Nutr Res Pract. 2007 Winter;1(4):247-253. English.
Published online Dec 31, 2007.
©2007 The Korean Nutrition Society and the Korean Society of Community Nutrition
other

Alpha-Tocopherol Transfer Protein (α-TTP): Insights from Alpha-Tocopherol Transfer Protein Knockout Mice

Yunsook Lim,1 and Maret G. Traber2
    • 1Department of Food and Nutrition, Kyung Hee University, Seoul 130-701, Korea.
    • 2Department of Nutrition and Food Management, Linus Pauling Institute, Oregon State University, Corvallis, OR 97330, USA.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Alpha-tocopherol transfer protein (α-TTP) is a liver cytosolic transport protein that faciliates α-tocopherol (α-T) transfer into liver secreted plasma lipoproteins. Genetic defects in α-TTP, like dietary vitamin E deficiency, are associated with infertility, muscular weakness and neurological disorders. Both human and α-TTP deficient (α-TTP-/-) mice exhibit severe plasma and tissue vitamin E deficiency that can be attenuated by sufficient dietary α-T supplementations. In this review, we summarize the literature concerning studies utilizing the α-TTP-/- mice. Levels of vitamin E in the α-TTP-/- mice do not appear to be directly related to the amounts of dietary α-T or to the levels of α-TTP protein in tissues. The α-TTP-/- mice appear to present a good model for investigating the specific role of α-T in tissue vitamin E metabolism. Furthermore, α-TTP-/- mice appear to be useful to elucidate functions of α-TTP beyond its well recognized functions of transferring α-T from liver to plasma lipoprotein fractions.

Keywords
α-tocopherol; α-tocopherol transfer protein; vitamin E

Introduction

Vitamin E is a generic term which describes a group of lipophilic chain breaking antioxidants in biological membranes and includes 4 tocopherols (α-, β-, γ- and δ-) and 4 tocotrienols (α-, β-, γ- and δ-) (Traber, 1999). Vitamin E plays an important role in reproduction and in maintaining the structural integrity of the cells (Evans & Bishop, 1922; Packer, 1991). α-tocopherol (α-T) is the dominant isoform found in human and animals. Indeed, in the 2000 Dietary Reference Intakes, only α-T was recommended as meeting human dietary vitamin E requirements (Food and Nutrition Board & Institute of Medicine, 2000). α-T functions additionally beyond its well recognized antioxidant role of smooth muscle cells. For example, α-T inhibits proliferation via a protein kinase C-dependent mechanism (Boscoboinik et al., 1991a; Boscoboinik et al., 1991b; Ricciarelli et al., 1998). However, α-T functions in most tissues remain to be fully clarified.

In humans, α-T deficiency occurs as a result of genetic defects in the hepatic α-tocopherol transfer protein (α-TTP) and causes a rare neurological disease, ataxia with vitamin E deficiency (AVED) (Ouahchi et al., 1995). Subsequently, α-TTP knock out (α-TTP-/-) mice were shown to exhibit many of the same phenotypes as humans with AVED (Yokota et al., 2001). In the human disease the neurologic abnormalities are a result of the α-T deficiency as neurologic manifestations can be ameliorated by a greatly augmented oral vitamin E intake. However, α-TTP functions remain to be completely clarified, especially in the few non-hepatic tissues where it has been detected. Although α-T enrichment of plasma is mediated by liver α-TTP, the α-TTP protein may also modulate tissue α-T levels (Arita et al., 1995; Traber & Arai, 1999). The molecular mechanisms of how α-T levels are regulated in different tissues and the extent to which α-TTP interrelate to these mechanisms, likewise remains incompletely understood. The following review will provide a summary of general characteristics and gene profiles of α-TTP-/- mice, and regulation of plasma and tissue levels of α-T manipulated by dietary α-T in α-TTP-/- mice.

α-T transfer protein

α-TTP is a 32 kDa cytosolic lipid binding protein known as a member of the cis-retinal binding motif sequence (CRAL-TRIO) protein family (Panagabko et al., 2003). α-TTP was first described in 1975 by Catignani et al. (1975) as a hepatic α-T binding protein. α-TTP is expressed primarily in liver, but has been detected in various tissues including lung, spleen, kidney, brain, adrenals, uterus (pregnant rodent) and placenta (Copp et al., 1999; Gohil et al., 2004; Hosomi et al., 1998; Jishage et al., 2001; Kaempf-Rotzoll et al., 2003; Kaempf-Rotzoll et al., 2002).

All forms of dietary vitamin E are absorbed and transported via chylomicrons through lymph into the circulation (Arita et al., 1995). Unlike non-specific absorption of vitamin E, α-TTP mediates the selective transport of α-T in its RRR- or 2R-forms into nascent very low density lipoproteins (VLDL) and is responsible for secretion of α-T from the liver into blood to maintain plasma α-T concentrations (Arita et al., 1995).

Liver α-TTP has been isolated and its cDNA sequences reported from a variety of species. The human gene has been localized to the 8q13.1-13.3 region of chromosome 8 (Arita et al., 1995; Doerflinger et al., 1995). α-TTP has been crystallized and the α-T binding pocket identified (Meier et al., 2003; Min et al., 2003). α-TTP has a hinge and a cover that closes entrapping α-T in the binding pocket that causes α-T to fold such that the 2-position is critical for its fit (Meier et al., 2003; Min et al., 2003).

α-TTP has ligand specificity and relative affinities towards different tocopherols (α-> β-> γ-> δ-T) in vitro (Hosomi et al., 1997). Tocopherol binding affinity and transfer activity of α-TTP and ligand selectivity discriminating physiological factor for α-T retention were also demonstrated in vitro models (Morley et al., 2004; Panagabko et al., 2003; Panagabko et al., 2002). However, binding affinity of α-TTP to α-T and roles of α-TTP beyond incorporation of α-T to very low density lipoproteins (VLDL) were not demonstrated in various tissues from in vivo models.

A recent study has shown that high concentration of hepatic α-tocopherol caused by high α-T injection increased hepatic α-T concentrations but didn't induce either hepatic α-TTP mRNA and protein levels in rats (Mustacich et al., 2006). The result suggests that hepatic α-T levels doesn't regulate α-TTP gene expression.

To elucidate the importance of vitamin E in vivo, α-TTP gene deletion leading to vitamin E deficiency was required due to difficulty of dietary depletion of vitamin E in humans and animals (Cuddihy et al., 2004). α-TTP-/- mice were generated by targeted disruption of α-TTP (Jishage et al., 2001; Terasawa et al., 2000).

Infertility in females with α-T deficiency

Vitamin E was first recognized as a "reproductive factor" in 1922 by Evans and Bishop (Evans and Bishop, 1922). Impaired fertility, such as resorption of the fetus, was reported in female α-TTP-/- mice (Jishage et al., 2001; Terasawa et al., 2000) while no signs of male infertility were detected (Terasawa et al., 2000). In pregnant α-TTP-/- mice, placental failure was effectively abrogated by dietary α-T (567 mg/kg diet) or a synthetic antioxidant (BO-653) (Jishage et al., 2001). Female rats with vitamin E deficiency demonstrated structural changes of ovary and uterus and abnormal estrus cycle (Das and Chowdhury, 1999). α-T levels increased in the placental membrane as gestation progressed during feto-placental development in normal humans and rodents (Jishage et al., 2001; Qanungo et al., 1999; Sen & Mukherjea, 1998). Moreover, α-TTP was not expressed in the placenta but in the uterus to supply α-T for placenta and fetus during the development of placental labyrinthine tropoblasts in mice (Jishage et al., 2001). These results suggest that α-TTP is a critical fertility factor to provide enough α-T for the maintenance of a regular estrous cycle and protection of placenta and fetus during pregnancy. It should be noted that mice and humans have very different placental structures and that α-TTP has been detected in human placenta (Muller-Schmehl et al., 2004).

Neurological disorders in α-T deficiency

Vitamin E deficiency is associated with neurological abnormalities in humans and animals suggesting α-T's importance in the central nervous systems. The α-TTP mRNA expression was demonstrated in cerebral cortex, especially Purkinje cells and in cerebellum (Hosomi et al., 1998) possibly to protect the brain from oxidative stress by providing α-T (Meier et al., 2003). AVED patients with genetic defects of α-TTP had very low or undetectable plasma α-T levels and progressive spinocerebellar dysfunctions (Hoshino et al., 1999; Hosomi et al., 1998; Ouahchi et al., 1995; Yokota et al., 1997; Yokota et al., 2000).

Although specific discrimination of α-T by α-TTP was shown in in vitro models (Hosomi et al., 1997; Panagabko et al., 2003; Panagabko et al., 2002) some human α-TTP mutants showed α-T transfer activity similar to that of wild type protein suggesting that α-TTP may have additional physiological functions in vivo (Morley et al., 2004). Therefore, we should consider the importance of an in vivo animal model to investigate effects of α-T in AVED and other possible genetic disorders in inbreeding mice as well.

Gene profiles related to neurodegeneration

α-TTP-/- mice showed many features of neurological diseases as seen in humans with AVED. Specifically, the mice with ataxia had very low plasma and brain α-T levels (Yokota et al., 2001). Gene profile studies in α-TTP-/- cortex identified putative genes affected by genetic deletion of α-TTP and may account for neurological disorders (Gohil et al., 2004; Gohil et al., 2003). These studies showed down-regulation in α-TTP-/- cortex of genes related to neurogenesis and increased expression of genes related to neurodegeneration. The authors suggested that some of the affected genes may be regulated by retinoic acid receptor related orphan receptor α. In female α-TTP-/- cerebral cortex, selective genes, such as myelin associated oligodendrocyte specific protein and proteolipid protein, associated with synaptogenesis and myelination were repressed. Induction of neurodegenerative genes, such as synulein-α, repression of synulein-β and induction of mitochondrial proapoptotic factor, cytochrome c, were also shown in α-TTP-/- cerebral cortex (Gohil et al., 2004; Gohil et al., 2003). Although apparent neurological signs were not reported in 3-4 months old mice (Gohil et al., 2003; Jishage et al., 2001; Terasawa et al., 2000; Yokota et al., 2001), neurological behaviors such as balance, coordination, strength, exploratory behavior and anxiety were affected in α-TTP-/- mice fed a α-T deficient or a normal diet up to at age of 6 months (Gohil et al., 2003; Jishage et al., 2001; Terasawa et al., 2000; Yokota et al., 2001). Learning and memory skills were significantly decreased in old female α-TTP-/- mice at age of 12 months old (Gohil et al., 2004). However, by the age of 18 months α-TTP-/- mice fed normal α-T diets developed abnormalities gradually while those fed deficient diets had obvious ataxia (Yokota et al., 2001). These differences in response along with the slow progression of abnormalities might be due to different dietary α-T concentrations and/or the duration of feeding, as well as different genetic backgrounds, ages and genders of mice (Table 1). The results suggest that the combination of genetic α-TTP defect (or deficiency) and dietary α-T deficiency would exacerbate neurodegeneration caused by prolonged α-T deficiency. The neurological abnormalities were prevented or attenuated when high dietary α-T was supplemented to α-TTP-/- mice, as well as to patients with AVED (Mariotti et al., 2004; Schuelke et al., 2000; Yokota et al., 2001; Yokota et al., 1997). Recent studies in phospholipid transfer protein (PLTP) deficient mice having brain α-T concentrations at higher levels than in apparently normal α-TTP -/-mice demonstrated vitamin E deficiency-related anxiety but did not show any evidence of neurological disorders, including neuromotor coordination (Desrumaux et al., 2005). It is likely that there will be tissue specific responses that are also dependent on α-T concentrations in various brain region.

Table 1
Relationship of plasma (µmol/l) and tissue α-T (nmol/g) and dietary α-T in α-TTP -/- mice

Gene profiles related to vasculogenesis, signal transduction pathways and cell proliferation

The gene profile studies also suggested that deletion of α-TTP leading to α-T deficiency causes tissue specific change of genes associated with vasculogenesis, signal transduction pathways and cell proliferation (Gohil et al., 2003; Vasu et al., 2007). In α-TTP-/- liver, changes in genes associated with neovascularization including quaking, NMDA receptor regulated gene 1 and hyalurinidase I were found. In addition, cell proliferation pathways such as growth factor binding proteins, mitogen activated protein kinase kinase 3 and apoptosis inhibitor 6 were activated in α-TTP-/- liver (Gohil et al., 2003). Moreover, genes related to signal transduction such as casein kinase 1, doublecortin and calcium/calmodulin-dependent protein kinase-like 1 were downregulated in heart gene profile study of α-TTP-/- mice (Vasu et al., 2007).

Taken together, the gene profile studies demonstrated that α-TTP-/- mice are a useful model to find target genes related to neurological disorders or neurodegeneration which could be prevented or attenuated by dietary vitamin E treatment and vasculogenesis, signal transduction pathways and cell proliferation. Further gene profile studies identifying gender differences in different feeding regimen (time and dose) at different age and verifying reversibility of the neurological symptoms are recommended for future application of dietary α-T in patients with AVED.

Oxidative stress, inflammatory and immune response related markers

Vitamin E is a widely accepted antioxidant and a potent anti-inflammatory agent. α-TTP mRNA level was decreased in rat liver exposed to hypoxia (Ban et al., 2002). α-TTP deletion increased levels of lipid peroxidation measured by thiobarbituric acid reactive substances (TBARS) and 4-hydroxy-2-nonenal (HNE) in brain and by malondialdehydes in plasma (Terasawa et al., 2000; Yokota et al., 2001). α-TTP deletion with vitamin E deficiency increased another lipid peroxidation biomarkers demonstrated by 8-iso-prostaglandin F (8-iso-PGF) and hydroxyoctadecadienoic acid (tHODE) in retina and retinal pigment epithelium (RPE) fraction (Tanito et al., 2007). Moreover, F2-isoprostanes levels in aortic lesions were increased in α-TTP-/-ApoE-/- mice, which are atherosclerosis susceptible and vitamin E deficient, when compared to that of α-TTP+/+ApoE-/- mice (Terasawa et al., 2000). α-TTP-/- mice challenged with lipopolysaccharide affected the inflammatory markers including heme oxygenase -1, inducible nitric oxide synthetase expression and nitric oxides (NOx) release in lung and liver (Schock et al., 2004).

Additionally, protein oxidation (protein carbonyl) in lung lavage fluid, lung and liver and protein kinase C activity were increased in polymorphonuclear cells of α-TTP-/- mice. However, levels of vitamin C and urate in lung tissues remained unchanged in α-TTP-/- mice (unpublished data).

As a supportive evidence, genes regulated by antioxidant response elements such as metallothionein II and glutathione-s-transferase π were selectively induced in α-TTP-/- liver. However, classical antioxidant enzymes such as catalase, gluthathione peroxidase, and superoxide dismutase were not induced in α-TTP-/- liver, lung and heart (Gohil et al., 2003; Oommen et al., 2007; Vasu et al., 2007). It was similar patterns shown in lung tissues of α-TTP+/+ mice fed an vitamin E deficient diet (Gohil et al., 2004) as shown in the lung tissues of α-TTP-/- mice (Gohil et al., 2007). Interestingly, genes involved in detoxification of xenobiotics (cytochrome p450 1a2 and 2d9) were induced in α-TTP-/- liver and herat (Gohil et al., 2003; Vasu et al., 2007).

Vit E has been known as a regulator of immune functions and vit E deficiency modulates immune responses (Beharka et al., 1997). The immunoglobulin genes including immunoglobulin kappa chain variable 8, immunoglobulin joining chain, immunoglobulin heavy chain, complement factor D were down-regulated (Vasu et al., 2007).

These results suggest that α-TTP may play a role in modulation of inflammatory-immune responses and antioxidant systems. Moreover, α-TTP-/- mice are mildly susceptible to oxidative stress and might use other indirect antioxidant defense systems instead of classical antioxidant defense mechanism. Therefore, α-TTP-/- mice are a practical model to discriminate antioxidant and anti-inflammatory functions α-T and to identify biomarkers of oxidative stress and inflammation in various disease states.

The relationship between dietary α-T and plasma or tissue α-T

α-TTP is the primary determinant of plasma α-T levels (Gohil et al., 2003; Jishage et al., 2001; Leonard et al., 2002; Schock et al., 2004; Terasawa et al., 2000; Yokota et al., 2001). Plasma and tissue α-T levels were very low in α-TTP-/- mice fed α-T deficient or normal diets but with vitamin E supplementation concentrations were comparable to those of wild-type mice fed normal diets (Table 1). During chylomicron catabolism vitamin E is transported to other lipoproteins bypassing the sorting function of α-TTP in the liver (Traber et al., 1990). If vitamin E intakes are sufficiently high, then α-TTP is not necessary to maintain plasma α-T concentrations.

Liver α-T levels in α-TTP-/- mice were 34-47% of those of wild-type mice when fed normal diets (30-150 mg/kg) (Table 1). Brain α-T concentrations in α-TTP-/- mice were less than 10% of those of wild-type mice fed normal diets (Traber et al., 2005). In long term (38 weeks) vitamin E deficient wild-type mice, the brain retained approximately 50% of its α-T while liver was approximately 95% deficient (Cuddihy et al., 2004; Traber et al., 2005). Interestingly, liver α-T in α-TTP-/- mice on normal diets was relatively well-maintained compared to wild-type mice with dietary vitamin E deficiency. This might be due to non-specific transport of α-T from gut to liver. Our group reported that α-T modulates cytochrome p 450 system especially, cyp3a, which is responsible for xenobiotic metabolism and is regulated by pregnane X receptor (Goodwin et al., 2002; Traber et al., 2005). This regulatory role of α-T in metabolizing toxic substances may be the reason that the liver maintains relatively high α-T levels compared to other organs. The results also suggest that α-TTP-/- mice are more susceptible to neurological disorders because brain α-T levels are lower in α-TTP-/- mice compared to dietary vitamin E deficient wild-type mice.

Lung, spleen, kidney and heart α-T levels in α-TTP-/- mice were very low (only 10-15%) compared with those of wild-type mice fed normal diets (Leonard et al., 2002; Traber et al., 2005). In chow (vitamin E, ~99 mg/kg)-fed mice for 27 weeks, α-T levels of α-TTP-/- liver, adipose tissue, adrenal gland and aorta were 15-35% of those of wild type mice (Terasawa et al., 2000). α-T levels were undetectable in the plasma (<0.46 µmol/l) and tissues (<0.2 nmol/g) of α-TTP-/- mice when fed α-T deficient diet for ~43 weeks. After α-T supplementation (600 mg/kg diet), α-T levels in α-TTP-/- mice increased to 70-90% of wild-type levels in the plasma (~3.5 µmol/l) and heart (~21 nmol/g) but were slightly increased in the cerebrum cortex (6.8%), cerebellum (19.6%), and spinal cord (23.8%) as compared to those of wild type mice fed normal diets (Yokota et al., 2001). Another study showed selective transport of RRR- or 2R-α-T from 1:1 mixture of natural and synthetic α-T by α-TTP and relatively higher plasma and tissue α-T levels compared to those of other studies in both α-TTP-/- and wild-type mice; these latter diets were high in polyunsaturated fat (Leonard et al., 2002).

Our group also demonstrated tissue specific gene expression profiles in 5 different tissues including liver, lung, brain, adrenal and spleen of α-TTP-/- mice (Gohil et al., 2004). The number of genes which are sensitive to α-TTP in tissues was not correlated with levels of α-T in the same tissues. Additionally, the levels of α-TTP expressed in tissues are not directly correlated with the levels of α-T (unpublished data). Each tissue might specifically preserve α-T for its own functions i.e. spleen and thymus for immune functions, plasma and liver for maintenance of homeostasis and lung and skin for protection from environmental insults such as ozone. In the plasma, the distribution of α-T to tissues is related to plasma levels of low density lipoprotein (LDL) and high density lipoprotein (HDL) which deliver α-T to tissues (Traber et al., 1992). Thus, α-T levels in plasma are dependent on plasma concentrations of these lipoproteins (Mardones et al., 2002). Scavenger receptor class B1 type (SR-B1) binds HDL, then mediates selective lipid uptake from HDL (Trigatti et al., 2000). SR-B1 knockout mice showed significant decreases in α-T levels in various tissues including liver, spleen and kidney. SR-B1 may be responsible for tissue α-T levels by regulating α-T influx into cells (Mardones et al., 2002). These results suggest that the levels of lipoproteins and SR-B1 maybe responsible for regulation of plasma and tissue α-T concentrations.

Variability of α-T levels shown in the same tissues treated with normal diets maybe due to the dietary form (natural or synthetic) of α-T, the amount of dietary fat in diets and/or the duration of dietary treatments. With α-T supplementation plasma and tissue levels of α-T in α-TTP-/- mice on the diets containing different levels of α-T showed considerable variability suggesting that these differences are likely to be tissue specific.

Since there are still limitations to evaluate the relationship between dietary α-T levels and tissue α-T concentrations, and the roles of α-TTP in different tissues further studies in this animal model are recommended.

Conclusions

α-TTP-/- mice are characterized by abnormalities that can be reversed, prevented or attenuated by high α-T supplementation. Additionally, specific genes sensitive to α-TTP were discussed. Plasma and tissue α-T levels in α-TTP-/- mice reflected dietary α-T levels in tissue specific manner.

These results emphasize the benefit of α-T supplementation in vitamin E deficiency caused by genetic defects of α-TTP in humans (AVED). However, α-TTP expression in various tissues leads to questions such as tissue specific α-T requirements and other α-TTP functions beyond binding and transferring α-T in cells. α-TTP-/- mice combined with dietary vitamin E deficiency appear to present an ideal model for investigating the specific roles of α-T and even other vitamin E analogues in different tissues. Furthermore, α-TTP-/- mice would be a good genetic model to identify genes associated with α-T in various diseases and for dissociating antioxidant functions of α-T from its non-antioxidant functions.

Notes

This work was supported in part by grants from the USDA NRICGP 2003-00915 and the NIH-NIEHS ES011985.

References

    1. Arita M, Sato Y, Miyata A, Tanabe T, Takahashi E, Kayden HJ, Arai H, Inoue K. Human alpha-tocopherol transfer protein: cDNA cloning, expression and chromosomal localization. Biochem J 1995;306:437–443.
    1. Ban R, Takitani K, Kim HS, Murata T, Morinobu T, Ogihara T, Tamai H. alpha-Tocopherol transfer protein expression in rat liver exposed to hyperoxia. Free Radic Res 2002;36:933–938.
    1. Beharka A, Redican S, Leka L, Meydani SN. Vitamin E status and immune function. Methods Enzymol 1997;282:247–263.
    1. Boscoboinik D, Szewczyk A, Azzi A. Alpha-tocopherol (vitamin E) regulates vascular smooth muscle cell proliferation and protein kinase C activity. Arch Biochem Biophys 1991a;286:264–269.
    1. Boscoboinik D, Szewczyk A, Hensey C, Azzi A. Inhibition of cell proliferation by alpha-tocopherol. Role of protein kinase C. J Biol Chem 1991b;266:6188–6194.
    1. Catignani GL. An alpha-tocopherol binding protein in rat river cytoplasm. Biochem Biophys Acta 1975;497:349–357.
    1. Copp RP, Wisniewski T, Hentati F, Larnaout A, Ben Hamida M, Kayden HJ. Localization of alpha-tocopherol transfer protein in the brains of patients with ataxia with vitamin E deficiency and other oxidative stress related neurodegenerative disorders. Brain Res 1999;822:80–87.
    1. Cuddihy SL, Musiek ES, Morrow JD, Dugan LL. Long-term vitamin e deficiency in mice decreases superoxide radical production in brain. Ann N Y Acad Sci 2004;1031:428–431.
    1. Das P, Chowdhury M. Vitamin E-deficiency induced changes in ovary and uterus. Mol Cell Biochem 1999;198:151–156.
    1. Desrumaux C, Risold PY, Schroeder H, Deckert V, Masson D, Athias A, Laplanche H, Le Guern N, Blache D, Jiang XC, Tall AR, Desor D, Lagrost L. Phospholipid transfer protein (PLTP) deficiency reduces brain vitamin E content and increases anxiety in mice. Faseb J 2005;19:296–297.
    1. Doerflinger N, Linder C, Ouahchi K, Gyapay G, Weissenbach J, Le Paslier D, Rigault P, Belal S, Ben Hamida C, Hentati F, et al. Ataxia with vitamin E deficiency: refinement of genetic localization and analysis of linkage disequilibrium by using new markers in 14 families. Am J Hum Genet 1995;56:1116–1124.
    1. Evans HM, Bishop KS. On the existence of a hitherto unrecognizd dietary factor essential for reproduction. Science 1922;56:650.
    1. Food and Nutrition Board & Institute of Medicine. Dietary Reference Intakes for Vitamin C, Vitamin E, Selenium, and Carotenoids. Washington. USA: National Academy Press; 2000.
    1. Gohil K, Godzdanker R, O'Roark E, Schock BC, Kaini RR, Packer L, Cross CE, Traber MG. [alpha]-Tocopherol Transfer Protein Deficiency in Mice Causes Multi-Organ Deregulation of Gene Networks and Behavioral Deficits with Age. Ann N Y Acad Sci 2004;1031:109–126.
    1. Gohil K, Oommen S, Vasu VT, Aung HH, Cross CE. Tocopherol transfer protein deficiency modifies nuclear receptor transcriptional networks in lungs: Modulation by cigarette smoke in vivo. Mol Aspects Med 2007;28:453–480.
    1. Gohil K, Schock BC, Chakraborty AA, Terasawa Y, Raber J, Farese RV Jr, Packer L, Cross CE, Traber MG. Gene expression profile of oxidant stress and neurodegeneration in transgenic mice deficient in alpha-tocopherol transfer protein. Free Radic Biol Med 2003;35:1343–1354.
    1. Goodwin B, Redinbo MR, Kliewer SA. Regulation of cyp3a gene transcription by the pregnane x receptor. Annu Rev Pharmacol Toxicol 2002;42:1–23.
    1. Hoshino M, Masuda N, Ito Y, Murata M, Goto J, Sakurai M, Kanazawa I. Ataxia with isolated vitamin E deficiency: a Japanese family carrying a novel mutation in the alpha-tocopherol transfer protein gene. Ann Neurol 1999;45:809–812.
    1. Hosomi A, Arita M, Sato Y, Kiyose C, Ueda T, Igarashi O, Arai H, Inoue K. Affinity for alpha-tocopherol transfer protein as a determinant of the biological activities of vitamin E analogs. FEBS Lett 1997;409:105–108.
    1. Hosomi A, Goto K, Kondo H, Iwatsubo T, Yokota T, Ogawa M, Arita M, Aoki J, Arai H, Inoue K. Localization of alpha-tocopherol transfer protein in rat brain. Neurosci Lett 1998;256:159–162.
    1. Jishage K, Arita M, Igarashi K, Iwata T, Watanabe M, Ogawa M, Ueda O, Kamada N, Inoue K, Arai H, Suzuki H. Alpha-tocopherol transfer protein is important for the normal development of placental labyrinthine trophoblasts in mice. J Biol Chem 2001;276:1669–1672.
    1. Kaempf-Rotzoll DE, Horiguchi M, Hashiguchi K, Aoki J, Tamai H, Linderkamp O, Arai H. Human placental trophoblast cells express alpha-tocopherol transfer protein. Placenta 2003;24:439–444.
    1. Kaempf-Rotzoll DE, Igarashi K, Aoki J, Jishage K, Suzuki H, Tamai H, Linderkamp O, Arai H. Alpha-tocopherol transfer protein is specifically localized at the implantation site of pregnant mouse uterus. Biol Reprod 2002;67:599–604.
    1. Leonard SW, Terasawa Y, Farese RV Jr, Traber MG. Incorporation of deuterated RRR- or all-rac-alpha-tocopherol in plasma and tissues of alpha-tocopherol transfer protein--null mice. Am J Clin Nutr 2002;75:555–560.
    1. Mardones P, Strobel P, Miranda S, Leighton F, Quinones V, Amigo L, Rozowski J, Krieger M, Rigotti A. Alpha-tocopherol metabolism is abnormal in scavenger receptor class B type I (SR-BI)-deficient mice. J Nutr 2002;132:443–449.
    1. Mariotti C, Gellera C, Rimoldi M, Mineri R, Uziel G, Zorzi G, Pareyson D, Piccolo G, Gambi D, Piacentini S, Squitieri F, Capra R, Castellotti B, Di Donato S. Ataxia with isolated vitamin E deficiency: neurological phenotype, clinical follow-up and novel mutations in TTPA gene in Italian families. Neurol Sci 2004;25:130–137.
    1. Meier R, Tomizaki T, Schulze-Briese C, Baumann U, Stocker A. The molecular basis of vitamin E retention: structure of human alpha-tocopherol transfer protein. J Mol Biol 2003;331:725–734.
    1. Min KC, Kovall RA, Hendrickson WA. Crystal structure of human alpha-tocopherol transfer protein bound to its ligand: implications for ataxia with vitamin E deficiency. Proc Natl Acad Sci USA 2003;100:14713–14718.
    1. Morley S, Panagabko C, Shineman D, Mani B, Stocker A, Atkinson J, Manor D. Molecular determinants of heritable vitamin E deficiency. Biochemistry 2004;43:4143–4149.
    1. Muller-Schmehl K, Beninde J, Finckh B, Florian S, Dudenhausen JW, Brigelius-Flohe R, Schuelke M. Localization of alpha-tocopherol transfer protein in trophoblast, fetal capillaries' endothelium and amnion epithelium of human term placenta. Free Radic Res 2004;38:413–420.
    1. Mustacich DJ, Leonard SW, Devereaux MW, Sokol RJ, Traber MG. Alpha-tocopherol regulation of hepatic cytochrome P450s and ABC transporters in rats. Free Radic Biol Med 2006;41:1069–1078.
    1. Oommen S, Vasu VT, Leonard SW, Traber MG, Cross CE, Gohil K. Genome wide responses of murine lungs to dietary alpha-tocopherol. Free Radic Res 2007;41:98–109.
    1. Ouahchi K, Arita M, Kayden H, Hentati F, Ben Hamida M, Sokol R, Arai H, Inoue K, Mandel JL, Koenig M. Ataxia with isolated vitamin E deficiency is caused by mutations in the alpha-tocopherol transfer protein. Nat Genet 1995;9:141–145.
    1. Packer L. Protective role of vitamin E in biological systems. Am J Clin Nutr 1991;53:1050S–1055S.
    1. Panagabko C, Morley S, Hernandez M, Cassolato P, Gordon H, Parsons R, Manor D, Atkinson J. Ligand specificity in the CRAL-TRIO protein family. Biochemistry 2003;42:6467–6474.
    1. Panagabko C, Morley S, Neely S, Lei H, Manor D, Atkinson J. Expression and refolding of recombinant human alpha-tocopherol transfer protein capable of specific alpha-tocopherol binding. Protein Expr Purif 2002;24:395–403.
    1. Qanungo S, Sen A, Mukherjea M. Antioxidant status and lipid peroxidation in human feto-placental unit. Clin Chim Acta 1999;285:1–12.
    1. Ricciarelli R, Tasinato A, Clement S, Ozer NK, Boscoboinik D, Azzi A. alpha-Tocopherol specifically inactivates cellular protein kinase C alpha by changing its phosphorylation state. Biochem J 1998;334:243–249.
    1. Schock BC, Van der Vliet A, Corbacho AM, Leonard SW, Finkelstein E, Valacchi G, Obermueller-Jevic U, Cross CE, Traber MG. Enhanced inflammatory responses in alpha-tocopherol transfer protein null mice. Arch Biochem Biophys 2004;423:162–169.
    1. Schuelke M, Elsner A, Finckh B, Kohlschutter A, Hubner C, Brigelius-Flohe R. Urinary alpha-tocopherol metabolites in alpha-tocopherol transfer protein-deficient patients. J Lipid Res 2000;41:1543–1551.
    1. Sen A, Mukherjea M. Modulation of bilayer fluidity by lipid peroxidation of human placental syncytiotrophoblast membranes during embryogenesis. Indian J Biochem Biophys 1998;35:216–223.
    1. Tanito M, Yoshida Y, Kaidzu S, Chen ZH, Cynshi O, Jishage K, Niki E, Ohira A. Acceleration of age-related changes in the retina in alpha-tocopherol transfer protein null mice fed a Vitamin E-deficient diet. Invest Ophthalmol Vis Sci 2007;48:396–404.
    1. Terasawa Y, Ladha Z, Leonard SW, Morrow JD, Newland D, Sanan D, Packer L, Traber MG, Farese RV Jr. Increased atherosclerosis in hyperlipidemic mice deficient in alpha-tocopherol transfer protein and vitamin E. Proc Natl Acad Sci U S A 2000;97:13830–13834.
    1. Traber MG. Vitamin E. In: Shils ME, Olson JA, Shike M, Ross AC, editors. Modern Nutrition in Health and Disease. Baltimore: Williams & Willkins; 1999. pp. 347-361.
    1. Traber MG, Arai H. Molecular mechanisms of vitamin E transport. Annu Rev Nutr 1999;19:343–355.
    1. Traber MG, Lane JC, Lagmay NR, Kayden HJ. Studies on the transfer of tocopherol between lipoproteins. Lipids 1992;27:657–663.
    1. Traber MG, Siddens LK, Leonard SW, Schock B, Gohil K, Krueger SK, Cross CE, Williams DE. Alpha-tocopherol modulates Cyp3a expression, increases gamma-CEHC production, and limits tissue gamma-tocopherol accumulation in mice fed high gamma-tocopherol diets. Free Radic Biol Med 2005;38:773–785.
    1. Traber MG, Sokol RJ, Burton GW, Ingold KU, Papas AM, Huffaker JE, Kayden HJ. Impaired ability of patients with familial isolated vitamin E deficiency to incorporate alpha-tocopherol into lipoproteins secreted by the liver. J Clin Invest 1990;85:397–407.
    1. Trigatti B, Rigotti A, Krieger M. The role of the high-density lipoprotein receptor SR-BI in cholesterol metabolism. Curr Opin Lipidol 2000;11:123–131.
    1. Vasu VT, Hobson B, Gohil K, Cross CE. Genome-wide screening of alpha-tocopherol sensitive genes in heart tissue from alpha-tocopherol transfer protein null mice (ATTP (-/-)). FEBS Lett 2007;581:1572–1578.
    1. Yokota T, Igarashi K, Uchihara T, Jishage K, Tomita H, Inaba A, Li Y, Arita M, Suzuki H, Mizusawa H, Arai H. Delayed-onset ataxia in mice lacking alpha-tocopherol transfer protein: model for neuronal degeneration caused by chronic oxidative stress. Proc Natl Acad Sci USA 2001;98:15185–15190.
    1. Yokota T, Shiojiri T, Gotoda T, Arita M, Arai H, Ohga T, Kanda T, Suzuki J, Imai T, Matsumoto H, Harino S, Kiyosawa M, Mizusawa H, Inoue K. Friedreich-like ataxia with retinitis pigmentosa caused by the His101Gln mutation of the alpha-tocopherol transfer protein gene. Ann Neurol 1997;41:826–832.
    1. Yokota T, Uchihara T, Kumagai J, Shiojiri T, Pang JJ, Arita M, Arai H, Hayashi M, Kiyosawa M, Okeda R, Mizusawa H. Postmortem study of ataxia with retinitis pigmentosa by mutation of the alpha-tocopherol transfer protein gene. J Neurol Neurosurg Psychiatry 2000;68:521–525.

Metrics
Share
Tables

1 / 1

PERMALINK