1
|
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
|
2
|
Li C, Li M, Sheng W, Zhou W, Zhang Z, Ji G, Zhang L. High dietary Fructose Drives Metabolic Dysfunction-Associated Steatotic Liver Disease via Activating ubiquitin-specific peptidase 2/11β-hydroxysteroid dehydrogenase type 1 Pathway in Mice. Int J Biol Sci 2024; 20:3480-3496. [PMID: 38993560 PMCID: PMC11234208 DOI: 10.7150/ijbs.97309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 06/08/2024] [Indexed: 07/13/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common cause of chronic liver-related morbidity and mortality. Though high fructose intake is acknowledged as a metabolic hazard, its role in the etiology of MASLD requires further clarification. Here, we demonstrated that high dietary fructose drives MASLD development and promotes MASLD progression in mice, and identified Usp2 as a fructose-responsive gene in the liver. Elevated USP2 levels were detected in the hepatocytes of MASLD mice; a similar increase was observed following fructose exposure in primary hepatocytes and mouse AML12 cells. Notably, hepatocytes overexpressing USP2 presented with exaggerated lipid accumulation and metabolic inflammation when exposed to fructose. Conversely, USP2 knockdown mitigated these fructose-induced changes. Furthermore, USP2 was found to activate the C/EBPα/11β-HSD1 signaling, which further impacted the equilibrium of cortisol and cortisone in the circulation of mice. Collectively, our findings revealed the role of dietary fructose in MASLD pathogenesis and identified the USP2-mediated C/EBPα/ 11β-HSD1 signaling as a potential target for the management of MASLD.
Collapse
Affiliation(s)
- Chunlin Li
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Meng Li
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Wei Sheng
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Wenjun Zhou
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- State Key Laboratory of Integration and Innovation of Classical Formula and Modern Chinese Medicine, China
| | - Ziqi Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- State Key Laboratory of Integration and Innovation of Classical Formula and Modern Chinese Medicine, China
| | - Li Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- State Key Laboratory of Integration and Innovation of Classical Formula and Modern Chinese Medicine, China
| |
Collapse
|
3
|
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
|
4
|
Li Y, Qi P, Song SY, Wang Y, Wang H, Cao P, Liu Y, Wang Y. Elucidating cuproptosis in metabolic dysfunction-associated steatotic liver disease. Biomed Pharmacother 2024; 174:116585. [PMID: 38615611 DOI: 10.1016/j.biopha.2024.116585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/07/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024] Open
Abstract
Emerging research into metabolic dysfunction-associated steatotic liver disease (MASLD) up until January 2024 has highlighted the critical role of cuproptosis, a unique cell death mechanism triggered by copper overload, in the disease's development. This connection offers new insights into MASLD's complex pathogenesis, pointing to copper accumulation as a key factor that disrupts lipid metabolism and insulin sensitivity. The identification of cuproptosis as a significant contributor to MASLD underscores the potential for targeting copper-mediated pathways for novel therapeutic approaches. This promising avenue suggests that managing copper levels could mitigate MASLD progression, offering a fresh perspective on treatment strategies. Further investigations into how cuproptosis influences MASLD are essential for unraveling the detailed mechanisms at play and for identifying effective interventions. The focus on copper's role in liver health opens up the possibility of developing targeted therapies that address the underlying causes of MASLD, moving beyond symptomatic treatment to tackle the root of the problem. The exploration of cuproptosis in the context of MASLD exemplifies the importance of understanding metal homeostasis in metabolic diseases and represents a significant step forward in the quest for more effective treatments. This research direction lights path for innovative MASLD management and reversal.
Collapse
Affiliation(s)
- Yamei Li
- Department of Rehabilitation, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ping Qi
- Department of Pediatrics, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | | | - Yiping Wang
- Department of Critical Care Medicine, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hailian Wang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Center of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, China
| | - Peng Cao
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Yu'e Liu
- Tongji University Cancer Center, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Yi Wang
- Department of Critical Care Medicine, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Center of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, China.
| |
Collapse
|
5
|
Zhang L, Yu S, Guan Y, Wang D, Yang J, Li J, Zhao W, Zhang F. Dual intervention on the gut and skin microbiota attenuates facial cutaneous aging. Food Funct 2024; 15:4246-4261. [PMID: 38526064 DOI: 10.1039/d3fo05473j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
The gut and skin microbiota are microbial barriers, resisting harmful foreign microorganisms and maintaining internal homeostasis. Dysbiosis of the gut and skin microbiota is involved in aging progression. However, interventions targeting facial skin wellness taking into account the gut-skin axis are scarce. In this study, the impact of an eight-week intervention with oral (O), topical (T), and both oral and topical (OT) xylo-oligosaccharides (XOS) by regulating gut and skin microbiota on facial cutaneous aging was investigated in a double-blind placebo-controlled trial in females. An increase in the proportion of participants with skin rejuvenation was observed, along with a significant reduction in facial pores after OT intervention. The reduction of cutaneous Cutibacterium by OT intervention was greater than that in the O and T groups. These interventions can change the skin microbial structure. Intestinal Bifidobacterium was enriched only by dual treatment with oral and topical XOS. Function prediction analysis revealed a decrease in K02770 encoding fructose-1-phosphate kinase involved in de novo lipid synthesis from fructose with dual intervention, suggesting that inhibition of lipophilic Cutibacterium may contribute to reducing facial pores. Overall, the dual XOS intervention approach is most effective for improving both gut and skin microbiota, as well as facial skin aging.
Collapse
Affiliation(s)
- Liujing Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.
| | - Shun Yu
- Department of Plastic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Yin Guan
- Huishan Community Health Service Center, Health Commission of Liangxi District, Wuxi, China
| | - Dan Wang
- Department of Plastic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Ju Yang
- Department of Nutrition, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Jingling Li
- Department of Plastic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Wei Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.
| | - Feng Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China.
- Department of Nutrition, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Functional Food Clinical Evaluation Center, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- School of Bioengineering, Jiangnan University, Wuxi, Jiangsu, China
- School of Environment and Civil Engineering, Jiangnan University, Wuxi, Jiangsu, China
- Chinese Society of Nutritional Oncology, Beijing, China
| |
Collapse
|
6
|
Zuriaga E, Santander S, Lomba L, Izquierdo-García E, Luesma MJ. Descriptive Analysis of Carrier and Affected Hereditary Fructose Intolerance in Women during Pregnancy. Healthcare (Basel) 2024; 12:573. [PMID: 38470684 PMCID: PMC10930640 DOI: 10.3390/healthcare12050573] [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: 01/11/2024] [Revised: 02/21/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
(1) Background: Hereditary fructose intolerance (HFI) is a rare autosomal recessive metabolic disorder resulting from aldolase B deficiency, requiring a fructose, sorbitol and sucrose (FSS)-free diet. Limited information exists on the relationship between pregnancy outcomes and HFI. This study aims to analyze pregnancy-related factors in a cohort of thirty Spanish women, with twenty-three being carriers and seven being HFI-affected (45 pregnancies). (2) Methods: A descriptive, cross-sectional and retrospective study utilized an anonymous questionnaire. (3) Results: Findings encompassed physical and emotional states, nutritional habits, pathology development and baby information. Notable results include improved physical and emotional states compared to the general population, with conventional analyses mostly within normal ranges. Persistent issues after pregnancy included hepatic steatosis, liver adenomas and hemangiomas. Carrier mothers' babies exhibited higher weight than those of patient mothers, while the weights of carrier children born with HFI were similar to disease-affected children. (4) Conclusions: Pregnant women with HFI did not significantly differ in physical and emotional states, except for nausea, vomiting, and cravings. Post-pregnancy, HFI patients and carriers exhibited persistent hepatic issues. Significantly, babies born to HFI-affected mothers had lower weights. This study sheds light on pregnancy outcomes in HFI, emphasizing potential complications and the need for ongoing monitoring and care.
Collapse
Affiliation(s)
- Estefanía Zuriaga
- Facultad de Ciencias de la Salud, Universidad San Jorge, Campus Universitario, Autov A23 km 299, 50830 Villanueva de Gállego Zaragoza, Spain; (E.Z.); (L.L.)
| | - Sonia Santander
- Faculty of Health and Sports Sciences, University of Zaragoza, 22002 Huesca, Spain
| | - Laura Lomba
- Facultad de Ciencias de la Salud, Universidad San Jorge, Campus Universitario, Autov A23 km 299, 50830 Villanueva de Gállego Zaragoza, Spain; (E.Z.); (L.L.)
| | | | - María José Luesma
- Department of Human Anatomy and Histology, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain;
| |
Collapse
|
7
|
Hong X, Edmondson AC, Strong A, Pomerantz D, Michl E, Berry G, He M. Combined PMM2-CDG and hereditary fructose intolerance in a patient with mild clinical presentation. Mol Genet Metab 2023; 140:107682. [PMID: 37597336 PMCID: PMC10840806 DOI: 10.1016/j.ymgme.2023.107682] [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: 04/30/2023] [Revised: 07/02/2023] [Accepted: 08/08/2023] [Indexed: 08/21/2023]
Abstract
We report a patient with an extremely rare, combined diagnosis of PMM2-CDG and hereditary fructose intolerance (HFI). By comparing with other patients, under-galactosylation was identified as a feature of HFI. Fructose/sorbitol/sucrose restriction was initiated right afterwards. The patient is at the mild end of the PMM2-CDG spectrum, raising the question of sorbitol's role in the pathogenesis of PMM2-CDG and whether fructose/sorbitol/sucrose restriction could benefit other PMM2-CDG patients. Additionally, epalrestat, an emerging potential PMM2-CDG therapy, may benefit HFI patients.
Collapse
Affiliation(s)
- Xinying Hong
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Andrew C Edmondson
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alanna Strong
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Daniel Pomerantz
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Emma Michl
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Gerard Berry
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Miao He
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
8
|
Beyzaei Z, Ezgu F, Imanieh MH, Haghighat M, Dehghani SM, Honar N, Geramizadeh B. Identification of a novel mutation in the ALDOB gene in hereditary fructose intolerance. J Pediatr Endocrinol Metab 2023; 36:331-334. [PMID: 36659819 DOI: 10.1515/jpem-2022-0566] [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: 11/05/2022] [Accepted: 12/15/2022] [Indexed: 01/21/2023]
Abstract
OBJECTIVES Hereditary fructose intolerance (HFI) is caused by aldolase B enzyme deficiency. There has been no report about HFI from Iran and the type of mutations has not been reported in the Iranian population so far. CASE PRESENTATION Herein we report a 2 year old girl presented with failure to thrive, hepatomegaly, and liver dysfunction. The primary impression has been hepatic glycogen storage disease type 1 or 6. This diagnosis was not confirmed by laboratory data and liver biopsy. Therefore, targeted-gene sequencing (TGS) covering 450 genes involved in inborn errors in metabolic diseases was performed. The results of TGS showed a rare novel homozygous pathogenic variant c.944del (p.Gly315ValfsTer15) in the ALDOB gene. CONCLUSIONS This report introduces a novel variant that expands the mutational spectrum of the ALDOB gene in patients with HFI.
Collapse
Affiliation(s)
- Zahra Beyzaei
- Shiraz Transplant Research Center (STRC), Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatih Ezgu
- Department of Pediatric Metabolism and Genetics, Gazi University Faculty of Medicine, Ankara, Türkiye
| | - Mohammad Hadi Imanieh
- Gastroenterology and Hepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahmoud Haghighat
- Gastroenterology and Hepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohsen Dehghani
- Gastroenterology and Hepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Naser Honar
- Gastroenterology and Hepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bita Geramizadeh
- Shiraz Transplant Research Center (STRC), Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pathology, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
9
|
Sukiasyan L. Fructose-Induced Alteration of the Heart and Vessels Homeostasis. Curr Probl Cardiol 2023; 48:101013. [PMID: 34637847 DOI: 10.1016/j.cpcardiol.2021.101013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 01/04/2023]
Abstract
To date, the role of uncontrolled sugar consumption in the triggering and progression of cardiovascular events is undeniable. Modern concepts offer a new hypothesis regarding the direct myocardiotoxic effects of fructose. Experimental studies have demonstrated that cardiomyocytes have a unique ability to transport and use fructose along with the expression of all components involved in fructose metabolism. The purpose of this review article is to assess and analyze the available knowledge on fructose-induced cardiotoxicity detection since understanding the pathophysiological mechanisms and pathobiochemical aspects will become the basis for the determination of a rational myocardioprotection regimen.
Collapse
Affiliation(s)
- Lilit Sukiasyan
- Yerevan State Medical University after M.Heratsi, Armenia; L. A. Orbeli Institute of Human Physiology, Armenia.
| |
Collapse
|
10
|
Brouwers MCGJ. Fructose 1-phosphate, an evolutionary signaling molecule of abundancy. Trends Endocrinol Metab 2022; 33:680-689. [PMID: 35995682 DOI: 10.1016/j.tem.2022.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 11/18/2022]
Abstract
Evidence is accumulating that specifically fructose exerts adverse cardiometabolic effects in humans. Recent experimental studies have shown that fructose not only serves as a substrate for, among others, intrahepatic lipid formation, but also has a signaling function. It is postulated that fructose 1-phosphate (F1-P) has evolved as a signaling molecule of abundancy that stimulates nutrient absorption, lipid storage, and reproduction. Such a role would provide an explanation for why fructose contributes to the pathogenesis of evolutionary mismatch diseases, including nonalcoholic fatty liver disease (NAFLD), cardiovascular disease, polycystic ovary syndrome (PCOS), and colorectal cancer, in the current era of nutritional abundance. It is anticipated that reducing F1-P, by either pharmacological inhibition of ketohexokinase (KHK) or societal measures, will mitigate the risk of these diseases.
Collapse
Affiliation(s)
- Martijn C G J Brouwers
- Department of Internal Medicine, Division of Endocrinology and Metabolic Disease, Maastricht University Medical Centre, Maastricht, The Netherlands; CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
11
|
Koene E, Schrauwen-Hinderling VB, Schrauwen P, Brouwers MCGJ. Novel insights in intestinal and hepatic fructose metabolism: from mice to men. Curr Opin Clin Nutr Metab Care 2022; 25:354-359. [PMID: 35838297 DOI: 10.1097/mco.0000000000000853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The rise in fructose consumption in parallel with the current epidemic of obesity and related cardiometabolic disease requires a better understanding of the pathophysiological pathways that are involved. RECENT FINDINGS Animal studies have shown that fructose has various effects on the intestines that subsequently affect intrahepatic lipid accumulation and inflammation. Fructose adversely affects the gut microbiome - as a producer of endotoxins and intermediates of de novo lipogenesis - and intestinal barrier function. Furthermore, intestinal fructose metabolism shields fructose away from the liver. Finally, fructose 1-phosphate (F1-P) serves as a signal molecule that promotes intestinal cell survival and, consequently, intestinal absorption capacity. Intervention and epidemiological studies have convincingly shown that fructose, particularly derived from sugar-sweetened beverages, stimulates de novo lipogenesis and intrahepatic lipid accumulation in humans. Of interest, individuals with aldolase B deficiency, who accumulate F1-P, are characterized by a greater intrahepatic lipid content. First phase II clinical trials have recently shown that reduction of F1-P, by inhibition of ketohexokinase, reduces intrahepatic lipid content. SUMMARY Experimental evidence supports current measures to reduce fructose intake, for example by the implementation of a tax on sugar-sweetened beverages, and pharmacological inhibition of fructose metabolism to reduce the global burden of cardiometabolic disease.
Collapse
Affiliation(s)
- Evi Koene
- Department of Nutrition and Movement Sciences
- School of Nutrition and Translational Research in Metabolism (NUTRIM)
| | - Vera B Schrauwen-Hinderling
- Department of Nutrition and Movement Sciences
- School of Nutrition and Translational Research in Metabolism (NUTRIM)
- Department of Radiology and Nuclear Medicine, Maastricht University
| | - Patrick Schrauwen
- Department of Nutrition and Movement Sciences
- School of Nutrition and Translational Research in Metabolism (NUTRIM)
| | - Martijn C G J Brouwers
- Division of Endocrinology and Metabolic Diseases, Department of Internal Medicine, Maastricht University Medical Center
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
12
|
Tang M, Chen X, Ni Q, Lu Y, Wu B, Wang H, Yin Z, Zhou W, Dong X. Estimation of hereditary fructose intolerance prevalence in the Chinese population. Orphanet J Rare Dis 2022; 17:326. [PMID: 36028839 PMCID: PMC9419342 DOI: 10.1186/s13023-022-02487-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 08/15/2022] [Indexed: 11/15/2022] Open
Abstract
Background Hereditary fructose intolerance (HFI) caused by aldolase B reduction or deficiency that results in fructose metabolism disorder. The disease prevalence in the Chinese population is unknown, which impedes the formulation of HFI screening and diagnosis strategies. Materials and methods By searching a local cohort (Chinese Children’s Rare Disease Genetic Testing Clinical Collaboration System, CCGT) and public databases (ClinVar and Human Gene Mutation Database) and reviewing HFI-related literature, we manually curated ALDOB pathogenic or likely pathogenic (P/LP) variants according to ACMG guidelines. Allele frequency (AF) information from the local database CCGT and the public databases HuaBiao and gnomAD for ALDOB P/LP variants was used to estimate and the HFI prevalence in the Chinese population and other populations by the Bayesian framework. We collected the genotype and clinical characteristics of HFI patients from the CCGT database and published literature to study genotype–phenotype relationships. Result In total, 81 variants of ALDOB were curated as P/LP. The estimated Chinese HFI prevalence was approximately 1/504,678, which was much lower than that for non-Finland European (1/23,147), Finnish in Finland (1/55,539), admixed American (1/132,801) and Ashkenazi Jewish (1/263,150) populations. By analyzing the genetic characteristics of ALDOB in the Chinese population, two variants (A338V, A338G) had significantly higher AFs in the Chinese population than in the non-Finland European population from gnomAD (all P values < 0.05). Five variants (A150P, A175D, N335K, R60*, R304Q) had significantly lower AFs (all P values < 0.1). The genotype–phenotype association analyses were based on 68 reported HFI patients from a literature review and the CCGT database. The results showed that patients carrying homozygous variant sites (especially A150P) were more likely to present nausea, and patients carrying two missense variant sites were more likely to present aversion to sweets and fruit (all P values < 0.05). Our research reveals that some gastrointestinal symptoms seem to be associated with certain genotypes. Conclusion The prevalence of HFI in the Chinese population is extremely low, and there is no need to add HFI testing to the current newborn screening programs if medical costs are considered. A genetic testing strategy is suggested for early diagnosis of HFI. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-022-02487-3.
Collapse
Affiliation(s)
- Meiling Tang
- Center for Molecular Medicine, Children's Hospital of Fudan University, Shanghai, China
| | - Xiang Chen
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Qi Ni
- Center for Molecular Medicine, Children's Hospital of Fudan University, Shanghai, China
| | - Yulan Lu
- Center for Molecular Medicine, Children's Hospital of Fudan University, Shanghai, China
| | - Bingbing Wu
- Center for Molecular Medicine, Children's Hospital of Fudan University, Shanghai, China
| | - Huijun Wang
- Center for Molecular Medicine, Children's Hospital of Fudan University, Shanghai, China
| | - Zhaoqing Yin
- Department of Pediatrics, Dehong Hospital of Kunming Medical University, Dehong, 678400, China
| | - Wenhao Zhou
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, 201102, China.
| | - Xinran Dong
- Center for Molecular Medicine, Children's Hospital of Fudan University, Shanghai, China.
| |
Collapse
|
13
|
Elseweidy MM, Elesawy AE, Sobh MS, Elnagar GM. Ellagic acid ameliorates high fructose-induced hyperuricemia and non-alcoholic fatty liver in Wistar rats: Focusing on the role of C1q/tumor necrosis factor-related protein-3 and ATP citrate lyase. Life Sci 2022; 305:120751. [PMID: 35780841 DOI: 10.1016/j.lfs.2022.120751] [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: 05/14/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 02/07/2023]
Abstract
AIMS High-fructose intake (HF) represents an inducible risk factor for non-alcoholic fatty liver disease (NAFLD). Present study aimed to illustrate the effect of HF diet (HFD) on the induction of NAFLD, hyperuricemia and role of ellagic acid as modulator. MAIN METHODS Twenty-four adult male albino rats were randomly divided into four groups (6/each). The first group received normal chow diet only while the others received 60 % HFD for 4 weeks and subdivided later into 3 groups. The first and second groups received allopurinol and ellagic acid, respectively while the third group received HFD only for extra 4 weeks. KEY FINDINGS Rats fed on HFD for 8 weeks displayed body weight gain, insulin resistance (IR), hyperglycemia, dyslipidemia, hyperuricemia with increased oxidative stress and hepatic lipogenic enzymes such as ATP citrate lyase (ACL), aldolase B, and fatty acid synthase (FAS), sterol regulatory element-binding protein 1 (SERBP-1c). C1q /tumor necrosis factor-related protein -3 (CTRP3), and phosphorylated AMP-activated protein kinase (p-AMPK) however showed significant decreases. Ellagic acid or allopurinol administration significantly decreased serum lipids, uric acid, glucose, insulin levels and hepatic contents of enzymes. Malondialdehyde (MDA), FAS, aldolase B, SERBP-1c, and xanthine oxidase (XO) hepatic contents showed significant decreases along with glutathione (GSH) increase as compared to fructose group where ellagic acid was more remarkable compared to allopurinol. SIGNIFICANCE Our findings indicated that ellagic acid had alleviated HFD-induced hyperuricemia, its associated NAFLD pattern as mediated through activation of CTRP3 and inhibition of ACL activities in a pattern more remarkable than allopurinol.
Collapse
Affiliation(s)
| | - Ahmed E Elesawy
- Biochemistry Department, Faculty of Pharmacy, Badr University in Cairo)BUC), Egypt
| | - Mohammed S Sobh
- Pathology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Gehad M Elnagar
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Egypt
| |
Collapse
|
14
|
Wright K, Nip KM, Kim JE, Cheng KM, Birol I. Seasonal and sex-dependent gene expression in emu (Dromaius novaehollandiae) fat tissues. Sci Rep 2022; 12:9419. [PMID: 35676317 PMCID: PMC9177602 DOI: 10.1038/s41598-022-13681-5] [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: 10/21/2021] [Accepted: 05/10/2022] [Indexed: 12/02/2022] Open
Abstract
Emu (Dromaius novaehollandiae) farming has been gaining wide interest for fat production. Oil rendered from this large flightless bird’s fat is valued for its anti-inflammatory and antioxidant properties for uses in therapeutics and cosmetics. We analyzed the seasonal and sex-dependent differentially expressed (DE) genes involved in fat metabolism in emus. Samples were taken from back and abdominal fat tissues of a single set of four male and four female emus in April, June, and November for RNA-sequencing. We found 100 DE genes (47 seasonally in males; 34 seasonally in females; 19 between sexes). Seasonally DE genes with significant difference between the sexes in gene ontology terms suggested integrin beta chain-2 (ITGB2) influences fat changes, in concordance with earlier studies. Six seasonally DE genes functioned in more than two enriched pathways (two female: angiopoietin-like 4 (ANGPTL4) and lipoprotein lipase (LPL); four male: lumican (LUM), osteoglycin (OGN), aldolase B (ALDOB), and solute carrier family 37 member 2 (SLC37A2)). Two sexually DE genes, follicle stimulating hormone receptor (FSHR) and perilipin 2 (PLIN2), had functional investigations supporting their influence on fat gain and loss. The results suggested these nine genes influence fat metabolism and deposition in emus.
Collapse
|
15
|
Rodríguez L, López A, Moyna G, Seoane GA, Davyt D, Vázquez Á, Hernández G, Carrera I. New Insights into the Chemical Composition of Ayahuasca. ACS OMEGA 2022; 7:12307-12317. [PMID: 35449956 PMCID: PMC9016809 DOI: 10.1021/acsomega.2c00795] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/21/2022] [Indexed: 06/14/2023]
Abstract
Ayahuasca is a psychedelic beverage originally from the Amazon rainforest used in different shamanic settings for medicinal, spiritual, and cultural purposes. It is prepared by boiling in water an admixture of the Amazonian vine Banisteriopsis caapi, which is a source of β-carboline alkaloids, with plants containing N,N-dimethyltryptamine, usually Psychotria viridis. While previous studies have focused on the detection and quantification of the alkaloids present in the drink, less attention has been given to other nonalkaloid components or the composition of the solids suspended in the beverage, which may also affect its psychoactive properties. In this study, we used nuclear magnetic resonance (NMR) and liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS) to study the composition of ayahuasca samples, to determine their alkaloid qualitative and quantitative profiles, as well as other major soluble and nonsoluble components. For the first time, fructose was detected as a major component of the samples, while harmine (a β-carboline previously described as an abundant alkaloid in ayahuasca) was found to be present in the solids suspended in the beverage. In addition, N,N-dimethyltryptamine (DMT), harmine, tetrahydroharmine, harmaline, and harmol were identified as the major alkaloids present in extracts of all samples. Finally, a novel, easy, and fast method using quantitative NMR was developed and validated to simultaneously quantify the content of these alkaloids found in each ayahuasca sample.
Collapse
Affiliation(s)
- Luisina Rodríguez
- Laboratorio
de Farmacognosia, Departamento de Química Orgánica, Universidad de la República, Avenue General Flores 2124, Montevideo 11800, Uruguay
- Laboratorio
de Síntesis Orgánica, Departamento de Química
Orgánica, Universidad de la República, Avenue General Flores 2124, Montevideo 11800, Uruguay
- Arché-
Núcleo Interdisciplinario de Estudios sobre Psicodélicos-
Espacio Interdisciplinario, Universidad
de la República, José Enrique Rodó 1843, Montevideo 11200, Uruguay
| | - Andrés López
- Departamento
de Química del Litoral, Centro Universitario Regional Litoral
Norte, Universidad de la República, Ruta 3, km 363, Paysandú 60000, Uruguay
| | - Guillermo Moyna
- Departamento
de Química del Litoral, Centro Universitario Regional Litoral
Norte, Universidad de la República, Ruta 3, km 363, Paysandú 60000, Uruguay
| | - Gustavo A. Seoane
- Laboratorio
de Síntesis Orgánica, Departamento de Química
Orgánica, Universidad de la República, Avenue General Flores 2124, Montevideo 11800, Uruguay
- Arché-
Núcleo Interdisciplinario de Estudios sobre Psicodélicos-
Espacio Interdisciplinario, Universidad
de la República, José Enrique Rodó 1843, Montevideo 11200, Uruguay
| | - Danilo Davyt
- Laboratorio
de Química Farmacéutica, Departamento de Química
Orgánica, Universidad de la República, Avenue General Flores 2124, Montevideo 11800, Uruguay
| | - Álvaro Vázquez
- Laboratorio
de Farmacognosia, Departamento de Química Orgánica, Universidad de la República, Avenue General Flores 2124, Montevideo 11800, Uruguay
- Arché-
Núcleo Interdisciplinario de Estudios sobre Psicodélicos-
Espacio Interdisciplinario, Universidad
de la República, José Enrique Rodó 1843, Montevideo 11200, Uruguay
| | - Gonzalo Hernández
- Laboratorio
de Resonancia Magnética Nuclear, Departamento de Química
Orgánica, Universidad de la República, Avenue General Flores 2124, Montevideo 11800, Uruguay
| | - Ignacio Carrera
- Laboratorio
de Síntesis Orgánica, Departamento de Química
Orgánica, Universidad de la República, Avenue General Flores 2124, Montevideo 11800, Uruguay
- Arché-
Núcleo Interdisciplinario de Estudios sobre Psicodélicos-
Espacio Interdisciplinario, Universidad
de la República, José Enrique Rodó 1843, Montevideo 11200, Uruguay
| |
Collapse
|
16
|
Gorce M, Lebigot E, Arion A, Brassier A, Cano A, De Lonlay P, Feillet F, Gay C, Labarthe F, Nassogne MC, Roche S, Roubertie A, Sacaze E, Touati G, Broué P. Fructose-1,6-bisphosphatase deficiency causes fatty liver disease and requires long-term hepatic follow-up. J Inherit Metab Dis 2022; 45:215-222. [PMID: 34687058 DOI: 10.1002/jimd.12452] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/15/2021] [Accepted: 10/20/2021] [Indexed: 11/06/2022]
Abstract
Liver disease, occurring during pediatric or adult age, is often of undetermined cause. Some cases are probably related to undiagnosed inherited metabolic disorders. Hepatic disorders associated with fructose-1,6-bisphosphatase deficiency, a gluconeogenesis defect, are not reported in the literature. These symptoms are mainly described during acute crises, and many reports do not mention them because hypoglycemia and hyperlactatemia are more frequently in the forefront. Herein, the liver manifestations of 18 patients affected with fructose-1,6-bisphosphatase deficiency are described and the corresponding literature is reviewed. Interestingly, all 18 patients had liver abnormalities either during follow-up (hepatomegaly [n = 8/18], elevation of transaminases [n = 6/15], bright liver [n = 7/11]) or during acute crises (hepatomegaly [n = 10/17], elevation of transaminases [n = 13/16], acute liver failure [n = 6/14], bright liver [n = 4/14]). Initial reports described cases of liver steatosis, when liver biopsy was necessary to confirm the diagnosis by an enzymatic study. There is no clear pathophysiological basis for this fatty liver disease but we postulate that endoplasmic reticulum stress and de novo lipogenesis activation could be key factors, as observed in FBP1 knockout mice. Liver steatosis may expose patients to severe long-term liver complications. As hypoglycemia becomes less frequent with age, most adult patients are no longer monitored by hepatologist. Signs of fructose-1,6-bisphosphatase deficiency may be subtle and can be missed in childhood. We suggest that fructose-1,6-bisphosphatase deficiency should be considered as an etiology of hepatic steatosis, and a liver monitoring protocol should be set up for these patients, during lifelong follow-up.
Collapse
Affiliation(s)
- Magali Gorce
- Centre de référence des maladies héréditaires du métabolisme, Unité pédiatrique de Gastro-entérologie, hépatologie, nutrition et Maladies héréditaires du métabolisme, CHU de Toulouse-Hôpital des Enfants, Toulouse, France
| | - Elise Lebigot
- APHP, CHU Bicêtre, Service de biochimie, Le Kremlin-Bicêtre, Paris, France
| | - Alina Arion
- Centre de compétence des maladies héréditaires du métabolisme, Service de pédiatrie médicale, CHU de Caen - Hôpital de la Côte de Nacre, Caen, France
| | - Anaïs Brassier
- Centre de référence des maladies héréditaires du métabolisme, Institut Imagine, Hôpital Necker, Université Paris-Descartes, Paris, France
| | - Aline Cano
- Centre de référence des maladies héréditaires du métabolisme, Service de neurologie pédiatrique, Pédiatrie spécialisée et médecine infantile, CHU de Marseille-Hôpital de la Timone, Marseille, France
| | - Pascale De Lonlay
- Centre de référence des maladies héréditaires du métabolisme, Institut Imagine, Hôpital Necker, Université Paris-Descartes, Paris, France
| | - François Feillet
- Centre de référence des maladies héréditaires du métabolisme, Service de médecine infantile, CHU Brabois Enfants, Vandœuvre-lès-Nancy, France
| | - Claire Gay
- Centre de compétence des maladies héréditaires du métabolisme, Service de Pédiatrie, CHU de Saint-Etienne-Hôpital Nord, Saint Etienne, France
| | - François Labarthe
- Centre de référence des maladies héréditaires du métabolisme, Service de Médecine pédiatrique, Pediatrics Departement, CHRU de Tours-Hôpital Clocheville, Tours, France
| | - Marie-Cécile Nassogne
- Service de neurologie pédiatrique et Centre de référence des maladies métaboliques héréditaires-Cliniques Universitaires Saint-Luc, UCLouvain, Bruxelles, Belgium
| | - Sandrine Roche
- Centre de compétence des maladies héréditaires du métabolisme, Service de pédiatrie médicale, CHU Bordeaux, hôpital Pellegrin, Bordeaux, France
| | - Agathe Roubertie
- Centre de compétence des maladies héréditaires du métabolisme, Département de neuropédiatrie, Pôle Femme Mère Enfant, CHRU de Montpellier-Hôpital Gui de Chauliac, Montpellier, France
| | - Elise Sacaze
- Centre de compétence des maladies héréditaires du métabolisme, Service de Pédiatrie, Pôle Femme-Mère-Enfant, CHRU de Brest-Hôpital Morvan, Brest, France
| | - Guy Touati
- Centre de référence des maladies héréditaires du métabolisme, Unité pédiatrique de Gastro-entérologie, hépatologie, nutrition et Maladies héréditaires du métabolisme, CHU de Toulouse-Hôpital des Enfants, Toulouse, France
| | - Pierre Broué
- Centre de référence des maladies héréditaires du métabolisme, Unité pédiatrique de Gastro-entérologie, hépatologie, nutrition et Maladies héréditaires du métabolisme, CHU de Toulouse-Hôpital des Enfants, Toulouse, France
| |
Collapse
|
17
|
Apgar TL, Sanders CR. Compendium of causative genes and their encoded proteins for common monogenic disorders. Protein Sci 2022; 31:75-91. [PMID: 34515378 PMCID: PMC8740837 DOI: 10.1002/pro.4183] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/29/2021] [Accepted: 08/31/2021] [Indexed: 01/19/2023]
Abstract
A compendium is presented of inherited monogenic disorders that have a prevalence of >1:20,000 in the human population, along with their causative genes and encoded proteins. "Simple" monogenic diseases are those for which the clinical features are caused by mutations impacting a single gene, usually in a manner that alters the sequence of the encoded protein. Of course, for a given "monogenic disorder", there is sometimes more than one potential disease gene, mutations in any one of which is sufficient to cause phenotypes of that disorder. Disease-causing mutations for monogenic disorders are usually passed on from generation to generation in a Mendelian fashion, and originate from spontaneous (de novo) germline founder mutations. In the past monogenic disorders have often been written off as targets for drug discovery because they sometimes are assumed to be rare disorders, for which the meager projected financial payoff of drug discovery and development has discouraged investment. However, not all monogenic diseases are rare. Here, we report that that currently available data identifies 72 disorders with a prevalence of at least 1 in 20,000 humans. For each, we tabulate the gene(s) for which mutations cause the spectrum of phenotypes associated with that disorder. We also identify the gene and protein that most commonly causes each disease. 34 of these disorders are caused exclusively by mutations in only a single gene and encoded protein.
Collapse
Affiliation(s)
- Tucker L. Apgar
- Department of Biochemistry and Center for Structural BiologyVanderbilt University School of Medicine Basic SciencesNashvilleTennesseeUSA
| | - Charles R. Sanders
- Department of Biochemistry and Center for Structural BiologyVanderbilt University School of Medicine Basic SciencesNashvilleTennesseeUSA
| |
Collapse
|
18
|
Bobrus-Chociej A, Pollak A, Kopiczko N, Flisiak-Jackiewicz M, Płoski R, Lebensztejn DM. Celiac Disease in Conjunction with Hereditary Fructose Intolerance as a Rare Cause of Liver Steatosis with Mild Hypertransaminasemia-A Case Report. Pediatr Rep 2021; 13:589-593. [PMID: 34842789 PMCID: PMC8628906 DOI: 10.3390/pediatric13040070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 12/11/2022] Open
Abstract
Celiac disease (CD) has been associated with several genetic and autoimmune disorders, but its association with hereditary fructose intolerance (HFI) is very rare. The possibility of an association between CD and HFI should be considered, especially in patients with a lack of improvement after a gluten-free diet. Children with HFI often present with a wide range of symptoms, however, data about a strong aversion to fruits and sweets may be helpful to establish the diagnosis. The diagnosis of HFI should be confirmed in genetic testing. Both CD and HFI may present with liver steatosis with hypertransaminasemia. In patients with these two disorders, the dietary restrictions of gluten and fructose improve clinical symptoms and protect them from secondary complications. We report the case of a child with the concurrence of these two disorders.
Collapse
Affiliation(s)
- Anna Bobrus-Chociej
- Department of Pediatrics, Gastroenterology, Hepatology, Nutrition and Allergology, Medical University of Białystok, Jana Kilińskiego 1, 15-089 Białystok, Poland
| | - Agnieszka Pollak
- Department of Medical Genetics, Medical University of Warsaw, Żwirki i Wigury 61, 02-091 Warsaw, Poland
| | - Natalia Kopiczko
- Department of Pediatrics, Gastroenterology, Hepatology, Nutrition and Allergology, Medical University of Białystok, Jana Kilińskiego 1, 15-089 Białystok, Poland
| | - Marta Flisiak-Jackiewicz
- Department of Pediatrics, Gastroenterology, Hepatology, Nutrition and Allergology, Medical University of Białystok, Jana Kilińskiego 1, 15-089 Białystok, Poland
| | - Rafał Płoski
- Department of Medical Genetics, Medical University of Warsaw, Żwirki i Wigury 61, 02-091 Warsaw, Poland
| | - Dariusz M Lebensztejn
- Department of Pediatrics, Gastroenterology, Hepatology, Nutrition and Allergology, Medical University of Białystok, Jana Kilińskiego 1, 15-089 Białystok, Poland
| |
Collapse
|
19
|
Ali Z, Saeed IM, Bybee KA, Thompson R, O'Keefe JH, Shafiq M, Saeed L, Zafar Y, Kennedy KF, Al-Sayyed L. Correlation of Hepatic Steatosis Among Cohabitants Using Hounsfield Unit From Coronary Computed Tomography. Cureus 2021; 13:e17834. [PMID: 34660042 PMCID: PMC8502746 DOI: 10.7759/cureus.17834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2021] [Indexed: 11/30/2022] Open
Abstract
Background Individuals living in the same household are exposed to common risk factors. We hypothesized that living with someone who has fatty liver disease increases the risk of having the same disease. Methods This was a retrospective study that included pairs of men and women who shared the same residential addresses, underwent screening non-contrast computed tomography for coronary calcium scoring and had Hounsfield Unit density for liver and spleen in the field of view available for measurement. The primary goal was to determine the association between hepatic steatosis and living in the same household. Secondary end-points compared to body mass index, triglyceride levels, type 2 diabetes mellitus (T2DM) and hypertension. Results Out of 1,362 cohabitant pairs, there were 202 couples with either the male or female having hepatic steatosis and 10 cohabitant pairs with both the male and female having hepatic steatosis. In 1,150 cohabitant pairs out of 1,362, neither man nor woman had hepatic steatosis. Pearson correlation coefficient (r) for hepatic steatosis between cohabitant pairs was 0.122 (p-value: < 0.001), suggesting that no correlation was found. Elevated triglyceride levels were prevalent among cohabitant pairs with hepatic steatosis, when compared to pairs without hepatic steatosis (p-value < 0.05). Female gender and having a diagnosis of hepatic steatosis also showed a strong association with higher body mass index, T2DM and hypertension (p-value < 0.05). Conclusion Despite the assumption of exposure to similar environmental factors, our results did not show any correlation of hepatic steatosis among the cohabitants.
Collapse
Affiliation(s)
- Zafar Ali
- Internal Medicine, The University of Kansas Medical Center, Kansas City, USA
| | | | - Kevin A Bybee
- Cardiovascular Disease, Saint Luke's Mid America Heart Institute, Kansas City, USA
| | - Randall Thompson
- Cardiovascular Disease, Saint Luke's Mid America Heart Institute, Kansas City, USA
| | - James H O'Keefe
- Cardiovascular Disease, Saint Luke's Mid America Heart Institute, Kansas City, USA
| | - Muhammad Shafiq
- Internal Medicine, The University of Kansas Medical Center, Kansas City, USA
| | - Lyla Saeed
- Internal Medicine, University of Missouri Kansas City School of Medicine, Kansas City, USA
| | - Yousaf Zafar
- Internal Medicine, Naples Community Healthcare, Naples, USA
| | - Kevin F Kennedy
- Biostatistics, Saint Luke's Mid America Heart Institute, Kansas City, USA
| | - Leen Al-Sayyed
- Gastroenterology and Hepatology, University of Missouri Kansas City School of Medicine, Kansas City, USA
| |
Collapse
|
20
|
Abstract
Accumulation of phosphorylated intermediates during cellular metabolism can have wide-ranging toxic effects on many organisms, including humans and the pathogens that infect them. These toxicities can be induced by feeding an upstream metabolite (a sugar, for instance) while simultaneously blocking the appropriate metabolic pathway with either a mutation or an enzyme inhibitor. Here, we survey the toxicities that can arise in the metabolism of glucose, galactose, fructose, fructose-asparagine, glycerol, trehalose, maltose, mannose, mannitol, arabinose, and rhamnose. Select enzymes in these metabolic pathways may serve as novel therapeutic targets. Some are conserved broadly among prokaryotes and eukaryotes (e.g., glucose and galactose) and are therefore unlikely to be viable drug targets. However, others are found only in bacteria (e.g., fructose-asparagine, rhamnose, and arabinose), and one is found in fungi but not in humans (trehalose). We discuss what is known about the mechanisms of toxicity and how resistance is achieved in order to identify the prospects and challenges associated with targeted exploitation of these pervasive metabolic vulnerabilities.
Collapse
|
21
|
Pinheiro FC, Sperb-Ludwig F, Schwartz IVD. Epidemiological aspects of hereditary fructose intolerance: A database study. Hum Mutat 2021; 42:1548-1566. [PMID: 34524712 DOI: 10.1002/humu.24282] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 09/05/2021] [Accepted: 09/11/2021] [Indexed: 12/11/2022]
Abstract
Hereditary fructose intolerance (HFI) is an inborn error of fructose metabolism of autosomal recessive inheritance caused by pathogenic variants in the ALDOB gene that lead to aldolase B deficiency in the liver, kidneys, and intestine. Patients manifest symptoms, such as ketotic hypoglycemia, vomiting, nausea, in addition to hepatomegaly and other liver and kidney dysfunctions. The treatment consists of a fructose-restricted diet, which results in a good prognosis. To analyze the distribution of ALDOB variants described in patients and to estimate the prevalence of HFI based on carrier frequency in the gnomAD database, a systematic review was conducted to assess ALDOB gene variants among patients with HFI. The prevalence of HFI was estimated from the carrier frequency of variants described in patients, as well as rare variants predicted as pathogenic by in silico tools. The p.(Ala150Pro) and p.(Ala175Asp) variants are the most frequent and are distributed worldwide. However, these variants have particular distribution patterns in Europe. The analysis of the prevalence of HFI showed that the inclusion of rare alleles predicted as pathogenic is a more informative approach for populations with few patients. The data show that HFI has a wide distribution and an estimated prevalence of ~1:10,000.
Collapse
Affiliation(s)
- Franciele C Pinheiro
- Post-Graduate Program in Genetics and Molecular Biology, Federal University of do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,BRAIN Laboratory, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil.,Federal University of Pampa, Itaqui, Rio Grande do Sul, Brazil
| | - Fernanda Sperb-Ludwig
- Post-Graduate Program in Genetics and Molecular Biology, Federal University of do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,BRAIN Laboratory, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Ida V D Schwartz
- Post-Graduate Program in Genetics and Molecular Biology, Federal University of do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,BRAIN Laboratory, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil.,Department of Genetics, Bioscience Institute, Federal University of do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
22
|
Gunduz M, Ünal-Uzun Ö, Koç N, Ceylaner S, Özaydın E, Kasapkara ÇS. Molecular and clinical findings of Turkish patients with hereditary fructose intolerance. J Pediatr Endocrinol Metab 2021; 34:1017-1022. [PMID: 34162028 DOI: 10.1515/jpem-2021-0303] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 05/25/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Hereditary fructose intolerance (HFI) is an autosomal recessive disorder caused by a deficiency in aldolase B that can result in hypoglycemia, nausea, vomiting, abdominal pain, liver and kidney dysfunction, coma, and even death. This study aims to represent the clinical features and molecular genetic analysis data of the patients diagnosed with HFI in our study population. METHODS The medical records of the 26 patients with HFI were evaluated retrospectively. Age, gender, clinical findings, metabolic crises, and the results of molecular analyses were recorded. RESULTS The patients with HFI had a good prognosis and the aversion to sugar-containing foods was the main complaint. Seven different variants were identified in the Aldolase B (ALDOB) gene in HFI patients. The most frequent mutations were p.Ala150Pro, p.Ala175Asp had a prevalence of 61 and 30%, respectively, in agreement with the literature and other known variants were found with minor frequencies c.360-363del4(3.8%), p.Asn335Lys(3.8%), and three novel mutations c.113-1_15del4 (3.8%), p.Ala338Val(7.6%), and p.Asp156His(3.8%) were identified at a heterozygous, homozygous, or compound heterozygous level. CONCLUSIONS This study results revealed three novel mutations in patients with HFI. On the basis of age of presentation, clinical symptoms, and metabolic crisis, there was no clear-cut genotype-phenotype correlation. This article also demonstrates the importance of screening suspected infants in cases of acute liver failure for prompt diagnosis and treatment of HFI.
Collapse
Affiliation(s)
- Mehmet Gunduz
- Ankara City Hospital, Department of Pediatric Metabolism, Ankara, Turkey
| | - Özlem Ünal-Uzun
- Kocaeli University, Department of Pediatric Metabolism, Kocaeli, Turkey
| | - Nevra Koç
- University of Health Sciences, Gulhane Health Sciences Faculty, Department of Nutrition and Dietetics, Ankara, Turkey
| | - Serdar Ceylaner
- Genetics, İntergen Genetic Diseases Diagnostic Center, Ankara, Turkey
| | - Eda Özaydın
- Ankara City Hospital, Department of Pediatrics, Ankara, Turkey
| | | |
Collapse
|
23
|
Koch F, Albrecht D, Görs S, Kuhla B. Jejunal mucosa proteomics unravel metabolic adaptive processes to mild chronic heat stress in dairy cows. Sci Rep 2021; 11:12484. [PMID: 34127774 PMCID: PMC8203643 DOI: 10.1038/s41598-021-92053-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/04/2021] [Indexed: 12/31/2022] Open
Abstract
Climate change affects the duration and intensity of heat waves during summer months and jeopardizes animal health and welfare. High ambient temperatures cause heat stress in dairy cows resulting in a reduction of milk yield, feed intake, and alterations in gut barrier function. The objectives of this study were to investigate the mucosal amino acid, glucose and lactate metabolism, as well as the proteomic response of the small intestine in heat stressed (HS) Holstein dairy cows. Cows of the HS group (n = 5) were exposed for 4 days to 28 °C (THI = 76) in a climate chamber. Percentage decrease in daily ad libitum intake of HS cows was calculated to provide isocaloric energy intake to pair-fed control cows kept at 15 °C (THI = 60) for 4 days. The metabolite, mRNA and proteomic analyses revealed that HS induced incorrect protein folding, cellular destabilization, increased proteolytic degradation and protein kinase inhibitor activity, reduced glycolysis, and activation of NF-κB signaling, uronate cycling, pentose phosphate pathway, fatty acid and amino acid catabolism, mitochondrial respiration, ATPase activity and the antioxidative defence system. Our results highlight adaptive metabolic and immune mechanisms attempting to maintain the biological function in the small intestine of heat-stressed dairy cows.
Collapse
Affiliation(s)
- Franziska Koch
- Institute of Nutritional Physiology "Oskar Kellner", Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Dirk Albrecht
- Institute of Microbiology, Ernst-Moritz-Arndt-University, Felix-Hausdorff-Straße 8, 17487, Greifswald, Germany
| | - Solvig Görs
- Institute of Nutritional Physiology "Oskar Kellner", Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Björn Kuhla
- Institute of Nutritional Physiology "Oskar Kellner", Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| |
Collapse
|
24
|
Diagnosis and management of secondary causes of steatohepatitis. J Hepatol 2021; 74:1455-1471. [PMID: 33577920 DOI: 10.1016/j.jhep.2021.01.045] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 01/09/2021] [Accepted: 01/25/2021] [Indexed: 02/06/2023]
Abstract
The term non-alcoholic fatty liver disease (NAFLD) was originally coined to describe hepatic fat deposition as part of the metabolic syndrome. However, a variety of rare hereditary liver and metabolic diseases, intestinal diseases, endocrine disorders and drugs may underlie, mimic, or aggravate NAFLD. In contrast to primary NAFLD, therapeutic interventions are available for many secondary causes of NAFLD. Accordingly, secondary causes of fatty liver disease should be considered during the diagnostic workup of patients with fatty liver disease, and treatment of the underlying disease should be started to halt disease progression. Common genetic variants in several genes involved in lipid handling and metabolism modulate the risk of progression from steatosis to fibrosis, cirrhosis and hepatocellular carcinoma development in NAFLD, alcohol-related liver disease and viral hepatitis. Hence, we speculate that genotyping of common risk variants for liver disease progression may be equally useful to gauge the likelihood of developing advanced liver disease in patients with secondary fatty liver disease.
Collapse
|
25
|
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: 2.3] [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
|
26
|
Shi YN, Liu YJ, Xie Z, Zhang WJ. Fructose and metabolic diseases: too much to be good. Chin Med J (Engl) 2021; 134:1276-1285. [PMID: 34010200 PMCID: PMC8183764 DOI: 10.1097/cm9.0000000000001545] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Indexed: 12/15/2022] Open
Abstract
ABSTRACT Excessive consumption of fructose, the sweetest of all naturally occurring carbohydrates, has been linked to worldwide epidemics of metabolic diseases in humans, and it is considered an independent risk factor for cardiovascular diseases. We provide an overview about the features of fructose metabolism, as well as potential mechanisms by which excessive fructose intake is associated with the pathogenesis of metabolic diseases both in humans and rodents. To accomplish this aim, we focus on illuminating the cellular and molecular mechanisms of fructose metabolism as well as its signaling effects on metabolic and cardiovascular homeostasis in health and disease, highlighting the role of carbohydrate-responsive element-binding protein in regulating fructose metabolism.
Collapse
Affiliation(s)
- Ya-Nan Shi
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin 300134, China
| | - Ya-Jin Liu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin 300134, China
| | - Zhifang Xie
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200240, China
| | - Weiping J. Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin 300134, China
- Department of Pathophysiology, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
27
|
Debray FG, Seyssel K, Fadeur M, Tappy L, Paquot N, Tran C. Effect of a high fructose diet on metabolic parameters in carriers for hereditary fructose intolerance. Clin Nutr 2021; 40:4246-4254. [PMID: 33551217 DOI: 10.1016/j.clnu.2021.01.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/04/2021] [Accepted: 01/17/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Hyperuricemia is an independent risk factor for the metabolic syndrome and cardiovascular disease. We hypothesized that asymptomatic carriers for hereditary fructose intolerance (OMIM 22960) would have increased uric acid and altered component of the metabolic syndrome when exposed to fructose overfeeding. METHODS Six heterozygotes for HFI (hHFI) and 6 controls (Ctrl) were studied in a randomized, controlled, crossover trial. Participants ingested two identical test meals containing 0.7 g kg-1 glucose and 0.7 g kg-1 fructose according to a cross-over design, once after a 7-day on a low fructose diet (LoFruD, <10 g/d) and on another occasion after 7 days on a high fructose diet (HiFruD, 1.4 g kg-1 day-1 fructose + 0.1 g kg-1 day-1 glucose). Uric acid, glucose, and insulin concentrations were monitored in fasting conditions and over 2 h postprandial, and insulin resistance indexes were calculated. RESULTS HiFruD increased fasting uric acid (p < 0.05) and reduced fasting insulin sensitivity estimated by the homeostasis model assessment (HOMA) for insulin resistance (p < 0.05), in both groups. Postprandial glucose concentrations were not different between hHFI and Ctrl. However HiFruD increased postprandial plasma uric acid, insulin and hepatic insulin resistance index (HIRI) in hHFI only (all p < 0.05). CONCLUSIONS Seven days of HiFruD increased fasting uric acid and slightly reduced fasting HOMA index in both groups. In contrast, HiFruD increased postprandial uric acid, insulin concentration and HIRI in hHFI only, suggesting that heterozygosity for pathogenic Aldolase B variants may confer an increased susceptibility to the effects of dietary fructose on uric acid and hepatic insulin sensitivity. This trial was registered at the U.S. Clinical Trials Registry as NCT03545581.
Collapse
Affiliation(s)
| | - Kevin Seyssel
- Department of Biomedical Sciences, University of Lausanne, 1005 Lausanne, Switzerland
| | - Marjorie Fadeur
- Division of Diabetes, Nutrition and Metabolic Diseases, Department of Medicine CHU Sart-Tilman and GIGA I3, Immunometabolism and Nutrition Unit, University of Liège, Liège, Belgium
| | - Luc Tappy
- Department of Biomedical Sciences, University of Lausanne, 1005 Lausanne, Switzerland
| | - Nicolas Paquot
- Division of Diabetes, Nutrition and Metabolic Diseases, Department of Medicine CHU Sart-Tilman and GIGA I3, Immunometabolism and Nutrition Unit, University of Liège, Liège, Belgium
| | - Christel Tran
- Center for Molecular Diseases, Division of Genetic Medicine, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
28
|
Simons N, Veeraiah P, Simons PIHG, Schaper NC, Kooi ME, Schrauwen-Hinderling VB, Feskens EJM, van der Ploeg EMC(L, Van den Eynde MDG, Schalkwijk CG, Stehouwer CDA, Brouwers MCGJ. Effects of fructose restriction on liver steatosis (FRUITLESS); a double-blind randomized controlled trial. Am J Clin Nutr 2020; 113:391-400. [PMID: 33381794 PMCID: PMC7851818 DOI: 10.1093/ajcn/nqaa332] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND There is an ongoing debate on whether fructose plays a role in the development of nonalcoholic fatty liver disease. OBJECTIVES The aim of this study was to investigate the effects of fructose restriction on intrahepatic lipid (IHL) content in a double-blind randomized controlled trial using an isocaloric comparator. METHODS Between March 2017 and October 2019, 44 adult overweight individuals with a fatty liver index ≥ 60 consumed a 6-wk fructose-restricted diet (<7.5 g/meal and <10 g/d) and were randomly assigned to supplementation with sachets of glucose (= intervention group) or fructose (= control group) 3 times daily. Participants and assessors were blinded to the allocation. IHL content, assessed by proton magnetic resonance spectroscopy, was the primary outcome and glucose tolerance and serum lipids were the secondary outcomes. All measurements were conducted in Maastricht University Medical Center. RESULTS Thirty-seven participants completed the study protocol. After 6 wk of fructose restriction, dietary fructose intake and urinary fructose excretion were significantly lower in the intervention group (difference: -57.0 g/d; 95% CI: -77.9, -39.5 g/d; and -38.8 μmol/d; 95% CI: -91.2, -10.7 μmol/d, respectively). Although IHL content decreased in both the intervention and control groups (P < 0.001 and P = 0.003, respectively), the change in IHL content was more pronounced in the intervention group (difference: -0.7% point, 95% CI: -2.0, -0.03% point). The changes in glucose tolerance and serum lipids were not significantly different between groups. CONCLUSIONS Six weeks of fructose restriction per se led to a small, but statistically significant, decrease in IHL content in comparison with an isocaloric control group.This trial was registered at clinicaltrials.gov as NCT03067428.
Collapse
Affiliation(s)
- Nynke Simons
- Division of Endocrinology and Metabolic Diseases, 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,CARIM School for Cardiovascular Diseases, Maastricht, The Netherlands
| | - Pandichelvam Veeraiah
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands,NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht, The Netherlands
| | - Pomme I H G Simons
- Division of Endocrinology and Metabolic Diseases, 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,CARIM School for Cardiovascular Diseases, Maastricht, The Netherlands
| | - Nicolaas C Schaper
- Division of Endocrinology and Metabolic Diseases, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands,CARIM School for Cardiovascular Diseases, Maastricht, The Netherlands,CAPHRI School for Public Health and Primary Care, Maastricht, The Netherlands
| | - M Eline Kooi
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands,NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht, The Netherlands
| | - Vera B Schrauwen-Hinderling
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands,NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht, The Netherlands,Department of Nutrition and Movement Sciences, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Edith J M Feskens
- Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | | | - Mathias D G Van den Eynde
- Laboratory for Metabolism and Vascular Medicine, Division of General Internal Medicine, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands,CARIM School for Cardiovascular Diseases, 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,CARIM School for Cardiovascular Diseases, 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,CARIM School for Cardiovascular Diseases, Maastricht, The Netherlands,Division of General Internal Medicine, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | | |
Collapse
|
29
|
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: 61] [Impact Index Per Article: 15.3] [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
|
30
|
Pinheiro FC, Sperb-Ludwig F, Schwartz IVD. KHK inhibition for the treatment of hereditary fructose intolerance and nonalcoholic fatty liver disease: a double-edged sword. Cell Mol Life Sci 2020; 77:3465-3466. [PMID: 32591859 PMCID: PMC11104886 DOI: 10.1007/s00018-020-03575-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/08/2020] [Accepted: 06/15/2020] [Indexed: 12/24/2022]
Affiliation(s)
- Franciele Cabral Pinheiro
- Graduate Program in Genetics and Molecular Biology, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul RS, Brazil.
- BRAIN Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil.
- Federal University of Pampa (Unipampa), Itaqui, RS, Brazil.
- Medical Genetic Service, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 350, Porto Alegre, RS, 90035-903, Brazil.
| | - Fernanda Sperb-Ludwig
- Graduate Program in Genetics and Molecular Biology, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul RS, Brazil
- BRAIN Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil
| | - Ida Vanessa Doederlein Schwartz
- Graduate Program in Genetics and Molecular Biology, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul RS, Brazil
- BRAIN Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil
- Genetics Department, Biosciences Institute, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
- Medical Genetics Department, HCPA, Porto Alegre, RS, Brazil
| |
Collapse
|
31
|
Agius L, Chachra SS, Ford BE. The Protective Role of the Carbohydrate Response Element Binding Protein in the Liver: The Metabolite Perspective. Front Endocrinol (Lausanne) 2020; 11:594041. [PMID: 33281747 PMCID: PMC7705168 DOI: 10.3389/fendo.2020.594041] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/16/2020] [Indexed: 12/15/2022] Open
Abstract
The Carbohydrate response element binding protein, ChREBP encoded by the MLXIPL gene, is a transcription factor that is expressed at high levels in the liver and has a prominent function during consumption of high-carbohydrate diets. ChREBP is activated by raised cellular levels of phosphate ester intermediates of glycolysis, gluconeogenesis and the pentose phosphate pathway. Its target genes include a wide range of enzymes and regulatory proteins, including G6pc, Gckr, Pklr, Prkaa1,2, and enzymes of lipogenesis. ChREBP activation cumulatively promotes increased disposal of phosphate ester intermediates to glucose, via glucose 6-phosphatase or to pyruvate via glycolysis with further metabolism by lipogenesis. Dietary fructose is metabolized in both the intestine and the liver and is more lipogenic than glucose. It also induces greater elevation in phosphate ester intermediates than glucose, and at high concentrations causes transient depletion of inorganic phosphate, compromised ATP homeostasis and degradation of adenine nucleotides to uric acid. ChREBP deficiency predisposes to fructose intolerance and compromised cellular phosphate ester and ATP homeostasis and thereby markedly aggravates the changes in metabolite levels caused by dietary fructose. The recent evidence that high fructose intake causes more severe hepatocyte damage in ChREBP-deficient models confirms the crucial protective role for ChREBP in maintaining intracellular phosphate homeostasis. The improved ATP homeostasis in hepatocytes isolated from mice after chronic activation of ChREBP with a glucokinase activator supports the role of ChREBP in the control of intracellular homeostasis. It is hypothesized that drugs that activate ChREBP confer a protective role in the liver particularly in compromised metabolic states.
Collapse
|
32
|
Hereditary fructose intolerance: a diagnostic trap in infants? PEDIATRU.RO 2020. [DOI: 10.26416/pedi.60.4.2020.4076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|