Buscar en
Gastroenterología y Hepatología
Toda la web
Inicio Gastroenterología y Hepatología Genetic factors associated with the presence and progression of nonalcoholic fat...
Información de la revista
Vol. 35. Núm. 1.
Páginas 32-41 (Enero 2012)
Compartir
Compartir
Descargar PDF
Más opciones de artículo
Visitas
7872
Vol. 35. Núm. 1.
Páginas 32-41 (Enero 2012)
Progress in hepatology
Acceso a texto completo
Genetic factors associated with the presence and progression of nonalcoholic fatty liver disease: A narrative review
Factores genéticos asociados con la presencia y progresión de la enfermedad hepática no alcohólica: Una revisión narrativa
Visitas
7872
Ruben Hernaeza,b,
Autor para correspondencia
rhernae1@jhmi.edu

Corresponding author.
a Department of Medicine, The Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
b Department of Medicine, Georgetown University Hospital/Washington Hospital Center, Washington, DC 20010, USA
Este artículo ha recibido
Información del artículo
Resumen
Texto completo
Bibliografía
Descargar PDF
Estadísticas
Tablas (2)
Table 1. Candidate gene studies in nonalcoholic fatty liver disease (last updated 7/27/11).
Table 2. Genome-wide association studies in NAFLD (last updated 7/27/11).
Mostrar másMostrar menos
Abstract

Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in the world. Whereas insulin resistance and obesity are considered major risk factors for the development and progression of NAFLD, the genetic underpinnings are unclear. Before 2008, candidate gene studies based on prior knowledge of pathophysiology of fatty liver yielded conflicting results. In 2008, Romeo et al. published the first genome wide association study and reported the strongest genetic signal for the presence of fatty liver (PNPLA3, patatin-like phospholipase domain containing 3; rs738409). Since then, two additional genome wide scans were published and identified 9 additional genetic variants. Whereas these results shed light into the understanding of the genetics of NAFLD, most of associations have not been replicated in independent samples and, therefore, remain undetermined the significance of these findings. This review aims to summarize the understanding of genetic epidemiology of NAFLD and highlights the gaps in knowledge.

Keywords:
Genetic association studies
Nonalcoholic fatty liver disease
Fatty liver
Disease progression
Prevalence
Review
Resumen

La enfermedad hepática no alcohólica (EHNA) es la enfermedad crónica del hígado más común en el mundo. Si bien se considera que la resistencia a la insulina y la obesidad son factores de riesgo importantes para el desarrollo y progresión de la misma, sus bases genéticas no están claras. Antes de 2008, los estudios sobre genes candidatos basados en conocimientos previos de la fisiopatología de la esteatosis hepática produjeron resultados contradictorios. En 2008, Romeo et al. publicaron el primer estudio de asociación amplia del genoma que presentaba indicios genéticos sólidos de la presencia de esteatosis hepática (PNPLA3, dominio de la fosfolipasa-patatina 3; rs738409). Desde entonces, se han publicado dos estudios adicionales de asociación amplia del genoma en los que se identificaron otras nueve variantes genéticas. Si bien estos resultados arrojan luz sobre la genética de la EHNA, la mayoría de las asociaciones no se han replicado en muestras independientes y, por tanto, la importancia de estos hallazgos continúa siendo incierta. Esta revisión pretende resumir los conocimientos actuales sobre la epidemiología genética de la EHNA y destaca las áreas de incertidumbre.

Palabras clave:
Estudios genéticos de asociación
Enfermedad grasa del hígado no alcohólica
Hígado graso
Progresión de la enfermedad
Prevalencia
Revisión
Texto completo
Nonalcoholic fatty liver disease is common and has a genetic basis

Nonalcoholic fatty liver disease (NAFLD), the most common chronic liver disease,1 represents a wide spectrum of disease characterized by the presence hepatic steatosis in the absence of significant alcohol consumption or other causes of liver disease.2,3 The pathogenic processes leading to steatosis, steatohepatitis (NASH) and fibrosis are multifactorial and involve both environmental and genetic factors.4,5 Obesity and type 2 diabetes/insulin resistance are the most common risk factors for the development and progression of NAFLD.6 Since both obesity and type 2 diabetes continue to rise, it is expected that NAFLD will reach epidemic proportions worldwide. The prevalence of ultrasound-defined NAFLD ranges from 14 to 45%.7 Extrapolating data from small studies and the current prevalence of obesity and type 2 diabetes, NAFLD was recently estimated to affect more than 30 million people in the U.S.8 In Spain, Caballeria et al. showed in a recent cross-sectional study a prevalence of 26% in individuals aged 15 and 85 years randomly selected from 25 primary healthcare centers,9 consistent with prior American estimates.

A genetic underpinning for NAFLD has been suggested by familial aggregation studies,10,11 heritability studies,12,13 candidate gene studies,4 genome-wide scans14–17 and expression studies.18–22 The presence of a genetic basis in NAFLD not only will shed light in the identification of individuals at risk to develop NAFLD and its progression, but also the dissection of NAFLD pathogenesis and the development of new therapies.

The ultimate goal of this review is to provide the big picture of the current understanding of genes associated with NAFLD so the reader is able to understand what the gaps are in the genetic epidemiology of NAFLD. This paper, however, will not assess other forms of fatty liver disease, expression studies or animal studies.

This work is structured in three parts: first, the author will provide some background concepts in the genetic epidemiology of NAFLD and the current understanding of NAFLD pathophysiology; second, the reviewer will highlight the most up-to-date findings in candidate gene studies and genome wide association studies (GWAS) in humans; the review will conclude with suggestions to conduct candidate gene studies. Compared to other reviews,23–25 the present work will not elaborate the mechanism for each specific gene but will provide references for the reader for a more careful evaluation.

Study designs and methodological problems in genetic epidemiology of NAFLD

NAFLD is considered a complex disease because it does have a genetic component but with no simple Mendelian pattern of single-gene inheritance such as Wilson disease. In NAFLD, multiple genes, polygenes, environmental factors, age effects, and their interactions, may be involved.26

Genetic epidemiology studies can be divided into two broad categories: (a) according to the relatedness amongst participants (family-based versus non-related or population-based); and (b) according to the knowledge of the genetic marker(s) used (hypothesis-driven or candidate gene study versus hypothesis free or genome wide association study). A candidate gene is defined as gene whose product is considered to play a role in disease pathogenesis.27 It is important to bear in mind that all studies provide key information and are not mutually exclusive. For instance, family-based studies are typically the first step to determine if a condition is genetic in nature, the model of inheritance, and to understand the proportion of phenotypic variance due to shared genetic factors (heritability). Family-based studies may use linkage analysis to identify the location of a major gene or, association analyses such as the transmission-disequilibrium test, candidate gene approach and a genome-wide association analysis.

Population-based studies, on the other hand, is the mainstream of studies in NAFLD and, until recently, candidate gene studies were the most common design using the “two hit” hypothesis as a framework.5

Both candidate gene and GWAS rely on the statistical association of genetic markers and the disease (phenotype) of interest. The most commonly used genetic markers are the single nucleotide polymorphisms or SNPs, defined as the change in a single base pair of the DNA common in more than 1% of the population.23

The major problem in both candidate gene and GWAS is the presence of false negative and false positive associations. Lack of statistical power leads to false negatives; that is, true associations are not detected in the study. So the researcher needs to estimate the number of participants to be recruited to circumvent such error using power calculators. In GWAS, where there is no hypothesis behind the statistical analysis, the problem is the opposite and SNPs are spuriously associated due to chance with the disease, even after statistical correction for multiple comparisons. Consequently, most of the journals extremely recommend authors to replicate their findings in a different sample before accepting the new SNP as truly associated with NAFLD.

Steatosis: a protective mechanism surrounded by multiple insults

The key pathological finding in fatty liver disease is the accumulation of triglycerides in the hepatocytes. This deposit is due to an imbalance between triglycerides acquisition and removal.28,29 Triglycerides are neutral lipids consisting of a glycerol backbone and three long-chain fatty acids. The major routes of free fatty acids (FFAs) are (a) dietary intake; (b) lipolysis from fat reservoirs, and (c) de novo lipogenesis. Animal studies and human inherited diseases have shown increased hepatic steatosis with increased triglyceride dietary intake, increased lipolysis from peripheral tissues, increased de novo lipogenesis, or prevention of triglyceride removal from the liver (i.e. decreased efflux or oxidation).28 The exact contribution of each pathway for the development of hepatic steatosis has been studied by Donnelly et al.30 who showed that 59% of hepatic fat derived from circulating FFAs (mainly lipolysis), 26% from de novo lipogenesis and 15% from diet.

Insulin resistance is the key etiopathogenic factor in the development of fatty liver. Whether insulin resistance is a cause of hepatic steatosis or vice versa is a matter of debate and has been reviewed recently.28 Current evidence suggest that insulin resistance comes first and leads to hepatic steatosis, as shown by animal models and human diseases where hepatic steatosis is not associated with insulin resistance and, conversely, human diseases with inherited insulin resistance leading to hepatic steatosis (e.g. AKT2 mutation).28 Opposite views, however, suggest that hepatic steatosis, possibly originated by impaired mitochondrial β-oxidation of fatty acids, leads to hepatic insulin resistance which, in turn, increases de novo fatty acid synthesis, and impairs glucogenogenesis, hepatic glucose uptake, and transport of lipoproteins perpetuating increase peripheral insulin resistance.7,29

In 1998, Day and James proposed a “two-hit model” for the development of steatohepatitis in which the first step was the accumulation of free fatty acids in the liver, and, from there, further insults induced inflammation, fibrosis and, eventually, cirrhosis.5 Nevertheless, Dr. Day's group updated their own model and suggested that “steatosis may be early adaptive response to hepatocyte stress through which potentially lipotoxic FFAs are partitioned into relatively stable intracellular triglyceride stores”.31 NAFLD is seen now “as a combination of effects of several fundamental biochemical and immunologic mechanisms of liver injury rather than adhering to a sequential ‘two-hit’ paradigm”31 and is being supported by other experts in the field.32 In Day et al.’s view, “insulin resistance promotes increased hepatic FFA flux (lipolysis, diet and lipogenesis) leading to hepatic steatosis, and is driven by (1) direct hepatocyte lipotoxicity, (2) hepatocellular oxidative stress secondary to free radicals produced during beta- and -FFA oxidation, (3) endotoxin/TLR4-induced inflammation, (4) cytokine release, and(5) endoplasmic reticulum (ER) stress”, leading, at the end, to inflammation, cellular damage, and progression to cirrhosis.

Genetic epidemiology of NAFLD: a systematic approach

To identify most of the available published literature in genetics of NAFLD, the author performed a systematic search using PUBMED by applying the following search engine: (polymorphism OR SNP) AND (“steatosis” OR “steatohepatitis” OR NAFLD OR NASH) without language restriction and until July 26th, 2011. The search yielded 224 references; of which 60 corresponded to candidate gene studies and are summarized in Table 1 and three genome wide association studies in Table 2. This search engine is limited by two major factors: on the one hand, it did not include EMBASE database (more sensitive for non-English papers and scientific meetings), and, on the other hand, there is still a risk by publication bias (negative studies are not published).

Table 1.

Candidate gene studies in nonalcoholic fatty liver disease (last updated 7/27/11).

Gene (HGNC)  SNP  Effect on steatosis  Inflammation/NASH  Fibrosis 
Hepatic lipid metabolism
PEMT (8830),36–39 phosphatidylethanolamine N-methyltransferase  rs7946  ↑∅↑  ↑   
MTTP (7467),40–44 microsomal triglyceride transfer protein  rs1800591  ∅∅  ∅↑↑↑↑   
MTTP37,45  rs3816873  ↑∅     
MTTP45  rs61733139  ∅     
APOC3 (610),46,47 apolipoprotein C-III  rs2854116rs2854117  ↑∅     
NR1I2 (7968),48 nuclear receptor subfamily 1, group I, member 2  rs7643645  ↑  ↑   
FABP2 (2556),49 fatty acid binding protein 2, intestinal  rs1799883  ↑    (Alcoholic cirrhosis)50 
DGAT (2843),51 acyl-CoA:diacylglycerol acyltransferase  rs1944438  ∅     
ACSL4 (3571),52 acyl-CoA synthetase long-chain family member 4  rs7887981  ↑     
ADRB3 (288),53 adrenergic, beta-3-, receptor  rs4994  ↑  ↑   
ADRB2 (286),54 adrenergic, beta-2-, receptor, surface  rs1042714  ↑    (Response to β-blockade in cirrhosis)55 
LIPC(6619),36,56 hepatic lipase  rs1800588  ↑↑     
APOE (613),57–60 apolipoprotein E  N/A  ↑↓∅↓  ↑  (Fibrosis progression in hepatitis C)61 
CLOCK (2082),62 clock homolog (mouse)  rs11932595rs6843722rs1554483rs6843722rs6850524rs1554483  ↑  ↑  ↑ 
Insulin sensitivity
ENPP1 (3356),40,63 ectonucleotide pyrophosphatase/phosphodiesterase 1 or PC-1  rs1044498  ∅∅  ∅∅  ↑ 
IRS1 (6125),63 insulin receptor substrate 1  rs1801278  ∅  ∅  ∅ 
ADIPOQ (13633),64–67 adiponectin, C1Q and collagen domain containing  rs2241766  ∅∅∅↑↑  ↑  ∅ 
ADIPOQ64–67  rs1501299  ↓∅∅↑  ∅↑∅   
ADIPOR1 (24040),52 adiponectin receptor 1  rs6666089  ∅     
ADIPOR2 (24041),52 adiponectin receptor 2  rs767870  ↓     
PPARA (9232),68,69 peroxisome proliferator-activated receptor alpha  rs1800206  ∅↑  ∅  ∅ 
PPARG (9236),36 peroxisome proliferative activated receptor, gamma  rs3856806  ∅     
PPARG68,70,71  rs1801282  ∅∅∅  ∅∅  ∅∅ 
PPARGC1A (9237),36 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha  rs8192687  ∅     
TCF7L2 (11641),72 transcription factor 7-like 2 (T-cell specific, HMG-box)  rs7903146  ↑  ↑  ↑ 
GCKR (4196),73 glucokinase (hexokinase 4) regulator,  rs780094  ↑     
MC4R (6932),74 melanocortin-4 receptor  rs17782313  ∅     
SPINK-1 (11244),75 serine protease inhibitor Kazal-1  N/A  ∅     
LEPR (6554),76 leptin receptor gene  N/A  ↑     
LEP (6553),36 leptin  rs7799039  ∅     
Genes influencing generation of reactive oxidant species, or cytokine genes
TNF (11892),36,67,77–80 tumor necrosis factor-alpha  rs180062  ∅∅∅ (∅ hepatitis C)81  ↑∅∅  ↑∅ (↑ alcoholic liver cirrhosis)82 
Tumor necrosis factor-alpha67,79,80  rs361525  ↑↑↑∅  ∅∅  ∅ 
Tumor necrosis factor-alpha67,79  rs1800630  ∅  ↑  ∅ 
Tumor necrosis factor-alpha79  rs1799964  ∅  ↑   
Tumor necrosis factor-alpha79  rs1799724  ∅  ∅   
Tumor necrosis factor-beta79  rs909253  ∅  ∅   
TNFSF10 (11925)83 tumor necrosis factor (ligand) superfamily, member 10  N/A  ↓     
IL-6 (6018)40  rs1800795  ↑  ↑  (↑ liver cirrhosis and cancer)84,85 
CD14 (1628)86  rs2569190  ∅  ↑  (∅ hepatitis C)87 
GCLC (4311),45,42 glutamate–cysteine ligase, catalytic subunit,  rs4140528  ∅  ↑   
SOD2 (11180),43,44,88 superoxide dismutase 2, mitochondrial  rs4880  ↑  ∅↑↑  ↑ 
HFE (4886),89 hemochromatosis  rs1800562 rs1799945  ∅  ∅   
UGT1A1 (12530),90 UDP glucuronosyltransferase 1 family, polypeptide A1  N/A  ↓     
UCP (12519),91,92 uncoupling protein 3 (mitochondrial, proton carrier)  rs1800849  ↑  ↑   
PTGS2 (9506),93 prostaglandin-endoperoxide synthase 2 (prostaglandin G/H synthase and cyclooxygenase)  rs689466  ↑     
ABCB11 (42),94 ATP-binding cassette, subfamily B, member 11  rs2287622  ∅  ∅  ∅ 
MIF (797),95 macrophage migration inhibitory factor (glycosylation-inhibiting factor),  rs755622  ∅  ∅  ∅ 
CYP2E1 (2631),96,97 cytochrome P450, family 2, subfamily E, polypeptide 1  rs2031920  ∅↓  ∅   
SERPINA1 (8941),98 serpin peptidase inhibitor, clade A (alpha-1 antiproteinase, antitrypsin), member 1  rs28929474  ↑  ↑   
MTHFR (7436),99 methylenetetrahydrofolate reductase  rs1801131  ↑  ↑  (↑ hepatocellular carcinoma)100 
IL1B (5992),53 interleukin 1, beta  rs16944  ↑  ↑   
TLR4 (11850),86 toll-like receptor 4  N/A    ∅  ∅ 
CFTR/MRP (53),101 ATP-binding cassette, sub-family C, member 2  rs17222723 rs8187710  ↑  ∅   
STAT3 (11364),102 signal transducer and activator of transcription 3 acute-phase response factor  rs6503695 rs9891119  ↑  ∅   
HP (5141),103 haptoglobin  N/A  ↑     
AGTR1 (336),104 angiotensin II receptor, type 1  rs3772622  ↑  ↑   
KLF6 (2235),105 Kruppel-like factor 6  rs3750861  ↑    ↑ 

Abbreviations: N/A: not available; ↑: increase; ∅: no association; ↓: decrease (protective).

Table 2.

Genome-wide association studies in NAFLD (last updated 7/27/11).

Gene (HGNC)  SNP  Effect on steatosis  Inflammation/NASH  Fibrosis 
PNPLA3 (18590), patatin-like phospholipase domain containing 315,16  rs738409  ↑↑     
PNPLA3 (18590), patatin-like phospholipase domain containing 315  rs6006460  ↓     
FDFT1 (3629),14 farnesyl diphosphate farnesyl transferase 1  rs2645424    ↑   
COL13A1 (2190),14 collagen, type XIII, alpha 1  rs1227756      ↑ 
PDGFA (8799),14 platelet-derived growth factor alpha polypeptide  rs343064      ↑ 
LTBP3 (6716),14 latent transforming growth factor beta binding protein 3  rs1227756    ↑   
EFCAB4B (28657),14 EF-hand calcium binding domain 4B  rs887304    ↑   
NCAN (2465),16 neurocan  rs2228603  ↑     
LYPLAL1 (20440),16 lysophospholipase-like 1  rs12137855  ↑     
GCKR (4196),16 glucokinase regulatory protein  rs780094  ↑     
PPP1R3B (14942),16 protein phosphatase 1, regulatory subunit 3b  rs4240624  ↑     

Abbreviations: N/A: not available; ↑: increase; ∅: no association; ↓: decrease (protective).

Genes (or nearby genes) are named using the HUGO Gene Nomenclature Committee (HGNC) and reported identification.33

Candidate genes studies in human NAFLD: hepatic lipid metabolism, insulin resistance and oxidative stress

Numerous candidate gene studies, applying this ‘multiple-hit’ hypothesis, have studied the effects of genes on NAFLD presence and progression. The reviewer has simplified the prior hypothesis into the following mechanisms (not mutually exclusive): genes involved in hepatic lipid metabolism (synthesis, storage, export, oxidation), genes implicated in insulin signaling (insulin resistance), and finally, genes involved in oxidative stress and inflammation (and therefore, most likely involved in progression to cirrhosis) (Table 1).

The reviewer included, when appropriate, other data from different phenotypes to illustrate the direction of association for a particular gene. The number of symbols represent how many studies have been done for a particular gene.

Genome wide association studies in NAFLD

Romeo et al. were the first to apply the GWAS method using a phenotype based on magnetic resonance spectroscopy.15 They studied 9,299 nonsynonymous sequence variations and identified a missense mutation [Ile148Met148 (I148M)] in patatin-like phospholipase domain-containing (PNPLA) 3 gene PNPLA3 (HCNG: 18590). PNPLA3, highly expressed in adipose tissue and liver, is regulated by insulin through a signaling cascade that includes LXR and SREBP-1c28 and, therefore, increased with feeding in animal studies.34 This mutation is, by far, the strongest genetic signal up to date and showed increased odds for fatty liver of 3.26 in the original report.15 In addition, the PNPLA3 gene could also be responsible for the difference in prevalence of fatty liver disease between ethnic groups. For instance, Mexican-Americans have more prevalence of the high risk allele, whereas African Americans, where fatty liver is known to be less frequent, had a protective variation for such.15 Future studies confirmed the association between this gene and the presence of fatty liver disease, including GWAS with liver enzymes17 and multiple case-control studies.35

Chalasani et al. described in 236 white female biopsy-proven NAFLD patients’ five new genetic variants associated with inflammation and fibrosis. They found that the NAFLD activity score (NAS), a pathological tool to measure changes in NAFLD during clinical trials, was associated with the gene FDFT1 (farnesyl diphosphate farnesyl transferase 1, HGNC: 3629); in addition, they found an association with lobular inflammation for the collagen gene COL13A1 (collagen, type XIII, alpha 1, HGNC: 2190), a SNP nearby the PDGFA gene (platelet-derived growth factor alpha polypeptide, HGNC: 8799); the LTBP3 (latent transforming growth factor beta binding protein 3, HGNC: 6716), and the EFCAB4B (EF-hand calcium binding domain 4B, HGNC: 28657).

The Genetics of Obesity-related Liver Disease (GOLD) Consortium is the last genome wide scan published up to date. We obtained the same association with PNPLA3 and four additional genetic variants including the PNPLA3 (HGNC: 18590), namely, NCAN (neurocan, HGNC: 2465), LYPLAL1 (lysophospholipase-like 1, HGNC: 20440); GCKR (glucokinase regulatory protein, HGNC: 4196); and the PPP1R3B (protein phosphatase 1, regulatory subunit 3b, HGNC: 14942). GCKR and PPP1R3B are key enzymes in de novo lipogenesis from glucose; LYPLAL1-related protein has been predicted to play a crucial part in consecutive steps in triglyceride breakdown. The role of NCAN, however, remains to be determined. Interestingly, we found that PNPLA3 and our other genes had a modest role for lipid metabolism suggesting that these genes, if they are involved in lipid metabolism, exert their effects within the liver through different mechanistic pathways than the observed ones by conventional laboratory lipid measurement.

There is no replication up to date of the aforementioned GWAS findings except for the PNPLA3 (rs738409). For this gene variant, a recent meta-analysis updated until January 2011 found an association with the presence of fat accumulation (GG homozygous showed 73% higher lipid fat content when compared with CC ones); 3.2-fold greater risk of higher necroinflammatory scores, 3.5-fold risk of NASH, and 3.2-fold greater risk of developing fibrosis when compared with CC homozygous.35

Future directions: candidate-genes are needed

The typical statement “more research is needed” is clearly shown by the examination of both tables. To better understand the genetic determinants of NAFLD, it is key to replicate prior studies. Whereas GWAS are expensive, require thousands of individuals and strong statistical and population genetics knowledge, the use of candidate gene studies in NAFLD is easy to implement and straightforward. The major caveat for these studies is the lack of power. For instance, assuming a genetic risk ratio of 1.5 for a given polymorphism (high for most of the replicated genetic studies), and a allele frequency about 20%, more than 400 patients and controls are required to give a study 90% power to detect a significant effect at the 5% level.27 Day27 and Bataller23 provide two outstanding reviews to guide the reader in the design of high quality candidate gene studies, step-by-step.

On the other hand, if the researcher is willing to invest his/her time to study new candidate genes, then Day proposes to find them by reviewing: “(1) Gene product considered to play a role in the disease; (2) gene is known to be mutated in a familial form of the disease; (3) gene knockout/overexpression in animal models influences disease development; (4) gene lies in a chromosomal region associated with disease in a linkage study; (5) gene expression is altered in microarray studies of tissue from patients with disease; (6) gene is identified in a phenotype-driven mouse mutagenesis study”27; and (7) gene identify in genome-wide association studies.

In conclusion, genes play a role in the development and progression of NAFLD; PNPLA3 is the strongest signal up to date but there are other numerous genes that have been described but not formally replicated. The future of genetic epidemiology will require replication and, ultimately, expression studies and animal models to know the molecular role of that particular genetic variant. The understanding of genetic determinants of NAFLD will help to identify individuals at risk and, potentially, new therapies to treat the most common chronic liver disease in the world.

Conflict of interest

The author declare no conflict of interest.

Acknowledgement

The author has been supported by the American Diabetes Association Mentor-Based Program (7-07-MN-08, PI: Dr. Frederick L. Brancati).

References
[1]
M. Lazo, J.M. Clark.
The epidemiology of nonalcoholic fatty liver disease: a global perspective.
Semin Liver Dis, 28 (2008), pp. 339-350
[2]
E.M. Brunt.
Nonalcoholic steatohepatitis: definition and pathology.
Semin Liver Dis, 21 (2001), pp. 3-16
[3]
A.J. Sanyal.
AGA technical review on nonalcoholic fatty liver disease.
Gastroenterology, 123 (2002), pp. 1705-1725
[4]
A.K. Daly, S. Ballestri, L. Carulli, P. Loria, C.P. Day.
Genetic determinants of susceptibility and severity in nonalcoholic fatty liver disease.
Expert Rev Gastroenterol Hepatol, 5 (2011), pp. 253-263
[5]
C.P. Day, O.F. James.
Steatohepatitis: a tale of two hits?.
Gastroenterology, 114 (1998), pp. 842-845
[6]
D. Moreno-Sanchez.
Epidemiology and natural history of primary nonalcoholic fatty liver disease.
Gastroenterol Hepatol, 29 (2006), pp. 244-254
[7]
G.C. Farrell, C.Z. Larter.
Nonalcoholic fatty liver disease: from steatosis to cirrhosis.
Hepatology, 43 (2006), pp. S99-S112
[8]
M. Charlton.
Cirrhosis and liver failure in nonalcoholic fatty liver disease: Molehill or mountain?.
Hepatology, 47 (2008), pp. 1431-1433
[9]
L. Caballeria, G. Pera, M.A. Auladell, P. Toran, L. Munoz, D. Miranda, et al.
Prevalence and factors associated with the presence of nonalcoholic fatty liver disease in an adult population in Spain.
Eur J Gastroenterol Hepatol, 22 (2010), pp. 24-32
[10]
M.F. Abdelmalek, C. Liu, J. Shuster, D.R. Nelson, N.R. Asal.
Familial aggregation of insulin resistance in first-degree relatives of patients with nonalcoholic fatty liver disease.
Clin Gastroenterol Hepatol, 4 (2006), pp. 1162-1169
[11]
I.R. Willner, B. Waters, S.R. Patil, A. Reuben, J. Morelli, C.A. Riely.
Ninety patients with nonalcoholic steatohepatitis: insulin resistance, familial tendency, and severity of disease.
Am J Gastroenterol, 96 (2001), pp. 2957-2961
[12]
M.C. Brouwers, M.M. van Greevenbroek, R.M. Cantor.
Heritability of nonalcoholic fatty liver disease.
Gastroenterology, 137 (2009), pp. 1536
[13]
J.B. Schwimmer, M.A. Celedon, J.E. Lavine, R. Salem, N. Campbell, N.J. Schork, et al.
Heritability of nonalcoholic fatty liver disease.
Gastroenterology, 136 (2009), pp. 1585-1592
[14]
N. Chalasani, X. Guo, R. Loomba, M.O. Goodarzi, T. Haritunians, S. Kwon, et al.
Genome-wide association study identifies variants associated with histologic features of nonalcoholic fatty liver disease.
Gastroenterology, 139 (2010), pp. 1567-1576
[15]
S. Romeo, J. Kozlitina, C. Xing, A. Pertsemlidis, D. Cox, L.A. Pennacchio, et al.
Genetic variation in PNPLA3 confers susceptibility to nonalcoholic fatty liver disease.
Nat Genet, 40 (2008), pp. 1461-1465
[16]
E.K. Speliotes, L.M. Yerges-Armstrong, J. Wu, R. Hernaez, L.J. Kim, C.D. Palmer, et al.
Genome-wide association analysis identifies variants associated with nonalcoholic fatty liver disease that have distinct effects on metabolic traits.
PLoS Genet, 7 (2011), pp. e1001324
[17]
X. Yuan, D. Waterworth, J.R. Perry, N. Lim, K. Song, J.C. Chambers, et al.
Population-based genome-wide association studies reveal six loci influencing plasma levels of liver enzymes.
Am J Hum Genet, 83 (2008), pp. 520-528
[18]
F. Caballero, A. Fernandez, A.M. De Lacy, J.C. Fernandez-Checa, J. Caballeria, C. Garcia-Ruiz.
Enhanced free cholesterol, SREBP-2 and StAR expression in human NASH.
J Hepatol, 50 (2009), pp. 789-796
[19]
N. Guillen, M.A. Navarro, C. Arnal, E. Noone, J.M. Rbones-Mainar, S. Acin, et al.
Microarray analysis of hepatic gene expression identifies new genes involved in steatotic liver.
Physiol Genomics, 37 (2009), pp. 187-198
[20]
M.E. Miquilena-Colina, E. Lima-Cabello, S. Sanchez-Campos, M.V. García-Mediavilla, M. Fernández-Bermejo, T. Lozano-Rodríguez, et al.
Hepatic fatty acid translocase CD36 upregulation is associated with insulin resistance, hyperinsulinaemia and increased steatosis in non-alcoholic steatohepatitis and chronic hepatitis C.
Gut, 60 (2011), pp. 1394-1402
[21]
M. Yoneda, H. Endo, H. Mawatari, Y. Nozaki, K. Fujita, T. Akiyama, et al.
Gene expression profiling of non-alcoholic steatohepatitis using gene set enrichment analysis.
Hepatol Res, 38 (2008), pp. 1204-1212
[22]
Z.M. Younossi, A. Baranova, K. Ziegler, G.L. Del, K. Schlauch, T.L. Born, et al.
A genomic and proteomic study of the spectrum of nonalcoholic fatty liver disease.
Hepatology, 42 (2005), pp. 665-674
[23]
R. Bataller, K.E. North, D.A. Brenner.
Genetic polymorphisms and the progression of liver fibrosis: a critical appraisal.
Hepatology, 37 (2003), pp. 493-503
[24]
C.P. Day.
The potential role of genes in nonalcoholic fatty liver disease.
Clin Liver Dis, 8 (2004), pp. 673-691
[25]
C.P. Day.
Genetic and environmental susceptibility to non-alcoholic fatty liver disease.
Dig Dis, 28 (2010), pp. 255-260
[26]
D.T. Thomas.
Statistical methods in genetic epidemiology.
1st ed., Oxford University Press, (2004),
[27]
C.P. Day.
Genetic studies to identify hepatic fibrosis genes and SNPs in human populations.
Methods Mol Med, 117 (2005), pp. 315-331
[28]
J.C. Cohen, J.D. Horton, H.H. Hobbs.
Human fatty liver disease: old questions and new insights.
Science, 332 (2011), pp. 1519-1523
[29]
A.J. Sanyal.
Mechanisms of disease: pathogenesis of nonalcoholic fatty liver disease.
Nat Clin Pract Gastroenterol Hepatol, 2 (2005), pp. 46-53
[30]
K.L. Donnelly, C.I. Smith, S.J. Schwarzenberg, J. Jessurun, M.D. Boldt, E.J. Parks.
Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease.
J Clin Invest, 115 (2005), pp. 1343-1351
[31]
Q.M. Anstee, A.K. Daly, C.P. Day.
Genetics of alcoholic and nonalcoholic fatty liver disease.
Semin Liver Dis, 31 (2011), pp. 128-146
[32]
A.M. Diehl.
Genetic susceptibility to hepatic steatosis.
N Engl J Med, 362 (2010), pp. 1142-1143
[33]
HUGO Gene Nomenclature Committee at the European Bioinformatics Institute.
HUGO Gene Nomenclature Committee (HGNC).
(2011),
[34]
Y. Huang, S. He, J.Z. Li, Y.K. Seo, T.F. Osborne, J.C. Cohen, et al.
A feed-forward loop amplifies nutritional regulation of PNPLA3.
Proc Natl Acad Sci USA, 107 (2010), pp. 7892-7897
[35]
S. Sookoian, C.J. Pirola.
Meta-analysis of the influence of I148M variant of patatin-like phospholipase domain containing 3 gene (PNPLA3) on the susceptibility and histological severity of nonalcoholic fatty liver disease.
Hepatology, 53 (2011), pp. 1883-1894
[36]
Y.J. Zhou, Y.Y. Li, Y.Q. Nie, H. Yang, Q. Zhan, J. Huang, et al.
Influence of polygenetic polymorphisms on the susceptibility to non-alcoholic fatty liver disease of Chinese people.
J Gastroenterol Hepatol, 25 (2010), pp. 772-777
[37]
D.W. Jun, J.H. Han, E.C. Jang, S.H. Kim, S.H. Kim, Y.J. Jo, et al.
Polymorphisms of microsomal triglyceride transfer protein gene and phosphatidylethanolamine N-methyltransferase gene in alcoholic and nonalcoholic fatty liver disease in Koreans.
Eur J Gastroenterol Hepatol, 21 (2009), pp. 667-672
[38]
H. Dong, J. Wang, C. Li, A. Hirose, Y. Nozaki, M. Takahashi, et al.
The phosphatidylethanolamine N-methyltransferase gene V175M single nucleotide polymorphism confers the susceptibility to NASH in Japanese population.
J Hepatol, 46 (2007), pp. 915-920
[39]
J. Song, K.A. da Costa, L.M. Fischer, M. Kohlmeier, L. Kwock, S. Wang, et al.
Polymorphism of the PEMT gene and susceptibility to nonalcoholic fatty liver disease (NAFLD).
FASEB J, 19 (2005), pp. 1266-1271
[40]
L. Carulli, I. Canedi, S. Rondinella, S. Lombardini, D. Ganazzi, S. Fargion, et al.
Genetic polymorphisms in non-alcoholic fatty liver disease: interleukin-6-174G/C polymorphism is associated with non-alcoholic steatohepatitis.
Dig Liver Dis, 41 (2009), pp. 823-828
[41]
G. Musso, R. Gambino, M. Cassader.
Lipoprotein metabolism mediates the association of MTP polymorphism with beta-cell dysfunction in healthy subjects and in nondiabetic normolipidemic patients with nonalcoholic steatohepatitis.
J Nutr Biochem, 21 (2010), pp. 834-840
[42]
C.P. Oliveira, J.T. Stefano, A.M. Cavaleiro, M.A. Zanella Fortes, S.M. Vieira, L. Rodrigues, et al.
Association of polymorphisms of glutamate–cystein ligase and microsomal triglyceride transfer protein genes in non-alcoholic fatty liver disease.
J Gastroenterol Hepatol, 25 (2010), pp. 357-361
[43]
N.M. El-Koofy, H.M. El-Karaksy, I.M. Mandour, G.M. Anwar, M.S. El-Raziky, A.M. El-Hennawy.
Genetic polymorphisms in non-alcoholic fatty liver disease in obese Egyptian children.
Saudi J Gastroenterol, 17 (2011), pp. 265-270
[44]
C. Namikawa, Z. Shu-Ping, J.R. Vyselaar, Y. Nozaki, Y. Nemoto, M. Ono, et al.
Polymorphisms of microsomal triglyceride transfer protein gene and manganese superoxide dismutase gene in non-alcoholic steatohepatitis.
J Hepatol, 40 (2004), pp. 781-786
[45]
M. Hashemi, H. Hoseini, P. Yaghmaei, A. Moazeni-Roodi, A. Bahari, N. Hashemzehi, et al.
Association of polymorphisms in glutamate–cysteine ligase catalytic subunit and microsomal triglyceride transfer protein genes with nonalcoholic fatty liver disease.
DNA Cell Biol, 30 (2011), pp. 569-575
[46]
K.F. Petersen, S. Dufour, A. Hariri, C. Nelson-Williams, J.N. Foo, X.M. Zhang, et al.
Apolipoprotein C3 gene variants in nonalcoholic fatty liver disease.
N Engl J Med, 362 (2010), pp. 1082-1089
[47]
J. Kozlitina, E. Boerwinkle, J.C. Cohen, H.H. Hobbs.
Dissociation between APOC3 variants, hepatic triglyceride content and insulin resistance.
Hepatology, 53 (2011), pp. 467-474
[48]
S. Sookoian, G.O. Castano, A.L. Burgueno, T.F. Gianotti, M.S. Rosselli, C.J. Pirola.
The nuclear receptor PXR gene variants are associated with liver injury in nonalcoholic fatty liver disease.
Pharmacogenet Genomics, 20 (2010), pp. 1-8
[49]
X. Peng, L. Zhang, Q. Wang, X. Cui.
Study on the relationship between FABP2 Ala54Thr polymorphism and the risk of non-alcoholic fatty liver diseases.
Wei Sheng Yan Jiu, 38 (2009), pp. 401-404
[50]
M.L. Salguero, R.E. Leon, A. Santos, S. Roman, J.E. Segura-Ortega, A. Panduro.
The role of FABP2 gene polymorphism in alcoholic cirrhosis.
Hepatol Res, 33 (2005), pp. 306-312
[51]
K. Kantartzis, F. Machicao, J. Machann, F. Schick, A. Fritsche, H.U. Haring, et al.
The DGAT2 gene is a candidate for the dissociation between fatty liver and insulin resistance in humans.
Clin Sci (Lond), 116 (2009), pp. 531-537
[52]
A. Kotronen, H. Yki-Jarvinen, A. Aminoff, R. Bergholm, K.H. Pietilainen, J. Westerbacka, et al.
Genetic variation in the ADIPOR2 gene is associated with liver fat content and its surrogate markers in three independent cohorts.
Eur J Endocrinol, 160 (2009), pp. 593-602
[53]
Y. Nozaki, T. Saibara, Y. Nemoto, M. Ono, N. Akisawa, S. Iwasaki, et al.
Polymorphisms of interleukin-1 beta and beta 3-adrenergic receptor in Japanese patients with nonalcoholic steatohepatitis.
Alcohol Clin Exp Res, 28 (2004), pp. 106S-110S
[54]
N. Iwamoto, Y. Ogawa, S. Kajihara, A. Hisatomi, T. Yasutake, T. Yoshimura, et al.
Gln27Glu beta2-adrenergic receptor variant is associated with hypertriglyceridemia and the development of fatty liver.
Clin Chim Acta, 314 (2001), pp. 85-91
[55]
J. Turnes, M. Hernandez-Guerra, J.G. Abraldes, P. Bellot, R. Oliva, J.C. Garcia-Pagan, et al.
Influence of beta-2 adrenergic receptor gene polymorphism on the hemodynamic response to propranolol in patients with cirrhosis.
Hepatology, 43 (2006), pp. 34-41
[56]
Q. Zhan, Y.Y. Li, Y.Q. Nie, Y.J. Zhou, Y.L. DU, W.H. Sha, et al.
Association of hepatic lipase gene promoter polymorphism −514C/T with nonalcoholic fatty liver disease.
Zhonghua Gan Zang Bing Za Zhi, 16 (2008), pp. 375-378
[57]
M.D. Demirag, H.I. Onen, M.Y. Karaoguz, I. Dogan, T. Karakan, A. Ekmekci, et al.
Apolipoprotein E gene polymorphism in nonalcoholic fatty liver disease.
Dig Dis Sci, 52 (2007), pp. 3399-3403
[58]
D.M. Lee, S.O. Lee, B.S. Mun, H.S. Ahn, H.Y. Park, H.S. Lee, et al.
Relation of apolipoprotein E polymorphism to clinically diagnosed fatty liver disease.
Taehan Kan Hakhoe Chi, 8 (2002), pp. 355-362
[59]
A. Sazci, G. Akpinar, C. Aygun, E. Ergul, O. Senturk, S. Hulagu.
Association of apolipoprotein E polymorphisms in patients with non-alcoholic steatohepatitis.
Dig Dis Sci, 53 (2008), pp. 3218-3224
[60]
M.H. Yang, H.J. Son, J.D. Sung, Y.H. Choi, K.C. Koh, B.C. Yoo, et al.
The relationship between apolipoprotein E polymorphism, lipoprotein (a) and fatty liver disease.
Hepatogastroenterology, 52 (2005), pp. 1832-1835
[61]
C. Fabris, C. Vandelli, P. Toniutto, R. Minisini, C. Colletta, E. Falleti, et al.
Apolipoprotein E genotypes modulate fibrosis progression in patients with chronic hepatitis C and persistently normal transaminases.
J Gastroenterol Hepatol, 26 (2011), pp. 328-333
[62]
S. Sookoian, G. Castano, C. Gemma, T.F. Gianotti, C.J. Pirola.
Common genetic variations in CLOCK transcription factor are associated with nonalcoholic fatty liver disease.
World J Gastroenterol, 13 (2007), pp. 4242-4248
[63]
P. Dongiovanni, L. Valenti, R. Rametta, A.K. Daly, V. Nobili, E. Mozzi, et al.
Genetic variants regulating insulin receptor signalling are associated with the severity of liver damage in patients with non-alcoholic fatty liver disease.
[64]
K. Tokushige, E. Hashimoto, H. Noto, S. Yatsuji, M. Taniai, N. Torii, et al.
Influence of adiponectin gene polymorphisms in Japanese patients with non-alcoholic fatty liver disease.
J Gastroenterol, 44 (2009), pp. 976-982
[65]
Z.L. Wang, B. Xia, U. Shrestha, L. Jiang, C.W. Ma, Q. Chen, et al.
Correlation between adiponectin polymorphisms and non-alcoholic fatty liver disease with or without metabolic syndrome in Chinese population.
J Endocrinol Invest, 31 (2008), pp. 1086-1091
[66]
G. Musso, R. Gambino, M.F. De, M. Durazzo, G. Pagano, M. Cassader.
Adiponectin gene polymorphisms modulate acute adiponectin response to dietary fat: Possible pathogenetic role in NASH.
Hepatology, 47 (2008), pp. 1167-1177
[67]
V.W. Wong, G.L. Wong, S.W. Tsang, A.Y. Hui, A.W. Chan, P.C. Choi, et al.
Genetic polymorphisms of adiponectin and tumor necrosis factor-alpha and nonalcoholic fatty liver disease in Chinese people.
J Gastroenterol Hepatol, 23 (2008), pp. 914-921
[68]
P. Dongiovanni, R. Rametta, A.L. Fracanzani, L. Benedan, V. Borroni, P. Maggioni, et al.
Lack of association between peroxisome proliferator-activated receptors alpha and gamma2 polymorphisms and progressive liver damage in patients with non-alcoholic fatty liver disease: a case control study.
BMC Gastroenterol, 10 (2010), pp. 102
[69]
S. Chen, Y. Li, S. Li, C. Yu.
A Val227Ala substitution in the peroxisome proliferator activated receptor alpha (PPAR alpha) gene associated with non-alcoholic fatty liver disease and decreased waist circumference and waist-to-hip ratio.
J Gastroenterol Hepatol, 23 (2008), pp. 1415-1418
[70]
A.C. Gupta, A.K. Chaudhory, Sukriti., C. Pande, P. Sakhuja, Y. Singh, et al.
Peroxisome proliferators-activated receptor gamma2 Pro12Ala variant is associated with body mass index in non-alcoholic fatty liver disease patients.
Hepatol Int, 5 (2010), pp. 575-580
[71]
J.W. Rey, A. Noetel, A. Hardt, A. Canbay, H. Alakus, H.A. Zur, et al.
Pro12Ala polymorphism of the peroxisome proliferator-activated receptor gamma2 in patients with fatty liver diseases.
World J Gastroenterol, 16 (2010), pp. 5830-5837
[72]
G. Musso, R. Gambino, G. Pacini, G. Pagano, M. Durazzo, M. Cassader.
Transcription factor 7-like 2 polymorphism modulates glucose and lipid homeostasis, adipokine profile, and hepatocyte apoptosis in NASH.
Hepatology, 49 (2009), pp. 426-435
[73]
Z. Yang, J. Wen, X. Tao, B. Lu, Y. Du, M. Wang, et al.
Genetic variation in the GCKR gene is associated with non-alcoholic fatty liver disease in Chinese people.
Mol Biol Rep, 38 (2011), pp. 1145-1150
[74]
A. Haupt, C. Thamer, M. Heni, O. Tschritter, J. Machann, F. Schick, et al.
Impact of variation near MC4R on whole-body fat distribution, liver fat, and weight loss.
Obesity (Silver Spring), 17 (2009), pp. 1942-1945
[75]
N. Oruc, O. Ozutemiz, A. Berdeli, G. Ersoz, F. Gunsar, Z. Karasu, et al.
Common SPINK-1 mutations do not predispose to the development of non-alcoholic fatty liver disease.
Ann Hepatol, 8 (2009), pp. 116-119
[76]
H. Lu, J. Sun, L. Sun, X. Shu, Y. Xu, D. Xie.
Polymorphism of human leptin receptor gene is associated with type 2 diabetic patients complicated with non-alcoholic fatty liver disease in China.
J Gastroenterol Hepatol, 24 (2009), pp. 228-232
[77]
R. Aller, D.A. De Luis, O. Izaola, S.M. Gonzalez, R. Conde, G.T. Alvarez, et al.
G308A polymorphism of TNF-alpha gene is associated with insulin resistance and histological changes in non alcoholic fatty liver disease patients.
Ann Hepatol, 9 (2010), pp. 439-444
[78]
Z.W. Hu, H.B. Luo, Y.M. Xu, J.W. Guo, X.L. Deng, Y.W. Tong, et al.
Tumor necrosis factor-alpha gene promoter polymorphisms in Chinese patients with nonalcoholic fatty liver diseases.
Acta Gastroenterol Belg, 72 (2009), pp. 215-221
[79]
K. Tokushige, M. Takakura, N. Tsuchiya-Matsushita, M. Taniai, E. Hashimoto, K. Shiratori.
Influence of TNF gene polymorphisms in Japanese patients with NASH and simple steatosis.
J Hepatol, 46 (2007), pp. 1104-1110
[80]
L. Valenti, A.L. Fracanzani, P. Dongiovanni, G. Santorelli, A. Branchi, E. Taioli, et al.
Tumor necrosis factor alpha promoter polymorphisms and insulin resistance in nonalcoholic fatty liver disease.
Gastroenterology, 122 (2002), pp. 274-280
[81]
D. Sanchez-Munoz, M. Romero-Gomez, M.F. Gonzalez-Escribano, B. Torres, V.M. Castellano-Megias, L. Gomez-Izquierdo, et al.
Tumour necrosis factor alpha polymorphisms are not involved in the development of steatosis in chronic hepatitis C.
Eur J Gastroenterol Hepatol, 16 (2004), pp. 761-765
[82]
I.J. Pastor, F.J. Laso, A. Romero, R. Gonzalez-Sarmiento.
−238 G>A polymorphism of tumor necrosis factor alpha gene (TNFA) is associated with alcoholic liver cirrhosis in alcoholic Spanish men.
Alcohol Clin Exp Res, 29 (2005), pp. 1928-1931
[83]
X. Yan, L. Xu, J. Qi, X. Liang, C. Ma, C. Guo, et al.
sTRAIL levels and TRAIL gene polymorphisms in Chinese patients with fatty liver disease.
Immunogenetics, 61 (2009), pp. 551-556
[84]
L. Giannitrapani, M. Soresi, A. Giacalone, M.E. Campagna, M. Marasa, M. Cervello, et al.
IL-6 −174G/C polymorphism and IL-6 serum levels in patients with liver cirrhosis and hepatocellular carcinoma.
OMICS, 15 (2011), pp. 183-186
[85]
E. Falleti, C. Fabris, P. Toniutto, E. Fontanini, A. Cussigh, D. Bitetto, et al.
Interleukin-6 polymorphisms and gender: relationship with the occurrence of hepatocellular carcinoma in patients with end-stage liver disease.
Oncology, 77 (2009), pp. 304-313
[86]
P. Brun, I. Castagliuolo, A.R. Floreani, A. Buda, L. Blasone, G. Palu, et al.
Increased risk of NASH in patients carrying the C(−159)T polymorphism in the CD14 gene promoter region.
[87]
E. Askar, G. Ramadori, S. Mihm.
Endotoxin receptor CD14 gene variants and histological features in chronic HCV infection.
World J Gastroenterol, 15 (2009), pp. 3884-3890
[88]
A. Al-Serri, Q.M. Anstee, L. Valenti, V. Nobili, J.B. Leathart, P. Dongiovanni, et al.
The sod2 c47t polymorphism influences nafld fibrosis severity: evidence from case–control and intra-familial allele association studies.
J Hepatol, (2011),
[89]
R. Hernaez, E. Yeung, J.M. Clark, K.V. Kowdley, F.L. Brancati, W.H. Kao.
Hemochromatosis gene and nonalcoholic fatty liver disease: a systematic review and meta-analysis.
J Hepatol, (2011),
[90]
Y.C. Lin, P.F. Chang, F.C. Hu, M.H. Chang, Y.H. Ni.
Variants in the UGT1A1 gene and the risk of pediatric nonalcoholic fatty liver disease.
Pediatrics, 124 (2009), pp. e1221-e1227
[91]
R. Aller, D.A. De Luis, O. Izaola, S.M. Gonzalez, R. Conde, T. Alvarez, et al.
Role of −55CT polymorphism of UCP3 gene on non alcoholic fatty liver disease and insulin resistance in patients with obesity.
Nutr Hosp, 25 (2010), pp. 572-576
[92]
G. Labruna, F. Pasanisi, C. Nardelli, G. Tarantino, D.F. Vitale, R. Bracale, et al.
UCP1 −3826 AG+GG genotypes, adiponectin, and leptin/adiponectin ratio in severe obesity.
J Endocrinol Invest, 32 (2009), pp. 525-529
[93]
M.B. Cao, Y.X. Yang, L. Dong.
Relationship between single nucleotide polymorphisms in the promoter of COX-2 gene and hereditariness to NAFLD.
Zhonghua Gan Zang Bing Za Zhi, 18 (2010), pp. 773-777
[94]
R. Iwata, K. Baur, B. Stieger, J.C. Mertens, A.K. Daly, P. Frei, et al.
A common polymorphism in the ABCB11 gene is associated with advanced fibrosis in hepatitis C but not in non-alcoholic fatty liver disease.
Clin Sci (Lond), 120 (2011), pp. 287-296
[95]
M. Akyildiz, F. Gunsar, D. Nart, O. Sahin, F. Yilmaz, S. Akay, et al.
Macrophage migration inhibitory factor expression and MIF gene −173 G/C polymorphism in nonalcoholic fatty liver disease.
Eur J Gastroenterol Hepatol, 22 (2010), pp. 192-198
[96]
Y.F. Piao, J.T. Li, Y. Shi.
Relationship between genetic polymorphism of cytochrome P450IIE1 and fatty liver.
World J Gastroenterol, 9 (2003), pp. 2612-2615
[97]
N.M. Varela, L.A. Quinones, M. Orellana, J. Poniachik, A. Csendes, G. Smok, et al.
Study of cytochrome P450 2E1 and its allele variants in liver injury of nondiabetic, nonalcoholic steatohepatitis obese women.
Biol Res, 41 (2008), pp. 81-92
[98]
L. Valenti, P. Dongiovanni, A. Piperno, A.L. Fracanzani, M. Maggioni, R. Rametta, et al.
Alpha 1-antitrypsin mutations in NAFLD: high prevalence and association with altered iron metabolism but not with liver damage.
Hepatology, 44 (2006), pp. 857-864
[99]
A. Sazci, E. Ergul, C. Aygun, G. Akpinar, O. Senturk, S. Hulagu.
Methylenetetrahydrofolate reductase gene polymorphisms in patients with nonalcoholic steatohepatitis (NASH).
Cell Biochem Funct, 26 (2008), pp. 291-296
[100]
C. Fabris, P. Toniutto, E. Falleti, E. Fontanini, A. Cussigh, D. Bitetto, et al.
MTHFR C677T polymorphism and risk of HCC in patients with liver cirrhosis: role of male gender and alcohol consumption.
Alcohol Clin Exp Res, 33 (2009), pp. 102-107
[101]
S. Sookoian, G. Castano, T.F. Gianotti, C. Gemma, C.J. Pirola.
Polymorphisms of MRP2 (ABCC2) are associated with susceptibility to nonalcoholic fatty liver disease.
J Nutr Biochem, 20 (2009), pp. 765-770
[102]
S. Sookoian, G. Castano, T.F. Gianotti, C. Gemma, M.S. Rosselli, C.J. Pirola.
Genetic variants in STAT3 are associated with nonalcoholic fatty liver disease.
Cytokine, 44 (2008), pp. 201-206
[103]
T. Nakagawa, Y. Muramoto, M. Hori, S. Mihara, T. Marubayashi, K. Nakagawa.
A preliminary investigation of the association between haptoglobin polymorphism, serum ferritin concentration and fatty liver disease.
Clin Chim Acta, 398 (2008), pp. 34-38
[104]
M. Yoneda, K. Hotta, Y. Nozaki, H. Endo, T. Uchiyama, H. Mawatari, et al.
Association between angiotensin II type 1 receptor polymorphisms and the occurrence of nonalcoholic fatty liver disease.
Liver Int, 29 (2009), pp. 1078-1085
[105]
L. Miele, G. Beale, G. Patman, V. Nobili, J. Leathart, A. Grieco, et al.
The Kruppel-like factor 6 genotype is associated with fibrosis in nonalcoholic fatty liver disease.
Gastroenterology, 135 (2008), pp. 282-291
Copyright © 2011. Elsevier España, S.L.. All rights reserved
Opciones de artículo
Herramientas
es en pt

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?

Você é um profissional de saúde habilitado a prescrever ou dispensar medicamentos