101
|
Sharpton SR, Schnabl B, Knight R, Loomba R. Current Concepts, Opportunities, and Challenges of Gut Microbiome-Based Personalized Medicine in Nonalcoholic Fatty Liver Disease. Cell Metab 2021; 33:21-32. [PMID: 33296678 PMCID: PMC8414992 DOI: 10.1016/j.cmet.2020.11.010] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 10/16/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NALFD) is now a leading cause of chronic liver disease worldwide, in part, as a consequence of rapidly rising levels of obesity and metabolic syndrome and is a major risk factor for cirrhosis, hepatocellular carcinoma, and liver-related mortality. From NAFLD stems a myriad of clinical challenges related to both diagnosis and management. A growing body of evidence suggests an intricate linkage between the gut microbiome and the pathogenesis of NAFLD. We highlight how our current knowledge of the gut-liver axis in NAFLD may be leveraged to develop gut microbiome-based personalized approaches for disease management, including its use as a non-invasive biomarker for diagnosis and staging, as a target for therapeutic modulation, and as a marker of drug response. We will also discuss current limitations of these microbiome-based approaches. Ultimately, a better understanding of microbiota-host interactions in NAFLD will inform the development of novel preventative strategies and precise therapeutic targets.
Collapse
Affiliation(s)
- S R Sharpton
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA; NAFLD Research Center, Division of Gastroenterology, University of California, San Diego, La Jolla, CA, USA
| | - B Schnabl
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA; Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, USA
| | - R Knight
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, USA; Department of Computer Science & Engineering, Jacobs School of Engineering, University of California, San Diego, La Jolla, CA, USA; Department of Bioengineering, Jacobs School of Engineering, University of California, San Diego, La Jolla, CA, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - R Loomba
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA; NAFLD Research Center, Division of Gastroenterology, University of California, San Diego, La Jolla, CA, USA; Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
102
|
Oliveira DT, Chaves-Filho AB, Yoshinaga MY, Paiva NCN, Carneiro CM, Miyamoto S, Festuccia WT, Guerra-Sá R. Liver lipidome signature and metabolic pathways in nonalcoholic fatty liver disease induced by a high-sugar diet. J Nutr Biochem 2021; 87:108519. [PMID: 33017610 DOI: 10.1016/j.jnutbio.2020.108519] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 08/25/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023]
Abstract
Dietary sugar is an important determinant of the development and progression of nonalcoholic fatty liver disease (NAFLD). However, the molecular mechanisms underlying the deleterious effects of sugar intake on NAFLD under energy-balanced conditions are still poorly understood. Here, we provide a comprehensive analysis of the liver lipidome and mechanistic insights into the pathogenesis of NAFLD induced by the chronic consumption of high-sugar diet (HSD). Newly weaned male Wistar rats were fed either a standard chow diet or an isocaloric HSD for 18 weeks. Livers were harvested for histological, oxidative stress, gene expression, and lipidomic analyses. Intake of HSD increased oxidative stress and induced severe liver injury, microvesicular steatosis, and ballooning degeneration of hepatocytes. Using untargeted lipidomics, we identified and quantified 362 lipid species in the liver. Rats fed with HSD displayed increased hepatic levels of triacylglycerol enriched in saturated and monounsaturated fatty acids, lipids related to mitochondrial function/structure (phosphatidylglycerol, cardiolipin, and ubiquinone), and acylcarnitine (an intermediate lipid of fatty acid beta-oxidation). HSD-fed animals also presented increased levels of some species of membrane lipids and a decreased content of phospholipids containing omega-6 fatty acids. These changes in the lipidome were associated with the downregulation of genes involved in fatty acid oxidation in the liver. In conclusion, our data suggest that the chronic intake of a HSD, even under isocaloric conditions, induces lipid overload, and inefficient/impaired fatty acid oxidation in the liver. Such events lead to marked disturbance in hepatic lipid metabolism and the development of NAFLD.
Collapse
Affiliation(s)
- Daiane T Oliveira
- Laboratório de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Farmacêuticas, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brasil.
| | - Adriano B Chaves-Filho
- Laboratório de Fisiologia Molecular e Metabolismo, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, São Paulo, Brasil.
| | - Marcos Y Yoshinaga
- Laboratório de Lipídeos Modificados, Departamento de Bioquímica, Universidade de São Paulo, São Paulo, Brasil.
| | - Nívia Carolina N Paiva
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brasil.
| | - Cláudia M Carneiro
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brasil.
| | - Sayuri Miyamoto
- Laboratório de Lipídeos Modificados, Departamento de Bioquímica, Universidade de São Paulo, São Paulo, Brasil.
| | - William T Festuccia
- Laboratório de Fisiologia Molecular e Metabolismo, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, São Paulo, Brasil.
| | - Renata Guerra-Sá
- Laboratório de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Farmacêuticas, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brasil.
| |
Collapse
|
103
|
Al-Baiaty FDR, Ismail A, Abdul Latiff Z, Muhammad Nawawi KN, Raja Ali RA, Mokhtar NM. Possible Hepatoprotective Effect of Tocotrienol-Rich Fraction Vitamin E in Non-alcoholic Fatty Liver Disease in Obese Children and Adolescents. Front Pediatr 2021; 9:667247. [PMID: 34307250 PMCID: PMC8295474 DOI: 10.3389/fped.2021.667247] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 06/09/2021] [Indexed: 12/23/2022] Open
Abstract
Obesity has become a worldwide health concern among the pediatric population. The prevalence of non-alcoholic fatty liver disease (NAFLD) is growing rapidly, alongside the high prevalence of obesity. NAFLD refers to a multifactorial disorder that includes simple steatosis to non-alcoholic steatohepatitis (NASH) with or devoid of fibrosis. NAFLD is regarded as a systemic disorder that influences glucose, lipid, and energy metabolism with hepatic manifestations. A sedentary lifestyle and poor choice of food remain the major contributors to the disease. Prompt and timely diagnosis of NAFLD among overweight children is crucial to prevent the progression of the condition. Yet, there has been no approved pharmacological treatment for NAFLD in adults or children. As indicated by clinical evidence, lifestyle modification plays a vital role as a primary form of therapy for managing and treating NAFLD. Emphasis is on the significance of caloric restriction, particularly macronutrients (fats, carbohydrates, and proteins) in altering the disease consequences. A growing number of studies are now focusing on establishing a link between vitamins and NAFLD. Different types of vitamin supplements have been shown to be effective in treating NAFLD. In this review, we elaborate on the potential role of vitamin E with a high content of tocotrienol as a therapeutic alternative in treating NAFLD in obese children.
Collapse
Affiliation(s)
- Farah D R Al-Baiaty
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Aziana Ismail
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Zarina Abdul Latiff
- Department of Pediatrics, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Khairul Najmi Muhammad Nawawi
- Gastroenterology Unit, Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.,GUT Research Group, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Raja Affendi Raja Ali
- Gastroenterology Unit, Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.,GUT Research Group, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Norfilza Mohd Mokhtar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.,GUT Research Group, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
104
|
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
|
105
|
Alberti G, Gana JC, Santos JL. Fructose, Omega 3 Fatty Acids, and Vitamin E: Involvement in Pediatric Non-Alcoholic Fatty Liver Disease. Nutrients 2020; 12:nu12113531. [PMID: 33212947 PMCID: PMC7698421 DOI: 10.3390/nu12113531] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/30/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is currently the most common form of liver disease in both adults and children, becoming the leading cause for liver transplant in many countries. Its prevalence has increased considerably in recent years, mainly due to the explosive increase in pediatric obesity rates. NAFLD is strongly associated with central obesity, diabetes, dyslipidemia and insulin resistance, and it has been considered as the hepatic manifestation of the metabolic syndrome. Its complex pathophysiology involves a series of metabolic, inflammatory and oxidative stress processes, among others. Given the sharp increase in the prevalence of NAFLD and the lack of an appropriate pharmacological approach, it is crucial to consider the prevention/management of the disease based on lifestyle modifications such as the adoption of a healthy nutrition pattern. Herein, we review the literature and discuss the role of three key nutrients involved in pediatric NAFLD: fructose and its participation in metabolism, Omega-3 fatty acids and its anti-inflammatory effects and vitamin E and its action on oxidative stress.
Collapse
Affiliation(s)
- Gigliola Alberti
- Gastroenterology and Nutrition Department, Division of Pediatrics, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile; (G.A.); (J.C.G.)
| | - Juan Cristóbal Gana
- Gastroenterology and Nutrition Department, Division of Pediatrics, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile; (G.A.); (J.C.G.)
| | - José L. Santos
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile
- Correspondence: ; Tel.: +56-2-2354-3868
| |
Collapse
|
106
|
Ultraprocessed Food: Addictive, Toxic, and Ready for Regulation. Nutrients 2020; 12:nu12113401. [PMID: 33167515 PMCID: PMC7694501 DOI: 10.3390/nu12113401] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 10/19/2020] [Accepted: 10/23/2020] [Indexed: 12/15/2022] Open
Abstract
Past public health crises (e.g., tobacco, alcohol, opioids, cholera, human immunodeficiency virus (HIV), lead, pollution, venereal disease, even coronavirus (COVID-19) have been met with interventions targeted both at the individual and all of society. While the healthcare community is very aware that the global pandemic of non-communicable diseases (NCDs) has its origins in our Western ultraprocessed food diet, society has been slow to initiate any interventions other than public education, which has been ineffective, in part due to food industry interference. This article provides the rationale for such public health interventions, by compiling the evidence that added sugar, and by proxy the ultraprocessed food category, meets the four criteria set by the public health community as necessary and sufficient for regulation—abuse, toxicity, ubiquity, and externalities (How does your consumption affect me?). To their credit, some countries have recently heeded this science and have instituted sugar taxation policies to help ameliorate NCDs within their borders. This article also supplies scientific counters to food industry talking points, and sample intervention strategies, in order to guide both scientists and policy makers in instituting further appropriate public health measures to quell this pandemic.
Collapse
|
107
|
Mandala A, Janssen RC, Palle S, Short KR, Friedman JE. Pediatric Non-Alcoholic Fatty Liver Disease: Nutritional Origins and Potential Molecular Mechanisms. Nutrients 2020; 12:E3166. [PMID: 33081177 PMCID: PMC7602751 DOI: 10.3390/nu12103166] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the number one chronic liver disease worldwide and is estimated to affect nearly 40% of obese youth and up to 10% of the general pediatric population without any obvious signs or symptoms. Although the early stages of NAFLD are reversible with diet and lifestyle modifications, detecting such stages is hindered by a lack of non-invasive methods of risk assessment and diagnosis. This absence of non-invasive means of diagnosis is directly related to the scarcity of long-term prospective studies of pediatric NAFLD in children and adolescents. In the majority of pediatric NAFLD cases, the mechanisms driving the origin and rapid progression of NAFLD remain unknown. The progression from NAFLD to non-alcoholic steatohepatitis (NASH) in youth is associated with unique histological features and possible immune processes and metabolic pathways that may reflect different mechanisms compared with adults. Recent data suggest that circulating microRNAs (miRNAs) are important new biomarkers underlying pathways of liver injury. Several factors may contribute to pediatric NAFLD development, including high-sugar diets, in utero exposures via epigenetic alterations, changes in the neonatal microbiome, and altered immune system development and mitochondrial function. This review focuses on the unique aspects of pediatric NAFLD and how nutritional exposures impact the immune system, mitochondria, and liver/gastrointestinal metabolic health. These factors highlight the need for answers to how NAFLD develops in children and for early stage-specific interventions.
Collapse
Affiliation(s)
- Ashok Mandala
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (A.M.); (R.C.J.); (K.R.S.)
| | - Rachel C. Janssen
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (A.M.); (R.C.J.); (K.R.S.)
| | - Sirish Palle
- Department of Pediatrics, Section of Gastroenterology, Hepatology & Nutrition, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Kevin R. Short
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (A.M.); (R.C.J.); (K.R.S.)
- Department of Pediatrics, Section of Diabetes and Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (A.M.); (R.C.J.); (K.R.S.)
- Department of Pediatrics, Section of Diabetes and Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
108
|
Trandafir LM, Frasinariu OE, Leon-Constantin MM, Chiriac Ş, Trandafirescu MF, Miron IC, Luca AC, Iordache AC, Cojocaru E. Pediatric nonalcoholic fatty liver disease - a changing diagnostic paradigm. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY = REVUE ROUMAINE DE MORPHOLOGIE ET EMBRYOLOGIE 2020; 61:1023-1031. [PMID: 34171051 PMCID: PMC8343491 DOI: 10.47162/rjme.61.4.04] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 05/22/2021] [Indexed: 11/05/2022]
Abstract
Worldwide, nonalcoholic fatty liver disease (NAFLD) has emerged as the leading cause of chronic liver disease in children and adolescents, but also as a real public health issue. Over the last decades, the increase in the rates of obesity and overweight in children has led to the increase in the worldwide prevalence of pediatric NAFLD. Detection of a hyperechoic appearance of the liver at ultrasounds or elevated levels of transaminases, identified during a routine control in children, suggests NAFLD. The disorder can be diagnosed with either non-invasive strategies or through liver biopsy, which further allows the identification of specific histological aspects, distinct from those found in adults. Since NAFLD is a clinically heterogeneous disease, there is an imperative need to identify noninvasive biomarkers and screening techniques for early diagnosis in children, in order to prevent metabolic and cardiovascular complications later in adulthood. This review emphasizes the main diagnosis tools in pediatric NAFLD, a systemic disorder with multifactorial pathogenesis and varying clinical manifestations.
Collapse
Affiliation(s)
- Laura Mihaela Trandafir
- Department of Mother and Child Medicine – Pediatrics, Grigore T. Popa University of Medicine and Pharmacy, Iaşi, Romania
| | - Otilia Elena Frasinariu
- Department of Mother and Child Medicine – Pediatrics, Grigore T. Popa University of Medicine and Pharmacy, Iaşi, Romania
| | | | - Ştefan Chiriac
- First Medical Department, Grigore T. Popa University of Medicine and Pharmacy, Iaşi, Romania
| | | | - Ingrith Crenguţa Miron
- Department of Mother and Child Medicine – Pediatrics, Grigore T. Popa University of Medicine and Pharmacy, Iaşi, Romania
| | - Alina Costina Luca
- Department of Mother and Child Medicine – Pediatrics, Grigore T. Popa University of Medicine and Pharmacy, Iaşi, Romania
| | - Alin Constantin Iordache
- Second Surgery Department – Neurosurgery, Grigore T. Popa University of Medicine and Pharmacy, Iaşi, Romania
| | - Elena Cojocaru
- Department of Morphofunctional Sciences I – Pathology, Grigore T. Popa University of Medicine and Pharmacy, Iaşi, Romania
| |
Collapse
|
109
|
Castillo‐Leon E, Cioffi CE, Vos MB. Perspectives on youth-onset nonalcoholic fatty liver disease. Endocrinol Diabetes Metab 2020; 3:e00184. [PMID: 33102800 PMCID: PMC7576279 DOI: 10.1002/edm2.184] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/07/2020] [Accepted: 07/11/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The prevalence and incidence of youth-onset nonalcoholic fatty liver disease (NAFLD) far exceeds other paediatric chronic liver diseases and represents a considerable public health issue globally. METHODS Here, we performed a narrative review of current knowledge regarding the epidemiology of paediatric NAFLD, selected concepts in pathogenesis, comorbidities, diagnosis, and management, and issues related to the transition to adulthood. RESULTS Paediatric NAFLD has become increasingly more prevalent, especially in certain subgroups, such as children with obesity and certain races/ethnicities. The pathophysiology of paediatric NAFLD is complex and multifactorial, driven by an interaction of environmental and genetic factors. Once developed, NAFLD in childhood is associated with type 2 diabetes, hypertension, increased cardiovascular disease risk, and end-stage liver disease. This predicts an increased burden of morbidity and mortality in adolescents and young adults. Early screening and diagnosis are therefore crucial, and the development of noninvasive biomarkers remains an active area of investigation. Currently, treatment strategies are focused on lifestyle changes, but there is also research interest in pharmacological and surgical options. In the transition from paediatric to adult care, there are several potential challenges/barriers to treatment and research is needed to understand how best to support patients during this time. CONCLUSIONS Our understanding of the epidemiology and pathophysiology of paediatric NAFLD has increased considerably over recent decades, but several critical knowledge gaps remain and must be addressed in order to better mitigate the short-term and long-term risks of youth-onset NAFLD.
Collapse
Affiliation(s)
| | - Catherine E. Cioffi
- Department of PediatricsEmory University School of MedicineAtlantaGAUSA
- Nutrition & Health Sciences Doctoral ProgramLaney Graduate SchoolEmory UniversityAtlantaGAUSA
| | - Miriam B. Vos
- Department of PediatricsEmory University School of MedicineAtlantaGAUSA
- Children's Healthcare of AtlantaAtlantaGAUSA
| |
Collapse
|
110
|
Futatsugi K, Smith AC, Tu M, Raymer B, Ahn K, Coffey SB, Dowling MS, Fernando DP, Gutierrez JA, Huard K, Jasti J, Kalgutkar AS, Knafels JD, Pandit J, Parris KD, Perez S, Pfefferkorn JA, Price DA, Ryder T, Shavnya A, Stock IA, Tsai AS, Tesz GJ, Thuma BA, Weng Y, Wisniewska HM, Xing G, Zhou J, Magee TV. Discovery of PF-06835919: A Potent Inhibitor of Ketohexokinase (KHK) for the Treatment of Metabolic Disorders Driven by the Overconsumption of Fructose. J Med Chem 2020; 63:13546-13560. [PMID: 32910646 DOI: 10.1021/acs.jmedchem.0c00944] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Increased fructose consumption and its subsequent metabolism have been implicated in metabolic disorders such as nonalcoholic fatty liver disease and steatohepatitis (NAFLD/NASH) and insulin resistance. Ketohexokinase (KHK) converts fructose to fructose-1-phosphate (F1P) in the first step of the metabolic cascade. Herein we report the discovery of a first-in-class KHK inhibitor, PF-06835919 (8), currently in phase 2 clinical trials. The discovery of 8 was built upon our originally reported, fragment-derived lead 1 and the recognition of an alternative, rotated binding mode upon changing the ribose-pocket binding moiety from a pyrrolidinyl to an azetidinyl ring system. This new binding mode enabled efficient exploration of the vector directed at the Arg-108 residue, leading to the identification of highly potent 3-azabicyclo[3.1.0]hexane acetic acid-based KHK inhibitors by combined use of parallel medicinal chemistry and structure-based drug design.
Collapse
Affiliation(s)
- Kentaro Futatsugi
- Pfizer Inc. Medicine Design, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Aaron C Smith
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Meihua Tu
- Pfizer Inc. Medicine Design, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Brian Raymer
- Pfizer Inc. Medicine Design, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Kay Ahn
- Pfizer Inc. Internal Medicine Research Unit, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Steven B Coffey
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Matthew S Dowling
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Dilinie P Fernando
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jemy A Gutierrez
- Pfizer Inc. Internal Medicine Research Unit, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Kim Huard
- Pfizer Inc. Medicine Design, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Jayasankar Jasti
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Amit S Kalgutkar
- Pfizer Inc. Medicine Design, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - John D Knafels
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jayvardhan Pandit
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kevin D Parris
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Sylvie Perez
- Pfizer Inc. Internal Medicine Research Unit, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Jeffrey A Pfefferkorn
- Pfizer Inc. Internal Medicine Research Unit, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - David A Price
- Pfizer Inc. Medicine Design, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Tim Ryder
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Andre Shavnya
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Ingrid A Stock
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Andy S Tsai
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Gregory J Tesz
- Pfizer Inc. Internal Medicine Research Unit, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Benjamin A Thuma
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Yan Weng
- Pfizer Inc. Medicine Design, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Hanna M Wisniewska
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Gang Xing
- Pfizer Inc. Internal Medicine Research Unit, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Jun Zhou
- Pfizer Inc. Drug Safety R&D, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Thomas V Magee
- Pfizer Inc. Medicine Design, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
111
|
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
|
112
|
The physio-metabolic effects of time-restricting liquid sugar intake to six-hour windows during the mouse active phase: The effects of active phase liquid sugar consumption. Physiol Behav 2020; 223:112905. [PMID: 32446780 DOI: 10.1016/j.physbeh.2020.112905] [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: 10/16/2019] [Revised: 03/12/2020] [Accepted: 04/02/2020] [Indexed: 11/22/2022]
Abstract
Obesity is a major public health concern and overconsumption of unhealthy fats and sugary beverages are contributing factors. Time-restricted feeding can reduce obesity-associated pathophysiological parameters by limiting the time of food consumption; however, the effects of time-restricted sugary water consumption are unknown. To examine whether liquid calorie restriction impacts metabolic health, we measured metabolic parameters in mice provided liquid sugar at various intervals during the active phase. The control (Con) group received tap water, the adlibitum fructose-glucose (ALFG) group received ad libitumsugar water and the early fructose-glucose (EFG) and late fructose-glucose (LFG) groups received liquid sugar during the first and last six hours of the active period, respectively. Each group was given free access to chow. Zeitgeber time (ZT) notation was used to set all experimental time points to lights on as ZT 0. The ALFG group exhibited elevated body and adipose tissue weights compared to the other groups and increased hepatic steatosis compared to the Con group. The ALFG group consumed more calories than the other groups during ZT 6-11, indicating that this window may be critical in the promotion of weight gain from liquid sugar consumption. The EFG group exhibited higher levels of energy expenditure than the Con and LFG groups during the first half of the active period (ZT 12-17); however, there was no difference among the groups during the second half of the active period (ZT18-23). In contrast, the EFG group exhibited lower respiratory exchange ratio than other groups during the inactive period as well as the second half of the active period, indicating that the EFG group had greater metabolic flexibility and utilized lipids when carbohydrates from liquid sugar were not available. Additionally, the EFG group was more insulin tolerant than the ALFG and Con groups. Our results support the hypothesis that time-restricted liquid calorie restriction aids in reducing the detrimental metabolic effects of sugary drink consumption.
Collapse
|
113
|
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common liver disease in childhood. There is an increase in disease prevalence and diagnoses as it is difficult to diagnose the problem. There are currently no effective medications. Management of NAFLD is a challenge for primary care clinicians and subspecialists. This paper provides guidelines for disease screening, diagnosis, management, and algorithm for subspecialty referral.
Collapse
Affiliation(s)
- Krista McNeice
- Department of Pediatric Gastroenterology, Dayton Children's Hospital, Dayton, OH 45404, United States.
| | - Kelly Sandberg
- Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| |
Collapse
|
114
|
Cao X, Gu Y, Bian S, Zhang Q, Meng G, Liu L, Wu H, Zhang S, Wang Y, Zhang T, Wang X, Sun S, Wang X, Jia Q, Song K, Niu K. Association between eating speed and newly diagnosed nonalcoholic fatty liver disease among the general population. Nutr Res 2020; 80:78-88. [PMID: 32736293 DOI: 10.1016/j.nutres.2020.06.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/09/2020] [Accepted: 06/17/2020] [Indexed: 12/11/2022]
Abstract
Fast eating speed is a risk factor for obesity, which is also closely related to nonalcoholic fatty liver disease (NAFLD), suggesting that fast eating speed may contribute to the development of NAFLD. But the extent to which obesity may mediate the association between eating speed and NAFLD is uncertain. We hypothesized that obesity plays a mediating role in the association between eating speed and prevalence of NAFLD in the general population. A cross-sectional study (n = 23,611) was conducted in a general population sample from Tianjin, China. We measured anthropometrics and biochemical variables. The self-reported eating speed per meal was recorded and classified into 4 categories: slow, medium, relatively fast, and very fast. NAFLD was diagnosed by liver ultrasonography. Multiple logistic regression analysis was used to assess the associations between the eating speed and the prevalence of NAFLD, as well as the mediation effects of obesity on the association between eating speed and NAFLD. The prevalence of newly diagnosed NAFLD was 19.0%. After adjusting for potentially confounding factors, the odds ratios (95% confidence interval) of NAFLD across categories of eating speed were 1.00 (reference), 1.39 (1.18-1.64), 1.71 (1.45-2.01), and 2.04 (1.70-2.46). All these significant odds ratios were attenuated to be nonsignificant by adjustment for body mass index and/or waist circumference. This is the first study to demonstrate that eating speed is not independently associated with increased risk of NAFLD.
Collapse
Affiliation(s)
- Xingqi Cao
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Yeqing Gu
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Shanshan Bian
- The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Qing Zhang
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Ge Meng
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Li Liu
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongmei Wu
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Shunming Zhang
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Yawen Wang
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Tingjing Zhang
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xuena Wang
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Shaomei Sun
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Xing Wang
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Qiyu Jia
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Kun Song
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Kaijun Niu
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China; Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China.
| |
Collapse
|
115
|
Nakagawa T, Lanaspa MA, Millan IS, Fini M, Rivard CJ, Sanchez-Lozada LG, Andres-Hernando A, Tolan DR, Johnson RJ. Fructose contributes to the Warburg effect for cancer growth. Cancer Metab 2020; 8:16. [PMID: 32670573 PMCID: PMC7350662 DOI: 10.1186/s40170-020-00222-9] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/01/2020] [Indexed: 12/13/2022] Open
Abstract
Obesity and metabolic syndrome are strongly associated with cancer, and these disorders may share a common mechanism. Recently, fructose has emerged as a driving force to develop obesity and metabolic syndrome. Thus, we assume that fructose may be the mechanism to explain why obesity and metabolic syndrome are linked with cancer. Clinical and experimental evidence showed that fructose intake was associated with cancer growth and that fructose transporters are upregulated in various malignant tumors. Interestingly, fructose metabolism can be driven under low oxygen conditions, accelerates glucose utilization, and exhibits distinct effects as compared to glucose, including production of uric acid and lactate as major byproducts. Fructose promotes the Warburg effect to preferentially downregulate mitochondrial respiration and increases aerobic glycolysis that may aid metastases that initially have low oxygen supply. In the process, uric acid may facilitate carcinogenesis by inhibiting the TCA cycle, stimulating cell proliferation by mitochondrial ROS, and blocking fatty acid oxidation. Lactate may also contribute to cancer growth by suppressing fat oxidation and inducing oncogene expression. The ability of fructose metabolism to directly stimulate the glycolytic pathway may have been protective for animals living with limited access to oxygen, but may be deleterious toward stimulating cancer growth and metastasis for humans in modern society. Blocking fructose metabolism may be a novel approach for the prevention and treatment of cancer.
Collapse
Affiliation(s)
- Takahiko Nakagawa
- Department of Nephrology, Rakuwakai Otowa Hospital, 2 Otowa-Chinji-cho, Yamashina-ku, Kyoto, Japan
- Department of Stem Cell Biology & Regenerative Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Miguel A. Lanaspa
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO USA
| | - Inigo San Millan
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Aurora, USA
| | - Mehdi Fini
- University of Colorado Cancer Center, Aurora, CO USA
| | | | - Laura G. Sanchez-Lozada
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología Ignacio Chavez, 14080 Mexico City, CP Mexico
| | - Ana Andres-Hernando
- Division of Renal Diseases and Hypertension, University of Colorado Denver, 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
| |
Collapse
|
116
|
Abstract
This JCL Roundtable discussion probes the knowledge of 3 experts in pediatric lipidology, an emerging discipline both in the United States and internationally. In the 1990s, only 3 US institutions could be said to have dedicated pediatric lipid clinics; that number has grown to 25 today. The Pediatric Atherosclerosis Prevention and Lipidology Group of the National Lipid Association has regular teleconferences to support advocacy and convey best practices. Guidelines for pediatric lipidology initially focused on low-density lipoprotein cholesterol in 1992 as part of the National Cholesterol Education Program. Today the most comprehensive coverage comes from the 2011 National Heart Lung and Blood Institute Pediatric Guidelines. Universal screening was recommended for children between ages 9 and 11 years and teenagers/young adults between 17 to 21 years, a position echoed as "may be recommended" by the 2018 AHA/ACC/Multisociety Cholesterol Guidelines. While pediatric lipidologists continue to treat uncommon genetic disorders, they increasingly confront an issue of epidemic proportions-dyslipidemia as the initial presentation of metabolic dysregulation associated with obesity. Consequences of such altered metabolism extend to atherosclerosis, diabetes, liver disease, and other serious problems in adult life. Pediatric lipid science and practice differ from adult experience in several ways, including importance of family and birth history as well as genetics/epigenetics, lack of general pediatricians' familiarity with lipid drugs, value of family counseling, need for biomarkers of early metabolic dysregulation, and anticipation of endpoints in adult life not fully defined by randomized clinical trials in children.
Collapse
|
117
|
Deletion of Fructokinase in the Liver or in the Intestine Reveals Differential Effects on Sugar-Induced Metabolic Dysfunction. Cell Metab 2020; 32:117-127.e3. [PMID: 32502381 PMCID: PMC7347444 DOI: 10.1016/j.cmet.2020.05.012] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/22/2020] [Accepted: 05/12/2020] [Indexed: 12/18/2022]
Abstract
Intake of fructose-containing sugars is strongly associated with metabolic syndrome. Compared with other sugars, dietary fructose is uniquely metabolized by fructokinase. However, the tissue-specific role of fructokinase in sugar-induced metabolic syndrome, and the specific roles of glucose and fructose in driving it, is not fully understood. Here, we show that in mice receiving excess fructose-glucose solutions, whole-body deletion of fructokinase, and thus full blockade of fructose metabolism, is sufficient to prevent metabolic syndrome. This protection is not only due to reduced fructose metabolism, but also due to decreased sugar intake. Furthermore, by using tissue-specific fructokinase-deficient mice, we determined that while sugar intake is controlled by intestinal fructokinase activity, metabolic syndrome is driven by fructose metabolism in the liver. Our findings show a two-pronged role for fructose metabolism in sugar-induced metabolic syndrome, one arm via the intestine that mediates sugar intake and a second arm in the liver that drives metabolic dysfunction.
Collapse
|
118
|
Ayenigbara IO. The Accumulation of Visceral Fat and Preventive Measures among the Elderly. CARDIOVASCULAR INNOVATIONS AND APPLICATIONS 2020. [DOI: 10.15212/cvia.2019.0573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Visceral fat is a specific fat that is produced in the body, transformed into cholesterol, and circulated in the blood to other parts of the body. The circulated cholesterol usually in the form of low-density lipoproteins forms plaque on the walls of the arteries, thereby constricting
and blocking them and preventing the free flow of nutrients to various vital organs in the body. Visceral fat is deleterious to the health of elderly people because it is mostly found in the region of the abdomen that houses vital organs such as the pancreas, liver, and digestive tract, and
it further affects the normal functioning of hormones in the body. Globally, 15 million people die of a noncommunicable disease (NCD) annually between the age of 30 years and the age of 69 years, and there is an increase in NCD morbidity among people older than 70 years. Accumulated visceral
fat in elderly people could have deleterious health consequences, as it is a predisposing factor for many other NCDs and chronic health conditions. Sedentary lifestyle, unhealthy eating, stress, and inactivity are the major causes of excessive visceral fat. However, measures to prevent the
accumulation of visceral fat are straightforward, and impressive results are achieved with regular physical exercise, healthy diet choices, and proper stress management.
Collapse
|
119
|
Goldner D, Lavine JE. Nonalcoholic Fatty Liver Disease in Children: Unique Considerations and Challenges. Gastroenterology 2020; 158:1967-1983.e1. [PMID: 32201176 DOI: 10.1053/j.gastro.2020.01.048] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/30/2019] [Accepted: 01/05/2020] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is increasing in prevalence in concert with the global epidemic of obesity and is being diagnosed at increasingly younger ages. The unique histologic features and early presentation of disease in pediatrics suggest that children and adults may differ with regard to etiopathogenesis, with children displaying a greater vulnerability to genetic and environmental factors. Of significant relevance to pediatrics, in utero and perinatal stressors may alter the lifelong health trajectory of a child, increasing the risk of NAFLD and other cardiometabolic diseases. The development and progression of disease in childhood is likely to carry increased risk of long-term morbidity. Novel biomarkers and therapeutic agents are needed to avoid the otherwise inevitable health and societal consequences of this rapidly expanding pediatric population.
Collapse
Affiliation(s)
- Dana Goldner
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Columbia University Medical Center, New York, New York
| | - Joel E Lavine
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Columbia University Medical Center, New York, New York.
| |
Collapse
|
120
|
Fappi A, Mittendorfer B. Different physiological mechanisms underlie an adverse cardiovascular disease risk profile in men and women. Proc Nutr Soc 2020; 79:210-218. [PMID: 31340878 PMCID: PMC7583670 DOI: 10.1017/s0029665119001022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
CVD affect about one-third of the population and are the leading cause of mortality. The prevalence of CVD is closely linked to the prevalence of obesity because obesity is commonly associated with metabolic abnormalities that are important risk factors for CVD, including insulin resistance, pre-diabetes, and type-2 diabetes, atherosclerotic dyslipidaemia, endothelial dysfunction and hypertension. Women have a more beneficial traditional CVD risk profile (lower fasting plasma glucose, less atherogenic lipid profile) and a lower absolute risk for CVD than men. However, the relative risk for CVD associated with hyperglycaemia and dyslipidaemia is several-fold higher in women than in men. The reasons for the sex differences in CVD risk associated with metabolic abnormalities are unclear but could be related to differences in the mechanisms that cause hyperglycaemia and dyslipidaemia in men and women, which could influence the pathogenic processes involved in CVD. In the present paper, we review the influence of a person's sex on key aspects of metabolism involved in the cardiometabolic disease process, including insulin action on endogenous glucose production, tissue glucose disposal, and adipose tissue lipolysis, insulin secretion and insulin plasma clearance, postprandial glucose, fatty acid, and triglyceride kinetics, hepatic lipid metabolism and myocardial substrate use. We conclude that there are marked differences in many aspects of metabolism in men and women that are not all attributable to differences in the sex hormone milieu. The mechanisms responsible for these differences and the clinical implications of these observations are unclear and require further investigation.
Collapse
Affiliation(s)
- Alan Fappi
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
| | - Bettina Mittendorfer
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
121
|
Decreased Consumption of Added Fructose Reduces Waist Circumference and Blood Glucose Concentration in Patients with Overweight and Obesity. The DISFRUTE Study: A Randomised Trial in Primary Care. Nutrients 2020; 12:nu12041149. [PMID: 32325919 PMCID: PMC7231003 DOI: 10.3390/nu12041149] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/08/2020] [Accepted: 04/15/2020] [Indexed: 11/27/2022] Open
Abstract
The relationship between fructose intake and insulin resistance remains controversial. Our purpose was to determine whether a reduction in dietary fructose is effective in decreasing insulin resistance (HOMA2-IR). This field trial was conducted on 438 adults with overweight and obese status, without diabetes. A total of 121 patients in a low fructose diet (LFD) group and 118 in a standard diet (SD) group completed the 24-week study. Both diets were prescribed with 30–40% of energy intake restriction. There were no between-group differences in HOMA2-IR. However, larger decreases were seen in the LFD group in waist circumference (−7.0 vs. −4.8 = −2.2 cms, 95% CI: −3.7, −0.7) and fasting blood glucose −0.25 vs. −0.11 = −0.14 mmol/L, 95% CI: −0.028, −0.02). The percentage of reduction in calorie intake was similar. Only were differences observed in the % energy intake for some nutrients: total fructose (−2 vs. −0.6 = −1.4, 95% CI: −2.6, −0.3), MUFA (−1.7 vs. −0.4 = −1.3, 95% CI: −2.4, −0.2), protein (5.1 vs. 3.6 = 1.4, 95% CI: 0.1, 2.7). The decrease in fructose consumption originated mainly from the reduction in added fructose (−2.8 vs. −1.9 = −0.9, 95% CI: −1.6, −0.03). These results were corroborated after multivariate adjustments. The low fructose diet did not reduce insulin resistance. However, it reduced waist circumference and fasting blood glucose concentration, which suggests a decrease in hepatic insulin resistance.
Collapse
|
122
|
Ye G, Gao H, Huang Q, Lin Y, Liao X, Zhang H, Yang BC. Metabolomic Characterization of Metabolic Disturbances in the Extracellular Microenvironment of Oleate-Treated Macrophages Using Gas Chromatography–Mass Spectrometry. ANAL LETT 2020. [DOI: 10.1080/00032719.2020.1750623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Guozhu Ye
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, China
| | - Han Gao
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qiansheng Huang
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, China
| | - Yi Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, China
| | - Xu Liao
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, China
| | - Han Zhang
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, China
| | - Bi-cheng Yang
- Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| |
Collapse
|
123
|
Dewdney B, Roberts A, Qiao L, George J, Hebbard L. A Sweet Connection? Fructose's Role in Hepatocellular Carcinoma. Biomolecules 2020; 10:E496. [PMID: 32218179 PMCID: PMC7226025 DOI: 10.3390/biom10040496] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 03/20/2020] [Accepted: 03/23/2020] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma is one of few cancer types that continues to grow in incidence and mortality worldwide. With the alarming increase in diabetes and obesity rates, the higher rates of hepatocellular carcinoma are a result of underlying non-alcoholic fatty liver disease. Many have attributed disease progression to an excess consumption of fructose sugar. Fructose has known toxic effects on the liver, including increased fatty acid production, increased oxidative stress, and insulin resistance. These effects have been linked to non-alcoholic fatty liver (NAFLD) disease and a progression to non-alcoholic steatohepatitis (NASH). While the literature suggests fructose may enhance liver cancer progression, the precise mechanisms in which fructose induces tumor formation remains largely unclear. In this review, we summarize the current understanding of fructose metabolism in liver disease and liver tumor development. Furthermore, we consider the latest knowledge of cancer cell metabolism and speculate on additional mechanisms of fructose metabolism in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Brittany Dewdney
- Molecular and Cell Biology, and The Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville QLD 4811, Australia; (B.D.); (A.R.)
| | - Alexandra Roberts
- Molecular and Cell Biology, and The Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville QLD 4811, Australia; (B.D.); (A.R.)
| | - Liang Qiao
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney NSW 2145, Australia; (L.Q.); (J.G.)
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney NSW 2145, Australia; (L.Q.); (J.G.)
| | - Lionel Hebbard
- Molecular and Cell Biology, and The Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville QLD 4811, Australia; (B.D.); (A.R.)
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney NSW 2145, Australia; (L.Q.); (J.G.)
| |
Collapse
|
124
|
Abstract
The relationship of evolution with diet and environment can provide insights into modern disease. Fossil evidence shows apes, and early human ancestors were fruit eaters living in environments with strongly seasonal climates. Rapid cooling at the end of the Middle Miocene (15-12 Ma: millions of years ago) increased seasonality in Africa and Europe, and ape survival may be linked with a mutation in uric acid metabolism. Climate stabilized in the later Miocene and Pliocene (12-5 Ma), and fossil apes and early hominins were both adapted for life on ground and in trees. Around 2.5 Ma, early species of Homo introduced more animal products into their diet, and this coincided with developing bipedalism, stone tool technology and increase in brain size. Early species of Homo such as Homo habilis still lived in woodland habitats, and the major habitat shift in human evolution occurred at 1.8 Ma with the origin of Homo erectus. Homo erectus had increased body size, greater hunting skills, a diet rich in meat, control of fire and understanding about cooking food, and moved from woodland to savannah. Group size may also have increased at the same time, facilitating the transmission of knowledge from one generation to the next. The earliest fossils of Homo sapiens appeared about 300 kyr, but they had separated from Neanderthals by 480 kyr or earlier. Their diet shifted towards grain-based foods about 100 kyr ago, and settled agriculture developed about 10 kyr ago. This pattern remains for many populations to this day and provides important insights into current burden of lifestyle diseases.
Collapse
Affiliation(s)
- P Andrews
- From the, Natural History Museum, London University College, London, UK
| | - R J Johnson
- University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
125
|
Epel ES, Hartman A, Jacobs LM, Leung C, Cohn MA, Jensen L, Ishkanian L, Wojcicki J, Mason AE, Lustig RH, Stanhope KL, Schmidt LA. Association of a Workplace Sales Ban on Sugar-Sweetened Beverages With Employee Consumption of Sugar-Sweetened Beverages and Health. JAMA Intern Med 2020; 180:9-16. [PMID: 31657840 PMCID: PMC6820289 DOI: 10.1001/jamainternmed.2019.4434] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
IMPORTANCE Reductions in sugar-sweetened beverage (SSB) intake can improve health, but are difficult for individuals to achieve on their own. OBJECTIVES To evaluate whether a workplace SSB sales ban was associated with SSB intake and cardiometabolic health among employees and whether a brief motivational intervention provides added benefits to the sales ban. DESIGN, SETTING, AND PARTICIPANTS This before-after study and additional randomized trial conducted from July 28, 2015, to October 16, 2016, at a Northern California university and hospital assessed SSB intake, anthropometrics, and cardiometabolic biomarkers among 214 full-time English-speaking employees who were frequent SSB consumers (≥360 mL [≥12 fl oz] per day) before and 10 months after implementation of an SSB sales ban in a large workplace, with half the employees randomized to receive a brief motivational intervention targeting SSB reduction. INTERVENTIONS The employer stopped selling SSBs in all workplace venues, and half the sample was randomized to receive a brief motivational intervention and the other half was a control group that did not receive the intervention. This intervention was modeled on standard brief motivational interventions for alcohol used in the workplace that promote health knowledge and goal setting. MAIN OUTCOMES AND MEASURES Outcomes included changes in SSB intake, Homeostatic Model Assessment of Insulin Resistance (HOMA-IR), and measures of abdominal adiposity. The primary associations tested were the correlation between changes in SSB intake and changes in HOMA-IR. RESULTS Among the 214 study participants, 124 (57.9%) were women, with a mean (SD) age of 41.2 (11.0) years and a baseline mean (SD) body mass index of 29.4 (6.5). They reported a mean daily intake of 1050 mL (35 fl oz) of SSBs at baseline and 540 mL (18 fl oz) at follow-up-a 510-mL (17-fl oz) (48.6%) decrease (P < .001). Reductions in SSB intake correlated with improvements in HOMA-IR (r = 0.16; P = .03). Those not randomized to receive the brief intervention reduced their SSB intake by a mean (SD) of 246.0 (84.0) mL (8.2 [2.8] fl oz), while those also receiving the brief intervention reduced SSB intake by 762.0 (84.0) mL (25.4 [2.8] fl oz). From baseline to follow-up, there were significant reductions in mean (SE) waist circumference (2.1 [2.8] cm; P < .001). CONCLUSIONS AND RELEVANCE This study's findings suggest that the workplace sales ban was associated with a reduction in SSB intake and a significant reduction in waist circumference among employees within 10 months. The randomized clinical trial portion of this study found that targeting those at high risk with a brief motivational intervention led to additional improvements. Workplace sales bans may offer a promising new private-sector strategy for reducing the health harms of SSB intake. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT02585336.
Collapse
Affiliation(s)
- Elissa S Epel
- Department of Psychiatry, University of California, San Francisco.,Center for Health and Community, University of California, San Francisco
| | - Alison Hartman
- Center for Health and Community, University of California, San Francisco
| | - Laurie M Jacobs
- Philip R. Lee Institute for Health Policy Studies, University of California, San Francisco
| | - Cindy Leung
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor
| | - Michael A Cohn
- Osher Center for Integrative Medicine, University of California, San Francisco
| | - Leeane Jensen
- Campus Life Services, UCSF Wellness Program, University of California, San Francisco
| | - Laura Ishkanian
- Campus Life Services, UCSF Wellness Program, University of California, San Francisco
| | - Janet Wojcicki
- Center for Health and Community, University of California, San Francisco.,Department of Pediatrics, University of California, San Francisco
| | - Ashley E Mason
- Department of Psychiatry, University of California, San Francisco.,Osher Center for Integrative Medicine, University of California, San Francisco
| | - Robert H Lustig
- Philip R. Lee Institute for Health Policy Studies, University of California, San Francisco.,Department of Pediatrics, University of California, San Francisco
| | - Kimber L Stanhope
- Department of Molecular Biosciences, University of California, Davis
| | - Laura A Schmidt
- Philip R. Lee Institute for Health Policy Studies, University of California, San Francisco.,Department of Anthropology, History and Social Medicine, University of California, San Francisco
| |
Collapse
|
126
|
Neuschwander-Tetri BA. Too Much Sugar-The Not-So-Sweet Reality of Its Impact on Our Health. Hepatology 2020; 71:377-379. [PMID: 31446629 DOI: 10.1002/hep.30910] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
127
|
Borén J, Packard CJ, Taskinen MR. The Roles of ApoC-III on the Metabolism of Triglyceride-Rich Lipoproteins in Humans. Front Endocrinol (Lausanne) 2020; 11:474. [PMID: 32849270 PMCID: PMC7399058 DOI: 10.3389/fendo.2020.00474] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death globally. It is well-established based on evidence accrued during the last three decades that high plasma concentrations of cholesterol-rich atherogenic lipoproteins are causatively linked to CVD, and that lowering these reduces atherosclerotic cardiovascular events in humans (1-9). Historically, most attention has been on low-density lipoproteins (LDL) since these are the most abundant atherogenic lipoproteins in the circulation, and thus the main carrier of cholesterol into the artery wall. However, with the rise of obesity and insulin resistance in many populations, there is increasing interest in the role of triglyceride-rich lipoproteins (TRLs) and their metabolic remnants, with accumulating evidence showing they too are causatively linked to CVD. Plasma triglyceride, measured either in the fasting or non-fasting state, is a useful index of the abundance of TRLs and recent research into the biology and genetics of triglyceride heritability has provided new insight into the causal relationship of TRLs with CVD. Of the genetic factors known to influence plasma triglyceride levels variation in APOC3- the gene for apolipoprotein (apo) C-III - has emerged as being particularly important as a regulator of triglyceride transport and a novel therapeutic target to reduce dyslipidaemia and CVD risk (10).
Collapse
Affiliation(s)
- Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
- *Correspondence: Jan Borén
| | - Chris J. Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Marja-Riitta Taskinen
- Research Programs Unit, Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland
| |
Collapse
|
128
|
Jones GM, Caccavello R, Palii SP, Pullinger CR, Kane JP, Mulligan K, Gugliucci A, Schwarz JM. Separation of postprandial lipoproteins: improved purification of chylomicrons using an ApoB100 immunoaffinity method. J Lipid Res 2019; 61:455-463. [PMID: 31888979 DOI: 10.1194/jlr.d119000121] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 12/23/2019] [Indexed: 11/20/2022] Open
Abstract
Elevated levels of triglyceride-rich lipoproteins (TRLs), both fasting and postprandial, are associated with increased risk for atherosclerosis. However, guidelines for treatment are defined solely by fasting lipid levels, even though postprandial lipids may be more informative. In the postprandial state, circulating lipids consist of dietary fat transported from the intestine in chylomicrons (CMs; containing ApoB48) and fat transported from the liver in VLDL (containing ApoB100). Research into the roles of endogenous versus dietary fat has been hindered because of the difficulty in separating these particles by ultracentrifugation. CM fractions have considerable contamination from VLDL (purity, 10%). To separate CMs from VLDL, we produced polyclonal antibodies against ApoB100 and generated immunoaffinity columns. TRLs isolated by ultracentrifugation of plasma were applied to these columns, and highly purified CMs were collected (purity, 90-94%). Overall eight healthy unmedicated adult volunteers (BMI, 27.2 ± 1.4 kg/m2; fasting triacylglycerol, 102.6 ± 19.5 mg/dl) participated in a feeding study, which contained an oral stable-isotope tracer (1-13C acetate). We then used this technique on plasma samples freshly collected during an 8 h human feeding study from a subset of four subjects. We analyzed fractionated lipoproteins by Western blot, isolated and derivatized triacylglycerols, and calculated fractional de novo lipogenesis. The results demonstrated effective separation of postprandial lipoproteins and substantially improved purity compared with ultracentrifugation protocols, using the immunoaffinity method. This method can be used to better delineate the role of dietary sugar and fat on postprandial lipids in cardiovascular risk and explore the potential role of CM remnants in atherosclerosis.
Collapse
Affiliation(s)
- Grace Marie Jones
- Departments of Basic Science College of Osteopathic Medicine, Touro University California, Vallejo, CA
| | - Russell Caccavello
- Research, College of Osteopathic Medicine, Touro University California, Vallejo, CA
| | - Sergiu P Palii
- Research, College of Osteopathic Medicine, Touro University California, Vallejo, CA
| | - Clive R Pullinger
- Cardiovascular Research Institute University of California, San Francisco, San Francisco, CA.,Departments of Physiological Nursing University of California, San Francisco, San Francisco, CA
| | - John P Kane
- Cardiovascular Research Institute University of California, San Francisco, San Francisco, CA.,Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA
| | - Kathleen Mulligan
- Research, College of Osteopathic Medicine, Touro University California, Vallejo, CA.,Department of Medicine, Division of Endocrinology, University of California, San Francisco, San Francisco, CA and Zuckerberg San Francisco General Hospital, San Francisco, CA
| | - Alejandro Gugliucci
- Research, College of Osteopathic Medicine, Touro University California, Vallejo, CA
| | - Jean-Marc Schwarz
- Departments of Basic Science College of Osteopathic Medicine, Touro University California, Vallejo, CA .,Department of Medicine, Division of Endocrinology, University of California, San Francisco, San Francisco, CA and Zuckerberg San Francisco General Hospital, San Francisco, CA
| |
Collapse
|
129
|
Castro-Quezada I, Flores-Guillén E, Núñez-Ortega PE, Irecta-Nájera CA, Sánchez-Chino XM, Mendez-Flores OG, Olivo-Vidal ZE, García-Miranda R, Solís-Hernández R, Ochoa-Díaz-López H. Dietary Carbohydrates and Insulin Resistance in Adolescents from Marginalized Areas of Chiapas, México. Nutrients 2019; 11:E3066. [PMID: 31888175 PMCID: PMC6950049 DOI: 10.3390/nu11123066] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/05/2019] [Accepted: 12/12/2019] [Indexed: 12/28/2022] Open
Abstract
Evidence of the role that dietary carbohydrates (total carbohydrates, dietary fiber, total sugars, dietary glycemic index (GI) and glycemic load (GL)) exerts on insulin levels in adolescents is controversial. Thus, the aim of this study was to assess the association between dietary carbohydrates and insulin resistance in adolescents from Chiapas, México. A cross-sectional study was conducted in 217 adolescents. Sociodemographic, anthropometric, dietary and biochemical data were obtained. Total carbohydrates, dietary fiber, total sugars, dietary GI and GL were calculated from 24 h recalls. Two validated cut-off points for the homeostasis model assessment of insulin resistance (HOMA-IR) were used as surrogates of insulin resistance. Fasting insulin levels ≥ 14.38 μU/mL were considered as abnormal. Multivariate logistic regression models were fitted to assess the association between tertiles of dietary carbohydrates and insulin resistance or hyperinsulinemia. In our study, adolescents with the highest dietary fiber intake had lower odds of HOMA-IR > 2.97 (OR = 0.34; 95% CI: 0.13-0.93) when adjusted for sex, age, body fat percentage and saturated fatty acids intake. No significant associations were found for the rest of the carbohydrate variables. In summary, high-fiber diets reduce the probability of insulin resistance in adolescents from marginalized areas of Chiapas, México.
Collapse
Affiliation(s)
- Itandehui Castro-Quezada
- Health Department, El Colegio de la Frontera Sur, Carr. Panamericana y Periférico Sur s/n, Barrio de María Auxiliadora, San Cristóbal de las Casas, Chiapas 29290, Mexico
| | - Elena Flores-Guillén
- Health Department, El Colegio de la Frontera Sur, Carr. Panamericana y Periférico Sur s/n, Barrio de María Auxiliadora, San Cristóbal de las Casas, Chiapas 29290, Mexico
- Faculty of Nutrition and Food Science, University of Science and Arts of Chiapas, Libramiento Norte-Poniente 1150, Col. Lajas Maciel, Tuxtla Gutiérrez, Chiapas 29039, Mexico
| | - Pilar E. Núñez-Ortega
- Health Department, El Colegio de la Frontera Sur, Carr. Panamericana y Periférico Sur s/n, Barrio de María Auxiliadora, San Cristóbal de las Casas, Chiapas 29290, Mexico
| | - César A. Irecta-Nájera
- Health Department, El Colegio de la Frontera Sur, Carr. A Reforma Km. 15.5 s/n, RA. Guineo 2da. Sección, Villahermosa, Tabasco 86280, Mexico
| | - Xariss M. Sánchez-Chino
- Cátedra-CONACyT, Health Department, El Colegio de la Frontera Sur, Unidad Villahermosa, Carretera a Reforma Km. 15.5 s/n, RA. Guineo 2da. Sección, Villahermosa, Tabasco 86280, Mexico
| | - Orquidia G. Mendez-Flores
- Cátedra-CONACyT, Health Department, El Colegio de la Frontera Sur, Carr. Panamericana y Periférico Sur s/n, Barrio de María Auxiliadora, San Cristóbal de las Casas, Chiapas 29290, Mexico
| | - Zendy E. Olivo-Vidal
- Health Department, El Colegio de la Frontera Sur, Carr. A Reforma Km. 15.5 s/n, RA. Guineo 2da. Sección, Villahermosa, Tabasco 86280, Mexico
| | - Rosario García-Miranda
- Health Department, El Colegio de la Frontera Sur, Carr. Panamericana y Periférico Sur s/n, Barrio de María Auxiliadora, San Cristóbal de las Casas, Chiapas 29290, Mexico
| | - Roberto Solís-Hernández
- Health Department, El Colegio de la Frontera Sur, Carr. Panamericana y Periférico Sur s/n, Barrio de María Auxiliadora, San Cristóbal de las Casas, Chiapas 29290, Mexico
| | - Héctor Ochoa-Díaz-López
- Health Department, El Colegio de la Frontera Sur, Carr. Panamericana y Periférico Sur s/n, Barrio de María Auxiliadora, San Cristóbal de las Casas, Chiapas 29290, Mexico
| |
Collapse
|
130
|
Hallsworth K, Adams LA. Lifestyle modification in NAFLD/NASH: Facts and figures. JHEP Rep 2019; 1:468-479. [PMID: 32039399 PMCID: PMC7005657 DOI: 10.1016/j.jhepr.2019.10.008] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/16/2019] [Accepted: 10/25/2019] [Indexed: 12/16/2022] Open
Abstract
The development of non-alcoholic fatty liver disease is closely linked to lifestyle factors, namely excessive caloric intake coupled with reduced physical activity and exercise. This review aims to examine the evidence behind lifestyle change as a tool to improve hepatic steatosis and liver histology in patients with non-alcoholic fatty liver disease/non-alcoholic steatohepatitis. Furthermore, potential barriers to adopting lifestyle changes and strategies to overcome these barriers in the clinical setting are discussed.
Collapse
Affiliation(s)
- Kate Hallsworth
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Liver Unit, The Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle, Upon Tyne, UK
- Corresponding author. Address: 4th Floor William Leech Building, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK. Tel.: +44 191 208 8882; fax: +44 191 208 5685.
| | - Leon A. Adams
- Medical School, The University of Western Australia, Perth, WA, Australia
- Department of Hepatology, Sir Charles Gairdner Hospital, Perth, WA, Australia
| |
Collapse
|
131
|
Iruarrizaga-Lejarreta M, Arretxe E, Alonso C. Using metabolomics to develop precision medicine strategies to treat nonalcoholic steatohepatitis. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019. [DOI: 10.1080/23808993.2019.1685379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
| | - Enara Arretxe
- OWL Metabolomics, Parque Tecnológico de Bizkaia, Derio, Spain
| | - Cristina Alonso
- OWL Metabolomics, Parque Tecnológico de Bizkaia, Derio, Spain
| |
Collapse
|
132
|
El-Agroudy NN, Kurzbach A, Rodionov RN, O'Sullivan J, Roden M, Birkenfeld AL, Pesta DH. Are Lifestyle Therapies Effective for NAFLD Treatment? Trends Endocrinol Metab 2019; 30:701-709. [PMID: 31422872 DOI: 10.1016/j.tem.2019.07.013] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/10/2019] [Accepted: 07/12/2019] [Indexed: 12/17/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is becoming the most common liver disorder worldwide. Specifically, nonalcoholic steatohepatitis (NASH) and fibrosis pose an enormous burden for patients and health-care systems. In the absence of approved pharmacological therapies, effective lifestyle interventions for NAFLD, such as dietary strategies and exercise training, are currently the therapeutic strategies of choice. This review covers the influence of macronutrient quality and quantity (i.e., low-carbohydrate and high-protein diets), for successful reduction of intrahepatocellular lipids (IHL). Moreover, we discuss the effectiveness of different modalities of physical exercising with and without weight loss. These lifestyle modifications not only provide strategies to reduce IHL but may also hold a still underestimated potential to induce improvement and/or even remission of NAFLD.
Collapse
Affiliation(s)
- Nermeen N El-Agroudy
- Medizinische Klinik III, Universitätsklinikum Carl Gustav Carus, TU Dresden, Dresden, Germany; German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany
| | - Anica Kurzbach
- Medizinische Klinik III, Universitätsklinikum Carl Gustav Carus, TU Dresden, Dresden, Germany; German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany
| | - Roman N Rodionov
- Medizinische Klinik III, Universitätsklinikum Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - John O'Sullivan
- Medizinische Klinik III, Universitätsklinikum Carl Gustav Carus, TU Dresden, Dresden, Germany; Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Michael Roden
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich-Heine University Düsseldorf, Germany; Institute for Clinical Diabetology and Leibniz Center for Diabetes Research at Heinrich-Heine University Düsseldorf, German Diabetes Center, Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany
| | - Andreas L Birkenfeld
- Medizinische Klinik III, Universitätsklinikum Carl Gustav Carus, TU Dresden, Dresden, Germany; German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany; Section of Diabetes and Nutritional Sciences, Rayne Institute, Denmark Hill Campus, King's College London, London, UK; Paul Langerhans Institute Dresden, Helmholtz Zentrum München at the TU Dresden, Dresden, Germany.
| | - Dominik H Pesta
- Institute for Clinical Diabetology and Leibniz Center for Diabetes Research at Heinrich-Heine University Düsseldorf, German Diabetes Center, Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany
| |
Collapse
|
133
|
Prinz P. The role of dietary sugars in health: molecular composition or just calories? Eur J Clin Nutr 2019; 73:1216-1223. [PMID: 30787473 PMCID: PMC6760629 DOI: 10.1038/s41430-019-0407-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/30/2019] [Accepted: 02/01/2019] [Indexed: 12/20/2022]
Abstract
This review will focus on the question of whether dietary sugars are a relevant determinant in the global rise of overweight and obesity in adults, adolescents, and children. Initially, the review describes the current definitions for sugars in the diet and makes reference to them while analyzing their role in overweight and obesity as well as diet-related diseases, including type 2 diabetes, cardiovascular diseases, non-alcoholic fatty liver disease and cancer. Second, it will focus particularly on sucrose and the question of whether it is the molecular composition of sucrose (glucose and fructose) or its energy content that promotes body weight gain and diet-related diseases. Finally, the review will clarify the molecular characteristics of sucrose regarding the release of the gastrointestinal glucose-dependent insulinotropic peptide (GIP) compared to other energy-providing nutrients and its relevance in metabolic diseases. Current data indicates that dietary sugars are only associated with an increase in obesity when consumed as an excess source of calories and with that an increase in the risk of diet-related diseases. Furthermore, it was shown that a diet rich in fat will stimulate GIP secretion more than a diet rich in sucrose. Taken together, current scientific evidence does not support the conclusion that dietary sugars per se are detrimental to human health.
Collapse
Affiliation(s)
- Philip Prinz
- Department of Nutritional Sciences, German Sugar Association, Berlin, Germany.
| |
Collapse
|
134
|
Taskinen MR, Packard CJ, Borén J. Dietary Fructose and the Metabolic Syndrome. Nutrients 2019; 11:nu11091987. [PMID: 31443567 PMCID: PMC6770027 DOI: 10.3390/nu11091987] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/06/2019] [Accepted: 08/08/2019] [Indexed: 12/16/2022] Open
Abstract
Consumption of fructose, the sweetest of all naturally occurring carbohydrates, has increased dramatically in the last 40 years and is today commonly used commercially in soft drinks, juice, and baked goods. These products comprise a large proportion of the modern diet, in particular in children, adolescents, and young adults. A large body of evidence associate consumption of fructose and other sugar-sweetened beverages with insulin resistance, intrahepatic lipid accumulation, and hypertriglyceridemia. In the long term, these risk factors may contribute to the development of type 2 diabetes and cardiovascular diseases. Fructose is absorbed in the small intestine and metabolized in the liver where it stimulates fructolysis, glycolysis, lipogenesis, and glucose production. This may result in hypertriglyceridemia and fatty liver. Therefore, understanding the mechanisms underlying intestinal and hepatic fructose metabolism is important. Here we review recent evidence linking excessive fructose consumption to health risk markers and development of components of the Metabolic Syndrome.
Collapse
Affiliation(s)
- Marja-Riitta Taskinen
- Research Program for Clinical and Molecular Medicine Unit, Diabetes and Obesity, University of Helsinki, 00029 Helsinki, Finland
| | - Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, 41345 Gothenburg, Sweden.
| |
Collapse
|
135
|
Utari A, Maududi MS, Kusumawati NRD, Mexitalia M. Effects of low glycemic index diet on insulin resistance among obese adolescent with non-alcoholic fatty liver disease: a randomized controlled trial. MEDICAL JOURNAL OF INDONESIA 2019. [DOI: 10.13181/mji.v28i2.2496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
BACKGROUND Obesity is strongly correlated with insulin resistance (IR) and nonalcoholic fatty liver disease (NAFLD). Some studies suggest that dietary intake with low glycemic index (GI) may prevent IR and reduce the incidences of NAFLD. This study was aimed to determine the effects of low GI diet on IR among obese adolescents with NAFLD.
METHODS This study was a randomized controlled trial conducted in two JuniorHigh Schools in Semarang, Indonesia. The subjects were 12–14 years obese students with NAFLD, which divided into intervention and control groups according to schools using block random allocation. The intervention group received nutrition education and lunch diet (low energy, low GI, and low fat); meanwhile, the control group only received nutrition education for 12 weeks. The biochemical evaluation included fasting blood glucose (FBG) and insulin levels. IR was assessed using homeostatic model assessment-insulin resistance (HOMA-IR).
RESULTS Thirty-two subjects were enrolled in this study, 16 of which were assigned to the intervention group and the other to the control group. After 12 weeks, the energy and carbohydrate intake reduced in the intervention group (p < 0.05), FBG remained unchanged, and HOMA-IR increased (4.9 [3.7]–7.2 [3.5]) compared to the control group (6.4 [4.9]–5.5 [2.8]) (p < 0.05). Meanwhile, within the control group, there were no significant differences in the energy and carbohydrate intake as well as biochemical variables.
CONCLUSIONS Low GI modification diet alone may not reduce IR in the obeseadolescents with NAFLD.
Collapse
|
136
|
Varsamis P, Formosa MF, Larsen RN, Reddy-Luthmoodoo M, Jennings GL, Cohen ND, Grace M, Hawley JA, Devlin BL, Owen N, Dunstan DW, Dempsey PC, Kingwell BA. Between-meal sucrose-sweetened beverage consumption impairs glycaemia and lipid metabolism during prolonged sitting: A randomized controlled trial. Clin Nutr 2019; 38:1536-1543. [PMID: 30217471 DOI: 10.1016/j.clnu.2018.08.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/08/2018] [Accepted: 08/20/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Chronic overconsumption of sugar-sweetened beverages (SSBs) is associated with unfavourable health effects, including promotion of obesity. However, the acute effects of consuming SSBs on glucose and lipid metabolism remain to be characterized in a real-world, post-prandial context of prolonged sitting. We quantified the acute effects of between-meal SSB consumption compared with water, on glucose and lipid metabolism in habitual soft drink consumers during prolonged sitting. METHODS Twenty-eight overweight or obese young adults [15 males; 23 ± 3 (mean ± SD) years, body mass index (BMI) 31.0 ± 3.6 kg/m2) participated. During uninterrupted sitting and following standardized breakfast and lunch meals, each participant completed two 7-h conditions on separate days in a randomized, crossover design study. For each condition, participants consumed either a sucrose SSB or water mid-morning and mid-afternoon. Peak responses and total area under the curve (tAUC) over 7 h for blood glucose, insulin, C-peptide, triglyceride and non-esterified fatty acid (NEFA) concentrations were quantified and compared. RESULTS Compared to water, SSB consumption significantly increased the peak responses for blood glucose (20 ± 4% (mean ± SEM)), insulin (43 ± 15%) and C-peptide (21 ± 6%) concentrations. The tAUC for all these parameters was also increased by SSB consumption. The tAUC for triglycerides was 15 ± 5% lower after SSBs and this was driven by males (P < 0.05), as females showed no difference between conditions. The tAUC for NEFAs was 13 ± 5% lower after the SSB condition (P < 0.05). CONCLUSIONS Between-meal SSB consumption significantly elevated plasma glucose responses, associated with a sustained elevation in plasma insulin throughout a day of prolonged sitting. The SSB-induced reduction in circulating triglycerides and NEFAs indicates significant modulation of lipid metabolism, particularly in males. These metabolic effects may contribute to the development of metabolic disease when SSB consumption is habitual and co-occurring with prolonged sitting. Clinical Trial Registry number: ACTRN12616000840482, https://anzctr.org.au/Trial/Registration/TrialReview.aspx?ACTRN=12616000840482.
Collapse
Affiliation(s)
- Pia Varsamis
- Baker Heart & Diabetes Institute, Melbourne, Australia; Department of Physiology, Faculty of Medicine, Nursing & Health Sciences, Monash University, Melbourne, Australia.
| | | | | | | | - Garry L Jennings
- Baker Heart & Diabetes Institute, Melbourne, Australia; Sydney Medical School, University of Sydney, Sydney, Australia
| | - Neale D Cohen
- Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Megan Grace
- Baker Heart & Diabetes Institute, Melbourne, Australia; Department of Physiology, Faculty of Medicine, Nursing & Health Sciences, Monash University, Melbourne, Australia
| | - John A Hawley
- Exercise & Nutrition Research Programme, Mary MacKillop Institute of Health Research, Australian Catholic University, Melbourne, Victoria, Australia
| | - Brooke L Devlin
- Exercise & Nutrition Research Programme, Mary MacKillop Institute of Health Research, Australian Catholic University, Melbourne, Victoria, Australia
| | - Neville Owen
- Baker Heart & Diabetes Institute, Melbourne, Australia; Central Clinical School, Faculty of Medicine, Nursing & Health Sciences, Monash University, Melbourne, Australia; School of Sport Science, Exercise and Health, The University of Western Australia, Perth, Australia; Swinburne University of Technology, Melbourne, Australia; School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - David W Dunstan
- Baker Heart & Diabetes Institute, Melbourne, Australia; Central Clinical School, Faculty of Medicine, Nursing & Health Sciences, Monash University, Melbourne, Australia; Centre of Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University, Melbourne, Australia; Exercise & Nutrition Research Programme, Mary MacKillop Institute of Health Research, Australian Catholic University, Melbourne, Victoria, Australia; School of Public Health, The University of Queensland, Brisbane, Australia; School of Sport Science, Exercise and Health, The University of Western Australia, Perth, Australia
| | - Paddy C Dempsey
- Baker Heart & Diabetes Institute, Melbourne, Australia; Swinburne University of Technology, Melbourne, Australia
| | - Bronwyn A Kingwell
- Baker Heart & Diabetes Institute, Melbourne, Australia; Department of Physiology, Faculty of Medicine, Nursing & Health Sciences, Monash University, Melbourne, Australia; Central Clinical School, Faculty of Medicine, Nursing & Health Sciences, Monash University, Melbourne, Australia
| |
Collapse
|
137
|
Galderisi A, Giannini C, Van Name M, Caprio S. Fructose Consumption Contributes to Hyperinsulinemia in Adolescents With Obesity Through a GLP-1-Mediated Mechanism. J Clin Endocrinol Metab 2019; 104:3481-3490. [PMID: 30938760 PMCID: PMC6599430 DOI: 10.1210/jc.2019-00161] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 03/26/2019] [Indexed: 01/03/2023]
Abstract
CONTEXT The consumption of high-fructose beverages is associated with a higher risk for obesity and diabetes. Fructose can stimulate glucagon-like peptide 1 (GLP-1) secretion in lean adults, in the absence of any anorexic effect. OBJECTIVE We hypothesized that the ingestion of glucose and fructose may differentially stimulate GLP-1 and insulin response in lean adolescents and adolescents with obesity. DESIGN We studied 14 lean adolescents [four females; 15.9 ± 1.6 years of age; body mass index (BMI), 21.8 ± 2.2 kg/m2] and 23 adolescents with obesity (five females; 15.1 ± 1.6 years of age; BMI, 34.5 ± 4.6 kg/m2). Participants underwent a baseline oral glucose tolerance test to determine their glucose tolerance and estimate insulin sensitivity and β-cell function [oral disposition index (oDIcpep)]. Eligible subjects received, in a double-blind, crossover design, 75 g of glucose or fructose. Plasma was obtained every 10 minutes for 60 minutes for the measures of glucose, insulin, and GLP-1 (radioimmunoassay) and glucose-dependent insulinotropic polypeptide (GIP; ELISA). Incremental glucose and hormone levels were compared between lean individuals and those with obesity by a linear mixed model. The relationship between GLP-1 increment and oDIcpep was evaluated by regression analysis. RESULTS Following the fructose challenge, plasma glucose excursions were similar in both groups, yet the adolescents with obesity exhibited a greater insulin (P < 0.001) and GLP-1 (P < 0.001) increase than did their lean peers. Changes in GIP were similar in both groups. After glucose ingestion, the GLP-1 response (P < 0.001) was higher in the lean group. The GLP-1 increment during 60 minutes from fructose drink was correlated with a lower oDIcpep (r2 = 0.22, P = 0.009). CONCLUSION Fructose, but not glucose, ingestion elicits a higher GLP-1 and insulin response in adolescents with obesity than in lean adolescents. Fructose consumption may contribute to the hyperinsulinemic phenotype of adolescent obesity through a GLP-1-mediated mechanism.
Collapse
Affiliation(s)
- Alfonso Galderisi
- Department of Pediatrics, Pediatrics Endocrinology and Diabetes Section, Yale School of Medicine, New Haven, Connecticut
- Department of Woman’s and Child’s Health, University of Padova, Padova, Italy
| | - Cosimo Giannini
- Department of Pediatrics, Pediatrics Endocrinology and Diabetes Section, Yale School of Medicine, New Haven, Connecticut
| | - Michelle Van Name
- Department of Pediatrics, Pediatrics Endocrinology and Diabetes Section, Yale School of Medicine, New Haven, Connecticut
| | - Sonia Caprio
- Department of Pediatrics, Pediatrics Endocrinology and Diabetes Section, Yale School of Medicine, New Haven, Connecticut
- Correspondence and Reprint Requests: Sonia Caprio, MD, Division of Pediatric Endocrinology, Department of Pediatrics, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520. E-mail:
| |
Collapse
|
138
|
Thomas DD, Corkey BE, Istfan NW, Apovian CM. Hyperinsulinemia: An Early Indicator of Metabolic Dysfunction. J Endocr Soc 2019; 3:1727-1747. [PMID: 31528832 PMCID: PMC6735759 DOI: 10.1210/js.2019-00065] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 07/18/2019] [Indexed: 02/06/2023] Open
Abstract
Hyperinsulinemia is strongly associated with type 2 diabetes. Racial and ethnic minority populations are disproportionately affected by diabetes and obesity-related complications. This mini-review provides an overview of the genetic and environmental factors associated with hyperinsulinemia with a focus on racial and ethnic differences and its metabolic consequences. The data used in this narrative review were collected through research in PubMed and reference review of relevant retrieved articles. Insulin secretion and clearance are regulated processes that influence the development and progression of hyperinsulinemia. Environmental, genetic, and dietary factors are associated with hyperinsulinemia. Certain pharmacotherapies for obesity and bariatric surgery are effective at mitigating hyperinsulinemia and are associated with improved metabolic health. Hyperinsulinemia is associated with many environmental and genetic factors that interact with a wide network of hormones. Recent studies have advanced our understanding of the factors affecting insulin secretion and clearance. Further basic and translational work on hyperinsulinemia may allow for earlier and more personalized treatments for obesity and metabolic diseases.
Collapse
Affiliation(s)
- Dylan D Thomas
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts
| | - Barbara E Corkey
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts
| | - Nawfal W Istfan
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts
| | - Caroline M Apovian
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts
| |
Collapse
|
139
|
Erkin-Cakmak A, Bains Y, Caccavello R, Noworolski SM, Schwarz JM, Mulligan K, Lustig RH, Gugliucci A. Isocaloric Fructose Restriction Reduces Serum d-Lactate Concentration in Children With Obesity and Metabolic Syndrome. J Clin Endocrinol Metab 2019; 104:3003-3011. [PMID: 30869790 PMCID: PMC6553815 DOI: 10.1210/jc.2018-02772] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 03/08/2019] [Indexed: 01/06/2023]
Abstract
OBJECTIVE To investigate the link between dietary sugar consumption and two separate pathogenetic mechanisms associated with metabolic syndrome: de novo lipogenesis (DNL) and nonenzymatic glycation. DESIGN AND PARTICIPANTS We assessed changes in serum d-lactate (the detoxification end-product of methylglyoxal) concentration in response to 9 days of isocaloric fructose restriction in 20 children with obesity and metabolic syndrome, and examined correlations with changes in DNL, liver fat, insulin sensitivity, and other metrics of hepatic metabolism. INTERVENTIONS Nine days of dietary sugar restriction, with substitution of equal amounts of refined starch. MAIN OUTCOME MEASURES On days 0 and 10, children had laboratory evaluation of d-lactate levels and other analytes, and underwent oral glucose tolerance testing, magnetic resonance spectroscopy to quantify fat depots, and 13C-acetate incorporation into triglyceride (TG) to measure DNL. RESULTS d-Lactate was associated with baseline liver fat fraction (P < 0.001) and visceral adipose tissue (P < 0.001) but not with subcutaneous adipose tissue. At baseline, d-lactate was positively correlated with DNL-area under the curve (AUC) (P = 0.003), liver fat fraction (P = 0.02), TG (P = 0.004), and TG/high-density lipoprotein ratio (P = 0.002). After 9 days of isocaloric fructose restriction, serum d-lactate levels reduced by 50% (P < 0.0001), and changes in d-lactate correlated with both changes in DNL-AUC and measures of insulin sensitivity. CONCLUSION Baseline correlation of d-lactate with DNL and measures of insulin sensitivity and reduction in d-lactate after 9 days of isocaloric fructose restriction suggest that DNL and nonenzymatic glycation are functionally linked via intermediary glycolysis in the pathogenesis of metabolic syndrome and point to fructose as a key dietary substrate that drives both pathways.
Collapse
Affiliation(s)
- Ayca Erkin-Cakmak
- Division of Pediatric Endocrinology, Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - Yasmin Bains
- Glycation, Oxidation and Disease Laboratory, Department of Research, Touro University California College of Osteopathic Medicine, Vallejo, California
| | - Russell Caccavello
- Glycation, Oxidation and Disease Laboratory, Department of Research, Touro University California College of Osteopathic Medicine, Vallejo, California
| | - Susan M Noworolski
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Jean-Marc Schwarz
- Division of Endocrinology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Kathleen Mulligan
- Division of Endocrinology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Robert H Lustig
- Division of Pediatric Endocrinology, Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - Alejandro Gugliucci
- Glycation, Oxidation and Disease Laboratory, Department of Research, Touro University California College of Osteopathic Medicine, Vallejo, California
- Correspondence and Reprint Requests: Alejandro Gugliucci, MD, PhD, Glycation, Oxidation, and Disease Laboratory, Department of Research, Touro University California College of Osteopathic Medicine, 1310 Club Drive, Vallejo, California 94592. E-mail:
| |
Collapse
|
140
|
Golonka R, Yeoh BS, Vijay-Kumar M. Dietary Additives and Supplements Revisited: The Fewer, the Safer for Liver and Gut Health. ACTA ACUST UNITED AC 2019; 5:303-316. [PMID: 32864300 DOI: 10.1007/s40495-019-00187-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Purpose of Review The supplementation of dietary additives into processed foods has exponentially increased in the past few decades. Similarly, the incidence rates of various diseases, including metabolic syndrome, gut dysbiosis and hepatocarcinogenesis, have been elevating. Current research reveals that there is a positive association between food additives and these pathophysiological diseases. This review highlights the research published within the past 5 years that elucidate and update the effects of dietary supplements on liver and intestinal health. Recent Findings Some of the key findings include: enterocyte dysfunction of fructose clearance causes non-alcoholic fatty liver disease (NAFLD); non-caloric sweeteners are hepatotoxic; dietary emulsifiers instigate gut dysbiosis and hepatocarcinogenesis; and certain prebiotics can induce cholestatic hepatocellular carcinoma (HCC) in gut dysbiotic mice. Overall, multiple reports suggest that the administration of purified, dietary supplements could cause functional damage to both the liver and gut. Summary The extraction of bioactive components from natural resources was considered a brilliant method to modulate human health. However, current research highlights that such purified components may negatively affect individuals with microbiotal dysbiosis, resulting in a deeper break of the symbiotic relationship between the host and gut microbiota, which can lead to repercussions on gut and liver health. Therefore, ingestion of these dietary additives should not go without some caution!
Collapse
Affiliation(s)
- Rachel Golonka
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Beng San Yeoh
- Graduate Program in Immunology & Infectious Disease, Pennsylvania State University, University Park, PA 16802, USA
| | - Matam Vijay-Kumar
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA.,Department of Medical Microbiology & Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| |
Collapse
|
141
|
Tappy L, Rosset R. Health outcomes of a high fructose intake: the importance of physical activity. J Physiol 2019; 597:3561-3571. [PMID: 31116420 PMCID: PMC6851848 DOI: 10.1113/jp278246] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 05/08/2019] [Indexed: 12/27/2022] Open
Abstract
Fructose metabolism is generally held to occur essentially in cells of the small bowel, the liver, and the kidneys expressing fructolytic enzymes (fructokinase, aldolase B and a triokinase). In these cells, fructose uptake and fructolysis are unregulated processes, resulting in the generation of intracellular triose phosphates proportionate to fructose intake. Triose phosphates are then processed into lactate, glucose and fatty acids to serve as metabolic substrates in other cells of the body. With small oral loads, fructose is mainly metabolized in the small bowel, while with larger loads fructose reaches the portal circulation and is largely extracted by the liver. A small portion, however, escapes liver extraction and is metabolized either in the kidneys or in other tissues through yet unspecified pathways. In sedentary subjects, consumption of a fructose-rich diet for several days stimulates hepatic de novo lipogenesis, increases intrahepatic fat and blood triglyceride concentrations, and impairs insulin effects on hepatic glucose production. All these effects can be prevented when high fructose intake is associated with increased levels of physical activity. There is also evidence that, during exercise, fructose carbons are efficiently transferred to skeletal muscle as glucose and lactate to be used for energy production. Glucose and lactate formed from fructose can also contribute to the re-synthesis of muscle glycogen after exercise. We therefore propose that the deleterious health effects of fructose are tightly related to an imbalance between fructose energy intake on one hand, and whole-body energy output related to a low physical activity on the other hand.
Collapse
Affiliation(s)
- Luc Tappy
- Department of Physiology, University of Lausanne, Lausanne, Switzerland.,Cardiometabolic Center, Broye Hospital, Estavayer-le-lac, Switzerland
| | - Robin Rosset
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
142
|
Hernández-Díazcouder A, Romero-Nava R, Carbó R, Sánchez-Lozada LG, Sánchez-Muñoz F. High Fructose Intake and Adipogenesis. Int J Mol Sci 2019; 20:E2787. [PMID: 31181590 PMCID: PMC6600229 DOI: 10.3390/ijms20112787] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 02/06/2023] Open
Abstract
In modern societies, high fructose intake from sugar-sweetened beverages has contributed to obesity development. In the diet, sucrose and high fructose corn syrup are the main sources of fructose and can be metabolized in the intestine and transported into the systemic circulation. The liver can metabolize around 70% of fructose intake, while the remaining is metabolized by other tissues. Several tissues including adipose tissue express the main fructose transporter GLUT5. In vivo, chronic fructose intake promotes white adipose tissue accumulation through activating adipogenesis. In vitro experiments have also demonstrated that fructose alone induces adipogenesis by several mechanisms, including (1) triglycerides and very-low-density lipoprotein (VLDL) production by fructose metabolism, (2) the stimulation of glucocorticoid activation by increasing 11β-HSD1 activity, and (3) the promotion of reactive oxygen species (ROS) production through uric acid, NOX and XOR expression, mTORC1 signaling and Ang II induction. Moreover, it has been observed that fructose induces adipogenesis through increased ACE2 expression, which promotes high Ang-(1-7) levels, and through the inhibition of the thermogenic program by regulating Sirt1 and UCP1. Finally, microRNAs may also be involved in regulating adipogenesis in high fructose intake conditions. In this paper, we propose further directions for research in fructose participation in adipogenesis.
Collapse
Affiliation(s)
- Adrián Hernández-Díazcouder
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico city 14080, Mexico.
- Departamento de Ciencias de la Salud, Área de Investigación Médica, Universidad Autónoma Metropolitana Iztapalapa, Mexico city 09340, Mexico.
| | - Rodrigo Romero-Nava
- Departamento de Ciencias de la Salud, Área de Investigación Médica, Universidad Autónoma Metropolitana Iztapalapa, Mexico city 09340, Mexico.
- Laboratorio de investigación en Farmacología, Hospital Infantil de México Federico Gómez, Mexico city 06720, Mexico.
- Sección de Postgraduados, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico city 11340, Mexico.
| | - Roxana Carbó
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico city 14080, Mexico.
| | - L Gabriela Sánchez-Lozada
- Laboratorio de Fisiopatología Renal, Departamento de Nefrología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico city 14080, Mexico.
| | - Fausto Sánchez-Muñoz
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico city 14080, Mexico.
- Sección de Postgraduados, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico city 11340, Mexico.
| |
Collapse
|
143
|
Zelber-Sagi S. Minding the Gap Between Clinical Trials and Treatment With the Mediterranean Dietary Pattern for Patients With Nonalcoholic Fatty Liver Disease. Clin Gastroenterol Hepatol 2019; 17:1248-1250. [PMID: 30648604 DOI: 10.1016/j.cgh.2019.01.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 12/22/2018] [Accepted: 01/06/2019] [Indexed: 02/07/2023]
Affiliation(s)
- Shira Zelber-Sagi
- School of Public Health, University of Haifa, Haifa, Israel; Liver Unit, Department of Gastroenterology, Tel Aviv Medical Center, Tel-Aviv, Israel
| |
Collapse
|
144
|
Sundborn G, Thornley S, Merriman TR, Lang B, King C, Lanaspa MA, Johnson RJ. Are Liquid Sugars Different from Solid Sugar in Their Ability to Cause Metabolic Syndrome? Obesity (Silver Spring) 2019; 27:879-887. [PMID: 31054268 DOI: 10.1002/oby.22472] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 01/29/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Intake of sugary drinks, especially soft drinks, carries increased risk for obesity and diabetes. This article reviews whether sugary drinks carry different risks for metabolic syndrome compared with foods that contain natural or added sugars. METHODS A narrative review was performed to evaluate differences between liquid and solid sugars in their ability to induce metabolic syndrome and to discuss potential mechanisms to account for the differences. RESULTS Epidemiological studies support liquid added sugars, such as soft drinks, as carrying greater risk for development of metabolic syndrome compared with solid sugar. Some studies suggest that fruit juice may also confer relatively higher risk for weight gain and insulin resistance compared with natural fruits. Experimental evidence suggests this may be due to differences in how fructose is metabolized. Fructose induces metabolic disease by reducing the energy levels in liver cells, mediated by the concentration of fructose to which the cells are exposed. The concentration relates to the quantity and speed at which fructose is ingested, absorbed, and metabolized. CONCLUSIONS Although reduced intake of added sugars (sucrose and high-fructose corn syrup) remains a general recommendation, there is evidence that sugary soft drinks may provide greater health risks relative to sugar-containing foods.
Collapse
Affiliation(s)
- Gerhard Sundborn
- Department of Pacific Health, The University of Auckland, Auckland, New Zealand
| | - Simon Thornley
- Auckland Regional Public Health Service, Auckland, New Zealand
| | - Tony R Merriman
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Bodo Lang
- Department of Marketing, Business School, The University of Auckland, Auckland, New Zealand
| | - Christopher King
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Miguel A Lanaspa
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
145
|
|
146
|
Abstract
Purpose of Review Apolipoprotein C-III (apoC-III) is known to inhibit lipoprotein lipase (LPL) and function as an important regulator of triglyceride metabolism. In addition, apoC-III has also more recently been identified as an important risk factor for cardiovascular disease. This review summarizes the mechanisms by which apoC-III induces hypertriglyceridemia and promotes atherogenesis, as well as the findings from recent clinical trials using novel strategies for lowering apoC-III. Recent Findings Genetic studies have identified subjects with heterozygote loss-of-function (LOF) mutations in APOC3, the gene coding for apoC-III. Clinical characterization of these individuals shows that the LOF variants associate with a low-risk lipoprotein profile, in particular reduced plasma triglycerides. Recent results also show that complete deficiency of apoC-III is not a lethal mutation and is associated with very rapid lipolysis of plasma triglyceride-rich lipoproteins (TRL). Ongoing trials based on emerging gene-silencing technologies show that intervention markedly lowers apoC-III levels and, consequently, plasma triglyceride. Unexpectedly, the evidence points to apoC-III not only inhibiting LPL activity but also suppressing removal of TRLs by LPL-independent pathways. Summary Available data clearly show that apoC-III is an important cardiovascular risk factor and that lifelong deficiency of apoC-III is cardioprotective. Novel therapies have been developed, and results from recent clinical trials indicate that effective reduction of plasma triglycerides by inhibition of apoC-III might be a promising strategy in management of severe hypertriglyceridemia and, more generally, a novel approach to CHD prevention in those with elevated plasma triglyceride.
Collapse
|
147
|
Castro MC, Villagarcía HG, Massa ML, Francini F. Alpha-lipoic acid and its protective role in fructose induced endocrine-metabolic disturbances. Food Funct 2019; 10:16-25. [PMID: 30575838 DOI: 10.1039/c8fo01856a] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In recent decades a worldwide increase has been reported in the consumption of unhealthy high calorie diets associated with marked changes in meal nutrient composition, such as a higher intake of refined carbohydrates, which leads to the speculatation that changes in food habits have contributed to the current epidemic of obesity and type 2 diabetes. Among these refined carbohydrates, fructose has been deeply investigated and murine models of high fructose diet have emerged as useful tools to study dietary-induced insulin resistance, impaired glucose tolerance, dyslipidemia and alterations in glucose metabolism. Since oxidative stress has been demonstrated to play a key pathogenic role in the alterations described above, several lines of research have focused on the possible preventive effects of antioxidant/redox state regulation therapy, among which alpha-lipoic acid has been extensively investigated. The following references discussed support the fact that co-administration of alpha-lipoic acid normalized the changes generated by fructose rich diets, thereby making this compound a good therapeutic tool, also administered as a food supplement, to prevent endocrine-metabolic disturbances triggered by high fructose associated with obesity and type 2 diabetes at an early stage of development (prediabetes).
Collapse
Affiliation(s)
- María Cecilia Castro
- CENEXA (Centro de Endocrinología Experimental y Aplicada, UNLP-CONICET La Plata-FCM) (Centro asociado CICPBA), 1900 La Plata, Argentina.
| | | | | | | |
Collapse
|
148
|
Complementary foods in baby food pouches: position statement from the Nutrition Commission of the German Society for Pediatrics and Adolescent Medicine (DGKJ, e.V.). Mol Cell Pediatr 2019; 6:2. [PMID: 30840172 PMCID: PMC6403273 DOI: 10.1186/s40348-019-0089-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 12/18/2022] Open
Abstract
Pureed complementary feeding products packed in squeezable plastic pouches, usually with a spout and a screw cap, have been increasingly marketed. The Committee on Nutrition recommends that infants and young children should not suck pureed or liquid complementary foods from baby food pouches. Complementary foods should be offered with a spoon or should be fed as finger foods. Infants and young children should be given the opportunity to get to know a variety of foods and food textures including pieces of foods, supported by responsive feeding between the child and their parents or caregivers. Complementary foods marketed in baby food pouches often have a high energy density and are predominantly extremely high in sugar content, with up to almost 90% of the total energy content. Regular consumption bears the risks of imbalanced nutrient provision and increased risks for dental caries and overweight. Complementary foods for infants and young children should have a balanced composition following the recommendations of the German Society of Pediatrics and Adolescent Medicine (DGKJ) and should contain only limited amounts of sugar. We discourage the feeding of pureed complementary foods from baby food pouches.
Collapse
|
149
|
Prinz P. The role of dietary sugars in health: molecular composition or just calories? Eur J Clin Nutr 2019. [PMID: 30787473 DOI: 10.1038/s41430-019-0407-z.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
This review will focus on the question of whether dietary sugars are a relevant determinant in the global rise of overweight and obesity in adults, adolescents, and children. Initially, the review describes the current definitions for sugars in the diet and makes reference to them while analyzing their role in overweight and obesity as well as diet-related diseases, including type 2 diabetes, cardiovascular diseases, non-alcoholic fatty liver disease and cancer. Second, it will focus particularly on sucrose and the question of whether it is the molecular composition of sucrose (glucose and fructose) or its energy content that promotes body weight gain and diet-related diseases. Finally, the review will clarify the molecular characteristics of sucrose regarding the release of the gastrointestinal glucose-dependent insulinotropic peptide (GIP) compared to other energy-providing nutrients and its relevance in metabolic diseases. Current data indicates that dietary sugars are only associated with an increase in obesity when consumed as an excess source of calories and with that an increase in the risk of diet-related diseases. Furthermore, it was shown that a diet rich in fat will stimulate GIP secretion more than a diet rich in sucrose. Taken together, current scientific evidence does not support the conclusion that dietary sugars per se are detrimental to human health.
Collapse
Affiliation(s)
- Philip Prinz
- Department of Nutritional Sciences, German Sugar Association, Berlin, Germany.
| |
Collapse
|
150
|
Sanchez-Lozada LG, Andres-Hernando A, Garcia-Arroyo FE, Cicerchi C, Li N, Kuwabara M, Roncal-Jimenez CA, Johnson RJ, Lanaspa MA. Uric acid activates aldose reductase and the polyol pathway for endogenous fructose and fat production causing development of fatty liver in rats. J Biol Chem 2019; 294:4272-4281. [PMID: 30651350 DOI: 10.1074/jbc.ra118.006158] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 01/10/2019] [Indexed: 12/18/2022] Open
Abstract
Dietary, fructose-containing sugars have been strongly associated with the development of nonalcoholic fatty liver disease (NAFLD). Recent studies suggest that fructose also can be produced via the polyol pathway in the liver, where it may induce hepatic fat accumulation. Moreover, fructose metabolism yields uric acid, which is highly associated with NAFLD. Here, using biochemical assays, reporter gene expression, and confocal fluorescence microscopy, we investigated whether uric acid regulates aldose reductase, a key enzyme in the polyol pathway. We evaluated whether soluble uric acid regulates aldose reductase expression both in cultured hepatocytes (HepG2 cells) and in the liver of hyperuricemic rats and whether this stimulation is associated with endogenous fructose production and fat accumulation. Uric acid dose-dependently stimulated aldose reductase expression in the HepG2 cells, and this stimulation was associated with endogenous fructose production and triglyceride accumulation. This stimulatory mechanism was mediated by uric acid-induced oxidative stress and stimulation of the transcription factor nuclear factor of activated T cells 5 (NFAT5). Uric acid also amplified the effects of elevated glucose levels to stimulate hepatocyte triglyceride accumulation. Hyperuricemic rats exhibited elevated hepatic aldose reductase expression, endogenous fructose accumulation, and fat buildup that was significantly reduced by co-administration of the xanthine oxidase inhibitor allopurinol. These results suggest that uric acid generated during fructose metabolism may act as a positive feedback mechanism that stimulates endogenous fructose production by stimulating aldose reductase in the polyol pathway. Our findings suggest an amplifying mechanism whereby soft drinks rich in glucose and fructose can induce NAFLD.
Collapse
Affiliation(s)
- Laura G Sanchez-Lozada
- From the Laboratory of Renal Physiopathology, Instituto Nacional de Cardiología Ignacio Chavez, CP 14080 Mexico City, Mexico and
| | - Ana Andres-Hernando
- the Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado, Aurora, Colorado 80045
| | - Fernando E Garcia-Arroyo
- From the Laboratory of Renal Physiopathology, Instituto Nacional de Cardiología Ignacio Chavez, CP 14080 Mexico City, Mexico and
| | - Christina Cicerchi
- the Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado, Aurora, Colorado 80045
| | - Nanxing Li
- the Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado, Aurora, Colorado 80045
| | - Masanari Kuwabara
- the Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado, Aurora, Colorado 80045
| | - Carlos A Roncal-Jimenez
- the Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado, Aurora, Colorado 80045
| | - Richard J Johnson
- the Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado, Aurora, Colorado 80045
| | - Miguel A Lanaspa
- the Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado, Aurora, Colorado 80045
| |
Collapse
|