1
|
Yoo J, Lee J, Ahn B, Han J, Lim MH. Multi-target-directed therapeutic strategies for Alzheimer's disease: controlling amyloid-β aggregation, metal ion homeostasis, and enzyme inhibition. Chem Sci 2025:d4sc06762b. [PMID: 39810997 PMCID: PMC11726323 DOI: 10.1039/d4sc06762b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative dementia, marked by progressive cognitive decline and memory impairment. Despite advances in therapeutic research, single-target-directed treatments often fall short in addressing the complex, multifactorial nature of AD. This arises from various pathological features, including amyloid-β (Aβ) aggregate deposition, metal ion dysregulation, oxidative stress, impaired neurotransmission, neuroinflammation, mitochondrial dysfunction, and neuronal cell death. This review illustrates their interrelationships, with a particular emphasis on the interplay among Aβ, metal ions, and AD-related enzymes, such as β-site amyloid precursor protein cleaving enzyme 1 (BACE1), matrix metalloproteinase 9 (MMP9), lysyl oxidase-like 2 (LOXL2), acetylcholinesterase (AChE), and monoamine oxidase B (MAOB). We further underscore the potential of therapeutic strategies that simultaneously inhibit Aβ aggregation and address other pathogenic mechanisms. These approaches offer a more comprehensive and effective method for combating AD, overcoming the limitations of conventional therapies.
Collapse
Affiliation(s)
- Jeasang Yoo
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Jimin Lee
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Byeongha Ahn
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Jiyeon Han
- Department of Applied Chemistry, University of Seoul Seoul 02504 Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| |
Collapse
|
2
|
Abedi A, Foroutan T, Mohaghegh Shalmani L, Dargahi L. Sex-dependent susceptibility to brain metabolic dysfunction and memory impairment in response to pre and postnatal high-fat diet. J Nutr Biochem 2024; 132:109675. [PMID: 38945454 DOI: 10.1016/j.jnutbio.2024.109675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/15/2024] [Accepted: 05/31/2024] [Indexed: 07/02/2024]
Abstract
The developing brain is sensitive to the impacts of early-life nutritional intake. This study investigates whether maternal high fat diet (HFD) causes glucose metabolism impairment, neuroinflammation, and memory impairment in immature and adult offspring, and whether it may be affected by postweaning diets in a sex-dependent manner in adult offspring. After weaning, female rats were fed HFD (55.9% fat) or normal chow diet (NCD; 10% fat) for 8 weeks before mating, during pregnancy, and lactation. On postnatal day 21 (PND21), the male and female offspring of both groups were split into two new groups, and NCD or HFD feeding was maintained until PND180. On PND21 and PND180, brain glucose metabolism, inflammation, and Alzheimer's pathology-related markers were by qPCR. In adult offspring, peripheral insulin resistance parameters, spatial memory performance, and brain glucose metabolism (18F-FDG-PET scan and protein levels of IDE and GLUT3) were assessed. Histological analysis was also performed on PND21 and adult offspring. On PND21, we found that maternal HFD affected transcript levels of glucose metabolism markers in both sexes. In adult offspring, more profoundly in males, postweaning HFD in combination with maternal HFD induced peripheral and brain metabolic disturbances, impaired memory performance and elevated inflammation, dementia risk markers, and neuronal loss. Our results suggest that maternal HFD affects brain glucose metabolism in the early ages of both sexes. Postweaning HFD sex-dependently causes brain metabolic dysfunction and memory impairment in later-life offspring; effects that can be worsened in combination with maternal HFD.
Collapse
Affiliation(s)
- Azam Abedi
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Tahereh Foroutan
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | - Leila Mohaghegh Shalmani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Lékó AH, Gregory-Flores A, Marchette RCN, Gomez JL, Vendruscolo JCM, Repunte-Canonigo V, Choung V, Deschaine SL, Whiting KE, Jackson SN, Cornejo MP, Perello M, You ZB, Eckhaus M, Rasineni K, Janda KD, Zorman B, Sumazin P, Koob GF, Michaelides M, Sanna PP, Vendruscolo LF, Leggio L. Genetic or pharmacological GHSR blockade has sexually dimorphic effects in rodents on a high-fat diet. Commun Biol 2024; 7:632. [PMID: 38796563 PMCID: PMC11127961 DOI: 10.1038/s42003-024-06303-5] [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: 09/29/2023] [Accepted: 05/08/2024] [Indexed: 05/28/2024] Open
Abstract
The stomach-derived hormone ghrelin regulates essential physiological functions. The ghrelin receptor (GHSR) has ligand-independent actions; therefore, GHSR gene deletion may be a reasonable approach to investigate the role of this system in feeding behaviors and diet-induced obesity (DIO). Here, we investigate the effects of a long-term (12-month) high-fat (HFD) versus regular diet on obesity-related measures in global GHSR-KO and wild-type (WT) Wistar male and female rats. Our main findings are that the GHSR gene deletion protects against DIO and decreases food intake during HFD in male but not in female rats. GHSR gene deletion increases thermogenesis and brain glucose uptake in male rats and modifies the effects of HFD on brain glucose metabolism in a sex-specific manner, as assessed with small animal positron emission tomography. We use RNA-sequencing to show that GHSR-KO rats have upregulated expression of genes responsible for fat oxidation in brown adipose tissue. Central administration of a novel GHSR inverse agonist, PF-5190457, attenuates ghrelin-induced food intake, but only in male, not in female mice. HFD-induced binge-like eating is reduced by inverse agonism in both sexes. Our results support GHSR as a promising target for new pharmacotherapies for obesity.
Collapse
Affiliation(s)
- András H Lékó
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
- Center on Compulsive Behaviors, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Adriana Gregory-Flores
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Renata C N Marchette
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Juan L Gomez
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, Neuroimaging Research Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Janaina C M Vendruscolo
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Vez Repunte-Canonigo
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Vicky Choung
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Sara L Deschaine
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
| | - Kimberly E Whiting
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Shelley N Jackson
- Translational Analytical Core, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Maria Paula Cornejo
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional La Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata, Argentina
| | - Mario Perello
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional La Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata, Argentina
| | - Zhi-Bing You
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Michael Eckhaus
- Pathology Service, Division of Veterinary Resources, Office of Research Services, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Karuna Rasineni
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kim D Janda
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Barry Zorman
- Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Pavel Sumazin
- Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - George F Koob
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, Neuroimaging Research Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Pietro P Sanna
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Leandro F Vendruscolo
- Stress and Addiction Neuroscience Unit, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA.
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA.
- Translational Analytical Core, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA.
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, RI, USA.
- Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
- Department of Neuroscience, Georgetown University Medical Center, Washington DC, USA.
| |
Collapse
|
4
|
Longo M, Paolini E, Meroni M, Jericó D, Córdoba KM, Battistin M, Gatti S, Di Pierro E, Fontanellas A, Dongiovanni P. The Alpha-Lipoic Acid Improves Glucose Metabolism and Hyperinsulinemia in Acute Intermittent Porphyria: A Nutritional Concept for the Management of Rare Disorders. Cell Mol Gastroenterol Hepatol 2023; 17:511-514. [PMID: 37979725 PMCID: PMC10884556 DOI: 10.1016/j.jcmgh.2023.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 11/20/2023]
Affiliation(s)
- Miriam Longo
- Medicine and Metabolic Diseases, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Erika Paolini
- Medicine and Metabolic Diseases, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Marica Meroni
- Medicine and Metabolic Diseases, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniel Jericó
- Hepatology: Porphyrias and Carcinogenesis Laboratory, CIMA-University of Navarra, Pamplona, Spain
| | - Karol M Córdoba
- Hepatology: Porphyrias and Carcinogenesis Laboratory, CIMA-University of Navarra, Pamplona, Spain
| | - Michele Battistin
- Center for Preclinical Research, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano Gatti
- Center for Preclinical Research, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Elena Di Pierro
- Medicine and Metabolic Diseases, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Antonio Fontanellas
- Hepatology: Porphyrias and Carcinogenesis Laboratory, Center for Applied Medical Research-University of Navarra, Pamplona, Spain; Navarra Institute for Health Research, Pamplona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Instituto de Salud Carlos III, Madrid, Spain
| | - Paola Dongiovanni
- Medicine and Metabolic Diseases, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
5
|
Leggio L, Leko A, Gregory-Flores A, Marchette R, Gomez J, Vendruscolo J, Repunte-Canonigo V, Chuong V, Deschaine S, Whiting K, Jackson S, Cornejo M, Perello M, You ZB, Eckhaus M, Janda K, Zorman B, Sumazin P, Koob G, Michaelides M, Sanna PP, Vendruscolo L. Genetic or pharmacological GHSR blockade has sexually dimorphic effects in rodents on a high-fat diet. RESEARCH SQUARE 2023:rs.3.rs-3236045. [PMID: 37886546 PMCID: PMC10602167 DOI: 10.21203/rs.3.rs-3236045/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
The stomach-derived hormone ghrelin regulates essential physiological functions. The ghrelin receptor (GHSR) has ligand-independent actions, therefore, GHSR gene deletion may be a reasonable approach to investigate the role of this system in feeding behaviors and diet-induced obesity (DIO). Here we investigated the effects of a long-term (12 month) high-fat (HFD) versus regular diet on obesity-related measures in global GHSR-KO and wild type (WT) Wistar male and female rats. Our main findings were that the GHSR gene deletion protects against DIO and decreases food intake during HFD in male but not in female rats. GHSR gene deletion increased thermogenesis and brain glucose uptake in male rats and modified the effects of HFD on brain glucose metabolism in a sex-specific manner, as assessed with small animal positron emission tomography. RNA-sequencing was also used to show that GHSR-KO rats had upregulated expression of genes responsible for fat oxidation in brown adipose tissue. Central administration of a novel GHSR inverse agonist, PF-5190457, attenuated ghrelin-induced food intake, but only in male, not in female mice. HFD-induced binge-like eating was reduced by inverse agonism in both sexes. Our results support GHSR as a promising target for new pharmacotherapies for obesity.
Collapse
|
6
|
Altunina N, Bondarchuk O. EFFECTS OF ALPHA-LIPOIC ACID ON GLYCEMIC STATUS IN 2 TYPE DIABETES PATIENTS WITH СHRONIC CORONARY SYNDROME. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2023; 75:3074-3079. [PMID: 36723330 DOI: 10.36740/wlek202212131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The aim: To study the possibilities of alpha-lipoic acid (ALA) to control the parameters of carbohydrate metabolism. PATIENTS AND METHODS Materials and methods: We examined 80 people with type 2 DM and coronary heart disease who suffered non-Q-myocardial infarction (non-Q-MI). All patients at the time of inclusion in the study received oral hypoglycemic agents, ACE inhibitor, β-blocker, statin and antiplatelet agent. 600 mg of ALA per day for 4 months was added to this treatment. After checking the patients for compliance with the criteria, they were divided into the main and experimental groups. The dosage of alpha-lipoic acid was determined for each of the groups. The results of the treatment were analyzed by determining the mean and standard deviations. RESULTS Results: At the end of the observation period, a significant decrease in the level of fasting glucose (FG) by 11.6% was found, which corresponded to the average size of the clinical effect. The values of glycosylated hemoglobin (HbA1c) and the insulin resistance index HOMA (HOMA-IR) showed only a tendency to decrease on the background of treatment. The effect of ALA on postprandial glycemia (PPG) and insulin levels was not detected in this study. CONCLUSION Conclusions: An additional 4-month dose of ALA in addition to baseline therapy showed a moderate effect on the decrease in FG concentration in the absence of significant dynamics in other parameters of glycemic control in the examined patients.
Collapse
|
7
|
Sánchez-Alegría K, Arias C. Functional consequences of brain exposure to saturated fatty acids: From energy metabolism and insulin resistance to neuronal damage. Endocrinol Diabetes Metab 2023; 6:e386. [PMID: 36321333 PMCID: PMC9836261 DOI: 10.1002/edm2.386] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/06/2022] [Accepted: 10/09/2022] [Indexed: 11/06/2022] Open
Abstract
INTRODUCTION Saturated fatty acids (FAs) are the main component of high-fat diets (HFDs), and high consumption has been associated with the development of insulin resistance, endoplasmic reticulum stress and mitochondrial dysfunction in neuronal cells. In particular, the reduction in neuronal insulin signaling seems to underlie the development of cognitive impairments and has been considered a risk factor for Alzheimer's disease (AD). METHODS This review summarized and critically analyzed the research that has impacted the field of saturated FA metabolism in neurons. RESULTS We reviewed the mechanisms for free FA transport from the systemic circulation to the brain and how they impact neuronal metabolism. Finally, we focused on the molecular and the physiopathological consequences of brain exposure to the most abundant FA in the HFD, palmitic acid (PA). CONCLUSION Understanding the mechanisms that lead to metabolic alterations in neurons induced by saturated FAs could help to develop several strategies for the prevention and treatment of cognitive impairment associated with insulin resistance, metabolic syndrome, or type II diabetes.
Collapse
Affiliation(s)
- Karina Sánchez-Alegría
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Clorinda Arias
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
8
|
Zhang W, Wang J, Wang L, Shi R, Chu C, Shi Z, Liu P, Li Y, Liu X, Liu Z. Alternate-day fasting prevents non-alcoholic fatty liver disease and working memory impairment in diet-induced obese mice. J Nutr Biochem 2022; 110:109146. [PMID: 36049672 DOI: 10.1016/j.jnutbio.2022.109146] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 04/28/2022] [Accepted: 08/10/2022] [Indexed: 01/13/2023]
Abstract
Alternate-day fasting (ADF) regimen has been reported to alleviate obesity and insulin resistance. However, the effects of ADF on preventing diet-induced non-alcoholic fatty liver disease (NAFLD) and related cognitive deficits are still elusive. In the present study, a high-fat diet (HFD)-induced obese (DIO) C57BL/6 mouse model was established. Mice were treated with a 4-week long ADF regimen and/or switching the diet to a standard diet. ADF reduced lipid accumulation, activated AMPK/ULK1 signaling, and suppressed the phosphorylation of mTOR. Also, ADF inhibited lipid accumulation and inflammatory responses in the white adipose tissue and down-regulated expressions of PPAR-γ and cytokines. Moreover, ADF improved the working memory and synaptic structure in the DIO mice and upregulated PSD-95 and BDNF in the hippocampus. ADF reduced oxidative stress and microglial over-activation in the CNS. These results suggest that ADF attenuates NAFLD development in the liver of DIO mice, which is related to the mediating effects of ADF on autophagy and energy metabolism. ADF also enhanced cognitive function, which could be partly explained by the down-regulated peripheral inflammatory responses. This study indicates that ADF could be a promising intervention for alleviating NAFLD development and cognitive decline.
Collapse
Affiliation(s)
- Wentong Zhang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Jun Wang
- Department of Digestive Diseases, Xijing Hospital, Xi'an, Shaanxi Province, China
| | - Luanfeng Wang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Renjie Shi
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Chuanqi Chu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Zhiling Shi
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Pujie Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Yitong Li
- Department of Food Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, USA
| | - Xuebo Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Zhigang Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China; Department of Food Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, USA.
| |
Collapse
|
9
|
Acupuncture Interventions for Alzheimer’s Disease and Vascular Cognitive Disorders: A Review of Mechanisms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6080282. [PMID: 36211826 PMCID: PMC9534683 DOI: 10.1155/2022/6080282] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/24/2022] [Accepted: 09/15/2022] [Indexed: 11/23/2022]
Abstract
Cognitive impairment (CI) related to Alzheimer's disease (AD) and vascular cognitive disorders (VCDs) has become a key problem worldwide. Importantly, CI is a neuropsychiatric abnormality mainly characterized by learning and memory impairments. The hippocampus is an important brain region controlling learning and memory. Recent studies have highlighted the effects of acupuncture on memory deficits in AD and VCDs. By reviewing the literature published on this topic in the past five years, the present study intends to summarize the effects of acupuncture on memory impairment in AD and VCDs. Focusing on hippocampal synaptic plasticity, we reviewed the mechanisms underlying the effects of acupuncture on memory impairments through regulation of synaptic proteins, AD characteristic proteins, intestinal microbiota, neuroinflammation, microRNA expression, orexin system, energy metabolism, etc., suggesting that hippocampal synaptic plasticity may be the common as well as the core link underlying the above mechanisms. We also discussed the potential strategies to improve the effect of acupuncture. Additionally, the effects of acupuncture on synaptic plasticity through the regulation of vascular–glia–neuron unit were further discussed.
Collapse
|
10
|
Sifat AE, Nozohouri S, Archie SR, Chowdhury EA, Abbruscato TJ. Brain Energy Metabolism in Ischemic Stroke: Effects of Smoking and Diabetes. Int J Mol Sci 2022; 23:ijms23158512. [PMID: 35955647 PMCID: PMC9369264 DOI: 10.3390/ijms23158512] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 02/06/2023] Open
Abstract
Proper regulation of energy metabolism in the brain is crucial for maintaining brain activity in physiological and different pathophysiological conditions. Ischemic stroke has a complex pathophysiology which includes perturbations in the brain energy metabolism processes which can contribute to worsening of brain injury and stroke outcome. Smoking and diabetes are common risk factors and comorbid conditions for ischemic stroke which have also been associated with disruptions in brain energy metabolism. Simultaneous presence of these conditions may further alter energy metabolism in the brain leading to a poor clinical prognosis after an ischemic stroke event. In this review, we discuss the possible effects of smoking and/or diabetes on brain glucose utilization and mitochondrial energy metabolism which, when present concurrently, may exacerbate energy metabolism in the ischemic brain. More research is needed to investigate brain glucose utilization and mitochondrial oxidative metabolism in ischemic stroke in the presence of smoking and/or diabetes, which would provide further insights on the pathophysiology of these comorbid conditions and facilitate the development of therapeutic interventions.
Collapse
|
11
|
Bai Y, Xin M, Lin J, Xie J, Lin R, Peng Z, Guo J, Bai W. Banana starch intervention ameliorates diabetes-induced mood disorders via modulation of the gut microbiota-brain axis in diabetic rats. FOOD AGR IMMUNOL 2022. [DOI: 10.1080/09540105.2022.2071846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Yongliang Bai
- School of Food Science and Engineering, Foshan University, Foshan, People’s Republic of China
- South China Research and Development Center for Food Safety, Foshan University, Foshan, People’s Republic of China
| | - Meiguo Xin
- School of Food Science and Engineering, Foshan University, Foshan, People’s Republic of China
- South China Research and Development Center for Food Safety, Foshan University, Foshan, People’s Republic of China
| | - Junming Lin
- School of Food Science and Engineering, Foshan University, Foshan, People’s Republic of China
| | - Jing Xie
- School of Food Science and Engineering, Foshan University, Foshan, People’s Republic of China
| | - Roumin Lin
- School of Food Science and Engineering, Foshan University, Foshan, People’s Republic of China
| | - Zhenshan Peng
- School of Food Science and Engineering, Foshan University, Foshan, People’s Republic of China
| | - Jingwen Guo
- School of Food Science and Engineering, Foshan University, Foshan, People’s Republic of China
| | - Weidong Bai
- College of Food Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, People’s Republic of China
| |
Collapse
|
12
|
Zhao Y, Jia M, Chen W, Liu Z. The neuroprotective effects of intermittent fasting on brain aging and neurodegenerative diseases via regulating mitochondrial function. Free Radic Biol Med 2022; 182:206-218. [PMID: 35218914 DOI: 10.1016/j.freeradbiomed.2022.02.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/29/2022] [Accepted: 02/21/2022] [Indexed: 12/11/2022]
Abstract
Intermittent fasting (IF) has been studied for its effects on lifespan and the prevention or delay of age-related diseases upon the regulation of metabolic pathways. Mitochondria participate in key metabolic pathways and play important roles in maintaining intracellular signaling networks that modulate various cellular functions. Mitochondrial dysfunction has been described as an early feature of brain aging and neurodegeneration. Although IF has been shown to prevent brain aging and neurodegeneration, the mechanism is still unclear. This review focuses on the mechanisms by which IF improves mitochondrial function, which plays a central role in brain aging and neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. The cellular and molecular mechanisms of IF in brain aging and neurodegeneration involve activation of adaptive cellular stress responses and signaling- and transcriptional pathways, thereby enhancing mitochondrial function, by promoting energy metabolism and reducing oxidant production.
Collapse
Affiliation(s)
- Yihang Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Mengzhen Jia
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Weixuan Chen
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Zhigang Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China; German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
| |
Collapse
|
13
|
Garcia-Serrano AM, Mohr AA, Philippe J, Skoug C, Spégel P, Duarte JMN. Cognitive Impairment and Metabolite Profile Alterations in the Hippocampus and Cortex of Male and Female Mice Exposed to a Fat and Sugar-Rich Diet are Normalized by Diet Reversal. Aging Dis 2022; 13:267-283. [PMID: 35111373 PMCID: PMC8782561 DOI: 10.14336/ad.2021.0720] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/20/2021] [Indexed: 12/17/2022] Open
Abstract
Diabetes impacts on brain metabolism, structure, and function. Alterations in brain metabolism have been observed in obesity and diabetes models induced by exposure to diets rich in saturated fat and/or sugar and have been linked to memory impairment. However, it remains to be determined whether brain dysfunction induced by obesogenic diets results from permanent brain alterations. We tested the hypothesis that an obesogenic diet (high-fat and high-sucrose diet; HFHSD) causes reversible changes in hippocampus and cortex metabolism and alterations in behavior. Mice were exposed to HFHSD for 24 weeks or for 16 weeks followed by 8 weeks of diet normalization. Development of the metabolic syndrome, changes in behavior, and brain metabolite profiles by magnetic resonance spectroscopy (MRS) were assessed longitudinally. Control mice were fed an ingredient-matched low-fat and low-sugar diet. Mice fed the HFHSD developed obesity, glucose intolerance and insulin resistance, with a more severe phenotype in male than female mice. Relative to controls, both male and female HFHSD-fed mice showed increased anxiety-like behavior, impaired memory in object recognition tasks, but preserved working spatial memory as evaluated by spontaneous alternation in a Y-maze. Alterations in the metabolite profiles were observed both in the hippocampus and cortex but were more distinct in the hippocampus. HFHSD-induced metabolic changes included altered levels of lactate, glutamate, GABA, glutathione, taurine, N-acetylaspartate, total creatine and total choline. Notably, HFHSD-induced metabolic syndrome, anxiety, memory impairment, and brain metabolic alterations recovered upon diet normalization for 8 weeks. In conclusion, cortical and hippocampal derangements induced by long-term HFHSD consumption are reversible rather than being the result of permanent tissue damage.
Collapse
Affiliation(s)
- Alba M Garcia-Serrano
- 1Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.,2Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Adélaïde A Mohr
- 3Institute of Physics, School of Basic Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Juliette Philippe
- 1Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.,2Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Cecilia Skoug
- 1Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.,2Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Peter Spégel
- 4Department of Chemistry, Centre for Analysis and Synthesis, Lund University, Lund Sweden
| | - João M N Duarte
- 1Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.,2Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| |
Collapse
|
14
|
Ko CY, Xu JH, Lo YM, Tu RS, Wu JSB, Huang WC, Shen SC. Alleviative Effect of Alpha-Lipoic Acid on Cognitive Impairment in High-Fat Diet and Streptozotocin-Induced Type 2 Diabetic Rats. Front Aging Neurosci 2021; 13:774477. [PMID: 34867302 PMCID: PMC8633445 DOI: 10.3389/fnagi.2021.774477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 10/21/2021] [Indexed: 02/01/2023] Open
Abstract
Background: The intricate relationship between type 2 diabetes mellitus (T2DM) and Alzheimer’s disease (AD) suggests that insulin is involved in modulating AD-related proteins. Alpha-lipoic acid (ALA) can improve insulin resistance (IR) in diabetic rats. However, the role of ALA in alleviating the cognitive decline of T2DM is not yet clear. This study examined the ameliorative effect of ALA on cognitive impairment, cerebral IR, and synaptic plasticity abnormalities in high-fat diet (HFD) plus streptozotocin (STZ) induced diabetic rats. Methods: The HFD/STZ-induced T2DM male Wistar rats were orally administered with ALA (50, 100, or 200 mg/kg BW) once a day for 13 weeks. Abilities of cognition were measured with a passive avoidance test and Morris water maze. Specimens of blood and brain were collected for biochemical analysis after the rats were sacrificed. Western blotting was used to determine protein expressions in the hippocampus and cortex in the insulin signaling pathways, long-term potentiation (LTP), and synaptic plasticity-related protein expressions. Results: Alpha-lipoic acid improved hyperinsulinemia and the higher levels of free fatty acids of the T2DM rats. Behavioral experiments showed that the administration of ALA improved cognitive impairment in HFD/STZ-induced T2DM rats. ALA ameliorated insulin-related pathway proteins [phosphoinositide 3-kinase (PI3K), phospho-protein kinase B (pAkt)/Akt, and insulin-degrading enzyme (IDE)] and the LTP pathway, as well as synaptic plasticity proteins (calmodulin-dependent protein kinase II, cyclic AMP response element-binding protein, and postsynaptic density protein-95) of the cerebral cortex or hippocampus in HFD/STZ-induced T2DM rats. Conclusion: Our findings suggested that ALA may ameliorate cognition impairment via alleviating cerebral IR improvement and cerebral synaptic plasticity in diabetic rats.
Collapse
Affiliation(s)
- Chih-Yuan Ko
- Department of Clinical Nutrition, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.,Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.,School of Public Health, Fujian Medical University, Fuzhou, China.,Respiratory Medicine Center of Fujian Province, Quanzhou, China
| | - Jian-Hua Xu
- Department of Tumor Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | | | - Rong-Syuan Tu
- Graduate Program of Nutrition Science, National Taiwan Normal University, Taipei City, Taiwan
| | - James Swi-Bea Wu
- Graduate Institute of Food Science and Technology, National Taiwan University, Taipei City, Taiwan
| | - Wen-Chung Huang
- Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan
| | - Szu-Chuan Shen
- Graduate Program of Nutrition Science, National Taiwan Normal University, Taipei City, Taiwan
| |
Collapse
|
15
|
Peng W, Tan C, Mo L, Jiang J, Zhou W, Du J, Zhou X, Liu X, Chen L. Glucose transporter 3 in neuronal glucose metabolism: Health and diseases. Metabolism 2021; 123:154869. [PMID: 34425073 DOI: 10.1016/j.metabol.2021.154869] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 07/22/2021] [Accepted: 08/17/2021] [Indexed: 12/14/2022]
Abstract
Neurons obtain glucose from extracellular environment for energy production mainly depending on glucose transporter 3 (GLUT3). GLUT3 uptakes glucose with high affinity and great transport capacity, and is important for neuronal energy metabolism. This review summarized the role of neuronal GLUT3 in brain metabolism, function and development under both physiological conditions and in diseases, aiming to provide insights into neuronal glucose metabolism and its effect on brain. GLUT3 stabilizes neuronal glucose uptake and utilization, influences brain development and function, and ameliorates aging-related manifestations. Neuronal GLUT3 is regulated by synaptic activity, hormones, nutrition, insulin and insulin-like growth factor 1 in physiological conditions, and is also upregulated by hypoxia-ischemia. GLUT3-related neuronal glucose and energy metabolism is possibly involved in the pathogenesis, pathophysiological mechanism, progression or prognosis of brain diseases, including Alzheimer's disease, Huntington's disease, attention-deficit/hyperactivity disorder and epilepsy. GLUT3 may be a promising therapeutic target of these diseases. This review also briefly discussed the role of other glucose transporters in neuronal glucose metabolism, which work together with GLUT3 to sustain and stabilize glucose and energy supply for neurons. Deficiency in these glucose transporters may also participate in brain diseases, especially GLUT1 and GLUT4.
Collapse
Affiliation(s)
- Wuxue Peng
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Changhong Tan
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lijuan Mo
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jin Jiang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wen Zhou
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Juncong Du
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xuan Zhou
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xi Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Lifen Chen
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
16
|
Jakaria M, Belaidi AA, Bush AI, Ayton S. Ferroptosis as a mechanism of neurodegeneration in Alzheimer's disease. J Neurochem 2021; 159:804-825. [PMID: 34553778 DOI: 10.1111/jnc.15519] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/07/2021] [Accepted: 09/14/2021] [Indexed: 01/19/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia, with complex pathophysiology that is not fully understood. While β-amyloid plaque and neurofibrillary tangles define the pathology of the disease, the mechanism of neurodegeneration is uncertain. Ferroptosis is an iron-mediated programmed cell death mechanism characterised by phospholipid peroxidation that has been observed in clinical AD samples. This review will outline the growing molecular and clinical evidence implicating ferroptosis in the pathogenesis of AD, with implications for disease-modifying therapies.
Collapse
Affiliation(s)
- Md Jakaria
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Abdel Ali Belaidi
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Scott Ayton
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
17
|
Sánchez-Alegría K, Bastián-Eugenio CE, Vaca L, Arias C. Palmitic acid induces insulin resistance by a mechanism associated with energy metabolism and calcium entry in neuronal cells. FASEB J 2021; 35:e21712. [PMID: 34110637 DOI: 10.1096/fj.202100243r] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/27/2021] [Accepted: 05/17/2021] [Indexed: 01/05/2023]
Abstract
Palmitic acid (PA) is a saturated fatty acid whose high consumption has been largely associated with the development of different metabolic alterations, such as insulin resistance, metabolic syndrome, and type 2 diabetes. Particularly in the brain, insulin signaling disruption has been linked to cognitive decline and is considered a risk factor for Alzheimer's disease. Cumulative evidence has demonstrated the participation of PA in the molecular cascade underlying cellular insulin resistance in peripheral tissues, but its role in the development of neuronal insulin resistance and the mechanisms involved are not fully understood. It has generally been accepted that the brain does not utilize fatty acids as a primary energy source, but recent evidence shows that neurons possess the machinery for fatty acid β-oxidation. However, it is still unclear under what conditions neurons use fatty acids as energy substrates and the implications of their oxidative metabolism in modifying insulin-stimulated effects. In the present work, we have found that neurons differentiated from human neuroblastoma MSN exposed to high but nontoxic concentrations of PA generate ATP through mitochondrial metabolism, which is associated with an increase in the cytosolic Ca2+ and diminished insulin signaling in neurons. These findings reveal a novel mechanism by which saturated fatty acids produce Ca2+ entry and insulin resistance that may play a causal role in increasing neuronal vulnerability associated with metabolic diseases.
Collapse
Affiliation(s)
- Karina Sánchez-Alegría
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Carlos Ernesto Bastián-Eugenio
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Luis Vaca
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Clorinda Arias
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
18
|
Zhao T, Ma D, Mulati A, Zhao B, Liu F, Liu X. Development of astaxanthin-loaded layer-by-layer emulsions: physicochemical properties and improvement of LPS-induced neuroinflammation in mice. Food Funct 2021; 12:5333-5350. [PMID: 33977957 DOI: 10.1039/d0fo03018j] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Astaxanthin (AST) has been shown to have neuroprotective effects; however, its bioavailability in vivo is low due to its hydrophobic properties. In this study, lactoferrin (LF) was prepared by heat-treatment at different temperatures, and on this basis, a layer-by-layer self-assembly method was used to construct double-layer emulsions with LF as the inner layer and polysaccharide (beet pectin, BP or carboxymethyl chitosan, CMCS) as the outer layer. Then AST was encapsulated in the emulsions and their physiochemical properties and function were investigated. The results indicated that high temperature heated LF (95 °C) showed a more stable structure than the lower temperature one, and the exposed internal nonpolar groups of LF could give the emulsion an enhanced stability. The rheology results showed that compared with CMCS, the double-layer emulsion formed by BP had a higher viscosity. In addition, the 95 °C LF-AST-BP emulsion showed the best stability among all the bilayer emulsions. The best emulsion was then used as a model drug to investigate its effects on lipopolysaccharide (LPS)-induced neuroinflammation and learning-memory loss in C57BL/6J mice. Through animal behavioral experiments, it was found that dietary supplementation with the AST emulsion could effectively improve the brain cognitive and learning memory impairment caused by inflammation. Transmission electron microscopy, mRNA and western blotting results also illustrated that the AST emulsion could alleviate neuroinflammation caused by LPS. This study provides a feasible scheme for exploring an AST loaded system and may be suitable for food and drug applications.
Collapse
Affiliation(s)
- Tong Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, PR China.
| | | | | | | | | | | |
Collapse
|
19
|
Mi Y, Qi G, Brinton RD, Yin F. Mitochondria-Targeted Therapeutics for Alzheimer's Disease: The Good, the Bad, the Potential. Antioxid Redox Signal 2021; 34:611-630. [PMID: 32143551 PMCID: PMC7891225 DOI: 10.1089/ars.2020.8070] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 03/02/2020] [Indexed: 12/12/2022]
Abstract
Significance: Alzheimer's disease (AD) is the leading cause of dementia. Thus far, 99.6% of clinical trials, including those targeting energy metabolism, have failed to exert disease-modifying efficacy. Altered mitochondrial function and disruption to the brain bioenergetic system have long-been documented as early events during the pathological progression of AD. Recent Advances: While therapeutic approaches that directly promote mitochondrial bioenergetic machinery or eliminate reactive oxygen species have exhibited limited translatability, emerging strategies targeting nonenergetic aspects of mitochondria provide novel therapeutic targets with the potential to modify AD risk and progression. Growing evidence also reveals a critical link between mitochondrial phenotype and neuroinflammation via metabolic reprogramming of glial cells. Critical Issues: Herein, we summarize major classes of mitochondrion-centered AD therapeutic strategies. In addition, the discrepancy in their efficacy when translated from preclinical models to clinical trials is addressed. Key factors that differentiate the responsiveness to bioenergetic interventions, including sex, apolipoprotein E genotype, and cellular diversity in the brain, are discussed. Future Directions: We propose that the future development of mitochondria-targeted AD therapeutics should consider the interactions between bioenergetics and other disease mechanisms, which may require cell-type-specific targeting to distinguish neurons and non-neuronal cells. Moreover, a successful strategy will likely include stratification by metabolic phenotype, which varies by sex and genetic risk profile and dynamically changes throughout the course of disease. As the network of mitochondrial integration expands across intracellular and systems level biology, assessment of intended, the good, versus unintended consequences, the bad, will be required to reach the potential of mitochondrial therapeutics.
Collapse
Affiliation(s)
- Yashi Mi
- Center for Innovation in Brain Science, University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Guoyuan Qi
- Center for Innovation in Brain Science, University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Roberta Diaz Brinton
- Center for Innovation in Brain Science, University of Arizona Health Sciences, Tucson, Arizona, USA
- Department of Pharmacology, College of Medicine Tucson, Tucson, Arizona, USA
- Department of Neurology, College of Medicine Tucson, Tucson, Arizona, USA
- Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, Arizona, USA
| | - Fei Yin
- Center for Innovation in Brain Science, University of Arizona Health Sciences, Tucson, Arizona, USA
- Department of Pharmacology, College of Medicine Tucson, Tucson, Arizona, USA
- Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
20
|
Képes Z, Aranyi C, Forgács A, Nagy F, Kukuts K, Hascsi Z, Esze R, Somodi S, Káplár M, Varga J, Emri M, Garai I. Glucose-level dependent brain hypometabolism in type 2 diabetes mellitus and obesity. Eur J Hybrid Imaging 2021; 5:3. [PMID: 34181137 PMCID: PMC8218076 DOI: 10.1186/s41824-021-00097-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Metabolic syndrome and its individual components lead to wide-ranging consequences, many of which affect the central nervous system. In this study, we compared the [18F]FDG regional brain metabolic pattern of participants with type 2 diabetes mellitus (T2DM) and non-DM obese individuals. METHODS In our prospective study, 51 patients with controlled T2DM (ages 50.6 ± 8.0 years) and 45 non-DM obese participants (ages 52.0 ± 9.6 years) were enrolled. Glucose levels measured before PET/CT examination (pre-PET glucose) as well as laboratory parameters assessing glucose and lipid status were determined. NeuroQ application (NeuroQTM 3.6, Syntermed, Philips) was used to evaluate regional brain metabolic differences. [18F]FDG PET/CT (AnyScan PC, Mediso) scans, estimating brain metabolism, were transformed to MNI152 brain map after T1 registration and used for SPM-based group comparison of brain metabolism corrected for pre-PET glucose, and correlation analysis with laboratory parameters. RESULTS NeuroQ analysis did not reveal significant regional metabolic defects in either group. Voxel-based group comparison revealed significantly (PFWE<0.05) decreased metabolism in the region of the precuneus and in the right superior frontal gyrus (rSFG) in the diabetic group as compared to the obese patients. Data analysis corrected for pre-PET glucose level showed a hypometabolic difference only in the rSFG in T2DM. Voxel-based correlation analysis showed significant negative correlation of the metabolism in the following brain regions with pre-PET glucose in diabetes: precuneus, left posterior orbital gyrus, right calcarine cortex and right orbital part of inferior frontal gyrus; whilst in the obese group only the right rolandic (pericentral) operculum proved to be sensitive to pre-PET glucose level. CONCLUSIONS To our knowledge, this is the first study to perform pre-PET glucose level corrected comparative analysis of brain metabolism in T2DM and obesity. We also examined the pre-PET glucose level dependency of regional cerebral metabolism in the two groups separately. Large-scale future studies are warranted to perform further correlation analysis with the aim of determining the effects of metabolic disturbances on brain metabolism.
Collapse
Affiliation(s)
- Z. Képes
- Faculty of Medicine, Department of Medical Imaging, Division of Nuclear Medicine and Translational Imaging, University of Debrecen, Nagyerdei krt. 98, Debrecen, Hungary
| | - Cs. Aranyi
- Faculty of Medicine, Department of Medical Imaging, Division of Nuclear Medicine and Translational Imaging, University of Debrecen, Nagyerdei krt. 98, Debrecen, Hungary
| | - A. Forgács
- Scanomed Ltd. Nuclear Medicine Centers, Nagyerdei krt. 98, Debrecen, Hungary
| | - F. Nagy
- Scanomed Ltd. Nuclear Medicine Centers, Nagyerdei krt. 98, Debrecen, Hungary
| | - K. Kukuts
- Scanomed Ltd. Nuclear Medicine Centers, Nagyerdei krt. 98, Debrecen, Hungary
| | - Zs. Hascsi
- Scanomed Ltd. Nuclear Medicine Centers, Nagyerdei krt. 98, Debrecen, Hungary
| | - R. Esze
- Faculty of Medicine, Department of Internal Medicine, University of Debrecen, Nagyerdei krt. 98, Debrecen, Hungary
| | - S. Somodi
- Faculty of Medicine, Department of Internal Medicine, University of Debrecen, Nagyerdei krt. 98, Debrecen, Hungary
| | - M. Káplár
- Faculty of Medicine, Department of Internal Medicine, University of Debrecen, Nagyerdei krt. 98, Debrecen, Hungary
| | - J. Varga
- Faculty of Medicine, Department of Medical Imaging, Division of Nuclear Medicine and Translational Imaging, University of Debrecen, Nagyerdei krt. 98, Debrecen, Hungary
| | - M. Emri
- Faculty of Medicine, Department of Medical Imaging, Division of Nuclear Medicine and Translational Imaging, University of Debrecen, Nagyerdei krt. 98, Debrecen, Hungary
| | - I. Garai
- Faculty of Medicine, Department of Medical Imaging, Division of Nuclear Medicine and Translational Imaging, University of Debrecen, Nagyerdei krt. 98, Debrecen, Hungary
- Scanomed Ltd. Nuclear Medicine Centers, Nagyerdei krt. 98, Debrecen, Hungary
| |
Collapse
|
21
|
Koekkoek LL, Unmehopa UA, Eggels L, Kool T, Lamuadni K, Diepenbroek C, Mul JD, Serlie MJ, la Fleur SE. A free-choice high-fat diet modulates the effects of a sucrose bolus on the expression of genes involved in glucose handling in the hypothalamus and nucleus accumbens. Physiol Behav 2020; 222:112936. [PMID: 32417644 DOI: 10.1016/j.physbeh.2020.112936] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 04/22/2020] [Accepted: 04/24/2020] [Indexed: 11/27/2022]
Abstract
The consumption of saturated fat and sucrose can have synergistic effects on the brain that do not occur when either nutrient is consumed by itself. In this study we hypothesize that saturated fat intake modulates glucose handling in the hypothalamus and nucleus accumbens, both brain areas highly involved in the control of food intake. To study this, male Wistar rats were given a free-choice high fat diet (fcHFD) or a control diet for two weeks. During the last seven days rats were given a daily bolus of either a 30% sucrose solution or water. Rats were sacrificed on day eight, 30 minutes after the onset of drinking. mRNA and protein levels of genes involved in glucose handling were assessed in the hypothalamus and nucleus accumbens. We found increased Glut3 and Glut4 mRNA in the hypothalamus of fcHFD-fed rats without an additional effect of the sucrose bolus. In the nucleus accumbens, the sucrose bolus increased Glut3 mRNA and decreased Glut4 mRNA independent of prior diet exposure. The ATP-sensitive potassium channel subunit Kir6.1 in the nucleus accumbens tended to be affected by the synergistic effects of a fcHFD and a sucrose bolus. These data suggest that acute glucose handling in the hypothalamus and nucleus accumbens may be affected by prior high fat exposure.
Collapse
Affiliation(s)
- L L Koekkoek
- Amsterdam University Medical Center, Location AMC, University of Amsterdam, Laboratory of Endocrinology, Dept. Clinical Chemistry, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Meibergdreef 9, Amsterdam, Netherlands; Amsterdam University Medical Center, Location AMC, University of Amsterdam, Dept Endocrinology and Metabolism, Neuroscience Amsterdam, Amsterdam Gastroenterology, Endocrinology and Metabolism, Meibergdreef 9, K2-283, 1105 AZ Amsterdam-Zuidoost, Amsterdam, Netherlands.; Metabolism and Reward Group, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, Amsterdam, Netherlands
| | - U A Unmehopa
- Amsterdam University Medical Center, Location AMC, University of Amsterdam, Laboratory of Endocrinology, Dept. Clinical Chemistry, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Meibergdreef 9, Amsterdam, Netherlands; Amsterdam University Medical Center, Location AMC, University of Amsterdam, Dept Endocrinology and Metabolism, Neuroscience Amsterdam, Amsterdam Gastroenterology, Endocrinology and Metabolism, Meibergdreef 9, K2-283, 1105 AZ Amsterdam-Zuidoost, Amsterdam, Netherlands
| | - L Eggels
- Amsterdam University Medical Center, Location AMC, University of Amsterdam, Laboratory of Endocrinology, Dept. Clinical Chemistry, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Meibergdreef 9, Amsterdam, Netherlands; Amsterdam University Medical Center, Location AMC, University of Amsterdam, Dept Endocrinology and Metabolism, Neuroscience Amsterdam, Amsterdam Gastroenterology, Endocrinology and Metabolism, Meibergdreef 9, K2-283, 1105 AZ Amsterdam-Zuidoost, Amsterdam, Netherlands.; Metabolism and Reward Group, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, Amsterdam, Netherlands
| | - T Kool
- Amsterdam University Medical Center, Location AMC, University of Amsterdam, Laboratory of Endocrinology, Dept. Clinical Chemistry, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Meibergdreef 9, Amsterdam, Netherlands; Amsterdam University Medical Center, Location AMC, University of Amsterdam, Dept Endocrinology and Metabolism, Neuroscience Amsterdam, Amsterdam Gastroenterology, Endocrinology and Metabolism, Meibergdreef 9, K2-283, 1105 AZ Amsterdam-Zuidoost, Amsterdam, Netherlands.; Metabolism and Reward Group, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, Amsterdam, Netherlands
| | - K Lamuadni
- Amsterdam University Medical Center, Location AMC, University of Amsterdam, Laboratory of Endocrinology, Dept. Clinical Chemistry, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Meibergdreef 9, Amsterdam, Netherlands; Amsterdam University Medical Center, Location AMC, University of Amsterdam, Dept Endocrinology and Metabolism, Neuroscience Amsterdam, Amsterdam Gastroenterology, Endocrinology and Metabolism, Meibergdreef 9, K2-283, 1105 AZ Amsterdam-Zuidoost, Amsterdam, Netherlands
| | - C Diepenbroek
- Amsterdam University Medical Center, Location AMC, University of Amsterdam, Laboratory of Endocrinology, Dept. Clinical Chemistry, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Meibergdreef 9, Amsterdam, Netherlands; Amsterdam University Medical Center, Location AMC, University of Amsterdam, Dept Endocrinology and Metabolism, Neuroscience Amsterdam, Amsterdam Gastroenterology, Endocrinology and Metabolism, Meibergdreef 9, K2-283, 1105 AZ Amsterdam-Zuidoost, Amsterdam, Netherlands.; Metabolism and Reward Group, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, Amsterdam, Netherlands
| | - J D Mul
- Amsterdam University Medical Center, Location AMC, University of Amsterdam, Laboratory of Endocrinology, Dept. Clinical Chemistry, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Meibergdreef 9, Amsterdam, Netherlands; Metabolism and Reward Group, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, Amsterdam, Netherlands; Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - M J Serlie
- Amsterdam University Medical Center, Location AMC, University of Amsterdam, Laboratory of Endocrinology, Dept. Clinical Chemistry, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Meibergdreef 9, Amsterdam, Netherlands
| | - S E la Fleur
- Amsterdam University Medical Center, Location AMC, University of Amsterdam, Laboratory of Endocrinology, Dept. Clinical Chemistry, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Meibergdreef 9, Amsterdam, Netherlands; Amsterdam University Medical Center, Location AMC, University of Amsterdam, Dept Endocrinology and Metabolism, Neuroscience Amsterdam, Amsterdam Gastroenterology, Endocrinology and Metabolism, Meibergdreef 9, K2-283, 1105 AZ Amsterdam-Zuidoost, Amsterdam, Netherlands.; Metabolism and Reward Group, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, Amsterdam, Netherlands.
| |
Collapse
|
22
|
Garcia-Serrano AM, Duarte JMN. Brain Metabolism Alterations in Type 2 Diabetes: What Did We Learn From Diet-Induced Diabetes Models? Front Neurosci 2020; 14:229. [PMID: 32265637 PMCID: PMC7101159 DOI: 10.3389/fnins.2020.00229] [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: 11/04/2019] [Accepted: 03/02/2020] [Indexed: 12/27/2022] Open
Abstract
Type 2 diabetes (T2D) is a metabolic disease with impact on brain function through mechanisms that include glucose toxicity, vascular damage and blood–brain barrier (BBB) impairments, mitochondrial dysfunction, oxidative stress, brain insulin resistance, synaptic failure, neuroinflammation, and gliosis. Rodent models have been developed for investigating T2D, and have contributed to our understanding of mechanisms involved in T2D-induced brain dysfunction. Namely, mice or rats exposed to diabetogenic diets that are rich in fat and/or sugar have been widely used since they develop memory impairment, especially in tasks that depend on hippocampal processing. Here we summarize main findings on brain energy metabolism alterations underlying dysfunction of neuronal and glial cells promoted by diet-induced metabolic syndrome that progresses to a T2D phenotype.
Collapse
Affiliation(s)
- Alba M Garcia-Serrano
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - João M N Duarte
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| |
Collapse
|
23
|
Liu Z, Dai X, Zhang H, Shi R, Hui Y, Jin X, Zhang W, Wang L, Wang Q, Wang D, Wang J, Tan X, Ren B, Liu X, Zhao T, Wang J, Pan J, Yuan T, Chu C, Lan L, Yin F, Cadenas E, Shi L, Zhao S, Liu X. Gut microbiota mediates intermittent-fasting alleviation of diabetes-induced cognitive impairment. Nat Commun 2020; 11:855. [PMID: 32071312 PMCID: PMC7029019 DOI: 10.1038/s41467-020-14676-4] [Citation(s) in RCA: 297] [Impact Index Per Article: 59.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 01/27/2020] [Indexed: 12/14/2022] Open
Abstract
Cognitive decline is one of the complications of type 2 diabetes (T2D). Intermittent fasting (IF) is a promising dietary intervention for alleviating T2D symptoms, but its protective effect on diabetes-driven cognitive dysfunction remains elusive. Here, we find that a 28-day IF regimen for diabetic mice improves behavioral impairment via a microbiota-metabolites-brain axis: IF enhances mitochondrial biogenesis and energy metabolism gene expression in hippocampus, re-structures the gut microbiota, and improves microbial metabolites that are related to cognitive function. Moreover, strong connections are observed between IF affected genes, microbiota and metabolites, as assessed by integrative modelling. Removing gut microbiota with antibiotics partly abolishes the neuroprotective effects of IF. Administration of 3-indolepropionic acid, serotonin, short chain fatty acids or tauroursodeoxycholic acid shows a similar effect to IF in terms of improving cognitive function. Together, our study purports the microbiota-metabolites-brain axis as a mechanism that can enable therapeutic strategies against metabolism-implicated cognitive pathophysiologies. Intermittent fasting (IF) has been shown beneficial in reducing metabolic diseases. Here, using a multi-omics approach in a T2D mouse model, the authors report that IF alters the composition of the gut microbiota and improves metabolic phenotypes that correlate with cognitive behavior.
Collapse
Affiliation(s)
- Zhigang Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China.
| | - Xiaoshuang Dai
- BGI Institute of Applied Agriculture, BGI-Shenzhen, Shenzhen, 518120, China
| | - Hongbo Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Renjie Shi
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Yan Hui
- BGI Institute of Applied Agriculture, BGI-Shenzhen, Shenzhen, 518120, China.,Department of Food Science, University of Copenhagen, Copenhagen, 1958, Denmark
| | - Xin Jin
- BGI Institute of Applied Agriculture, BGI-Shenzhen, Shenzhen, 518120, China
| | - Wentong Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Luanfeng Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Qianxu Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Danna Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Jia Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Xintong Tan
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Bo Ren
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Xiaoning Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Tong Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Jiamin Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Junru Pan
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Tian Yuan
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Chuanqi Chu
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Lei Lan
- BGI Institute of Applied Agriculture, BGI-Shenzhen, Shenzhen, 518120, China
| | - Fei Yin
- Center for Innovation in Brain Science and Department of Pharmacology, University of Arizona, Tucson, 85721, AZ, USA
| | - Enrique Cadenas
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, 90089, CA, USA
| | - Lin Shi
- Division Food and Nutrition Science, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, SE-412 96, Sweden.
| | - Shancen Zhao
- BGI Institute of Applied Agriculture, BGI-Shenzhen, Shenzhen, 518120, China.
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
24
|
Samad HA, Konyak YY, Latheef SK, Kumar A, Khan IA, Verma V, Chouhan VS, Verma MR, Maurya VP, Kumar P, Sarkar M, Singh G. Alpha lipoic acid supplementation ameliorates the wrath of simulated tropical heat and humidity stress in male Murrah buffaloes. INTERNATIONAL JOURNAL OF BIOMETEOROLOGY 2019; 63:1331-1346. [PMID: 31280374 DOI: 10.1007/s00484-019-01750-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 05/17/2019] [Accepted: 06/14/2019] [Indexed: 06/09/2023]
Abstract
A supplement which ameliorates temperature-humidity menace in food producing livestock is a prerequisite to develop climate smart agricultural packages. A study was conducted to investigate the heat stress ameliorative efficacy of alpha lipoic acid (ALA) in male Murrah water buffaloes (Bubalus bubalis). Eighteen animals (293.61 ± 4.66Kg Bwt) were randomly allocated into three groups (n = 6); NHSC (non-heat-stressed control), HS (heat-stressed) and HSLA (heat-stressed-supplemented with ALA@32 mg/kg Bwt orally) based on the temperature humidity index (THI) and ALA supplementation. HS and HSLA were exposed to simulated heat challenge in a climatically controlled chamber (40 °C) for 21 consecutive days, 6 h daily. Physiological responses viz. Respiration rate (RR), Pulse rate (PR) and Rectal temperature (RT) were recorded daily before and after heat exposure. Blood samples were collected at the end of heat exposure on days 1, 6, 11, 16, and 21 and on day 28 (7th day post exposure which is considered as recovery) for peripheral blood mononuclear cells (PBMCs) separation, followed by RNA and Protein extraction for Real time quantitative PCR and Western blot analysis respectively, of heat shock proteins (HSPs). Two-way repeated measure ANOVA was performed between groups at different experimental periods. RR (post exposure) in HS and HSLA was significantly higher (P < 0.05) than NHSC from day 1 onwards but HSLA varied significantly from the HS 8th day onwards. Post exposure RT and PR in both HS and HSLA varied (P < 0.05) from NHSC throughout the study; but between HS and HSLA, RT significantly varied on initial 2 days and last 6 days (from days 16 to 21). HSP70 mRNA expression significantly up regulated in high THI groups with respect to the low THI group throughout the experimental period. During chronic stress (days 16 and 21) HSP70 significantly (P < 0.05) increased in HS but not in HSLA (P > 0.05) with respect to NHSC. ALA supplementation up-regulates and sustains (P < 0.05) the expression of HSP90 in HSLA in comparison to the HS and NHSC. HSP105 expression was significantly up-regulated (P < 0.05) in HS on days 16 and 21 (during long-term exposure) but only on day 21 (P < 0.05) in HSLA. HSP70, HSP90, and HSP105 protein expression dynamics were akin to the mRNA transcript data between the study groups. In conclusion, supplementing ALA ameliorates the deleterious effect of heat stress as reflected by improved physiological and cellular responses. ALA supplementation improved cellular antioxidant status and sustained otherwise easily decaying heat shock responses which concertedly hasten the baton change from a limited window of thermo tolerance to long run acclimatization.
Collapse
Affiliation(s)
- H A Samad
- Division of Physiology & Climatology, ICAR-IndianVeterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Y Y Konyak
- Division of Physiology & Climatology, ICAR-IndianVeterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - S K Latheef
- Division of Pathology, ICAR-IndianVeterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - A Kumar
- Division of Physiology & Climatology, ICAR-IndianVeterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - I A Khan
- Dolphin PG Institue of Biomedical & Natural Science, Dehradun, Uttarakhand, India
| | - V Verma
- Division of Physiology & Climatology, ICAR-IndianVeterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - V S Chouhan
- Division of Physiology & Climatology, ICAR-IndianVeterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - M R Verma
- Division of Livestock economics and statistics, ICAR-IndianVeterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - V P Maurya
- Division of Physiology & Climatology, ICAR-IndianVeterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Puneet Kumar
- Division of Physiology & Climatology, ICAR-IndianVeterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - M Sarkar
- Division of Physiology & Climatology, ICAR-IndianVeterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - G Singh
- Division of Physiology & Climatology, ICAR-IndianVeterinary Research Institute, Bareilly, Uttar Pradesh, India.
| |
Collapse
|
25
|
Panzhinskiy E, Bashir R, Bagchi D, Nair S. Effect of Curcumin and α-Lipoic Acid in Attenuating Weight Gain and Adiposity. J Am Coll Nutr 2019; 38:493-498. [PMID: 30620684 DOI: 10.1080/07315724.2018.1557572] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 11/29/2018] [Accepted: 12/05/2018] [Indexed: 12/31/2022]
Abstract
Objective: Obesity is growing at epidemic proportions worldwide. Natural compounds curcumin and α-lipoic acid have been shown to reduce body-weight gain in both preclinical and clinical studies. This study examined the effect of a combination of curcumin and α-lipoic acid on weight gain and adiposity in high-fat-diet (HFD)-fed mice. Methods: C57BL6 mice (7 weeks old) were randomly assigned to receive either HFD (60% fat) or a normal diet (ND, 10% fat) for a 12-week period, following which the mice receiving HFD were further assigned to supplemental curcumin (0.07%), α-lipoic acid (0.2%), or a combination of curcumin and α-lipoic acid formulated into the HFD for a further 12 weeks. Food intake and body mass were determined on a weekly basis. Body fat composition was determined by dual energy X-ray absorptiometry. Results: Treatment with both curcumin and α-lipoic acid significantly reduced body weight gain in HFD-treated mice, and the combination was more effective in attenuating body weight compared to the individual agents. Food intake and caloric intake were significantly lower in the mice that received α-lipoic acid. Percentage body fat and fat mass and lean body mass, which were increased following HFD feeding, were attenuated in the mice receiving curcumin and the combination. Lean mass was also elevated in the mice that were subjected to an HFD, which was unaltered by curcumin or the combination. Conclusions: Taken together, the combination of curcumin and α-lipoic acid exhibits an additive effect in reducing weight gain and adiposity in response to high-fat feeding.
Collapse
Affiliation(s)
- Evgeniy Panzhinskiy
- a Division of Pharmaceutical Sciences, School of Pharmacy, College of Health Sciences, University of Wyoming, College of Health Sciences , Laramie , Wyoming , USA
- b Diabetes Research Group, University of British Columbia, Life Sciences Institute , Vancouver , British Columbia , Canada
| | - Raza Bashir
- c Iovate Health Sciences International Inc ., Oakville , Ontario , Canada
| | - Debasis Bagchi
- d College of Pharmacy, University of Houston , Houston , Texas , USA
| | - Sreejayan Nair
- a Division of Pharmaceutical Sciences, School of Pharmacy, College of Health Sciences, University of Wyoming, College of Health Sciences , Laramie , Wyoming , USA
| |
Collapse
|
26
|
Sharman MJ, Verdile G, Kirubakaran S, Parenti C, Singh A, Watt G, Karl T, Chang D, Li CG, Münch G. Targeting Inflammatory Pathways in Alzheimer's Disease: A Focus on Natural Products and Phytomedicines. CNS Drugs 2019; 33:457-480. [PMID: 30900203 DOI: 10.1007/s40263-019-00619-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Studies of the brains of Alzheimer's disease (AD) patients have revealed key neuropathological features, such as the deposition of aggregates of insoluble amyloid-β (Aβ) peptides and neurofibrillary tangles (NFTs). These pathological protein deposits, including Aβ peptides (which form senile plaques) and hyperphosphorylated tau (which aggregates into NFTs), have been assumed to be 'the cause of AD'. Aβ has been extensively targeted to develop an effective disease-modifying therapy, but with limited clinical success. Emerging therapies are also now targeting further pathological processes in AD, including neuroinflammation. This review focuses on the inflammatory and oxidative stress-related changes that occur in AD, and discusses some emerging anti-inflammatory natural products and phytomedicines. Many of the promising compounds are cytokine-suppressive anti-inflammatory drugs (CSAIDs), which target the proinflammatory AP1 and nuclear factor-κB signalling pathways and inhibit the expression of many proinflammatory cytokines, such as interleukin (IL)-1, IL-6, tumour necrosis factor-α, or nitric oxide produced by inducible nitric oxide synthase. However, many of these phytomedicines have not been tested in rigorous clinical trials in AD patients. It is not yet clear if the active compounds reach an effective concentration in the brain (due to limited bioavailability) or if they can slow down AD progression in long-term trials. The authors suggest that it is crucial for both the pharmacological and complementary medicine industries to conduct and fund those studies to significantly advance the field.
Collapse
Affiliation(s)
- Matthew J Sharman
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Locked Bag 1322, Launceston, TAS, 7250, Australia
| | - Giuseppe Verdile
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, 6102, Australia
| | - Shanmugam Kirubakaran
- Department of Pharmacology, School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia
| | - Cristina Parenti
- Department of Pharmacology, School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia
| | - Ahilya Singh
- Department of Pharmacology, School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia
| | - Georgina Watt
- Department of Behavioural Neuroscience, School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia
| | - Tim Karl
- Department of Behavioural Neuroscience, School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia
| | - Dennis Chang
- NICM Health Research Institute, Western Sydney University, Campbelltown, NSW, 2560, Australia.,School of Science and Health, Western Sydney University, Campbelltown, NSW, 2560, Australia
| | - Chun Guang Li
- NICM Health Research Institute, Western Sydney University, Campbelltown, NSW, 2560, Australia
| | - Gerald Münch
- Department of Pharmacology, School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia. .,NICM Health Research Institute, Western Sydney University, Campbelltown, NSW, 2560, Australia. .,Pharmacology Unit, School of Medicine, Western Sydney University, Locked Bag 1797, Penrith, NSW, 2751, Australia.
| |
Collapse
|
27
|
Brain metabolic and functional alterations in a liver-specific PTEN knockout mouse model. PLoS One 2018; 13:e0204043. [PMID: 30235271 PMCID: PMC6147462 DOI: 10.1371/journal.pone.0204043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 08/31/2018] [Indexed: 01/11/2023] Open
Abstract
Insulin resistance-as observed in aging, diabetes, obesity, and other pathophysiological situations, affects brain function, for insulin signaling is responsible for neuronal glucose transport and control of energy homeostasis and is involved in the regulation of neuronal growth and synaptic plasticity. This study investigates brain metabolism and function in a liver-specific Phosphatase and Tensin Homologue (Pten) knockout mouse model (Liver-PtenKO), a negative regulator of insulin signaling. The Liver-PtenKO mouse model showed an increased flux of glucose into the liver-thus resulting in an overall hypoglycemic and hypoinsulinemic state-and significantly lower hepatic production of the ketone body beta-hydroxybutyrate (as compared with age-matched control mice). The Liver-PtenKO mice exhibited increased brain glucose uptake, improved rate of glycolysis and flux of metabolites in the TCA cycle, and improved synaptic plasticity in the hippocampus. Brain slices from both control- and Liver-PtenKO mice responded to the addition of insulin (in terms of pAKT/AKT levels), thereby neglecting an insulin resistance scenario. This study underscores the significance of insulin signaling in brain bioenergetics and function and helps recognize deficits in diseases associated with insulin resistance.
Collapse
|
28
|
Effects of early-life malnutrition on neurodevelopment and neuropsychiatric disorders and the potential mechanisms. Prog Neuropsychopharmacol Biol Psychiatry 2018; 83:64-75. [PMID: 29287829 DOI: 10.1016/j.pnpbp.2017.12.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 12/21/2017] [Accepted: 12/24/2017] [Indexed: 02/08/2023]
Abstract
Lines of evidence have demonstrated that early-life malnutrition is highly correlated with neurodevelopment and adulthood neuropsychiatric disorders, while some findings are conflicting with each other. In addition, the biological mechanisms are less investigated. We systematically reviewed the evidence linking early-life nutrition status with neurodevelopment and clinical observations in human and animal models. We summarized the effects of special nutritious on neuropsychiatric disorders and explored the underlying potential mechanisms. The further understanding of the biological regulation of early-life nutritional status on neurodevelopment might shed light on precision nutrition at an integrative systems biology framework.
Collapse
|
29
|
Zhao B, Liu H, Wang J, Liu P, Tan X, Ren B, Liu Z, Liu X. Lycopene Supplementation Attenuates Oxidative Stress, Neuroinflammation, and Cognitive Impairment in Aged CD-1 Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:3127-3136. [PMID: 29509007 DOI: 10.1021/acs.jafc.7b05770] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The carotenoid pigment lycopene (LYC) possesses several properties, including antioxidative, anti-inflammatory, and neuroprotective properties. This study examined the effects of dietary supplementation with LYC on age-induced cognitive impairment, and the potential underlying mechanisms. Behavioral tests revealed that chronic LYC supplementation alleviated age-associated memory loss and cognitive defects. Histological and immunofluorescence-staining results indicated that LYC treatment reversed age-associated neuronal damage and synaptic dysfunctions in the brain. Additionally, LYC supplementation decreased age-associated oxidative stress via suppression of malondialdehyde levels, which increased glutathione, catalase, and superoxide dismutase activities and the levels of antioxidant-enzyme mRNAs, including those of heme oxygenase 1 and NAD-(P)-H-quinone oxidoreductase-1. Furthermore, LYC supplementation significantly reduced age-associated neuroinflammation by inhibiting microgliosis (Iba-1) and downregulating related inflammatory mediators. Moreover, LYC lowered the accumulation of Aβ1-42 in the brains of aged CD-1 mice. Therefore, LYC has the potential for use in the treatment of several age-associated chronic diseases.
Collapse
Affiliation(s)
- Beita Zhao
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering , Northwest A&F University , Xinong Road 22 , Yangling 712100 , China
| | - Hua Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering , Northwest A&F University , Xinong Road 22 , Yangling 712100 , China
| | - Jiamin Wang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering , Northwest A&F University , Xinong Road 22 , Yangling 712100 , China
| | - Pujie Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering , Northwest A&F University , Xinong Road 22 , Yangling 712100 , China
| | - Xintong Tan
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering , Northwest A&F University , Xinong Road 22 , Yangling 712100 , China
| | - Bo Ren
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering , Northwest A&F University , Xinong Road 22 , Yangling 712100 , China
| | - Zhigang Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering , Northwest A&F University , Xinong Road 22 , Yangling 712100 , China
| | - Xuebo Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering , Northwest A&F University , Xinong Road 22 , Yangling 712100 , China
| |
Collapse
|
30
|
Wang J, Li L, Wang Z, Cui Y, Tan X, Yuan T, Liu Q, Liu Z, Liu X. Supplementation of lycopene attenuates lipopolysaccharide-induced amyloidogenesis and cognitive impairments via mediating neuroinflammation and oxidative stress. J Nutr Biochem 2018; 56:16-25. [PMID: 29454265 DOI: 10.1016/j.jnutbio.2018.01.009] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/19/2017] [Accepted: 01/16/2018] [Indexed: 01/13/2023]
Abstract
Neuroinflammation is documented to be the major culprit of Alzheimer's disease. Lycopene (LYC), a fat soluble carotenoid, exhibits neuroprotective function in several neurodegenerative disorders. However, the effects of LYC to countering systemic inflammation-induced amyloidogenesis and memory deficiency remain to be elucidated. In current study, 3-month-old male C57BL/6J mice were treated with 0.03% LYC (w/w, mixed into normal chow) for 5 weeks. The mice were then treated by intraperitoneal injection of LPS (0.25mg/kg) for 9 days. It was found that LYC inhibited LPS-induced memory loss by behavior tests including Y-maze test and Morris water test. Meanwhile, LYC prevented LPS-induced accumulation of Aβ, levels of amyloid precursor protein (APP), and suppressed neuronal β-secretase BACE1 and elevated the expressions of α-secretase ADAM10. Furthermore, LYC down-regulated the expression of IBA-1 (a marker of microglia activation), reduced the levels of inflammatory mediators and inhibited oxidative stress in LPS-treated mice. Moreover, LYC suppressed the phosphorylation of MAPKs, NFκB, and activated Nrf2 signaling pathways in LPS-treated BV2 microglial cells. Therefore, our study indicated that LYC could ameliorate LPS-induced neuroinflammation, oxidative stress, amyloidogenesis and cognitive impairments possibly through mediating MAPKs, NFκB and Nrf2 signaling pathways, indicating that LYC might be a nutritional preventive strategy in neuroinflammation-related diseases such as AD.
Collapse
Affiliation(s)
- Jia Wang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Lixia Li
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Zhuo Wang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Yifan Cui
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Xintong Tan
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Tian Yuan
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Qian Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Zhigang Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China.
| | - Xuebo Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China.
| |
Collapse
|