1
|
Flores Monar GV, Sanchez Cruz C, Calderon Martinez E. Mindful Eating: A Deep Insight Into Fructose Metabolism and Its Effects on Appetite Regulation and Brain Function. J Nutr Metab 2025; 2025:5571686. [PMID: 40297675 PMCID: PMC12037248 DOI: 10.1155/jnme/5571686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Fructose, a common sweetener in modern diets, has profound effects on both metabolism and brain function, primarily due to its distinct metabolic pathways. Unlike glucose, fructose bypasses critical regulatory steps in metabolism, particularly the phosphofructokinase-1 (PFK-1) feedback inhibition, leading to uncontrolled metabolism and increased fat storage. This review delves into the metabolic consequences of fructose consumption, including its limited role in directly stimulating insulin secretion, which affects satiety signaling and contributes to increased food intake. The small intestine initially helps metabolize ingested fructose, shielding the liver and brain from excessive exposure. However, when consumed in excess, particularly in diets high in processed foods, this protective mechanism becomes overwhelmed, contributing to metabolic disorders such as insulin resistance, obesity, and fatty liver disease. The review also explores fructose's impact on the brain, with a focus on the hippocampus, a key region for memory and learning. Chronic high fructose intake has been linked to mitochondrial dysfunction, increased production of reactive oxygen species (ROS), and neuroinflammation, all of which contribute to cognitive decline and impairments in memory and learning. Additionally, fructose-induced alterations in insulin signaling in the brain are associated with increased risk for neurodegenerative diseases. These findings underscore the potential long-term neurological consequences of excessive fructose intake and highlight the need for further human studies to assess the full spectrum of its effects on brain health. Addressing the rising consumption of fructose, particularly in processed foods, is essential for developing targeted strategies to mitigate its adverse metabolic and cognitive outcomes.
Collapse
Affiliation(s)
| | - Camila Sanchez Cruz
- Faculty of Medicine, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México, Mexico
| | | |
Collapse
|
2
|
Al‐Ibraheem AMT, Hameed AAZ, Marsool MDM, Jain H, Prajjwal P, Khazmi I, Nazzal RS, AL‐Najati HMH, Al‐Zuhairi BHYK, Razzaq M, Abd ZB, Marsool ADM, wahedaldin AI, Amir O. Exercise-Induced cytokines, diet, and inflammation and their role in adipose tissue metabolism. Health Sci Rep 2024; 7:e70034. [PMID: 39221051 PMCID: PMC11365580 DOI: 10.1002/hsr2.70034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 04/23/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Obesity poses a significant global health challenge, necessitating effective prevention and treatment strategies. Exercise and diet are recognized as pivotal interventions in combating obesity. This study reviews the literature concerning the impact of exercise-induced cytokines, dietary factors, and inflammation on adipose tissue metabolism, shedding light on potential pathways for therapeutic intervention. METHODOLOGY A comprehensive review of relevant literature was conducted to elucidate the role of exercise-induced cytokines, including interleukin-6 (IL-6), interleukin-15 (IL-15), brain-derived neurotrophic factor (BDNF), irisin, myostatin, fibroblast growth factor 21 (FGF21), follistatin (FST), and angiopoietin-like 4 (ANGPTL4), in adipose tissue metabolism. Various databases were systematically searched using predefined search terms to identify relevant studies. Articles selected for inclusion underwent thorough analysis to extract pertinent data on the mechanisms underlying the influence of these cytokines on adipose tissue metabolism. RESULTS AND DISCUSSION Exercise-induced cytokines exert profound effects on adipose tissue metabolism, influencing energy expenditure (EE), thermogenesis, fat loss, and adipogenesis. For instance, IL-6 activates AMP-activated protein kinase (AMPK), promoting fatty acid oxidation and reducing lipogenesis. IL-15 upregulates peroxisome proliferator-activated receptor delta (PPARδ), stimulating fatty acid catabolism and suppressing lipogenesis. BDNF enhances AMPK-dependent fat oxidation, while irisin induces the browning of white adipose tissue (WAT), augmenting thermogenesis. Moreover, myostatin, FGF21, FST, and ANGPTL4 each play distinct roles in modulating adipose tissue metabolism, impacting factors such as fatty acid oxidation, adipogenesis, and lipid uptake. The elucidation of these pathways offers valuable insights into the complex interplay between exercise, cytokines, and adipose tissue metabolism, thereby informing the development of targeted obesity management strategies. CONCLUSION Understanding the mechanisms by which exercise-induced cytokines regulate adipose tissue metabolism is critical for devising effective obesity prevention and treatment modalities. Harnessing the therapeutic potential of exercise-induced cytokines, in conjunction with dietary interventions, holds promise for mitigating the global burden of obesity. Further research is warranted to delineate the precise mechanisms underlying the interactions between exercise, cytokines, and adipose tissue metabolism.
Collapse
Affiliation(s)
| | | | | | - Hritvik Jain
- All India Institute of Medical SciencesJodhpurIndia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Purnell JQ, le Roux CW. Hypothalamic control of body fat mass by food intake: The key to understanding why obesity should be treated as a disease. Diabetes Obes Metab 2024; 26 Suppl 2:3-12. [PMID: 38351898 DOI: 10.1111/dom.15478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/06/2024] [Accepted: 01/18/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Hypothalamic centres have been recognized to play a central role in body weight regulation for nearly 70 years. AIMS In this review, we will explore the current undersanding of the role the hypothalamus plays in controlling food intake behaviours. MATERIALS AND METHODS Review of relevant literature from PubMed searches and review article citations. RESULTS Beginning with autopsy studies showing destructive hypothalamic lesions in patients manifesting hyperphagia and rapid weight gain, followed by animal lesioning studies pinpointing adjacent hypothalamic sites as the 'satiety' centre and the 'feeding' centre of the brain, the neurocircuitry that governs our body weight is now understood to consist of a complex, interconnected network, including the hypothalamus and extending to cortical sites, reward centres and brainstem. Neurons in these sites receive afferent signals from the gastrointestinal tract and adipose tissue indicating food availability, calorie content, as well as body fat mass. DISCUSSION Integration of these complex signals leads to modulation of the two prime effector systems that defend a body fat mass set point: food intake and energy expenditure. CONCLUSION Understanding the hypothalamic control of food intake forms the foundation for understanding and managing obesity as a chronic disease.
Collapse
Affiliation(s)
- Jonathan Q Purnell
- Department of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Carel W le Roux
- School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
4
|
Endy EJ, Yi SY, Steffen BT, Shikany JM, Jacobs DR, Goins RK, Steffen LM. Added sugar intake is associated with weight gain and risk of developing obesity over 30 years: The CARDIA study. Nutr Metab Cardiovasc Dis 2024; 34:466-474. [PMID: 38195258 PMCID: PMC11253751 DOI: 10.1016/j.numecd.2023.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 10/20/2023] [Accepted: 10/20/2023] [Indexed: 01/11/2024]
Abstract
BACKGROUND AND AIMS Numerous prospective studies have examined sugar sweetened beverage (SSB) intake associated with weight gain or incident obesity. Because SSB accounts for only 33 % of added sugar (AS) intake, we investigated the associations of AS intake with change in weight and waist circumference and risk of developing obesity. METHODS AND RESULTS At baseline (1985-86) Black and White women and men, aged 18-30 years, enrolled in the Coronary Artery Risk Development in Young Adults (CARDIA) study and were followed for 30 years (2015-16). A diet history assessed dietary intake 3 times over 20 years. Multivariable linear regression evaluated the associations of change in weight (n = 3306) and waist circumference (n = 3296) across quartiles of AS, adjusting for demographics, lifestyle factors, and anthropometrics. Proportional hazards regression analysis evaluated the associations of time-varying cumulative AS intake with risk of incident obesity (n = 4023) and abdominal obesity (n = 3449), adjusting for the same factors. Over 30 years of follow-up, greater AS intake was associated with gaining 2.3 kg more weight (ptrend = 0.01) and 2.2 cm greater change in waist circumference (ptrend = 0.005) as well as increased risk of incident obesity (HR 1.28; 95 % CI: 1.08-1.53) and incident abdominal obesity (HR 1.27; 95 % CI:1.02-1.60). CONCLUSION Our findings are consistent with recommendations from the 2020-2025 U S. Dietary Guidelines for Americans to limit daily AS intake.
Collapse
Affiliation(s)
- Emily J Endy
- University of Minnesota College of Liberal Arts, Minneapolis, MN, USA
| | - So-Yun Yi
- University of Minnesota School of Public Health, Division of Epidemiology and Community Health, Minneapolis, MN, USA
| | - Brian T Steffen
- University of Minnesota School of Medicine, Department of Surgery, Minneapolis, MN, USA
| | - James M Shikany
- University of Alabama at Birmingham School of Medicine, Division of Preventive Medicine, Birmingham, AL, USA
| | - David R Jacobs
- University of Minnesota School of Public Health, Division of Epidemiology and Community Health, Minneapolis, MN, USA
| | - Rae K Goins
- University of Minnesota School of Public Health, Division of Epidemiology and Community Health, Minneapolis, MN, USA
| | - Lyn M Steffen
- University of Minnesota School of Public Health, Division of Epidemiology and Community Health, Minneapolis, MN, USA.
| |
Collapse
|
5
|
Yang W, Jiang W, Guo S. Regulation of Macronutrients in Insulin Resistance and Glucose Homeostasis during Type 2 Diabetes Mellitus. Nutrients 2023; 15:4671. [PMID: 37960324 PMCID: PMC10647592 DOI: 10.3390/nu15214671] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
Insulin resistance is an important feature of metabolic syndrome and a precursor of type 2 diabetes mellitus (T2DM). Overnutrition-induced obesity is a major risk factor for the development of insulin resistance and T2DM. The intake of macronutrients plays a key role in maintaining energy balance. The components of macronutrients distinctly regulate insulin sensitivity and glucose homeostasis. Precisely adjusting the beneficial food compound intake is important for the prevention of insulin resistance and T2DM. Here, we reviewed the effects of different components of macronutrients on insulin sensitivity and their underlying mechanisms, including fructose, dietary fiber, saturated and unsaturated fatty acids, and amino acids. Understanding the diet-gene interaction will help us to better uncover the molecular mechanisms of T2DM and promote the application of precision nutrition in practice by integrating multi-omics analysis.
Collapse
Affiliation(s)
| | | | - Shaodong Guo
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX 77843, USA; (W.Y.); (W.J.)
| |
Collapse
|
6
|
Johnson RJ, Lanaspa MA, Sanchez-Lozada LG, Tolan D, Nakagawa T, Ishimoto T, Andres-Hernando A, Rodriguez-Iturbe B, Stenvinkel P. The fructose survival hypothesis for obesity. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220230. [PMID: 37482773 PMCID: PMC10363705 DOI: 10.1098/rstb.2022.0230] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 05/04/2023] [Indexed: 07/25/2023] Open
Abstract
The fructose survival hypothesis proposes that obesity and metabolic disorders may have developed from over-stimulation of an evolutionary-based biologic response (survival switch) that aims to protect animals in advance of crisis. The response is characterized by hunger, thirst, foraging, weight gain, fat accumulation, insulin resistance, systemic inflammation and increased blood pressure. The process is initiated by the ingestion of fructose or by stimulating endogenous fructose production via the polyol pathway. Unlike other nutrients, fructose reduces the active energy (adenosine triphosphate) in the cell, while blocking its regeneration from fat stores. This is mediated by intracellular uric acid, mitochondrial oxidative stress, the inhibition of AMP kinase and stimulation of vasopressin. Mitochondrial oxidative phosphorylation is suppressed, and glycolysis stimulated. While this response is aimed to be modest and short-lived, the response in humans is exaggerated due to gain of 'thrifty genes' coupled with a western diet rich in foods that contain or generate fructose. We propose excessive fructose metabolism not only explains obesity but the epidemics of diabetes, hypertension, non-alcoholic fatty liver disease, obesity-associated cancers, vascular and Alzheimer's dementia, and even ageing. Moreover, the hypothesis unites current hypotheses on obesity. Reducing activation and/or blocking this pathway and stimulating mitochondrial regeneration may benefit health-span. This article is part of a discussion meeting issue 'Causes of obesity: theories, conjectures and evidence (Part I)'.
Collapse
Affiliation(s)
- Richard J. Johnson
- Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, CO 80016, USA
| | - Miguel A. Lanaspa
- Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, CO 80016, USA
| | - L. Gabriela Sanchez-Lozada
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología ‘Ignacio Chavez’, Mexico City 14080, Mexico
| | - Dean Tolan
- Biology Department, Boston University, Boston, MA 02215, USA
| | - Takahiko Nakagawa
- Department of Nephrology, Rakuwakai-Otowa Hospital, Kyoto 607-8062, Japan
| | - Takuji Ishimoto
- Department of Nephrology and Rheumatology, Aichi Medical University, Aichi 480-1103, Japan
| | - Ana Andres-Hernando
- Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, CO 80016, USA
| | - Bernardo Rodriguez-Iturbe
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición ‘Salvador Zubirán’, Mexico City 14080, Mexico
| | - Peter Stenvinkel
- Department of Renal Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
| |
Collapse
|
7
|
Cargnin-Carvalho A, da Silva MR, Costa AB, Engel NA, Farias BX, Bressan JB, Backes KM, de Souza F, da Rosa N, de Oliveira Junior AN, Goldim MPDS, Correa MEAB, Venturini LM, Fortunato JJ, Prophiro JS, Petronilho F, Silveira PCL, Ferreira GK, Rezin GT. High concentrations of fructose cause brain damage in mice. Biochem Cell Biol 2023; 101:313-325. [PMID: 36947832 DOI: 10.1139/bcb-2022-0088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023] Open
Abstract
Excessive fructose consumption is associated with the incidence of obesity and systemic inflammation, resulting in increased oxidative damage and failure to the function of brain structures. Thus, we hypothesized that fructose consumption will significantly increase inflammation, oxidative damage, and mitochondrial dysfunction in the mouse brain and, consequently, memory damage. The effects of different fructose concentrations on inflammatory and biochemical parameters in the mouse brain were evaluated. Male Swiss mice were randomized into four groups: control, with exclusive water intake, 5%, 10%, and 20% fructose group. The 10% and 20% fructose groups showed an increase in epididymal fat, in addition to higher food consumption. Inflammatory markers were increased in epididymal fat and in some brain structures. In the evaluation of oxidative damage, it was possible to observe significant increases in the hypothalamus, prefrontal cortex, and hippocampus. In the epididymal fat and in the prefrontal cortex, there was a decrease in the activity of the mitochondrial respiratory chain complexes and an increase in the striatum. Furthermore, short memory was impaired in the 10% and 20% groups but not long memory. In conclusion, excess fructose consumption can cause fat accumulation, inflammation, oxidative damage, and mitochondrial dysfunction, which can damage brain structures and consequently memory.
Collapse
Affiliation(s)
- Anderson Cargnin-Carvalho
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Mariella Reinol da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Ana Beatriz Costa
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Nicole Alessandra Engel
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Bianca Xavier Farias
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Joice Benedet Bressan
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Kassiane Mathiola Backes
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Francielly de Souza
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Naiana da Rosa
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Aloir Neri de Oliveira Junior
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Mariana Pereira de Souza Goldim
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | | | - Ligia Milanez Venturini
- Laboratory of Experimental Phisiopatology, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Santa Catarina, Brazil
| | - Jucélia Jeremias Fortunato
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Josiane Somariva Prophiro
- Immunoparasitology Research Group, Postgraduate Program in Health Sciences, Universidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Fabrícia Petronilho
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Paulo Cesar Lock Silveira
- Laboratory of Experimental Phisiopatology, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Santa Catarina, Brazil
| | | | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| |
Collapse
|
8
|
Johnson RJ, Lee SMK, Sánchez-Lozada LG, Kanbay M, Bansal A, Tolan DR, Bjornstad P, Lanaspa MA, Maesaka J. Fructose: A New Variable to Consider in SIADH and the Hyponatremia Associated With Long-Distance Running? Am J Kidney Dis 2023; 82:105-112. [PMID: 36940740 PMCID: PMC10330032 DOI: 10.1053/j.ajkd.2023.01.443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 01/01/2023] [Indexed: 03/23/2023]
Abstract
Fructose has recently been proposed to stimulate vasopressin secretion in humans. Fructose-induced vasopressin secretion is not only postulated to result from ingestion of fructose-containing drinks but may also occur from endogenous fructose production via activation of the polyol pathway. This raises the question of whether fructose might be involved in some cases of vasopressin-induced hyponatremia, especially in situations where the cause is not fully known such as in the syndrome of inappropriate secretion of diuretic hormone (SIADH) and exercise-associated hyponatremia, which has been observed in marathon runners. Here we discuss the new science of fructose and vasopressin, and how it may play a role in some of these conditions, as well as in the complications associated with rapid treatment (such as the osmotic demyelination syndrome). Studies to test the role of fructose could provide new pathophysiologic insights as well as novel potential treatment strategies for these common conditions.
Collapse
Affiliation(s)
- Richard J Johnson
- Division of Renal Diseases and Hypertension, Anschutz Medical Campus, University of Colorado, Aurora, Colorado.
| | | | | | - Mehmet Kanbay
- Division of Nephrology, Department of Medicine, School of Medicine, Koc University, Istanbul, Turkey
| | - Anip Bansal
- Division of Renal Diseases and Hypertension, Anschutz Medical Campus, University of Colorado, Aurora, Colorado
| | - Dean R Tolan
- Biology Department, Boston University, Boston Massachusetts
| | - Petter Bjornstad
- Division of Renal Diseases and Hypertension, Anschutz Medical Campus, University of Colorado, Aurora, Colorado; Section of Endocrinology, Department of Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, Colorado
| | - Miguel A Lanaspa
- Division of Renal Diseases and Hypertension, Anschutz Medical Campus, University of Colorado, Aurora, Colorado
| | - John Maesaka
- Department of Medicine and Division of Nephrology and Hypertension, NYU Langone Hospitals, Mineola, New York
| |
Collapse
|
9
|
Li RJW, Chiu JFM, Bruce K, Zhang SY, Barros DR, Yue JTY, Lam TKT. A glucose-sensing mechanism with glucose transporter-1 and pyruvate kinase in the area postrema regulates hepatic glucose production in rats. J Biol Chem 2023; 299:104633. [PMID: 36963496 PMCID: PMC10149203 DOI: 10.1016/j.jbc.2023.104633] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/26/2023] Open
Abstract
The area postrema (AP) of the brain is exposed to circulating metabolites and hormones. However, whether AP detects glucose changes to exert biological responses remains unknown. Its neighboring nuclei, the nucleus tractus solitarius (NTS), responds to acute glucose infusion by inhibiting hepatic glucose production, but the mechanism also remains elusive. Herein, we characterized AP and NTS glucose-sensing mechanisms. Infusion of glucose into the AP, like the NTS, of chow rats suppressed glucose production during the pancreatic (basal insulin)-euglycemic clamps. Glucose transporter-1 or pyruvate kinase lentiviral-mediated knockdown in the AP negated AP glucose infusion to lower glucose production, while the glucoregulatory effect of NTS glucose infusion was also negated by knocking down glucose transporter-1 or pyruvate kinase in the NTS. Furthermore, we determined that high-fat (HF) feeding disrupts glucose infusion to lower glucose production in association with a modest reduction in expression of glucose transporter-1, but not pyruvate kinase, in the AP and NTS. However, pyruvate dehydrogenase activator dichloroacetate infusion into the AP or NTS that enhanced downstream pyruvate metabolism and recapitulated the glucoregulatory effect of glucose in chow rats still failed to lower glucose production in HF rats. We discovered that a glucose transporter-1 and pyruvate kinase-dependent glucose-sensing mechanism in the AP (as well as the NTS) lowers glucose production in chow rats, and that HF disrupts the glucose-sensing mechanism that is downstream of pyruvate metabolism in the AP and NTS. These findings highlight the role of AP and NTS in mediating glucose to regulate hepatic glucose production.
Collapse
Affiliation(s)
- Rosa J W Li
- Department of Physiology, University of Toronto, Toronto, Canada; Toronto General Hospital Research Institute, UHN, Toronto, Canada
| | - Jennifer F M Chiu
- Toronto General Hospital Research Institute, UHN, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Kyla Bruce
- Toronto General Hospital Research Institute, UHN, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Song-Yang Zhang
- Toronto General Hospital Research Institute, UHN, Toronto, Canada
| | - Daniel R Barros
- Department of Physiology, University of Toronto, Toronto, Canada; Toronto General Hospital Research Institute, UHN, Toronto, Canada
| | - Jessica T Y Yue
- Department of Physiology, University of Alberta, Edmonton, Canada
| | - Tony K T Lam
- Department of Physiology, University of Toronto, Toronto, Canada; Toronto General Hospital Research Institute, UHN, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada; Banting and Best Diabetes Centre, University of Toronto, Toronto, Canada.
| |
Collapse
|
10
|
Singh R, Gupta V, Kumar A, Singh K. 2-Deoxy-D-Glucose: A Novel Pharmacological Agent for Killing Hypoxic Tumor Cells, Oxygen Dependence-Lowering in Covid-19, and Other Pharmacological Activities. Adv Pharmacol Pharm Sci 2023; 2023:9993386. [PMID: 36911357 PMCID: PMC9998157 DOI: 10.1155/2023/9993386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/02/2023] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
The nonmetabolizable glucose analog 2-deoxy-D-glucose (2-DG) has shown promising pharmacological activities, including inhibition of cancerous cell growth and N-glycosylation. It has been used as a glycolysis inhibitor and as a potential energy restriction mimetic agent, inhibiting pathogen-associated molecular patterns. Radioisotope derivatives of 2-DG have applications as tracers. Recently, 2-DG has been used as an anti-COVID-19 drug to lower the need for supplemental oxygen. In the present review, various pharmaceutical properties of 2-DG are discussed.
Collapse
Affiliation(s)
- Raman Singh
- Division Chemistry & Toxicology, WTL-Clean and Renewable Energy Pvt. Ltd., New Delhi, India
| | - Vidushi Gupta
- Department of Chemistry, Indian Institute of Science Education and Research, Mohali, Punjab, India
| | - Antresh Kumar
- Department of Biochemistry, Central University of Haryana, Jant-Pali, Mahendergarh, Haryana 123031, India
| | - Kuldeep Singh
- Department of Applied Chemistry, Amity University Madhya Pradesh, Gwalior, MP 474005, India
| |
Collapse
|
11
|
Johnson RJ, Tolan DR, Bredesen D, Nagel M, Sánchez-Lozada LG, Fini M, Burtis S, Lanaspa MA, Perlmutter D. Could Alzheimer's disease be a maladaptation of an evolutionary survival pathway mediated by intracerebral fructose and uric acid metabolism? Am J Clin Nutr 2023; 117:455-466. [PMID: 36774227 PMCID: PMC10196606 DOI: 10.1016/j.ajcnut.2023.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/21/2022] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
An important aspect of survival is to assure enough food, water, and oxygen. Here, we describe a recently discovered response that favors survival in times of scarcity, and it is initiated by either ingestion or production of fructose. Unlike glucose, which is a source for immediate energy needs, fructose metabolism results in an orchestrated response to encourage food and water intake, reduce resting metabolism, stimulate fat and glycogen accumulation, and induce insulin resistance as a means to reduce metabolism and preserve glucose supply for the brain. How this survival mechanism affects brain metabolism, which in a resting human amounts to 20% of the overall energy demand, is only beginning to be understood. Here, we review and extend a previous hypothesis that this survival mechanism has a major role in the development of Alzheimer's disease and may account for many of the early features, including cerebral glucose hypometabolism, mitochondrial dysfunction, and neuroinflammation. We propose that the pathway can be engaged in multiple ways, including diets high in sugar, high glycemic carbohydrates, and salt. In summary, we propose that Alzheimer's disease may be the consequence of a maladaptation to an evolutionary-based survival pathway and what had served to enhance survival acutely becomes injurious when engaged for extensive periods. Although more studies are needed on the role of fructose metabolism and its metabolite, uric acid, in Alzheimer's disease, we suggest that both dietary and pharmacologic trials to reduce fructose exposure or block fructose metabolism should be performed to determine whether there is potential benefit in the prevention, management, or treatment of this disease.
Collapse
Affiliation(s)
- Richard J Johnson
- Department of Medicine, Rocky Mountain VA Medical Center, Aurora, CO, USA; Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, CO, USA.
| | - Dean R Tolan
- Biology Department, Boston University, Boston, MA, USA
| | - Dale Bredesen
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Maria Nagel
- Department of Neurology, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - Laura G Sánchez-Lozada
- Department of Cardio-Renal Physiopathology, National Institute of Cardiology Ignacio Chávez, Mexico City, Mexico
| | - Mehdi Fini
- Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | | | - Miguel A Lanaspa
- Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | | |
Collapse
|
12
|
Garcia-Chica J, Paraiso WKD, Zagmutt S, Fosch A, Reguera AC, Alzina S, Sánchez-García L, Fukushima S, Toh K, Casals N, Serra D, Herrero L, Garcia J, Kataoka K, Ariza X, Quader S, Rodríguez-Rodríguez R. Nanomedicine targeting brain lipid metabolism as a feasible approach for controlling the energy balance. Biomater Sci 2023; 11:2336-2347. [PMID: 36804651 DOI: 10.1039/d2bm01751b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Targeting brain lipid metabolism is a promising strategy to regulate the energy balance and fight metabolic diseases such as obesity. The development of stable platforms for selective delivery of drugs, particularly to the hypothalamus, is a challenge but a possible solution for these metabolic diseases. Attenuating fatty acid oxidation in the hypothalamus via CPT1A inhibition leads to satiety, but this target is difficult to reach in vivo with the current drugs. We propose using an advanced crosslinked polymeric micelle-type nanomedicine that can stably load the CPT1A inhibitor C75-CoA for in vivo control of the energy balance. Central administration of the nanomedicine induced a rapid attenuation of food intake and body weight in mice via regulation of appetite-related neuropeptides and neuronal activation of specific hypothalamic regions driving changes in the liver and adipose tissue. This nanomedicine targeting brain lipid metabolism was successful in the modulation of food intake and peripheral metabolism in mice.
Collapse
Affiliation(s)
- Jesús Garcia-Chica
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, E-08195, Spain.
| | - West Kristian Dizon Paraiso
- Innovation Center of Nanomedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan.
| | - Sebastián Zagmutt
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, E-08195, Spain.
| | - Anna Fosch
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, E-08195, Spain.
| | - Ana Cristina Reguera
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, E-08195, Spain.
| | - Sara Alzina
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, E-08195, Spain.
| | - Laura Sánchez-García
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, E-08195, Spain.
| | - Shigeto Fukushima
- Innovation Center of Nanomedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan.
| | - Kazuko Toh
- Innovation Center of Nanomedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan.
| | - Núria Casals
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, E-08195, Spain. .,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, E-28029, Spain
| | - Dolors Serra
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, E-28029, Spain.,Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, E-08028, Spain
| | - Laura Herrero
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, E-28029, Spain.,Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, E-08028, Spain
| | - Jordi Garcia
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, E-28029, Spain.,Department of Inorganic and Organic Chemistry, Faculty of Chemistry, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona (UB), Barcelona, E-08028, Spain
| | - Kazunori Kataoka
- Innovation Center of Nanomedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan.
| | - Xavier Ariza
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, E-28029, Spain.,Department of Inorganic and Organic Chemistry, Faculty of Chemistry, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona (UB), Barcelona, E-08028, Spain
| | - Sabina Quader
- Innovation Center of Nanomedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan.
| | - Rosalía Rodríguez-Rodríguez
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, E-08195, Spain. .,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, E-28029, Spain
| |
Collapse
|
13
|
D’Ambrosio C, Cigliano L, Mazzoli A, Matuozzo M, Nazzaro M, Scaloni A, Iossa S, Spagnuolo MS. Fructose Diet-Associated Molecular Alterations in Hypothalamus of Adolescent Rats: A Proteomic Approach. Nutrients 2023; 15:nu15020475. [PMID: 36678346 PMCID: PMC9862284 DOI: 10.3390/nu15020475] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/13/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The enhanced consumption of fructose as added sugar represents a major health concern. Due to the complexity and multiplicity of hypothalamic functions, we aim to point out early molecular alterations triggered by a sugar-rich diet throughout adolescence, and to verify their persistence until the young adulthood phase. METHODS Thirty days old rats received a high-fructose or control diet for 3 weeks. At the end of the experimental period, treated animals were switched to the control diet for further 3 weeks, and then analyzed in comparison with those that were fed the control diet for the entire experimental period. RESULTS Quantitative proteomics identified 19 differentially represented proteins, between control and fructose-fed groups, belonging to intermediate filament cytoskeleton, neurofilament, pore complex and mitochondrial respiratory chain complexes. Western blotting analysis confirmed proteomic data, evidencing a decreased abundance of mitochondrial respiratory complexes and voltage-dependent anion channel 1, the coregulator of mitochondrial biogenesis PGC-1α, and the protein subunit of neurofilaments α-internexin in fructose-fed rats. Diet-associated hypothalamic inflammation was also detected. Finally, the amount of brain-derived neurotrophic factor and its high-affinity receptor TrkB, as well as of synaptophysin, synaptotagmin, and post-synaptic protein PSD-95 was reduced in sugar-fed rats. Notably, deregulated levels of all proteins were fully rescued after switching to the control diet. CONCLUSIONS A short-term fructose-rich diet in adolescent rats induces hypothalamic inflammation and highly affects mitochondrial and cytoskeletal compartments, as well as the level of specific markers of brain function; above-reported effects are reverted after switching animals to the control diet.
Collapse
Affiliation(s)
- Chiara D’Ambrosio
- Institute for the Animal Production System in the Mediterranean Environment, National Research Council, 80055 Portici, Italy
| | - Luisa Cigliano
- Department of Biology, University of Naples Federico II, 80121 Naples, Italy
| | - Arianna Mazzoli
- Department of Biology, University of Naples Federico II, 80121 Naples, Italy
| | - Monica Matuozzo
- Institute for the Animal Production System in the Mediterranean Environment, National Research Council, 80055 Portici, Italy
| | - Martina Nazzaro
- Department of Biology, University of Naples Federico II, 80121 Naples, Italy
| | - Andrea Scaloni
- Institute for the Animal Production System in the Mediterranean Environment, National Research Council, 80055 Portici, Italy
| | - Susanna Iossa
- Department of Biology, University of Naples Federico II, 80121 Naples, Italy
| | - Maria Stefania Spagnuolo
- Institute for the Animal Production System in the Mediterranean Environment, National Research Council, 80055 Portici, Italy
- Correspondence:
| |
Collapse
|
14
|
Spagnuolo MS, Mazzoli A, Nazzaro M, Troise AD, Gatto C, Tonini C, Colardo M, Segatto M, Scaloni A, Pallottini V, Iossa S, Cigliano L. Long-Lasting Impact of Sugar Intake on Neurotrophins and Neurotransmitters from Adolescence to Young Adulthood in Rat Frontal Cortex. Mol Neurobiol 2023; 60:1004-1020. [PMID: 36394711 PMCID: PMC9849314 DOI: 10.1007/s12035-022-03115-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 11/01/2022] [Indexed: 11/18/2022]
Abstract
The detrimental impact of fructose, a widely used sweetener in industrial foods, was previously evidenced on various brain regions. Although adolescents are among the highest consumers of sweet foods, whether brain alterations induced by the sugar intake during this age persist until young adulthood or are rescued returning to a healthy diet remains largely unexplored. To shed light on this issue, just weaned rats were fed with a fructose-rich or control diet for 3 weeks. At the end of the treatment, fructose-fed rats underwent a control diet for a further 3 weeks until young adulthood phase and compared with animals that received from the beginning the healthy control diet. We focused on the consequences induced by the sugar on the main neurotrophins and neurotransmitters in the frontal cortex, as its maturation continues until late adolescence, thus being the last brain region to achieve a full maturity. We observed that fructose intake induces inflammation and oxidative stress, alteration of mitochondrial function, and changes of brain-derived neurotrophic factor (BDNF) and neurotrophin receptors, synaptic proteins, acetylcholine, dopamine, and glutamate levels, as well as increased formation of the glycation end-products Nε-carboxymethyllysine (CML) and Nε-carboxyethyllysine (CEL). Importantly, many of these alterations (BDNF, CML, CEL, acetylcholinesterase activity, dysregulation of neurotransmitters levels) persisted after switching to the control diet, thus pointing out to the adolescence as a critical phase, in which extreme attention should be devoted to limit an excessive consumption of sweet foods that can affect brain physiology also in the long term.
Collapse
Affiliation(s)
- Maria Stefania Spagnuolo
- grid.419162.90000 0004 1781 6305Institute for the Animal Production System in the Mediterranean Environment, National Research Council, P.le E.Fermi 1, 80055 Portici, Italy
| | - Arianna Mazzoli
- grid.4691.a0000 0001 0790 385XDepartment of Biology, University of Naples Federico II, Complesso Universitario Monte S. Angelo, Edificio 7, Via Cintia - I-80126, Naples, Italy
| | - Martina Nazzaro
- grid.4691.a0000 0001 0790 385XDepartment of Biology, University of Naples Federico II, Complesso Universitario Monte S. Angelo, Edificio 7, Via Cintia - I-80126, Naples, Italy
| | - Antonio Dario Troise
- grid.419162.90000 0004 1781 6305Institute for the Animal Production System in the Mediterranean Environment, National Research Council, P.le E.Fermi 1, 80055 Portici, Italy
| | - Cristina Gatto
- grid.4691.a0000 0001 0790 385XDepartment of Biology, University of Naples Federico II, Complesso Universitario Monte S. Angelo, Edificio 7, Via Cintia - I-80126, Naples, Italy
| | - Claudia Tonini
- grid.8509.40000000121622106Department of Science, Biomedical and Technology Science Section, University Roma Tre, Rome, Italy
| | - Mayra Colardo
- grid.10373.360000000122055422Department of Biosciences and Territory, University of Molise, Pesche, Italy
| | - Marco Segatto
- grid.10373.360000000122055422Department of Biosciences and Territory, University of Molise, Pesche, Italy
| | - Andrea Scaloni
- grid.419162.90000 0004 1781 6305Institute for the Animal Production System in the Mediterranean Environment, National Research Council, P.le E.Fermi 1, 80055 Portici, Italy
| | - Valentina Pallottini
- grid.8509.40000000121622106Department of Science, Biomedical and Technology Science Section, University Roma Tre, Rome, Italy ,grid.417778.a0000 0001 0692 3437Neuroendocrinology Metabolism and Neuropharmacology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Susanna Iossa
- grid.4691.a0000 0001 0790 385XDepartment of Biology, University of Naples Federico II, Complesso Universitario Monte S. Angelo, Edificio 7, Via Cintia - I-80126, Naples, Italy
| | - Luisa Cigliano
- grid.4691.a0000 0001 0790 385XDepartment of Biology, University of Naples Federico II, Complesso Universitario Monte S. Angelo, Edificio 7, Via Cintia - I-80126, Naples, Italy
| |
Collapse
|
15
|
Ouchi Y, Komaki Y, Shimizu K, Fukano N, Sugino T, Shiraishi JI, Chowdhury VS, Bungo T. Comparison of oral administration of fructose and glucose on food intake and physiological parameters in broiler chicks. Poult Sci 2022; 102:102249. [PMID: 36335736 PMCID: PMC9640322 DOI: 10.1016/j.psj.2022.102249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/02/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022] Open
Abstract
Like glucose, fructose is a monosaccharide, but the mechanisms of its absorption and metabolism in the body are very different between the 2 molecules. In this study, we investigated the effects of oral administration of glucose and fructose on food intake, diencephalic gene expression, and plasma metabolite concentrations in broiler chicks. The animals used in this study were 4-day-old male broiler chicks (Ross 308). They were given glucose, fructose (200 mg/ 0.5 mL/ bird), or a similar volume of distilled water orally after 6 h fasting. After treatment, measurements of food intake (at 0, 30, and 60 min), and blood glucose as well as insulin concentrations were measured over time; however, diencephalic (hypothalamus) gene expression and plasma metabolites were measured at 30 min. The results showed that glucose administration suppressed food intake, but fructose administration did not suppress food intake and it was at the same level as distilled water administration. In addition, fructose administration did not increase plasma glucose and insulin levels as did glucose administration. In the diencephalon, expression levels of genes related to the melanocortin system were unaffected by the treatment, while gene expression levels related to intracellular energy regulation, such as AMP-activated protein kinase were affected by the glucose treatment in the fasted chicks. These results suggest that fructose administration does not suppress feeding behavior as a result of possible reduction in the energy levels in the diencephalon and associated energy metabolism.
Collapse
Affiliation(s)
- Yoshimitsu Ouchi
- Faculty of Veterinary Medicine, Okayama University of Science, Imabari 794-8555, Japan
| | - Yoshinori Komaki
- Graduate School of Bioresource Science, Hiroshima University, Higashi-Hiroshima 739-8528, Japan
| | - Kensuke Shimizu
- Graduate School of Bioresource Science, Hiroshima University, Higashi-Hiroshima 739-8528, Japan
| | - Natsuki Fukano
- Graduate School of Bioresource Science, Hiroshima University, Higashi-Hiroshima 739-8528, Japan
| | - Toshihisa Sugino
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima 739-8528, Japan
| | - Jun-ichi Shiraishi
- Department of Animal Science, Nippon Veterinary and Life Science University, Musashino 180-8602, Japan
| | - Vishwajit S. Chowdhury
- Faculty of Arts and Science, Graduate School of Bioresource and Bioenvironmental Science, Kyushu University, Fukuoka 819-0395, Japan
| | - Takashi Bungo
- Faculty of Veterinary Medicine, Okayama University of Science, Imabari 794-8555, Japan,Corresponding author:
| |
Collapse
|
16
|
Glucose and fructose directly stimulate brain-derived neurotrophic factor gene expression in microglia. Neuroreport 2022; 33:583-589. [DOI: 10.1097/wnr.0000000000001820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
17
|
Wu Z, Rao S, Li J, Ding N, Chen J, Feng L, Ma S, Hu C, Dai H, Wen L, Jiang Q, Deng J, Deng M, Tan C. Dietary adenosine 5’-monophosphate supplementation increases food intake and remodels energy expenditure in mice. Food Nutr Res 2022; 66:7680. [PMID: 35844957 PMCID: PMC9250134 DOI: 10.29219/fnr.v66.7680] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/30/2021] [Accepted: 10/04/2021] [Indexed: 11/20/2022] Open
Abstract
Background Methods Results Conclusions
Collapse
Affiliation(s)
- Zifang Wu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Sujuan Rao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jiaying Li
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Ning Ding
- Guangzhou Customs Technology Center, 510623, China
| | - Jianzhao Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Li Feng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Shuo Ma
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Chengjun Hu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Haonan Dai
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Lijun Wen
- Guangdong Hinabiotech Co., Ltd., Guangzhou, China
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jinping Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
- Ming Deng,
| | - Ming Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
- Ming Deng,
| | - Chengquan Tan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
- Chengquan Tan, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.
| |
Collapse
|
18
|
Aldhshan MS, Jhanji G, Poritsanos NJ, Mizuno TM. Glucose Stimulates Glial Cell Line-Derived Neurotrophic Factor Gene Expression in Microglia through a GLUT5-Independent Mechanism. Int J Mol Sci 2022; 23:ijms23137073. [PMID: 35806073 PMCID: PMC9266953 DOI: 10.3390/ijms23137073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 06/20/2022] [Accepted: 06/24/2022] [Indexed: 01/27/2023] Open
Abstract
Feeding-regulating neurotrophic factors are expressed in both neurons and glial cells. However, nutritional regulation of anorexigenic glial cell line-derived neurotrophic factor (GDNF) and orexigenic mesencephalic astrocyte-derived neurotrophic factor (MANF) expression in specific cell types remains poorly understood. Hypothalamic glucose sensing plays a critical role in the regulation of food intake. It has been theorized that local glucose concentration modulates microglial activity partially via glucose transporter 5 (GLUT5). We hypothesized that an increased local glucose concentration stimulates GDNF expression while inhibiting MANF expression in the hypothalamus and microglia via GLUT5. The present study investigated the effect of glucose on Gdnf and Manf mRNA expression in the mouse hypothalamus and murine microglial cell line SIM-A9. Intracerebroventricular glucose treatment significantly increased Gdnf mRNA levels in the hypothalamus without altering Manf mRNA levels. Exposure to high glucose caused a significant increase in Gdnf mRNA expression and a time-dependent change in Manf mRNA expression in SIM-A9 cells. GLUT5 inhibitor treatment did not block glucose-induced Gdnf mRNA expression in these cells. These findings suggest that microglia are responsive to changes in the local glucose concentration and increased local glucose availability stimulates the expression of microglial GNDF through a GLUT5-independent mechanism, contributing to glucose-induced feeding suppression.
Collapse
|
19
|
A Short-Term Sucrose Diet Impacts Cell Proliferation of Neural Precursors in the Adult Hypothalamus. Nutrients 2022; 14:nu14132564. [PMID: 35807744 PMCID: PMC9268421 DOI: 10.3390/nu14132564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 11/16/2022] Open
Abstract
Radial glia-like cells in the hypothalamus and dorsal vagal complex are neural precursors (NPs) located near subventricular organs: median eminence and area postrema, respectively. Their strategic position can detect blood-borne nutrients, hormones, and mitogenic signals. Hypothalamic NPs increase their proliferation with a mechanism that involves hemichannel (HC) activity. NPs can originate new neurons in response to a short-term high-fat diet as a compensatory mechanism. The effects of high carbohydrate Western diets on adult neurogenesis are unknown. Although sugars are usually consumed as sucrose, more free fructose is now incorporated into food items. Here, we studied the proliferation of both types of NPs in Sprague Dawley rats exposed to a short-term high sucrose diet (HSD) and a control diet. In tanycyte cultures, we evaluated the effects of glucose and fructose and a mix of both hexoses on HC activity. In rats fed an HSD, we observed an increase in the proliferative state of both precursors. Glucose, either in the presence or absence of fructose, but not fructose alone, induced in vitro HC activity. These results should broaden the understanding of the nutrient monitoring capacity of NPs in reacting to changes in feeding behavior, specifically to high sugar western diets.
Collapse
|
20
|
Duan Y, Du Y, Yi Z, Wang Z, Pei X, Wei X, Li M. Systematic Metabolic Engineering for the Production of Azaphilones in Monascus purpureus HJ11. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:1589-1600. [PMID: 35085438 DOI: 10.1021/acs.jafc.1c07588] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Fungal azaphilones have attracted considerable interest as they exhibit great potential in food and pharmacological industries. However, there is a severe bottleneck in the low production in wild strains and the ability to genetically engineer azaphilone-producing fungi. Using Monascus azaphilones (MAs) as an example, we demonstrate a systematic metabolic engineering strategy for improving the production of MAs. In this study, Monascus purpureus HJ11 was systematically engineered through a combination of promoter engineering, gene knockout, rate-limiting enzyme overexpression, repression of the competing pathway, enzyme engineering, and metabolic rebalance. The maximum yield and titer of MAs successfully increased to 906 mg/g dry cell weight (DCW) and 14.6 g/L, respectively, 2.6 and 3.7 times higher than those reported in the literature. Our successful model not only offers a practical and efficient way to improve the azaphilone production but also sheds light on the potential of systematic metabolic engineering in nonmodel fungi as a chassis for the production of high-value chemicals.
Collapse
Affiliation(s)
- Yali Duan
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China
| | - Yun Du
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China
| | - Zhiqiang Yi
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China
| | - Zhe Wang
- Polytechnic Institute, Zhejiang University, Hangzhou 310015, China
| | - Xiaolin Pei
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 310012, China
| | - Xuetuan Wei
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China
| | - Mu Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China
| |
Collapse
|
21
|
Silencing gut CCK cells alters gut reaction to sugar. Nat Neurosci 2022; 25:136-138. [PMID: 35027762 DOI: 10.1038/s41593-021-00998-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
22
|
Musso PY, Junca P, Gordon MD. A neural circuit linking two sugar sensors regulates satiety-dependent fructose drive in Drosophila. SCIENCE ADVANCES 2021; 7:eabj0186. [PMID: 34851668 PMCID: PMC8635442 DOI: 10.1126/sciadv.abj0186] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
In flies, neuronal sensors detect prandial changes in circulating fructose levels and either sustain or terminate feeding, depending on internal state. Here, we describe a three-part neural circuit that imparts satiety-dependent modulation of fructose sensing. We show that dorsal fan-shaped body neurons display oscillatory calcium activity when hemolymph glucose is high and that these oscillations require glutamatergic input from SLP-AB or “Janus” neurons projecting from the protocerebrum to the asymmetric body. Suppression of activity in this circuit, either by starvation or by genetic silencing, promotes specific drive for fructose ingestion. This is achieved through neuropeptidergic signaling by tachykinin, which is released from the fan-shaped body when glycemia is high. Tachykinin, in turn, signals to Gr43a-positive fructose sensors to modulate their response to fructose. Together, our results demonstrate how a three-layer neural circuit links the detection of two sugars to produce precise satiety-dependent control of feeding behavior.
Collapse
|
23
|
Li J, Long J, Zhang Q, Shen H, Guo AY, Ma Z, Zhang G. Hypothalamic long noncoding RNA AK044061 is involved in the development of dietary obesity in mice. Int J Obes (Lond) 2021; 45:2638-2647. [PMID: 34446844 DOI: 10.1038/s41366-021-00945-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 08/16/2021] [Accepted: 08/16/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) have been implicated in various important biological processes, however, its role in energy balance and obesity remains largely unknown. METHODS Differentially expressed lncRNAs in the hypothalamus of diet-induced obesity (DIO) mice versus chow-fed mice were identified by RNA sequencing. Lentivirus-mediated overexpression and knockdown of a novel lncRNA, AK044061, were used to assess its role in energy balance and the development of DIO. RNA immunoprecipitation (RIP) and pull down assays were carried out to analyze the interaction between lncRNA AK044061 and RelA, an NF-κB subunit. RESULTS LncRNA AK044061 was upregulated in the hypothalamus of DIO mice. Acute intracerebroventricular (i.c.v.) infusion of glucose reduced the expression of lncRNA AK044061, whereas an overnight of fasting enhanced its expression. RNA in situ hybridization data showed that AK044061 was expressed in the neurons of the arcuate nucleus (ARC). Lentivirus-mediated overexpression of AK044061 in ARC cells, or in the neurons of the ARC nucleus led to an obesity-like phenotype and related metabolic disorders. Furthermore, knockdown of lncRNA AK044061 in Agouti-related peptide (AgRP)-expressing neurons mitigated DIO and its related metabolic dysregulations. In mechanism, we showed that lncRNA AK044061 was associated with RelA and could enhance the NF-κB reporter activity. The effect of lncRNA AK044061 on energy balance is mediated by NF-κB. CONCLUSIONS Our findings suggest that excessive lncRNA AK044061 in the ARC nucleus leads to energy imbalance and obesity. LncRNA AK044061 expressed in the AgRP neurons is important in the development of dietary obesity in mice.
Collapse
Affiliation(s)
- Juan Li
- Key Laboratory of Environmental Health, Ministry of Education, Department of Toxicology, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jinlie Long
- Key Laboratory of Environmental Health, Ministry of Education, Department of Toxicology, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qiong Zhang
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongyuan Shen
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore, Singapore
| | - An-Yuan Guo
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Zhaowu Ma
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China.
| | - Guo Zhang
- Key Laboratory of Environmental Health, Ministry of Education, Department of Toxicology, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
24
|
Regulation of the Fructose Transporter Gene Slc2a5 Expression by Glucose in Cultured Microglial Cells. Int J Mol Sci 2021; 22:ijms222312668. [PMID: 34884473 PMCID: PMC8657830 DOI: 10.3390/ijms222312668] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 01/16/2023] Open
Abstract
Microglia play a role in the regulation of metabolism and pathogenesis of obesity. Microglial activity is altered in response to changes in diet and the body’s metabolic state. Solute carrier family 2 member 5 (Slc2a5) that encodes glucose transporter 5 (GLUT5) is a fructose transporter primarily expressed in microglia within the central nervous system. However, little is known about the nutritional regulation of Slc2a5 expression in microglia and its role in the regulation of metabolism. The present study aimed to address the hypothesis that nutrients affect microglial activity by altering the expression of glucose transporter genes. Murine microglial cell line SIM-A9 cells and primary microglia from mouse brain were exposed to different concentrations of glucose and levels of microglial activation markers and glucose transporter genes were measured. High concentration of glucose increased levels of the immediate-early gene product c-Fos, a marker of cell activation, Slc2a5 mRNA, and pro-inflammatory cytokine genes in microglial cells in a time-dependent manner, while fructose failed to cause these changes. Glucose-induced changes in pro-inflammatory gene expression were partially attenuated in SIM-A9 cells treated with the GLUT5 inhibitor. These findings suggest that an increase in local glucose availability leads to the activation of microglia by controlling their carbohydrate sensing mechanism through both GLUT5-dependent and –independent mechanisms.
Collapse
|
25
|
Jiang H, Lin Q, Ma L, Luo S, Jiang X, Fang J, Lu Z. Fructose and fructose kinase in cancer and other pathologies. J Genet Genomics 2021; 48:531-539. [PMID: 34326012 DOI: 10.1016/j.jgg.2021.06.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 12/14/2022]
Abstract
Fructose metabolism and fructose kinase KHK-C/A are key factors in the development of lipid oversynthesis-promoted metabolic disorders and cancer. Here, we summarize and discuss the current knowledge about the specific features of fructose metabolism and the distinct roles of KHK-C and KHK-A in metabolic liver diseases and their relevant metabolic disorders and cancer, and we highlight the specific protein kinase activity of KHK-A in tumor development. In addition, different approaches that have been used to inhibit KHK and the exploration of KHK inhibitors in clinical treatment are introduced.
Collapse
Affiliation(s)
- Hongfei Jiang
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao 266061, China
| | - Qian Lin
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao 266061, China
| | - Leina Ma
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao 266061, China
| | - Shudi Luo
- Zhejiang University Cancer Center, Hangzhou 310029, China
| | - Xiaoming Jiang
- Zhejiang University Cancer Center, Hangzhou 310029, China
| | - Jing Fang
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao 266061, China.
| | - Zhimin Lu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China; Zhejiang University Cancer Center, Hangzhou 310029, China.
| |
Collapse
|
26
|
Neural mechanisms underlying the role of fructose in overfeeding. Neurosci Biobehav Rev 2021; 128:346-357. [PMID: 34182019 DOI: 10.1016/j.neubiorev.2021.06.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 06/16/2021] [Accepted: 06/23/2021] [Indexed: 10/21/2022]
Abstract
Fructose consumption has been linked with metabolic syndrome and obesity. Fructose-based sweeteners like high fructose corn syrup taste sweeter, improve food palatability, and are increasingly prevalent in our diet. The increase in fructose consumption precedes the rise in obesity and is a contributing driver to the obesity epidemic worldwide. The role of dietary fructose in obesity can be multifactorial by promoting visceral adiposity, hypertension, and insulin resistance. Interestingly, one emergent finding from human and animal studies is that dietary fructose promotes overfeeding. As the brain is a critical regulator of food intake, we reviewed the evidence that fructose can act in the brain and elucidated the major brain systems underlying fructose-induced overfeeding. We found that fructose acts on multiple interdependent brain systems to increase orexigenic drive and the incentive salience of food while decreasing the latency between food bouts and reducing cognitive control to disinhibit feeding. We concluded that the collective actions of fructose may promote feeding behavior by producing a hunger-like state in the brain.
Collapse
|
27
|
Amorim R, Simões ICM, Veloso C, Carvalho A, Simões RF, Pereira FB, Thiel T, Normann A, Morais C, Jurado AS, Wieckowski MR, Teixeira J, Oliveira PJ. Exploratory Data Analysis of Cell and Mitochondrial High-Fat, High-Sugar Toxicity on Human HepG2 Cells. Nutrients 2021; 13:nu13051723. [PMID: 34069635 PMCID: PMC8161147 DOI: 10.3390/nu13051723] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/06/2021] [Accepted: 05/17/2021] [Indexed: 12/13/2022] Open
Abstract
Non-alcoholic steatohepatitis (NASH), one of the deleterious stages of non-alcoholic fatty liver disease, remains a significant cause of liver-related morbidity and mortality worldwide. In the current work, we used an exploratory data analysis to investigate time-dependent cellular and mitochondrial effects of different supra-physiological fatty acids (FA) overload strategies, in the presence or absence of fructose (F), on human hepatoma-derived HepG2 cells. We measured intracellular neutral lipid content and reactive oxygen species (ROS) levels, mitochondrial respiration and morphology, and caspases activity and cell death. FA-treatments induced a time-dependent increase in neutral lipid content, which was paralleled by an increase in ROS. Fructose, by itself, did not increase intracellular lipid content nor aggravated the effects of palmitic acid (PA) or free fatty acids mixture (FFA), although it led to an up-expression of hepatic fructokinase. Instead, F decreased mitochondrial phospholipid content, as well as OXPHOS subunits levels. Increased lipid accumulation and ROS in FA-treatments preceded mitochondrial dysfunction, comprising altered mitochondrial membrane potential (ΔΨm) and morphology, and decreased oxygen consumption rates, especially with PA. Consequently, supra-physiological PA alone or combined with F prompted the activation of caspase pathways leading to a time-dependent decrease in cell viability. Exploratory data analysis methods support this conclusion by clearly identifying the effects of FA treatments. In fact, unsupervised learning algorithms created homogeneous and cohesive clusters, with a clear separation between PA and FFA treated samples to identify a minimal subset of critical mitochondrial markers in order to attain a feasible model to predict cell death in NAFLD or for high throughput screening of possible therapeutic agents, with particular focus in measuring mitochondrial function.
Collapse
Affiliation(s)
- Ricardo Amorim
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC-Biotech, Biocant Park, 3060-197 Cantanhede, Portugal; (R.A.); (C.V.); (A.C.); (R.F.S.); (J.T.)
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
- PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3004-531 Coimbra, Portugal
| | - Inês C. M. Simões
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 02-093 Warsaw, Poland; (I.C.M.S.); (M.R.W.)
| | - Caroline Veloso
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC-Biotech, Biocant Park, 3060-197 Cantanhede, Portugal; (R.A.); (C.V.); (A.C.); (R.F.S.); (J.T.)
| | - Adriana Carvalho
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC-Biotech, Biocant Park, 3060-197 Cantanhede, Portugal; (R.A.); (C.V.); (A.C.); (R.F.S.); (J.T.)
- PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3004-531 Coimbra, Portugal
| | - Rui F. Simões
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC-Biotech, Biocant Park, 3060-197 Cantanhede, Portugal; (R.A.); (C.V.); (A.C.); (R.F.S.); (J.T.)
- PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3004-531 Coimbra, Portugal
| | - Francisco B. Pereira
- Center for Informatics and Systems, University of Coimbra, Polo II, Pinhal de Marrocos, 3030-290 Coimbra, Portugal;
- Coimbra Polytechnic-ISEC, 3030-190 Coimbra, Portugal
| | - Theresa Thiel
- Mediagnostic, D-72770 Reutlingen, Germany; (T.T.); (A.N.)
| | - Andrea Normann
- Mediagnostic, D-72770 Reutlingen, Germany; (T.T.); (A.N.)
| | - Catarina Morais
- Center for Neuroscience and Cell Biology, Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal; (C.M.); (A.S.J.)
| | - Amália S. Jurado
- Center for Neuroscience and Cell Biology, Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal; (C.M.); (A.S.J.)
| | - Mariusz R. Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 02-093 Warsaw, Poland; (I.C.M.S.); (M.R.W.)
| | - José Teixeira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC-Biotech, Biocant Park, 3060-197 Cantanhede, Portugal; (R.A.); (C.V.); (A.C.); (R.F.S.); (J.T.)
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Paulo J. Oliveira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC-Biotech, Biocant Park, 3060-197 Cantanhede, Portugal; (R.A.); (C.V.); (A.C.); (R.F.S.); (J.T.)
- Correspondence:
| |
Collapse
|
28
|
Schneider ACR, de Moura AC, Carvalho FB, Alves T, Meurer F, Porawski M, da Silveira TR. Effect of Melatonin on the Reduction of Hepatic Steatosis and Intestinal Leptin Expression in Zebrafish Exposed to Fructose. Zebrafish 2021; 18:184-189. [PMID: 33983041 DOI: 10.1089/zeb.2020.1910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Melatonin is a hormone related to circadian rhythms and has potential clinical applications. Our objectives were to verify the effect of melatonin on the liver of zebrafish exposed to fructose and evaluate the expression of appetite-related genes (leptin, ghrelin, and melanocortin receptor 4 [MC4R]). Animals were divided into three groups: control (CG, n = 25), fructose (FG, n = 25), and fructose+melatonin (FMG, n = 25). The study was carried out in 8 weeks. FG and FMG were exposed to 2% fructose and FMG treated with 1 μM of melatonin. Histological liver studies and gene expression analyses of Leptin, Ghrelin, and MC4R (liver and intestines) were performed. FG developed hepatic steatosis, which did not occur with CG and FMG. Genetic expression of hepatic leptin and MC4R did not show significant difference among the groups. Animals exposed to fructose (FG) presented an increased expression of intestinal leptin compared to those administered with melatonin. Animals exposed to fructose gained weight and developed an important hepatic steatosis, but melatonin reduced significantly the hepatic damage. Intestinal leptin showed increased expression in the group exposed to fructose.
Collapse
Affiliation(s)
- Ana Claudia Reis Schneider
- Programa de Pós-Graduação em Pediatria, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Ana Carolina de Moura
- Laboratório de Fisiologia Comportamental e Metabólica, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Fabiano Barbosa Carvalho
- Laboratório de Pesquisa em Patologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Thiago Alves
- Grupo de Pesquisa Experimental em Zebrafish/Biotério, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Fabio Meurer
- Campus Avançado de Jandaia do Sul, Universidade Federal do Paraná, Jandaia do Sul, Rio Grande do Sul, Brazil
| | - Marilene Porawski
- Programa de Pós-graduação em Hepatologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Themis Reverbel da Silveira
- Programa de Pós-Graduação em Pediatria, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
29
|
Gomez-Pinilla F, Cipolat RP, Royes LFF. Dietary fructose as a model to explore the influence of peripheral metabolism on brain function and plasticity. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166036. [PMID: 33508421 DOI: 10.1016/j.bbadis.2020.166036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023]
Abstract
High consumption of fructose has paralleled an explosion in metabolic disorders including obesity and type 2 diabetes. Even more problematic, sustained consumption of fructose is perceived as a threat for brain function and development of neurological disorders. The action of fructose on peripheral organs is an excellent model to understand how systemic physiology impacts the brain. Given the recognized action of fructose on liver metabolism, here we discuss mechanisms by which fructose can impact the brain by interacting with liver and other organs. The interaction between peripheral and central mechanisms is a suitable target to reduce the pathophysiological consequences of neurological disorders.
Collapse
Affiliation(s)
- Fernando Gomez-Pinilla
- Department of Neurosurgery, UCLA Brain Injury Research Center, University of California Los Angeles, USA; Department of Integrative Biology and Physiology, UCLA Brain Injury Research Center, University of California Los Angeles, USA.
| | - Rafael Parcianello Cipolat
- Exercise Biochemistry Laboratory, Center of Physical Education and Sports, Federal University of Santa Maria - UFSM, Santa Maria, RS, Brazil
| | - Luiz Fernando Freire Royes
- Exercise Biochemistry Laboratory, Center of Physical Education and Sports, Federal University of Santa Maria - UFSM, Santa Maria, RS, Brazil
| |
Collapse
|
30
|
Sweet but Bitter: Focus on Fructose Impact on Brain Function in Rodent Models. Nutrients 2020; 13:nu13010001. [PMID: 33374894 PMCID: PMC7821920 DOI: 10.3390/nu13010001] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022] Open
Abstract
Fructose consumption has drastically increased during the last decades due to the extensive commercial use of high-fructose corn syrup as a sweetener for beverages, snacks and baked goods. Fructose overconsumption is known to induce obesity, dyslipidemia, insulin resistance and inflammation, and its metabolism is considered partially responsible for its role in several metabolic diseases. Indeed, the primary metabolites and by-products of gut and hepatic fructolysis may impair the functions of extrahepatic tissues and organs. However, fructose itself causes an adenosine triphosphate (ATP) depletion that triggers inflammation and oxidative stress. Many studies have dealt with the effects of this sugar on various organs, while the impact of fructose on brain function is, to date, less explored, despite the relevance of this issue. Notably, fructose transporters and fructose metabolizing enzymes are present in brain cells. In addition, it has emerged that fructose consumption, even in the short term, can adversely influence brain health by promoting neuroinflammation, brain mitochondrial dysfunction and oxidative stress, as well as insulin resistance. Fructose influence on synaptic plasticity and cognition, with a major impact on critical regions for learning and memory, was also reported. In this review, we discuss emerging data about fructose effects on brain health in rodent models, with special reference to the regulation of food intake, inflammation, mitochondrial function and oxidative stress, insulin signaling and cognitive function.
Collapse
|
31
|
Fadó R, Rodríguez-Rodríguez R, Casals N. The return of malonyl-CoA to the brain: Cognition and other stories. Prog Lipid Res 2020; 81:101071. [PMID: 33186641 DOI: 10.1016/j.plipres.2020.101071] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/06/2020] [Accepted: 11/08/2020] [Indexed: 12/11/2022]
Abstract
Nutrients, hormones and the energy sensor AMP-activated protein kinase (AMPK) tightly regulate the intracellular levels of the metabolic intermediary malonyl-CoA, which is a precursor of fatty acid synthesis and a negative regulator of fatty acid oxidation. In the brain, the involvement of malonyl-CoA in the control of food intake and energy homeostasis has been known for decades. However, recent data uncover a new role in cognition and brain development. The sensing of malonyl-CoA by carnitine palmitoyltransferase 1 (CPT1) proteins regulates a variety of functions, such as the fate of neuronal stem cell precursors, the motility of lysosomes in developing axons, the trafficking of glutamate receptors to the neuron surface (necessary for proper synaptic function) and the metabolic coupling between astrocytes and neurons. We discuss the relevance of those recent findings evidencing how nutrients and metabolic disorders impact cognition. We also enumerate all nutritional and hormonal conditions that are known to regulate malonyl-CoA levels in the brain, reflect on protein malonylation as a new post-translational modification, and give a reasoned vision of the opportunities and challenges that future research in the field could address.
Collapse
Affiliation(s)
- Rut Fadó
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain; Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Bellaterra, Cerdanyola del Vallès, Spain.
| | - Rosalía Rodríguez-Rodríguez
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain.
| | - Núria Casals
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain.
| |
Collapse
|
32
|
Helsley RN, Moreau F, Gupta MK, Radulescu A, DeBosch B, Softic S. Tissue-Specific Fructose Metabolism in Obesity and Diabetes. Curr Diab Rep 2020; 20:64. [PMID: 33057854 DOI: 10.1007/s11892-020-01342-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/10/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW The objective of this review is to provide up-to-date and comprehensive discussion of tissue-specific fructose metabolism in the context of diabetes, dyslipidemia, and nonalcoholic fatty liver disease (NAFLD). RECENT FINDINGS Increased intake of dietary fructose is a risk factor for a myriad of metabolic complications. Tissue-specific fructose metabolism has not been well delineated in terms of its contribution to detrimental health effects associated with fructose intake. Since inhibitors targeting fructose metabolism are being developed for the management of NAFLD and diabetes, it is essential to recognize how inability of one tissue to metabolize fructose may affect metabolism in the other tissues. The primary sites of fructose metabolism are the liver, intestine, and kidney. Skeletal muscle and adipose tissue can also metabolize a large portion of fructose load, especially in the setting of ketohexokinase deficiency, the rate-limiting enzyme of fructose metabolism. Fructose can also be sensed by the pancreas and the brain, where it can influence essential functions involved in energy homeostasis. Lastly, fructose is metabolized by the testes, red blood cells, and lens of the eye where it may contribute to infertility, advanced glycation end products, and cataracts, respectively. An increase in sugar intake, particularly fructose, has been associated with the development of obesity and its complications. Inhibition of fructose utilization in tissues primary responsible for its metabolism alters consumption in other tissues, which have not been traditionally regarded as important depots of fructose metabolism.
Collapse
Affiliation(s)
- Robert N Helsley
- Division of Pediatric Gastroenterology, Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
| | - Francois Moreau
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Manoj K Gupta
- Islet Cell and Regenerative Medicine, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA, 02215, USA
| | - Aurelia Radulescu
- Department of Pediatrics, University of Kentucky College of Medicine and Kentucky Children's Hospital, Lexington, KY, 40536, USA
| | - Brian DeBosch
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63131, USA
| | - Samir Softic
- Division of Pediatric Gastroenterology, Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, 40506, USA.
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, 138 Leader Ave, Lexington, KY, 40506, USA.
| |
Collapse
|
33
|
Chiba Y, Murakami R, Matsumoto K, Wakamatsu K, Nonaka W, Uemura N, Yanase K, Kamada M, Ueno M. Glucose, Fructose, and Urate Transporters in the Choroid Plexus Epithelium. Int J Mol Sci 2020; 21:E7230. [PMID: 33008107 PMCID: PMC7582461 DOI: 10.3390/ijms21197230] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 02/07/2023] Open
Abstract
The choroid plexus plays a central role in the regulation of the microenvironment of the central nervous system by secreting the majority of the cerebrospinal fluid and controlling its composition, despite that it only represents approximately 1% of the total brain weight. In addition to a variety of transporter and channel proteins for solutes and water, the choroid plexus epithelial cells are equipped with glucose, fructose, and urate transporters that are used as energy sources or antioxidative neuroprotective substrates. This review focuses on the recent advances in the understanding of the transporters of the SLC2A and SLC5A families (GLUT1, SGLT2, GLUT5, GLUT8, and GLUT9), as well as on the urate-transporting URAT1 and BCRP/ABCG2, which are expressed in choroid plexus epithelial cells. The glucose, fructose, and urate transporters repertoire in the choroid plexus epithelium share similar features with the renal proximal tubular epithelium, although some of these transporters exhibit inversely polarized submembrane localization. Since choroid plexus epithelial cells have high energy demands for proper functioning, a decline in the expression and function of these transporters can contribute to the process of age-associated brain impairment and pathophysiology of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yoichi Chiba
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (K.M.); (K.W.)
| | - Ryuta Murakami
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (K.M.); (K.W.)
| | - Koichi Matsumoto
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (K.M.); (K.W.)
| | - Keiji Wakamatsu
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (K.M.); (K.W.)
| | - Wakako Nonaka
- Department of Supportive and Promotive Medicine of the Municipal Hospital, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan;
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Naoya Uemura
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan; (N.U.); (K.Y.)
| | - Ken Yanase
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan; (N.U.); (K.Y.)
| | - Masaki Kamada
- Department of Neurological Intractable Disease Research, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan;
| | - Masaki Ueno
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (K.M.); (K.W.)
| |
Collapse
|
34
|
Cargnin-Carvalho A, de Mello AH, Bressan JB, Backes KM, Uberti MF, Fogaça JB, da Rosa Turatti C, Cavalheiro EKFF, Vilela TC, Rezin GT. Can fructose influence the development of obesity mediated through hypothalamic alterations? J Neurosci Res 2020; 98:1662-1668. [PMID: 32524664 DOI: 10.1002/jnr.24628] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 03/16/2020] [Accepted: 03/30/2020] [Indexed: 12/13/2022]
Abstract
Epidemiological data from the last decades point to an exponential growth in the number of obese people. Different behavioral factors, mainly associated with food consumption, appear to contribute significantly to its development. Concomitant with increased obesity rates, an increase in the consumption of fructose has been observed; therefore, fructose consumption has been implicated as an important obesogenic factor. However, changes in brain activity due to fructose consumption are possible, especially in relation to hypothalamic satiety mechanisms. In addition, the obese state may provide an environment of chronic inflammation and further contribute to the discontinuation of satiety mechanisms in the hypothalamus. We briefly review the intrinsic alterations to the increased adipose tissue, its connections with the hypothalamus in the control of energy signaling mechanisms and, consequently, the participation of fructose as a co-adjuvant or trigger. Presenting the current context with clinical trials involving human and animal studies, we seek to contribute to a better understanding of the role of fructose in the progression of obesity.
Collapse
Affiliation(s)
- Anderson Cargnin-Carvalho
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina at Tubarão, Santa Catarina, Brazil
| | - Aline Haas de Mello
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina at Tubarão, Santa Catarina, Brazil
| | - Joice Benedet Bressan
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina at Tubarão, Santa Catarina, Brazil
| | - Kassiane Mathiola Backes
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina at Tubarão, Santa Catarina, Brazil
| | - Marcela Fornari Uberti
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina at Tubarão, Santa Catarina, Brazil
| | - Jéssica Benedet Fogaça
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina at Tubarão, Santa Catarina, Brazil
| | - Cristini da Rosa Turatti
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina at Tubarão, Santa Catarina, Brazil
| | - Eulla Keimili Fernandes Ferreira Cavalheiro
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina at Tubarão, Santa Catarina, Brazil
| | - Thais Ceresér Vilela
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina at Tubarão, Santa Catarina, Brazil
| | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina at Tubarão, Santa Catarina, Brazil
| |
Collapse
|
35
|
Ao H, Li J, Li O, Su M, Gao X. Fructose vs glucose decreased liking/wanting and subsequent intake of high-energy foods in young women. Nutr Res 2020; 78:60-71. [PMID: 32516689 DOI: 10.1016/j.nutres.2020.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 03/11/2020] [Accepted: 05/06/2020] [Indexed: 12/25/2022]
Abstract
Recent research on the health impacts of added sugar has prompted the comparison of the effects of its 2 major components: glucose and fructose. Fructose was identified as a risk factor for obesity and metabolic syndrome. However, because of the differences in metabolic responses and responsivity of reward circuitry to palatable food, it is unknown if glucose and fructose induce similar appetite-related responses in humans with varying weights. This study compared the behavioral responses to food in young women of a healthy weight (n = 31) and with excess weight (n = 28). We hypothesized that (1) the inhibitory effect of glucose (vs fructose) on food-related responses would be greater in subjects of a healthy weight than in those with overweight/obesity and (2) subjects with overweight/obesity would exhibit a stronger preference for food than subjects with a healthy weight. After an overnight fast, the subjects ingested a glucose or equienergetic fructose beverage on 2 separate days, respectively. Then, they completed liking and wanting ratings and 2 decision-making tasks followed by ad libitum food intake. The results revealed that fructose reduced both liking and wanting for food in subjects with overweight/obesity and also decreased energy intake in all subjects. Relative to the healthy-weight group, subjects with overweight/obesity preferred the immediate reward. Moreover, only in the healthy-weight group were liking and wanting scores for food positively associated with actual food consumption. Overall, fructose (vs glucose) showed an acute inhibitory effect on appetite-related responses in subjects with excess weight.
Collapse
Affiliation(s)
- Hua Ao
- Faculty of Psychology, Southwest University, Chongqing, China; Key Laboratory of Cognition and Personality, Southwest University, Chongqing, China.
| | - Jiachun Li
- Faculty of Psychology, Southwest University, Chongqing, China.
| | - Ouwen Li
- Faculty of Psychology, Southwest University, Chongqing, China.
| | - Manyi Su
- Faculty of Psychology, Southwest University, Chongqing, China.
| | - Xiao Gao
- Faculty of Psychology, Southwest University, Chongqing, China; Key Laboratory of Cognition and Personality, Southwest University, Chongqing, China.
| |
Collapse
|
36
|
Brun T, Jiménez-Sánchez C, Madsen JGS, Hadadi N, Duhamel D, Bartley C, Oberhauser L, Trajkovski M, Mandrup S, Maechler P. AMPK Profiling in Rodent and Human Pancreatic Beta-Cells under Nutrient-Rich Metabolic Stress. Int J Mol Sci 2020; 21:ijms21113982. [PMID: 32492936 PMCID: PMC7312098 DOI: 10.3390/ijms21113982] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic exposure of pancreatic β-cells to elevated nutrient levels impairs their function and potentially induces apoptosis. Like in other cell types, AMPK is activated in β-cells under conditions of nutrient deprivation, while little is known on AMPK responses to metabolic stresses. Here, we first reviewed recent studies on the role of AMPK activation in β-cells. Then, we investigated the expression profile of AMPK pathways in β-cells following metabolic stresses. INS-1E β-cells and human islets were exposed for 3 days to glucose (5.5–25 mM), palmitate or oleate (0.4 mM), and fructose (5.5 mM). Following these treatments, we analyzed transcript levels of INS-1E β-cells by qRT-PCR and of human islets by RNA-Seq; with a special focus on AMPK-associated genes, such as the AMPK catalytic subunits α1 (Prkaa1) and α2 (Prkaa2). AMPKα and pAMPKα were also evaluated at the protein level by immunoblotting. Chronic exposure to the different metabolic stresses, known to alter glucose-stimulated insulin secretion, did not change AMPK expression, either in insulinoma cells or in human islets. Expression profile of the six AMPK subunits was marginally modified by the different diabetogenic conditions. However, the expression of some upstream kinases and downstream AMPK targets, including K-ATP channel subunits, exhibited stress-specific signatures. Interestingly, at the protein level, chronic fructose treatment favored fasting-like phenotype in human islets, as witnessed by AMPK activation. Collectively, previously published and present data indicate that, in the β-cell, AMPK activation might be implicated in the pre-diabetic state, potentially as a protective mechanism.
Collapse
Affiliation(s)
- Thierry Brun
- Department of Cell Physiology and Metabolism & Faculty Diabetes Center, University of Geneva Medical Center, 1206 Geneva, Switzerland; (T.B.); (C.J.-S.); (N.H.); (D.D.); (C.B.); (L.O.); (M.T.)
| | - Cecilia Jiménez-Sánchez
- Department of Cell Physiology and Metabolism & Faculty Diabetes Center, University of Geneva Medical Center, 1206 Geneva, Switzerland; (T.B.); (C.J.-S.); (N.H.); (D.D.); (C.B.); (L.O.); (M.T.)
| | - Jesper Grud Skat Madsen
- Functional Genomics and Metabolism Research Unit, Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark; (J.G.S.M.); (S.M.)
| | - Noushin Hadadi
- Department of Cell Physiology and Metabolism & Faculty Diabetes Center, University of Geneva Medical Center, 1206 Geneva, Switzerland; (T.B.); (C.J.-S.); (N.H.); (D.D.); (C.B.); (L.O.); (M.T.)
| | - Dominique Duhamel
- Department of Cell Physiology and Metabolism & Faculty Diabetes Center, University of Geneva Medical Center, 1206 Geneva, Switzerland; (T.B.); (C.J.-S.); (N.H.); (D.D.); (C.B.); (L.O.); (M.T.)
| | - Clarissa Bartley
- Department of Cell Physiology and Metabolism & Faculty Diabetes Center, University of Geneva Medical Center, 1206 Geneva, Switzerland; (T.B.); (C.J.-S.); (N.H.); (D.D.); (C.B.); (L.O.); (M.T.)
| | - Lucie Oberhauser
- Department of Cell Physiology and Metabolism & Faculty Diabetes Center, University of Geneva Medical Center, 1206 Geneva, Switzerland; (T.B.); (C.J.-S.); (N.H.); (D.D.); (C.B.); (L.O.); (M.T.)
| | - Mirko Trajkovski
- Department of Cell Physiology and Metabolism & Faculty Diabetes Center, University of Geneva Medical Center, 1206 Geneva, Switzerland; (T.B.); (C.J.-S.); (N.H.); (D.D.); (C.B.); (L.O.); (M.T.)
| | - Susanne Mandrup
- Functional Genomics and Metabolism Research Unit, Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark; (J.G.S.M.); (S.M.)
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism & Faculty Diabetes Center, University of Geneva Medical Center, 1206 Geneva, Switzerland; (T.B.); (C.J.-S.); (N.H.); (D.D.); (C.B.); (L.O.); (M.T.)
- Correspondence:
| |
Collapse
|
37
|
Dietary Fructose Intake and Hippocampal Structure and Connectivity during Childhood. Nutrients 2020; 12:nu12040909. [PMID: 32224933 PMCID: PMC7230400 DOI: 10.3390/nu12040909] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/21/2020] [Accepted: 03/24/2020] [Indexed: 01/06/2023] Open
Abstract
In rodent literature, there is evidence that excessive fructose consumption during development has a detrimental impact on hippocampal structure and function. In this study of 103 children ages 7–11 years old, we investigated whether dietary fructose intake was related to alterations in hippocampal volume and connectivity in humans. To examine if these associations were specific to fructose or were related to dietary sugars intake in general, we explored relationships between dietary intake of added sugars and the monosaccharide, glucose, on the same brain measures. We found that increased dietary intake of fructose, measured as a percentage of total calories, was associated with both an increase in the volume of the CA2/3 subfield of the right hippocampus and increased axial, radial, and mean diffusivity in the prefrontal connections of the right cingulum. These findings are consistent with the idea that increased fructose consumption during childhood may be associated with an inflammatory process, and/or decreases or delays in myelination and/or pruning. Increased habitual consumption of glucose or added sugar in general were associated with an increased volume of right CA2/3, but not with any changes in the connectivity of the hippocampus. These findings support animal data suggesting that higher dietary intake of added sugars, particularly fructose, are associated with alterations in hippocampal structure and connectivity during childhood.
Collapse
|
38
|
Merino B, Fernández-Díaz CM, Cózar-Castellano I, Perdomo G. Intestinal Fructose and Glucose Metabolism in Health and Disease. Nutrients 2019; 12:E94. [PMID: 31905727 PMCID: PMC7019254 DOI: 10.3390/nu12010094] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 12/26/2019] [Accepted: 12/26/2019] [Indexed: 02/06/2023] Open
Abstract
The worldwide epidemics of obesity and diabetes have been linked to increased sugar consumption in humans. Here, we review fructose and glucose metabolism, as well as potential molecular mechanisms by which excessive sugar consumption is associated to metabolic diseases and insulin resistance in humans. To this end, we focus on understanding molecular and cellular mechanisms of fructose and glucose transport and sensing in the intestine, the intracellular signaling effects of dietary sugar metabolism, and its impact on glucose homeostasis in health and disease. Finally, the peripheral and central effects of dietary sugars on the gut-brain axis will be reviewed.
Collapse
Affiliation(s)
- Beatriz Merino
- Instituto de Biología y Genética Molecular-IBGM (CSIC-Universidad de Valladolid), Valladolid 47003, Spain; (B.M.); (C.M.F.-D.); (G.P.)
| | - Cristina M. Fernández-Díaz
- Instituto de Biología y Genética Molecular-IBGM (CSIC-Universidad de Valladolid), Valladolid 47003, Spain; (B.M.); (C.M.F.-D.); (G.P.)
| | - Irene Cózar-Castellano
- Instituto de Biología y Genética Molecular-IBGM (CSIC-Universidad de Valladolid), Valladolid 47003, Spain; (B.M.); (C.M.F.-D.); (G.P.)
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid 28029, Spain
| | - German Perdomo
- Instituto de Biología y Genética Molecular-IBGM (CSIC-Universidad de Valladolid), Valladolid 47003, Spain; (B.M.); (C.M.F.-D.); (G.P.)
- Departamento de Ciencias de la Salud, Universidad de Burgos, Burgos 09001, Spain
| |
Collapse
|
39
|
Fructose increases the activity of sodium hydrogen exchanger in renal proximal tubules that is dependent on ketohexokinase. J Nutr Biochem 2019; 71:54-62. [DOI: 10.1016/j.jnutbio.2019.05.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 11/22/2022]
|
40
|
The impact of sugar consumption on stress driven, emotional and addictive behaviors. Neurosci Biobehav Rev 2019; 103:178-199. [DOI: 10.1016/j.neubiorev.2019.05.021] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 03/14/2019] [Accepted: 05/19/2019] [Indexed: 12/20/2022]
|
41
|
Bartley C, Brun T, Oberhauser L, Grimaldi M, Molica F, Kwak BR, Bosco D, Chanson M, Maechler P. Chronic fructose renders pancreatic β-cells hyper-responsive to glucose-stimulated insulin secretion through extracellular ATP signaling. Am J Physiol Endocrinol Metab 2019; 317:E25-E41. [PMID: 30912960 DOI: 10.1152/ajpendo.00456.2018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Fructose is widely used as a sweetener in processed food and is also associated with metabolic disorders, such as obesity. However, the underlying cellular mechanisms remain unclear, in particular, regarding the pancreatic β-cell. Here, we investigated the effects of chronic exposure to fructose on the function of insulinoma cells and isolated mouse and human pancreatic islets. Although fructose per se did not acutely stimulate insulin exocytosis, our data show that chronic fructose rendered rodent and human β-cells hyper-responsive to intermediate physiological glucose concentrations. Fructose exposure reduced intracellular ATP levels without affecting mitochondrial function, induced AMP-activated protein kinase activation, and favored ATP release from the β-cells upon acute glucose stimulation. The resulting increase in extracellular ATP, mediated by pannexin1 (Panx1) channels, activated the calcium-mobilizer P2Y purinergic receptors. Immunodetection revealed the presence of both Panx1 channels and P2Y1 receptors in β-cells. Addition of an ectonucleotidase inhibitor or P2Y1 agonists to naïve β-cells potentiated insulin secretion stimulated by intermediate glucose, mimicking the fructose treatment. Conversely, the P2Y1 antagonist and Panx1 inhibitor reversed the effects of fructose, as confirmed using Panx1-null islets and by the clearance of extracellular ATP by apyrase. These results reveal an important function of ATP signaling in pancreatic β-cells mediating fructose-induced hyper-responsiveness.
Collapse
Affiliation(s)
- Clarissa Bartley
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center , Geneva , Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center , Geneva , Switzerland
| | - Thierry Brun
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center , Geneva , Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center , Geneva , Switzerland
| | - Lucie Oberhauser
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center , Geneva , Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center , Geneva , Switzerland
| | - Mariagrazia Grimaldi
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center , Geneva , Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center , Geneva , Switzerland
| | - Filippo Molica
- Department of Pathology and Immunology, University of Geneva Medical Center , Geneva , Switzerland
| | - Brenda R Kwak
- Department of Pathology and Immunology, University of Geneva Medical Center , Geneva , Switzerland
- Division of Cardiology, University of Geneva Medical Center , Geneva , Switzerland
| | - Domenico Bosco
- Faculty Diabetes Center, University of Geneva Medical Center , Geneva , Switzerland
- Department of Surgery, Cell Isolation and Transplantation Center, Geneva University Hospital , Geneva , Switzerland
| | - Marc Chanson
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center , Geneva , Switzerland
- Department of Pediatrics, Geneva University Hospital , Geneva , Switzerland
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center , Geneva , Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center , Geneva , Switzerland
| |
Collapse
|
42
|
Sato T, Watanabe Y, Nishimura Y, Inoue M, Morita A, Miura S. Acute fructose intake suppresses fasting-induced hepatic gluconeogenesis through the AKT-FoxO1 pathway. Biochem Biophys Rep 2019; 18:100638. [PMID: 31032430 PMCID: PMC6479072 DOI: 10.1016/j.bbrep.2019.100638] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 03/19/2019] [Accepted: 04/08/2019] [Indexed: 11/21/2022] Open
Abstract
Excessive intake of fructose increases lipogenesis in the liver, leading to hepatic lipid accumulation and development of fatty liver disease. Metabolic alterations in the liver due to fructose intake have been reported in many studies, but the effect of fructose administration on hepatic gluconeogenesis is not fully understood. The aim of this study was to evaluate the acute effects of fructose administration on fasting-induced hepatic gluconeogenesis. C57BL/6J mice were administered fructose solution after 14 h of fasting and plasma insulin, glucose, free fatty acids, and ketone bodies were analysed. We also measured phosphorylated AKT and forkhead box O (FoxO) 1 protein levels and gene expression related to gluconeogenesis in the liver. Furthermore, we measured glucose production from pyruvate after fructose administration. Glucose-administered mice were used as controls. Fructose administration enhanced phosphorylation of AKT in the liver, without increase of blood insulin levels. Blood free fatty acids and ketone bodies concentrations were as high as those in the fasting group after fructose administration, suggesting that insulin-induced inhibition of lipolysis did not occur in mice administered with fructose. Fructose also enhanced phosphorylation of FoxO1 and suppressed gluconeogenic gene expression, glucose-6-phosphatase activity, and glucose production from pyruvate. The present study suggests that acute fructose administration suppresses fasting-induced hepatic gluconeogenesis in an insulin-independent manner.
Collapse
Key Words
- AKT
- CREB, cAMP response element binding protein
- ChREBP, carbohydrate response element binding protein
- EDTA, ethylenediaminetetraacetic acid
- FFA, free fatty acid
- FoxO, forkhead box O
- FoxO1
- Fructose
- G6Pase
- G6Pase, glucose-6-phosphatase
- Gluconeogenesis
- Insulin
- PEPCK, phosphoenolpyruvate carboxykinase
- PGC-1α, peroxisome proliferator-activated receptor gamma coactivator-1 alpha
- PI3K, phosphoinositide-3-kinase
- PIP 3, phosphatidylinositol-(3,4,5)-trisphosphate
- SREBP, sterol-regulatory element binding protein
Collapse
Affiliation(s)
- Tomoki Sato
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
- Research Fellow of Japan Society for the Promotion of Science, 5-3-1 Kojimachi, Chiyoda-ku, Tokyo, 102-0083, Japan
| | - Yui Watanabe
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Yuri Nishimura
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Mizuki Inoue
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Akihito Morita
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Shinji Miura
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| |
Collapse
|
43
|
Huerta-Ávila EE, Ramírez-Silva I, Torres-Sánchez LE, Díaz-Benítez CE, Orbe-Orihuela YC, Lagunas-Martínez A, Galván-Portillo M, Flores M, Cruz M, Burguete-García AI. High Relative Abundance of Lactobacillus reuteri and Fructose Intake are Associated with Adiposity and Cardiometabolic Risk Factors in Children from Mexico City. Nutrients 2019; 11:nu11061207. [PMID: 31141963 PMCID: PMC6627236 DOI: 10.3390/nu11061207] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/10/2019] [Accepted: 05/10/2019] [Indexed: 01/04/2023] Open
Abstract
In Mexico, 3 of 10 children are overweight. Fructose intake and relative abundance (RA) of Lactobacillus reuteri (L. reuteri) in the intestinal microbiota are associated with obesity and diabetes in adults, but studies in children are limited. This study evaluates the association between fructose intake and L. reuteri RA with adiposity and cardiometabolic risk markers in Mexican children dietary information, microbiota profiles, adiposity indicators (Body Mass Index, BMI and Waste Circumference, WC), and cardiometabolic markers were analyzed in 1087 children aged 6–12 years. Linear regression and path analysis models were used. High-tertile fructose intake and L. reuteri RA were positively associated with BMI (βTertil 3 vs. Tertil 1= 0.24 (95% CI, 0.04; 0.44) and βT3 vs. T1 = 0.52 (95% CI, 0.32; 0.72)) and WC (βT3 vs. T1 = 2.40 (95% CI, 0.93; 3.83) and βT3 vs. T1 = 3.40 (95% CI, 1.95; 4.90)), respectively. Also, these factors mediated by adiposity were positively correlated with high triglycerides and insulin concentrations and HOMA-IR (p ≤ 0.03) and negatively associated with HDL-C concentration (p < 0.01). High-tertile fructose intake and L. reuteri RA were directly associated with adiposity and indirectly associated though adiposity with metabolic disorders in children. In conclusion, fructose intake and L. reuteri RA were directly associated with adiposity and indirectly associated with metabolic disorders in children, mediated by adiposity.
Collapse
Affiliation(s)
- Eira E Huerta-Ávila
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos 62100, México.
| | - Ivonne Ramírez-Silva
- Centro de Investigación sobre Nutrición y Salud, Instituto Nacional de Salud Pública, Cuernavaca, Morelos 62100, México.
| | - Luisa E Torres-Sánchez
- Centro de Investigación Salud Poblacional, Instituto Nacional de Salud Pública, Cuernavaca, Morelos 62100, México.
| | - Cinthya E Díaz-Benítez
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos 62100, México.
| | - Yaneth C Orbe-Orihuela
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos 62100, México.
| | - Alfredo Lagunas-Martínez
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos 62100, México.
| | - Marcia Galván-Portillo
- Centro de Investigación Salud Poblacional, Instituto Nacional de Salud Pública, Cuernavaca, Morelos 62100, México.
| | - Mario Flores
- Centro de Investigación sobre Nutrición y Salud, Instituto Nacional de Salud Pública, Cuernavaca, Morelos 62100, México.
| | - Miguel Cruz
- Unidad de Investigación Médica en Bioquímica, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Juárez, Ciudad de México, CDMX 06600, México.
| | - Ana I Burguete-García
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos 62100, México.
| |
Collapse
|
44
|
Hung Y, Wijnhoven HAH, Visser M, Verbeke W. Appetite and Protein Intake Strata of Older Adults in the European Union: Socio-Demographic and Health Characteristics, Diet-Related and Physical Activity Behaviours. Nutrients 2019; 11:nu11040777. [PMID: 30987278 PMCID: PMC6521269 DOI: 10.3390/nu11040777] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/24/2019] [Accepted: 03/28/2019] [Indexed: 12/25/2022] Open
Abstract
Considerable efforts have been directed towards stimulating healthy ageing regarding protein intake and malnutrition, yet large-scale consumer studies are scarce and fragmented. This study aims to profile older adults in the European Union (EU) according to appetite (poor/good) and protein intake (lower/higher) strata, and to identify dietary and physical activity behaviours. A survey with older (aged 65 years or above) adults (n = 1825) in five EU countries (Netherlands, United Kingdom, Finland, Spain and Poland) was conducted in June 2017. Four appetite and protein intake strata were identified based on simplified nutritional appetite questionnaire (SNAQ) scores (≤14 versus >14) and the probability of a protein intake below 1.0 g/kg adjusted BW/day (≥0.3 versus <0.3) based on the 14-item Pro55+ screener: "appi"-Poor appetite and lower level of protein intake (12.2%); "APpi"-Good appetite but lower level of protein intake (25.5%); "apPI"-Poor appetite but higher level of protein intake (14.8%); and "APPI"-Good appetite and higher level of protein intake (47.5%). The stratum of older adults with a poor appetite and lower level of protein intake (12.2%) is characterized by a larger share of people aged 70 years or above, living in the UK or Finland, having an education below tertiary level, who reported some or severe financial difficulties, having less knowledge about dietary protein and being fussier about food. This stratum also tends to have a higher risk of malnutrition in general, oral-health related problems, experience more difficulties in mobility and meal preparation, lower confidence in their ability to engage in physical activities in difficult situations, and a lower readiness to follow dietary advice. Two multivariate linear regression models were used to identify the behavioural determinants that might explain the probability of lower protein intake, stratified by appetite status. This study provides an overview and highlights the similarities and differences in the strata profiles. Recommendations for optimal dietary and physical activity strategies to prevent protein malnutrition were derived, discussed and tailored according to older adults' profiles.
Collapse
Affiliation(s)
- Yung Hung
- Department of Agricultural Economics, Ghent University, Coupure links 653, 9000 Ghent, Belgium.
| | - Hanneke A H Wijnhoven
- Department of Health Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam Public Health research institute, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands.
| | - Marjolein Visser
- Department of Health Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam Public Health research institute, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands.
| | - Wim Verbeke
- Department of Agricultural Economics, Ghent University, Coupure links 653, 9000 Ghent, Belgium.
| |
Collapse
|
45
|
Wang J, Matias J, Gilbert ER, Tachibana T, Cline MA. Hypothalamic mechanisms associated with corticotropin-releasing factor-induced anorexia in chicks. Neuropeptides 2019; 74:95-102. [PMID: 30739813 DOI: 10.1016/j.npep.2019.01.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/04/2019] [Accepted: 01/10/2019] [Indexed: 01/07/2023]
Abstract
Central administration of corticotropin-releasing factor (CRF), a 41-amino acid peptide, is associated with potent anorexigenic effects in rodents and chickens. However, the mechanism underlying this effect remains unclear. Hence, the objective of the current study was to elucidate the hypothalamic mechanisms that mediate CRF-induced anorexia in 4 day-old Cobb-500 chicks. After intracerebroventricular (ICV) injection of 0.02 nmol of CRF, CRF-injected chicks ate less than vehicle chicks while no effect on water intake was observed at 30 min post-injection. In subsequent experiments, the hypothalamus samples were processed at 60 min post-injection. The CRF-injected chicks had more c-Fos immunoreactive cells in the arcuate nucleus (ARC), dorsomedial nucleus (DMN), ventromedial hypothalamus (VMH), and paraventricular nucleus (PVN) of the hypothalamus than vehicle-treated chicks. CRF injection was associated with decreased whole hypothalamic mRNA abundance of neuropeptide Y receptor sub-type 1 (NPYR1). In the ARC, CRF-injected chicks expressed more CRF and CRF receptor sub-type 2 (CRFR2) mRNA but less agouti-related peptide (AgRP), NPY, and NPYR1 mRNA than vehicle-injected chicks. CRF-treated chicks expressed greater amounts of CRFR2 and mesotocin mRNA than vehicle chicks in the PVN and VMH, respectively. In the DMN, CRF injection was associated with reduced NPYR1 mRNA. In conclusion, the results provide insights into understanding CRF-induced hypothalamic actions and suggest that the anorexigenic effect of CRF involves increased CRFR2-mediated signaling in the ARC and PVN that overrides the effects of NPY and other orexigenic factors.
Collapse
Affiliation(s)
- Jinxin Wang
- Department of Animal and Poultry Sciences, School of Neuroscience, USA
| | - Justin Matias
- Department of Animal and Poultry Sciences, School of Neuroscience, USA
| | - Elizabeth R Gilbert
- Department of Animal and Poultry Sciences, School of Neuroscience, USA; Virginia Polytechnic Institute and State University, Blacksburg 24061, VA, USA
| | - Tetsuya Tachibana
- Department of Agrobiological Science, Faculty of Agriculture, Ehime University, Matsuyama 790-8566, Japan
| | - Mark A Cline
- Department of Animal and Poultry Sciences, School of Neuroscience, USA; Virginia Polytechnic Institute and State University, Blacksburg 24061, VA, USA.
| |
Collapse
|
46
|
Boskovic M, Bundalo M, Zivkovic M, Stanisic J, Kostic M, Koricanac G, Stankovic A. Estradiol ameliorates antioxidant axis SIRT1-FoxO3a-MnSOD/catalase in the heart of fructose-fed ovariectomized rats. J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.11.053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
47
|
Haghi J, Eteraf-Oskouei T, Najafi M. Effects of Postconditioning with Fructose on Arrhythmias and the Size of Infarct Caused by Global Ischemia and Reperfusion in Isolated Rat Heart. Adv Pharm Bull 2018; 8:57-62. [PMID: 29670839 PMCID: PMC5896396 DOI: 10.15171/apb.2018.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 12/28/2017] [Accepted: 01/01/2018] [Indexed: 12/28/2022] Open
Abstract
Purpose: In the present study, postconditioning effect of fructose against ischemia/reperfusion (I/R)-induced arrhythmias and infarct size were investigated in isolated rat heart. Methods: The isolated hearts were divided into 7 groups, mounted on a Langendorff apparatus at constant pressure then subjected to 30 min zero flow global ischemia followed by 120 min reperfusion. In the control group, normal Krebs–Henseleit (K/H) solution was perfused into the hearts throughout the experiment. In two separate sets of experiments, the treatment groups received 12, 24 and 48 mM of fructose with/without normal glucose in K/H solution for 20 min at the beginning of reperfusion. Cardiac arrhythmias including number of ventricular tachycardia (VT), total ventricular ectopic beats, incidence and duration of VT, reversible and irreversible ventricular fibrillation were recorded and analyzed during the first 30 min of reperfusion. Computerized planimetry method was used to determine volume and percentage of infarct size. Results: Administration of fructose as a postconditioning agent clearly reduced volume and percentage of infarct size in the all treatment groups. The effect was statistically significant especially in the hearts that treated by fructose plus glucose (P<0.05). However, fructose alone or its co-administration with glucose had no significant inhibitory effect against reperfusion arrhythmias. Conclusion: The results showed that perfusion of high concentration of fructose alone or coincident with glucose in globally ischemic-reperfused isolated rat hearts can reduce infarct size without inhibitory effect against reperfusion arrhythmias. Probably, fructose by providing adequate ATP for cardiac functions may inhibit necrosis and death of cardiomyocytes during I/R.
Collapse
Affiliation(s)
- Jila Haghi
- Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tahereh Eteraf-Oskouei
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Moslem Najafi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
48
|
Dietary influences on cognition. Physiol Behav 2018; 192:118-126. [PMID: 29501837 DOI: 10.1016/j.physbeh.2018.02.052] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/27/2018] [Accepted: 02/28/2018] [Indexed: 01/01/2023]
Abstract
Obesity is a world-wide crisis with profound healthcare and socio-economic implications and it is now clear that the central nervous system (CNS) is a target for the complications of metabolic disorders like obesity. In addition to decreases in physical activity and sedentary lifestyles, diet is proposed to be an important contributor to the etiology and progression of obesity. Unfortunately, there are gaps in our knowledge base related to how dietary choices impact the structural and functional integrity of the CNS. For example, while chronic consumption of hypercaloric diets (increased sugars and fat) contribute to increases in body weight and adiposity characteristic of metabolic disorders, the mechanistic basis for neurocognitive deficits in obesity remains to be determined. In addition, studies indicate that acute consumption of hypercaloric diets impairs performance in a wide variety of cognitive domains, even in normal non-obese control subjects. These results from the clinical and basic science literature indicate that diet can have rapid, as well as long lasting effects on cognitive function. This review summarizes our symposium at the 2017 Society for the Study of Ingestive Behavior (SSIB) meeting that discussed these effects of diet on cognition. Collectively, this review highlights the need for integrated and comprehensive approaches to more fully determine how diet impacts behavior and cognition under physiological conditions and in metabolic disorders like type 2 diabetes mellitus (T2DM) and obesity.
Collapse
|
49
|
High fructose diet-induced metabolic syndrome: Pathophysiological mechanism and treatment by traditional Chinese medicine. Pharmacol Res 2018; 130:438-450. [PMID: 29471102 DOI: 10.1016/j.phrs.2018.02.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/09/2018] [Accepted: 02/14/2018] [Indexed: 02/08/2023]
Abstract
Fructose is a natural monosaccharide broadly used in modern society. Over the past few decades, epidemiological studies have demonstrated that high fructose intake is an etiological factor of metabolic syndrome (MetS). This review highlights research advances on fructose-induced MetS, especially the underlying pathophysiological mechanism as well as pharmacotherapy by traditional Chinese medicine (TCM), using the PubMed, Web of science, China National Knowledge Infrastructure, China Science and Technology Journal and Wanfang Data. This review focuses on de novo lipogenesis (DNL) and uric acid (UA) production, two unique features of fructolysis different from glucose glycolysis. High level of DNL and UA production can result in insulin resistance, the key pathological event in developing MetS, mostly through oxidative stress and inflammation. Some other pathologies like the disturbance in brain and gut microbiota in the development of fructose-induced MetS in the past years, are also discussed. In management of MetS, TCM is an excellent representative in alternative and complementary medicine with a complete theory system and substantial herbal remedies. TCMs against MetS or MetS components, including Chinese patent medicines, TCM compound formulas, single TCM herbs and active compounds of TCM herbs, are reviewed on their effects and molecular mechanisms. TCMs with hypouricemic activity, which specially target fructose-induced MetS, are highlighted. And new technologies and strategies (such as high-throughput assay and systems biology) in this field are further discussed. In summary, fructose-induced MetS is a multifactorial disorder with the underlying complex mechanisms. Current clinical and pre-clinical evidence supports the potential of TCMs in management of MetS. Additionally, TCMs may show some advantages against complex MetS as their holistic feature through multiple target actions. However, further work is needed to confirm the effectivity and safety of TCMs by high-standard clinical trials, clarify the molecular mechanisms, and develop new anti-MetS drugs by development and application of optimized and feasible strategies and methods.
Collapse
|
50
|
Batista LO, Ramos VW, Rosas Fernández MA, Concha Vilca CM, Albuquerque KTD. Oral solution of fructose promotes SREBP-1c high-expression in the hypothalamus of Wistar rats. Nutr Neurosci 2018; 22:648-654. [PMID: 29366380 DOI: 10.1080/1028415x.2018.1427659] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Objective: We evaluate whether the consumption of fructose for 8 weeks affects enzymes and transcription factors of the lipogenic and inflammatory pathways in the hypothalamus of Wistar rats. Methods: At 30 days, the animals were divided into groups: Control (C) and Fructose (F) and maintained with free access to feed and filtered water (C) or aqueous solution of purified fructose at 20% (F). RT-PCR and Western blotting were performed for the target genes and proteins. Results: In F group, results showed a lower feed intake, an increase in glycemia (146.20 ± 6.09 vs. 102.32 ± 4.58; n: 9) and triacylglycerol (F: 191.65 ± 13.51 vs. C: 131.69 ± 6.49; n: 9) and there was no difference in water and energy consumption. We identified a higher content of acetyl-CoA carboxylase (ACC) (F: 133.93 ± 5.58 vs. C: 100 ± 0.0; n: 9-10) and NFκB (F: 125.5 ± 8.85 vs. C: 100 ± 0; n: 14) in group F, whereas fatty acid synthase (FAS) was lower (F: 85.90 ± 4.81 vs. C: 100 ± 0.0; n: 4-6). SREBP-1c gene expression was higher in F vs. C group (F: 4.08 ± 0.44 vs. C: 1.13 ± 0.15; n: 5-6), although we did not found difference between groups in the gene expression for ACC, SREBP-2, and NFκB. Discussion: Dietary fructose can change important lipogenic and inflammatory factors in the hypothalamus of rats and it leads to regulation of transcription factors before changes in body mass are evident.
Collapse
Affiliation(s)
- Leandro Oliveira Batista
- a Laboratório de Nutrição Experimental, Universidade Federal do Rio de Janeiro - Campus UFRJ Macaé , Av. Aloísio da Silva Gomes, 50 - Macaé, 27930-560 Rio de Janeiro , Brazil.,b Programa de Pós-graduação em Nutrição, Universidade Federal do Rio de Janeiro , Av. Carlos Chagas Filho, 373, CCS, Bloco J / 2° andar, 21941-902 Rio de Janeiro , Brazil
| | - Viviane Wagner Ramos
- a Laboratório de Nutrição Experimental, Universidade Federal do Rio de Janeiro - Campus UFRJ Macaé , Av. Aloísio da Silva Gomes, 50 - Macaé, 27930-560 Rio de Janeiro , Brazil.,b Programa de Pós-graduação em Nutrição, Universidade Federal do Rio de Janeiro , Av. Carlos Chagas Filho, 373, CCS, Bloco J / 2° andar, 21941-902 Rio de Janeiro , Brazil
| | - Mariana Alejandra Rosas Fernández
- a Laboratório de Nutrição Experimental, Universidade Federal do Rio de Janeiro - Campus UFRJ Macaé , Av. Aloísio da Silva Gomes, 50 - Macaé, 27930-560 Rio de Janeiro , Brazil.,c Programa de Pós-graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro - Campus UFRJ Macaé , Av. Aloísio da Silva Gomes, 50 - Macaé, 27930-560 Rio de Janeiro , Brazil
| | - Carlos Marcelo Concha Vilca
- a Laboratório de Nutrição Experimental, Universidade Federal do Rio de Janeiro - Campus UFRJ Macaé , Av. Aloísio da Silva Gomes, 50 - Macaé, 27930-560 Rio de Janeiro , Brazil.,b Programa de Pós-graduação em Nutrição, Universidade Federal do Rio de Janeiro , Av. Carlos Chagas Filho, 373, CCS, Bloco J / 2° andar, 21941-902 Rio de Janeiro , Brazil
| | - Kelse Tibau de Albuquerque
- a Laboratório de Nutrição Experimental, Universidade Federal do Rio de Janeiro - Campus UFRJ Macaé , Av. Aloísio da Silva Gomes, 50 - Macaé, 27930-560 Rio de Janeiro , Brazil.,b Programa de Pós-graduação em Nutrição, Universidade Federal do Rio de Janeiro , Av. Carlos Chagas Filho, 373, CCS, Bloco J / 2° andar, 21941-902 Rio de Janeiro , Brazil.,c Programa de Pós-graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro - Campus UFRJ Macaé , Av. Aloísio da Silva Gomes, 50 - Macaé, 27930-560 Rio de Janeiro , Brazil
| |
Collapse
|