1
|
Buziau AM, Lefeber DJ, Cassiman D, Rubio‐Gozalbo ME, Kwast H, Tolan DR, Schalkwijk CG, Brouwers MCGJ. Aldolase B Deficient Mice Are Characterized by Hepatic Nucleotide Sugar Abnormalities. J Inherit Metab Dis 2025; 48:e12836. [PMID: 39727106 PMCID: PMC11672228 DOI: 10.1002/jimd.12836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024]
Abstract
Hereditary fructose intolerance (HFI) is characterized by liver damage and a secondary defect in N-linked glycosylation due to impairment of mannose phosphate isomerase (MPI). Mannose treatment has been shown to be an effective treatment in a primary defect in MPI (i.e., MPI-CDG), which is also characterized by liver damage. Therefore, the aims of this study were to determine: (1) hepatic nucleotide sugar levels, and (2), the effect of mannose supplementation on hepatic nucleotide sugar levels and liver fat, in a mouse model for HFI. Aldolase B deficient mice (Aldob-/-) were treated for four weeks with 5% mannose via the drinking water and compared to Aldob-/- mice and wildtype mice treated with regular drinking water. We found that hepatic GDP-mannose and hepatic GDP-fucose were lower in water-treated Aldob-/- mice when compared to water-treated wildtype mice (p = 0.002 and p = 0.002, respectively), consistent with impaired N-linked glycosylation. Of interest, multiple other hepatic nucleotide sugars not involved in N-linked glycosylation, such as hepatic UDP-glucuronic acid, UDP-xylose, CMP-N-acetyl-beta-neuraminic acid, and CDP-ribitol (p = 0.002, p = 0.003, p = 0.002, p = 0.002), were found to have altered levels as well. However, mannose treatment did not correct the reduction in hepatic GDP-mannose levels, nor was liver fat affected. Aldob-/- mice are characterized by hepatic nucleotide sugar abnormalities, but these were not abrogated by mannose treatment. Future studies are needed to identify the underlying mechanisms responsible for the abnormal hepatic nucleotide sugar pattern and intrahepatic lipid accumulation in HFI. Trial Registration: PCT ID: PCTE0000340, this animal experiment is registered at (https://preclinicaltrials.eu/).
Collapse
Affiliation(s)
- Amée M. Buziau
- Department of Internal Medicine, Division of Endocrinology and Metabolic DiseaseMaastricht University Medical Center+MaastrichtThe Netherlands
- Cardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
- Department of Internal Medicine, Division of General Internal Medicine, Laboratory for Metabolism and Vascular MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - Dirk J. Lefeber
- Translational Metabolic Laboratory, Department of Human GeneticsRadboud University Medical CenterNijmegenThe Netherlands
- Department of Neurology, Donders Institute for Brain, Cognition and BehaviorRadboud University Medical CenterNijmegenThe Netherlands
| | - D. Cassiman
- Department of Gastroenterology‐Hepatology and Metabolic CenterUniversity Hospital LeuvenLeuvenBelgium
| | - M. Estela Rubio‐Gozalbo
- Department of Pediatrics and Clinical GeneticsMaastricht University Medical Center+MaastrichtThe Netherlands
| | - Hanneke Kwast
- Translational Metabolic Laboratory, Department of Human GeneticsRadboud University Medical CenterNijmegenThe Netherlands
| | - Dean R. Tolan
- Department of BiologyBoston UniversityBostonMassachusettsUSA
| | - Casper G. Schalkwijk
- Cardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
- Department of Internal Medicine, Division of General Internal Medicine, Laboratory for Metabolism and Vascular MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - Martijn C. G. J. Brouwers
- Department of Internal Medicine, Division of Endocrinology and Metabolic DiseaseMaastricht University Medical Center+MaastrichtThe Netherlands
- Cardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
- Department of Internal MedicineMaastricht University Medical Center+MaastrichtThe Netherlands
- United for Metabolic DiseasesThe Netherlands
| |
Collapse
|
2
|
Buziau AM, Oosterveer MH, Wouters K, Bos T, Tolan DR, Agius L, Ford BE, Cassiman D, Stehouwer CDA, Schalkwijk CG, Brouwers MCGJ. Hepatic glucokinase regulatory protein and carbohydrate response element binding protein attenuation reduce de novo lipogenesis but do not mitigate intrahepatic triglyceride accumulation in Aldob deficiency. Mol Metab 2024; 87:101984. [PMID: 38972375 PMCID: PMC11300931 DOI: 10.1016/j.molmet.2024.101984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/15/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024] Open
Abstract
OBJECTIVE Stable isotope studies have shown that hepatic de novo lipogenesis (DNL) plays an important role in the pathogenesis of intrahepatic lipid (IHL) deposition. Furthermore, previous research has demonstrated that fructose 1-phosphate (F1P) not only serves as a substrate for DNL, but also acts as a signalling metabolite that stimulates DNL from glucose. The aim of this study was to elucidate the mediators of F1P-stimulated DNL, with special focus on two key regulators of intrahepatic glucose metabolism, i.e., glucokinase regulatory protein (GKRP) and carbohydrate response element binding protein (ChREBP). METHODS Aldolase B deficient mice (Aldob-/-), characterized by hepatocellular F1P accumulation, enhanced DNL, and hepatic steatosis, were either crossed with GKRP deficient mice (Gckr-/-) or treated with short hairpin RNAs directed against hepatic ChREBP. RESULTS Aldob-/- mice showed higher rates of de novo palmitate synthesis from glucose when compared to wildtype mice (p < 0.001). Gckr knockout reduced de novo palmitate synthesis in Aldob-/- mice (p = 0.017), without affecting the hepatic mRNA expression of enzymes involved in DNL. In contrast, hepatic ChREBP knockdown normalized the hepatic mRNA expression levels of enzymes involved in DNL and reduced fractional DNL in Aldob-/- mice (p < 0.05). Of interest, despite downregulation of DNL in response to Gckr and ChREBP attenuation, no reduction in intrahepatic triglyceride levels was observed. CONCLUSIONS Both GKRP and ChREBP mediate F1P-stimulated DNL in aldolase B deficient mice. Further studies are needed to unravel the role of GKRP and hepatic ChREBP in regulating IHL accumulation in aldolase B deficiency.
Collapse
Affiliation(s)
- Amée M Buziau
- Department of Internal Medicine, Division of Endocrinology and Metabolic Disease, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of Internal Medicine, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Maaike H Oosterveer
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Kristiaan Wouters
- Department of Internal Medicine, Division of Endocrinology and Metabolic Disease, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of Internal Medicine, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Trijnie Bos
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Dean R Tolan
- Department of Biology, Boston University, Boston, MA, USA
| | - Loranne Agius
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Brian E Ford
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - David Cassiman
- Department of Gastroenterology-Hepatology and Metabolic Center, University Hospital Leuven, Leuven, Belgium
| | - Coen D A Stehouwer
- Department of Internal Medicine, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Casper G Schalkwijk
- Department of Internal Medicine, Division of Endocrinology and Metabolic Disease, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of Internal Medicine, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Martijn C G J Brouwers
- Department of Internal Medicine, Division of Endocrinology and Metabolic Disease, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of Internal Medicine, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands; Department of Internal Medicine, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht University Medical Center+, Maastricht, the Netherlands.
| |
Collapse
|
3
|
Lodge M, Dykes R, Kennedy A. Regulation of Fructose Metabolism in Nonalcoholic Fatty Liver Disease. Biomolecules 2024; 14:845. [PMID: 39062559 PMCID: PMC11274671 DOI: 10.3390/biom14070845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Elevations in fructose consumption have been reported to contribute significantly to an increased incidence of obesity and metabolic diseases in industrial countries. Mechanistically, a high fructose intake leads to the dysregulation of glucose, triglyceride, and cholesterol metabolism in the liver, and causes elevations in inflammation and drives the progression of nonalcoholic fatty liver disease (NAFLD). A high fructose consumption is considered to be toxic to the body, and there are ongoing measures to develop pharmaceutical therapies targeting fructose metabolism. Although a large amount of work has summarized the effects fructose exposure within the intestine, liver, and kidney, there remains a gap in our knowledge regarding how fructose both indirectly and directly influences immune cell recruitment, activation, and function in metabolic tissues, which are essential to tissue and systemic inflammation. The most recent literature demonstrates that direct fructose exposure regulates oxidative metabolism in macrophages, leading to inflammation. The present review highlights (1) the mechanisms by which fructose metabolism impacts crosstalk between tissues, nonparenchymal cells, microbes, and immune cells; (2) the direct impact of fructose on immune cell metabolism and function; and (3) therapeutic targets of fructose metabolism to treat NAFLD. In addition, the review highlights how fructose disrupts liver tissue homeostasis and identifies new therapeutic targets for treating NAFLD and obesity.
Collapse
Affiliation(s)
| | | | - Arion Kennedy
- Department of Molecular and Structural Biochemistry, North Carolina State University, 128 Polk Hall Campus, Box 7622, Raleigh, NC 27695, USA
| |
Collapse
|
4
|
Andres-Hernando A, Orlicky DJ, Kuwabara M, Fini MA, Tolan DR, Johnson RJ, Lanaspa MA. Activation of AMPD2 drives metabolic dysregulation and liver disease in mice with hereditary fructose intolerance. Commun Biol 2024; 7:849. [PMID: 38992061 PMCID: PMC11239681 DOI: 10.1038/s42003-024-06539-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 07/03/2024] [Indexed: 07/13/2024] Open
Abstract
Hereditary fructose intolerance (HFI) is a painful and potentially lethal genetic disease caused by a mutation in aldolase B resulting in accumulation of fructose-1-phosphate (F1P). No cure exists for HFI and treatment is limited to avoid exposure to fructose and sugar. Using aldolase B deficient mice, here we identify a yet unrecognized metabolic event activated in HFI and associated with the progression of the disease. Besides the accumulation of F1P, here we show that the activation of the purine degradation pathway is a common feature in aldolase B deficient mice exposed to fructose. The purine degradation pathway is a metabolic route initiated by adenosine monophosphate deaminase 2 (AMPD2) that regulates overall energy balance. We demonstrate that very low amounts of fructose are sufficient to activate AMPD2 in these mice via a phosphate trap. While blocking AMPD2 do not impact F1P accumulation and the risk of hypoglycemia, its deletion in hepatocytes markedly improves the metabolic dysregulation induced by fructose and corrects fat and glycogen storage while significantly increasing the voluntary tolerance of these mice to fructose. In summary, we provide evidence for a critical pathway activated in HFI that could be targeted to improve the metabolic consequences associated with fructose consumption.
Collapse
Affiliation(s)
- Ana Andres-Hernando
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Denver, Aurora, CO, USA
| | - David J Orlicky
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Masanari Kuwabara
- Department of Cardiology, Toranomon Hospital, Tokyo, Japan
- Division of Public Health, Center for Community Medicine, Jichi Medical University, Tochigi, Japan
| | - Mehdi A Fini
- Division of Pulmonary and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dean R Tolan
- Department of Biology, Boston University, Boston, MA, USA
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, USA
| | - Miguel A Lanaspa
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Denver, Aurora, CO, USA.
| |
Collapse
|
5
|
Garbowski L, Walasek M, Firszt R, Chilińska-Kopko E, Błażejewska-Gała P, Popielnicki D, Dzięcioł-Anikiej Z. A Case Study of a Rare Disease (Fructosemia) Diagnosed in a Patient with Abdominal Pain. J Clin Med 2024; 13:3394. [PMID: 38929922 PMCID: PMC11204229 DOI: 10.3390/jcm13123394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/22/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Hereditary fructose intolerance is a rare genetic disorder that is inherited in an autosomal recessive manner, with mutations sometimes occurring spontaneously. Consuming fructose triggers biochemical abnormalities, disrupting liver processes like glycogenolysis and gluconeogenesis. Recent studies have revealed elevated intrahepatic fat levels in affected individuals. Symptoms include aversion to fructose-containing foods, hypoglycemia, liver and kidney dysfunction, and growth delays, with severe cases leading to liver enlargement, fatty liver disease, kidney failure, and life-threatening hypoglycemia. In this case study, we present a 20-month-old child with symptoms including difficulty passing stool, abdominal rigidity, abdominal pain with bloating and hypoglycemia. Initial clinical findings revealed elevated liver enzymes, a mildly enlarged hyperechoic liver, hypercholesterolemia, and borderline alpha-fetoprotein values. Diagnostic assessments identified hereditary fructose intolerance (HFI) with pathogenic variants in the ALDOB gene, along with a diagnosis of celiac disease. Genetic testing of the parents revealed carrier status for pathological aldolase B genes. This case underscores the importance of comprehensive clinical evaluation and genetic testing in pediatric patients with complex metabolic presentations.
Collapse
Affiliation(s)
- Leszek Garbowski
- Public Independent Healthcare Services of the Ministry of Internal Affairs and Administration in Białystok, 15-471 Białystok, Poland
- Department of Human Anatomy, Medical University of Białystok, 15-089 Białystok, Poland (P.B.-G.); (D.P.)
| | - Marzena Walasek
- Public Independent Healthcare Services of the Ministry of Internal Affairs and Administration in Białystok, 15-471 Białystok, Poland
| | - Rafał Firszt
- Department of Ornamental Plants and Garden Art, Faculty of Biotechnology and Horticulture, University of Agriculture in Krakow, 31-425 Kraków, Poland;
| | - Ewelina Chilińska-Kopko
- Department of Human Anatomy, Medical University of Białystok, 15-089 Białystok, Poland (P.B.-G.); (D.P.)
| | - Paulina Błażejewska-Gała
- Department of Human Anatomy, Medical University of Białystok, 15-089 Białystok, Poland (P.B.-G.); (D.P.)
- Department of Neonatology and Newborn Intensive Care, University Clinical Hospital in Białystok, 15-276 Białystok, Poland
| | - Daniel Popielnicki
- Department of Human Anatomy, Medical University of Białystok, 15-089 Białystok, Poland (P.B.-G.); (D.P.)
| | - Zofia Dzięcioł-Anikiej
- Department of Rehabilitation, University Clinical Hospital in Białystok, 15-276 Białystok, Poland
| |
Collapse
|
6
|
Andres-Hernando A, Orlicky DJ, Kuwabara M, Cicerchi C, Pedler M, Petrash MJ, Johnson RJ, Tolan DR, Lanaspa MA. Endogenous Fructose Production and Metabolism Drive Metabolic Dysregulation and Liver Disease in Mice with Hereditary Fructose Intolerance. Nutrients 2023; 15:4376. [PMID: 37892451 PMCID: PMC10609559 DOI: 10.3390/nu15204376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/06/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Excessive intake of sugar, and particularly fructose, is closely associated with the development and progression of metabolic syndrome in humans and animal models. However, genetic disorders in fructose metabolism have very different consequences. While the deficiency of fructokinase, the first enzyme involved in fructose metabolism, is benign and somewhat desirable, missense mutations in the second enzyme, aldolase B, causes a very dramatic and sometimes lethal condition known as hereditary fructose intolerance (HFI). To date, there is no cure for HFI, and treatment is limited to avoiding fructose and sugar. Because of this, for subjects with HFI, glucose is their sole source of carbohydrates in the diet. However, clinical symptoms still occur, suggesting that either low amounts of fructose are still being consumed or, alternatively, fructose is being produced endogenously in the body. Here, we demonstrate that as a consequence of consuming high glycemic foods, the polyol pathway, a metabolic route in which fructose is produced from glucose, is activated, triggering a deleterious mechanism whereby glucose, sorbitol and alcohol induce severe liver disease and growth retardation in aldolase B knockout mice. We show that generically and pharmacologically blocking this pathway significantly improves metabolic dysfunction and thriving and increases the tolerance of aldolase B knockout mice to dietary triggers of endogenous fructose production.
Collapse
Affiliation(s)
- Ana Andres-Hernando
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Denver, Aurora, CO 80045, USA;
| | - David J. Orlicky
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO 80045, USA;
| | - Masanari Kuwabara
- Department of Cardiology, Toranomon Hospital, Tokyo 105-8470, Japan;
- Division of Public Health, Center for Community Medicine, Jichi Medical University, Tochigi 329-0431, Japan
| | - Christina Cicerchi
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO 80045, USA; (C.C.); (R.J.J.)
| | - Michelle Pedler
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO 80045, USA; (M.P.); (M.J.P.)
| | - Mark J. Petrash
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO 80045, USA; (M.P.); (M.J.P.)
| | - Richard J. Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO 80045, USA; (C.C.); (R.J.J.)
| | - Dean R. Tolan
- Department of Biology, Boston University, Boston, MA 02215, USA;
| | - Miguel A. Lanaspa
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Denver, Aurora, CO 80045, USA;
| |
Collapse
|
7
|
Gu L, Zhu Y, Watari K, Lee M, Liu J, Perez S, Thai M, Mayfield JE, Zhang B, Cunha E Rocha K, Li F, Kim LC, Jones AC, Wierzbicki IH, Liu X, Newton AC, Kisseleva T, Lee JH, Ying W, Gonzalez DJ, Saltiel AR, Simon MC, Karin M. Fructose-1,6-bisphosphatase is a nonenzymatic safety valve that curtails AKT activation to prevent insulin hyperresponsiveness. Cell Metab 2023; 35:1009-1021.e9. [PMID: 37084733 PMCID: PMC10430883 DOI: 10.1016/j.cmet.2023.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/16/2023] [Accepted: 03/30/2023] [Indexed: 04/23/2023]
Abstract
Insulin inhibits gluconeogenesis and stimulates glucose conversion to glycogen and lipids. How these activities are coordinated to prevent hypoglycemia and hepatosteatosis is unclear. Fructose-1,6-bisphosphatase (FBP1) is rate controlling for gluconeogenesis. However, inborn human FBP1 deficiency does not cause hypoglycemia unless accompanied by fasting or starvation, which also trigger paradoxical hepatomegaly, hepatosteatosis, and hyperlipidemia. Hepatocyte FBP1-ablated mice exhibit identical fasting-conditional pathologies along with AKT hyperactivation, whose inhibition reversed hepatomegaly, hepatosteatosis, and hyperlipidemia but not hypoglycemia. Surprisingly, fasting-mediated AKT hyperactivation is insulin dependent. Independently of its catalytic activity, FBP1 prevents insulin hyperresponsiveness by forming a stable complex with AKT, PP2A-C, and aldolase B (ALDOB), which specifically accelerates AKT dephosphorylation. Enhanced by fasting and weakened by elevated insulin, FBP1:PP2A-C:ALDOB:AKT complex formation, which is disrupted by human FBP1 deficiency mutations or a C-terminal FBP1 truncation, prevents insulin-triggered liver pathologies and maintains lipid and glucose homeostasis. Conversely, an FBP1-derived complex disrupting peptide reverses diet-induced insulin resistance.
Collapse
Affiliation(s)
- Li Gu
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yahui Zhu
- School of Medicine, Chongqing University, Chongqing 400030, China
| | - Kosuke Watari
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Maiya Lee
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Junlai Liu
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sofia Perez
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Melinda Thai
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Joshua E Mayfield
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Bichen Zhang
- Division of Endocrinology & Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Karina Cunha E Rocha
- Division of Endocrinology & Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Fuming Li
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200438, China
| | - Laura C Kim
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexander C Jones
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Igor H Wierzbicki
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Xiao Liu
- Division of Endocrinology & Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Surgery, University of California, San Diego, La Jolla, CA 92093, USA
| | - Alexandra C Newton
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Tatiana Kisseleva
- Department of Surgery, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jun Hee Lee
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Wei Ying
- Division of Endocrinology & Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - David J Gonzalez
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Alan R Saltiel
- Division of Endocrinology & Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
8
|
Zhao N, Xu H. Pan-cancer analysis of aldolase B gene as a novel prognostic biomarker for human cancers. Medicine (Baltimore) 2023; 102:e33577. [PMID: 37083815 PMCID: PMC10118374 DOI: 10.1097/md.0000000000033577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/30/2023] [Indexed: 04/22/2023] Open
Abstract
Aldolase B (ALDOB) gene is essential for the process of glycolysis and differentially expressed in cancers. The aims of this study were to explore the potential role of ALDOB in pan-cancer, in order to deepen the research on the pathological mechanism of cancer. Hence, we used several online tools (TIMER2, GEPIA2, UALCAN, cBioPortal, and MXPRESS) and R language to identify the correlation between the ALDOB expression and survival analysis, genetic alteration, DNA methylation, and immune cell infiltration based on The Cancer Genome Atlas project. The results showed that ALDOB was lowly expressed in pan-cancer. Survival analysis revealed that low expression of ALDOB was markedly related with poor clinical prognosis, while the genetic alteration within ALDOB changed along with the difference of overall survival (OS) and disease-free survival (DFS) prognosis in several cancers. A possible relationship between DNA methylation and ALDOB expression for several tumors was found. Besides, ALDOB expression was confirmed to be associated with tumor immune cell infiltration, especially in breast invasive carcinoma (BRCA), esophageal carcinoma (ESCA), and testicular germ cell tumors (TGCT) cases. Further, the enrichment analysis demonstrated that metabolic pathway was closely related to ALDOB expression. Our results provide a comprehensive pan-cancer analysis and suggest ALDOB could act as a promising tumor predictive biomarker for human cancer.
Collapse
Affiliation(s)
- Nannan Zhao
- School of Food and Biology Engineering, Xuzhou University of Technology, Xuzhou, China
| | - Haixu Xu
- School of Food and Biology Engineering, Xuzhou University of Technology, Xuzhou, China
| |
Collapse
|
9
|
Iizuka K. Recent Progress on Fructose Metabolism-Chrebp, Fructolysis, and Polyol Pathway. Nutrients 2023; 15:nu15071778. [PMID: 37049617 PMCID: PMC10096667 DOI: 10.3390/nu15071778] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/26/2023] [Accepted: 03/29/2023] [Indexed: 04/14/2023] Open
Abstract
Excess fructose intake is associated with obesity, fatty liver, tooth decay, cancer, and cardiovascular diseases. Even after the ingestion of fructose, fructose concentration in the portal blood is never high; fructose is further metabolized in the liver, and the blood fructose concentration is 1/100th of the glucose concentration. It was previously thought that fructose was metabolized in the liver and not in the small intestine, but it has been reported that metabolism in the small intestine also plays an important role in fructose metabolism. Glut5 knockout mice exhibit poor fructose absorption. In addition, endogenous fructose production via the polyol pathway has also received attention; gene deletion of aldose reductase (Ar), ketohexokinase (Khk), and triokinase (Tkfc) has been found to prevent the development of fructose-induced liver lipidosis. Carbohydrate response element-binding protein (Chrebp) regulates the expression of Glut5, Khk, aldolase b, and Tkfc. We review fructose metabolism with a focus on the roles of the glucose-activating transcription factor Chrebp, fructolysis, and the polyol pathway.
Collapse
Affiliation(s)
- Katsumi Iizuka
- Department of Clinical Nutrition, Fujita Health University, Toyoake 470-1192, Japan
- Food and Nutrition Service Department, Fujita Health University Hospital, Toyoake 470-1192, Japan
| |
Collapse
|
10
|
Comparative Proteomic Analysis of Liver Tissues and Serum in db/db Mice. Int J Mol Sci 2022; 23:ijms23179687. [PMID: 36077090 PMCID: PMC9455973 DOI: 10.3390/ijms23179687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/20/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Background and Aims: Non-alcoholic fatty liver disease (NAFLD) affects one-quarter of individuals worldwide. Liver biopsy, as the current reliable method for NAFLD evaluation, causes low patient acceptance because of the nature of invasive sampling. Therefore, sensitive non-invasive serum biomarkers are urgently needed. Results: The serum gene ontology (GO) classification and Kyoto encyclopedia of genes and genomes (KEGG) analysis revealed the DEPs enriched in pathways including JAK-STAT and FoxO. GO analysis indicated that serum DEPs were mainly involved in the cellular process, metabolic process, response to stimulus, and biological regulation. Hepatic proteomic KEGG analysis revealed the DEPs were mainly enriched in the PPAR signaling pathway, retinol metabolism, glycine, serine, and threonine metabolism, fatty acid elongation, biosynthesis of unsaturated fatty acids, glutathione metabolism, and steroid hormone biosynthesis. GO analysis revealed that DEPs predominantly participated in cellular, biological regulation, multicellular organismal, localization, signaling, multi-organism, and immune system processes. Protein-protein interaction (PPI) implied diverse clusters of the DEPs. Besides, the paralleled changes of the common upregulated and downregulated DEPs existed in both the liver and serum were validated in the mRNA expression of NRP1, MUP3, SERPINA1E, ALPL, and ALDOB as observed in our proteomic screening. Methods: We conducted hepatic and serum proteomic analysis based on the leptin-receptor-deficient mouse (db/db), a well-established diabetic mouse model with overt obesity and NAFLD. The results show differentially expressed proteins (DEPs) in hepatic and serum proteomic analysis. A parallel reaction monitor (PRM) confirmed the authenticity of the selected DEPs. Conclusion: These results are supposed to offer sensitive non-invasive serum biomarkers for diabetes and NAFLD.
Collapse
|
11
|
Abstract
The consumption of fructose as sugar and high-fructose corn syrup has markedly increased during the past several decades. This trend coincides with the exponential rise of metabolic diseases, including obesity, nonalcoholic fatty liver disease, cardiovascular disease, and diabetes. While the biochemical pathways of fructose metabolism were elucidated in the early 1990s, organismal-level fructose metabolism and its whole-body pathophysiological impacts have been only recently investigated. In this review, we discuss the history of fructose consumption, biochemical and molecular pathways involved in fructose metabolism in different organs and gut microbiota, the role of fructose in the pathogenesis of metabolic diseases, and the remaining questions to treat such diseases.
Collapse
Affiliation(s)
- Sunhee Jung
- Department of Biological Chemistry, University of California, Irvine, California, USA
| | - Hosung Bae
- Department of Biological Chemistry, University of California, Irvine, California, USA
| | - Won-Suk Song
- Department of Biological Chemistry, University of California, Irvine, California, USA;,Institute of Bioengineering, Bio-MAX, Seoul National University, Seoul, South Korea
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California, Irvine, California, USA;,Chao Family Comprehensive Cancer Center, University of California, Irvine, California, USA,Center for Complex Biological Systems, University of California, Irvine, California, USA,Center for Epigenetics and Metabolism, University of California, Irvine, California, USA
| |
Collapse
|
12
|
Singh SK, Sarma MS. Hereditary fructose intolerance: A comprehensive review. World J Clin Pediatr 2022; 11:321-329. [PMID: 36052111 PMCID: PMC9331401 DOI: 10.5409/wjcp.v11.i4.321] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 05/08/2022] [Accepted: 06/20/2022] [Indexed: 02/06/2023] Open
Abstract
Hereditary fructose intolerance (HFI) is a rare autosomal recessive inherited disorder that occurs due to the mutation of enzyme aldolase B located on chromosome 9q22.3. A fructose load leads to the rapid accumulation of fructose 1-phosphate and manifests with its downstream effects. Most commonly children are affected with gastrointestinal symptoms, feeding issues, aversion to sweets and hypoglycemia. Liver manifestations include an asymptomatic increase of transaminases, steatohepatitis and rarely liver failure. Renal involvement usually occurs in the form of proximal renal tubular acidosis and may lead to chronic renal insufficiency. For confirmation, a genetic test is favored over the measurement of aldolase B activity in the liver biopsy specimen. The crux of HFI management lies in the absolute avoidance of foods containing fructose, sucrose, and sorbitol (FSS). There are many dilemmas regarding tolerance, dietary restriction and occurrence of steatohepatitis. Patients with HFI who adhere strictly to FSS free diet have an excellent prognosis with a normal lifespan. This review attempts to increase awareness and provide a comprehensive review of this rare but treatable disorder.
Collapse
Affiliation(s)
- Sumit Kumar Singh
- Department of Pediatrics, Sri Aurobindo Medical College and PGI, Indore 453555, Madhya Pradesh, India
| | - Moinak Sen Sarma
- Department of Pediatric Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| |
Collapse
|
13
|
Febbraio MA, Karin M. "Sweet death": Fructose as a metabolic toxin that targets the gut-liver axis. Cell Metab 2021; 33:2316-2328. [PMID: 34619076 PMCID: PMC8665123 DOI: 10.1016/j.cmet.2021.09.004] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/30/2021] [Accepted: 09/09/2021] [Indexed: 02/07/2023]
Abstract
Glucose and fructose are closely related simple sugars, but fructose has been associated more closely with metabolic disease. Until the 1960s, the major dietary source of fructose was fruit, but subsequently, high-fructose corn syrup (HFCS) became a dominant component of the Western diet. The exponential increase in HFCS consumption correlates with the increased incidence of obesity and type 2 diabetes mellitus, but the mechanistic link between these metabolic diseases and fructose remains tenuous. Although dietary fructose was thought to be metabolized exclusively in the liver, evidence has emerged that it is also metabolized in the small intestine and leads to intestinal epithelial barrier deterioration. Along with the clinical manifestations of hereditary fructose intolerance, these findings suggest that, along with the direct effect of fructose on liver metabolism, the gut-liver axis plays a key role in fructose metabolism and pathology. Here, we summarize recent studies on fructose biology and pathology and discuss new opportunities for prevention and treatment of diseases associated with high-fructose consumption.
Collapse
Affiliation(s)
- Mark A Febbraio
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.
| | - Michael Karin
- Department of Pharmacology, School of Medicine, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
14
|
Herman MA, Birnbaum MJ. Molecular aspects of fructose metabolism and metabolic disease. Cell Metab 2021; 33:2329-2354. [PMID: 34619074 PMCID: PMC8665132 DOI: 10.1016/j.cmet.2021.09.010] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/02/2021] [Accepted: 09/13/2021] [Indexed: 02/06/2023]
Abstract
Excessive sugar consumption is increasingly considered as a contributor to the emerging epidemics of obesity and the associated cardiometabolic disease. Sugar is added to the diet in the form of sucrose or high-fructose corn syrup, both of which comprise nearly equal amounts of glucose and fructose. The unique aspects of fructose metabolism and properties of fructose-derived metabolites allow for fructose to serve as a physiological signal of normal dietary sugar consumption. However, when fructose is consumed in excess, these unique properties may contribute to the pathogenesis of cardiometabolic disease. Here, we review the biochemistry, genetics, and physiology of fructose metabolism and consider mechanisms by which excessive fructose consumption may contribute to metabolic disease. Lastly, we consider new therapeutic options for the treatment of metabolic disease based upon this knowledge.
Collapse
Affiliation(s)
- Mark A Herman
- Division of Endocrinology, Metabolism, and Nutrition, Duke University, Durham, NC, USA; Duke Molecular Physiology Institute, Duke University, Durham, NC, USA; Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA.
| | | |
Collapse
|
15
|
Liu G, Wang N, Zhang C, Li M, He X, Yin C, Tu Q, Shen X, Zhang L, Lv J, Wang Y, Jiang H, Chen S, Li N, Tao Y, Yin H. Fructose-1,6-Bisphosphate Aldolase B Depletion Promotes Hepatocellular Carcinogenesis Through Activating Insulin Receptor Signaling and Lipogenesis. Hepatology 2021; 74:3037-3055. [PMID: 34292642 DOI: 10.1002/hep.32064] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 06/11/2021] [Accepted: 06/28/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Insulin receptor (IR) transduces cell surface signal through phosphoinositide 3-kinase (PI3K)-AKT pathways or translocates to the nucleus and binds to the promoters to regulate genes associated with insulin actions, including de novo lipogenesis (DNL). Chronic activation of IR signaling drives malignant transformation, but the underlying mechanisms remain poorly defined. Down-regulation of fructose-1,6-bisphosphate aldolase (ALDO) B in hepatocellular carcinoma (HCC) is correlated with poor prognosis. We aim to study whether and how ALDOB is involved in IR signaling in HCC. APPROACH AND RESULTS Global or liver-specific ALDOB knockout (L-ALDOB-/- ) mice were used in N-diethylnitrosamine (DEN)-induced HCC models, whereas restoration of ALDOB expression was achieved in L-ALDOB-/- mice by adeno-associated virus (AAV). 13 C6 -glucose was employed in metabolic flux analysis to track the de novo fatty acid synthesis from glucose, and nontargeted lipidomics and targeted fatty acid analysis using mass spectrometry were performed. We found that ALDOB physically interacts with IR and attenuates IR signaling through down-regulating PI3K-AKT pathways and suppressing IR nuclear translocation. ALDOB depletion or disruption of IR/ALDOB interaction in ALDOB mutants promotes DNL and tumorigenesis, which is significantly attenuated with ALDOB restoration in L-ALDOB-/- mice. Notably, attenuated IR/ALDOB interaction in ALDOB-R46A mutant exhibits more significant tumorigenesis than releasing ALDOB/AKT interaction in ALDOB-R43A, whereas knockdown IR sufficiently diminishes tumor-promoting effects in both mutants. Furthermore, inhibiting phosphorylated AKT or fatty acid synthase significantly attenuates HCC in L-ALDOB-/- mice. Consistently, ALDOB down-regulation is correlated with up-regulation of IR signaling and DNL in human HCC tumor tissues. CONCLUSIONS Our study reports a mechanism by which loss of ALDOB activates IR signaling primarily through releasing IR/ALDOB interaction to promote DNL and HCC, highlighting a potential therapeutic strategy in HCC.
Collapse
Affiliation(s)
- Guijun Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health (SINH), University of the Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS), Shanghai, China
| | - Ningning Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health (SINH), University of the Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS), Shanghai, China
| | - Cunzhen Zhang
- Department of Hepatic Surgery I (Ward l), Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Min Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health (SINH), University of the Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS), Shanghai, China
| | - Xuxiao He
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health (SINH), University of the Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS), Shanghai, China
| | - Chunzhao Yin
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Qiaochu Tu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xia Shen
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Lili Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health (SINH), University of the Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS), Shanghai, China
| | - Jingwen Lv
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health (SINH), University of the Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS), Shanghai, China
| | - Yongqiang Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health (SINH), University of the Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS), Shanghai, China
| | - Huimin Jiang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health (SINH), University of the Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS), Shanghai, China
| | - Shiting Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health (SINH), University of the Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS), Shanghai, China
| | - Nan Li
- Department of Hepatic Surgery I (Ward l), Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Yongzhen Tao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health (SINH), University of the Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS), Shanghai, China
| | - Huiyong Yin
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health (SINH), University of the Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS), Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China
| |
Collapse
|
16
|
Fructose and Mannose in Inborn Errors of Metabolism and Cancer. Metabolites 2021; 11:metabo11080479. [PMID: 34436420 PMCID: PMC8397987 DOI: 10.3390/metabo11080479] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 07/21/2021] [Accepted: 07/21/2021] [Indexed: 12/19/2022] Open
Abstract
History suggests that tasteful properties of sugar have been domesticated as far back as 8000 BCE. With origins in New Guinea, the cultivation of sugar quickly spread over centuries of conquest and trade. The product, which quickly integrated into common foods and onto kitchen tables, is sucrose, which is made up of glucose and fructose dimers. While sugar is commonly associated with flavor, there is a myriad of biochemical properties that explain how sugars as biological molecules function in physiological contexts. Substantial research and reviews have been done on the role of glucose in disease. This review aims to describe the role of its isomers, fructose and mannose, in the context of inborn errors of metabolism and other metabolic diseases, such as cancer. While structurally similar, fructose and mannose give rise to very differing biochemical properties and understanding these differences will guide the development of more effective therapies for metabolic disease. We will discuss pathophysiology linked to perturbations in fructose and mannose metabolism, diagnostic tools, and treatment options of the diseases.
Collapse
|
17
|
Kading J, Finck BN, DeBosch BJ. Targeting hepatocyte carbohydrate transport to mimic fasting and calorie restriction. FEBS J 2021; 288:3784-3798. [PMID: 32654397 PMCID: PMC8662989 DOI: 10.1111/febs.15482] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/02/2020] [Accepted: 07/09/2020] [Indexed: 12/14/2022]
Abstract
The pervasion of three daily meals and snacks is a relatively new introduction to our shared experience and is coincident with an epidemic rise in obesity and cardiometabolic disorders of overnutrition. The past two decades have yielded convincing evidence regarding the adaptive, protective effects of calorie restriction (CR) and intermittent fasting (IF) against cardiometabolic, neurodegenerative, proteostatic, and inflammatory diseases. Yet, durable adherence to intensive lifestyle changes is rarely attainable. New evidence now demonstrates that restricting carbohydrate entry into the hepatocyte by itself mimics several key signaling responses and physiological outcomes of IF and CR. This discovery raises the intriguing proposition that targeting hepatocyte carbohydrate transport to mimic fasting and caloric restriction can abate cardiometabolic and perhaps other fasting-treatable diseases. Here, we review the metabolic and signaling fates of a hepatocyte carbohydrate, identify evidence to target the key mediators within these pathways, and provide rationale and data to highlight carbohydrate transport as a broad, proximal intervention to block the deleterious sequelae of hepatic glucose and fructose metabolism.
Collapse
Affiliation(s)
- Jacqueline Kading
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian N. Finck
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian J DeBosch
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
18
|
Federico A, Rosato V, Masarone M, Torre P, Dallio M, Romeo M, Persico M. The Role of Fructose in Non-Alcoholic Steatohepatitis: Old Relationship and New Insights. Nutrients 2021; 13:1314. [PMID: 33923525 PMCID: PMC8074203 DOI: 10.3390/nu13041314] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/12/2021] [Accepted: 04/14/2021] [Indexed: 12/22/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) represents the result of hepatic fat overload not due to alcohol consumption and potentially evolving to advanced fibrosis, cirrhosis, and hepatocellular carcinoma. Fructose is a naturally occurring simple sugar widely used in food industry linked to glucose to form sucrose, largely contained in hypercaloric food and beverages. An increasing amount of evidence in scientific literature highlighted a detrimental effect of dietary fructose consumption on metabolic disorders such as insulin resistance, obesity, hepatic steatosis, and NAFLD-related fibrosis as well. An excessive fructose consumption has been associated with NAFLD development and progression to more clinically severe phenotypes by exerting various toxic effects, including increased fatty acid production, oxidative stress, and worsening insulin resistance. Furthermore, some studies in this context demonstrated even a crucial role in liver cancer progression. Despite this compelling evidence, the molecular mechanisms by which fructose elicits those effects on liver metabolism remain unclear. Emerging data suggest that dietary fructose may directly alter the expression of genes involved in lipid metabolism, including those that increase hepatic fat accumulation or reduce hepatic fat removal. This review aimed to summarize the current understanding of fructose metabolism on NAFLD pathogenesis and progression.
Collapse
Affiliation(s)
- Alessandro Federico
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.D.); (M.R.)
| | - Valerio Rosato
- Internal Medicine and Hepatology Division, Department of Medicine, Surgery and Odontostomatology, “Scuola Medica Salernitana”, University of Salerno, 84084 Salerno, Italy; (V.R.); (M.M.); (P.T.); (M.P.)
- Liver Unit, Ospedale Evangelico Betania, 80147 Naples, Italy
| | - Mario Masarone
- Internal Medicine and Hepatology Division, Department of Medicine, Surgery and Odontostomatology, “Scuola Medica Salernitana”, University of Salerno, 84084 Salerno, Italy; (V.R.); (M.M.); (P.T.); (M.P.)
| | - Pietro Torre
- Internal Medicine and Hepatology Division, Department of Medicine, Surgery and Odontostomatology, “Scuola Medica Salernitana”, University of Salerno, 84084 Salerno, Italy; (V.R.); (M.M.); (P.T.); (M.P.)
| | - Marcello Dallio
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.D.); (M.R.)
| | - Mario Romeo
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.D.); (M.R.)
| | - Marcello Persico
- Internal Medicine and Hepatology Division, Department of Medicine, Surgery and Odontostomatology, “Scuola Medica Salernitana”, University of Salerno, 84084 Salerno, Italy; (V.R.); (M.M.); (P.T.); (M.P.)
| |
Collapse
|
19
|
Singh MK, Jayarajan R, Varshney S, Upadrasta S, Singh A, Yadav R, Scaria V, Sengupta S, Shanmugam D, Shalimar, Sivasubbu S, Gandotra S, Sachidanandan C. Chronic systemic exposure to IL6 leads to deregulation of glycolysis and fat accumulation in the zebrafish liver. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158905. [PMID: 33582286 DOI: 10.1016/j.bbalip.2021.158905] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 01/21/2021] [Accepted: 02/09/2021] [Indexed: 02/08/2023]
Abstract
Inflammation is a constant in Non-Alcoholic Fatty Liver Disease (NAFLD), although their relationship is unclear. In a transgenic zebrafish system with chronic systemic overexpression of human IL6 (IL6-OE) we show that inflammation can cause intra-hepatic accumulation of triglycerides. Transcriptomics and proteomics analysis of the IL6-OE liver revealed a deregulation of glycolysis/gluconeogenesis pathway, especially a striking down regulation of the glycolytic enzyme aldolase b. Metabolomics analysis by mass spectrometry showed accumulation of hexose monophosphates and their derivatives, which can act as precursors for triglyceride synthesis. Our results suggest that IL6-driven repression of glycolysis/gluconeogenesis, specifically aldolase b, may be a novel mechanism for fatty liver. This mechanism may be relevant for NAFLD in lean individuals, an emerging class of NAFLD prevalent more in Asian Indian populations.
Collapse
Affiliation(s)
- Manoj K Singh
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rijith Jayarajan
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India
| | - Swati Varshney
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sindhuri Upadrasta
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; CSIR-National Chemical Laboratory, Pune, India
| | - Archana Singh
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rajni Yadav
- All India Institute of Medical Sciences, New Delhi, India
| | - Vinod Scaria
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shantanu Sengupta
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Dhanasekaran Shanmugam
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; CSIR-National Chemical Laboratory, Pune, India
| | - Shalimar
- All India Institute of Medical Sciences, New Delhi, India
| | - Sridhar Sivasubbu
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sheetal Gandotra
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Chetana Sachidanandan
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
20
|
He X, Li M, Yu H, Liu G, Wang N, Yin C, Tu Q, Narla G, Tao Y, Cheng S, Yin H. Loss of hepatic aldolase B activates Akt and promotes hepatocellular carcinogenesis by destabilizing the Aldob/Akt/PP2A protein complex. PLoS Biol 2020; 18:e3000803. [PMID: 33275593 PMCID: PMC7744066 DOI: 10.1371/journal.pbio.3000803] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 12/16/2020] [Accepted: 11/13/2020] [Indexed: 12/21/2022] Open
Abstract
Loss of hepatic fructose-1, 6-bisphosphate aldolase B (Aldob) leads to a paradoxical up-regulation of glucose metabolism to favor hepatocellular carcinogenesis (HCC), but the upstream signaling events remain poorly defined. Akt is highly activated in HCC, and targeting Akt is being explored as a potential therapy for HCC. Herein, we demonstrate that Aldob suppresses Akt activity and tumor growth through a protein complex containing Aldob, Akt, and protein phosphatase 2A (PP2A), leading to inhibition of cell viability, cell cycle progression, glucose uptake, and metabolism. Interestingly, Aldob directly interacts with phosphorylated Akt (p-Akt) and promotes the recruitment of PP2A to dephosphorylate p-Akt, and this scaffolding effect of Aldob is independent of its enzymatic activity. Loss of Aldob or disruption of Aldob/Akt interaction in Aldob R304A mutant restores Akt activity and tumor-promoting effects. Consistently, Aldob and p-Akt expression are inversely correlated in human HCC tissues, and Aldob down-regulation coupled with p-Akt up-regulation predicts a poor prognosis for HCC. We have further discovered that Akt inhibition or a specific small-molecule activator of PP2A (SMAP) efficiently attenuates HCC tumorigenesis in xenograft mouse models. Our work reveals a novel nonenzymatic role of Aldob in negative regulation of Akt activation, suggesting that directly inhibiting Akt activity or through reactivating PP2A may be a potential therapeutic approach for HCC treatment.
Collapse
Affiliation(s)
- Xuxiao He
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences (CAS), Shanghai, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Min Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences (CAS), Shanghai, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Hongming Yu
- The Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Guijun Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences (CAS), Shanghai, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Ningning Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences (CAS), Shanghai, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Chunzhao Yin
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Qiaochu Tu
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Goutham Narla
- Division of Genetic Medicine, Department of International Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Yongzhen Tao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences (CAS), Shanghai, China
| | - Shuqun Cheng
- The Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Huiyong Yin
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences (CAS), Shanghai, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China
| |
Collapse
|
21
|
Skenderian S, Park G, Jang C. Organismal Fructose Metabolism in Health and Non-Alcoholic Fatty Liver Disease. BIOLOGY 2020; 9:E405. [PMID: 33218081 PMCID: PMC7698815 DOI: 10.3390/biology9110405] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 02/07/2023]
Abstract
NAFLD has alarmingly increased, yet FDA-approved drugs are still lacking. An excessive intake of fructose, especially in liquid form, is a dietary risk factor of NAFLD. While fructose metabolism has been studied for decades, it is still controversial how fructose intake can cause NAFLD. It has long been believed that fructose metabolism solely happens in the liver and accordingly, numerous studies have investigated liver fructose metabolism using primary hepatocytes or liver cell lines in culture. While cultured cells are useful for studying detailed signaling pathways and metabolism in a cell-autonomous manner, it is equally important to understand fructose metabolism at the whole-body level in live organisms. In this regard, recent in vivo studies using genetically modified mice and stable isotope tracing have tremendously expanded our understanding of the complex interaction between fructose-catabolizing organs and gut microbiota. Here, we discuss how the aberrant distribution of fructose metabolism between organs and gut microbiota can contribute to NAFLD. We also address potential therapeutic interventions of fructose-elicited NAFLD.
Collapse
Affiliation(s)
- Shea Skenderian
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA;
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA;
| | - Grace Park
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA;
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA;
| |
Collapse
|
22
|
Zhang C, Li L, Zhang Y, Zeng C. Recent advances in fructose intake and risk of hyperuricemia. Biomed Pharmacother 2020; 131:110795. [PMID: 33152951 DOI: 10.1016/j.biopha.2020.110795] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/17/2020] [Accepted: 09/19/2020] [Indexed: 12/17/2022] Open
Abstract
With the widespread popularity of hyperuricemia, it has become a severe threat to human public health. Accumulating evidence suggests that dietary fructose has a close relationship with hyperuricemia, but the role of fructose intake in hyperuricemia remains unclear. Hyperuricemia is characterized by excessive production and deposition of urate crystals. Metabolism of fructose leads to the increased serum concentration of urate. In this review, we depict an update of fructose consumption worldwide and the epidemiology of hyperuricemia and summarize the progress in studying the relationship between fructose intake and the risk of hyperuricemia. This review highlights the metabolic process of fructose in the liver, small intestine, and kidney. Furthermore, we discuss molecular insights on fructose metabolism to reveal the underlying mechanism of fructose metabolism. Additionally, we elaborate on the effect of fructose metabolism on hyperuricemia to deeply understand the pathogenesis of hyperuricemia caused by fructose intake. Fructose consumption has a close correlation with an enhanced risk of developing hyperuricemia. More prospective studies are inevitable to understand the role of fructose intake in the development of hyperuricemia.
Collapse
Affiliation(s)
- Congwang Zhang
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, Guangdong, 518110, PR China
| | - Lijun Li
- Department of Quality Control, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, Guangdong, 518110, PR China
| | - Yipeng Zhang
- Clinical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, Guangdong, 518110, PR China
| | - Changchun Zeng
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, Guangdong, 518110, PR China.
| |
Collapse
|
23
|
Aldolase B suppresses hepatocellular carcinogenesis by inhibiting G6PD and pentose phosphate pathways. ACTA ACUST UNITED AC 2020; 1:735-747. [DOI: 10.1038/s43018-020-0086-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 05/27/2020] [Indexed: 12/11/2022]
|
24
|
Buziau AM, Schalkwijk CG, Stehouwer CDA, Tolan DR, Brouwers MCGJ. Recent advances in the pathogenesis of hereditary fructose intolerance: implications for its treatment and the understanding of fructose-induced non-alcoholic fatty liver disease. Cell Mol Life Sci 2020; 77:1709-1719. [PMID: 31713637 PMCID: PMC11105038 DOI: 10.1007/s00018-019-03348-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 10/02/2019] [Accepted: 10/16/2019] [Indexed: 12/31/2022]
Abstract
Hereditary fructose intolerance (HFI) is a rare inborn disease characterized by a deficiency in aldolase B, which catalyzes the cleavage of fructose 1,6-bisphosphate and fructose 1-phosphate (Fru 1P) to triose molecules. In patients with HFI, ingestion of fructose results in accumulation of Fru 1P and depletion of ATP, which are believed to cause symptoms, such as nausea, vomiting, hypoglycemia, and liver and kidney failure. These sequelae can be prevented by a fructose-restricted diet. Recent studies in aldolase B-deficient mice and HFI patients have provided more insight into the pathogenesis of HFI, in particular the liver phenotype. Both aldolase B-deficient mice (fed a very low fructose diet) and HFI patients (treated with a fructose-restricted diet) displayed greater intrahepatic fat content when compared to controls. The liver phenotype in aldolase B-deficient mice was prevented by reduction in intrahepatic Fru 1P concentrations by crossing these mice with mice deficient for ketohexokinase, the enzyme that catalyzes the synthesis of Fru 1P. These new findings not only provide a potential novel treatment for HFI, but lend insight into the pathogenesis of fructose-induced non-alcoholic fatty liver disease (NAFLD), which has raised to epidemic proportions in Western society. This narrative review summarizes the most recent advances in the pathogenesis of HFI and discusses the implications for the understanding and treatment of fructose-induced NAFLD.
Collapse
Affiliation(s)
- Amée M Buziau
- Division of Endocrinology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Laboratory for Metabolism and Vascular Medicine, Division of General Internal Medicine, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| | - Casper G Schalkwijk
- Laboratory for Metabolism and Vascular Medicine, Division of General Internal Medicine, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| | - Coen D A Stehouwer
- Laboratory for Metabolism and Vascular Medicine, Division of General Internal Medicine, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
- Division of General Internal Medicine, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Dean R Tolan
- Department of Biology, Boston University, Boston, MA, USA.
| | - Martijn C G J Brouwers
- Division of Endocrinology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands.
- Laboratory for Metabolism and Vascular Medicine, Division of General Internal Medicine, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands.
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands.
| |
Collapse
|
25
|
Abstract
Irritable bowel syndrome (IBS) is a chronic disorder characterised by recurrent abdominal pain or discomfort and transit disturbances with heterogeneous pathophysiological mechanisms. The link between food and gastrointestinal (GI) symptoms is often reported by patients with IBS and the role of fructose has recently been highlighted. Fructose malabsorption can easily be assessed by hydrogen and/or methane breath test in response to 25 g fructose; and its prevalence is about 22 % in patients with IBS. The mechanism of fructose-related symptoms is incompletely understood. Osmotic load, fermentation and visceral hypersensitivity are likely to participate in GI symptoms in the IBS population and may be triggered or worsened by fructose. A low-fructose diet could be integrated in the overall treatment strategy, but its role and implication in the improvement of IBS symptoms should be evaluated. In the present review, we discuss fructose malabsorption in adult patients with IBS and the interest of a low-fructose diet in order to underline the important role of fructose in IBS.
Collapse
|
26
|
Non-alcoholic fatty liver in hereditary fructose intolerance. Clin Nutr 2020; 39:455-459. [DOI: 10.1016/j.clnu.2019.02.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 02/06/2019] [Accepted: 02/10/2019] [Indexed: 02/05/2023]
|
27
|
|
28
|
Bu P, Chen KY, Xiang K, Johnson C, Crown SB, Rakhilin N, Ai Y, Wang L, Xi R, Astapova I, Han Y, Li J, Barth BB, Lu M, Gao Z, Mines R, Zhang L, Herman M, Hsu D, Zhang GF, Shen X. Aldolase B-Mediated Fructose Metabolism Drives Metabolic Reprogramming of Colon Cancer Liver Metastasis. Cell Metab 2018; 27:1249-1262.e4. [PMID: 29706565 PMCID: PMC5990465 DOI: 10.1016/j.cmet.2018.04.003] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 01/18/2018] [Accepted: 04/02/2018] [Indexed: 02/07/2023]
Abstract
Cancer metastasis accounts for the majority of cancer-related deaths and remains a clinical challenge. Metastatic cancer cells generally resemble cells of the primary cancer, but they may be influenced by the milieu of the organs they colonize. Here, we show that colorectal cancer cells undergo metabolic reprogramming after they metastasize and colonize the liver, a key metabolic organ. In particular, via GATA6, metastatic cells in the liver upregulate the enzyme aldolase B (ALDOB), which enhances fructose metabolism and provides fuel for major pathways of central carbon metabolism during tumor cell proliferation. Targeting ALDOB or reducing dietary fructose significantly reduces liver metastatic growth but has little effect on the primary tumor. Our findings suggest that metastatic cells can take advantage of reprogrammed metabolism in their new microenvironment, especially in a metabolically active organ such as the liver. Manipulation of involved pathways may affect the course of metastatic growth.
Collapse
Affiliation(s)
- Pengcheng Bu
- Key Laboratory of RNA Biology, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA.
| | - Kai-Yuan Chen
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Kun Xiang
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Christelle Johnson
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; School of Electrical and Computer Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Scott B Crown
- Molecular Physiology Institute, Duke University, Durham, NC 27701, USA
| | - Nikolai Rakhilin
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; School of Electrical and Computer Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Yiwei Ai
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Lihua Wang
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Rui Xi
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Inna Astapova
- Molecular Physiology Institute, Duke University, Durham, NC 27701, USA; Department of Medicine, Duke University, Durham, NC 27701, USA
| | - Yan Han
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Jiahe Li
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Bradley B Barth
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Min Lu
- Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, NC 27710, USA
| | - Ziyang Gao
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Robert Mines
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Liwen Zhang
- Key Laboratory of RNA Biology, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mark Herman
- Molecular Physiology Institute, Duke University, Durham, NC 27701, USA; Department of Medicine, Duke University, Durham, NC 27701, USA
| | - David Hsu
- Department of Medicine, Duke University, Durham, NC 27701, USA; Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, NC 27710, USA
| | - Guo-Fang Zhang
- Molecular Physiology Institute, Duke University, Durham, NC 27701, USA; Department of Medicine, Duke University, Durham, NC 27701, USA
| | - Xiling Shen
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
29
|
Lanaspa MA, Andres-Hernando A, Orlicky DJ, Cicerchi C, Jang C, Li N, Milagres T, Kuwabara M, Wempe MF, Rabinowitz JD, Johnson RJ, Tolan DR. Ketohexokinase C blockade ameliorates fructose-induced metabolic dysfunction in fructose-sensitive mice. J Clin Invest 2018. [PMID: 29533924 DOI: 10.1172/jci94427] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Increasing evidence suggests a role for excessive intake of fructose in the Western diet as a contributor to the current epidemics of metabolic syndrome and obesity. Hereditary fructose intolerance (HFI) is a difficult and potentially lethal orphan disease associated with impaired fructose metabolism. In HFI, the deficiency of aldolase B results in the accumulation of intracellular phosphorylated fructose, leading to phosphate sequestration and depletion, increased adenosine triphosphate (ATP) turnover, and a plethora of conditions that lead to clinical manifestations such as fatty liver, hyperuricemia, Fanconi syndrome, and severe hypoglycemia. Unfortunately, there is currently no treatment for HFI, and avoiding sugar and fructose has become challenging in our society. In this report, through use of genetically modified mice and pharmacological inhibitors, we demonstrate that the absence or inhibition of ketohexokinase (Khk), an enzyme upstream of aldolase B, is sufficient to prevent hypoglycemia and liver and intestinal injury associated with HFI. Herein we provide evidence for the first time to our knowledge of a potential therapeutic approach for HFI. Mechanistically, our studies suggest that it is the inhibition of the Khk C isoform, not the A isoform, that protects animals from HFI.
Collapse
Affiliation(s)
- Miguel A Lanaspa
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, Colorado, USA
| | - Ana Andres-Hernando
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, Colorado, USA
| | - David J Orlicky
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, Colorado, USA
| | - Christina Cicerchi
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, Colorado, USA
| | - Cholsoon Jang
- Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, USA
| | - Nanxing Li
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, Colorado, USA
| | - Tamara Milagres
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, Colorado, USA
| | - Masanari Kuwabara
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, Colorado, USA
| | - Michael F Wempe
- Department of Pharmacology, University of Colorado, Aurora, Colorado, USA
| | - Joshua D Rabinowitz
- Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, USA
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, Colorado, USA
| | - Dean R Tolan
- Department of Biology, Boston University, Boston, Massachusetts, USA
| |
Collapse
|
30
|
Hannou SA, Haslam DE, McKeown NM, Herman MA. Fructose metabolism and metabolic disease. J Clin Invest 2018; 128:545-555. [PMID: 29388924 DOI: 10.1172/jci96702] [Citation(s) in RCA: 307] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Increased sugar consumption is increasingly considered to be a contributor to the worldwide epidemics of obesity and diabetes and their associated cardiometabolic risks. As a result of its unique metabolic properties, the fructose component of sugar may be particularly harmful. Diets high in fructose can rapidly produce all of the key features of the metabolic syndrome. Here we review the biology of fructose metabolism as well as potential mechanisms by which excessive fructose consumption may contribute to cardiometabolic disease.
Collapse
Affiliation(s)
- Sarah A Hannou
- Division of Endocrinology and Metabolism and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Danielle E Haslam
- Nutritional Epidemiology Program, Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts, USA
| | - Nicola M McKeown
- Nutritional Epidemiology Program, Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts, USA
| | - Mark A Herman
- Division of Endocrinology and Metabolism and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
31
|
Chen W, Lau SW, Fan Y, Wu RSS, Ge W. Juvenile exposure to bisphenol A promotes ovarian differentiation but suppresses its growth - Potential involvement of pituitary follicle-stimulating hormone. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2017; 193:111-121. [PMID: 29055862 DOI: 10.1016/j.aquatox.2017.10.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/08/2017] [Accepted: 10/12/2017] [Indexed: 06/07/2023]
Abstract
Bisphenol A (BPA), a plastic monomer and plasticizer, is commonly used in plastics industry, and it has been well documented to be an estrogenic endocrine disrupter. In the present study, we investigated the effect of early (juvenile) exposure to BPA on the hypothalamic-pituitary-gonad (HPG) axis in the zebrafish. Estradiol (E2) and testosterone (T) were also included as positive and negative controls respectively. Juvenile zebrafish were exposed to BPA (1 and 10μM), E2 (10nM) and T (10nM) from 20 to 40 dpf (days post-fertilization), the period of sex/gonadal differentiation, followed by histological and expression analyses at 40 dpf. The ovary and hepatic proteomes were also analyzed by mass spectrometry. Our results showed that 20day exposure to BPA and E2 increased the ratio of females; however, they both significantly suppressed ovarian growth. Meanwhile, BPA and E2 significantly suppressed fshb but stimulated lhb expression in the pituitary. These effects did not seem to involve the hypothalamus because neither BPA nor E2 altered the expression of kiss1, kiss2, gnrh2 and gnrh3 in the hypothalamus. At the ovary level, BPA and E2 both decreased lhcgr expression. Interestingly, E2 and BPA displayed different effects in the liver. E2 induced a significant hepatic hypertrophy; however, BPA had no such effect. Analysis of hepatic proteomes revealed distinct protein profiles in the E2 group as compared with the others, especially fructose-bisphospahte aldolase B. These results indicated that BPA has estrogenic effects on female reproduction, but it does not mimic all E2 actions. Our data in the zebrafish suggest that sex differentiation involves estrogens and it is a sensitive window for evaluating estrogenic activities of compounds and their impacts on wildlife reproduction.
Collapse
Affiliation(s)
- Weiting Chen
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| | - Shuk-Wa Lau
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Yuqin Fan
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| | - Rudolf S S Wu
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Wei Ge
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| |
Collapse
|
32
|
Inborn Errors of Fructose Metabolism. What Can We Learn from Them? Nutrients 2017; 9:nu9040356. [PMID: 28368361 PMCID: PMC5409695 DOI: 10.3390/nu9040356] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 03/27/2017] [Accepted: 03/28/2017] [Indexed: 12/18/2022] Open
Abstract
Fructose is one of the main sweetening agents in the human diet and its ingestion is increasing globally. Dietary sugar has particular effects on those whose capacity to metabolize fructose is limited. If intolerance to carbohydrates is a frequent finding in children, inborn errors of carbohydrate metabolism are rare conditions. Three inborn errors are known in the pathway of fructose metabolism; (1) essential or benign fructosuria due to fructokinase deficiency; (2) hereditary fructose intolerance; and (3) fructose-1,6-bisphosphatase deficiency. In this review the focus is set on the description of the clinical symptoms and biochemical anomalies in the three inborn errors of metabolism. The potential toxic effects of fructose in healthy humans also are discussed. Studies conducted in patients with inborn errors of fructose metabolism helped to understand fructose metabolism and its potential toxicity in healthy human. Influence of fructose on the glycolytic pathway and on purine catabolism is the cause of hypoglycemia, lactic acidosis and hyperuricemia. The discovery that fructose-mediated generation of uric acid may have a causal role in diabetes and obesity provided new understandings into pathogenesis for these frequent diseases.
Collapse
|
33
|
Edwards CH, Rossi M, Corpe CP, Butterworth PJ, Ellis PR. The role of sugars and sweeteners in food, diet and health: Alternatives for the future. Trends Food Sci Technol 2016. [DOI: 10.1016/j.tifs.2016.07.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
34
|
Herman MA, Samuel VT. The Sweet Path to Metabolic Demise: Fructose and Lipid Synthesis. Trends Endocrinol Metab 2016; 27:719-730. [PMID: 27387598 PMCID: PMC5035631 DOI: 10.1016/j.tem.2016.06.005] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 06/06/2016] [Accepted: 06/09/2016] [Indexed: 01/04/2023]
Abstract
Epidemiological studies link fructose consumption with metabolic disease, an association attributable in part to fructose-mediated lipogenesis. The mechanisms governing fructose-induced lipogenesis and disease remain debated. Acutely, fructose increases de novo lipogenesis through the efficient and uninhibited action of ketohexokinase and aldolase B which yields substrates for fatty-acid synthesis. Chronic fructose consumption further enhances the capacity for hepatic fructose metabolism by activating several key transcription factors (i.e., SREBP1c and ChREBP) which augment the expression of lipogenic enzymes, increasing lipogenesis and further compounding hypertriglyceridemia and hepatic steatosis. Hepatic insulin resistance develops from diacylglycerol-PKCɛ-mediated impairment of insulin signaling and possibly additional mechanisms. Initiatives that decrease fructose consumption and therapies that block fructose-mediated lipogenesis will be necessary to avert future metabolic pandemics.
Collapse
Affiliation(s)
- Mark A Herman
- Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA.
| | - Varman T Samuel
- Yale University School of Medicine, 950 Campbell Avenue, West Haven, CT 06516, USA; Veterans Affairs Connecticut Healthcare System, 950 Campbell Avenue, West Haven, CT 06516, USA.
| |
Collapse
|