and heparan sulfate proteogycans in the liver, increased apoE expression could potentially target more VLDL and remnants for hepatic degradation, thus reducing VLDL and LDL levels. Again, proof-of-principle studies in relevant animal models are needed to address this question. Specifically, more data are needed on the effects of hepatic apoE gene transfer on lipoprotein metabolism in dyslipidemic animal models other than apoE deficiency.

Disorders associated primarily with low-HDL cholesterol

Lecithin:cholesterol acyltransferase deficiency

Unesterified cholesterol is esterified to cholesteryl ester in the blood by the lipoprotein-associated enzyme lecithin: cholesterol acyltransferase (LCAT). Complete LCAT deficiency is is characterized by markedly reduced HDL cholesterol levels (less than 10 mg/dl), corneal opacities, anemia, and progressive proteinuria and renal insufficiency eventually leading to end-stage renal disease [43]. Several different mutations in the LCAT gene have been described in patients with LCAT deficiency. LCAT knockout mice also have reduced HDL cholesterol levels [44], although the development of renal disease has not been reported. Because this disorder has no known therapy to prevent the progressive renal disease, it is a candidate for the development of gene therapy. Although LCAT is normally synthesized in the liver, because it is a secreted protein it could theoretically be made in other tissues such as muscle after gene transfer.

Tangier disease

Tangier disease is a rare genetic disorder associated with markedly reduced HDL cholesterol levels (less than 5 mg/dl), the accumulation of cholesterol in macrophages and related cells, neuropathy, and premature atherosclerosis [45]. It is caused by mutations in the ATP-binding cassette protein 1 (ABC1), a cellular protein that promotes the efflux of cellular cholesterol and phospholipids to nascent HDL and lipid-poor apolipoprotein A-I (apoA-I) [46]. ABC1 knockout mice also have markedly reduced HDL cholesterol levels and cholesterol storage in macrophages and macrophage-like cells [47]. A naturally occurring model of Tangier disease in chickens has been reported [48]. Gene transfer of ABC1 to macrophages or bone marrow stem cells would be likely to ameliorate the symptoms of this disorder, but no proof-of-principle studies have yet been reported in animals.

Importantly, heterozygotes for ABC1 mutations have significantly reduced HDL cholesterol levels (less than 10th percentile) [49] and are at increased risk for atherosclerotic vascular disease [50]. There are no drug therapies that raise HDL cholesterol levels in these patients. Macrophage-directed gene therapy with ABC1 could therefore be an approach to this disorder as well.

ApoA-I deficiency

ApoA-I is the major protein in HDL; a genetic deficiency of apoA-I results in markedly reduced HDL and seems, at least in some kindreds, to increase the risk for coronary artery disease [51]. ApoA-I knockout mice have extremely low levels of HDL cholesterol and although chow-fed mice do not develop atherosclerosis [52], the absence of apoA-I enhances the progression of atheroclerosis in hyperlipidemic mouse models [53,54]. The expression of human apoA-I in apoA-I knockout mice by using a first-generation adenovirus transiently restored HDL cholesterol levels to the normal range [55]. In apoA-I-deficient patients, gene transfer of apoA-I to liver (or possibly muscle) would be expected to increase HDL cholesterol levels and over time would reduce the risk for atherosclerotic disease. However, apoA-I-deficient patients are very rare, making this disease somewhat less attractive as a target for the development of gene therapy.

Other disorders of reduced HDL cholesterol

Reduced levels of HDL cholesterol are common, and most patients with low HDL do not have mutations in LCAT, ABC1, or apoA-I. However, it is possible that gene transfer to express any of these genes could nevertheless be therapeutically useful. For example, by converting unesterified to esterified cholesterol, LCAT is believed to facilitate the process of 'reverse cholesterol transport'. Transgenic over-expression of human LCAT increased HDL cholesterol levels in mice [56,57,58] and rabbits [59,60]. Administration of a first-generation adenovirus encoding human LCAT in human apoA-I transgenic mice resulted in a substantial increase in HDL cholesterol and human apoA-I levels [61]. Although transgenic LCAT overexpression increased atherosclerosis in cholesterol-fed mice [62], co-expression with cholesteryl ester transfer protein (CETP) reduced atherosclerosis in mice [63]. Furthermore, transgenic expression of LCAT in cholesterol-fed rabbits reduced atherosclerosis [64]. Thus, LCAT might be a candidate for gene therapy to inhibit atherosclerosis in humans. Similarly, gene therapy to augment ABC1 expression in macrophages might be expected to reduce atherosclerosis even in patients who do not have ABC1 mutations. Animal experiments are needed to test this hypothesis.

Perhaps the best evidence for this concept exists for apoA-I. A variety of evidence in animals indicates that intervention to increase the expression of apoA-I is likely to be associated with reduced atherosclerosis. Weekly intravenous injection of human HDL into cholesterol-fed rabbits resulted in the regression of atherosclerotic lesions [65] and regular intravenous injection of rabbit apoA-I into cholesterol-fed rabbits resulted in the reduced progression of atherosclerotic lesions [66]. Hepatic over-expression of human apoA-I in trangenic mice reduced atherosclerosis in C57BL/6 mice fed with a high-fat diet [67] and in apoE-deficient mice [68,69]. Transgenic over-expression of human apoA-I in the livers of hyperlipidemic WHHL rabbits reduced the development of aortic atherosclerosis [70]. Gene transfer of human apoA-I with a first-generation adenoviral vector in human apoA-I-transgenic/apoE-deficient mice reduced the progression of atherosclerosis [71]. Administration of a second-generation adenovirus resulted in the significant regression of pre-existing atherosclerotic lesions in mice deficient in low-density lipoprotein receptor that were fed with a Western diet [72]. These results indicate the feasibility of apoA-I gene transfer as a strategy for affecting atherosclerosis. ApoE is another candidate for gene transfer and overexpression as a strategy to reduce atherosclerosis [73].

Other disorders associated with defects in lipid and lipoprotein metabolism

Wolman disease and cholesteryl ester storage disease

Lysosomal acid lipase (LAL) is required for the hydrolysis of lipoprotein-derived cholesteryl esters and triglycerides in the lysosome. Wolman disease and cholesteryl ester storage disease are autosomal recessive disorders caused by mutations in LAL. Wolman disease is the rare and more severe form in which infants develop hepatosplenomegaly, steatorrhea, adrenal calcification, failure to thrive, and early death by the second year of life. Patients with cholesteryl ester storage disease have hepatomegaly and hyperlipidemia in childhood and often progress to fibrotic liver disease. The reason for the differences in phenotype might be related to the specific molecular defects underlying the LAL deficiency [74]. A natural rat model of this disorder [75] and the acid lipase knockout mouse [76] each have many of the pathologic features of the human disease. Correction of the LAL deficiency in fibroblasts obtained from Wolman disease patients by using a recombinant adenovirus reduced the lipid storage and increased cell growth [77]. LAL deficiency disorders are attractive for gene therapy because the restoration of a relatively small amount of LAL expression in the liver would be expected to ameliorate the disease.

Abetalipoproteinemia

The microsomal transfer protein (MTP) mediates the microsomal transport of lipids in the intestine and liver and is necessary for the normal formation and secretion of chylomicrons in the enterocyte and VLDL in the hepatocyte [78]. Abetalipoproteinemia is a rare autosomal recessive disease caused by mutations in MTP. Abetalipoproteinemia is characterized by progressive spinocerebellar degeneration and pigmented degenerative retinopathy, leading to severe ataxia, blindness, and eventually premature death. These symptoms are the result of defects in the absorption and transport of fat-soluble vitamins, especially vitamin E, which requires the VLDL-LDL system for adequate transport to peripheral tissues. Therapy with high-dose vitamin E and other fat-soluble vitamins can slow but not prevent the progression of these symptoms. Knockout of MTP is lethal during embryogenesis [79] but the MTP gene was deleted in the liver after birth, creating a mouse model of hepatic MTP deficiency [80]. A recombinant adenovirus was used to overexpress MTP in wild-type mice, resulting in increased hepatic VLDL production [81]. Abetalipoproteinemia is an attractive candidate for the development of gene therapy, because even a relatively low level of MTP expression in the liver would be expected to result in some VLDL production and therefore a substantial improvement in vitamin E transport and potentially slowing or halting the progression of neurological and retinal symtoms.

Issues relevant to the development of gene therapy for lipid disorders

Effective gene therapy for the lipid disorders described above will require stable gene expression. Recent progress in vector development has focused on the development of vectors with a potential for long-term expression. Recombinant adenoviral vectors elicit a cellular immune response to adenoviral proteins, limiting gene expression and posing safety issues. Helper-dependent adenoviruses are fully deleted of adenoviral genes, thus removing the trigger of the immune response. A single intravenous injection of helper-dependent adenovirus results in less hepatotoxicity and considerably longer hepatic expression than earlier-generation adenoviral vectors [82]. Neutralizing antibodies against adenoviral capsid proteins are also generated by the injection of recombinant adenoviruses, preventing successful re-administration [83]. Transient immunosuppression at the time of vector administration permits subsequent administration and repeat transgene expression in liver [84]. The use of different adenoviral serotypes might also provide a strategy for overcoming neutralizing antibodies [85].

Recombinant AAV is an attractive vector for gene transfer because, like the helper-dependent adenovirus, it does not contain sequences encoding viral proteins [86]. Gene expression in the liver by using AAV vectors is dependent on the promoter [87]. AAV was used to express human factor IX in liver at therapeutic concentrations providing the stable correction of hemophilia B in mice and dogs [88,89]. AAV was also used to express human factor IX in muscle to achieve therapeutic plasma concentrations in mice, dogs, and humans [28]. Although AAV is promising, higher levels of gene expression will probably be required for the therapy of most of the lipid disorders described above.

Conclusion

Lipid disorders are common and associated with serious consequences such as atherosclerotic vascular disease and others specific to individual disorders. Current approaches to the treatment of lipid disorders are effective in many but not all patients. New approaches to refractory hypercholesterolemia, severe hypertriglyceridemia, low levels of HDL cholesterol, and certain inherited disorders of intracellular lipid metabolism are needed. Somatic gene therapy is a viable approach to the therapy of several lipid disorders for which therapies are currently inadequate. The molecular etiology and pathophysiology of most of the candidate diseases are very well understood. Animal models exist for the diseases and in many cases preclinical proof-of-principle studies have already been performed. In contrast with many other situations, overexpression of the therapeutic gene is unlikely in most cases to have any deleterious consequences and thus regulated expression is not required. Finally, many of the same genes that could be used to treat genetic deficiency disorders are also attractive candidates for genetic overexpression in persons who do not have a specific gene deficiency disorder but who have other causes of severe hyperlipidemia or low-HDL cholesterol. There has been progress in the development of vectors with less immunogenicicity that provide long-term gene expression. The next decade is therefore likely to witness several clinical trials of gene therapy for lipid disorders.

References

  1. Grundy SM: Statin trials and goals of cholesterol-lowering therapy. Circulation. 1998, 97: 1436-1439.

    Article  CAS  PubMed  Google Scholar 

  2. Gotto AM: Cholesterol management in theory and practice. Circulation. 1997, 96: 4424-4430.

    Article  PubMed  Google Scholar 

  3. Hokanson JE, Austin MA: Plasma triglyceride level is a risk factor for cardiovascular disease independent of high-density lipoprotein cholesterol level: a meta-analysis of population-based prospective studies. J Cardiovasc Risk. 1996, 3: 213-219.

    CAS  PubMed  Google Scholar 

  4. Gordon DJ, Rifkind BM: High-density lipoproteins - the clinical implications of recent studies. N Engl J Med. 1989, 321: 1311-1316.

    Article  CAS  PubMed  Google Scholar 

  5. Genest J, Bard JM, Fruchart JC, Ordovas JM, Schaefer EJ: Familial hypoalphalipoproteinemia in premature coronary artery disease. Arterioscler Thromb. 1993, 13: 1728-1737.

    Article  PubMed  Google Scholar 

  6. Rader DJ: Pathophysiology and management of low high-density lipoprotein cholesterol. Am J Cardiol. 1999, 83: 22F-24F. 10.1016/S0002-9149(99)00210-6.

    Article  CAS  PubMed  Google Scholar 

  7. Hobbs HH, Brown MS, Goldstein JL: Molecular genetics of the LDL receptor gene in familial hypercholesterolemia. Hum Mut. 1992, 1: 445-446.

    Article  CAS  PubMed  Google Scholar 

  8. Gordon BR, Stein E, Jones P, Illingworth DR: Indications for low-density liproprotein apheresis. Am J Cardiol. 1994, 74: 1109-1112.

    Article  CAS  PubMed  Google Scholar 

  9. Chowdhury JR, Grossman M, Gupta S, Chowdhury NR, Baker J, Wilson JM: Long-term improvement of hypercholesterolemia after ex vivo gene therapy in LDLR-deficient rabbits. Science. 1991, 254: 1802-1805.

    Article  CAS  PubMed  Google Scholar 

  10. Kozarsky KF, McKinley DR, Austin LL, Raper SE, Stratford-Perricaudet LD, Wilson JM: In vivo correction of low density lipoprotein receptor deficiency in the Watanabe heritable hyperlipidemic rabbit with recombinant adenoviruses. J Biol Chem. 1994, 269: 13695-13702.

    CAS  PubMed  Google Scholar 

  11. Brown DR, Brousseau ME, Shamburek RD, Talley GD, Meyn S, Demosky SJJ, Santamarina-Fojo S, Brewer HBJ, Hoeg JM: Adenoviral delivery of low-density lipoprotein receptors to hyperlipidemic rabbits: receptor expression modulates high-density lipoproteins. Metabolism. 1996, 45: 1447-1457.

    Article  CAS  PubMed  Google Scholar 

  12. Ishibashi S, Brown M, Goldstein J, Gerard R, Hammer R, Herz J: Hypercholesterolemia in low density lipoprotein receptor knockout mice and its reversal by adenovirus-mediated gene delivery. J Clin Invest. 1993, 92: 883-893.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  13. Stein CS, Martins I, Davidson BL: Long-term reversal of hypercholesterolemia in low density lipoprotein receptor (LDLR)-deficient mice by adenovirus-mediated LDLR gene transfer combined with CD154 blockade. J Gene Med. 2000, 2: 41-51. 10.1002/(SICI)1521-2254(200001/02)2:1<41::AID-JGM79>3.3.CO;2-G.

    Article  CAS  PubMed  Google Scholar 

  14. Kozarsky K, Jooss K, Donahee M, Strauss JF, Wilson JM: Effective treatment of familial hypercholesterolemia in the mouse model using adenovirus-mediated transfer of the VLDL receptor gene. Nat Genet . 1996, 13: 54-62.

    Article  CAS  PubMed  Google Scholar 

  15. Chen SJ, Rader DJ, Tazelaar J, Kawashiri M, Gao G, Wilson JM: Prolonged correction of hyperlipidemia in mice with familial hypercholesterolemia using an adeno-associated viral vector expressing very-low-density lipoprotein receptor. Mol Ther. 2000, 2: 256-261. 10.1006/mthe.2000.0122.

    Article  CAS  PubMed  Google Scholar 

  16. Parise F, Simone L, Croce MA, Ghisellini M, Battini R, Borghi S, Tiozzo R, Ferrari S, Calandra S, Ferrari S: Construction and in vitro functional evaluation of a low-density lipoprotein receptor/transferrin fusion protein as a therapeutic tool for familial hypercholesterolemia. Hum Gene Ther. 1999, 10: 1219-1228. 10.1089/10430349950018201.

    Article  CAS  PubMed  Google Scholar 

  17. Grossman M, Rader DJ, Muller DWM, Kolansky DM, Kozarsky K, Clark BJ, Stein EA, Lupien PJ, Brewer HBJ, Raper SE, Wilson JM: A pilot study of ex vivo gene therapy for homozygous familial hypercholesterolaemia. Nat Med. 1995, 1: 1148-1154.

    Article  CAS  PubMed  Google Scholar 

  18. Fojo SS, Brewer HB: The familial hyperchylomicronemia syndrome. J Am Med Ass. 1991, 265: 904-908. 10.1001/jama.265.7.904.

    Article  Google Scholar 

  19. Weinstock PH, Bisgaier CL, Aalto-Setala K, Radner H, Ramakrishnan R, Levak-Frank S, Essenburg AD, Zechner R, Breslow JL: Severe hypertriglyceridemia, reduced high density lipoprotein, and neonatal death in lipoprotein lipase knockout mice. J Clin Invest. 1995, 96: 2555-2568.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  20. Levak-Frank S, Weinstock PH, Hayek T, Verdery R, Hofmann W, Ramakrishnan R, Sattler W, Breslow JL, Zechner R: Induced mutant mice expressing lipoprotein lipase exclusively in muscle have subnormal triglycerides yet reduced high density lipoprotein cholesterol levels in plasma. J Biol Chem. 1997, 272: 17182-17190. 10.1074/jbc.272.27.17182.

    Article  CAS  PubMed  Google Scholar 

  21. Ginzinger DG, Lewis ME, Ma Y, Jones BR, Liu G, Jones SD: A mutation in the lipoprotein lipase gene is the molecular basis of chylomicronemia in a colony of domestic cats. J Clin Invest. 1996, 97: 1257-1266.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  22. Christophersen B, Nordstoga K, Shen Y, Olivecrona T, Olivecrona G: Lipoprotein lipase deficiency with pancreatitis in mink: biochemical characterization and pathology. J Lipid Res. 1997, 38: 837-846.

    CAS  PubMed  Google Scholar 

  23. Liu G, Ashbourne Excoffon KJ, Wilson JE, McManus BM, Rogers QR, Miao L, Kastelein JJ, Lewis ME, Hayden MR: Phenotypic correction of feline lipoprotein lipase deficiency by adenoviral gene transfer. Hum Gene Ther. 2000, 11: 21-32. 10.1089/10430340050016120.

    Article  CAS  PubMed  Google Scholar 

  24. Kessler P, Podsakoff G, Chen X, McQuiston S, Colosi P, Matelis L, Kurtzman G, Byrne BJ: Gene delivery to skeletal muscle results in sustained expression and systemic delivery of a therapeutic protein. Proc Natl Acad Sci USA. 1996, 93: 14082-14087. 10.1073/pnas.93.24.14082.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  25. Herzog R, Hagstrom JN, Kung S, Tai S, Wilson JM, Fisher K, High KA: Stable gene transfer and expression of human blood coagulation factor IX after intramuscular injection of recombinant adeno-associated virus. Proc Natl Acad Sci USA. 1997, 94: 5804-5809. 10.1073/pnas.94.11.5804.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  26. Murphy J, Zhou S, Giese K, Williams L, Escobedo J, Dwarki V: Long-term correction of obesity and diabetes in genetically obese mice by a single intramuscular injection of recombinant adeno-associated virus encoding mouse leptin. Proc Natl Acad Sci USA. 1997, 94: 13921-13926. 10.1073/pnas.94.25.13921.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  27. Herzog RW, Yang EY, Couto LB, Hagstrom JN, Elwell D, Fields PA, Burton M, Bellinger DA, Read MS, Brinkhous KM, Podsakoff GM, Nichols TC, Kurtzman GJ, High KA: Long-term correction of canine hemophilia B by gene transfer of blood coagulation factor IX mediated by adeno-associated viral vector. Nat Med. 1999, 5: 56-63. 10.1038/4743.

    Article  CAS  PubMed  Google Scholar 

  28. Kay MA, Manno CS, Ragni MV, Larson PJ, Couto LB, McClelland A, Glader B, Chew AJ, Tai SJ, Herzog RW, Arruda V, Johnson F, Scallan C, Skarsgard E, Flake AW, High KA: Evidence for gene transfer and expression of factor IX in haemophilia B patients treated with an AAV vector. Nat Genet. 2000, 24: 257-261. 10.1038/73464.

    Article  CAS  PubMed  Google Scholar 

  29. Ashbourne-Excoffon KJD, Liu G, Miao L, Wilson JE, McManus B, Semenkovich CF, Coleman T, Benoit P, Duverger N, Branellec D, Denefle P, Hayden MR, Lewis MES: Correction of hypertriglyceridemia and impaired fat tolerance in lipoprotein lipase deficient mice by adenovirus-mediated expression of human lipoprotein lipase. Arterioscler Thromb Vasc Biol. 1997, 17: 2532-2539.

    Article  Google Scholar 

  30. Zsigmond E, Kobayashi K, Tzung KW, Li L, Fuke Y, Chan L: Adenovirus-mediated gene transfer of human lipoprotein lipase ameliorates the hyperlipidemias associated with apolipoprotein E and LDL receptor deficiencies in mice. Hum Gene Ther. 1997, 8: 1921-1933.

    Article  CAS  PubMed  Google Scholar 

  31. Mahley RW: Apolipoprotein E: cholesterol transport protein with expanding role in cell biology. Science. 1988, 240: 622-630.

    Article  CAS  PubMed  Google Scholar 

  32. Schaefer EJ, Gregg RE, Ghiselli G, Forte TM, Ordovas JM, Zech LA, Brewer HB: Familial apolipoprotein E deficiency. J Clin Invest. 1986, 78: 1206-1219.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  33. Breslow JL: Mouse models of atherosclerosis. Science . 1996, 272: 685-688.

    Article  CAS  PubMed  Google Scholar 

  34. Stevenson S, Marshall-Neff J, Teng B, Lee C, Roy S, McClelland A: Phenotypic correction of hypercholesterolemia in apoE-deficient mice by adenovirus-mediated in vivo gene transfer. Arterioscler Thromb Vasc Biol. 1995, 15: 479-484.

    Article  CAS  PubMed  Google Scholar 

  35. Kashyap VS, Santamarina-Fojo S, Brown DR, Parrott CL, Applebaum-Bowden D, Meyn S, Talley G, Paigen B, Maeda N, Brewer HB: Apolipoprotein E deficiency in mice: gene replacement and prevention of atherosclerosis using adenovirus vectors. J Clin Invest. 1995, 96: 1612-1620.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  36. Tsukamoto K, Smith P, Glick JM, Rader D: Liver-directed gene transfer and prolonged expression of three major human apoE isoforms in apoE-deficient mice. J Clin Invest. 1997, 100: 107-114.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  37. Tsukamoto K, Tangirala R, Chun SH, Puré E, Rader DJ: Rapid regression of atherosclerosis induced by liver-directed gene transfer of apolipoprotein E in apoE-deficient mice. Arterioscler Thromb Vasc Biol. 1999, 19: 2162-2170.

    Article  CAS  PubMed  Google Scholar 

  38. Desurmont C, Caillaud JM, Emmanuel F, Benoit P, Fruchart JC, Castro G, Branellec D, Heard JM, Duverger N: Complete atherosclerosis regression after human ApoE gene transfer in ApoE-deficient/ nude mice. Arterioscler Thromb Vasc Biol. 2000, 20: 435-442.

    Article  CAS  PubMed  Google Scholar 

  39. Linton MF, Atkinson JB, Fazio S: Prevention of atherosclerosis in apolipoprotein E-deficient mice by bone marrow transplantation. Science. 1995, 267: 1034-1037.

    Article  CAS  PubMed  Google Scholar 

  40. Mahley RW, Huang Y, Rall SC: Pathogenesis of type III hyper-lipoproteinemia (dysbetalipoproteinemia): questions, quandaries, and paradoxes. J Lipid Res. 1999, 40: 1933-1949.

    CAS  PubMed  Google Scholar 

  41. Grundy SM, Chait A, Brunzell JD: Familial combined hyperlipidemia workshop. Arteriosclerosis. 1987, 7: 203-207.

    Google Scholar 

  42. Abbate SL, Brunzell JD: Pathophysiology of hyperlipidemia in diabetes mellitus. J Cardiovasc Pharm Ther. 1990, 16: S1-S7.

    Article  Google Scholar 

  43. Kuivenhoven JA, Pritchard H, Hill J, Frohlich J, Assmann G, Kastelein J: The molecular pathology of lecithin: cholesterol acyltransferase (LCAT) deficiency syndromes. J Lipid Res. 1997, 38: 191-205.

    CAS  PubMed  Google Scholar 

  44. Ng DS, Francone OL, Forte TM, Zhang J, Haghpassand M, Rubin EM: Disruption of the murine lecithin: cholesterol acyltransferase gene causes impairment of adrenal lipid delivery and up-regulation of scavenger receptor class B type I. J Biol Chem. 1997, 272: 15777-15781. 10.1074/jbc.272.25.15777.

    Article  CAS  PubMed  Google Scholar 

  45. Hobbs HH, Rader DJ: ABC1: connecting yellow tonsils, neuropathy, and very low HDL. J Clin Invest. 1999, 104: 1015-1017.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  46. Young SG, Fielding CJ: The ABCs of cholesterol efflux. Nat Genet. 1999, 22: 316-318. 10.1038/11878.

    Article  CAS  PubMed  Google Scholar 

  47. McNeish J, Aiello RJ, Guyot D, Turi T, Gabel C, Aldinger C, Hoppe KL, Roach ML, Royer LJ, de Wet J, Broccardo C, Chimini G, Francone OL: High density lipoprotein deficiency and foam cell accumulation in mice with targeted disruption of ATP-binding cassette transporter-1. Proc Natl Acad Sci USA. 2000, 97: 4245-4250. 10.1073/pnas.97.8.4245.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  48. Schreyer SA, Hart LK, Attie AD: Hypercatabolism of lipoprotein-free apolipoprotein A-I in HDL-deficient mutant chickens. Arterioscler Thromb. 1994, 14: 2053-2059.

    Article  CAS  PubMed  Google Scholar 

  49. Wilson AB, Marcil M, Clee SM, Zhang LH, Roomp K, van Dam M, Yu L, Collins JA, Molhuizen HOF, Loubser O, Ouelette BFF, Fichter K, Ashbourne-Excoffon KJD, Sensen CW, Scherer S, Mott S, Denis M, Martindale D, Frohlich J, Morgan K, Koop B, Pimstone S, Kastelein JJP, Genest J, Hayden MR: Mutations in ABC1 in Tangier disease and familial high-density lipoprotein deficiency. Nat Genet. 1999, 22: 336-345. 10.1038/11905.

    Article  Google Scholar 

  50. Schaefer EJ, Zech LA, Schwartz DE, Brewer HBJ: Coronary heart disease prevalence and other clinical features in familial high-density lipoprotein deficiency (Tangier disease). Ann Intern Med. 1980, 93: 261-266.

    Article  CAS  PubMed  Google Scholar 

  51. Funke H: Genetic determinants of high density lipoprotein levels. Curr Opin Lipidol. 1997, 8: 189-196.

    Article  CAS  PubMed  Google Scholar 

  52. Li H, Reddick R, Maeda N: Lack of apoA-I is not associated with increased susceptibility to atherosclerosis in mice. Arterioscler Thromb. 1993, 13: 1814-1821.

    Article  CAS  PubMed  Google Scholar 

  53. Hughes SD, Verstuyft J, Rubin EM: HDL deficiency in genetically engineered mice requires elevated LDL to accelerate atherogenesis. Arterioscler Thromb Vasc Biol. 1997, 17: 1725-1729.

    Article  CAS  PubMed  Google Scholar 

  54. Voyiaziakis E, Goldberg IJ, Plump AS, Rubin EM, Breslow JL, Huang LS: ApoA-I deficiency causes both hypertriglyceridemia and increased atherosclerosis in human apoB transgenic mice. J Lipid Res. 1998, 39: 313-321.

    CAS  PubMed  Google Scholar 

  55. Webb NR, de Beer MC, van der Wethuyzen DR, Kindy MS, Banka CL, Tsukamoto K, Rader DJ, de Beer FC: Adenoviral vector-mediated overexpression of serum amyloid A in apoA-I-deficient mice. J Lipid Res. 1997, 38: 1583-1590.

    CAS  PubMed  Google Scholar 

  56. Mehlum A, Staels B, Duverger N, Tailleux A, Castro G, Fievet C, Luc G, Fruchart JC, Olivecrona G, Skretting G: Tissue-specific expression of the human gene for lecithin: cholesterol acyltransferase in transgenic mice alters blood lipids, lipoproteins and lipases towards a less atherogenic profile. Eur J Biochem. 1995, 230: 567-575.

    Article  CAS  PubMed  Google Scholar 

  57. Francone OL, Haghpassand M, Bennett JA, Royer L, McNeish J: Expression of human lecithin: cholesterol acyltransferase in transgenic mice: effects on cholesterol efflux, esterification, and transport. J Lipid Res. 1997, 38: 813-822.

    CAS  PubMed  Google Scholar 

  58. Vaisman BL, Klein HG, Rouis M, Berard AM, Kindt MR, Talley GD, Meyn SM, Hoyt RFJ, Marcovina SM, Albers JJ: Overexpression of human lecithin cholesterol acyltransferase leads to hyperalphalipoproteinemia in transgenic mice. J Biol Chem. 1995, 270: 12269-12275. 10.1074/jbc.270.20.12269.

    Article  CAS  PubMed  Google Scholar 

  59. Hoeg JM, Vaisman BL, Demosky SJ, Meyn SM, Talley GD, Hoyt RF, Feldman S, Berard AM, Sakai N, Wood D, Brousseau ME, Marcovina S, Brewer HB, Santamarina-Fojo S: Lecithin:cholesterol acyltransferase overexpression generates hyperalpha-lipoproteinemia and a nonatherogenic lipoprotein pattern in transgenic rabbits. J Biol Chem. 1996, 271: 4396-4402. 10.1074/jbc.271.8.4396.

    Article  CAS  PubMed  Google Scholar 

  60. Brousseau ME, Wang J, Demosky SJ, Vaisman BL, Talley GD, Santamarina-Fojo S, Brewer HB, Hoeg JM: Correction of hypoalphalipoproteinemia in LDL receptor-deficient rabbits by lecithin: cholesterol acyltransferase. J Lipid Res. 1998, 39: 1558-1567.

    CAS  PubMed  Google Scholar 

  61. Seguret-Mace S, Latta-Mahieu M, Castro G, Luc G, Fruchart JC, Rubin E, Denefle P, Duverger N: Potential gene therapy for lecithin-cholesterol acyltransferase (LCAT)-deficient and hypoalphalipoproteinemic patients with adenovirus-mediated transfer of human LCAT gene. Circulation. 1996, 94: 2177-2184.

    Article  CAS  PubMed  Google Scholar 

  62. Berard AM, Foger B, Remaley A, Vaisman BL, Talley G, Paigen B, Hoyt RF, Brewer HB, Santamarina-Fojo S: High plasma HDL concentration associated with enhanced atherosclerosis in transgenic mice overexpressing lecithin-cholesteryl acyltransferase. Nat Med. 1997, 3: 744-749.

    Article  CAS  PubMed  Google Scholar 

  63. Foger B, Chase M, Amar MJ, Vaisman BL, Shamburek RD, Paigen B, Fruchart-Najib J, Paiz JA, Koch CA, Hoyt RF, Brewer HB, Santamarina-Fojo S: Cholesteryl ester transfer protein corrects dysfunctional high density lipoproteins and reduces aortic atherosclerosis in lecithin cholesterol acyltransferase transgenic mice. J Biol Chem. 1999, 274: 36912-36920. 10.1074/jbc.274.52.36912.

    Article  CAS  PubMed  Google Scholar 

  64. Hoeg JM, Santamarina-Fojo S, Berard A, Cornhill JF, Herderick E, Feldman S, Haudenschild C, Vaisman B, Hoyt RF, Demosky S, Kauffman R, Hazel C, Marcovina S, Brewer HB: Overexpression of lecithin:cholesterol acyltransferase in transgenic rabbits prevents diet-induced atherosclerosis. Proc Natl Acad Sci USA. 1996, 93: 11448-11453. 10.1073/pnas.93.21.11448.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  65. Badimon JJ, Badimon L, Fuster V: Regression of atherosclerotic lesions by high density lipoprotein plasma fraction in the cholesterol-fed rabbit. J Clin Invest. 1990, 85: 1234-1243.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  66. Miyazaki A, Sakuma S, Morikawa W, Takiue T, Miake F, Terano T, Sakai M, Hakamata H, Sakamoto Y, Naito M, Ruan Y, Takahashi K, Ohta T, Horiuchi S: Intravenous injection of rabbot apolipoprotein A-I inhibits the progression of atherosclerosis in cholesterol-fed rabbits. Arterioscler Thromb Vasc Biol. 1995, 15: 1882-1888.

    Article  CAS  PubMed  Google Scholar 

  67. Rubin E, Krauss R, Spangler E, Verstuyft J, Clift S: Inhibition of early atherogenesis in transgenic mice by human apolipoprotein AI. Nature. 1991, 353: 265-267. 10.1038/353265a0.

    Article  CAS  PubMed  Google Scholar 

  68. Plump A, Scott C, Breslow J: Human apolipoprotein A-I gene expression increases high density lipoprotein and suppresses atherosclerosis in the apolipoprotein E-deficient mouse. Proc Natl Acad Sci USA . 1994, 91: 9607-9611.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  69. Paszty C, Maeda N, Verstuyft J, Rubin EM: Apolipoprotein AI trans-gene corrects apolipoprotein E deficiency-induced atherosclerosis in mice. J Clin Invest. 1994, 94: 899-903.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  70. Duverger N, Kruth H, Emmanuel F, Caillaud J, Viglietta C, Castro G, Tailleux A, Fievet C, Fruchart J, Houdebine LM, Denefle P: Inhibition of atherosclerosis development in cholesterol-fed human apolipoprotein A-I-transgenic rabbits. Circulation. 1996, 94: 713-717.

    Article  CAS  PubMed  Google Scholar 

  71. Benoit P, Emmanuel F, Caillaud JM, Bassinet L, Castro G, Gallix P, Fruchart JC, Branellec D, Denefle P, Duverger N: Somatic gene transfer of human apoA-I inhibits atherosclerosis progression in mouse models. Circulation. 1999, 99: 105-110.

    Article  CAS  PubMed  Google Scholar 

  72. Tangirala RK, Tsukamoto K, Chun SH, Usher D, Puré E, Rader DJ: Regression of atherosclerosis induced by liver-directed gene transfer of apolipoprotein A-I in mice. Circulation. 1999, 100: 1816-1822.

    Article  CAS  PubMed  Google Scholar 

  73. Rader DJ: Gene therapy for atherosclerosis. Curr Res Mol Therapeut. 1998, 1: 680-689.

    CAS  Google Scholar 

  74. Lohse P, Maas S, Sewell AC, van Diggelen OP, Seidel D: Molecular defects underlying Wolman disease appear to be more heterogeneous than those resulting in cholesteryl ester storage disease. J Lipid Res. 1999, 40: 221-228.

    CAS  PubMed  Google Scholar 

  75. Yoshida Y, Kuriyama M: Genetic lipid storage disease with lysosomal acid lipase deficiency in rats. Lab Anim Sci. 1990, 40: 486-489.

    CAS  PubMed  Google Scholar 

  76. Du H, Duanmu M, Witte D, Grabowski GA: Targeted disruption of the mouse lysosomal acid lipase gene: long-term survival with massive cholesteryl ester and triglyceride storage. Hum Mol Genet. 1998, 7: 1347-1354. 10.1093/hmg/7.9.1347.

    Article  CAS  PubMed  Google Scholar 

  77. Tietge UJ, Sun G, Czarnecki S, Yu QC, Lohse P, Du H, Grabowski GA, Glick JM, Rader DJ: Phenotypic correction of lipid storage and growth arrest in Wolman disease fibroblasts by gene transfer of lysosomal acid lipase. Hum Gene Ther. 2000,

    Google Scholar 

  78. Gordon DA: Recent advances in elucidating the role of the microsomal triglyceride transfer protein in apolipoprotein B lipoprotein assembly. Curr Opin Lipidol. 1997, 8: 131-137.

    Article  CAS  PubMed  Google Scholar 

  79. Raabe M, Flynn LM, Zlot CH, Wong JS, Veniant MM, Hamilton RL, Young SG: Knockout of the abetalipoproteinemia gene in mice: reduced lipoprotein secretion in heterozygotes and embryonic lethality in homozygotes. Proc Natl Acad Sci USA. 1998, 95: 8686-8691. 10.1073/pnas.95.15.8686.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  80. Chang BH, Liao W, Li L, Nakamuta M, Mack D, Chan L: Liver-specific inactivation of the abetalipoproteinemia gene completely abrogates very low density lipoprotein/low density lipoprotein production in a viable conditional knockout mouse. J Biol Chem . 1999, 274: 6051-6055. 10.1074/jbc.274.10.6051.

    Article  CAS  PubMed  Google Scholar 

  81. Tietge UJF, Bakillah A, Maugeais C, Tsukamoto K, Hussain MM, Rader DJ: Hepatic overexpression of microsomal triglyceride transfer protein (MTP) results in increased in vivo secretion of VLDL triglycerides and apolipoprotein B. J Lipid Res. 1999, 40: 2134-2139.

    CAS  PubMed  Google Scholar 

  82. Morsy MA, Gu M, Motzel S, Zhao J, Lin J, Su Q, Allen H, Franlin L, Parks RJ, Graham FL, Kochanek S, Bett AJ, Caskey CT: An adenoviral vector deleted for all viral coding sequences results in enhanced safety and extended expression of a leptin transgene. Proc Natl Acad Sci USA . 1998, 95: 7866-7871. 10.1073/pnas.95.14.7866.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  83. Chirmule N, Xiao W, Truneh A, Schnell MA, Hughes JV, Zoltick P, Wilson JM: Humoral immunity to adeno-associated virus type 2 vectors following administration to murine and nonhuman primate muscle. J Virol. 2000, 74: 2420-2425. 10.1128/JVI.74.5.2420-2425.2000.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  84. Ilan Y, Jona VK, Sengupta K, Davidson A, Horwitz MS, Roy-Chowdhury N, Roy-Chowdhury J: Transient immunosuppression with FK506 permits long-term expression of therapeutic genes introduced into the liver using recombinant adenoviruses in the rat. Hepatology. 1997, 26: 949-956.

    Article  CAS  PubMed  Google Scholar 

  85. Mack CA, Song WR, Carpenter H, Wickham TJ, Kovesdi I, Harvey BG, Magovern CJ, Isom OW, Rosengart T, Falck-Pedersen E, Hackett NR, Crystal RG, Mastrangeli A: Circumvention of anti-adenovirus neutralizing immunity by administration of an adenoviral vector of an alternate serotype. Hum Gene Ther. 1997, 8: 99-109.

    Article  CAS  PubMed  Google Scholar 

  86. Kotin RM: Prospects for the use of adeno-associated virus as a vector for human gene therapy. Hum Gene Ther. 1994, 5: 793-801.

    Article  CAS  PubMed  Google Scholar 

  87. Xiao W, Berta SC, Lu MM, Moscioni AD, Tazelaar J, Wilson JM: Adeno-associated virus as a vector for liver-directed gene therapy. J Virol. 1998, 72: 10222-10226.

    PubMed Central  CAS  PubMed  Google Scholar 

  88. Snyder RO, Miao C, Meuse L, Tubb J, Donahue BA, Lin HF, Stafford DW, Patel S, Thompson AR, Nichols T, Read MS, Bellinger DA, Brinkhous KM, Kay MA: Correction of hemophilia B in canine and murine models using recombinant adeno-associated viral vectors. Nat Med. 1999, 5: 64-70. 10.1038/4751.

    Article  CAS  PubMed  Google Scholar 

  89. Wang L, Nichols TC, Read MS, Bellinger DA, Verma IM: Sustained expression of therapeutic level of factor IX in hemophilia B dogs by AAV-mediated gene therapy in liver. Mol Ther. 2000, 1: 154-158. 10.1006/mthe.2000.0031.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Daniel J Rader.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kawashiri, Ma., Rader, D.J. Gene therapy for lipid disorders. Trials 1, 120 (2000). https://0-doi-org.brum.beds.ac.uk/10.1186/cvm-1-2-120

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/cvm-1-2-120

Keywords