1
|
Wen B, Han X, Gong J, Wang P, Sun W, Xu C, Shan A, Wang X, Luan H, Li S, Li R, Guo J, Chen R, Li C, Sun Y, Lv S, Wei C. Nutrition: A non-negligible factor in the pathogenesis and treatment of Alzheimer's disease. Alzheimers Dement 2025; 21:e14547. [PMID: 39868840 PMCID: PMC11863745 DOI: 10.1002/alz.14547] [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: 11/14/2024] [Revised: 12/16/2024] [Accepted: 12/21/2024] [Indexed: 01/28/2025]
Abstract
Alzheimer's disease (AD) is a degenerative disease characterized by progressive cognitive dysfunction. The strong link between nutrition and the occurrence and progression of AD pathology has been well documented. Poor nutritional status accelerates AD progress by potentially aggravating amyloid beta (Aβ) and tau deposition, exacerbating oxidative stress response, modulating the microbiota-gut-brain axis, and disrupting blood-brain barrier function. The advanced stage of AD tends to lead to malnutrition due to cognitive impairments, sensory dysfunctions, brain atrophy, and behavioral and psychological symptoms of dementia (BPSD). This, in turn, produces a vicious cycle between malnutrition and AD. This review discusses how nutritional factors and AD deteriorate each other from the early stage of AD to the terminal stages of AD, focusing on the potential of different levels of nutritional factors, ranging from micronutrients to diet patterns. This review provides novel insights into reducing the risk of AD, delaying its progression, and improving prognosis. HIGHLIGHTS: Two-fifths of Alzheimer's disease (AD) cases worldwide have been attributed to potentially modifiable risk factors. Up to ≈26% of community-dwelling patients with AD are malnourished, compared to 7%∼76% of institutionalized patients. Undernutrition effects the onset, progression, and prognosis of AD through multiple mechanisms. Various levels of nutritional supports were confirmed to be protective factors for AD via specific mechanisms.
Collapse
Affiliation(s)
- Boye Wen
- Innovation Center for Neurological Disorders and Department of NeurologyXuanwu HospitalCapital Medical UniversityNational Clinical Research Center for Geriatric DiseasesXicheng DistrictBeijingChina
| | - Xiaodong Han
- Innovation Center for Neurological Disorders and Department of NeurologyXuanwu HospitalCapital Medical UniversityNational Clinical Research Center for Geriatric DiseasesXicheng DistrictBeijingChina
| | - Jin Gong
- College of Integrated Traditional Chinese and Western MedicineChangchun University of Chinese MedicineJingyue National High‐tech Industrial Development ZoneChangchunChina
| | - Pin Wang
- Innovation Center for Neurological Disorders and Department of NeurologyXuanwu HospitalCapital Medical UniversityNational Clinical Research Center for Geriatric DiseasesXicheng DistrictBeijingChina
| | - Wenxian Sun
- Innovation Center for Neurological Disorders and Department of NeurologyXuanwu HospitalCapital Medical UniversityNational Clinical Research Center for Geriatric DiseasesXicheng DistrictBeijingChina
| | - Chang Xu
- Innovation Center for Neurological Disorders and Department of NeurologyXuanwu HospitalCapital Medical UniversityNational Clinical Research Center for Geriatric DiseasesXicheng DistrictBeijingChina
| | - Aidi Shan
- Innovation Center for Neurological Disorders and Department of NeurologyXuanwu HospitalCapital Medical UniversityNational Clinical Research Center for Geriatric DiseasesXicheng DistrictBeijingChina
| | - Xin Wang
- Innovation Center for Neurological Disorders and Department of NeurologyXuanwu HospitalCapital Medical UniversityNational Clinical Research Center for Geriatric DiseasesXicheng DistrictBeijingChina
| | - Heya Luan
- Innovation Center for Neurological Disorders and Department of NeurologyXuanwu HospitalCapital Medical UniversityNational Clinical Research Center for Geriatric DiseasesXicheng DistrictBeijingChina
| | - Shaoqi Li
- College of Integrated Traditional Chinese and Western MedicineChangchun University of Chinese MedicineJingyue National High‐tech Industrial Development ZoneChangchunChina
| | - Ruina Li
- School of Biological Science and Medical EngineeringBeihang UniversityHaidian DistrictBeijingChina
| | - Jinxuan Guo
- College of Integrated Traditional Chinese and Western MedicineChangchun University of Chinese MedicineJingyue National High‐tech Industrial Development ZoneChangchunChina
| | - Runqi Chen
- School of Biological Science and Medical EngineeringBeihang UniversityHaidian DistrictBeijingChina
| | - Chuqiao Li
- Innovation Center for Neurological Disorders and Department of NeurologyXuanwu HospitalCapital Medical UniversityNational Clinical Research Center for Geriatric DiseasesXicheng DistrictBeijingChina
| | - Yao Sun
- Innovation Center for Neurological Disorders and Department of NeurologyXuanwu HospitalCapital Medical UniversityNational Clinical Research Center for Geriatric DiseasesXicheng DistrictBeijingChina
| | - Sirong Lv
- Innovation Center for Neurological Disorders and Department of NeurologyXuanwu HospitalCapital Medical UniversityNational Clinical Research Center for Geriatric DiseasesXicheng DistrictBeijingChina
| | - Cuibai Wei
- Innovation Center for Neurological Disorders and Department of NeurologyXuanwu HospitalCapital Medical UniversityNational Clinical Research Center for Geriatric DiseasesXicheng DistrictBeijingChina
| |
Collapse
|
2
|
O'Connor LC, Kang WK, Vo P, Spinelli JB, Alkema MJ, Byrne AB. Comamonas aquatica inhibits TIR-1/SARM1 induced axon degeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.20.622298. [PMID: 39605655 PMCID: PMC11601612 DOI: 10.1101/2024.11.20.622298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Emerging evidence suggests the microbiome critically influences the onset and progression of neurodegenerative diseases; however, the identity of neuroprotective bacteria and the molecular mechanisms that respond within the host remain largely unknown. We took advantage of Caenorhabditis elegans' well characterized nervous system and ability to eat uni-bacterial diets to determine how metabolites and neuroprotective molecules from single species of bacteria suppress degeneration of motor neurons. We found Comamonas aquatica significantly protects against degeneration induced by overexpressing a key regulator of axon degeneration, TIR-1/SARM1. Genetic analyses and metabolomics reveal Comamonas protects against neurodegeneration by providing sufficient Vitamin B12 to activate METR-1/MTR methionine synthase in the intestine, which then lowers toxic levels of homocysteine in TIR-1-expressing animals. Defining a molecular pathway between Comamonas and neurodegeneration adds significantly to our understanding of gut-brain interactions and, given the prominent role of homocysteine in neurodegenerative disorders, reveals how such a bacterium could protect against disease.
Collapse
|
3
|
Ekundayo BE, Adewale OB, Obafemi BA, Afolabi OB, Obafemi TO. Management of Alzheimer's disease and related neurotoxic pathologies: Role of thiamine, pyridoxine and cobalamin. Eur J Pharmacol 2024; 982:176958. [PMID: 39209095 DOI: 10.1016/j.ejphar.2024.176958] [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: 05/17/2024] [Revised: 08/10/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Alzheimer's disease (AD) remains one of the most debilitating disease and most common neurological disorder in the world at large. However, with many years of multiple research and billions of dollars invested for the purpose of research, not many therapeutic options exist for the management of this disease. As at 2023, the number has only increased to 7, one of which is a combination of two existing therapies. However, research has continued still in the search for a cure. The roles and functions of thiamine, pyridoxine and cobalamin in the proper function of the nervous system has been well researched over time and their role in the management of neurological diseases have been of interest in the last decade. This review describes the roles of the aforementioned chemicals in the management of different models of AD and AD-like pathologies as mono-therapeutic agents and prospective adjuvant for combination therapy.
Collapse
Affiliation(s)
| | | | - Blessing Ariyo Obafemi
- Department of Medical Biochemistry Afe Babalola University, PMB 5454, Ado-Ekiti, Nigeria
| | | | - Tajudeen Olabisi Obafemi
- Department of Biochemistry Afe Babalola University, PMB 5454, Ado-Ekiti, Nigeria; Department of Life and Consumer Sciences, School of Agriculture and Life Sciences, University of South Florida Park, Roodepoort, 1709, Johannesburg, South Africa
| |
Collapse
|
4
|
Anjaneyulu J, Godbole A. Small organism models for mode of action research on anti-ageing and nootropic herbs, foods, and formulations. Nutr Neurosci 2024:1-19. [PMID: 39432435 DOI: 10.1080/1028415x.2024.2409128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
With global increase in ageing population along with increasing age-related neurodegenerative diseases (NDs), development of sustainable, safe and effective solutions for promoting healthy ageing and preventing diseases has become a priority. Traditional healthcare systems/medicines prescribe several herbs, foods and formulations to promote healthy ageing and prevent and/or treat age-related diseases. However, the scientific data elucidating their mechanism of action is very limited and deeper research using different models is warranted for timely and wider use. The clinical studies and research with higher model organisms, although useful, have several practical, technical, and financial limitations. Conversely, small organism models like Yeast, Roundworm, Fruit fly, and Zebrafish, which have genetic similarities to humans, can replicate the disease features and provide behavioural, cellular and molecular insights. The common features of ageing and NDs, like amyloid protein aggregations, oxidative stress, energy dysregulation, inflammation and neurodegeneration can be mimicked in the small organism models for Alzheimer's, Parkinson's, Huntington's diseases, and Amyotrophic Lateral Sclerosis. This review focuses on small organism model- based research unveiling interesting modes of action and synergistic effects of herbal extracts, foods, and formulations, which are indicated especially for healthy ageing and management of NDs. This will provide leads for the quick and sustainable development of scientifically evaluated solutions for clinically relevant, age-related conditions.
Collapse
Affiliation(s)
- Jalagam Anjaneyulu
- The University of Trans-disciplinary Health Sciences and Technology (TDU), Bengaluru, India
| | - Ashwini Godbole
- The University of Trans-disciplinary Health Sciences and Technology (TDU), Bengaluru, India
| |
Collapse
|
5
|
Lu S, Zhao Q, Guan Y, Sun Z, Li W, Guo S, Zhang A. The communication mechanism of the gut-brain axis and its effect on central nervous system diseases: A systematic review. Biomed Pharmacother 2024; 178:117207. [PMID: 39067168 DOI: 10.1016/j.biopha.2024.117207] [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: 05/13/2024] [Revised: 07/15/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024] Open
Abstract
Gut microbiota is involved in intricate and active metabolic processes the host's brain function, especially its role in immune responses, secondary metabolism, and symbiotic connections with the host. Gut microbiota can promote the production of essential metabolites, neurotransmitters, and other neuroactive chemicals that affect the development and treatment of central nervous system diseases. This article introduces the relevant pathways and manners of the communication between the brain and gut, summarizes a comprehensive overview of the current research status of key gut microbiota metabolites that affect the functions of the nervous system, revealing those adverse factors that affect typical communication between the brain-gut axis, and outlining the efforts made by researchers to alleviate these neurological diseases through targeted microbial interventions. The relevant pathways and manners of communication between the brain and gut contribute to the experimental design of new treatment plans and drug development. The factors that may cause changes in gut microbiota and affect metabolites, as well as current intervention methods are summarized, which helps improve gut microbiota brain dialogue, prevent adverse triggering factors from interfering with the gut microbiota system, and minimize neuropathological changes.
Collapse
Affiliation(s)
- Shengwen Lu
- Department of Pharmaceutical Analysis, GAP Center, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Qiqi Zhao
- Department of Pharmaceutical Analysis, GAP Center, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Yu Guan
- Department of Pharmaceutical Analysis, GAP Center, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Zhiwen Sun
- Department of Gastroenterology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Wenhao Li
- School of Basic Medical Science of Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Sifan Guo
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China
| | - Aihua Zhang
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China; Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China; INTI International University, Nilai 71800, Malaysia.
| |
Collapse
|
6
|
Ge Y, Chen S, Wu B, Zhang Y, Wang J, He X, Liu W, Chen Y, Ou Y, Shen X, Huang Y, Gan Y, Yang L, Ma L, Ma Y, Chen K, Chen S, Cui M, Tan L, Dong Q, Zhao Q, Wang Y, Jia J, Yu J. Genome-wide meta-analysis identifies ancestry-specific loci for Alzheimer's disease. Alzheimers Dement 2024; 20:6243-6256. [PMID: 39023044 PMCID: PMC11497642 DOI: 10.1002/alz.14121] [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: 12/13/2023] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 07/20/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a devastating neurological disease with complex genetic etiology. Yet most known loci have only identified from the late-onset type AD in populations of European ancestry. METHODS We performed a two-stage genome-wide association study (GWAS) of AD totaling 6878 Chinese and 63,926 European individuals. RESULTS In addition to the apolipoprotein E (APOE) locus, our GWAS of two independent Chinese samples uncovered three novel AD susceptibility loci (KIAA2013, SLC52A3, and TCN2) and a novel ancestry-specific variant within EGFR (rs1815157). More replicated variants were observed in the Chinese (31%) than in the European samples (15%). In combining genome-wide associations and functional annotations, EGFR and TCN2 were prioritized as two of the most biologically significant genes. Phenome-wide Mendelian randomization suggests that high mean corpuscular hemoglobin concentration might protect against AD. DISCUSSION The current study reveals novel AD susceptibility loci, emphasizes the importance of diverse populations in AD genetic research, and advances our understanding of disease etiology. HIGHLIGHTS Loci KIAA2013, SLC52A3, and TCN2 were associated with Alzheimer's disease (AD) in Chinese populations. rs1815157 within the EGFR locus was associated with AD in Chinese populations. The genetic architecture of AD varied between Chinese and European populations. EGFR and TCN2 were prioritized as two of the most biologically significant genes. High mean corpuscular hemoglobin concentrations might have protective effects against AD.
Collapse
Affiliation(s)
- Yi‐Jun Ge
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Shi‐Dong Chen
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Bang‐Sheng Wu
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Ya‐Ru Zhang
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Jun Wang
- Department of Neurology and Centre for Clinical NeuroscienceDaping HospitalThird Military Medical UniversityChongqingChina
| | - Xiao‐Yu He
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Wei‐Shi Liu
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Yi‐Lin Chen
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Ya‐Nan Ou
- Department of NeurologyQingdao Municipal HospitalQingdao UniversityQingdaoChina
| | - Xue‐Ning Shen
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Yu‐Yuan Huang
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Yi‐Han Gan
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Liu Yang
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Ling‐Zhi Ma
- Department of NeurologyQingdao Municipal HospitalQingdao UniversityQingdaoChina
| | - Ya‐Hui Ma
- Department of NeurologyThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Ke‐Liang Chen
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Shu‐Fen Chen
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Mei Cui
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Lan Tan
- Department of NeurologyQingdao Municipal HospitalQingdao UniversityQingdaoChina
| | - Qiang Dong
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Qian‐Hua Zhao
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Yan‐Jiang Wang
- Department of Neurology and Centre for Clinical NeuroscienceDaping HospitalThird Military Medical UniversityChongqingChina
| | - Jian‐Ping Jia
- Innovation Center for Neurological Disorders and Department of NeurologyNational Clinical Research Center for Geriatric DiseasesXuanwu HospitalCapital Medical UniversityBeijingChina
| | - Jin‐Tai Yu
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| |
Collapse
|
7
|
Zhao R. Can exercise benefits be harnessed with drugs? A new way to combat neurodegenerative diseases by boosting neurogenesis. Transl Neurodegener 2024; 13:36. [PMID: 39049102 PMCID: PMC11271207 DOI: 10.1186/s40035-024-00428-7] [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: 02/02/2024] [Accepted: 07/01/2024] [Indexed: 07/27/2024] Open
Abstract
Adult hippocampal neurogenesis (AHN) is affected by multiple factors, such as enriched environment, exercise, ageing, and neurodegenerative disorders. Neurodegenerative disorders can impair AHN, leading to progressive neuronal loss and cognitive decline. Compelling evidence suggests that individuals engaged in regular exercise exhibit higher production of proteins that are essential for AHN and memory. Interestingly, specific molecules that mediate the effects of exercise have shown effectiveness in promoting AHN and cognition in different transgenic animal models. Despite these advancements, the precise mechanisms by which exercise mimetics induce AHN remain partially understood. Recently, some novel exercise molecules have been tested and the underlying mechanisms have been proposed, involving intercommunications between multiple organs such as muscle-brain crosstalk, liver-brain crosstalk, and gut-brain crosstalk. In this review, we will discuss the current evidence regarding the effects and potential mechanisms of exercise mimetics on AHN and cognition in various neurological disorders. Opportunities, challenges, and future directions in this research field are also discussed.
Collapse
Affiliation(s)
- Renqing Zhao
- College of Physical Education, Yangzhou University, 88 South Daxue Road, Yangzhou, 225009, China.
| |
Collapse
|
8
|
Lee YT, Senturk M, Guan Y, Wang MC. Bacteria-organelle communication in physiology and disease. J Cell Biol 2024; 223:e202310134. [PMID: 38748249 PMCID: PMC11096858 DOI: 10.1083/jcb.202310134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 04/03/2024] [Accepted: 05/03/2024] [Indexed: 05/18/2024] Open
Abstract
Bacteria, omnipresent in our environment and coexisting within our body, exert dual beneficial and pathogenic influences. These microorganisms engage in intricate interactions with the human body, impacting both human health and disease. Simultaneously, certain organelles within our cells share an evolutionary relationship with bacteria, particularly mitochondria, best known for their energy production role and their dynamic interaction with each other and other organelles. In recent years, communication between bacteria and mitochondria has emerged as a new mechanism for regulating the host's physiology and pathology. In this review, we delve into the dynamic communications between bacteria and host mitochondria, shedding light on their collaborative regulation of host immune response, metabolism, aging, and longevity. Additionally, we discuss bacterial interactions with other organelles, including chloroplasts, lysosomes, and the endoplasmic reticulum (ER).
Collapse
Affiliation(s)
- Yi-Tang Lee
- Waisman Center, University of Wisconsin, Madison, WI, USA
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Integrative Program of Molecular and Biochemical Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Mumine Senturk
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
| | - Youchen Guan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Meng C. Wang
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| |
Collapse
|
9
|
Lee CY, Chan L, Hu CJ, Hong CT, Chen JH. Role of vitamin B12 and folic acid in treatment of Alzheimer's disease: a meta-analysis of randomized control trials. Aging (Albany NY) 2024; 16:7856-7869. [PMID: 38700503 PMCID: PMC11132008 DOI: 10.18632/aging.205788] [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: 09/05/2023] [Accepted: 03/28/2024] [Indexed: 05/05/2024]
Abstract
Vitamin B12 and folic acid could reduce blood homocysteine levels, which was thought to slow down the progression of Alzheimer's disease (AD), but previous studies regarding the effect of vitamin B12 and folic acid in treatment of AD have not reached conclusive results. We searched PubMed and Embase until January 12, 2023. Only randomized control trials involving participants clearly diagnosed with AD and who received vitamin B12 and folic acid were enrolled. Five studies that met the criteria were selected for inclusion in the meta-analysis. Changes in cognitive function were measured based on either the Mini-Mental State Examination (MMSE) or the Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog). Changes in daily life function and the level of blood homocysteine were also investigated. After a 6-month treatment, administration of vitamin B12 and folic acid improved the MMSE scores more than placebo did (SMD = 0.21, 95% CI = 0.01 to 0.32, p = 0.04) but did not significantly affect ADAS-Cog scores (SMD = 0.06, 95% CI = -0.22 to 0.33, p = 0.68) or measures of daily life function. Blood homocysteine levels were significantly decreased after vitamin B12 and folic acid treatment. Participants with AD who received 6 months of vitamin B12 and folic acid supplementation had better MMSE scores but had no difference in ADAS-Cog scores. Daily life function did not improve after treatment.
Collapse
Affiliation(s)
- Chih-Ying Lee
- Department of Neurology, Shuang-Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | - Lung Chan
- Department of Neurology, Shuang-Ho Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chaur-Jong Hu
- Department of Neurology, Shuang-Ho Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chien-Tai Hong
- Department of Neurology, Shuang-Ho Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jia-Hung Chen
- Department of Neurology, Shuang-Ho Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
10
|
Wang C, Yang M, Liu D, Zheng C. Metabolic rescue of α-synuclein-induced neurodegeneration through propionate supplementation and intestine-neuron signaling in C. elegans. Cell Rep 2024; 43:113865. [PMID: 38412096 DOI: 10.1016/j.celrep.2024.113865] [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: 07/31/2023] [Revised: 01/14/2024] [Accepted: 02/08/2024] [Indexed: 02/29/2024] Open
Abstract
Microbial metabolites that can modulate neurodegeneration are promising therapeutic targets. Here, we found that the short-chain fatty acid propionate protects against α-synuclein-induced neuronal death and locomotion defects in a Caenorhabditis elegans model of Parkinson's disease (PD) through bidirectional regulation between the intestine and neurons. Both depletion of dietary vitamin B12, which induces propionate breakdown, and propionate supplementation suppress neurodegeneration and reverse PD-associated transcriptomic aberrations. Neuronal α-synuclein aggregation induces intestinal mitochondrial unfolded protein response (mitoUPR), which leads to reduced propionate levels that trigger transcriptional reprogramming in the intestine and cause defects in energy production. Weakened intestinal metabolism exacerbates neurodegeneration through interorgan signaling. Genetically enhancing propionate production or overexpressing metabolic regulators downstream of propionate in the intestine rescues neurodegeneration, which then relieves mitoUPR. Importantly, propionate supplementation suppresses neurodegeneration without reducing α-synuclein aggregation, demonstrating metabolic rescue of neuronal proteotoxicity downstream of protein aggregates. Our study highlights the involvement of small metabolites in the gut-brain interaction in neurodegenerative diseases.
Collapse
Affiliation(s)
- Chenyin Wang
- School of Biological Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Meigui Yang
- School of Biological Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Dongyao Liu
- School of Biological Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Chaogu Zheng
- School of Biological Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| |
Collapse
|
11
|
Yadav DK, Chang AC, Grooms NWF, Chung SH, Gabel CV. O-GlcNAc signaling increases neuron regeneration through one-carbon metabolism in Caenorhabditis elegans. eLife 2024; 13:e86478. [PMID: 38334260 PMCID: PMC10857789 DOI: 10.7554/elife.86478] [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: 01/28/2023] [Accepted: 11/17/2023] [Indexed: 02/10/2024] Open
Abstract
Cellular metabolism plays an essential role in the regrowth and regeneration of a neuron following physical injury. Yet, our knowledge of the specific metabolic pathways that are beneficial to neuron regeneration remains sparse. Previously, we have shown that modulation of O-linked β-N-acetylglucosamine (O-GlcNAc) signaling, a ubiquitous post-translational modification that acts as a cellular nutrient sensor, can significantly enhance in vivo neuron regeneration. Here, we define the specific metabolic pathway by which O-GlcNAc transferase (ogt-1) loss of function mediates increased regenerative outgrowth. Performing in vivo laser axotomy and measuring subsequent regeneration of individual neurons in C. elegans, we find that glycolysis, serine synthesis pathway (SSP), one-carbon metabolism (OCM), and the downstream transsulfuration metabolic pathway (TSP) are all essential in this process. The regenerative effects of ogt-1 mutation are abrogated by genetic and/or pharmacological disruption of OCM and the SSP linking OCM to glycolysis. Testing downstream branches of this pathway, we find that enhanced regeneration is dependent only on the vitamin B12 independent shunt pathway. These results are further supported by RNA sequencing that reveals dramatic transcriptional changes by the ogt-1 mutation, in the genes involved in glycolysis, OCM, TSP, and ATP metabolism. Strikingly, the beneficial effects of the ogt-1 mutation can be recapitulated by simple metabolic supplementation of the OCM metabolite methionine in wild-type animals. Taken together, these data unearth the metabolic pathways involved in the increased regenerative capacity of a damaged neuron in ogt-1 animals and highlight the therapeutic possibilities of OCM and its related pathways in the treatment of neuronal injury.
Collapse
Affiliation(s)
- Dilip Kumar Yadav
- Department of Pharmacology, Physiology and Biophysics, Chobanian & Avedisian School of Medicine, Boston UniversityBostonUnited States
| | - Andrew C Chang
- Department of Pharmacology, Physiology and Biophysics, Chobanian & Avedisian School of Medicine, Boston UniversityBostonUnited States
| | - Noa WF Grooms
- Department of Bioengineering, Northeastern UniversityBostonUnited States
| | - Samuel H Chung
- Department of Bioengineering, Northeastern UniversityBostonUnited States
| | - Christopher V Gabel
- Department of Pharmacology, Physiology and Biophysics, Chobanian & Avedisian School of Medicine, Boston UniversityBostonUnited States
- Neurophotonics Center, Boston UniversityBostonUnited States
| |
Collapse
|
12
|
Zhao R. Exercise mimetics: a novel strategy to combat neuroinflammation and Alzheimer's disease. J Neuroinflammation 2024; 21:40. [PMID: 38308368 PMCID: PMC10837901 DOI: 10.1186/s12974-024-03031-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/25/2024] [Indexed: 02/04/2024] Open
Abstract
Neuroinflammation is a pathological hallmark of Alzheimer's disease (AD), characterized by the stimulation of resident immune cells of the brain and the penetration of peripheral immune cells. These inflammatory processes facilitate the deposition of amyloid-beta (Aβ) plaques and the abnormal hyperphosphorylation of tau protein. Managing neuroinflammation to restore immune homeostasis and decrease neuronal damage is a therapeutic approach for AD. One way to achieve this is through exercise, which can improve brain function and protect against neuroinflammation, oxidative stress, and synaptic dysfunction in AD models. The neuroprotective impact of exercise is regulated by various molecular factors that can be activated in the same way as exercise by the administration of their mimetics. Recent evidence has proven some exercise mimetics effective in alleviating neuroinflammation and AD, and, additionally, they are a helpful alternative option for patients who are unable to perform regular physical exercise to manage neurodegenerative disorders. This review focuses on the current state of knowledge on exercise mimetics, including their efficacy, regulatory mechanisms, progress, challenges, limitations, and future guidance for their application in AD therapy.
Collapse
Affiliation(s)
- Renqing Zhao
- College of Physical Education, Yangzhou University, Yangzhou, China.
| |
Collapse
|
13
|
Mishra E, Thakur MK. Vitamin B 12-folic acid supplementation improves memory by altering mitochondrial dynamics, dendritic arborization, and neurodegeneration in old and amnesic male mice. J Nutr Biochem 2024; 124:109536. [PMID: 37981108 DOI: 10.1016/j.jnutbio.2023.109536] [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: 06/25/2023] [Revised: 10/28/2023] [Accepted: 11/15/2023] [Indexed: 11/21/2023]
Abstract
Memory impairment during aging and amnesia is attributed to compromised mitochondrial dynamics and mitophagy and other mitochondrial quality control mechanisms. Mitochondrial dynamics involves the continuous process of fission and fusion of mitochondria within a cell and is a fundamental mechanism for regulating mitochondrial quality and function. An extensive range of potential nutritional supplements has been shown to improve mitochondrial health, synaptic plasticity, and cognitive functions. Previous findings revealed that supplementation of vitamin B12-folic acid reduces locomotor deficits and mitochondrial abnormalities but enhances mitochondrial and neuronal health. The present study aims to explore the impact of combined vitamin B12-folic acid supplementation on mitochondrial dynamics, neuronal health, and memory decline in old age and scopolamine-induced amnesia, which remains elusive. The results demonstrated that supplementation led to a noteworthy increase in recognition and spatial memory and expression of memory-related protein BDNF in old and amnesic mice. Moreover, the decrease in the fragmented mitochondrial number was validated by the downregulation of mitochondrial fission p-Drp1 (S616) protein and the increase in elongated mitochondria by the upregulation of mitochondrial fusion Mfn2 protein. The increased spine density and dendritic arborization in old and amnesic mice upon supplementation were confirmed by the enhanced expression level of PSD95 and synaptophysin. Furthermore, supplementation reduced ROS production, inhibited Caspase-3 activation, mitigated neurodegeneration, and enhanced mitochondrial membrane potential, ATP production, Vdac1 expression, myelination, in old and amnesic mice. Collectively, our findings imply that combined supplementation of vitamin B12-folic acid improves mitochondrial dynamics and neuronal health, and leads to recovery of memory during old age and amnesia.
Collapse
Affiliation(s)
- Ela Mishra
- Department of Zoology, Biochemistry and Molecular Biology Laboratory, Centre of Advanced Study, Institute of Science, Banaras Hindu University, Varanasi, India.
| | - Mahendra Kumar Thakur
- Department of Zoology, Biochemistry and Molecular Biology Laboratory, Centre of Advanced Study, Institute of Science, Banaras Hindu University, Varanasi, India.
| |
Collapse
|
14
|
Zhu X, Xia Y, Wang H, Shi L, Yin H, Gu M, Yan F. PM 2.5 induced neurotoxicity through unbalancing vitamin B12 metabolism by gut microbiota disturbance. Gut Microbes 2023; 15:2267186. [PMID: 37842922 PMCID: PMC10580859 DOI: 10.1080/19490976.2023.2267186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 10/02/2023] [Indexed: 10/17/2023] Open
Abstract
Fine particulate matter (PM2.5) in the atmosphere is easily accompanied by toxic and harmful substances, causing serious harm to human health, including cognitive impairment. Vitamin B12 (VitB12) is an essential micronutrient that is synthesized by bacteria and contributes to neurotransmitter synthesis as a nutrition and signaling molecule. However, the relationship between VitB12 attenuation of cognitive impairment and intestinal microbiota regulation in PM2.5 exposure has not been elucidated. In this study, we demonstrated that PM2.5 caused behavioral defects and neuronal damage in Caenorhabditis elegans (C. elegans), along with significant gene expression changes in neurotransmitter receptors and a decrease in VitB12 content, causing behavioral defects and neuronal damage in C. elegans. Methylcobalamin (MeCbl), a VitB12 analog, alleviated PM2.5-induced neurotoxicity in C. elegans. Moreover, using in vivo and in vitro models, we discovered that long-term exposure to PM2.5 led to changes in the structure of the gut microbiota, resulting in an imbalance of the VitB12-associated metabolic pathway followed by cognitive impairment. MeCbl supplementation could increase the diversity of the bacteria, reduce harmful substance contents, and restore the concentration of short-chain fatty acids (SCFAs) and neurotransmitters to the level of the control group to some degree. Here, a new target to mitigate the harm caused by PM2.5 was discovered, supplying MeCbl for relieving intestinal and intracellular neurotransmitter disorders. Our results also provide a reference for the use of VitB12 to target the adjustment of the human intestinal microbiota to improve metabolic disorders in people exposed to PM2.5.
Collapse
Affiliation(s)
- Xuan Zhu
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, People’s Republic of China
- Zhejiang Provincial Key Laboratory of Food Safety, Zhejiang Gongshang University, Hangzhou, China
| | - Yanting Xia
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, People’s Republic of China
| | - Huanhuan Wang
- School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
- Laboratory animal center, Hangzhou Normal University, Hangzhou, China
| | - Lihua Shi
- Weifang Elbe Health Food Co. Ltd, Weifang, China
| | - Hongping Yin
- School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
- Laboratory animal center, Hangzhou Normal University, Hangzhou, China
| | - Meier Gu
- School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
- Laboratory animal center, Hangzhou Normal University, Hangzhou, China
| | - Fujie Yan
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
15
|
Maynard AG, Petrova B, Kanarek N. Notes from the 2022 Folate, Vitamin B12, and One-Carbon Metabolism Conference. Metabolites 2023; 13:486. [PMID: 37110145 PMCID: PMC10147059 DOI: 10.3390/metabo13040486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
Here, we present notes from the Folate, Vitamin B12, and One-Carbon Metabolism Conference organized by The Federation of American Societies for Experimental Biology (FASEB), held in Asheville, North Carolina, USA, 14-19 August 2022. We aim to share the most recent findings in the field with members of our scientific community who did not attend the meeting and who are interested in the research that was presented. The research described includes discussions of one-carbon metabolism at the biochemical and physiological levels and studies of the role of folate and B12 in development and in the adult, and from bacteria to mammals. Furthermore, the summarized studies address the role of one-carbon metabolism in disease, including COVID-19, neurodegeneration, and cancer.
Collapse
Affiliation(s)
- Adam G. Maynard
- Department of Pathology, Boston Children’s Hospital, Boston, MA 02115, USA
- Graduate Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Boryana Petrova
- Department of Pathology, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Naama Kanarek
- Department of Pathology, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| |
Collapse
|
16
|
Li Q, Xiao M, Li N, Cai W, Zhao C, Liu B, Zeng F. Application of
Caenorhabditis elegans
in the evaluation of food nutrition: A review. EFOOD 2023. [DOI: 10.1002/efd2.68] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
Affiliation(s)
- Quancen Li
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Meifang Xiao
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Na Li
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Wenwen Cai
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Chao Zhao
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
- College of Marine Sciences Fujian Agriculture and Forestry University Fuzhou China
- Engineering Research Center of Fujian Subtropical Fruit and Vegetable Processing Fujian Agriculture and Forestry University Fuzhou China
| | - Bin Liu
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
- Engineering Research Center of Fujian Subtropical Fruit and Vegetable Processing Fujian Agriculture and Forestry University Fuzhou China
- National Engineering Research Center of JUNCAO Technology Fujian Agriculture and Forestry University Fuzhou China
| | - Feng Zeng
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
- Engineering Research Center of Fujian Subtropical Fruit and Vegetable Processing Fujian Agriculture and Forestry University Fuzhou China
| |
Collapse
|
17
|
Wang L, Zhou C, Yu H, Hao L, Ju M, Feng W, Guo Z, Sun X, Fan Q, Xiao R. Vitamin D, Folic Acid and Vitamin B 12 Can Reverse Vitamin D Deficiency-Induced Learning and Memory Impairment by Altering 27-Hydroxycholesterol and S-Adenosylmethionine. Nutrients 2022; 15:nu15010132. [PMID: 36615790 PMCID: PMC9824694 DOI: 10.3390/nu15010132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/09/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
The cholesterol-oxidized metabolite 27-hydroxycholesterol (27-OHC) is synthesized by CYP27A1, which is a key factor in vitamin D and oxysterol metabolism. Both vitamin D and 27-OHC are considered to play important roles in Alzheimer’s disease (AD). The study aims to research the effects of co-supplementation of vitamin D, folic acid, and vitamin B12 on learning and memory ability in vitamin D-deficient mice, and to explore the underlying mechanism. In this study, C57BL/6J mice were fed a vitamin D-deficient diet for 13 weeks to establish a vitamin D-deficient mice model. The vitamin D-deficient mice were then orally gavaged with vitamin D (VD), folic acid (FA), and vitamin B12 (VB12) alone or together for eight weeks. Following the gavage, the learning and memory ability of the mice were evaluated by Morris Water Maze and Novel object recognition test. The CYP27A1-related gene and protein expressions in the liver and brain were determined by qRT-PCR. The serum level of 27-OHC was detected by HPLC-MS. Serum levels of 25(OH)D, homocysteine (Hcy), and S-Adenosylmethionine (SAM) were measured by ELISA. After feeding with the vitamin D-deficient diet, the mice performed longer latency to a platform (p < 0.001), lower average speed (p = 0.026) in the Morris Water Maze, a lower time discrimination index (p = 0.009) in Novel object recognition, and performances were reversed after vitamin D, folic acid and vitamin B12 supplementation alone or together (p < 0.05). The gene expressions of CYP27A1 in the liver and brain were upregulated in the vitamin D-deficiency (VDD) group compared with the control (CON) group (p = 0.015), while it was downregulated in VDD + VD and VDD + VD-FA/VB12 groups compared with the VDD group (p < 0.05), with a similar trend in the protein expression of CYP27A1. The serum levels of 27-OHC were higher in the VDD group, compared with CON, VDD + VD, and VDD + VD-FA/VB12 group (p < 0.05), and a similar trend was found in the brain. The serum 25(OH)D levels were significantly decreased in the vitamin D-deficiency group (p = 0.008), and increased in the vitamin D-supplemented group (p < 0.001). The serum levels of SAM were higher in the B vitamins-supplemented group, compared with CON and VDD groups (p < 0.05). This study suggests that CYP27A1 expression may be involved in the mechanism of learning and memory impairment induced by vitamin D deficiency. Co-supplementation with vitamin D, folic acid, and vitamin B12 significantly reverses this effect by affecting the expression of CYP27A1, which in turn regulates the metabolism of 27-OHC, 25(OH)D, and SAM.
Collapse
Affiliation(s)
- Lijing Wang
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Cui Zhou
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Huiyan Yu
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Ling Hao
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Mengwei Ju
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Wenjing Feng
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Zhiting Guo
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Xuejing Sun
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Qiushi Fan
- Medical Nutrition, College of Allied Health Professions, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Rong Xiao
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, China
- Correspondence: ; Tel.: +86-010-83911512; Fax: +86-010-83911512
| |
Collapse
|
18
|
Wu X, Han Z, Liu B, Yu D, Sun J, Ge L, Tang W, Liu S. Gut microbiota contributes to the methionine metabolism in host. Front Microbiol 2022; 13:1065668. [PMID: 36620044 PMCID: PMC9815504 DOI: 10.3389/fmicb.2022.1065668] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Methionine (Met) metabolism provides methyl groups for many important physiological processes and is implicated in multiple inflammatory diseases associated with the disrupted intestinal microbiota; nevertheless, whether intestinal microbiota determines Met metabolism in the host remains largely unknown. Here, we found that gut microbiota is responsible for host Met metabolism by using various animal models, including germ-free (GF) pigs and mice. Specifically, the Met levels are elevated in both GF pigs and GF mice that mainly metabolized to S-adenosine methionine (SAM) in the liver. Furthermore, antibiotic clearance experiments demonstrate that the loss of certain ampicillin- or neomycin-sensitive gut microbiota causes decreased Met in murine colon. Overall, our study suggests that gut microbiota mediates Met metabolism in the host and is a prospective target for the treatment of Met metabolism-related diseases.
Collapse
Affiliation(s)
- Xiaoyan Wu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Ziyi Han
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Bingnan Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Dongming Yu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jing Sun
- Chongqing Academy of Animal Sciences, Chongqing, China
| | - Liangpeng Ge
- Chongqing Academy of Animal Sciences, Chongqing, China
| | - Wenjie Tang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, China,Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animtech Feed Co., Ltd., Chengdu, China,*Correspondence: Wenjie Tang, ; Shaojuan Liu,
| | - Shaojuan Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China,*Correspondence: Wenjie Tang, ; Shaojuan Liu,
| |
Collapse
|
19
|
Li J, Yang G, Shi W, Fang X, Han L, Cao Y. Anti-Alzheimer's disease active components screened out and identified from Hedyotis diffusa combining bioaffinity ultrafiltration LC-MS with acetylcholinesterase. JOURNAL OF ETHNOPHARMACOLOGY 2022; 296:115460. [PMID: 35714878 DOI: 10.1016/j.jep.2022.115460] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/30/2022] [Accepted: 06/11/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hedyotis diffusa is a traditional ethnomedicinal plant in local communities in northeastern Asia and used to treat inflammation, nervous breakdown, among others. In recent years, it has been applied in the treatment of Alzheimer's disease (AD), while the specific chemical components responsible for the activity remain need to be explored. AIM OF THE STUDY To prepare, screen and identify the potential anti-AD active components from Hedyotis diffusa. MATERIALS AND METHODS The acetylcholinesterase (AChE) inhibitory activity of four different extracts of Hedyotis diffusa were initially assessed using a spectrophotometric Ellman's method. A more accurate LC-MS/MS screening method combining functional enzyme assay and affinity ultrafiltration (AU) screening assay was developed and applied for the screening of natural compound inhibitors of AChE from Hedyotis diffusa. The binding mode was further investigated between protein and ligands via molecular docking. Subsequently, CL4176, a transgenic nematode model for AD, was used for activity validation of one of these components. RESULTS N-butanol extract of Hedyotis diffusa (NHD) appeared significant inhibitory activities on AChE, were chosen to delve deeper. Five bioactive components targeting AChE were screened out and identified using AU coupled to liquid chromatography-mass spectrometry. Molecular docking technique further confirmed the results of the screening assay. Finally, quercetin-3-O-sophoroside (QS) was confirmed as a potent anti-AD agent by in vivo experiments in C. elegans. CONCLUSION This study explores a new idea for screening anti-AD active components from traditional medicine. The findings provide a molecular structure and bioactivity basis for future potential applications of Hedyotis diffusa in medical industries.
Collapse
Affiliation(s)
- Jing Li
- Key Laboratory of Education Ministry on Traditional Chinese Medicine Resource and Compound Prescription, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Guangyi Yang
- Key Laboratory of Education Ministry on Traditional Chinese Medicine Resource and Compound Prescription, Hubei University of Chinese Medicine, Wuhan, 430065, China; Shenzhen Bao'an Traditional Chinese Medical Hospital, Shenzhen, 518000, China
| | - Wenfeng Shi
- Key Laboratory of Education Ministry on Traditional Chinese Medicine Resource and Compound Prescription, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xiaoping Fang
- Key Laboratory of Education Ministry on Traditional Chinese Medicine Resource and Compound Prescription, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Lintao Han
- Key Laboratory of Education Ministry on Traditional Chinese Medicine Resource and Compound Prescription, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Yan Cao
- Key Laboratory of Education Ministry on Traditional Chinese Medicine Resource and Compound Prescription, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| |
Collapse
|
20
|
Qin S, Wang Y, Li L, Liu J, Xiao C, Duan D, Hao W, Qin C, Chen J, Yao L, Zhang R, You J, Zheng JS, Shen E, Wu L. Early-life vitamin B12 orchestrates lipid peroxidation to ensure reproductive success via SBP-1/SREBP1 in Caenorhabditis elegans. Cell Rep 2022; 40:111381. [PMID: 36130518 DOI: 10.1016/j.celrep.2022.111381] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 07/05/2022] [Accepted: 08/27/2022] [Indexed: 11/03/2022] Open
Abstract
Vitamin B12 (B12) deficiency is a critical problem worldwide. Such deficiency in infants has long been known to increase the propensity to develop obesity and diabetes later in life through unclear mechanisms. Here, we establish a Caenorhabditis elegans model to study how early-life B12 impacts adult health. We find that early-life B12 deficiency causes increased lipogenesis and lipid peroxidation in adult worms, which in turn induces germline defects through ferroptosis. Mechanistically, we show the central role of the methionine cycle-SBP-1/SREBP1-lipogenesis axis in programming adult traits by early-life B12. Moreover, SBP-1/SREBP1 participates in a crucial feedback loop with NHR-114/HNF4 to maintain cellular B12 homeostasis. Inhibition of SBP-1/SREBP1-lipogenesis signaling and ferroptosis later in life can reverse disorders in adulthood when B12 cannot. Overall, this study provides mechanistic insights into the life-course effects of early-life B12 on the programming of adult health and identifies potential targets for future interventions for adiposity and infertility.
Collapse
Affiliation(s)
- Shenlu Qin
- Fudan University, Shanghai, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Yihan Wang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Lili Li
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Junli Liu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Congmei Xiao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Duo Duan
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Wanyu Hao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Chunxia Qin
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Jie Chen
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Luxia Yao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Runshuai Zhang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Jia You
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Ju-Sheng Zheng
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Enzhi Shen
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Lianfeng Wu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| |
Collapse
|
21
|
Zhao Y, Dong X, Chen B, Zhang Y, Meng S, Guo F, Guo X, Zhu J, Wang H, Cui H, Li S. Blood levels of circulating methionine components in Alzheimer’s disease and mild cognitive impairment: A systematic review and meta-analysis. Front Aging Neurosci 2022; 14:934070. [PMID: 35936764 PMCID: PMC9354989 DOI: 10.3389/fnagi.2022.934070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundCirculating methionine components have been reported to be associated with Alzheimer’s disease (AD) and mild cognitive impairment (MCI), although outcomes are not always consistent.Materials and methodsDatabase searching was conducted using PubMed, Embase, Cochrane Library, and Web of Science from inception to 26 December 2021. In this study, two reviewers independently identified eligible articles and extracted the data. We used Joanna Briggs Institute (JBI) Critical Appraisal tools to assess the overall quality of the included studies. STATA software was employed to perform meta-analysis evaluating the standardized mean difference (SMD) with its 95% confidence intervals (CIs) using random-effects models. Evidence quality was assessed using the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) criteria.ResultsTotally, 30 observational studies were eligible for inclusion. Compared with cognitively normal controls, patients with AD had increased homocysteine (Hcy) levels in the blood [standardized mean difference (SMD) = 0.59, 95% confidence interval [CI]: 0.36–0.82, P = 0.000], plasma (SMD = 0.39, 95% CI: 0.23–0.55, P = 0.000), and serum (SMD = 1.56, 95% CI: 0.59–2.95, P = 0.002). Patients with MCI were not significantly different from controls (SMD = 0.26, 95% CI: –0.07–0.58, P = 0.127). Patients with AD or MCI did not significantly differ from controls of blood vitamin B12 levels, AD (SMD = –0.05, 95% CI: –0.19–0.08, P = 0.440), or MCI (SMD = 0.01, 95% CI: –0.16–0.17, P = 0.94). Some cohort studies have suggested that higher Hcy, methionine, and S-adenosylmethionine levels may accelerate cognitive decline in patients with MCI or AD, and vitamin B12 deficiency is a risk factor for the disease; however, the results of other studies were inconsistent. According to the GRADE system, all these outcomes scored very low to low quality, and no high-quality evidence was found.ConclusionOnly Hcy levels in the plasma and serum were found to be inversely related to the risk of AD. However, due to the low quality of supporting these results, high-quality studies are needed to verify these findings.Systematic Review Registrationhttp://www.crd.york.ac.uk/PROSPERO/, identifier CRD42022308961.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
- School of Nursing, Hebei Medical University, Shijiazhuang, China
| | - Xinyi Dong
- School of Nursing, Hebei Medical University, Shijiazhuang, China
| | - Bingyu Chen
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
| | - Yizhou Zhang
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, China
| | - Sijia Meng
- School of Nursing, Hebei Medical University, Shijiazhuang, China
| | - Fangzhen Guo
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
| | - Xiaojing Guo
- School of Nursing, Hebei Medical University, Shijiazhuang, China
| | - Jialei Zhu
- School of Nursing, Hebei Medical University, Shijiazhuang, China
| | - Haoyue Wang
- School of Nursing, Hebei Medical University, Shijiazhuang, China
| | - Huixian Cui
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, China
- Huixian Cui,
| | - Sha Li
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, China
- *Correspondence: Sha Li,
| |
Collapse
|
22
|
Chondroitin sulfate E alleviates β-amyloid toxicity in transgenic Caenorhabditis elegans by inhibiting its aggregation. Int J Biol Macromol 2022; 209:1280-1287. [PMID: 35461860 DOI: 10.1016/j.ijbiomac.2022.04.124] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/14/2022] [Accepted: 04/17/2022] [Indexed: 01/13/2023]
Abstract
Chondroitin sulfate E (CS-E), which is characterized by oversulfated disaccharide units, has been shown to regulate neuronal adhesion, neurite outgrowth and exert neuroprotective effects. In view of these findings, here we investigated the anti-Alzheimer's disease (AD) activities of CSE by using transgenic Caenorhabditis elegans model of Alzheimer's disease. The behavioral experiments demonstrated that CSE at the concentration of 1 mg/ml significantly delayed the worm paralysis caused by Aβ aggregation as compared with control group. Western blot analysis revealed that the level of small oligomers in the transgenic C. elegans was significantly reduced upon treatment with CSE. The number of Aβ plaque deposits in transgenic worm was significantly decreased. In addition, CSE also protected the worms from oxidative stress and rescued chemotaxis dysfunction in transgenic strain CL2355. Taken together, these data suggested that CSE could protect against Aβ-induced toxicity in C. elegans. These results offer valuable evidence for the future use of CSE in the development of agents for the treatment of AD.
Collapse
|
23
|
G G, Singh J. Dithiothreitol causes toxicity in C. elegans by modulating the methionine-homocysteine cycle. eLife 2022; 11:76021. [PMID: 35438636 PMCID: PMC9090326 DOI: 10.7554/elife.76021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 04/17/2022] [Indexed: 11/13/2022] Open
Abstract
The redox reagent dithiothreitol (DTT) causes stress in the endoplasmic reticulum (ER) by disrupting its oxidative protein folding environment, which results in the accumulation and misfolding of the newly synthesized proteins. DTT may potentially impact cellular physiology by ER-independent mechanisms; however, such mechanisms remain poorly characterized. Using the nematode model Caenorhabditis elegans, here we show that DTT toxicity is modulated by the bacterial diet. Specifically, the dietary component vitamin B12 alleviates DTT toxicity in a methionine synthase-dependent manner. Using a forward genetic screen, we discover that loss-of-function of R08E5.3, an S-adenosylmethionine (SAM)-dependent methyltransferase, confers DTT resistance. DTT upregulates R08E5.3 expression and modulates the activity of the methionine–homocysteine cycle. Employing genetic and biochemical studies, we establish that DTT toxicity is a result of the depletion of SAM. Finally, we show that a functional IRE-1/XBP-1 unfolded protein response pathway is required to counteract toxicity at high, but not low, DTT concentrations. Animal and plant cells synthesize a significant fraction of their proteins on a structure known as the endoplasmic reticulum. Researchers often use the molecule dithiothreitol to specifically target this compartment and learn more about its role. The toxin works by disturbing the complex chemical environment present in the reticulum, which is required for the proteins to assemble properly. However, it is important to clarify whether dithiothreitol could also affect other parts of the cell, as this could give rise to misleading results. To explore this possibility, Gokul G and Jogender Singh studied the effects of dithiothreitol on the millimetre-long roundworm Caenorhabditis elegans. Their experiments revealed that vitamin B12 could protect against dithiothreitol toxicity via a complex cascade of molecular events which reduced the levels of an important regulatory molecule known as S-adenosylmethionine. Crucially, the chemical reactions that dithiothreitol targeted took place outside the reticulum, suggesting that the toxin impairs processes in the wider cell. These results suggest that dithiothreitol should be reconsidered for use in endoplasmic reticulum studies. However, they also imply that this toxin could be beneficial in small doses, as a reduced concentration of S-adenosylmethionine increases lifespan and health in a variety of organisms.
Collapse
Affiliation(s)
- Gokul G
- Indian Institute of Science Education and Research, Bhopal, Bhopal, India
| | - Jogender Singh
- Indian Institute of Science Education and Research, Mohali, Mohali, India
| |
Collapse
|
24
|
Kirchweger B, Klein-Junior LC, Pretsch D, Chen Y, Cretton S, Gasper AL, Heyden YV, Christen P, Kirchmair J, Henriques AT, Rollinger JM. Azepine-Indole Alkaloids From Psychotria nemorosa Modulate 5-HT 2A Receptors and Prevent in vivo Protein Toxicity in Transgenic Caenorhabditis elegans. Front Neurosci 2022; 16:826289. [PMID: 35360162 PMCID: PMC8963987 DOI: 10.3389/fnins.2022.826289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/18/2022] [Indexed: 11/26/2022] Open
Abstract
Nemorosine A (1) and fargesine (2), the main azepine-indole alkaloids of Psychotria nemorosa, were explored for their pharmacological profile on neurodegenerative disorders (NDs) applying a combined in silico-in vitro-in vivo approach. By using 1 and 2 as queries for similarity-based searches of the ChEMBL database, structurally related compounds were identified to modulate the 5-HT2A receptor; in vitro experiments confirmed an agonistic effect for 1 and 2 (24 and 36% at 10 μM, respectively), which might be linked to cognition-enhancing properties. This and the previously reported target profile of 1 and 2, which also includes BuChE and MAO-A inhibition, prompted the evaluation of these compounds in several Caenorhabditis elegans models linked to 5-HT modulation and proteotoxicity. On C. elegans transgenic strain CL4659, which expresses amyloid beta (Aβ) in muscle cells leading to a phenotypic paralysis, 1 and 2 reduced Aβ proteotoxicity by reducing the percentage of paralyzed worms to 51%. Treatment of the NL5901 strain, in which α-synuclein is yellow fluorescent protein (YFP)-tagged, with 1 and 2 (10 μM) significantly reduced the α-synuclein expression. Both alkaloids were further able to significantly extend the time of metallothionein induction, which is associated with reduced neurodegeneration of aged brain tissue. These results add to the multitarget profiles of 1 and 2 and corroborate their potential in the treatment of NDs.
Collapse
Affiliation(s)
- Benjamin Kirchweger
- Department of Pharmaceutical Sciences, Division of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Luiz C. Klein-Junior
- School of Health Sciences, Universidade do Vale do Itajaí (UNIVALI), Itajaí, Brazil
- Laboratory of Pharmacognosy and Quality Control of Phytomedicines, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Dagmar Pretsch
- Department of Pharmaceutical Sciences, Division of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Ya Chen
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Sylvian Cretton
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - André L. Gasper
- Herbarium Dr. Roberto Miguel Klein, Department of Natural Sciences, Universidade Regional de Blumenau (FURB), Blumenau, Brazil
| | - Yvan Vander Heyden
- Department of Analytical Chemistry, Applied Chemometrics and Molecular Modeling, Center for Pharmaceutical Research (CePhaR), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Philippe Christen
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Johannes Kirchmair
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Amélia T. Henriques
- Laboratory of Pharmacognosy and Quality Control of Phytomedicines, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Judith M. Rollinger
- Department of Pharmaceutical Sciences, Division of Pharmacognosy, University of Vienna, Vienna, Austria
| |
Collapse
|
25
|
Chen Y, Culetto E, Legouis R. The strange case of Drp1 in autophagy: Jekyll and Hyde? Bioessays 2022; 44:e2100271. [PMID: 35166388 DOI: 10.1002/bies.202100271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 12/27/2022]
Abstract
There is a debate regarding the function of Drp1, a GTPase involved in mitochondrial fission, during the elimination of mitochondria by autophagy. A number of experiments indicate that Drp1 is needed to eliminate mitochondria during mitophagy, either by reducing the mitochondrial size or by providing a noncanonical mitophagy function. Yet, other convincing experimental results support the conclusion that Drp1 is not necessary. Here, we review the possible functions for Drp1 in mitophagy and autophagy, depending on tissues, organisms and stresses, and discuss these apparent discrepancies. In this regard, it appears that the reduction of mitochondria size is often required for mitophagy but not always in a Drp1-dependent manner. Finally, we speculate on Drp1-independent mitochondrial fission mechanism that may take place during mitophagy and on noncanonical roles, which Drp1 may play such as modulating organelle contact sites dynamic during the autophagosome formation.
Collapse
Affiliation(s)
- Yanfang Chen
- College of Life Sciences, Animal Ressources Center, Nankai University, Tianjin, China
| | - Emmanuel Culetto
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, Paris, France.,INSERM, U1280, Gif-sur-Yvette, France
| | - Renaud Legouis
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, Paris, France.,INSERM, U1280, Gif-sur-Yvette, France
| |
Collapse
|
26
|
Modeling Alzheimer's Disease in Caenorhabditis elegans. Biomedicines 2022; 10:biomedicines10020288. [PMID: 35203497 PMCID: PMC8869312 DOI: 10.3390/biomedicines10020288] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/18/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer’s disease (AD) is the most frequent cause of dementia. After decades of research, we know the importance of the accumulation of protein aggregates such as β-amyloid peptide and phosphorylated tau. We also know that mutations in certain proteins generate early-onset Alzheimer’s disease (EOAD), and many other genes modulate the disease in its sporadic form. However, the precise molecular mechanisms underlying AD pathology are still unclear. Because of ethical limitations, we need to use animal models to investigate these processes. The nematode Caenorhabditis elegans has received considerable attention in the last 25 years, since the first AD models overexpressing Aβ peptide were described. We review here the main results obtained using this model to study AD. We include works studying the basic molecular mechanisms of the disease, as well as those searching for new therapeutic targets. Although this model also has important limitations, the ability of this nematode to generate knock-out or overexpression models of any gene, single or combined, and to carry out toxicity, recovery or survival studies in short timeframes with many individuals and at low cost is difficult to overcome. We can predict that its use as a model for various diseases will certainly continue to increase.
Collapse
|
27
|
Lauer AA, Grimm HS, Apel B, Golobrodska N, Kruse L, Ratanski E, Schulten N, Schwarze L, Slawik T, Sperlich S, Vohla A, Grimm MOW. Mechanistic Link between Vitamin B12 and Alzheimer's Disease. Biomolecules 2022; 12:129. [PMID: 35053277 PMCID: PMC8774227 DOI: 10.3390/biom12010129] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 01/27/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia in the elderly population, affecting over 55 million people worldwide. Histopathological hallmarks of this multifactorial disease are an increased plaque burden and tangles in the brains of affected individuals. Several lines of evidence indicate that B12 hypovitaminosis is linked to AD. In this review, the biochemical pathways involved in AD that are affected by vitamin B12, focusing on APP processing, Aβ fibrillization, Aβ-induced oxidative damage as well as tau hyperphosphorylation and tau aggregation, are summarized. Besides the mechanistic link, an overview of clinical studies utilizing vitamin B supplementation are given, and a potential link between diseases and medication resulting in a reduced vitamin B12 level and AD are discussed. Besides the disease-mediated B12 hypovitaminosis, the reduction in vitamin B12 levels caused by an increasing change in dietary preferences has been gaining in relevance. In particular, vegetarian and vegan diets are associated with vitamin B12 deficiency, and therefore might have potential implications for AD. In conclusion, our review emphasizes the important role of vitamin B12 in AD, which is particularly important, as even in industrialized countries a large proportion of the population might not be sufficiently supplied with vitamin B12.
Collapse
Affiliation(s)
- Anna Andrea Lauer
- Experimental Neurology, Saarland University, 66424 Homburg, Germany; (A.A.L.); (H.S.G.)
| | - Heike Sabine Grimm
- Experimental Neurology, Saarland University, 66424 Homburg, Germany; (A.A.L.); (H.S.G.)
| | - Birgit Apel
- Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany; (B.A.); (N.G.); (L.K.); (E.R.); (N.S.); (L.S.); (T.S.); (S.S.); (A.V.)
| | - Nataliya Golobrodska
- Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany; (B.A.); (N.G.); (L.K.); (E.R.); (N.S.); (L.S.); (T.S.); (S.S.); (A.V.)
| | - Lara Kruse
- Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany; (B.A.); (N.G.); (L.K.); (E.R.); (N.S.); (L.S.); (T.S.); (S.S.); (A.V.)
| | - Elina Ratanski
- Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany; (B.A.); (N.G.); (L.K.); (E.R.); (N.S.); (L.S.); (T.S.); (S.S.); (A.V.)
| | - Noemi Schulten
- Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany; (B.A.); (N.G.); (L.K.); (E.R.); (N.S.); (L.S.); (T.S.); (S.S.); (A.V.)
| | - Laura Schwarze
- Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany; (B.A.); (N.G.); (L.K.); (E.R.); (N.S.); (L.S.); (T.S.); (S.S.); (A.V.)
| | - Thomas Slawik
- Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany; (B.A.); (N.G.); (L.K.); (E.R.); (N.S.); (L.S.); (T.S.); (S.S.); (A.V.)
| | - Saskia Sperlich
- Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany; (B.A.); (N.G.); (L.K.); (E.R.); (N.S.); (L.S.); (T.S.); (S.S.); (A.V.)
| | - Antonia Vohla
- Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany; (B.A.); (N.G.); (L.K.); (E.R.); (N.S.); (L.S.); (T.S.); (S.S.); (A.V.)
| | - Marcus Otto Walter Grimm
- Experimental Neurology, Saarland University, 66424 Homburg, Germany; (A.A.L.); (H.S.G.)
- Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany; (B.A.); (N.G.); (L.K.); (E.R.); (N.S.); (L.S.); (T.S.); (S.S.); (A.V.)
- Deutsches Institut für DemenzPrävention, Saarland University, 66424 Homburg, Germany
| |
Collapse
|
28
|
Lionaki E, Ploumi C, Tavernarakis N. One-Carbon Metabolism: Pulling the Strings behind Aging and Neurodegeneration. Cells 2022; 11:cells11020214. [PMID: 35053330 PMCID: PMC8773781 DOI: 10.3390/cells11020214] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 01/27/2023] Open
Abstract
One-carbon metabolism (OCM) is a network of biochemical reactions delivering one-carbon units to various biosynthetic pathways. The folate cycle and methionine cycle are the two key modules of this network that regulate purine and thymidine synthesis, amino acid homeostasis, and epigenetic mechanisms. Intersection with the transsulfuration pathway supports glutathione production and regulation of the cellular redox state. Dietary intake of micronutrients, such as folates and amino acids, directly contributes to OCM, thereby adapting the cellular metabolic state to environmental inputs. The contribution of OCM to cellular proliferation during development and in adult proliferative tissues is well established. Nevertheless, accumulating evidence reveals the pivotal role of OCM in cellular homeostasis of non-proliferative tissues and in coordination of signaling cascades that regulate energy homeostasis and longevity. In this review, we summarize the current knowledge on OCM and related pathways and discuss how this metabolic network may impact longevity and neurodegeneration across species.
Collapse
Affiliation(s)
- Eirini Lionaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 70013 Heraklion, Crete, Greece; (E.L.); (C.P.)
| | - Christina Ploumi
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 70013 Heraklion, Crete, Greece; (E.L.); (C.P.)
- Department of Basic Sciences, Faculty of Medicine, University of Crete, 70013 Heraklion, Crete, Greece
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 70013 Heraklion, Crete, Greece; (E.L.); (C.P.)
- Department of Basic Sciences, Faculty of Medicine, University of Crete, 70013 Heraklion, Crete, Greece
- Correspondence: ; Tel.: +30-2810-391069
| |
Collapse
|
29
|
An R, Li D, Dong Y, She Q, Zhou T, Nie X, Pan R, Deng Y. Methylcobalamin Protects Melanocytes from H 2O 2-Induced Oxidative Stress by Activating the Nrf2/HO-1 Pathway. Drug Des Devel Ther 2021; 15:4837-4848. [PMID: 34876806 PMCID: PMC8643160 DOI: 10.2147/dddt.s336066] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/11/2021] [Indexed: 12/28/2022] Open
Abstract
Purpose Oxidative stress is considered a major determinant in the pathogenesis of vitiligo. Methylcobalamin (MeCbl) is an activated form of vitamin B12 that regulates inflammatory factors, counters oxidative stress, and reduces apoptosis in many disease models. However, the specific mechanism of MeCbl repigmentation against vitiligo is unknown. In this study, we explored the effect of MeCbl on melanocytes following hydrogen peroxide (H2O2)-induced oxidative stress. Methods We established an oxidative stress model using the immortalized human normal melanocyte cell line PIG1. We used a Cell Counting Kit-8 (CCK-8) to detect drug cytotoxicity, and we measured the melanin content of cells using the NaOH method. Intracellular oxidative damage was assessed by flow cytometry and antioxidant enzyme detection kits. In addition, we assessed the presence of apoptosis by flow cytometry and Western blots. We explored the underlying mechanisms of MeCbl during oxidative stress in melanocytes by analyzing the results of experiments based on real-time quantitative polymerase chain reaction (RT-qPCR), Western blotting, and laser scanning confocal immunofluorescence microscopy. Finally, we repeated the experiments after applying an inhibitor to block the Nrf2 pathway. Results We found that MeCbl treatment enhanced cell viability, increased melanin content, reduced intracellular reactive oxygen species (ROS) accumulation, increased the activities of antioxidant enzyme superoxide dismutase (SOD) and catalase (CAT), reduced melanocyte apoptosis, and up-regulated the expression of the Nrf2/HO-1 pathway. Moreover, the protective effects of MeCbl were significantly weakened after inhibiting the Nrf2/HO-1 pathway. Conclusion Our results indicate that MeCbl attenuated the H2O2-induced oxidative stress in melanocytes by activating the Nrf2/HO-1 pathway, this suggests that MeCbl may be an effective treatment against vitiligo.
Collapse
Affiliation(s)
- Ran An
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.,Department of Dermatology, Children's Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Dong Li
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yingying Dong
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Qiuyun She
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Ting Zhou
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Xiaoqi Nie
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Ronghua Pan
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yunhua Deng
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| |
Collapse
|