1
|
Xie M, Gu S, Liu Y, Yang H, Wang Y, Yin W, Hong Y, Lu W, He C, Li L, Zhao L, Zhang J, Liu H, Lan T, Li S, Wang Q. 2-Hydroxyisobutyric acid targeted binding to MT-ND3 boosts mitochondrial respiratory chain homeostasis in hippocampus to rescue diabetic cognitive impairment. Redox Biol 2025; 79:103446. [PMID: 39631248 PMCID: PMC11664011 DOI: 10.1016/j.redox.2024.103446] [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: 09/29/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND The prevalence of diabetic cognitive impairment (DCI) is significant, some studies have shown that it is related to mitochondrial respiratory chain homeostasis, but the specific mechanism is not clear. 2-hydroxyisobutyric acid (2-HIBA) is a novel short-chain fatty acid with potential applications in the treatment of metabolic diseases because it can regulate mitochondrial disorders. Our aim was to explore a novel mechanism of action for 2-HIBA in the treatment of DCI in mitochondrial respiratory chain homeostasis. METHODS Metabolic substances and differentially active metabolic pathways in the serum of diseased mice were identified based on multi-omics analysis. The nanoLC-Obitrap-MS technology was utilized to detect the content of selected small molecules with differential metabolic activity in the hippocampus and mitochondria of mice to evaluate their permeability through the blood-brain barrier (BBB) and outer mitochondrial membrane. A combination of behavioral, proteomic, and molecular biology approaches was used to explore specific regulatory mechanisms and identify potential pharmacological targets. Additionally, using techniques such as protein thermal shift, drug affinity responsive target stability (DARTS), hydrolase stability, and surface plasmon resonance (SPR) experiments, we demonstrated the direct binding effects of small molecule metabolites with protein targets. RESULTS 2-HIBA was found to directly ameliorate cognitive dysfunction in db/db mice by penetrating the blood-brain barrier and reversing the decrease in the protein content of NADH dehydrogenase 3 (MT-ND3) in the hippocampus through direct binding to ND3. This action helps maintain the stability of NAD+/NADH and regulate the mitochondrial respiratory chain balance. Furthermore, a combined medication plant agonist of 2-HIBA can enhance the expression of MT-ND3, thereby improving cognitive dysfunction in mice. CONCLUSION MT-ND3 is a crucial target for improving diabetic cognitive dysfunction, and 2-HIBA can directly bind to the MT-ND3 protein to alleviate the functional impairment of the mitochondrial respiratory chain in mice to treat DCI.
Collapse
Affiliation(s)
- Minzhen Xie
- Department of Medicinal Chemistry and Natural Medicinal Chemistry, College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Nangang District, Harbin City, Heilongjiang Province, 150081, China; State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Siqi Gu
- Department of Medicinal Chemistry and Natural Medicinal Chemistry, College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Nangang District, Harbin City, Heilongjiang Province, 150081, China
| | - Yan Liu
- Key Laboratory of Chinese Materia Medica, Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Xiangfang District, Harbin City, Heilongjiang Province, 150040, China
| | - Haolin Yang
- Beijing University of Chinese Medicine, Beijing, 100000, China
| | - Yuqi Wang
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Nangang District, Harbin City, Heilongjiang Province, 150081, China
| | - Wei Yin
- Department of Medicinal Chemistry and Natural Medicinal Chemistry, College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Nangang District, Harbin City, Heilongjiang Province, 150081, China
| | - Yang Hong
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Wanying Lu
- Department of Medicinal Chemistry and Natural Medicinal Chemistry, College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Nangang District, Harbin City, Heilongjiang Province, 150081, China
| | - Chengbing He
- Department of Medicinal Chemistry and Natural Medicinal Chemistry, College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Nangang District, Harbin City, Heilongjiang Province, 150081, China
| | - Lin Li
- Department of Medicinal Chemistry and Natural Medicinal Chemistry, College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Nangang District, Harbin City, Heilongjiang Province, 150081, China
| | - Limin Zhao
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Nangang District, Harbin City, Heilongjiang Province, 150081, China
| | - Jianjia Zhang
- Key Laboratory of Chinese Materia Medica, Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Xiangfang District, Harbin City, Heilongjiang Province, 150040, China
| | - Heng Liu
- Department of Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Nangang District, Harbin City, Heilongjiang Province, 150081, China.
| | - Tian Lan
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China; Department of Pharmacology, College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Nangang District, Harbin City, Heilongjiang Province, 150081, China.
| | - Shuijie Li
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Nangang District, Harbin City, Heilongjiang Province, 150081, China.
| | - Qi Wang
- Department of Medicinal Chemistry and Natural Medicinal Chemistry, College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Nangang District, Harbin City, Heilongjiang Province, 150081, China.
| |
Collapse
|
2
|
Liang Z, Deng L, Zhou X, Zhang Z, Zhao W. Comprehensive Overview of Ketone Bodies in Cancer Metabolism: Mechanisms and Application. Biomedicines 2025; 13:210. [PMID: 39857793 PMCID: PMC11760447 DOI: 10.3390/biomedicines13010210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Reprogramming energy metabolism is pivotal to tumor development. Ketone bodies (KBs), which are generated during lipid metabolism, are fundamental bioactive molecules that can be modulated to satisfy the escalating metabolic needs of cancer cells. At present, a burgeoning body of research is concentrating on the metabolism of KBs within tumors, investigating their roles as signaling mediators, drivers of post-translational modifications, and regulators of inflammation and oxidative stress. The ketogenic diet (KD) may enhance the sensitivity of various cancers to standard therapies, such as chemotherapy and radiotherapy, by exploiting the reprogrammed metabolism of cancer cells and shifting the metabolic state from glucose reliance to KB utilization, rendering it a promising candidate for adjunct cancer therapy. Nonetheless, numerous questions remain regarding the expression of key metabolic genes across different tumors, the regulation of their activities, and the impact of individual KBs on various tumor types. Further investigation is imperative to resolve the conflicting data concerning KB synthesis and functionality within tumors. This review aims to encapsulate the intricate roles of KBs in cancer metabolism, elucidating a comprehensive grasp of their mechanisms and highlighting emerging clinical applications, thereby setting the stage for future investigations into their therapeutic potential.
Collapse
Affiliation(s)
- Ziyuan Liang
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment, Guangxi Medical University, Ministry of Education, Nanning 530021, China; (Z.L.); (L.D.); (X.Z.)
- Life Science Institute, Guangxi Medical University, Nanning 530021, China
| | - Lixian Deng
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment, Guangxi Medical University, Ministry of Education, Nanning 530021, China; (Z.L.); (L.D.); (X.Z.)
- Life Science Institute, Guangxi Medical University, Nanning 530021, China
| | - Xiaoying Zhou
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment, Guangxi Medical University, Ministry of Education, Nanning 530021, China; (Z.L.); (L.D.); (X.Z.)
- Life Science Institute, Guangxi Medical University, Nanning 530021, China
| | - Zhe Zhang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Weilin Zhao
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
3
|
Wang J, Zhang Y, Chen L, Liu X. Reconstructing Molecular Networks by Causal Diffusion Do-Calculus Analysis with Deep Learning. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2409170. [PMID: 39440482 PMCID: PMC11633463 DOI: 10.1002/advs.202409170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/01/2024] [Indexed: 10/25/2024]
Abstract
Quantifying molecular regulations between genes/molecules causally from observed data is crucial for elucidating the molecular mechanisms underlying biological processes at the network level. Presently, most methods for inferring gene regulatory and biological networks rely on association studies or observational causal-analysis approaches. This study introduces a novel approach that combines intervention operations and diffusion models within a do-calculus framework by deep learning, i.e., Causal Diffusion Do-calculus (CDD) analysis, to infer causal networks between molecules. CDD can extract causal relations from observed data owing to its intervention operations, thereby significantly enhancing the accuracy and generalizability of causal network inference. Computationally, CDD has been applied to both simulated data and real omics data, which demonstrates that CDD outperforms existing methods in accurately inferring gene regulatory networks and identifying causal links from genes to disease phenotypes. Especially, compared with the Mendelian randomization algorithm and other existing methods, the CDD can reliably identify the disease genes or molecules for complex diseases with better performances. In addition, the causal analysis between various diseases and the potential factors in different populations from the UK Biobank database is also conducted, which further validated the effectiveness of CDD.
Collapse
Affiliation(s)
- Jiachen Wang
- Key Laboratory of Systems Health Science of Zhejiang ProvinceSchool of Life ScienceHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhou310024China
| | - Yuelei Zhang
- Key Laboratory of Systems Health Science of Zhejiang ProvinceSchool of Life ScienceHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhou310024China
| | - Luonan Chen
- Key Laboratory of Systems Health Science of Zhejiang ProvinceSchool of Life ScienceHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhou310024China
- Key Laboratory of Systems BiologyShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghai200031China
| | - Xiaoping Liu
- Key Laboratory of Systems Health Science of Zhejiang ProvinceSchool of Life ScienceHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhou310024China
| |
Collapse
|
4
|
Liang R, Li Y, Li J, Zhang S, Gao Y, Tan F, Feng Y, Chen Y, Wang F, Jiang T, Kang X. Metabolomic Profiling of Cerebrospinal Fluid Reveals Metabolite Biomarkers in Tick-Borne Encephalitis Patient. J Med Virol 2024; 96:e70082. [PMID: 39569456 PMCID: PMC11579828 DOI: 10.1002/jmv.70082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/20/2024] [Accepted: 11/06/2024] [Indexed: 11/22/2024]
Abstract
Tick-borne encephalitis virus (TBEV) can cause life-threatening central nervous system infection. Changes in cerebrospinal fluid (CSF) metabolites may reflect critical aspects of host responses and end-organ damage in neuro infection and neuroinflammation. In this study, we applied an untargeted metabolomics screen of CSF samples to investigate the metabolites profile and explore biomarkers for TBEV infection. By analyzing CSF samples from 77 patients with TBEV infection and 23 without TBEV infection, tryptophan metabolism and Citrate cycle were found to be the top important metabolic pathways in differentiating the control and case groups; acetoacetate, 5'-deoxy-5'-(methylthio)-adenosine, 3-methyl-2-oxobutanoic acid, and so forth. were identified to be metabolic biomarkers (|log 2 ${\mathrm{log}}_{2}$ FC|> 1, VIP > 1, FDR < 0.05) in CSF and clearly separated the TBEV infection from the noninfected samples. Moreover, four metabolites were identified to be associated with fatal outcome, including kynurenic acid, 5-hydroxyindole-3-acetic acid, DL-tryptophan, indole-3-acrylic acid, demonstrating the potential predictive biomarkers for severe TBEV infection. This study explored the metabolic profile of TBEV infection in CSF samples and identified candidate biomarkers for TBEV infection, which might be useful in target screening for differential diagnosis and therapeutic inter-vention.
Collapse
Affiliation(s)
- Runxin Liang
- State Key Laboratory of Pathogen and BiosecurityThe Academy of Military Medical ScienceBeijingChina
| | - Yuchang Li
- State Key Laboratory of Pathogen and BiosecurityThe Academy of Military Medical ScienceBeijingChina
| | - Jing Li
- State Key Laboratory of Pathogen and BiosecurityThe Academy of Military Medical ScienceBeijingChina
| | - Sen Zhang
- State Key Laboratory of Pathogen and BiosecurityThe Academy of Military Medical ScienceBeijingChina
| | - Yanhong Gao
- Laboratory Department of the First Medical CenterChinese PLA General HospitalBeijingChina
| | - Fuli Tan
- State Key Laboratory of Pathogen and BiosecurityThe Academy of Military Medical ScienceBeijingChina
| | - Ye Feng
- State Key Laboratory of Pathogen and BiosecurityThe Academy of Military Medical ScienceBeijingChina
| | - Yuehong Chen
- State Key Laboratory of Pathogen and BiosecurityThe Academy of Military Medical ScienceBeijingChina
| | - Fei Wang
- State Key Laboratory of Pathogen and BiosecurityThe Academy of Military Medical ScienceBeijingChina
| | - Tao Jiang
- State Key Laboratory of Pathogen and BiosecurityThe Academy of Military Medical ScienceBeijingChina
| | - Xiaoping Kang
- State Key Laboratory of Pathogen and BiosecurityThe Academy of Military Medical ScienceBeijingChina
| |
Collapse
|
5
|
Malinowska D, Żendzian-Piotrowska M. Ketogenic Diet: A Review of Composition Diversity, Mechanism of Action and Clinical Application. J Nutr Metab 2024; 2024:6666171. [PMID: 39463845 PMCID: PMC11511599 DOI: 10.1155/2024/6666171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 05/16/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024] Open
Abstract
The ketogenic diet (KD) is a special high-fat, very low-carbohydrate diet with the amount of protein adjusted to one's requirements. By lowering the supply of carbohydrates, this diet induces a considerable change in metabolism (of protein and fat) and increases the production of ketone bodies. The purpose of this article is to review the diversity of composition, mechanism of action, clinical application and risk associated with the KD. In the last decade, more and more results of the diet's effects on obesity, diabetes and neurological disorders, among other examples have appeared. The beneficial effects of the KD on neurological diseases are related to the reconstruction of myelin sheaths of neurons, reduction of neuron inflammation, decreased production of reactive oxygen species, support of dopamine production, repair of damaged mitochondria and formation of new ones. Minimizing the intake of carbohydrates results in the reduced absorption of simple sugars, thereby decreasing blood glucose levels and fluctuations of glycaemia in diabetes. Studies on obesity indicate an advantage of the KD over other diets in terms of weight loss. This may be due to the upregulation of the biological activity of appetite-controlling hormones, or to decreased lipogenesis, intensified lipolysis and increased metabolic costs of gluconeogenesis. However, it is important to be aware of the side effects of the KD. These include disorders of the digestive system as well as headaches, irritability, fatigue, the occurrence of vitamin and mineral deficiencies and worsened lipid profile. Further studies aimed to determine long-term effects of the KD are required.
Collapse
Affiliation(s)
- Dominika Malinowska
- Medical University of Bialystok, Department of Hygiene, Epidemiology and Ergonomy, ul. Jana Kilińskiego 1, Białystok 15-089, Poland
| | - Małgorzata Żendzian-Piotrowska
- Medical University of Bialystok, Department of Hygiene, Epidemiology and Ergonomy, ul. Jana Kilińskiego 1, Białystok 15-089, Poland
| |
Collapse
|
6
|
Chen Y, Lin J, Tao M. Association between cheese and fish consumption and the occurrence of depression based on European population: mediating role of metabolites. Front Nutr 2024; 11:1322254. [PMID: 38694223 PMCID: PMC11061354 DOI: 10.3389/fnut.2024.1322254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 03/28/2024] [Indexed: 05/04/2024] Open
Abstract
Background The consumption of cheese and fish has been linked to the onset of depression. However, the connection between consuming cheese, consuming fish, experiencing depression, and the pathways that mediate this relationship remains unclear. The purpose of this research was to investigate the potential association between the consumption of cheese and fish and the occurrence of depression. Moreover, it is important to identify any metabolites that might be involved and understand their respective roles and functions. Methods A two-step, two-sample Mendelian randomization (MR) study was conducted using genome-wide association study (GWAS) data on cheese, non-oily fish, and oily fish consumption and depression, along with 12 alternate mediators. The study included a total of 451,486 participants in the cheese consumption group, 460,880 in the non-oily fish consumption group, 460,443 in the oily fish consumption group, and 322,580 with a diagnosis of depression. The single nucleotide polymorphism (SNP) estimates were pooled using inverse-variance weighted, weighted median, MR-Egger, simple mode, and weighted mode. Results The data we collected suggested that consuming more cheese correlated with a lower likelihood of experiencing depression (OR: 0.95; 95% CI: 0.92 to 0.98). Neither non-oily fish nor oily fish consumption was directly linked to depression onset (p = 0.08, p = 0.78, respectively). Although there was a direct causal relationship with depression, the mediating relationship of triglycerides (TG), total cholesterol in large HDL, cholesterol to total lipids ratio in large HDL, free cholesterol to total lipids ratio in large HDL, glycine, and phospholipids to total lipids ratio in very large HDL of cheese intake on depression risk were - 0.002 (95% CI: -0.023 - 0.020), -0.002 (95% CI: -0.049 - 0.045), -0.001 (95% CI: -0.033 - 0.031), -0.001 (95% CI: -0.018 - 0.015), 0.001 (95% CI: -0.035 - 0.037), and - 0.001 (95% CI: -0.024 - 0.021), respectively. The mediating relationship of uridine, free cholesterol to total lipids ratio in large HDL, total cholesterol in large HDL, acetoacetate, and 3-hydroxybutyrate (3-HB) between non-oily fish consumption and depression risk were 0.016 (95% CI: -0.008 - 0.040), 0.011 (95% CI: -1.269 - 1.290), 0.010 (95% CI: -1.316 - 1.335), 0.011 (95% CI: -0.089 - 0.110), and 0.008 (95% CI: -0.051 - 0.068), respectively. The mediation effect of uridine and free cholesterol to total lipids ratio in large HDL between intake of oily fish and the risk of depression was found to be 0.006 (95% CI: -0.015 - 0.028) and - 0.002 (95% CI: -0.020 - 0.017), respectively. The correlation between eating cheese and experiencing depression persisted even when adjusting for other variables like Indian snacks, mango consumption, sushi consumption, and unsalted peanuts using multivariable MR. Conclusion The consumption of cheese and fish influenced the likelihood of experiencing depression, and this may be mediated by certain metabolites in the body. Our study provided a new perspective on the clinical treatment of depression.
Collapse
Affiliation(s)
- Yan Chen
- Second Clinical Medical School, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jixin Lin
- Second Clinical Medical School, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ming Tao
- Second Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
7
|
Han K, Meadows AM, Rodman MJ, Russo AC, Sharma R, Singh K, Hassanzadeh S, Dagur PK, Huffstutler RD, Krause FN, Griffin JL, Baumer Y, Powell-Wiley TM, Sack MN. Propionate functions as a feeding state-dependent regulatory metabolite to counter proinflammatory signaling linked to nutrient load and obesity. J Leukoc Biol 2024; 115:738-749. [PMID: 38207130 PMCID: PMC10980352 DOI: 10.1093/jleuko/qiae006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 12/18/2023] [Accepted: 12/22/2023] [Indexed: 01/13/2024] Open
Abstract
Generally, fasting and refeeding confer anti- and proinflammatory effects, respectively. In humans, these caloric-load interventions function, in part, via regulation of CD4+ T cell biology. However, mechanisms orchestrating this regulation remain incomplete. We employed integrative bioinformatics of RNA sequencing and high-performance liquid chromatography-mass spectrometry data to measure serum metabolites and gene expression of peripheral blood mononuclear cells isolated from fasting and refeeding in volunteers to identify nutrient-load metabolite-driven immunoregulation. Propionate, a short chain fatty acid (SCFA), and the SCFA-sensing G protein-coupled receptor 43 (ffar2) were coordinately and inversely regulated by fasting and refeeding. Propionate and free fatty acid receptor agonists decreased interferon-γ and interleukin-17 and significantly blunted histone deacetylase activity in CD4+ T cells. Furthermore, propionate blunted nuclear factor κB activity and diminished interleukin-6 release. In parallel, propionate reduced phosphorylation of canonical T helper 1 (TH1) and TH17 regulators, STAT1 and STAT3, respectively. Conversely, knockdown of free fatty acid receptors significantly attenuated the anti-inflammatory role of propionate. Interestingly, propionate recapitulated the blunting of CD4+ TH cell activation in primary cells from obese individuals, extending the role of this metabolite to a disease associated with low-grade inflammation. Together, these data identify a nutrient-load responsive SCFA-G protein-coupled receptor linked pathway to regulate CD4+ TH cell immune responsiveness.
Collapse
Affiliation(s)
- Kim Han
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Room 5-3342, Bld 10-CRC, 10 Center Drive, Bethesda, MD 20817, United States
| | - Allison M Meadows
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Room 5-3342, Bld 10-CRC, 10 Center Drive, Bethesda, MD 20817, United States
- Department of Biochemistry, University of Cambridge, Sanger Bld, 80 Tennis Ct Rd, Cambridge CB2 1GA, United Kingdom
| | - Matthew J Rodman
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Room 5-3342, Bld 10-CRC, 10 Center Drive, Bethesda, MD 20817, United States
| | - Anna Chiara Russo
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Room 5-3342, Bld 10-CRC, 10 Center Drive, Bethesda, MD 20817, United States
| | - Rahul Sharma
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Room 5-3342, Bld 10-CRC, 10 Center Drive, Bethesda, MD 20817, United States
| | - Komudi Singh
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Room 5-3342, Bld 10-CRC, 10 Center Drive, Bethesda, MD 20817, United States
| | - Shahin Hassanzadeh
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Room 5-3342, Bld 10-CRC, 10 Center Drive, Bethesda, MD 20817, United States
| | - Pradeep K Dagur
- Flow Cytometry Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, United States
| | - Rebecca D Huffstutler
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, United States
| | - Fynn N Krause
- Department of Biochemistry, University of Cambridge, Sanger Bld, 80 Tennis Ct Rd, Cambridge CB2 1GA, United Kingdom
| | - Julian L Griffin
- Department of Biochemistry, University of Cambridge, Sanger Bld, 80 Tennis Ct Rd, Cambridge CB2 1GA, United Kingdom
- Rowett Institute, School of Medicine, Medical Sciences and Nutrition, Foresterhill Campus, University of Aberdeen, Ashgrove Rd W, Aberdeen AB25 2ZD, United Kingdom
| | - Yvonne Baumer
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, United States
| | - Tiffany M Powell-Wiley
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, United States
| | - Michael N Sack
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Room 5-3342, Bld 10-CRC, 10 Center Drive, Bethesda, MD 20817, United States
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, United States
| |
Collapse
|
8
|
Al-Kuraishy HM, Jabir MS, Albuhadily AK, Al-Gareeb AI, Jawad SF, Swelum AA, Hadi NR. Role of ketogenic diet in neurodegenerative diseases focusing on Alzheimer diseases: The guardian angle. Ageing Res Rev 2024; 95:102233. [PMID: 38360180 DOI: 10.1016/j.arr.2024.102233] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/11/2024] [Accepted: 02/12/2024] [Indexed: 02/17/2024]
Abstract
The ketogenic diet (KD) is a low-carbohydrate, adequate protein and high-fat diet. KD is primarily used to treat refractory epilepsy. KD was shown to be effective in treating different neurodegenerative diseases. Alzheimer disease (AD) is the first common neurodegenerative disease in the world characterized by memory and cognitive impairment. However, the underlying mechanism of KD in controlling of AD and other neurodegenerative diseases are not discussed widely. Therefore, this review aims to revise the fundamental mechanism of KD in different neurodegenerative diseases focusing on the AD. KD induces a fasting-like which modulates the central and peripheral metabolism by regulating mitochondrial dysfunction, oxidative stress, inflammation, gut-flora, and autophagy in different neurodegenerative diseases. Different studies highlighted that KD improves AD neuropathology by regulating synaptic neurotransmission and inhibiting of neuroinflammation and oxidative stress. In conclusion, KD improves cognitive function and attenuates the progression of AD neuropathology by reducing oxidative stress, mitochondrial dysfunction, and enhancing neuronal autophagy and brain BDNF.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq.
| | - Majid S Jabir
- Department of Applied Science, University of Technology Iraq.
| | - Ali K Albuhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq.
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq; Jabir Ibn Hayyan Medical University, Al-Ameer Qu./Najaf-iraq, PO.Box13, Kufa, Iraq.
| | - Sabrean F Jawad
- Department of Pharmacy, Al-Mustaqbal University College, Hillah, Babylon, 51001, Iraq.
| | - Ayman A Swelum
- Department of Animal Production, King Saud University, Riyadh, Saudi Arabia.
| | | |
Collapse
|
9
|
Ramezani M, Fernando M, Eslick S, Asih PR, Shadfar S, Bandara EMS, Hillebrandt H, Meghwar S, Shahriari M, Chatterjee P, Thota R, Dias CB, Garg ML, Martins RN. Ketone bodies mediate alterations in brain energy metabolism and biomarkers of Alzheimer's disease. Front Neurosci 2023; 17:1297984. [PMID: 38033541 PMCID: PMC10687427 DOI: 10.3389/fnins.2023.1297984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. AD is a progressive neurodegenerative disorder characterized by cognitive dysfunction, including learning and memory deficits, and behavioral changes. Neuropathology hallmarks of AD such as amyloid beta (Aβ) plaques and neurofibrillary tangles containing the neuron-specific protein tau is associated with changes in fluid biomarkers including Aβ, phosphorylated tau (p-tau)-181, p-tau 231, p-tau 217, glial fibrillary acidic protein (GFAP), and neurofilament light (NFL). Another pathological feature of AD is neural damage and hyperactivation of astrocytes, that can cause increased pro-inflammatory mediators and oxidative stress. In addition, reduced brain glucose metabolism and mitochondrial dysfunction appears up to 15 years before the onset of clinical AD symptoms. As glucose utilization is compromised in the brain of patients with AD, ketone bodies (KBs) may serve as an alternative source of energy. KBs are generated from the β-oxidation of fatty acids, which are enhanced following consumption of ketogenic diets with high fat, moderate protein, and low carbohydrate. KBs have been shown to cross the blood brain barrier to improve brain energy metabolism. This review comprehensively summarizes the current literature on how increasing KBs support brain energy metabolism. In addition, for the first time, this review discusses the effects of ketogenic diet on the putative AD biomarkers such as Aβ, tau (mainly p-tau 181), GFAP, and NFL, and discusses the role of KBs on neuroinflammation, oxidative stress, and mitochondrial metabolism.
Collapse
Affiliation(s)
- Matin Ramezani
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Macquarie, NSW, Australia
| | - Malika Fernando
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Macquarie, NSW, Australia
| | - Shaun Eslick
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Macquarie, NSW, Australia
| | - Prita R. Asih
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Macquarie, NSW, Australia
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Sina Shadfar
- Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | | | - Heidi Hillebrandt
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Macquarie, NSW, Australia
| | - Silochna Meghwar
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Macquarie, NSW, Australia
| | - Maryam Shahriari
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Macquarie, NSW, Australia
| | - Pratishtha Chatterjee
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Macquarie, NSW, Australia
| | - Rohith Thota
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Macquarie, NSW, Australia
| | - Cintia B. Dias
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Macquarie, NSW, Australia
| | - Manohar L. Garg
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Macquarie, NSW, Australia
| | - Ralph N. Martins
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Macquarie, NSW, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| |
Collapse
|
10
|
Li T, Yang S, Liu X, Li Y, Gu Z, Jiang Z. Dietary neoagarotetraose extends lifespan and impedes brain aging in mice via regulation of microbiota-gut-brain axis. J Adv Res 2023; 52:119-134. [PMID: 37085001 PMCID: PMC10555787 DOI: 10.1016/j.jare.2023.04.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 03/31/2023] [Accepted: 04/16/2023] [Indexed: 04/23/2023] Open
Abstract
INTRODUCTION Dietary oligosaccharides can impact the gut microbiota and confer tremendous health benefits. OBJECTIVES The aim of this study was to determine the impact of a novel functional oligosaccharide, neoagarotetraose (NAT), on aging in mice. METHODS 8-month-old C57BL/6J mice as the natural aging mice model were orally administered with NAT for 12 months. The preventive effect of NAT in Alzheimer's disease (AD) mice was further evaluated. Aging related indicators, neuropathology, gut microbiota and short-chain fatty acids (SCFAs) in cecal contents were analyzed. RESULTS NAT treatment extended the lifespan of these mice by up to 33.3 %. Furthermore, these mice showed the improved aging characteristics and decreased injuries in cerebral neurons. Dietary NAT significantly delayed DNA damage in the brain, and inhibited reduction of tight junction protein in the colon. A significant increase at gut bacterial genus level (such as Lactobacillus, Butyricimonas, and Akkermansia) accompanied by increasing concentrations of SCFAs in cecal contents was observed after NAT treatment. Functional profiling of gut microbiota composition indicated that NAT treatment regulated the glucolipid and bile acid-related metabolic pathways. Interestingly, NAT treatment ameliorated cognitive impairment, attenuated amyloid-β (Aβ) and Tau pathology, and regulated the gut microbiota composition and SCFAs receptor-related pathway of Alzheimer's disease (AD) mice. CONCLUSION NAT mitigated age-associated cerebral injury in mice through gut-brain axis. The findings provide novel evidence for the effect of NAT on anti-aging, and highlight the potential application of NAT as an effective intervention against age-related diseases.
Collapse
Affiliation(s)
- Tao Li
- Key Laboratory of Food Bioengineering (China National Light Industry), College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Shaoqing Yang
- Key Laboratory of Food Bioengineering (China National Light Industry), College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Xiaoyan Liu
- Key Laboratory of Food Bioengineering (China National Light Industry), College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yanxiao Li
- Department of Nutrition and Health, College of Engineering, China Agricultural University, Beijing, China
| | - Zhenglong Gu
- Division of Nutritional Sciences, Cornell University, NY, USA; Greater Bay Area Institute of Precision Medicine (Guangzhou), Nansha District, Guangzhou 511400, China; Institute of Life Sciences, Fudan University, Shanghai 200433, China.
| | - Zhengqiang Jiang
- Key Laboratory of Food Bioengineering (China National Light Industry), College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.
| |
Collapse
|
11
|
Dong Y, Zou Z, Deng P, Fan X, Li C. Circulating metabolites and depression: a bidirectional Mendelian randomization. Front Neurosci 2023; 17:1146613. [PMID: 37152596 PMCID: PMC10160621 DOI: 10.3389/fnins.2023.1146613] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/29/2023] [Indexed: 05/09/2023] Open
Abstract
Background Studies have shown an association between depression and circulating metabolites, but the causal relationship between them has not been elucidated. The purpose of this study was to elucidate the causal relationship between circulating metabolites and depression and to explore the role of circulating metabolites in depression. Methods In this study, the top single-nucleotide polymorphisms (SNPs) associated with circulating metabolites (n = 24,925) and depression (n = 322,580) were obtained based on the publicly available genome-wide association study using two-sample Mendelian randomization (MR). SNP estimates were summarized through inverse variance weighted, MR Egger, weighted median, MR pleiotropy residual sum and outlier, and "leave-one-out" methods. Results Apolipoprotein A-I (OR 0.990, 95% CI 981-0.999) and glutamine (OR 0.985, 95% CI 0.972-0.997) had protective causal effects on depression, whereas acetoacetate (OR 1.021, 95% CI 1.009-1.034), glycoproteins (OR 1.005, 95% CI 1.000-1.009), isoleucine (OR 1.013, 95% CI 1.002-1.024), and urea (OR 1.020, 95% CI 1.000-1.039) had an anti-protective effect on depression. Reversed MR showed no effect of depression on the seven circulating metabolites. Conclusion In this study, MR analysis showed that apolipoprotein A-I and glutamine had a protective effect on depression, and acetoacetate, glycoprotein, isoleucine, glucose, and urea may be risk factors for depression. Therefore, further research must be conducted to translate the findings into practice.
Collapse
Affiliation(s)
- Yankai Dong
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zengxiao Zou
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Pin Deng
- Department of Hand and Foot Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Xiaoping Fan
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Xiaoping Fan
| | - Chunlin Li
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- *Correspondence: Chunlin Li
| |
Collapse
|