Английская Википедия:Apolipoprotein E

Материал из Онлайн справочника
Перейти к навигацииПерейти к поиску

Шаблон:Short description Шаблон:Cs1 config Шаблон:Infobox gene

Apolipoprotein E (Apo-E) is a protein involved in the metabolism of fats in the body of mammals. A subtype is implicated in the Alzheimer's disease and cardiovascular diseases.[1] It is encoded in humans by the gene APOE.

Apo-E belongs to a family of fat-binding proteins called apolipoproteins. In the circulation, it is present as part of several classes of lipoprotein particles, including chylomicron remnants, VLDL, IDL, and some HDL.[2] APOE interacts significantly with the low-density lipoprotein receptor (LDLR), which is essential for the normal processing (catabolism) of triglyceride-rich lipoproteins.[3] In peripheral tissues, APOE is primarily produced by the liver and macrophages, and mediates cholesterol metabolism. In the central nervous system, Apo-E is mainly produced by astrocytes and transports cholesterol to neurons[4] via APOE receptors, which are members of the low density lipoprotein receptor gene family.[5] Apo-E is the principal cholesterol carrier in the brain.[6] APOE qualifies as a checkpoint inhibitor of the classical complement pathway by complex formation with activated C1q.[7]

Evolution

Apolipoproteins are not unique to mammals. Many terrestrial and marine vertebrates have versions of them.[8] It is believed that APOE arose via gene duplications of APOC1 before the fish-tetrapod split c. 400 million years ago. Proteins similar in function have been found in choanoflagellates, suggesting that they are a very old class of proteins predating the dawn of all living animals.[9]

The three major human alleles (E4, E3, E2) arose after the primate-human split around 7.5 million years ago. These alleles are the by-product of non-synonymous mutations which led to changes in functionality. The first allele to emerge was E4. After the primate-human split, there were four amino acid changes in the human lineage, three of which had no effect on protein function (V174L, A18T, A135V). The fourth substitution (T61R) traded a threonine for an arginine altering the protein's functionality. This substitution occurred somewhere in the 6 million year gap between the primate-human split and the Denisovan-human split, since exactly the same substitutions were found in Denisovan APOE.[10]

About 220,000 years ago, a cysteine to arginine substitution took place at amino acid 112 (Cys112Arg) of the APOE4 gene, and this resulted in the E3 allele. Finally, 80,000 years ago, another arginine to cysteine substitution at amino acid 158 (Arg158Cys) of the APOE3 gene created the E2 allele.[11][9]

Structure

Gene

The gene, APOE, is mapped to chromosome 19 in a cluster with apolipoprotein C1 (APOC1) and the apolipoprotein C2 (APOC2). The APOE gene consists of four exons and three introns, totaling 3597 base pairs. APOE is transcriptionally activated by the liver X receptor (an important regulator of cholesterol, fatty acid, and glucose homeostasis) and peroxisome proliferator-activated receptor γ, nuclear receptors that form heterodimers with retinoid X receptors.[12] In melanocytic cells APOE gene expression may be regulated by MITF.[13]

Protein

APOE is 299 amino acids long and contains multiple amphipathic α-helices. According to crystallography studies, a hinge region connects the N- and C-terminal regions of the protein. The N-terminal region (residues 1–167) forms an anti-parallel four-helix bundle such that the non-polar sides face inside the protein. Meanwhile, the C-terminal domain (residues 206–299) contains three α-helices which form a large exposed hydrophobic surface and interact with those in the N-terminal helix bundle domain through hydrogen bonds and salt-bridges. The C-terminal region also contains a low density lipoprotein receptor (LDLR)-binding site.[14]

Polymorphisms

Шаблон:Infobox single nucleotide polymorphism APOE is polymorphic,[15][16] with three major alleles (epsilon 2, epsilon 3, and epsilon 4): APOE-ε2 (cys112, cys158), APOE-ε3 (cys112, arg158), and APOE-ε4 (arg112, arg158).[1][17][18] Although these allelic forms differ from each other by only one or two amino acids at positions 112 and 158,[19][20][21] these differences alter APOE structure and function.

There are several low-frequency polymorphisms of APOE. APOE5 comes in two subtypes E5f and E5s, based on migration rates. APOE5 E5f and APOE7 combined were found in 2.8% of Japanese males.[22]Шаблон:Unreliable medical source APOE7 is a mutation of APOE3 with two lysine residues replacing glutamic acid residues at positions 244 and 245.[23]

Polymorphism Worldwide allele frequency Disease relevance
ε2 (rs7412-T, rs429358-T) 8.4%[5] This variant of the apoprotein binds poorly to cell surface receptors while E3 and E4 bind well.[24] E2 is associated with both increased and decreased risk for atherosclerosis. Individuals with an E2/E2 combination may clear dietary fat slowly and be at greater risk for early vascular disease and the genetic disorder type III hyperlipoproteinemia—94.4% of people with such disease are E2/E2 but only ~2% of E2/E2 develop it, so other environmental and genetic factors are likely to be involved (such as cholesterol in the diet and age).[25][26][27] E2 has also been implicated in Parkinson's disease,[28] but this finding was not replicated in a larger population association study.[29]
ε3 (rs7412-C, rs429358-T) 77.9%[5] This variant is considered the "neutral" APOE genotype.
Шаблон:Anchorε4 (rs7412-C, rs429358-C) 13.7%[5]

E4 has been implicated in atherosclerosis,[30][31] Alzheimer's disease,[32][33] impaired cognitive function,[34][35] reduced hippocampal volume,[36] HIV,[37] faster disease progression in multiple sclerosis,[38][39] unfavorable outcome after traumatic brain injury,[40] ischemic cerebrovascular disease,[41] sleep apnea,[42][43] both the extension and shortening of telomeres,[44][45][46][47] reduced neurite outgrowth,[48] and COVID-19.[49] However, E4 has also been associated with enhanced vitamin D and calcium status,[50] higher fecundity,[51] protection against early childhood infection and malnutrition,[52] and decreased fetal, perinatal, and infant mortality.[53]

Much remains to be learned about the APOE isoforms, including the interaction of other protective genes.[54] Indeed, the apolipoprotein ε4 isoform is more protective against cognitive decline than other isoforms in some cases,[54] so caution is advised before making determinant statements about the influence of APOE polymorphisms on cognition, development of Alzheimer's disease, cardiovascular disease, telomere shortening, etc. Many of the studies cited that purport these adverse outcomes are from single studies that have not been replicated and the research is based on unchecked assumptions about this isoform. As of 2007, there was no evidence that APOE polymorphisms influence cognition in younger age groups (other than possible increased episodic memory ability and neural efficiency in younger APOE4 age groups), nor that the APOE4 isoform places individuals at increased risk for any infectious disease.[55]

However, the association between the APOE4 allele and Alzheimer's disease has been shown to be weaker in minority groups differently compared to their Caucasian counterparts.[5] Hispanics/Latinos and African Americans who were homozygous for the APOE4 allele had 2.2 and 5.7 times the odds, respectively of developing Alzheimer's disease.[56][5] The APOE4 allele has an even stronger effect in East Asian populations, with Japanese populations have 33 times the odds compared to other populations.[57] Caucasians who were homozygous for the allele had 12.5 times the odds.[56][5]

Function

APOE transports lipids, fat-soluble vitamins, and cholesterol into the lymph system and then into the blood. It is synthesized principally in the liver, but has also been found in other tissues such as the brain, kidneys, and spleen.[17] In the nervous system, non-neuronal cell types, most notably astroglia and microglia, are the primary producers of APOE, while neurons preferentially express the receptors for APOE.[58] There are seven currently identified mammalian receptors for APOE which belong to the evolutionarily conserved LDLR family.[59]

APOE was initially recognized for its importance in lipoprotein metabolism and cardiovascular disease. Defects in APOE result in familial dysbetalipoproteinemia aka type III hyperlipoproteinemia (HLP III), in which increased plasma cholesterol and triglycerides are the consequence of impaired clearance of chylomicron, VLDL and LDL.[60][3] More recently, it has been studied for its role in several biological processes not directly related to lipoprotein transport, including Alzheimer's disease (AD), immunoregulation, and cognition.[1] Though the exact mechanisms remain to be elucidated, isoform 4 of APOE, encoded by an APOE allele, has been associated with increased calcium ion levels and apoptosis following mechanical injury.[61]

In the field of immune regulation, a growing number of studies point to APOE's interaction with many immunological processes, including suppressing T cell proliferation, macrophage functioning regulation, lipid antigen presentation facilitation (by CD1)[62] to natural killer T cell as well as modulation of inflammation and oxidation.[63] APOE is produced by macrophages and APOE secretion has been shown to be restricted to classical monocytes in PBMC, and the secretion of APOE by monocytes is down regulated by inflammatory cytokines and upregulated by TGF-beta.[64]

Clinical significance

Alzheimer's disease

As of 2012, the E4 variant was the largest known genetic risk factor for late-onset sporadic Alzheimer's disease (AD) in a variety of ethnic groups.[65] However, the E4 variant does not correlate with risk in every population. Nigerian people have the highest observed frequency of the APOE4 allele in world populations,[66] but AD is rare among them.[66][67] This may be due to their low cholesterol levels.[66][67][68][69] Caucasian and Japanese carriers of two E4 alleles have between 10 and 30 times the risk of developing AD by 75 years of age, as compared to those not carrying any E4 alleles. This may be caused by an interaction with amyloid.[70] Alzheimer's disease is characterized by build-ups of aggregates of the peptide beta-amyloid. Apolipoprotein E enhances proteolytic break-down of this peptide, both within and between cells. The isoform APOE-ε4 is not as effective as the others at promoting these reactions, resulting in increased vulnerability to AD in individuals with that gene variation.[71]

Recently, the amyloid hypothesis of Alzheimer's disease has been questioned, and an article in Science claimed that "Just as removing smoke does not extinguish a fire, reducing amyloid plaques may not affect the course of Alzheimer's disease."[72] The role that the E4 variant carries can still be fully explained even in the absence of a valid amyloid hypothesis given the fact that reelin signaling emerges to be one of the key processes involved in Alzheimer's disease[73] and the E4 variant is shown to interact with ApoER2, one of the neuronal reelin receptors, thereby obstructing reelin signaling.[73]

Although 40–65% of AD patients have at least one copy of the ε4 allele, APOE4 is not a determinant of the disease. At least one-third of patients with AD are APOE4 negative and some APOE4 homozygotes never develop the disease. Yet those with two ε4 alleles have up to 20 times the risk of developing AD.[74] There is also evidence that the APOE2 allele may serve a protective role in AD.[75] Thus, the genotype most at risk for Alzheimer's disease and at an earlier age is APOE4,4. Using genotype APOE3,3 as a benchmark (with the persons who have this genotype regarded as having a risk level of 1.0) and for white populations only, individuals with genotype APOE4,4 have an odds ratio of 14.9 of developing Alzheimer's disease. Individuals with the APOE3,4 genotype face an odds ratio of 3.2, and people with a copy of the 2 allele and the 4 allele (APOE2,4), have an odds ratio of 2.6. Persons with one copy each of the 2 allele and the 3 allele (APOE2,3) have an odds ratio of 0.6. Persons with two copies of the 2 allele (APOE2,2) also have an odds ratio of 0.6.[76]

Estimated worldwide human allele frequencies of APOE in Caucasian population[76]
Allele ε2 ε3 ε4
General Frequency 8.4% 77.9% 13.7%
AD Frequency 3.9% 59.4% 36.7%

While ApoE4 has been found to greatly increase the odds that an individual will develop Alzheimer's, a 2002 study concluded, that in persons with any combination of APOE alleles, high serum total cholesterol and high blood pressure in mid-life are independent risk factors which together can nearly triple the risk that the individual will later develop AD.[69] Projecting from their data, some researchers have suggested that lowering serum cholesterol levels may reduce a person's risk for Alzheimer's disease, even if they have two ApoE4 alleles, thus reducing the risk from nine or ten times the odds of getting AD down to just two times the odds.[69]

Women are more likely to develop AD than men across most ages and APOE genotypes. Premorbid women with the ε4 allele have significantly more neurological dysfunction than men.[77]

APOE-ε4 increases the risk not only for AD but also for dementia in pure alpha-synucleinopathies.[78] The influence of APOE-ε4 on hippocampal atrophy was suggested to be more predominant early in the course of AD at milder stages prior to more widespread neurodegeneration.[79]

Atherosclerosis

Knockout mice that lack the apolipoprotein-E gene (APOE−/−) develop extreme hypercholesterolemia when fed a high-fat diet.[80]

Malaria

APOE−/− knockout mice show marked attenuation of cerebral malaria and increased survival, as well as decreased sequestration of parasites and T cells within the brain, likely due to protection of the blood–brain barrier.[81] Human studies have shown that the APOE2 polymorphism correlates with earlier infection, and APOE3/4 polymorphisms increase likelihood of severe malaria.[82]

Lyme Disease

Borrelia burgdorferi, the causative agent of Lyme disease, is a host-adapted pathogen that acquires environmental cholesterol to form glycolipids for use in cell membrane maintenance. In one experiment in 2015, mice engineered with apoE deficiency were infected with Borrelia spirochetes. The knockout mice suffered from an increased spirochete burden in joints, as well as inflamed ankles, when compared with wild-type mice. This study suggests that apoE deficiency (and potentially other hyperlipidemias) may be a risk factor in the pathogenicity of Lyme disease.

Interactions

Interactive pathway map

Шаблон:StatinPathway WP430

References

Шаблон:Reflist

Further reading

Шаблон:Refbegin

Шаблон:Refend

External links

Шаблон:PDB Gallery Шаблон:Lipoproteins Шаблон:Longevity

  1. 1,0 1,1 1,2 Шаблон:Cite book
  2. Шаблон:Cite journal
  3. 3,0 3,1 Шаблон:Cite web
  4. Шаблон:Cite journal
  5. 5,0 5,1 5,2 5,3 5,4 5,5 5,6 Шаблон:Cite journal
  6. Шаблон:Cite journal
  7. Шаблон:Cite journal
  8. Шаблон:Cite journal
  9. 9,0 9,1 Шаблон:Cite journal
  10. Шаблон:Cite journal
  11. Шаблон:Cite journal
  12. Шаблон:Cite journal
  13. Шаблон:Cite journal
  14. Шаблон:Cite journal
  15. Шаблон:Cite journal
  16. Шаблон:Cite journal
  17. 17,0 17,1 Шаблон:Cite book
  18. Шаблон:Cite journal
  19. Шаблон:OMIM
  20. Шаблон:OMIM
  21. Шаблон:Cite journal
  22. Шаблон:Cite journal
  23. Шаблон:Cite journal
  24. Шаблон:Cite journal
  25. Шаблон:Cite journal
  26. Шаблон:Cite journal
  27. Шаблон:Cite journal
  28. Шаблон:Cite journal
  29. Шаблон:Cite journal
  30. Шаблон:Cite journal
  31. Шаблон:Cite journal
  32. Шаблон:Cite journal
  33. Шаблон:Cite journal
  34. Шаблон:Cite journal
  35. Шаблон:Cite journal
  36. Шаблон:Cite journal
  37. Шаблон:Cite journal
  38. Шаблон:Cite journal
  39. Шаблон:Cite journal
  40. Шаблон:Cite journal
  41. Шаблон:Cite journal
  42. Шаблон:Cite journal
  43. Шаблон:Cite journal
  44. Шаблон:Cite journal
  45. Шаблон:Cite journal
  46. Шаблон:Cite journal
  47. Шаблон:Cite journal
  48. Шаблон:Cite journal
  49. Шаблон:Cite journal
  50. Шаблон:Cite journal
  51. Шаблон:Cite journal
  52. Шаблон:Cite journal
  53. Шаблон:Cite journal
  54. 54,0 54,1 Sundermann EE, Wang C, Katz M, et al. Cholesteryl ester transfer protein genotype modifies the effect of apolipoprotein ε4 on memory decline in older adults. Neurobiol Aging. 2016;41:200.e7-200.e12. doi:10.1016/j.neurobiolaging.2016.02.006
  55. Шаблон:Cite journal
  56. 56,0 56,1 Шаблон:Cite journal
  57. Шаблон:Cite journal
  58. Шаблон:Cite journal
  59. Шаблон:Cite journal
  60. Шаблон:Cite journal
  61. Шаблон:Cite journal
  62. Шаблон:Cite journal
  63. Шаблон:Cite journal
  64. Шаблон:Cite journal
  65. Шаблон:Cite journal
  66. 66,0 66,1 66,2 Шаблон:Cite journal
  67. 67,0 67,1 Шаблон:Cite journal
  68. Шаблон:Cite journal
  69. 69,0 69,1 69,2 Шаблон:Cite journal
  70. Шаблон:Cite journal
  71. Шаблон:Cite journal
  72. Шаблон:Cite journal
  73. 73,0 73,1 Шаблон:Cite journal
  74. Шаблон:Cite journal
  75. Шаблон:Cite journal
  76. 76,0 76,1 Шаблон:Cite journal
  77. Шаблон:Cite journal
  78. Шаблон:Cite journal
  79. Шаблон:Cite journal
  80. Шаблон:Cite journal
  81. Шаблон:Cite journal
  82. Шаблон:Cite journal