Next Article in Journal
Peripheral Blood Mononuclear Cell Metabolism Acutely Adapted to Postprandial Transition and Mainly Reflected Metabolic Adipose Tissue Adaptations to a High-Fat Diet in Minipigs
Next Article in Special Issue
Sugar Responses of Human Enterochromaffin Cells Depend on Gut Region, Sex, and Body Mass
Previous Article in Journal
Lipoprotein Subclass Profile after Progressive Energy Deficits Induced by Calorie Restriction or Exercise
Previous Article in Special Issue
Eight-Week Consumption of High-Sucrose Diet Has a Pro-Oxidant Effect and Alters the Function of the Salivary Glands of Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Copper-Fructose Interactions: A Novel Mechanism in the Pathogenesis of NAFLD

1
Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA
2
Hepatobiology&Toxicology Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
3
Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30307, USA
4
Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
5
Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA
6
University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
7
Robley Rex Veterans Affairs Medical Center, Louisville, KY 40206, USA
*
Author to whom correspondence should be addressed.
Nutrients 2018, 10(11), 1815; https://doi.org/10.3390/nu10111815
Submission received: 18 October 2018 / Revised: 8 November 2018 / Accepted: 16 November 2018 / Published: 21 November 2018
(This article belongs to the Special Issue Fructose and Glucose for Human Health)

Abstract

:
Compelling epidemiologic data support the critical role of dietary fructose in the epidemic of obesity, metabolic syndrome and nonalcoholic fatty liver disease (NAFLD). The metabolic effects of fructose on the development of metabolic syndrome and NAFLD are not completely understood. High fructose intake impairs copper status, and copper-fructose interactions have been well documented in rats. Altered copper-fructose metabolism leads to exacerbated experimental metabolic syndrome and NAFLD. A growing body of evidence has demonstrated that copper levels are low in NAFLD patients. Moreover, hepatic and serum copper levels are inversely correlated with the severity of NAFLD. Thus, high fructose consumption and low copper availability are considered two important risk factors in NAFLD. However, the causal effect of copper-fructose interactions as well as the effects of fructose intake on copper status remain to be evaluated in humans. The aim of this review is to summarize the role of copper-fructose interactions in the pathogenesis of the metabolic syndrome and discuss the potential underlying mechanisms. This review will shed light on the role of copper homeostasis and high fructose intake and point to copper-fructose interactions as novel mechanisms in the fructose induced NAFLD.

1. Introduction

Accumulating evidence has shown that increased fructose consumption parallels the rises in the obesity epidemic, metabolic syndrome and NAFLD in the United States and worldwide [1,2,3,4,5,6,7,8]. Moreover, fructose consumption is higher in patients with NAFLD compared to healthy controls and is associated with severity of fibrosis, suggesting that high fructose intake may be an important risk factor for the development and progression of NAFLD [9,10,11,12]. Fructose is consumed mainly as added sugars, such as sucrose and high-fructose corn syrup (HFCS), which represents 45% and 41% of the total added sugars ingested, respectively [13].
Fructose is distinct from glucose due to its unique metabolism [14] and limited absorption [15,16]. The role of fructose in the induction of components of metabolic syndrome, as well as NAFLD, has been well documented in numerous animal studies [17,18,19,20]. A recent study demonstrated dietary fructose is primarily metabolized in the small intestine. However, excess fructose ingestion can saturate intestinal clearance capacity; it then reaches the liver and colon microbiota where it is metabolized [21]. In line with these data, depletion or knockdown of ketohexokinase (KHK), a key enzyme of fructose metabolism, markedly attenuated high fructose diet-induced NAFLD, obesity and other metabolic effects [22,23,24]. Similarly, toll like receptor 4 (TLR4) mutation or oral antibiotics protected against high fructose diet-induced NAFLD in mice [17,25], highlighting the importance of the gut-liver axis in the pathogenesis of dietary fructose associated NAFLD and metabolic syndrome. Despite the major progress that has been made over the past two decades, the mechanisms underlying fructose-induced NAFLD and metabolic syndrome are still incompletely understood. Even with increased de novo lipogenesis, only a small amount of fructose (<1%) ingested is converted to plasma triglyceride [26]. Thus, it was proposed that the general activation of lipogenesis and blockade of fatty acid oxidation signaling might account for the fructose induced fat accumulation in the liver [27]. However, a gap still remains in our understanding of increased de novo lipogenesis and hepatic fat accumulation during ingestion of fructose.
Although compelling epidemiologic data support the critical role of dietary fructose in the epidemic of the metabolic syndrome [2,8,28,29,30,31], a causal link between fructose consumption and the metabolic syndrome has not been firmly established in human studies [32,33,34,35,36]. Most of studies on fructose are limited by a short term of study and/or a small study population. Moreover, hypercaloric effects cannot be excluded in some of the studies [37]. Of note, isocaloric dietary fructose restriction has been reported to be beneficial in improving obesity and metabolic parameters [38,39,40]. Animal studies also showed the essential role of fructose in the methionine-choline-deficient (MCD) diet-induced nonalcoholic steatohepatitis (NASH) model [41,42]. Thus, another key issue is the complexity of the etiology of NAFLD, which involves multiple metabolic effects. One crucial factor is the nutrition interactions. Results from animal studies demonstrated that when fructose is ingested with fat, more severe hepatic steatosis and liver injury or fibrosis was induced compared to high fat diet alone, even when consumed isocalorically [43]. Similarly, the metabolic effects of fructose are more obvious in obese patients with NAFLD and insulin resistance [12,32,44,45,46], suggesting potential interactions between fructose and other metabolites or a complex interplay with other metabolic pathways. We and others have demonstrated copper-fructose interactions in inducing the components of metabolic syndrome and NAFLD in rat model [47,48,49,50,51].
A growing body of evidence indicates that hepatic copper level is lower in NAFLD patients, and steatosis grades inversely correlate with hepatic copper content [52,53,54,55,56]. Moreover, dietary copper restriction induces hepatic steatosis and insulin resistance in rats, suggesting that copper availability may be involved in the development of NAFLD [52]. The mechanism leading to low copper levels in NAFLD patients is not clear. Multiple factors can lead to copper deficiency, including the amount of copper in the diet. The Western diet often is low in copper [57,58]. Other factors, including bariatric surgery, pancreatoduodenectomy, excessive use of denture cream high in zinc and excessive intake of soft drinks, with added fructose, can also induce copper deficiency by impairing copper absorption [59,60,61,62,63,64,65,66,67].
Collectively, high fructose consumption and inadequate copper intake represent two important nutritional problems in the United States. Although copper-fructose interactions have been well documented in experimental models [48,49,50,51,68,69,70,71,72], limited data are available from human studies. In this review, we will discuss the role of copper-fructose interactions in the pathogenesis of the metabolic syndrome and NAFLD and discuss potential underlying mechanisms.

2. Epidemiology of NAFLD, Fructose Consumption, and Dietary Copper Intake

NAFLD is now the most common liver disease in the United States, and accounts for more than 75% of chronic liver diseases. In contrast with the other chronic liver diseases, whose prevalence has remained stable, the prevalence of NAFLD increased steadily from 5.51% (1988–1994) to 9.84% (1999–2004) to 11.01% (2005–2008) [73]. The most recent study showed that global prevalence of NAFLD is 25.24%, and is 24.13% in North America [8]. The increased prevalence of NAFLD parallels the increases in obesity, type 2 diabetes (T2D), insulin resistance and hypertension [8,73]—all hallmarks of the metabolic syndrome. In particular, the prevalence of suspected NAFLD in adolescents increased at an alarming rate, from 3.9% to 10.7% over the past 20 years [7]. NASH, a more advanced stage of NAFLD, is the third most common indication for liver transplantation in the United States [74]. Of note, NAFLD and NASH exhibit age and sex differences, with a higher prevalence in males than in females in both adolescents and adults until the age of 60. After age 60, the prevalence of NASH is higher in women [75,76]. Based on NHANES III (1988–1994) data, mean fructose consumption was 54.7 g per day and accounted for 10.2% of total caloric intake [4], about 50% higher than the mean reported from the 1970’s (37g/day) [5]. Consumption was highest among adolescents at 72.8 g/day (12.1 % of total calories) [4]. Although evidence showed that consumption of sugar-sweetened beverages (SSBs), which are the leading source of added sugars in the diet, has declined recently [77,78], it remains high among children and adolescents [79]. Moreover, time-trend data over the past 3 decades have shown that the increased consumption of SSBs parallels the obesity epidemic and is associated with increased T2D risk in the United States [2]. The prevalence of obesity increased from approximately 5% (early 1970’s) to 17% (2011–2014) in children and adolescents, and from 15% (late 1970’s) to 36.5% (2011–2014) in adults in the United States [80]. The rise of SSBs intake is mainly due to the dramatic increased consumption of HFCS, which is the primary sweetener in SSBs [2,13]. The most common forms of HFCS contain either 42% (HFCS-42) or 55% (HFCS-55) fructose, along with glucose and water, with HFCS-55 being the most common used form [13,81]. Therefore, SSBs appear to be the major source of dietary fructose. It was estimated that 184,000 global deaths in 2010 were attributable to consumption of sugary beverages, with 72.3% from diabetes mellitus, 24.2% from cardiovascular diseases (CVDs) and 3.5% from cancer. United States is ranked second in SSB-related mortality among the 20 most populous countries in the world [82]. Accordingly, Dietary Guidelines for Americans recommends decreasing added sugars from 25% (2010-2015) to less than 10% (2015–2020) of calories per day [31,83].
The Recommended Dietary Allowance (RDA) for copper in adult men and women is 0.9 mg/day and the Estimated Average Requirement (EAR) for copper is 0.7 mg /day [84]. Klevay summarized the data from NHANES III and found that at least one fourth of adults consume less than the EAR in both the United States and Canada [59,85]. A recent study revealed that 62% and 36% of diets of 80 randomly selected adults in Baltimore were below the RDA and EAR, respectively [86], suggesting that the Western diet is often low in copper. Secondary copper deficiency can be caused by factors such as gastric bypass surgery and high zinc exposure [87,88].
Copper status is affected by age, gender and hormone use. Plasma copper concentrations and ceruloplasmin levels were higher in women than in men [89]. Lack of good biomarkers make it challenging to monitor marginal copper deficiency status [88]. The copper-containing enzyme activities in blood cells, such as erythrocyte copper/zinc-superoxide dismutase (SOD1) and platelet or leukocyte cytochrome c oxidase (COX), are sensitive to changes in copper stores and are better indicators of copper status than plasma copper level and ceruloplasmin activity [89,90]. A recent study by Heffern et al. revealed that Copper-Caged Luciferin-1(CCL-1), a bioluminescent reporter, can be used for tissue-specific copper in vivo imaging, thus to monitor copper levels in living animals. They found that hepatic copper levels were markedly decreased in diet-induced NAFLD mice model [91].

3. Fructose Absorption, Metabolism and Metabolic Fate

Dietary fructose is absorbed in the small intestine, and the absorption of fructose is greater in the proximal and middle than in the distal small intestine [92]. A murine study with oral gavage of low dose 13C-fructose demonstrated that labeled fructose 1-phosphate (F1P), a specific metabolite of fructose, predominantly localized in the small intestine (jejunum ˃ duodenum ˃ ileum). When high dose of 13C-fructose was orally gavaged to mice, the majority of labeled F1P was detected in the jejunum and only a small amount of labeled F1P accumulated in the liver, suggesting that most of ingested fructose is metabolized in the small intestine and only excess fructose intake flows to the liver [21]. Fructose is absorbed into enterocytes by the fructose transporter GLUT5 in the apical membrane and exits to the portal blood via GLUT2 in the basolateral membrane of enterocytes. The absorption of fructose in the liver is mediated by GLUT2 [93]. Recent studies demonstrated that GLUT8 also play an important role in the hepatic and intestinal fructose absorption. Moreover, GLUT8 mediated fructose absorption exhibits sex differences [94,95,96]. Of note, the distribution of GLUT2 and GLUT8 in the liver as shown by mRNA abundance is very similar between humans and mice [95]. GLUT5 is mainly expressed in the small intestine and kidney, while the major sites of GLUT2 expression are the liver, pancreas, intestine, kidney, and brain. The distribution of GLUT5 in the small intestine exhibits a regional pattern which is greater in the proximal segment compared to the distal segment [97]. Moreover, GLUT5 is inducible and dramatically stimulated by early introduction of dietary fructose [97]. Glut5 deletion resulted in more than a 75% reduction in fructose absorption in the small intestine and a decrease of 90% of serum fructose concentration compared to wild-type mice [98]. In addition, GLUT5 gene expression is tightly regulated by glucocorticoid and thyroid hormones and circadian rhythm [97,99].
Fructose is metabolized by KHK or fructokinase. [23,100,101]. Fructose is phosphorylated by KHK at the 1-position to generate F1P, which consumes ATP and phosphate. F1P is then cleaved to glyceraldehyde and dihydroxyacetone phosphate (DHAP) by aldolase B. At this point, glucose metabolism and fructose metabolism converge. Unlike glycolysis, fructolysis bypasses phosphofructokinase, a rate-limiting step in glycolysis, to circumvent feedback inhibition. Moreover, KHK is much faster than hexokinase in phosphorylating their substrates [14], thus leading to rapid ATP depletion and phosphate consumption [102]. There are two KHK isoforms, KHK-C and -A, and both can metabolize fructose, but KHK-C is considered the primary enzyme involved in fructose metabolism because of its lower Michaelis constant (Km) [23,101]. Depletion of KHK-C or KHK-A and -C, but not KHK-A alone, protects against fructose-induced metabolic syndrome [23,103,104]. A reduction of intracellular phosphate leads to the activation of adenosine monophosphate deaminase (AMPD), which converts AMP to inosine monophosphate (IMP). IMP is subsequently converted to hypoxanthine and then to xanthine, ultimately leading to the generation of uric acid [105,106,107] (Figure 1). Inhibition of xanthine oxidase (XO), a rate-limiting enzyme that catalyzes uric acid production, protects against hepatic steatosis in mice [108].
The metabolic fate of fructose has been shown by studies with an isotope tracer. After ingestion of fructose, approximately 50% is converted into glucose, 25% is converted into lactate, 17% is converted to glycogen, and only less than 1% is converted to plasma triglyceride. However, most of the tracer studies are short-term studies. Longer term effects of fructose on the de novo lipogenesis need to be evaluated [13,26]. It appears that fructose-induced fatty liver is unlikely the direct effect of fructose metabolism. This leads to the hypothesis that the activation of lipogenesis and blockade of fatty acid oxidation signaling might account for the hepatic steatosis induced by fructose metabolism [27]. Overall, knowing the fundamentals of fructose biochemistry is crucial for the understanding of fructose induced metabolic disorders.
Recently, work from animal studies demonstrated that endogenous fructose, generated from polyol pathway, plays a critical role in the development of metabolic syndrome and NASH (in addition to dietary fructose) [22,104]. The polyol pathway is an alternate route of glucose metabolism. The rate-limiting step of this polyol pathway is the reduction of glucose to sorbitol which is catalyzed by aldose reductase (AR). Under normoglycemia, AR-catalyzed reduction is less than 3% of total glucose utilization, whereas more than 30% glucose is used by AR under hyperglycemia [109,110].
The causal role of fructose in the pathogenesis of NAFLD has been demonstrated in numerous animal studies [17,25,111,112] and has been reviewed [113,114,115,116]. In this review, we will focus on the copper-fructose interactions and NAFLD.

4. Copper Absorption, Distribution and Utilization

Copper is an essential trace element. It serves as a cofactor for a number of enzymes, including COX, SOD1, ceruloplasmin, hephaestin, and lysyl oxidase, etc., which are involved in mitochondrial respiration, antioxidant defense, copper and iron export, connective tissue maturation, etc. [117]. In addition, copper also functions as a cellular signal to regulate cellular and molecular events, such as proteasome degradation of copper chaperone for SOD1 (CCS) and hypoxia inducible factor-1 (HIF-1) activation [118,119]. Mammals acquire copper through the diet. Copper absorption, distribution, and utilization are tightly regulated to maintain copper homeostasis. Dietary copper is primarily absorbed from the small intestine via copper transporter 1 (Ctr1). Ctr1 is considered the major copper transporter in mammalian cells [120,121,122]. Mice with intestinal-specific deletion of Ctr1 died of severe weight loss within three weeks, and these mice can be rescued by a single intraperitoneal injection of copper [123]. Similarly, cardiac-specific knockout of the Ctr1 results in cardiac copper deficiency and severe cardiomyopathy [124], suggesting that Ctr1 is required for copper absorption. Ctr2 was originally identified based on its sequence homology to Ctr1. However, Ctr1 and Ctr2 exhibit distinct functions in copper metabolism. Ctr2 knockout mice exhibit increased tissue copper levels and are defective in accumulation of truncated Ctr1. Thus, Ctr2 plays a significant role in Ctr1 degradation and functions as a regulator of Ctr1 [125,126]. In human adults, the amount of copper absorption is inversely correlated with dietary copper intake; high dietary copper intake results in low copper absorption [127]. After import, the copper ion in the cytoplasm is stored either in a complex with metallothioneins (MT) mediated by GSH, or is distributed to proteins or organelles by specific Cu chaperone proteins that function in the delivery of Cu to mitochondrial COX (via Cox17), to SOD1 (via CCS), and to the cytosolic Cu binding domain of the P-type Cu-transporting ATPases, ATP7A or ATP7B (via Atox1) [128]. ATP7A and ATP7B are required for transport of copper into the trans-Golgi network (TGN) for biosynthesis of several secreted cuproenzymes and for efflux of copper. ATP7A is required for copper efflux in the intestine and ATP7B is required for the biliary excretion of excess copper in the liver [128] (Figure 2).

5. Copper Homeostasis and NAFLD

Disturbance of copper homeostasis is associated with a variety of clinical manifestations. In this review, we focus on the copper dyshomeostasis associated NAFLD and its risk factors.
Analysis of 124 adult biopsy-proven NAFLD patients revealed that serum copper as well as liver copper levels are lower compared to healthy controls and patients with other types of liver diseases, including hepatitis C virus (HCV) infection, autoimmune hepatitis, and alcoholic liver disease. Among these NAFLD patients, NASH patients displayed even lower hepatic copper levels than those with simple steatosis. Hepatic copper level is lower in NAFLD patients with the metabolic syndrome and T2D compared to those without metabolic syndrome and T2D [52]. Moreover, NAFLD patients with lower serum copper and lower liver copper exhibited higher serum ferritin levels and hepatic iron levels, which were associated with decreased mRNA expression of liver ferroportin-1 (FP-1) [53]. Similar results were obtained from pediatric NAFLD patients [54,56]. More severe NAFLD (NAFLD activity score, NAS ≥5) patients, particularly in those with ballooning hepatocytes, displayed significantly lower serum copper and ceruloplasmin levels compared to the patients with less severe NAFLD (NAS ˂ 5) [56]. A recent study from 751 Korean adults revealed that lower hair copper concentration was associated with higher body mass index, waist circumference, blood pressure, and lower levels of high-density lipoprotein cholesterol. Of note, NAFLD patients displayed significantly lower hair copper concentrations [129]. Moreover, dietary copper restriction induces hepatic steatosis and insulin resistance in rats, further suggesting that copper availability may be involved in the development of NAFLD [52].
Mutations in the ATP7B gene leads to Wilson’s disease (WD), an inherited autosomal recessive disorder of copper dyshomeostasis, characterized by excessive hepatic copper accumulation and decreased serum ceruloplasmin levels. In the earlier stage, it manifests as hepatic steatosis which is often indistinguishable from NAFLD [130]; it may progress to hepatic fibrosis and cirrhosis, and eventually liver failure [131]. The mechanism(s) by which hepatic copper accumulation leads to hepatic steatosis are not clear, but likely involve mitochondrial damage [132], global DNA hypomethylation [133], and/or nuclear receptors [134]. In addition to WD, evidence from animal studies indicates a critical role of copper homeostasis in the pathogenesis of liver fibrosis. Bile duct ligation results in copper accumulation in the liver. High copper levels were also observed in the late stage of NAFLD patients, including cirrhosis and hepatocellular carcinoma [135]. Treatment with a copper chelator protects against bile duct ligation-induced liver fibrosis. However, overdose of copper chelator results in copper deficiency and accentuates liver injury and fibrosis [136,137]. Thus, both copper deficiency and excess may lead to hepatic steatosis and, in some cases, more severe distinct liver pathology. The relationship between copper and NAFLD has recently been reviewed [138,139].

6. Copper-Fructose Interactions.

Extensive studies in 1980’s demonstrated that dietary copper-fructose interactions worsened copper deficiency-induced metabolic syndrome. The severity of experimental copper deficiency was exacerbated by a diet containing high fructose compared to animals with diets containing high glucose or starch [50,51,69,70,71], and this was characterized by lower body weight and hematocrit, and increased liver weight, blood urea nitrogen, ammonia, cholesterol and triglycerides. Switching the type of dietary carbohydrate from fructose to either starch or glucose ameliorated the severity of copper deficiency [50]. In line with animal studies, a human study demonstrated that adult males displayed significantly reduced SOD1 activity in erythrocytes after consumption of a low copper (1.03 mg/day/2850 kcal) and high fructose (20% calorie) diet for 11 weeks compared to those who consumed diets with low copper and starch [60], suggesting dietary fructose intake can affect indices of copper status.

6.1. Copper-Fructose Interaction and NAFLD

Our studies demonstrated that dietary high fructose intake further impaired copper status and exacerbated liver injury and fat accumulation in marginally copper deficient rats (Figure 3) [48]. Similar results were obtained in rats fed with high sucrose and copper deficient diet [140]. Moreover, we found that not only high dietary fructose (30% (w/v) fructose in the drinking water) impairs copper status, but also modest fructose consumption (3% (w/v) fructose in the drinking water) has a similar adverse effect on copper status [48,49]. The limitation of these studies is the AIN-76 based rodent diet which contains 49% sucrose, which could be a potential confounding factor. However, when extra fructose was given from drinking water, it still worsened the copper status.
Of note, the expression of copper transporter, Ctr-1, in duodenum was markedly upregulated when animals were exposed to a marginal copper deficient diet, and this upregulation was abrogated by high fructose feeding [48], suggesting that high fructose intake may impair copper absorption, which is likely a mechanism underlying copper-fructose interactions. Results from previous studies also support the concept that impaired copper absorption from gut might account for the more severe copper deficiency associated with copper-fructose interaction [61,62]. How dietary fructose impairs copper absorption and whether it is mediated by Ctr-1 remain open questions.
The mechanisms by which copper-fructose interaction induces NAFLD are not clear. Marginal copper deficient diet with high fructose feeding (CuMF) significantly upregulates hepatic fatty acid synthase (FAS) protein expression compared to either marginal copper deficient diet or high fructose feeding alone [48]. Copper-fructose interaction induced hepatic steatosis is completely abrogated by Kupffer cell (KC) depletion, which is associated with the downregulation of hepatic sterol regulatory element-binding protein-1 (SREBP-1) [48,141]. Upregulation of FAS and SREBP-1 by copper deficiency was also observed in other studies [142,143,144]. Pretreatment of KCs isolated from CuMF rats with an intracellular lysosomal iron chelator significantly attenuated lipopolysaccharide (LPS)-induced monocyte chemoattractant protein-1 (MCP-1) production in culture medium, suggesting that the MCP-1 signaling pathway was mediated, at least partially, by intracellular iron (141). A role for MCP-1 in inducing steatosis in hepatocytes has been described [145,146]. The precise mechanism underlying the role of KC in CuMF induced hepatic steatosis remain to be defined.
Iron overload is considered as a partial potential mechanism underlying copper deficiency and fructose induced metabolic syndrome [147,148,149,150]. We showed that marginal copper deficient and high fructose diet markedly increased liver iron level (Figure 4) [49] as well as plasma ferritin level in rats [48]. Similarly, NAFLD patients with low copper levels had hepatic iron overload [52,53]. Mechanism(s) by which copper deficiency induces iron overload have been partially elucidated. Cellular iron export requires members of a family of copper-containing ferroxidases, including ceruloplasmin and hephaestin which oxidize iron from the ferrous to ferric forms. The ferric form of iron binds to Apo-transferrin, thereby facilitating transferrin delivery to peripheral organs. Hephaestin functions to move iron across the basolateral membrane of intestinal epithelial cells into the circulation. Hephaestin-deficient mice display iron deficiency anemia with accumulation of iron in enterocytes [151]. Ceruloplasmin exerts its action on intestinal iron absorption, iron release from macrophages and hepatocytes [152,153]. A clinical phenotype of NAFLD that we regularly see is a young adult male with modestly decreased serum ceruloplasmin, increased serum ferritin, and high fructose intake via sugared pop.
Decreased activities of cuproenzymes, such as SOD1 and COX [154,155], may lead to decreased antioxidant defense and mitochondrial dysfunction, which are likely mechanisms leading to liver injury and hepatic fat accumulation. A previous study showed that the hepatocytes from rats with moderate copper deficiency (liver copper level of 4–8 µg/g dry weight, equal to marginal copper deficiency in our study) [48] have enlarged, bizarre-shaped mitochondria and disarranged endoplasmic reticulum (ER) as assessed by electron microscopy [156]. In rats with severe copper deficiency (liver copper level of ˂2 µg/g dry weight), the hepatocyte ultrastructure displayed dramatic changes characterized by the giant, misshapen mitochondria which occupy most of the cytoplasmic space and squeeze out and obscure otherwise normal-looking organelles. The mitochondrial matrix is less dense than normal [156]. These apparent morphological alterations of mitochondria appear to be linked to their abnormal functions. Whether and how severe copper deficiency affects ER and lysosome function leading to ER stress and defective autophagy remain elusive.
Pharmacological suppression of systemic copper levels with a chelating drug impaired mitochondrial energy metabolism and decreased ATP levels despite induction of glycolysis [157]. Of note, it is well documented that fructose metabolism also leads to ATP depletion [102,105,106]. Thus, one may postulate the additive or synergistic effect of copper deficiency and high fructose intake could be lethal. In fact, this effect has been demonstrated in experimental animals [158,159]. However, the effects of severe copper deficiency in rats fed with fructose can be reversed by replacing fructose with either glucose or starch [50,71].
Fructose and glucose are distinct in several aspects, including intestinal absorption, metabolic pathways and the organ of its major metabolism. The unique features of fructose absorption and metabolism provide clues for mechanisms of copper-fructose interactions. Rats treated with allopurinol, a competitive inhibitor of xanthine oxidase, displayed improved symptoms induced by copper deficient and high fructose diet, including anemia and decreased mortality, and this was associated with a dramatic reduction of uric acid. The beneficial role of allopurinol is likely attributable to protection against the catabolism of purines and increased nucleotides pool [160], suggesting the complexity of copper-fructose interactions in NAFLD.
Copper is required for the activity of COX, and copper deficiency was associated with decreased COX activity in multiple organs, including heart, liver, intestine, in mouse and rat models [124,154,161]. Whether or not copper deficiency induced metabolic phenotypic alteration through COX deficiency mediated mitochondrial dysfunction and the potential molecular mechanisms are not clear. COX (or complex IV) is the terminal enzyme of the electron transport chain in the inner mitochondrial membrane and catalyzes the transfer of electrons from reduced cytochrome c to molecular oxygen. Complex IV is composed of 14 subunits, and three of these (subunits I–III) form the highly conserved catalytic core of the enzyme encoded by mitochondrial DNA. The remaining less conserved subunits are encoded by nuclear genomes and were considered to be related to structural stability and enzyme activity. Highly conserved domains within subunit I include two heme moieties (heme a and a3) and a copper binding site (CuB), and subunit II also contains a copper binding site (CuA). The assembly of the complex IV protein is achieved by more than 20 different assembly proteins [162,163]. SCO2 encodes a copper chaperone required for the insertion of copper into the active site of subunit II of complex IV, but it is not essential for complete holoenzyme formation. SCO2 deficient mice exhibit increased adiposity, hepatic steatosis and insulin resistance along with 20%–60% reduction in complex IV activity [164]. In vitro research in human myoblasts demonstrated that COX deficiency due to mutations in SCO2 can be rescued by copper supplementation [165]. Whether the copper deficiency-induced reduction in COX activity is through SCO2 remains to be elucidated. However, mice lacking SURF1, a complex IV assembly protein, displayed an improved metabolic phenotype, including reduced adiposity, increased insulin sensitivity, and mitochondrial biogenesis despite of more than 50% reduction in COX activity [166,167,168].
In addition, previous studies from ATP7B knockout mice revealed that copper accumulation dysregulated nuclear receptors which contribute to liver function and lipid metabolism, such liver X receptor (LXR), farnesoid X receptor (FXR), retinoid X receptor (RXR), and small heterodimer partner (SHP) [134,169]. However, the effects of copper deficiency and high fructose on the regulation of nuclear receptors remain elusive.

6.2. Copper-Fructose Interaction and Hyperlipidemia

Copper-fructose interactions-induced hypercholesterolemia and hypertriglyceridemia have been well demonstrated [48,50,72,142,147,148,170,171,172,173]. In a population-based cohort study with 1197 subjects, dietary copper intake was inversely associated with plasma total cholesterol and LDL-cholesterol. Serum copper levels from a randomly identified subgroup of 231 men were also inversely associated with plasma total cholesterol and LDL-cholesterol, implying a crucial role of copper in cholesterol metabolism [170]. Rats exposed to a copper deficient diet for 3–4 weeks developed hypercholesterolemia, and this effect was more significant when the diet carbohydrate component was solely fructose, but not the starch, suggesting that a copper-fructose interaction is instrumental in the development of hypercholesterolemia. Moreover, hypercholesterolemia is further worsened by a diet high in saturated fat, but not polyunsaturated fat. However, copper-fructose interaction induced hypertriglyceridemia can be exacerbated by both high saturated fat diet and high polyunsaturated fat diet [72,171,173]. Of note, both hypertriglyceridemia and hypercholesterolemia are associated with hepatic iron overload and are ameliorated by dietary iron restriction [147,148]. Restriction of dietary iron intake significantly decreased blood cholesterol and triglyceride levels associated with decreased lipid peroxidation in rats fed with a copper deficient and high fructose diet. Similarly, the severity of copper deficiency was attenuated by the iron chelator, deferoxamine [147,150,174]. Moreover, increased iron intake further increased blood cholesterol and triglyceride levels in copper deficient diet fed rats [148]. Copper deficiency induced hypercholesterolemia is likely due to increased cholesterol synthesis [172]. Hepatocytes isolated from rats fed with a copper deficient diet for 7-8 weeks exhibited 90% reduction of copper content compared to those from adequate copper fed rats. After three hours incubation, these cells displayed 2–3 fold increase in the intracellular glutathione (GSH) synthesis rate along with the increased intracellular and extracellular GSH [175]. Treatment with L-buthionine sulfoximine (BSO), a specific GSH synthesis inhibitor, abolished the hypercholesterolemia and increased HMG-CoA reductase (HMGCR) activity in rats fed with copper deficient diet [142]. These results suggest that copper deficiency induced hypercholesterolemia and increased HMG-CoA are the consequence of increased GSH synthesis. Moreover, the induction of FAS expression was also prevented by BSO in copper deficient rats [144]. One hypothesized mechanism for the increased synthesis of GSH is a compensatory mechanism to the decreased antioxidant defenses due to the decreased cuproenzymes [48,154].

6.3. Copper-Fructose Interaction and Glucose Tolerance

Copper-deficient rats displayed improved glucose tolerance when they were switched from high fructose diet to high glucose diet for four weeks after being fed with a high fructose diet for five weeks. Similarly, changing the dietary carbohydrates in the copper-deficient diet from fructose to starch increased insulin levels and decreased blood glucose in response to a glucose tolerance test compared to rats continuously fed fructose. These results suggest that the copper-fructose interaction was more diabetogenic compared to copper-glucose. [71]. In addition, a copper deficient or a marginally copper deficient diet induced impaired glucose tolerance compared to an adequate copper diet, suggesting that copper deficiency may interfere with glucose utilization [176,177].

6.4. Copper-Fructose Interaction and Gut Permeability

Our recent study demonstrated that expression of the tight junction proteins, claudin-1 and occludin, was significantly downregulated in the ileum of rats fed with marginal copper deficient diet. This effect was synergistically or additively enhanced by high fructose feeding, suggesting copper-fructose interaction in the small intestine may play a vital role in gut barrier function [47]. A recent study showed that the metabolism of microbiota-derived butyrate in the gut epithelial cells through β-oxidation results in the depletion of oxygen and contributes to the maintenance of “physiologic hypoxia”, which, in turn, leads to the stabilization of HIF-1 [178]. HIF-1 is a transcription factor which plays a central role in the protection of gut barrier function in multiple ways, including transcriptional regulation of tight junction protein expression [179,180], induction of Tregs activation [181], and differentiation via transcriptional regulation of FoxP3 [182]. It is known that copper is required for the activation of HIF-1 [119,183]. Our previous study demonstrated that the fecal short chain fatty acid (SCFA), butyrate, was significantly decreased in high fructose fed male rats [184]. However, questions of whether or not decreased fecal SCFAs play a causal role and whether or not copper-fructose interaction induced gut barrier dysfunction is mediated by HIF-1 remain to be elucidated. In addition, in vitro studies demonstrated that increased copper concentration in the culture medium may induce Caco-2 cell apoptosis and increased permeability of the Caco-2 cell monolayer [185,186,187]. Collectively, copper homeostasis plays a crucial role in maintaining intestinal integrity.

6.5. Copper-Fructose Interaction and Gut Microbiome

Our data showed that the gut microbiome of rats fed with 30% fructose (w/v) in the drinking water and AIN-76 based rodent diet (ad libitum) for four weeks exhibited an obesity phenotype characterized by a markedly increased ratio of Firmicutes/Bacteroides, and this effect was further exacerbated with a marginal copper deficient diet, associated with increased gut permeability, exacerbated hepatic steatosis and liver injury [47,48]. These findings indicate that copper-fructose interaction may alter the gut microbiome. The mechanisms involved are not clear. Several lines of evidence indicate that copper might be involved in the regulation of gut microbiota and gut barrier function. First, copper has been used as an antimicrobial agent throughout the ages [188], and the response to copper stress varies among different bacteria species [189,190]. Second, one of the copper containing enzymes, diamine oxidase, was found in high concentrations in intestinal mucosa and its circulating enzyme activity serves as a marker of mucosal maturation and integrity, as does the copper level [191,192,193]. Thus, decreased copper levels may exacerbate dietary fructose-induced gut microbiota dysbiosis and/or gut barrier dysfunction. Whether copper-fructose interaction induced gut barrier dysfunction is the direct role of copper-fructose interaction in the intestine and/or mediated by the gut microbiota requires further investigation.

6.6. Sex Difference in the Copper-Fructose Interaction

Sex differences in the metabolic effects of fructose and/or copper deficiency have long been noted in the animal studies [158,194,195,196] as well as in humans [197,198], with males being sensitive to the deleterious effects of fructose and/or copper deficiency, and females being protected, which is consistent with the sex differences in NAFLD prevalence [75,199]. However, the mechanistic link between fructose, copper deficiency and sex is not well established. Experimental study from rats implies the level of testosterone in the males may play a role in the severity of copper deficiency [195]. In line with this, evidence from a murine study demonstrated that testosterone robustly suppressed hepcidin transcription through epidermal growth factor receptor (Egfr) signaling, and these suppressive effects were more obvious in male mice than in female mice [200]. Our work also showed significantly decreased plasma hepcidin levels in CuMF male rats compared to controls [141]. Sex differences in the copper-fructose interaction were also shown with regard to the enzyme activities involved in fructose metabolism pathway and their relevant metabolites [201,202]. A previous study showed that female rats displayed lower hepatic KHK and triose kinase activities, but higher triose phosphate isomerase activity compared to male rats in response to high fructose with either adequate copper or copper deficient diet [201]. Moreover, F1P levels were elevated to a greater extent in male rats compared to female rats on copper deficient diet [202]. In addition, high fructose feeding resulted in markedly elevated serum uric acid levels in male rats compared to female rats, and it was further increased by copper deficient diet compared to adequate copper diet [201]. However, inhibition of uric acid generation with allopurinol showed beneficial effects on copper-fructose interactions [160]. Recent studies pointed out that sexual dimorphism in glycerol metabolism and aquaglyceroporins (AQPs) contribute to the lower prevalence of NAFLD in premenopausal women as well as in rodents. However, whether these mechanisms contribute to the sex difference in the copper-fructose interactions remain to be determined (reviewed by Rodriguez et al.) [203]. Collectively, a sex difference in copper-fructose interactions likely contributes to sex variances in fructose metabolism and susceptibility to NAFLD/metabolic syndrome.

7. Conclusions

High fructose consumption and low copper availability are two risk factors identified in NAFLD patients. Evidence of copper-fructose interactions comes largely from animal studies. Hepatic iron overload and mitochondrial dysfunction are two important mechanisms. The causal role of high fructose consumption on the impaired copper status in humans as well as copper-fructose interactions in the pathogenesis of NAFLD patients remain to be firmly established. Therefore, larger cohort studies are needed to examine the correlation between copper status and fructose consumption in healthy controls, obese and NAFLD patients. However, we suggest that there are multiple NAFLD phenotypes, with one such NAFLD phenotype being relatively young males with high sugar sweetened beverage (and high fructose) consumption and modestly depressed serum copper/ceruloplasmin. A beneficial role for restricting dietary fructose intake to improve obesity and the metabolic syndrome has been clearly demonstrated and further studies may confirm the additional role of low copper availability.

Author Contributions

M.S., draft and design the manuscript; M.B.V., revise and proofreading; C.J.M., revise and proofreading.

Funding

This work was funded by National Institutes of Health (NIH). MS was supported in part by the Pilot Project Program of NIH grant under award number P20GM113226 (CJM); CJM was funded by NIH P50AA024337, RO1AA015970, PO1AA017103, R37AA010762, RO1AA018869, RC2AA019385, P30AA019360, R01AA018016, RO1DK071765, the Veterans Administration, and UofL Clinical and Translational Sciences Pilots, “High fructose feeding induces copper deficiency: A novel mechanism for obesity related hyperlipidemia and fatty liver”.

Acknowledgments

We thank Marion McClain and Russell Prough for careful reading of this manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

NAFLDnonalcoholic fatty liver disease
KCKupffer cell
HFCShigh-fructose corn syrup
KHKketohexokinase
TLR4toll like receptor 4
MCDmethionine choline deficient
NASHnonalcoholic steatohepatitis
T2Dtype 2 diabetes
SSBssugar-sweetened beverages
CVDscardiovascular diseases
RDARecommended Dietary Allowance
EAREstimated Average Requirement
SOD1copper/zinc-superoxide dismutase
COXcytochrome c oxidase
F1Pfructose 1-phosphate
DHAPdihydroxyacetone phosphate
AMPDadenosine monophosphate deaminase
IMPinosine monophosphate
AMPKadenosine monophosphate-activated protein kinase
XOxanthine oxidase
ARaldose reductase
CCScopper chaperone for SOD1
HIF-1hypoxia inducible factor-1
Ctr1copper transporter 1
MTmetallothionein
Atox1antioxidant protein 1
TGNtrans-Golgi network
HCVhepatitis C virus
FP-1ferroportin-1
NASNAFLD Activity Score
WDWilson’s disease
FASfatty acid synthase
SREBP-1sterol regulatory element-binding protein-1
BSOL-buthionine sulfoximine
LPSlipopolysaccharide
MCP-1monocyte chemoattractant protein-1
LXRliver X receptor
FXRfarnesoid X receptor
RXRretinoid X receptor
SHPsmall heterodimer partner
ERendoplasmic reticulum
GSHglutathione
GSSGglutathione disulfide
HMGCRHMG-CoA reductase
GPxglutathione peroxidase
SCFAshort chain fatty acid
Egfrepidermal growth factor receptor
AQPsaquaglyceroporins

References

  1. Johnson, R.J.; Segal, M.S.; Sautin, Y.; Nakagawa, T.; Feig, D.I.; Kang, D.H.; Gersch, M.S.; Benner, S.; Sánchez-Lozada, L.G. Potential role of sugar (fructose) in the epidemic of hypertension, obesity and the metabolic syndrome, diabetes, kidney disease, and cardiovascular disease. Am. J. Clin. Nutr. 2007, 86, 899–906. [Google Scholar] [PubMed]
  2. Hu, F.B.; Malik, V.S. Sugar-sweetened beverages and risk of obesity and type 2 diabetes: Epidemiologic evidence. Physiol. Behav. 2010, 100, 47–54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Bray, G.A.; Nielsen, S.J.; Popkin, B.M. Consumption of high-fructose corn syrup in beverages may play a role in the epidemic of obesity. Am. J. Clin. Nutr. 2004, 79, 537–543. [Google Scholar] [CrossRef] [PubMed]
  4. Vos, M.B.; Kimmons, J.E.; Gillespie, C.; Welsh, J.; Blanck, H.M. Dietary fructose consumption among US children and adults: The third national health and nutrition examination survey. Medscape J. Med. 2008, 10, 160. [Google Scholar] [PubMed]
  5. Park, Y.K.; Yetley, E.A. Intakes and food sources of fructose in the United States. Am. J. Clin. Nutr. 1993, 58, 737S–747S. [Google Scholar] [CrossRef] [PubMed]
  6. Marriott, B.P.; Cole, N.; Lee, E. National estimates of dietary fructose intake increased from 1977 to 2004 in the United States. J. Nutr. 2009, 139, 1228S–1235S. [Google Scholar] [CrossRef] [PubMed]
  7. Welsh, J.A.; Karpen, S.; Vos, M.B. Increasing prevalence of nonalcoholic fatty liver disease among United States adolescents, 1988–1994 to 2007–2010. J. Pediatr. 2013, 162, 496–500. [Google Scholar] [CrossRef] [PubMed]
  8. Younossi, Z.M.; Koenig, A.B.; Abdelatif, D.; Fazel, Y.; Henry, L.; Wymer, M. Global epidemiology of nonalcoholic fatty liver disease—Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 2016, 64, 73–84. [Google Scholar] [CrossRef] [PubMed]
  9. Assy, N.; Nasser, G.; Kamayse, I.; Nseir, W.; Beniashvili, Z.; Djibre, A.; Grosovski, M. Soft drink consumption linked with fatty liver in the absence of traditional risk factors. Can. J. Gastroenterol. 2008, 22, 811–816. [Google Scholar] [CrossRef] [PubMed]
  10. Ouyang, X.; Cirillo, P.; Sautin, Y.; McCall, S.; Bruchette, J.L.; Diehl, A.M.; Johnson, R.J.; Abdelmalek, M.F. Fructose consumption as a risk factor for non-alcoholic fatty liver disease. J. Hepatol. 2008, 48, 993–999. [Google Scholar] [CrossRef] [PubMed]
  11. Lim, J.S.; Mietus-Snyder, M.; Valente, A.; Schwarz, J.M.; Lustig, R.H. The role of fructose in the pathogenesis of NAFLD and the metabolic syndrome. Nat. Rev. Gastroenterol. Hepatol. 2010, 7, 251–264. [Google Scholar] [CrossRef] [PubMed]
  12. Abdelmalek, M.F.; Suzuki, A.; Guy, C.; Unalp-Arida, A.; Colvin, R.; Johnson, R.J.; Diehl, A.M. Increased fructose consumption is associated with fibrosis severity in patients with nonalcoholic fatty liver disease. Hepatology 2010, 51, 1961–1971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Tappy, L.; Le, K.A. Metabolic effects of fructose and the worldwide increase in obesity. Physiol. Rev. 2010, 90, 23–46. [Google Scholar] [CrossRef] [PubMed]
  14. Heinz, F.; Lamprecht, W.; Kirsch, J. Enzymes of fructose metabolism in human liver. J. Clin. Investig. 1968, 47, 1826–1832. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Fujisawa, T.; Riby, J.; Kretchmer, N. Intestinal absorption of fructose in the rat. Gastroenterology 1991, 101, 360–367. [Google Scholar] [CrossRef]
  16. Riby, J.E.; Fujisawa, T.; Kretchmer, N. Fructose absorption. Am. J. Clin. Nutr. 1993, 58, 748S–753S. [Google Scholar] [CrossRef] [PubMed]
  17. Spruss, A.; Kanuri, G.; Wagnerberger, S.; Haub, S.; Bischoff, S.C.; Bergheim, I. Toll-like receptor 4 is involved in the development of fructose-induced hepatic steatosis in mice. Hepatology 2009, 50, 1094–1104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Wada, T.; Kenmochi, H.; Miyashita, Y.; Sasaki, M.; Ojima, M.; Sasahara, M.; Koya, D.; Tsuneki, H.; Sasaoka, T. Spironolactone improves glucose and lipid metabolism by ameliorating hepatic steatosis and inflammation and suppressing enhanced gluconeogenesis induced by high-fat and high-fructose diet. Endocrinology 2010, 151, 2040–2049. [Google Scholar] [CrossRef] [PubMed]
  19. Kelley, G.L.; Allan, G.; Azhar, S. High dietary fructose induces a hepatic stress response resulting in cholesterol and lipid dysregulation. Endocrinology 2004, 145, 548–555. [Google Scholar] [CrossRef] [PubMed]
  20. Park, O.J.; Cesar, D.; Faix, D.; Wu, K.; Shackleton, C.H.; Hellerstein, M.K. Mechanisms of fructose-induced hypertriglyceridaemia in the rat. Activation of hepatic pyruvate dehydrogenase through inhibition of pyruvate dehydrogenase kinase. Biochem. J. 1992, 282, 753–757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Jang, C.; Hui, S.; Lu, W.; Cowan, A.J.; Morscher, R.J.; Lee, G.; Liu, W.; Tesz, G.J.; Birnbaum, M.J.; Rabinowitz, J.D. The Small intestine converts dietary fructose into glucose and organic acids. Cell MeTable 2018, 27, 351–361. [Google Scholar] [CrossRef] [PubMed]
  22. Ishimoto, T.; Lanaspa, M.A.; Rivard, C.J.; Roncal-Jimenez, C.A.; Orlicky, D.J.; Cicerchi, C.; McMahan, R.H.; Abdelmalek, M.F.; Rosen, H.R.; Jackman, M.R.; et al. High-fat and high-sucrose (western) diet induces steatohepatitis that is dependent on fructokinase. Hepatology 2013, 58, 1632–1643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Ishimoto, T.; Lanaspa, M.A.; Le, M.T.; Garcia, G.E.; Diggle, C.P.; Maclean, P.S.; Jackman, M.R.; Asipu, A.; Roncal-Jimenez, C.A.; Kosugi, T.; et al. Opposing effects of fructokinase C and A isoforms on fructose-induced metabolic syndrome in mice. Proc. Natl. Acad. Sci. USA 2012, 109, 4320–4325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Softic, S.; Gupta, M.K.; Wang, G.X.; Fujisaka, S.; O’Neill, B.T.; Rao, T.N.; Willoughby, J.; Harbison, C.; Fitzgerald, K.; Ilkayeva, O.; et al. Divergent effects of glucose and fructose on hepatic lipogenesis and insulin signaling. J. Clin. Investig. 2017, 127, 4059–4074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Bergheim, I.; Weber, S.; Vos, M.; Kramer, S.; Volynets, V.; Kaserouni, S.; McClain, C.J.; Bischoff, S.C. Antibiotics protect against fructose-induced hepatic lipid accumulation in mice: Role of endotoxin. J. Hepatol. 2008, 48, 983–992. [Google Scholar] [CrossRef] [PubMed]
  26. Sun, S.Z.; Empie, M.W. Fructose metabolism in humans—What isotopic tracer studies tell us. Nutr. MeTable 2012, 9, 89. [Google Scholar] [CrossRef] [PubMed]
  27. Jensen, T.; Abdelmalek, M.F.; Sullivan, S.; Nadeau, K.J.; Green, M.; Roncal, C.; Nakagawa, T.; Kuwabara, M.; Sato, Y.; Kang, D.H.; et al. Fructose and sugar: A major mediator of non-alcoholic fatty liver disease. J. Hepatol. 2018, 68, 1063–1075. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Malik, V.S.; Pan, A.; Willett, W.C.; Hu, F.B. Sugar-sweetened beverages and weight gain in children and adults: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2013, 98, 1084–1102. [Google Scholar] [CrossRef] [PubMed]
  29. Malik, V.S.; Popkin, B.M.; Bray, G.A.; Despres, J.P.; Hu, F.B. Sugar-sweetened beverages, obesity, type 2 diabetes mellitus, and cardiovascular disease risk. Circulation 2010, 121, 1356–1364. [Google Scholar] [CrossRef] [PubMed]
  30. Malik, V.S.; Schulze, M.B.; Hu, F.B. Intake of sugar-sweetened beverages and weight gain: A systematic review. Am. J. Clin. Nutr. 2006, 84, 274–288. [Google Scholar] [CrossRef] [PubMed]
  31. Stanhope, K.L. Role of fructose-containing sugars in the epidemics of obesity and metabolic syndrome. Annu. Rev. Med. 2012, 63, 329–343. [Google Scholar] [CrossRef] [PubMed]
  32. Stanhope, K.L.; Schwarz, J.M.; Keim, N.L.; Griffen, S.C.; Bremer, A.A.; Graham, J.L.; Hatcher, B.; Cox, C.L.; Dyachenko, A.; Zhang, W.; et al. Consuming fructose-sweetened, not glucose-sweetened, beverages increases visceral adiposity and lipids and decreases insulin sensitivity in overweight/obese humans. J. Clin. Investig. 2009, 119, 1322–1334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Maersk, M.; Belza, A.; Stodkilde-Jorgensen, H.; Ringgaard, S.; Chabanova, E.; Thomsen, H.; Pedersen, S.B.; Astrup, A.; Richelsen, B. Sucrose-sweetened beverages increase fat storage in the liver, muscle, and visceral fat depot: A 6-mo randomized intervention study. Am. J. Clin. Nutr. 2012, 95, 283–289. [Google Scholar] [CrossRef] [PubMed]
  34. Le, K.A.; Faeh, D.; Stettler, R.; Ith, M.; Kreis, R.; Vermathen, P.; Boesch, C.; Ravussin, E.; Tappy, L. A 4-wk high-fructose diet alters lipid metabolism without affecting insulin sensitivity or ectopic lipids in healthy humans. Am. J. Clin. Nutr. 2006, 84, 1374–1379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Johnston, R.D.; Stephenson, M.C.; Crossland, H.; Cordon, S.M.; Palcidi, E.; Cox, E.F.; Taylor, M.A.; Aithal, G.P.; Macdonald, I.A. No difference between high-fructose and high-glucose diets on liver triacylglycerol or biochemistry in healthy overweight men. Gastroenterology 2013, 145, 1016–1025. [Google Scholar] [CrossRef] [PubMed]
  36. Kuzma, J.N.; Cromer, G.; Hagman, D.K.; Breymeyer, K.L.; Roth, C.L.; Foster-Schubert, K.E.; Holte, S.E.; Weigle, D.S.; Kratz, M. No differential effect of beverages sweetened with fructose, high-fructose corn syrup, or glucose on systemic or adipose tissue inflammation in normal-weight to obese adults: A randomized controlled trial. Am. J. Clin. Nutr. 2016, 104, 306–314. [Google Scholar] [CrossRef] [PubMed]
  37. Ter Horst, K.W.; Schene, M.R.; Holman, R.; Romijn, J.A.; Serlie, M.J. Effect of fructose consumption on insulin sensitivity in nondiabetic subjects: A systematic review and meta-analysis of diet-intervention trials. Am. J. Clin. Nutr. 2016, 104, 1562–1576. [Google Scholar] [CrossRef] [PubMed]
  38. Lustig, R.H.; Mulligan, K.; Noworolski, S.M.; Tai, V.W.; Wen, M.J.; Erkin-Cakmak, A.; Gugliucci, A.; Schwarz, J.M. Isocaloric fructose restriction and metabolic improvement in children with obesity and metabolic syndrome. Obesity (Silver Spring) 2016, 24, 453–460. [Google Scholar] [CrossRef] [PubMed]
  39. Schwarz, J.M.; Noworolski, S.M.; Erkin-Cakmak, A.; Korn, N.J.; Wen, M.J.; Tai, V.W.; Jones, G.M.; Palii, S.P.; Velasco-Alin, M.; Pan, K.; et al. Effects of dietary fructose restriction on liver fat, de novo lipogenesis, and insulin kinetics in children with obesity. Gastroenterology 2017, 153, 743–752. [Google Scholar] [CrossRef] [PubMed]
  40. Campos, V.; Despland, C.; Brandejsky, V.; Kreis, R.; Schneiter, P.; Chiolero, A.; Boesch, C.; Tappy, L. Sugar- and artificially sweetened beverages and intrahepatic fat: A randomized controlled trial. Obesity 2015, 23, 2335–2339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Pickens, M.K.; Ogata, H.; Soon, R.K.; Grenert, J.P.; Maher, J.J. Dietary fructose exacerbates hepatocellular injury when incorporated into a methionine-choline-deficient diet. Liver Int. 2010, 30, 1229–1239. [Google Scholar] [CrossRef] [PubMed]
  42. Pickens, M.K.; Yan, J.S.; Ng, R.K.; Ogata, H.; Grenert, J.P.; Beysen, C.; Turner, S.M.; Maher, J.J. Dietary sucrose is essential to the development of liver injury in the methionine-choline-deficient model of steatohepatitis. J. Lipid Res. 2009, 50, 2072–2082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Kohli, R.; Kirby, M.; Xanthakos, S.A.; Softic, S.; Feldstein, A.E.; Saxena, V.; Tang, P.H.; Miles, L.; Miles, M.V.; Balistreri, W.F.; et al. High-fructose, medium chain trans fat diet induces liver fibrosis and elevates plasma coenzyme Q9 in a novel murine model of obesity and nonalcoholic steatohepatitis. Hepatology 2010, 52, 934–944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Jin, R.; Le, N.A.; Liu, S.; Farkas Epperson, M.; Ziegler, T.R.; Welsh, J.A.; Jones, D.P.; McClain, C.J.; Vos, M.B. Children with NAFLD are more sensitive to the adverse metabolic effects of fructose beverages than children without NAFLD. J. Clin. Endocrinol. MeTable 2012, 97, E1088–E1098. [Google Scholar] [CrossRef] [PubMed]
  45. Teff, K.L.; Grudziak, J.; Townsend, R.R.; Dunn, T.N.; Grant, R.W.; Adams, S.H.; Keim, N.L.; Cummings, B.P.; Stanhope, K.L.; Havel, P.J. Endocrine and metabolic effects of consuming fructose- and glucose-sweetened beverages with meals in obese men and women: Influence of insulin resistance on plasma triglyceride responses. J. Clin. Endocrinol. MeTable 2009, 94, 1562–1569. [Google Scholar] [CrossRef] [PubMed]
  46. Abdelmalek, M.F.; Lazo, M.; Horska, A.; Bonekamp, S.; Lipkin, E.W.; Balasubramanyam, A.; Bantle, J.P.; Johnson, R.J.; Diehl, A.M.; Clark, J.M.; et al. Higher dietary fructose is associated with impaired hepatic adenosine triphosphate homeostasis in obese individuals with type 2 diabetes. Hepatology 2012, 56, 952–960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Song, M.; Li, X.; Zhang, X.; Shi, H.; Vos, M.B.; Wei, X.; Wang, Y.; Gao, H.; Rouchka, E.C.; Yin, X.; et al. Dietary copper-fructose interactions alter gut microbial activity in male rats. Am. J. Physiol. Gastrointest. Liver Physiol. 2018, 314, G119–G130. [Google Scholar] [CrossRef] [PubMed]
  48. Song, M.; Schuschke, D.A.; Zhou, Z.; Chen, T.; Pierce, W.M., Jr.; Wang, R.; Johnson, W.T.; McClain, C.J. High fructose feeding induces copper deficiency in Sprague-Dawley rats: A novel mechanism for obesity related fatty liver. J. Hepatol 2012, 56, 433–440. [Google Scholar] [CrossRef] [PubMed]
  49. Song, M.; Schuschke, D.A.; Zhou, Z.; Chen, T.; Shi, X.; Zhang, J.; Zhang, X.; Pierce, W.M., Jr.; Johnson, W.T.; Vos, M.B.; et al. Modest fructose beverage intake causes liver injury and fat accumulation in marginal copper deficient rats. Obesity (Silver Spring) 2013, 21, 1669–1675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Fields, M.; Ferretti, R.J.; Reiser, S.; Smith, J.C., Jr. The severity of copper deficiency in rats is determined by the type of dietary carbohydrate. Proc. Soc. Exp. Biol. Med. 1984, 175, 530–537. [Google Scholar] [CrossRef] [PubMed]
  51. Fields, M.; Ferretti, R.J.; Smith, J.C., Jr.; Reiser, S. The interaction of type of dietary carbohydrates with copper deficiency. Am. J. Clin. Nutr. 1984, 39, 289–295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Aigner, E.; Strasser, M.; Haufe, H.; Sonnweber, T.; Hohla, F.; Stadlmayr, A.; Solioz, M.; Tilg, H.; Patsch, W.; Weiss, G.; et al. A role for low hepatic copper concentrations in nonalcoholic Fatty liver disease. Am. J. Gastroenterol. 2010, 105, 1978–1985. [Google Scholar] [CrossRef] [PubMed]
  53. Aigner, E.; Theurl, I.; Haufe, H.; Seifert, M.; Hohla, F.; Scharinger, L.; Stickel, F.; Mourlane, F.; Weiss, G.; Datz, C. Copper availability contributes to iron perturbations in human nonalcoholic fatty liver disease. Gastroenterology 2008, 135, 680–688. [Google Scholar] [CrossRef] [PubMed]
  54. Mendoza, M.; Caltharp, S.; Song, M.; Collin, L.; Konomi, J.V.; McClain, C.J.; Vos, M.B. Low Hepatic Tissue Copper in Pediatric Nonalcoholic Fatty Liver Disease. J. Pediatr. Gastroenterol. Nutr. 2017, 65, 89–92. [Google Scholar] [CrossRef] [PubMed]
  55. Stattermayer, A.F.; Traussnigg, S.; Aigner, E.; Kienbacher, C.; Huber-Schonauer, U.; Steindl-Munda, P.; Stadlmayr, A.; Wrba, F.; Trauner, M.; Datz, C.; et al. Low hepatic copper content and PNPLA3 polymorphism in non-alcoholic fatty liver disease in patients without metabolic syndrome. J. Trace Elem. Med. Biol. 2017, 39, 100–107. [Google Scholar] [CrossRef] [PubMed]
  56. Nobili, V.; Siotto, M.; Bedogni, G.; Rava, L.; Pietrobattista, A.; Panera, N.; Alisi, A.; Squitti, R. Levels of serum ceruloplasmin associate with pediatric nonalcoholic fatty liver disease. J. Pediatr. Gastroenterol. Nutr. 2013, 56, 370–375. [Google Scholar] [CrossRef] [PubMed]
  57. Holden, J.M.; Wolf, W.R.; Mertz, W. Zinc and copper in self-selected diets. J. Am. Diet. Assoc. 1979, 75, 23–28. [Google Scholar] [PubMed]
  58. Klevay, L.M.; Reck, S.J.; Jacob, R.A.; Logan, G.M., Jr.; Munoz, J.M.; Sandstead, H.H. The human requirement for copper. I. Healthy men fed conventional, American diets. Am. J. Clin. Nutr. 1980, 33, 45–50. [Google Scholar] [CrossRef] [PubMed]
  59. Klevay, L.M. Is the Western diet adequate in copper? J. Trace Elem. Med. Biol. 2011, 25, 204–212. [Google Scholar] [CrossRef] [PubMed]
  60. Reiser, S.; Smith, J.C., Jr.; Mertz, W.; Holbrook, J.T.; Scholfield, D.J.; Powell, A.S.; Canfield, W.K.; Canary, J.J. Indices of copper status in humans consuming a typical American diet containing either fructose or starch. Am. J. Clin. Nutr. 1985, 42, 242–251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Fields, M.; Holbrook, J.; Scholfield, D.; Smith, J.C., Jr.; Reiser, S. Effect of fructose or starch on copper-67 absorption and excretion by the rat. J. Nutr. 1986, 116, 625–632. [Google Scholar] [PubMed]
  62. Holbrook, J.; Fields, M.; Smith, J.C., Jr.; Reiser, S. Tissue distribution and excretion of copper-67 intraperitoneally administered to rats fed fructose or starch. J. Nutr. 1986, 116, 831–838. [Google Scholar] [PubMed]
  63. Ernst, B.; Thurnheer, M.; Schultes, B. Copper deficiency after gastric bypass surgery. Obesity. (Silver Spring) 2009, 17, 1980–1981. [Google Scholar] [CrossRef] [PubMed]
  64. Kumar, P.; Hamza, N.; Madhok, B.; De Alwis, N.; Sharma, M.; Miras, A.D.; Mahawar, K.K. Copper Deficiency after Gastric Bypass for Morbid Obesity: A Systematic Review. Obes. Surg. 2016, 26, 1335–1342. [Google Scholar] [CrossRef] [PubMed]
  65. Hedera, P.; Peltier, A.; Fink, J.K.; Wilcock, S.; London, Z.; Brewer, G.J. Myelopolyneuropathy and pancytopenia due to copper deficiency and high zinc levels of unknown origin II. The denture cream is a primary source of excessive zinc. Neurotoxicology 2009, 30, 996–999. [Google Scholar] [CrossRef] [PubMed]
  66. Spain, R.I.; Leist, T.P.; De Sousa, E.A. When metals compete: A case of copper-deficiency myeloneuropathy and anemia. Nat. Clin. Pract. Neurol. 2009, 5, 106–111. [Google Scholar] [CrossRef] [PubMed]
  67. Fujii, Y.; Nanashima, A.; Hiyoshi, M.; Imamura, N.; Yano, K.; Hamada, T. Risk factors for development of nonalcoholic fatty liver disease after pancreatoduodenectomy. Ann. Gastroenterol. Surg. 2017, 1, 226–231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Fields, M.; Ferretti, R.J.; Judge, J.M.; Smith, J.C.; Reiser, S. Effects of different dietary carbohydrates on hepatic enzymes of copper-deficient rats. Proc. Soc. Exp. Biol. Med. 1985, 178, 362–366. [Google Scholar] [CrossRef] [PubMed]
  69. Fields, M.; Ferretti, R.J.; Smith, J.C., Jr.; Reiser, S. Effect of copper deficiency on metabolism and mortality in rats fed sucrose or starch diets. J. Nutr. 1983, 113, 1335–1345. [Google Scholar] [CrossRef] [PubMed]
  70. Fields, M.; Ferretti, R.J.; Smith, J.C., Jr.; Reiser, S. Effect of dietary carbohydrates and copper status on blood pressure of rats. Life Sci. 1984, 34, 763–769. [Google Scholar] [CrossRef]
  71. Fields, M.; Ferretti, R.J.; Smith, J.C., Jr.; Reiser, S. Impairment of glucose tolerance in copper-deficient rats: Dependency on the type of dietary carbohydrate. J. Nutr. 1984, 114, 393–397. [Google Scholar] [CrossRef] [PubMed]
  72. Fields, M.; Lewis, C.G. Dietary fructose but not starch is responsible for hyperlipidemia associated with copper deficiency in rats: Effect of high-fat diet. J. Am. Coll. Nutr. 1999, 18, 83–87. [Google Scholar] [CrossRef] [PubMed]
  73. Younossi, Z.M.; Stepanova, M.; Afendy, M.; Fang, Y.; Younossi, Y.; Mir, H.; Srishord, M. Changes in the prevalence of the most common causes of chronic liver diseases in the United States from 1988 to 2008. Clin. Gastroenterol. Hepatol. 2011, 9, 524.e1–530.e1. [Google Scholar] [CrossRef] [PubMed]
  74. Charlton, M.R.; Burns, J.M.; Pedersen, R.A.; Watt, K.D.; Heimbach, J.K.; Dierkhising, R.A. Frequency and outcomes of liver transplantation for nonalcoholic steatohepatitis in the United States. Gastroenterology 2011, 141, 1249–1253. [Google Scholar] [CrossRef] [PubMed]
  75. Fraser, A.; Longnecker, M.P.; Lawlor, D.A. Prevalence of elevated alanine aminotransferase among US adolescents and associated factors: NHANES 1999–2004. Gastroenterology 2007, 133, 1814–1820. [Google Scholar] [CrossRef] [PubMed]
  76. Hashimoto, E.; Tokushige, K. Prevalence, gender, ethnic variations, and prognosis of NASH. J. Gastroenterol. 2011, 46 (Suppl. 1), 63–69. [Google Scholar] [CrossRef] [PubMed]
  77. Rosinger, A.; Herrick, K.; Gahche, J.; Park, S. Sugar-sweetened Beverage Consumption Among, U.S. Adults, 2011–2014. NCHS Data Briefs 2017, 270, 1–8. [Google Scholar]
  78. Rosinger, A.; Herrick, K.; Gahche, J.; Park, S. Sugar-sweetened Beverage Consumption Among, U.S.; Youth, 2011–2014. NCHS Data Briefs 2017, 271, 1–8. [Google Scholar]
  79. Bleich, S.N.; Vercammen, K.A. The negative impact of sugar-sweetened beverages on children’s health: An update of the literature. BMC Obes. 2018, 5, 6. [Google Scholar] [CrossRef] [PubMed]
  80. Ogden, C.L.; Carroll, M.D.; Fryar, C.D.; Flegal, K.M. Prevalence of Obesity Among Adults and Youth: United States, 2011–2014. NCHS Data Briefs 2015, 219, 1–8. [Google Scholar]
  81. Malik, V.S.; Hu, F.B. Fructose and Cardiometabolic HealthWhat the Evidence from Sugar-Sweetened Beverages Tells US. J. Am. Coll. Cardiol. 2015, 66, 1615–1624. [Google Scholar] [CrossRef] [PubMed]
  82. Singh, G.M.; Micha, R.; Khatibzadeh, S.; Lim, S.; Ezzati, M.; Mozaffarian, D. Estimated Global, Regional, and National Disease Burdens Related to Sugar-Sweetened Beverage Consumption in 2010. Circulation 2015, 132, 639–666. [Google Scholar] [CrossRef] [PubMed]
  83. Frank, A.P.; Clegg, D.J. Dietary Guidelines for Americans—Eat Less Sugar. JAMA 2016, 315, 1196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Institute of Medicine Panel on Micronutrients. Dietary Reference Intakes for Vitamin A, Vitamin K, Arsenic, Boron, Chromium, Copper, Iodine, Iron, Manganese, Molybdenum, Nickel, Silicon, Vanadium, and Zinc; National Academies Press: Washington, DC, USA, 2001; Copyright 2001 by the National Academy of Sciences. All rights reserved., 2001. [Google Scholar]
  85. Klevay, L.M. Lack of a recommended dietary allowance for copper may be hazardous to your health. J. Am. Coll. Nutr. 1998, 17, 322–326. [Google Scholar] [CrossRef] [PubMed]
  86. Pang, Y.; MacIntosh, D.L.; Ryan, P.B. A longitudinal investigation of aggregate oral intake of copper. J. Nutr. 2001, 131, 2171–2176. [Google Scholar] [CrossRef] [PubMed]
  87. Prohaska, J.R. Impact of copper limitation on expression and function of multicopper oxidases (ferroxidases). Adv. Nutr. 2011, 2, 89–95. [Google Scholar] [CrossRef] [PubMed]
  88. Prohaska, J.R. Impact of copper deficiency in humans. Ann. N. Y. Acad. Sci. 2014, 1314, 1–5. [Google Scholar] [CrossRef] [PubMed]
  89. Milne, D.B.; Johnson, P.E. Assessment of copper status: Effect of age and gender on reference ranges in healthy adults. Clin. Chem. 1993, 39, 883–887. [Google Scholar] [PubMed]
  90. Milne, D.B. Assessment of copper nutritional status. Clin. Chem. 1994, 40, 1479–1484. [Google Scholar] [PubMed]
  91. Heffern, M.C.; Park, H.M.; Au-Yeung, H.Y.; Van de Bittner, G.C.; Ackerman, C.M.; Stahl, A.; Chang, C.J. In vivo bioluminescence imaging reveals copper deficiency in a murine model of nonalcoholic fatty liver disease. Proc. Natl. Acad. Sci. USA 2016, 113, 14219–14224. [Google Scholar] [CrossRef] [PubMed]
  92. David, E.S.; Cingari, D.S.; Ferraris, R.P. Dietary induction of intestinal fructose absorption in weaning rats. Pediatr. Res. 1995, 37, 777–782. [Google Scholar] [CrossRef] [PubMed]
  93. Douard, V.; Ferraris, R.P. The role of fructose transporters in diseases linked to excessive fructose intake. J. Physiol. 2013, 591, 401–414. [Google Scholar] [CrossRef] [PubMed]
  94. DeBosch, B.J.; Chen, Z.; Finck, B.N.; Chi, M.; Moley, K.H. Glucose transporter-8 (GLUT8) mediates glucose intolerance and dyslipidemia in high-fructose diet-fed male mice. Mol. Endocrinol. 2013, 27, 1887–1896. [Google Scholar] [CrossRef] [PubMed]
  95. Debosch, B.J.; Chen, Z.; Saben, J.L.; Finck, B.N.; Moley, K.H. Glucose transporter 8 (GLUT8) mediates fructose-induced de novo lipogenesis and macrosteatosis. J. Biol. Chem. 2014, 289, 10989–10998. [Google Scholar] [CrossRef] [PubMed]
  96. DeBosch, B.J.; Chi, M.; Moley, K.H. Glucose transporter 8 (GLUT8) regulates enterocyte fructose transport and global mammalian fructose utilization. Endocrinology 2012, 153, 4181–4191. [Google Scholar] [CrossRef] [PubMed]
  97. Douard, V.; Ferraris, R.P. Regulation of the fructose transporter GLUT5 in health and disease. Am. J. Physiol. Endocrinol. MeTable 2008, 295, E227–E237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Barone, S.; Fussell, S.L.; Singh, A.K.; Lucas, F.; Xu, J.; Kim, C.; Wu, X.; Yu, Y.; Amlal, H.; Seidler, U.; et al. Slc2a5 (Glut5) is essential for the absorption of fructose in the intestine and generation of fructose-induced hypertension. J. Biol. Chem. 2009, 284, 5056–5066. [Google Scholar] [CrossRef] [PubMed]
  99. Douard, V.; Choi, H.I.; Elshenawy, S.; Lagunoff, D.; Ferraris, R.P. Developmental reprogramming of rat GLUT5 requires glucocorticoid receptor translocation to the nucleus. J. Physiol. 2008, 586, 3657–3673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Diggle, C.P.; Shires, M.; Leitch, D.; Brooke, D.; Carr, I.M.; Markham, A.F.; Hayward, B.E.; Asipu, A.; Bonthron, D.T. Ketohexokinase: Expression and localization of the principal fructose-metabolizing enzyme. J. Histochem. Cytochem. 2009, 57, 763–774. [Google Scholar] [CrossRef] [PubMed]
  101. Diggle, C.P.; Shires, M.; McRae, C.; Crellin, D.; Fisher, J.; Carr, I.M.; Markham, A.F.; Hayward, B.E.; Asipu, A.; Bonthron, D.T. Both isoforms of ketohexokinase are dispensable for normal growth and development. Physiol. Genomics 2010, 42A, 235–243. [Google Scholar] [CrossRef] [PubMed]
  102. Raivio, K.O.; Kekomaki, M.P.; Maenpaa, P.H. Depletion of liver adenine nucleotides induced by D-fructose. Dose-dependence and specificity of the fructose effect. Biochem. Pharmacol. 1969, 18, 2615–2624. [Google Scholar] [CrossRef]
  103. Lanaspa, M.A.; Andres-Hernando, A.; Orlicky, D.J.; Cicerchi, C.; Jang, C.; Li, N.; Milagres, T.; Kuwabara, M.; Wempe, M.F.; Rabinowitz, J.D.; et al. Ketohexokinase C blockade ameliorates fructose-induced metabolic dysfunction in fructose-sensitive mice. J. Clin. Investig. 2018, 128, 2226–2238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Lanaspa, M.A.; Ishimoto, T.; Li, N.; Cicerchi, C.; Orlicky, D.J.; Ruzycki, P.; Rivard, C.; Inaba, S.; Roncal-Jimenez, C.A.; Bales, E.S.; et al. Endogenous fructose production and metabolism in the liver contributes to the development of metabolic syndrome. Nat. Commun. 2013, 4, 2434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Maenpaa, P.H.; Raivio, K.O.; Kekomaki, M.P. Liver adenine nucleotides: Fructose-induced depletion and its effect on protein synthesis. Science 1968, 161, 1253–1254. [Google Scholar] [CrossRef] [PubMed]
  106. Kurtz, T.W.; Kabra, P.M.; Booth, B.E.; Al-Bander, H.A.; Portale, A.A.; Serena, B.G.; Tsai, H.C.; Morris, R.C., Jr. Liquid-chromatographic measurements of inosine, hypoxanthine, and xanthine in studies of fructose-induced degradation of adenine nucleotides in humans and rats. Clin. Chem. 1986, 32, 782–786. [Google Scholar] [PubMed]
  107. van den Berghe, G.; Bronfman, M.; Vanneste, R.; Hers, H.G. The mechanism of adenosine triphosphate depletion in the liver after a load of fructose. A kinetic study of liver adenylate deaminase. Biochem. J. 1977, 162, 601–609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Xu, C.; Wan, X.; Xu, L.; Weng, H.; Yan, M.; Miao, M.; Sun, Y.; Xu, G.; Dooley, S.; Li, Y.; et al. Xanthine oxidase in non-alcoholic fatty liver disease and hyperuricemia: One stone hits two birds. J. Hepatol. 2015, 62, 1412–1419. [Google Scholar] [CrossRef] [PubMed]
  109. Gonzalez, R.G.; Barnett, P.; Aguayo, J.; Cheng, H.M.; Chylack, L.T., Jr. Direct measurement of polyol pathway activity in the ocular lens. Diabetes 1984, 33, 196–199. [Google Scholar] [CrossRef] [PubMed]
  110. Morrison, A.D.; Clements, R.S., Jr.; Travis, S.B.; Oski, F.; Winegrad, A.I. Glucose utilization by the polyol pathway in human erythrocytes. Biochem. Biophys. Res. Commun. 1970, 40, 199–205. [Google Scholar] [CrossRef]
  111. Sapp, V.; Gaffney, L.; EauClaire, S.F.; Matthews, R.P. Fructose leads to hepatic steatosis in zebrafish that is reversed by mechanistic target of rapamycin (mTOR) inhibition. Hepatology 2014, 60, 1581–1592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Woods, A.; Williams, J.R.; Muckett, P.J.; Mayer, F.V.; Liljevald, M.; Bohlooly, Y.M.; Carling, D. Liver-Specific Activation of AMPK Prevents Steatosis on a High-Fructose Diet. Cell Rep. 2017, 18, 3043–3051. [Google Scholar] [CrossRef] [PubMed]
  113. Hannou, S.A.; Haslam, D.E.; McKeown, N.M.; Herman, M.A. Fructose metabolism and metabolic disease. J. Clin. Investig. 2018, 128, 545–555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Vos, M.B.; Lavine, J.E. Dietary fructose in nonalcoholic fatty liver disease. Hepatology 2013, 57, 2525–2531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Tappy, L. Fructose-containing caloric sweeteners as a cause of obesity and metabolic disorders. J. Exp. Biol. 2018, 221, 164202. [Google Scholar] [CrossRef] [PubMed]
  116. Ter Horst, K.W.; Serlie, M.J. Fructose Consumption, Lipogenesis, and Non-Alcoholic Fatty Liver Disease. Nutrients 2017, 9, 981. [Google Scholar] [CrossRef] [PubMed]
  117. Bonham, M.; O’Connor, J.M.; Hannigan, B.M.; Strain, J.J. The immune system as a physiological indicator of marginal copper status? Br. J. Nutr. 2002, 87, 393–403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Bertinato, J.; L’Abbe, M.R. Copper modulates the degradation of copper chaperone for Cu,Zn superoxide dismutase by the 26 S. proteosome. J. Biol. Chem. 2003, 278, 35071–35078. [Google Scholar] [CrossRef] [PubMed]
  119. Martin, F.; Linden, T.; Katschinski, D.M.; Oehme, F.; Flamme, I.; Mukhopadhyay, C.K.; Eckhardt, K.; Tröger, J.; Barth, S.; Camenisch, G.; et al. Copper-dependent activation of hypoxia-inducible factor (HIF)-1: Implications for ceruloplasmin regulation. Blood 2005, 105, 4613–4619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Gupta, A.; Lutsenko, S. Human copper transporters: Mechanism, role in human diseases and therapeutic potential. Future Med. Chem. 2009, 1, 1125–1142. [Google Scholar] [CrossRef] [PubMed]
  121. Maryon, E.B.; Molloy, S.A.; Ivy, K.; Yu, H.; Kaplan, J.H. Rate and regulation of copper transport by human copper transporter 1 (hCTR1). J. Biol. Chem. 2013, 288, 18035–18046. [Google Scholar] [CrossRef] [PubMed]
  122. Ohrvik, H.; Thiele, D.J. How copper traverses cellular membranes through the mammalian copper transporter 1, Ctr1. Ann. N. Y. Acad. Sci. 2014, 1314, 32–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Nose, Y.; Kim, B.E.; Thiele, D.J. Ctr1 drives intestinal copper absorption and is essential for growth, iron metabolism, and neonatal cardiac function. Cell MeTable 2006, 4, 235–244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Kim, B.E.; Turski, M.L.; Nose, Y.; Casad, M.; Rockman, H.A.; Thiele, D.J. Cardiac copper deficiency activates a systemic signaling mechanism that communicates with the copper acquisition and storage organs. Cell MeTable 2010, 11, 353–363. [Google Scholar] [CrossRef] [PubMed]
  125. Logeman, B.L.; Wood, L.K.; Lee, J.; Thiele, D.J. Gene duplication and neo-functionalization in the evolutionary and functional divergence of the metazoan copper transporters Ctr1 and Ctr2. J. Biol. Chem. 2017, 292, 11531–11546. [Google Scholar] [CrossRef] [PubMed]
  126. Ohrvik, H.; Nose, Y.; Wood, L.K.; Kim, B.E.; Gleber, S.C.; Ralle, M.; Thiele, D.J. Ctr2 regulates biogenesis of a cleaved form of mammalian Ctr1 metal transporter lacking the copper- and cisplatin-binding ecto-domain. Proc. Natl. Acad. Sci. USA 2013, 110, E4279–E4288. [Google Scholar] [CrossRef] [PubMed]
  127. van den Berghe, P.V.; Klomp, L.W. New developments in the regulation of intestinal copper absorption. Nutr. Rev. 2009, 67, 658–672. [Google Scholar] [CrossRef] [PubMed]
  128. Lalioti, V.; Muruais, G.; Tsuchiya, Y.; Pulido, D.; Sandoval, I.V. Molecular mechanisms of copper homeostasis. Front. Biosci. (Landmark Ed.) 2009, 14, 4878–4903. [Google Scholar] [CrossRef] [PubMed]
  129. Lee, S.H.; Kim, M.J.; Kim, Y.S.; Chun, H.; Won, B.Y.; Lee, J.H.; Han, K.; Rim, K.-S.; Park, K.-C. Low hair copper concentration is related to a high risk of nonalcoholic fatty liver disease in adults. J. Trace Elem. Med. Biol. 2018, 50, 28–33. [Google Scholar] [CrossRef] [PubMed]
  130. Stattermayer, A.F.; Traussnigg, S.; Dienes, H.P.; Aigner, E.; Stauber, R.; Lackner, K.; Hofer, H.; Stift, J.; Wrba, F.; Stadlmayr, A.; et al. Hepatic steatosis in Wilson disease—Role of copper and PNPLA3 mutations. J. Hepatol. 2015, 63, 156–163. [Google Scholar] [CrossRef] [PubMed]
  131. Ala, A.; Walker, A.P.; Ashkan, K.; Dooley, J.S.; Schilsky, M.L. Wilson’s disease. Lancet 2007, 369, 397–408. [Google Scholar] [CrossRef]
  132. Huster, D. Structural and metabolic changes in Atp7b-/- mouse liver and potential for new interventions in Wilson’s disease. Ann. N. Y. Acad. Sci. 2014, 1315, 37–44. [Google Scholar] [CrossRef] [PubMed]
  133. Medici, V.; Shibata, N.M.; Kharbanda, K.K.; LaSalle, J.M.; Woods, R.; Liu, S.; Engelberg, J.A.; Devaraj, S.; Török, N.J.; Jiang, J.X.; et al. Wilson’s disease: Changes in methionine metabolism and inflammation affect global DNA methylation in early liver disease. Hepatology 2013, 57, 555–565. [Google Scholar] [CrossRef] [PubMed]
  134. Wooton-Kee, C.R.; Jain, A.K.; Wagner, M.; Grusak, M.A.; Finegold, M.J.; Lutsenko, S.; Moore, D.D. Elevated copper impairs hepatic nuclear receptor function in Wilson’s disease. J. Clin. Investig. 2015, 125, 3449–3460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Porcu, C.; Antonucci, L.; Barbaro, B.; Illi, B.; Nasi, S.; Martini, M.; Licata, A.; Miele, L.; Grieco, A.; Balsano, C. Copper/MYC/CTR1 interplay: A dangerous relationship in hepatocellular carcinoma. Oncotarget 2018, 9, 9325–9343. [Google Scholar] [CrossRef] [PubMed]
  136. Song, M.; Song, Z.; Barve, S.; Zhang, J.; Chen, T.; Liu, M.; Arteel, G.E.; Brewer, G.J.; McClain, C.J. Tetrathiomolybdate protects against bile duct ligation-induced cholestatic liver injury and fibrosis. J. Pharmacol. Exp. Ther. 2008, 325, 409–416. [Google Scholar] [CrossRef] [PubMed]
  137. Song, M.; Zhou, Z.; Chen, T.; Zhang, J.; McClain, C.J. Copper deficiency exacerbates bile duct ligation-induced liver injury and fibrosis in rats. J. Pharmacol. Exp. Ther. 2011, 339, 298–306. [Google Scholar] [CrossRef] [PubMed]
  138. Antonucci, L.; Porcu, C.; Iannucci, G.; Balsano, C.; Barbaro, B. Non-Alcoholic Fatty Liver Disease and Nutritional Implications: Special Focus on Copper. Nutrients 2017, 9, 1137. [Google Scholar] [CrossRef] [PubMed]
  139. Morrell, A.; Tallino, S.; Yu, L.; Burkhead, J.L. The role of insufficient copper in lipid synthesis and fatty-liver disease. IUBMB Life 2017, 69, 263–270. [Google Scholar] [CrossRef] [PubMed]
  140. Tallino, S.; Duffy, M.; Ralle, M.; Cortes, M.P.; Latorre, M.; Burkhead, J.L. Nutrigenomics analysis reveals that copper deficiency and dietary sucrose up-regulate inflammation, fibrosis and lipogenic pathways in a mature rat model of nonalcoholic fatty liver disease. J. Nutr. Biochem. 2015, 26, 996–1006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Song, M.; Schuschke, D.A.; Zhou, Z.; Zhong, W.; Zhang, J.; Zhang, X.; Wang, Y.; McClain, C.J. Kupffer cell depletion protects against the steatosis, but not the liver damage, induced by marginal-copper, high-fructose diet in male rats. Am. J. Physiol. Gastrointest. Liver Physiol. 2015, 308, G934–G945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Kim, S.; Chao, P.Y.; Allen, K.G. Inhibition of elevated hepatic glutathione abolishes copper deficiency cholesterolemia. FASEB J. 1992, 6, 2467–2471. [Google Scholar] [CrossRef] [PubMed]
  143. Tang, Z.; Gasperkova, D.; Xu, J.; Baillie, R.; Lee, J.H.; Clarke, S.D. Copper deficiency induces hepatic fatty acid synthase gene transcription in rats by increasing the nuclear content of mature sterol regulatory element binding protein 1. J. Nutr. 2000, 130, 2915–2921. [Google Scholar] [CrossRef] [PubMed]
  144. Wilson, J.; Kim, S.; Allen, K.G.; Baillie, R.; Clarke, S.D. Hepatic fatty acid synthase gene transcription is induced by a dietary copper deficiency. Am. J. Physiol. 1997, 272, E1124–E1129. [Google Scholar] [CrossRef] [PubMed]
  145. Clement, S.; Juge-Aubry, C.; Sgroi, A.; Conzelmann, S.; Pazienza, V.; Pittet-Cuenod, B.; Meier, C.A.; Negro, F. Monocyte chemoattractant protein-1 secreted by adipose tissue induces direct lipid accumulation in hepatocytes. Hepatology 2008, 48, 799–807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Mandrekar, P.; Ambade, A.; Lim, A.; Szabo, G.; Catalano, D. An essential role for monocyte chemoattractant protein-1 in alcoholic liver injury: Regulation of proinflammatory cytokines and hepatic steatosis in mice. Hepatology 2011, 54, 2185–2197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Fields, M.; Lewis, C.G. Hepatic iron overload may contribute to hypertriglyceridemia and hypercholesterolemia in copper-deficient rats. Metabolism 1997, 46, 377–381. [Google Scholar] [CrossRef]
  148. Fields, M.; Lewis, C.G. Level of dietary iron, not type of dietary fat, is hyperlipidemic in copper-deficient rats. J. Am. Coll. Nutr. 1999, 18, 353–357. [Google Scholar] [CrossRef] [PubMed]
  149. Fields, M.; Lewis, C.G.; Lure, M.D.; Burns, W.A. Dietary ferric vs. ferrous iron in copper-deficient rats fed fructose-based diets. J. Am. Coll. Nutr. 1995, 14, 399–403. [Google Scholar] [CrossRef] [PubMed]
  150. Fields, M.; Lewis, C.G.; Lure, M.D.; Burns, W.A.; Antholine, W.E. The severity of copper deficiency can be ameliorated by deferoxamine. Metabolism 1991, 40, 105–109. [Google Scholar] [CrossRef]
  151. Vulpe, C.D.; Kuo, Y.M.; Murphy, T.L.; Cowley, L.; Askwith, C.; Libina, N.; Gitschier, J.; Anderson, G.J. Hephaestin, a ceruloplasmin homologue implicated in intestinal iron transport, is defective in the sla mouse. Nat. Genet. 1999, 21, 195–199. [Google Scholar] [CrossRef] [PubMed]
  152. Cherukuri, S.; Potla, R.; Sarkar, J.; Nurko, S.; Harris, Z.L.; Fox, P.L. Unexpected role of ceruloplasmin in intestinal iron absorption. Cell MeTable 2005, 2, 309–319. [Google Scholar] [CrossRef] [PubMed]
  153. Harris, Z.L.; Durley, A.P.; Man, T.K.; Gitlin, J.D. Targeted gene disruption reveals an essential role for ceruloplasmin in cellular iron efflux. Proc. Natl. Acad. Sci. USA 1999, 96, 10812–10817. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Prohaska, J.R. Changes in Cu,Zn-superoxide dismutase, cytochrome c oxidase, glutathione peroxidase and glutathione transferase activities in copper-deficient mice and rats. J. Nutr. 1991, 121, 355–363. [Google Scholar] [CrossRef] [PubMed]
  155. Dallman, P.R. Cytochrome oxidase repair during treatment of copper deficiency: Relation to mitochondrial turnover. J. Clin. Investig. 1967, 46, 1819–1827. [Google Scholar] [CrossRef] [PubMed]
  156. Gallagher, C.H.; Reeve, V.E.; Wright, R. Copper deficiency in the rat. Effect on the ultrastructure of hepatocytes. Aust. J. Exp. Biol. Med. Sci. 1973, 51, 181–189. [Google Scholar] [CrossRef] [PubMed]
  157. Ishida, S.; Andreux, P.; Poitry-Yamate, C.; Auwerx, J.; Hanahan, D. Bioavailable copper modulates oxidative phosphorylation and growth of tumors. Proc. Natl. Acad. Sci. USA 2013, 110, 19507–19512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Fields, M.; Lewis, C.; Scholfield, D.J.; Powell, A.S.; Rose, A.J.; Reiser, S.; Smith, J.C. Female rats are protected against the fructose induced mortality of copper deficiency. Proc. Soc. Exp. Biol. Med. 1986, 183, 145–149. [Google Scholar] [CrossRef] [PubMed]
  159. Xue, Q.; Aliabadi, H.; Hallfrisch, J. Effects of dietary galactose and fructose on rats fed diets marginal or adequate in copper for 9–21 months. Nutr. Res. 2001, 21, 1078–1087. [Google Scholar] [CrossRef]
  160. Fields, M.; Lewis, C.G.; Lure, M.D. Allopurinol, an inhibitor of xanthine oxidase, reduces uric acid levels and modifies the signs associated with copper deficiency in rats fed fructose. Free Radic. Biol. Med. 1996, 20, 595–600. [Google Scholar] [CrossRef]
  161. Rossi, L.; Lippe, G.; Marchese, E.; De Martino, A.; Mavelli, I.; Rotilio, G.; Ciriolo, M.R. Decrease of cytochrome c oxidase protein in heart mitochondria of copper-deficient rats. Biometals 1998, 11, 207–212. [Google Scholar] [CrossRef] [PubMed]
  162. Baker, Z.N.; Cobine, P.A.; Leary, S.C. The mitochondrion: A central architect of copper homeostasis. Metallomics 2017, 9, 1501–1512. [Google Scholar] [CrossRef] [PubMed]
  163. Leary, S.C. Redox regulation of SCO protein function: Controlling copper at a mitochondrial crossroad. Antioxid. Redox Signal. 2010, 13, 1403–1416. [Google Scholar] [CrossRef] [PubMed]
  164. Hill, S.; Deepa, S.S.; Sataranatarajan, K.; Premkumar, P.; Pulliam, D.; Liu, Y.; Soto, V.Y.; Fischer, K.E.; Van Remmen, H. Sco2 deficient mice develop increased adiposity and insulin resistance. Mol. Cell. Endocrinol. 2017, 455, 103–114. [Google Scholar] [CrossRef] [PubMed]
  165. Jaksch, M.; Paret, C.; Stucka, R.; Horn, N.; Muller-Hocker, J.; Horvath, R.; Trepesch, N.; Stecker, G.; Freisinger, P.; Thirion, C.; et al. Cytochrome c oxidase deficiency due to mutations in SCO2, encoding a mitochondrial copper-binding protein, is rescued by copper in human myoblasts. Hum. Mol. Genet. 2001, 10, 3025–3035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Deepa, S.S.; Pharaoh, G.; Kinter, M.; Diaz, V.; Fok, W.C.; Riddle, K.; Pulliam, D.; Hill, S.; Fischer, K.E.; Soto, V.; et al. Lifelong reduction in complex IV induces tissue-specific metabolic effects but does not reduce lifespan or healthspan in mice. Aging Cell 2018, e12769. [Google Scholar] [CrossRef] [PubMed]
  167. Deepa, S.S.; Pulliam, D.; Hill, S.; Shi, Y.; Walsh, M.E.; Salmon, A.; Sloane, L.; Zhang, N.; Zeviani, M.; Viscomi, C.; et al. Improved insulin sensitivity associated with reduced mitochondrial complex IV assembly and activity. FASEB J. 2013, 27, 1371–1380. [Google Scholar] [CrossRef] [PubMed]
  168. Pulliam, D.A.; Deepa, S.S.; Liu, Y.; Hill, S.; Lin, A.L.; Bhattacharya, A.; Shi, Y.; Sloane, L.; Viscomi, C.; Zeviani, M.; et al. Complex IV-deficient Surf1(-/-) mice initiate mitochondrial stress responses. Biochem. J. 2014, 462, 359–371. [Google Scholar] [CrossRef] [PubMed]
  169. Hamilton, J.P.; Koganti, L.; Muchenditsi, A.; Pendyala, V.S.; Huso, D.; Hankin, J.; Murphy, R.C.; Huster, D.; Merle, U.; Mangels, C.; et al. Activation of liver X receptor/retinoid X receptor pathway ameliorates liver disease in Atp7B-/- (Wilson disease) mice. Hepatology 2016, 63, 1828–1841. [Google Scholar] [CrossRef] [PubMed]
  170. Bo, S.; Durazzo, M.; Gambino, R.; Berutti, C.; Milanesio, N.; Caropreso, A.; Gentile, L.; Cassader, M.; Cavallo-Perin, P.; Pagano, G. Associations of dietary and serum copper with inflammation, oxidative stress, and metabolic variables in adults. J. Nutr. 2008, 138, 305–310. [Google Scholar] [CrossRef] [PubMed]
  171. Fields, M.; Lure, M.D.; Lewis, C.G. Effect of saturated versus unsaturated fat on the pathogenesis of copper deficiency in rats. J. Nutr. Biochem. 1996, 7, 246–251. [Google Scholar] [CrossRef]
  172. Allen, K.G.; Klevay, L.M. Copper deficiency and cholesterol metabolism in the rat. Atherosclerosis 1978, 31, 259–271. [Google Scholar] [CrossRef]
  173. Al-Othman, A.A.; Rosenstein, F.; Lei, K.Y. Pool size and concentration of plasma cholesterol are increased and tissue copper levels are reduced during early stages of copper deficiency in rats. J. Nutr. 1994, 124, 628–635. [Google Scholar] [CrossRef] [PubMed]
  174. Fields, M.; Lewis, C.G.; Lure, M.D.; Burns, W.A.; Antholine, W.E. Low dietary iron prevents free radical formation and heart pathology of copper-deficient rats fed fructose. Proc. Soc. Exp. Biol. Med. 1993, 202, 225–232. [Google Scholar] [CrossRef] [PubMed]
  175. Chao, P.Y.; Allen, K.G. Glutathione production in copper-deficient isolated rat hepatocytes. Free Radic. Biol. Med. 1992, 12, 145–150. [Google Scholar] [PubMed]
  176. Hassel, C.A.; Marchello, J.A.; Lei, K.Y. Impaired glucose tolerance in copper-deficient rats. J. Nutr. 1983, 113, 1081–1083. [Google Scholar] [CrossRef] [PubMed]
  177. Fields, M.; Lewis, C.G.; Lure, M.D. Responses of insulin to oral glucose and fructose loads in marginally copper-deficient rats fed starch or fructose. Nutrition 1996, 12, 524–528. [Google Scholar] [CrossRef]
  178. Kelly, C.J.; Zheng, L.; Campbell, E.L.; Saeedi, B.; Scholz, C.C.; Bayless, A.J.; Wilson, K.E.; Glover, L.E.; Kominsky, D.J.; Magnuson, A.; et al. Crosstalk between Microbiota-Derived Short-Chain Fatty Acids and Intestinal Epithelial HIF Augments Tissue Barrier Function. Cell Host Microbe 2015, 17, 662–671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Karhausen, J.; Furuta, G.T.; Tomaszewski, J.E.; Johnson, R.S.; Colgan, S.P.; Haase, V.H. Epithelial hypoxia-inducible factor-1 is protective in murine experimental colitis. J. Clin. Investig. 2004, 114, 1098–1106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Saeedi, B.J.; Kao, D.J.; Kitzenberg, D.A.; Dobrinskikh, E.; Schwisow, K.D.; Masterson, J.C.; Kendrick, A.A.; Kelly, C.J.; Bayless, A.J.; Kominsky, D.J.; et al. HIF-dependent regulation of claudin-1 is central to intestinal epithelial tight junction integrity. Mol. Biol. Cell 2015, 26, 2252–2262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Fluck, K.; Breves, G.; Fandrey, J.; Winning, S. Hypoxia-inducible factor 1 in dendritic cells is crucial for the activation of protective regulatory T cells in murine colitis. Mucosal Immunol. 2016, 9, 379–390. [Google Scholar] [CrossRef] [PubMed]
  182. Clambey, E.T.; McNamee, E.N.; Westrich, J.A.; Glover, L.E.; Campbell, E.L.; Jedlicka, P.; de Zoeten, E.F.; Cambier, J.C.; Stenmark, K.R.; Colgan, S.P.; et al. Hypoxia-inducible factor-1 alpha-dependent induction of FoxP3 drives regulatory T-cell abundance and function during inflammatory hypoxia of the mucosa. Proc. Natl. Acad. Sci. USA 2012, 109, E2784–E2793. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Feng, W.; Ye, F.; Xue, W.; Zhou, Z.; Kang, Y.J. Copper regulation of hypoxia-inducible factor-1 activity. Mol. Pharmacol. 2009, 75, 174–182. [Google Scholar] [CrossRef] [PubMed]
  184. Wei, X.; Song, M.; Yin, X.; Schuschke, D.A.; Koo, I.; McClain, C.J.; Zhang, X. Effects of Dietary Different Doses of Copper and High Fructose Feeding on Rat Fecal Metabolome. J. Proteome Res. 2015, 14, 4050–4058. [Google Scholar] [CrossRef] [PubMed]
  185. Ferruzza, S.; Scacchi, M.; Scarino, M.L.; Sambuy, Y. Iron and copper alter tight junction permeability in human intestinal Caco-2 cells by distinct mechanisms. Toxicol. In Vitro 2002, 16, 399–404. [Google Scholar] [CrossRef]
  186. Rossi, A.; Poverini, R.; Di Lullo, G.; Modesti, A.; Modica, A.; Scarino, M.L. Heavy metal toxicity following apical and basolateral exposure in the human intestinal cell line Caco-2. Toxicol. In Vitro 1996, 10, 27–36. [Google Scholar] [CrossRef]
  187. Santos, S.; Silva, A.M.; Matos, M.; Monteiro, S.M.; Alvaro, A.R. Copper induced apoptosis in Caco-2 and Hep-G2 cells: Expression of caspases 3, 8 and 9, AIF and p53. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2016, 185–186, 138–146. [Google Scholar] [CrossRef] [PubMed]
  188. Hodgkinson, V.; Petris, M.J. Copper homeostasis at the host-pathogen interface. J. Biol. Chem. 2012, 287, 13549–13555. [Google Scholar] [CrossRef] [PubMed]
  189. Pontel, L.B.; Soncini, F.C. Alternative periplasmic copper-resistance mechanisms in Gram negative bacteria. Mol. Microbiol. 2009, 73, 212–225. [Google Scholar] [CrossRef] [PubMed]
  190. Solioz, M.; Abicht, H.K.; Mermod, M.; Mancini, S. Response of gram-positive bacteria to copper stress. J. Biol. Inorg. Chem. 2010, 15, 3–14. [Google Scholar] [CrossRef] [PubMed]
  191. Kehoe, C.A.; Faughnan, M.S.; Gilmore, W.S.; Coulter, J.S.; Howard, A.N.; Strain, J.J. Plasma diamine oxidase activity is greater in copper-adequate than copper-marginal or copper-deficient rats. J. Nutr. 2000, 130, 30–33. [Google Scholar] [CrossRef] [PubMed]
  192. Legleiter, L.R.; Spears, J.W. Plasma diamine oxidase: A biomarker of copper deficiency in the bovine. J. Anim. Sci. 2007, 85, 2198–2204. [Google Scholar] [CrossRef] [PubMed]
  193. Luk, G.D.; Bayless, T.M.; Baylin, S.B. Diamine oxidase (histaminase). A circulating marker for rat intestinal mucosal maturation and integrity. J. Clin. Investig. 1980, 66, 66–70. [Google Scholar] [CrossRef] [PubMed]
  194. Galipeau, D.; Verma, S.; McNeill, J.H. Female rats are protected against fructose-induced changes in metabolism and blood pressure. Am. J. Physiol. Heart Circ. Physiol. 2002, 283, H2478–H2484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Fields, M.; Lewis, C.G.; Beal, T.; Scholfield, D.; Patterson, K.; Smith, J.C.; Reiser, S. Sexual differences in the expression of copper deficiency in rats. Proc. Soc. Exp. Biol. Med. 1987, 186, 183–187. [Google Scholar] [CrossRef] [PubMed]
  196. Fields, M.; Lewis, C.G.; Lure, M.; Antholine, W.E. The influence of gender on developing copper deficiency and on free radical generation of rats fed a fructose diet. Metabolism 1992, 41, 989–994. [Google Scholar] [CrossRef]
  197. Bantle, J.P.; Raatz, S.K.; Thomas, W.; Georgopoulos, A. Effects of dietary fructose on plasma lipids in healthy subjects. Am. J. Clin. Nutr. 2000, 72, 1128–1134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Couchepin, C.; Le, K.A.; Bortolotti, M.; da Encarnacao, J.A.; Oboni, J.B.; Tran, C.; Schneiter, P.; Tappy, L. Markedly blunted metabolic effects of fructose in healthy young female subjects compared with male subjects. Diabetes Care 2008, 31, 1254–1256. [Google Scholar] [CrossRef] [PubMed]
  199. Pan, J.J.; Fallon, M.B. Gender and racial differences in nonalcoholic fatty liver disease. World J. Hepatol. 2014, 6, 274–283. [Google Scholar] [CrossRef] [PubMed]
  200. Latour, C.; Kautz, L.; Besson-Fournier, C.; Island, M.L.; Canonne-Hergaux, F.; Loreal, O.; Ganz, T.; Coppin, H.; Roth, M.P. Testosterone perturbs systemic iron balance through activation of epidermal growth factor receptor signaling in the liver and repression of hepcidin. Hepatology 2014, 59, 683–694. [Google Scholar] [CrossRef] [PubMed]
  201. Werman, M.J.; Bhathena, S.J. Fructose metabolizing enzymes in the rat liver and metabolic parameters: Interactions between dietary copper, type of carbohydrates, and gender. J. Nutr. Biochem. 1995, 6, 373–379. [Google Scholar] [CrossRef] [Green Version]
  202. Millo, H.; Werman, M.J. Hepatic fructose-metabolizing enzymes and related metabolites: Role of dietary copper and gender. J. Nutr. Biochem. 2000, 11, 374–381. [Google Scholar] [CrossRef]
  203. Rodriguez, A.; Marinelli, R.A.; Tesse, A.; Fruhbeck, G.; Calamita, G. Sexual Dimorphism of Adipose and Hepatic Aquaglyceroporins in Health and Metabolic Disorders. Front. Endocrinol. (Lausanne) 2015, 6, 171. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Fructose and Glucose metabolism. AR, aldose reductase; SDH, sorbitol dehydrogenase; KHK, ketohexokinase; PFK, phosphofructokinase; AMPD, adenosine monophosphate deaminase; IMP, inosine monophosphate; XO, xanthine oxidase; ACC, acetyl-CoA carboxylase; FAS, fatty acid synthase; ACSL, long chain acyl-CoA synthetase.
Figure 1. Fructose and Glucose metabolism. AR, aldose reductase; SDH, sorbitol dehydrogenase; KHK, ketohexokinase; PFK, phosphofructokinase; AMPD, adenosine monophosphate deaminase; IMP, inosine monophosphate; XO, xanthine oxidase; ACC, acetyl-CoA carboxylase; FAS, fatty acid synthase; ACSL, long chain acyl-CoA synthetase.
Nutrients 10 01815 g001
Figure 2. Cellular copper distribution. Ctr1, copper transporter 1; MT, metallothionein; GSH, glutathione; CCS, copper chaperone for SOD1; COX, cytochrome c oxidase; Atox1, antioxidant protein 1.
Figure 2. Cellular copper distribution. Ctr1, copper transporter 1; MT, metallothionein; GSH, glutathione; CCS, copper chaperone for SOD1; COX, cytochrome c oxidase; Atox1, antioxidant protein 1.
Nutrients 10 01815 g002
Figure 3. Effects of marginal copper deficiency and fructose feeding on liver injury and lipid accumulation in male weanling Sprague-Dawley rats. (A) Plasma AST. (B) Representative photomicrographs of the H&E and Oil Red O staining of liver section (200×). Data represent means ± SD (n = 5–9) and analyzed by two-way ANOVA, * p < 0.05; #, interaction between copper and fructose is significant (p < 0.05). AST, aspartate aminotransferase; A, adequate copper diet; M, marginal copper deficient diet; AF, adequate copper diet + 30% fructose drinking; MF, marginal copper deficient diet + 30% fructose drinking.
Figure 3. Effects of marginal copper deficiency and fructose feeding on liver injury and lipid accumulation in male weanling Sprague-Dawley rats. (A) Plasma AST. (B) Representative photomicrographs of the H&E and Oil Red O staining of liver section (200×). Data represent means ± SD (n = 5–9) and analyzed by two-way ANOVA, * p < 0.05; #, interaction between copper and fructose is significant (p < 0.05). AST, aspartate aminotransferase; A, adequate copper diet; M, marginal copper deficient diet; AF, adequate copper diet + 30% fructose drinking; MF, marginal copper deficient diet + 30% fructose drinking.
Nutrients 10 01815 g003
Figure 4. Effect of fructose feeding on liver iron in male weanling Sprague-Dawley rats. Data represent means ± SD (n = 5–10) and analyzed by two-way ANOVA, * p < 0.05; A, adequate copper diet; M, marginal copper deficient diet; AF, adequate copper diet + 3% fructose drinking; MF, marginal copper deficient diet + 3% fructose drinking.
Figure 4. Effect of fructose feeding on liver iron in male weanling Sprague-Dawley rats. Data represent means ± SD (n = 5–10) and analyzed by two-way ANOVA, * p < 0.05; A, adequate copper diet; M, marginal copper deficient diet; AF, adequate copper diet + 3% fructose drinking; MF, marginal copper deficient diet + 3% fructose drinking.
Nutrients 10 01815 g004

Share and Cite

MDPI and ACS Style

Song, M.; Vos, M.B.; McClain, C.J. Copper-Fructose Interactions: A Novel Mechanism in the Pathogenesis of NAFLD. Nutrients 2018, 10, 1815. https://doi.org/10.3390/nu10111815

AMA Style

Song M, Vos MB, McClain CJ. Copper-Fructose Interactions: A Novel Mechanism in the Pathogenesis of NAFLD. Nutrients. 2018; 10(11):1815. https://doi.org/10.3390/nu10111815

Chicago/Turabian Style

Song, Ming, Miriam B. Vos, and Craig J. McClain. 2018. "Copper-Fructose Interactions: A Novel Mechanism in the Pathogenesis of NAFLD" Nutrients 10, no. 11: 1815. https://doi.org/10.3390/nu10111815

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop