101
|
Xu Lou I, Ali K, Chen Q. Effect of nutrition in Alzheimer's disease: A systematic review. Front Neurosci 2023; 17:1147177. [PMID: 37214392 PMCID: PMC10194838 DOI: 10.3389/fnins.2023.1147177] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/01/2023] [Indexed: 05/24/2023] Open
Abstract
Background and objective Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by declining cognitive ability. Currently, there are no effective treatments for this condition. However, certain measures, such as nutritional interventions, can slow disease progression. Therefore, the objective of this systematic review was to identify and map the updates of the last 5 years regarding the nutritional status and nutritional interventions associated with AD patients. Study design A systematic review. Methods A search was conducted for randomized clinical trials, systematic reviews, and meta-analyses investigating the association between nutritional interventions and AD published between 2018 and 2022 in the PubMed, Web of Science, Scopus, and Cochrane Library databases. A total of 38 studies were identified, of which 17 were randomized clinical trials, and 21 were systematic reviews and/or meta-analyses. Results The results show that the western diet pattern is a risk factor for developing AD. In contrast, the Mediterranean diet, ketogenic diet, and supplementation with omega-3 fatty acids and probiotics are protective factors. This effect is significant only in cases of mild-to-moderate AD. Conclusion Certain nutritional interventions may slow the progression of AD and improve cognitive function and quality of life. Further research is required to draw more definitive conclusions.
Collapse
Affiliation(s)
- Inmaculada Xu Lou
- International Education College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Department of Cardiology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Kamran Ali
- Department of Oncology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qilan Chen
- Department of Cardiology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
102
|
Li D, Yang H, Lyu M, Zhou L, Zhang Y, Kang C, Wang J, Wang Y. Association between Behavioural Risks and Alzheimer's Disease: Elucidated with an Integrated Analysis of Gene Expression Patterns and Molecular Mechanisms. Neurosci Biobehav Rev 2023; 150:105207. [PMID: 37146892 DOI: 10.1016/j.neubiorev.2023.105207] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/12/2023] [Accepted: 04/30/2023] [Indexed: 05/07/2023]
Abstract
Alzheimer's disease (AD) remains a global health challenge. Previous studies have reported linkages between AD and multiple behavioural risk exposures, however, the underlying biological mechanisms and crucial genes of gene expression patterns driven by behavioural risks on the onset or progression of AD remains ambiguous. In this study, we performed an integrated analysis on the influence of behavioural risks including smoking, excessive alcohol consumption, physical inactivity, and non-healthy dietary pattern on AD with a comprehensive strategy. Our results demonstrated that multiple behavioural risk exposures could independently or collectively influence diverse hierarchical levels of gene expression patterns through multiple biological mechanisms such as Wnt, mitogen-activated protein kinase (MAPK), AMP-activated protein kinase (AMPK), nuclear factor (NF)-κB, phosphatidylinositol 3-kinase (PI3K)-Akt, and insulin (INS) signalling pathways-mediated pathological processes, thereby prodromally or intermediately impacting AD. Our study provided insights into understanding the association of behavioural risk exposures with AD and informative support for further studies.
Collapse
Affiliation(s)
- Dun Li
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Hongxi Yang
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Mingqian Lyu
- Department of Computer Science, RWTH Aachen University, Aachen, 52062, Germany
| | - Lihui Zhou
- School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Yuan Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Chunsheng Kang
- epartment of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Ju Wang
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, 300070, China
| | - Yaogang Wang
- School of Integrative Medicine, Public Health Science and Engineering College, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; School of Public Health, Tianjin Medical University, Tianjin, 300070, China; National Institute of Health Data Science at Peking University, Peking University, Beijing, 100191, China.
| |
Collapse
|
103
|
Zhang Y, Shen Y, Liufu N, Liu L, Li W, Shi Z, Zheng H, Mei X, Chen CY, Jiang Z, Abtahi S, Dong Y, Liang F, Shi Y, Cheng L, Yang G, Kang JX, Wilkinson J, Xie Z. Transmission of Alzheimer's Disease-Associated Microbiota Dysbiosis and its Impact on Cognitive Function: Evidence from Mouse Models and Human Patients. RESEARCH SQUARE 2023:rs.3.rs-2790988. [PMID: 37162940 PMCID: PMC10168447 DOI: 10.21203/rs.3.rs-2790988/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Spouses of Alzheimer's disease (AD) patients are at higher risk of developing AD dementia, but the reasons and underlying mechanism are unknown. One potential factor is gut microbiota dysbiosis, which has been associated with AD. However, it remains unclear whether the gut microbiota dysbiosis can be transmitted to non-AD individuals and contribute to the development of AD pathogenesis and cognitive impairment. The present study found that co-housing wild-type mice with AD transgenic mice or giving them AD transgenic mice feces caused AD-associated gut microbiota dysbiosis, Tau phosphorylation, and cognitive impairment. Gavage with Lactobacillus and Bifidobacterium restored these changes. The oral and gut microbiota of AD patient partners resembled that of AD patients but differed from healthy controls, indicating the transmission of oral and gut microbiota and its impact on cognitive function. The underlying mechanism of these findings includes that the butyric acid-mediated acetylation of GSK3β at lysine 15 regulated its phosphorylation at serine 9, consequently impacting Tau phosphorylation. These results provide insight into a potential link between gut microbiota dysbiosis and AD and underscore the need for further research in this area.
Collapse
Affiliation(s)
| | - Yuan Shen
- Tenth People's Hospital of Tongji University
| | | | | | - Wei Li
- Massachusetts General Hospital
| | | | | | | | | | | | | | - Yuanlin Dong
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School
| | | | | | | | - Guang Yang
- Department of Anesthesiology, Columbia University
| | | | | | | |
Collapse
|
104
|
Li J, Zhang F, Zhao L, Dong C. Microbiota-gut-brain axis and related therapeutics in Alzheimer's disease: prospects for multitherapy and inflammation control. Rev Neurosci 2023:revneuro-2023-0006. [PMID: 37076953 DOI: 10.1515/revneuro-2023-0006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/26/2023] [Indexed: 04/21/2023]
Abstract
Alzheimer's disease (AD) is the most common type of dementia in the elderly and causes neurodegeneration, leading to memory loss, behavioral disorder, and psychiatric impairment. One potential mechanism contributing to the pathogenesis of AD may be the imbalance in gut microbiota, local and systemic inflammation, and dysregulation of the microbiota-gut-brain axis (MGBA). Most of the AD drugs approved for clinical use today are symptomatic treatments that do not improve AD pathologic changes. As a result, researchers are exploring novel therapeutic modalities. Treatments involving the MGBA include antibiotics, probiotics, transplantation of fecal microbiota, botanical products, and others. However, single-treatment modalities are not as effective as expected, and a combination therapy is gaining momentum. The purpose of this review is to summarize recent advances in MGBA-related pathological mechanisms and treatment modalities in AD and to propose a new concept of combination therapy. "MGBA-based multitherapy" is an emerging view of treatment in which classic symptomatic treatments and MGBA-based therapeutic modalities are used in combination. Donepezil and memantine are two commonly used drugs in AD treatment. On the basis of the single/combined use of these two drugs, two/more additional drugs and treatment modalities that target the MGBA are chosen based on the characteristics of the patient's condition as an adjuvant treatment, as well as the maintenance of good lifestyle habits. "MGBA-based multitherapy" offers new insights for the treatment of cognitive impairment in AD patients and is expected to show good therapeutic results.
Collapse
Affiliation(s)
- Jiahao Li
- Department of Neurology, The First Affiliated Hospital, Dalian Medical University, No. 222 Zhongshan Road, Dalian 116011, China
| | - Feng Zhang
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Li Zhao
- Department of Neurology, The First Affiliated Hospital, Dalian Medical University, No. 222 Zhongshan Road, Dalian 116011, China
| | - Chunbo Dong
- Department of Neurology, The First Affiliated Hospital, Dalian Medical University, No. 222 Zhongshan Road, Dalian 116011, China
| |
Collapse
|
105
|
Afsar A, Chacon Castro MDC, Soladogun AS, Zhang L. Recent Development in the Understanding of Molecular and Cellular Mechanisms Underlying the Etiopathogenesis of Alzheimer's Disease. Int J Mol Sci 2023; 24:7258. [PMID: 37108421 PMCID: PMC10138573 DOI: 10.3390/ijms24087258] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that leads to dementia and patient death. AD is characterized by intracellular neurofibrillary tangles, extracellular amyloid beta (Aβ) plaque deposition, and neurodegeneration. Diverse alterations have been associated with AD progression, including genetic mutations, neuroinflammation, blood-brain barrier (BBB) impairment, mitochondrial dysfunction, oxidative stress, and metal ion imbalance.Additionally, recent studies have shown an association between altered heme metabolism and AD. Unfortunately, decades of research and drug development have not produced any effective treatments for AD. Therefore, understanding the cellular and molecular mechanisms underlying AD pathology and identifying potential therapeutic targets are crucial for AD drug development. This review discusses the most common alterations associated with AD and promising therapeutic targets for AD drug discovery. Furthermore, it highlights the role of heme in AD development and summarizes mathematical models of AD, including a stochastic mathematical model of AD and mathematical models of the effect of Aβ on AD. We also summarize the potential treatment strategies that these models can offer in clinical trials.
Collapse
Affiliation(s)
| | | | | | - Li Zhang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
106
|
Giovana Maciel Reis C, Rocha-Gomes A, Escobar Teixeira A, Gomes de Oliveira D, Mainy Oliveira Santiago C, Alves da Silva A, Regina Riul T, de Jesus Oliveira E. Short-term Cafeteria Diet Is Associated with Fat Mass Accumulation, Systemic and Amygdala Inflammation, and Anxiety-like Behavior in Adult Male Wistar Rats. Neuroscience 2023; 515:37-52. [PMID: 36773840 DOI: 10.1016/j.neuroscience.2023.02.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/31/2023] [Accepted: 02/03/2023] [Indexed: 02/12/2023]
Abstract
Obesity is linked to metabolic, hormonal and biochemical alterations, and is also a risk factor for behavioral disorders. Evidence suggests that these disorders may be related to the consumption of hypercaloric diets, fat mass accumulation and changes in inflammation and redox status. Although much is known about the chronic effects of hypercaloric diets on mental health, few studies have evaluated the consequences of short-term exposure of these diets on behavior. The aim of this study was to evaluate nutritional, behavioral (anxiety-like), inflammatory and redox status parameters in adult male Wistar rats exposed to short-term cafeteria diet. Adult Wistar male rats (90 days-old; n = 12/group) received, during 14 days, the diets: Control- standard diet; Simple Cafeteria Diet (SCD)- homogeneous cafeteria diet. Varied Cafeteria Diet (VCD)- cafeteria diet with rotation and variation. Nutritional analyzes and tests for anxiety-like behaviors were performed, in addition to inflammatory and redox status measurements in blood and amygdala. The SCD group showed higher fat energy intake, while the VCD group consumed more energy from carbohydrates. SCD and VCD showed higher fat mass accumulation, in addition to higher levels of TNFα, INFγ, TBARS and FRAP in the blood. Also, SCD and VCD groups reported high levels of TNFα in the amygdala. Regarding behavioral evaluations, SCD and VCD groups showed anxiogenesis in the elevated plus maze, light-dark box, and open field tests. Therefore, the two cafeteria diets induced obesity and systemic inflammation, which in turn, resulted in an increase in amygdala TNFα levels and anxiety-like behaviors in Wistar rats.
Collapse
Affiliation(s)
- Clarisse Giovana Maciel Reis
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil; Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil
| | - Arthur Rocha-Gomes
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (USP), Brazil
| | - Amanda Escobar Teixeira
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil
| | - Dalila Gomes de Oliveira
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil; Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil
| | - Camilla Mainy Oliveira Santiago
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil; Programa de Pós-Graduação em Ciências da Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil
| | - Alexandre Alves da Silva
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil; Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil
| | - Tania Regina Riul
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil; Programa de Pós-Graduação em Ciências da Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil
| | - Eduardo de Jesus Oliveira
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil.
| |
Collapse
|
107
|
Nemeth Z, Patonai A, Simon-Szabó L, Takács I. Interplay of Vitamin D and SIRT1 in Tissue-Specific Metabolism-Potential Roles in Prevention and Treatment of Non-Communicable Diseases Including Cancer. Int J Mol Sci 2023; 24:ijms24076154. [PMID: 37047134 PMCID: PMC10094444 DOI: 10.3390/ijms24076154] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 04/14/2023] Open
Abstract
The importance of the prevention and control of non-communicable diseases, including obesity, metabolic syndrome, type 2 diabetes, cardiovascular diseases, and cancer, is increasing as a requirement of the aging population in developed countries and the sustainability of healthcare. Similarly, the 2013-2030 action plan of the WHO for the prevention and control of non-communicable diseases seeks these achievements. Adequate lifestyle changes, alone or with the necessary treatments, could reduce the risk of mortality or the deterioration of quality of life. In our recent work, we summarized the role of two central factors, i.e., appropriate levels of vitamin D and SIRT1, which are connected to adequate lifestyles with beneficial effects on the prevention and control of non-communicable diseases. Both of these factors have received increased attention in relation to the COVID-19 pandemic as they both take part in regulation of the main metabolic processes, i.e., lipid/glucose/energy homeostasis, oxidative stress, redox balance, and cell fate, as well as in the healthy regulation of the immune system. Vitamin D and SIRT1 have direct and indirect influence of the regulation of transcription and epigenetic changes and are related to cytoplasmic signaling pathways such as PLC/DAG/IP3/PKC/MAPK, MEK/Erk, insulin/mTOR/cell growth, proliferation; leptin/PI3K-Akt-mTORC1, Akt/NFĸB/COX-2, NFĸB/TNFα, IL-6, IL-8, IL-1β, and AMPK/PGC-1α/GLUT4, among others. Through their proper regulation, they maintain normal body weight, lipid profile, insulin secretion and sensitivity, balance between the pro- and anti-inflammatory processes under normal conditions and infections, maintain endothelial health; balance cell differentiation, proliferation, and fate; and balance the circadian rhythm of the cellular metabolism. The role of these two molecules is interconnected in the molecular network, and they regulate each other in several layers of the homeostasis of energy and the cellular metabolism. Both have a central role in the maintenance of healthy and balanced immune regulation and redox reactions; therefore, they could constitute promising targets either for prevention or as complementary therapies to achieve a better quality of life, at any age, for healthy people and patients under chronic conditions.
Collapse
Affiliation(s)
- Zsuzsanna Nemeth
- Department of Internal Medicine and Oncology, Semmelweis University, Koranyi S. u 2/a, 1083 Budapest, Hungary
| | - Attila Patonai
- Department of Surgery, Transplantation and Gastroenterology, Semmelweis University, Ulloi u. 78, 1082 Budapest, Hungary
| | - Laura Simon-Szabó
- Department of Molecular Biology, Semmelweis University, Tuzolto u. 37-47, 1094 Budapest, Hungary
| | - István Takács
- Department of Internal Medicine and Oncology, Semmelweis University, Koranyi S. u 2/a, 1083 Budapest, Hungary
| |
Collapse
|
108
|
Effects of High-Fat and High-Fat High-Sugar Diets in the Anxiety, Learning and Memory, and in the Hippocampus Neurogenesis and Neuroinflammation of Aged Rats. Nutrients 2023; 15:nu15061370. [PMID: 36986100 PMCID: PMC10053405 DOI: 10.3390/nu15061370] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
High-caloric diets induce several deleterious alterations in the human body, including the brain. However, information on the effects of these diets on the elderly brain is scarce. Therefore, we studied the effects of 2 months of treatment with high-fat (HF) and high-fat-high-sugar (HFHS) diets on aged male Wistar rats at 18 months. Anxiety levels were analyzed using the open-field and plus-maze tests, while learning and memory processes were analyzed using the Morris water maze test. We also analyzed neurogenesis using doublecortin (DCX) and neuroinflammation using glial fibrillary acidic protein (GFAP). In aged rats, the HFHS diet impaired spatial learning, memory, and working memory and increased anxiety levels, associated with a reduction in the number of DCX cells and an increase in GFAP cells in the hippocampus. In contrast, the effects of the HF diet were lighter, impairing spatial memory and working memory, and associated with a reduction in DCX cells in the hippocampus. Thus, our results suggest that aged rats are highly susceptible to high-caloric diets, even if they only started in the elderly, with an impact on cognition and emotions. Furthermore, diets rich in saturated fats and sugar are more detrimental to aged rats than high-fat diets are.
Collapse
|
109
|
Circular RNAs in Parkinson's Disease: Reliable Biological Markers and Targets for Rehabilitation. Mol Neurobiol 2023; 60:3261-3276. [PMID: 36840847 DOI: 10.1007/s12035-023-03268-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 02/14/2023] [Indexed: 02/26/2023]
Abstract
In clinical practice, the underlying pathogenesis of Parkinson's disease (PD) remains unknown. Circular RNAs (circRNAs) have good biological properties and can be used as biological marker. Rehabilitation as a third treatment alongside drug and surgery has been shown to be clinically effective, but biomarkers of rehabilitation efficiency at genetic level is still lacking. In this study, we identified differentially expressed circRNAs in peripheral blood exosomes between PD patients and health controls (HCs) and determined whether these circRNAs changed after rehabilitation, to explore the competing RNA networks and epigenetic mechanisms affected. We found that there were 558 upregulated and 609 downregulated circRNAs in PD patients compared to HCs, 3398 upregulated and 479 downregulated circRNAs in PD patients after rehabilitation compared to them before rehabilitation, along with 3721 upregulated and 635 downregulated circRNAs in PD patients after rehabilitation compared to HCs. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that differentially expressed circRNAs may affect the stability of the cellular actin backbone and synaptic structure by influencing the aggregation of α-synuclein (a-syn). We selected two circRNAs overexpressed in PD patients for validation (hsa_circ_0001535 and hsa_circ_0000437); the results revealed that their expression levels were all reduced to varying degrees (p < 0.05) after rehabilitation. After network analysis, we believe that hsa_circ_0001535 may be related to the aggregation of a-syn, while hsa_circ_0000437 may act on hsa-let-7b-5p or hsa-let-7c-5p through sponge effect to cause inflammatory response. Our findings suggest that rehabilitation can mitigate the pathological process of PD by epigenetic means.
Collapse
|
110
|
Rice-memolin, a novel peptide derived from rice bran, improves cognitive function after oral administration in mice. Sci Rep 2023; 13:2887. [PMID: 36807368 PMCID: PMC9938899 DOI: 10.1038/s41598-023-30021-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/14/2023] [Indexed: 02/20/2023] Open
Abstract
Many people eat polished rice, while rice bran, a by-product known to be rich in protein and expected to have potential functions for health benefits, has not been effectively utilized. In this study, we determined that orally administered Val-Tyr-Thr-Pro-Gly (VYTPG) derived from rice bran protein improved cognitive decline in mice fed a high-fat diet (HFD). It was demonstrated that VYTPG was released from model peptides corresponding to fragment sequences of original rice proteins (Os01g0941500, Os01g0872700, and allergenic protein) after treatment with thermolysin, a microorganism-derived enzyme often used in industrial scale processes. The thermolysin digest also improved cognitive decline after oral administration in mice. Because VYTPG (1.0 mg/kg) potently improved cognitive decline and is enzymatically produced from the rice bran, we named it rice-memolin. Next, we investigated the mechanisms underlying the cognitive decline improvement associated with rice-memolin. Methyllycaconitine, an antagonist for α7 nicotinic acetylcholine receptor, suppressed the rice-memolin-induced effect, suggesting that rice-memolin improved cognitive decline coupled to the acetylcholine system. Rice-memolin increased the number of 5-bromo-2'-deoxyuridine (BrdU)-positive cells and promoted the mRNA expression of EGF and FGF-2 in the hippocampus, implying that these neurotropic factors play a role in hippocampal neurogenesis after rice-memolin administration. Epidemiologic studies demonstrated that diabetes is a risk factor for dementia; therefore, we also examined the effect of rice-memolin on glucose metabolism. Rice-memolin improved glucose intolerance. In conclusion, we identified a novel rice-derived peptide that can improve cognitive decline. The mechanisms are associated with acetylcholine and hippocampal neurogenesis. Rice-memolin is the first rice-brain-derived peptide able to improve cognitive decline.
Collapse
|
111
|
Sebastian MJ, Khan SKA, Pappachan JM, Jeeyavudeen MS. Diabetes and cognitive function: An evidence-based current perspective. World J Diabetes 2023; 14:92-109. [PMID: 36926658 PMCID: PMC10011899 DOI: 10.4239/wjd.v14.i2.92] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/26/2022] [Accepted: 01/16/2023] [Indexed: 02/14/2023] Open
Abstract
Several epidemiological studies have clearly identified diabetes mellitus (DM) as a major risk factor for cognitive dysfunction, and it is going to be a major public health issue in the coming years because of the alarming rise in diabetes prevalence across the world. Brain and neural tissues predominantly depend on glucose as energy substrate and hence, any alterations in carbohydrate meta-bolism can directly impact on cerebral functional output including cognition, executive capacity, and memory. DM affects neuronal function and mental capacity in several ways, some of which include hypoperfusion of the brain tissues from cerebrovascular disease, diabetes-related alterations of glucose transporters causing abnormalities in neuronal glucose uptake and metabolism, local hyper- and hypometabolism of brain areas from insulin resistance, and recurrent hypoglycemic episodes inherent to pharmacotherapy of diabetes resulting in neuronal damage. Cognitive decline can further worsen diabetes care as DM is a disease largely self-managed by patients. Therefore, it is crucial to understand the pathobiology of cognitive dysfunction in relation to DM and its management for optimal long-term care plan for patients. A thorough appraisal of normal metabolic characteristics of the brain, how alterations in neural metabolism affects cognition, the diagnostic algorithm for patients with diabetes and dementia, and the management and prognosis of patients when they have this dangerous combination of illnesses is imperative in this context. This evidence-based narrative with the back-up of latest clinical trial reviews elaborates the current understanding on diabetes and cognitive function to empower physicians to manage their patients in day-to-day clinical practice.
Collapse
Affiliation(s)
| | - Shahanas KA Khan
- Department of Endocrinology and Metabolism, Lancashire Teaching Hospitals NHS Trust, Preston PR2 9HT, United Kingdom
| | - Joseph M Pappachan
- Department of Endocrinology and Metabolism, Lancashire Teaching Hospitals NHS Trust, Preston PR2 9HT, United Kingdom
- Faculty of Science, Manchester Metropolitan University, Manchester M15 6BH, United Kingdom
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - Mohammad Sadiq Jeeyavudeen
- Department of Endocrinology and Metabolism, University Hospitals of Edinburgh, Edinburgh EH16 4SA, United Kingdom
| |
Collapse
|
112
|
Rinoldi C, Ziai Y, Zargarian SS, Nakielski P, Zembrzycki K, Haghighat Bayan MA, Zakrzewska AB, Fiorelli R, Lanzi M, Kostrzewska-Księżyk A, Czajkowski R, Kublik E, Kaczmarek L, Pierini F. In Vivo Chronic Brain Cortex Signal Recording Based on a Soft Conductive Hydrogel Biointerface. ACS APPLIED MATERIALS & INTERFACES 2023; 15:6283-6296. [PMID: 36576451 DOI: 10.1021/acsami.2c17025] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
In neuroscience, the acquisition of neural signals from the brain cortex is crucial to analyze brain processes, detect neurological disorders, and offer therapeutic brain-computer interfaces. The design of neural interfaces conformable to the brain tissue is one of today's major challenges since the insufficient biocompatibility of those systems provokes a fibrotic encapsulation response, leading to an inaccurate signal recording and tissue damage precluding long-term/permanent implants. The design and production of a novel soft neural biointerface made of polyacrylamide hydrogels loaded with plasmonic silver nanocubes are reported herein. Hydrogels are surrounded by a silicon-based template as a supporting element for guaranteeing an intimate neural-hydrogel contact while making possible stable recordings from specific sites in the brain cortex. The nanostructured hydrogels show superior electroconductivity while mimicking the mechanical characteristics of the brain tissue. Furthermore, in vitro biological tests performed by culturing neural progenitor cells demonstrate the biocompatibility of hydrogels along with neuronal differentiation. In vivo chronic neuroinflammation tests on a mouse model show no adverse immune response toward the nanostructured hydrogel-based neural interface. Additionally, electrocorticography acquisitions indicate that the proposed platform permits long-term efficient recordings of neural signals, revealing the suitability of the system as a chronic neural biointerface.
Collapse
Affiliation(s)
- Chiara Rinoldi
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw02-106, Poland
| | - Yasamin Ziai
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw02-106, Poland
| | - Seyed Shahrooz Zargarian
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw02-106, Poland
| | - Paweł Nakielski
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw02-106, Poland
| | - Krzysztof Zembrzycki
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw02-106, Poland
| | - Mohammad Ali Haghighat Bayan
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw02-106, Poland
| | - Anna Beata Zakrzewska
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw02-106, Poland
| | - Roberto Fiorelli
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw02-106, Poland
| | - Massimiliano Lanzi
- Department of Industrial Chemistry "Toso Montanari", Alma Mater Studiorum University of Bologna, Bologna40136, Italy
| | | | - Rafał Czajkowski
- Nencki Institute of Experimental Biology Polish Academy of Sciences, Warsaw02-093, Poland
| | - Ewa Kublik
- Nencki Institute of Experimental Biology Polish Academy of Sciences, Warsaw02-093, Poland
| | - Leszek Kaczmarek
- Nencki Institute of Experimental Biology Polish Academy of Sciences, Warsaw02-093, Poland
| | - Filippo Pierini
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw02-106, Poland
| |
Collapse
|
113
|
Chandra S, Sisodia SS, Vassar RJ. The gut microbiome in Alzheimer's disease: what we know and what remains to be explored. Mol Neurodegener 2023; 18:9. [PMID: 36721148 PMCID: PMC9889249 DOI: 10.1186/s13024-023-00595-7] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/06/2023] [Indexed: 02/02/2023] Open
Abstract
Alzheimer's disease (AD), the most common cause of dementia, results in a sustained decline in cognition. There are currently few effective disease modifying therapies for AD, but insights into the mechanisms that mediate the onset and progression of disease may lead to new, effective therapeutic strategies. Amyloid beta oligomers and plaques, tau aggregates, and neuroinflammation play a critical role in neurodegeneration and impact clinical AD progression. The upstream modulators of these pathological features have not been fully clarified, but recent evidence indicates that the gut microbiome (GMB) may have an influence on these features and therefore may influence AD progression in human patients. In this review, we summarize studies that have identified alterations in the GMB that correlate with pathophysiology in AD patients and AD mouse models. Additionally, we discuss findings with GMB manipulations in AD models and potential GMB-targeted therapeutics for AD. Lastly, we discuss diet, sleep, and exercise as potential modifiers of the relationship between the GMB and AD and conclude with future directions and recommendations for further studies of this topic.
Collapse
Affiliation(s)
- Sidhanth Chandra
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
- Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Sangram S. Sisodia
- Department of Neurobiology, University of Chicago, Chicago, IL 60637 USA
| | - Robert J. Vassar
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| |
Collapse
|
114
|
Ghrelin system in Alzheimer's disease. Curr Opin Neurobiol 2023; 78:102655. [PMID: 36527939 PMCID: PMC10395051 DOI: 10.1016/j.conb.2022.102655] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/16/2022] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia in seniors. Current efforts to understand the etiopathogenesis of this neurodegenerative disorder have brought forth questions about systemic factors in the development of AD. Ghrelin is a brain-gut peptide that is activated by ghrelin O-acyltransferase (GOAT) and signals via its receptor, growth hormone secretagogue receptor (GHSR). With increasing recognition of the neurotropic effects of ghrelin, the role of ghrelin system deregulation in the development of AD has been accentuated in recent years. In this review, we summarized recent research progress regarding the mechanisms of ghrelin signaling dysregulation and its contribution to AD brain pathology. In addition, we also discussed the therapeutic potential of strategies targeting ghrelin signaling for the treatment of this neurological disease.
Collapse
|
115
|
Pleiotrophin deficiency protects against high-fat diet-induced neuroinflammation: Implications for brain mitochondrial dysfunction and aberrant protein aggregation. Food Chem Toxicol 2023; 172:113578. [PMID: 36566969 DOI: 10.1016/j.fct.2022.113578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/10/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Metabolic Syndrome (MetS) is a risk factor for the development of neurodegenerative diseases. Neuroinflammation associated with MetS may contribute significantly to neurodegeneration. Pleiotrophin (PTN) is a neurotrophic factor that modulates neuroinflammation and is a key player in regulating energy metabolism and thermogenesis, suggesting that PTN could be important in the connection between MetS and neuroinflammation. We have now used a high-fat diet (HFD)-induced obesity model in Ptn-/- mice. HFD and Ptn deletion caused alterations in circulating hormones including GIP, leptin and resistin. HFD produced in Ptn+/+ mice a neuroinflammatory state as observed in cerebral quantifications of proinflammatory markers, including Il1β, Tnfα and Ccl2. The upregulation of neuroinflammatory markers was prevented in Ptn-/- mice. Changes induced by HFD in genes related to mitochondrial biogenesis and dynamics were less pronounced in the brain of Ptn-/- mice and were accompanied by significant increases in the protein expression of mitochondrial oxidative phosphorylation (OXPHOS) complexes I and IV. HFD-induced changes in genes related to the elimination of protein aggregates were also less pronounced in the brain of Ptn-/- mice. This study provides substantial evidence that Ptn deletion protects against HFD-induced neuroinflammation, mitochondrial dysfunction, and aberrant protein aggregation, prominent features in neurodegenerative diseases.
Collapse
|
116
|
Liu Y, Tan Y, Zhang Z, Li H, Yi M, Zhang Z, Hui S, Peng W. Neuroimmune mechanisms underlying Alzheimer's disease: Insights into central and peripheral immune cell crosstalk. Ageing Res Rev 2023; 84:101831. [PMID: 36565960 DOI: 10.1016/j.arr.2022.101831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD) is a highly life-threatening neurodegenerative disease. Dysregulation of the immune system plays a critical role in promoting AD, which has attracted extensive attention recently. Central and peripheral immune responses are involved in the pathogenesis of AD. Immune changes precede Aβ-associated senile plaque formation and tau-related neurofibrillary tangles, which are the recognised pathological features of AD. Therefore, elucidating immune-related mechanisms underlying the development of AD can help to prevent and treat AD at the source by blocking its progression before the development of pathological changes. To understand the specific pathogenesis of AD, it is important to examine the role of central and peripheral immunity in AD. This review summarises immune-related mechanisms underlying the pathogenesis of AD, focusing on the effect of various central and peripheral immune cells, and describes the possible crosstalk between central and peripheral immunity during the development of AD. This review provides novel insights into the treatment of AD and offers a new direction for immune-related research on AD in the future.
Collapse
Affiliation(s)
- Yuqing Liu
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.
| | - Yejun Tan
- School of Mathematics, University of Minnesota Twin Cities, Minneapolis, MN, USA.
| | - Zheyu Zhang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.
| | - Hongli Li
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.
| | - Min Yi
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.
| | - Zhen Zhang
- YangSheng College of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, Guizhou, China.
| | - Shan Hui
- Department of Geratology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, China.
| | - Weijun Peng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.
| |
Collapse
|
117
|
Chen J, Xiao Y, Li D, Zhang S, Wu Y, Zhang Q, Bai W. New insights into the mechanisms of high-fat diet mediated gut microbiota in chronic diseases. IMETA 2023; 2:e69. [PMID: 38868334 PMCID: PMC10989969 DOI: 10.1002/imt2.69] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 10/13/2022] [Accepted: 11/06/2022] [Indexed: 06/14/2024]
Abstract
High-fat diet (HFD) has been recognized as a primary factor in the risk of chronic disease. Obesity, diabetes, gastrointestinal diseases, neurodegenerative diseases, and cardiovascular diseases have long been known as chronic diseases with high worldwide incidence. In this review, the influences of gut microbiota and their corresponding bacterial metabolites on the mechanisms of HFD-induced chronic diseases are systematically summarized. Gut microbiota imbalance is also known to increase susceptibility to diseases. Several studies have proven that HFD has a negative impact on gut microbiota, also exacerbating the course of many chronic diseases through increased populations of Erysipelotrichaceae, facultative anaerobic bacteria, and opportunistic pathogens. Since bile acids, lipopolysaccharide, short-chain fatty acids, and trimethylamine N-oxide have long been known as common features of bacterial metabolites, we will explore the possibility of synergistic mechanisms among those metabolites and gut microbiota in the context of HFD-induced chronic diseases. Recent literature concerning the mechanistic actions of HFD-mediated gut microbiota have been collected from PubMed, Google Scholar, and Scopus. The aim of this review is to provide new insights into those mechanisms and to point out the potential biomarkers of HFD-mediated gut microbiota.
Collapse
Affiliation(s)
- Jiali Chen
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid DetectionJinan UniversityGuangzhouChina
- School of Chinese Medicine, Centre for Cancer and Inflammation ResearchHong Kong Baptist UniversityHong KongChina
| | - Yuhang Xiao
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid DetectionJinan UniversityGuangzhouChina
| | - Dongmei Li
- Department of Microbiology & ImmunologyGeorgetown University Medical CenterWashingtonDistrict of ColumbiaUSA
| | - Shiqing Zhang
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
| | - Yingzi Wu
- School of Chinese Medicine, Centre for Cancer and Inflammation ResearchHong Kong Baptist UniversityHong KongChina
| | - Qing Zhang
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid DetectionJinan UniversityGuangzhouChina
| | - Weibin Bai
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid DetectionJinan UniversityGuangzhouChina
| |
Collapse
|
118
|
Brikou D, Charisis S, Drouka A, Christodoulakou SM, Ntanasi E, Mamalaki E, Constadinides VC, Scarmeas N, Yannakoulia M. Daily Energy Intake Distribution and Cognitive Performance in Non-Demented Individuals. Nutrients 2023; 15:nu15030673. [PMID: 36771379 PMCID: PMC9921864 DOI: 10.3390/nu15030673] [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: 12/18/2022] [Revised: 01/18/2023] [Accepted: 01/21/2023] [Indexed: 02/01/2023] Open
Abstract
Cognitive disorders have become important public health issues around the world. Studies evaluating the association between cognitive decline and food timing are lacking. The objective of this study was to examine the potential association between energy intake distribution during the day and cognitive performance in cognitively healthy and mildly cognitive impaired individuals. Data were derived from the ongoing Albion study which includes people aged 40 years or older who have a positive family history of cognitive disorder or concern about their cognitive status. A thorough dietary and cognitive assessment was performed. Participants consuming low energy intake at the beginning of the day or high energy at the end of the day had higher cognitive function compared to participants characterized by the opposite pattern. This trend remained statistically significant even after adjustment for potential confounders (p = 0.043). This study suggests that individuals with worse cognitive function may choose to eat earlier during the day, when cognitive performance is better, and it might be hypothesized that a meal pattern characterized by high energy consumption at the beginning of the day or low energy at the end of the day could be a marker of cognitive impairment.
Collapse
Affiliation(s)
- Dora Brikou
- Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece
| | - Sokratis Charisis
- Department of Neurology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Archontoula Drouka
- Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece
| | | | - Eva Ntanasi
- Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece
- 1st Department of Neurology, Aiginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Eirini Mamalaki
- Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece
| | - Vasilios C. Constadinides
- 1st Department of Neurology, Aiginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Nikolaos Scarmeas
- 1st Department of Neurology, Aiginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
- The Gertrude H. Sergievsky Center, Taub Institute for Research in Alzheimer’s Disease and the Aging Brain, Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Mary Yannakoulia
- Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece
- Correspondence:
| |
Collapse
|
119
|
Liang S, Wang L, Wu X, Hu X, Wang T, Jin F. The different trends in the burden of neurological and mental disorders following dietary transition in China, the USA, and the world: An extension analysis for the Global Burden of Disease Study 2019. Front Nutr 2023; 9:957688. [PMID: 36698474 PMCID: PMC9869872 DOI: 10.3389/fnut.2022.957688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 12/06/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction The highly processed western diet is substituting the low-processed traditional diet in the last decades globally. Increasing research found that a diet with poor quality such as western diet disrupts gut microbiota and increases the susceptibility to various neurological and mental disorders, while a balanced diet regulates gut microbiota and prevents and alleviates the neurological and mental disorders. Yet, there is limited research on the association between the disease burden expanding of neurological and mental disorders with a dietary transition. Methods We compared the disability-adjusted life-years (DALYs) trend by age for neurological and mental disorders in China, in the United States of America (USA), and across the world from 1990 to 2019, evaluated the dietary transition in the past 60 years, and analyzed the association between the burden trend of the two disorders with the changes in diet composition and food production. Results We identified an age-related upward pattern in disease burden in China. Compared with the USA and the world, the Chinese neurological and mental disorders DALY percent was least in the generation over 75 but rapidly increased in younger generations and surpassed the USA and/or the world in the last decades. The age-related upward pattern in Chinese disease burdens had not only shown in the presence of cardiovascular diseases, neoplasms, and diabetes mellitus but also appeared in the presence of depressive disorders, Parkinson's disease, Alzheimer's disease and other dementias, schizophrenia, headache disorders, anxiety disorders, conduct disorders, autism spectrum disorders, and eating disorders, successively. Additionally, the upward trend was associated with the dramatic dietary transition including a reduction in dietary quality and food production sustainability, during which the younger generation is more affected than the older. Following the increase in total calorie intake, alcohol intake, ratios of animal to vegetal foods, and poultry meat to pulses, the burdens of the above diseases continuously rose. Then, following the rise of the ratios of meat to pulses, eggs to pulses, and pork to pulses, the usage of fertilizers, the farming density of pigs, and the burdens of the above disease except diabetes mellitus were also ever-increasing. Even the usage of pesticides was positively correlated with the burdens of Parkinson's disease, schizophrenia, cardiovascular diseases, and neoplasms. Contrary to China, the corresponding burdens of the USA trended to reduce with the improvements in diet quality and food production sustainability. Discussion Our results suggest that improving diet quality and food production sustainability might be a promising way to stop the expanding burdens of neurological and mental disorders.
Collapse
Affiliation(s)
- Shan Liang
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Microbial Physiological and Metabolic Engineering, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Gut-brain Psychology Laboratory, Beijing, China
| | - Li Wang
- Department for the History of Science and Scientific Archaeology, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaoli Wu
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Gut-brain Psychology Laboratory, Beijing, China
| | - Xu Hu
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Gut-brain Psychology Laboratory, Beijing, China
| | - Tao Wang
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Gut-brain Psychology Laboratory, Beijing, China
| | - Feng Jin
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Gut-brain Psychology Laboratory, Beijing, China
| |
Collapse
|
120
|
Liu Q, Li Z, Huang L, Zhou D, Fu J, Duan H, Wang Z, Yang T, Zhao J, Li W, Liu H, Ma F, Sun C, Wang G, Du Y, Zhang M, Chen Y, Huang G. Telomere and mitochondria mediated the association between dietary inflammatory index and mild cognitive impairment: A prospective cohort study. Immun Ageing 2023; 20:1. [PMID: 36604719 PMCID: PMC9813461 DOI: 10.1186/s12979-022-00326-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/28/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND Diet and chronic inflammation might play a major role in the pathogenesis of mild cognitive impairment (MCI). In addition, peripheral blood leukocyte telomere length (LTL) and mitochondrial DNA copy number (mtDNAcn) might mediate the relationship between inflammation and MCI risk. The purpose of the present study is to evaluate whether inflammatory potential of diet assessed by dietary inflammatory index (DII), chronic inflammation, peripheral blood LTL, and mtDNAcn were associated with the risk of MCI. RESULTS A population-based cohort study was conducted with a total of 2944 participants. During a median follow-up of 2 years, 438 (14.90%) individuals were new-onset MCI. After adjustment, a higher score of DII (hazard ratio [HR]: 1.056, 95% CI: 1.005, 1.109), a higher log systemic immune inflammation index (SII) (HR: 1.333, 95% CI: 1.089, 1.633) and log system inflammation response index (SIRI) (HR: 1.487, 95% CI: 1.024, 2.161) predicted elevated risk of MCI. An increased mtDNAcn (HR: 0.843, 95% CI: 0.712, 0.997), but not LTL, predicted a decreased risk of MCI. Negative associations of log SII with LTL (β:-0.359, 95% CI: -0.445, -0.273) and mtDNAcn (β:-0.048, 95% CI: -0.090, -0.006) were found. Additionally, negative associations of log SIRI with LTL (β: -0.035, 95% CI: -0.052, -0.017) and mtDNAcn (β:-0.136, 95% CI: -0.216, -0.056) were also found. Path analysis suggested that SIRI, LTL, and mtDNAcn, in series, have mediation roles in the association between DII score and MCI risk. CONCLUSIONS Higher DII, SII, and SIRI might predict a greater risk of MCI, while a longer LTL and an increased mtDNAcn were linked to a reduced risk of MCI among the older population. LTL and mtDNAcn could play mediation roles in the association between DII and MCI risk.
Collapse
Affiliation(s)
- Qian Liu
- grid.265021.20000 0000 9792 1228Department of Nutrition & Food Science, School of Public Health, Tianjin Medical University, Tianjin, China ,grid.265021.20000 0000 9792 1228Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Zhenshu Li
- grid.265021.20000 0000 9792 1228Department of Nutrition & Food Science, School of Public Health, Tianjin Medical University, Tianjin, China ,grid.265021.20000 0000 9792 1228Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Ling Huang
- grid.265021.20000 0000 9792 1228Department of Nutrition & Food Science, School of Public Health, Tianjin Medical University, Tianjin, China ,grid.265021.20000 0000 9792 1228Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Dezheng Zhou
- grid.265021.20000 0000 9792 1228Department of Nutrition & Food Science, School of Public Health, Tianjin Medical University, Tianjin, China ,grid.265021.20000 0000 9792 1228Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Jingzhu Fu
- grid.265021.20000 0000 9792 1228Department of Nutrition & Food Science, School of Public Health, Tianjin Medical University, Tianjin, China ,grid.265021.20000 0000 9792 1228Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Huilian Duan
- grid.265021.20000 0000 9792 1228Department of Nutrition & Food Science, School of Public Health, Tianjin Medical University, Tianjin, China ,grid.265021.20000 0000 9792 1228Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Zehao Wang
- grid.265021.20000 0000 9792 1228Department of Nutrition & Food Science, School of Public Health, Tianjin Medical University, Tianjin, China ,grid.265021.20000 0000 9792 1228Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Tong Yang
- grid.265021.20000 0000 9792 1228Department of Nutrition & Food Science, School of Public Health, Tianjin Medical University, Tianjin, China ,grid.265021.20000 0000 9792 1228Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Jing Zhao
- grid.265021.20000 0000 9792 1228Department of Nutrition & Food Science, School of Public Health, Tianjin Medical University, Tianjin, China ,grid.265021.20000 0000 9792 1228Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Wen Li
- grid.265021.20000 0000 9792 1228Department of Nutrition & Food Science, School of Public Health, Tianjin Medical University, Tianjin, China ,grid.265021.20000 0000 9792 1228Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Huan Liu
- grid.265021.20000 0000 9792 1228Department of Nutrition & Food Science, School of Public Health, Tianjin Medical University, Tianjin, China ,grid.265021.20000 0000 9792 1228Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Fei Ma
- grid.265021.20000 0000 9792 1228Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China ,grid.265021.20000 0000 9792 1228Department of Epidemiology & Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Changqing Sun
- grid.265021.20000 0000 9792 1228Neurosurgical Department of Baodi Clinical College, Tianjin Medical University, Tianjin, China
| | - Guangshun Wang
- grid.265021.20000 0000 9792 1228Department of Tumor, Baodi Clinical College of Tianjin Medical University, Tianjin, China
| | - Yue Du
- grid.265021.20000 0000 9792 1228Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China ,grid.265021.20000 0000 9792 1228Department of Social Medicine and Health Management, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Meilin Zhang
- grid.265021.20000 0000 9792 1228Department of Nutrition & Food Science, School of Public Health, Tianjin Medical University, Tianjin, China ,grid.265021.20000 0000 9792 1228Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Yongjie Chen
- grid.265021.20000 0000 9792 1228Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China ,grid.265021.20000 0000 9792 1228Department of Epidemiology & Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Guowei Huang
- grid.265021.20000 0000 9792 1228Department of Nutrition & Food Science, School of Public Health, Tianjin Medical University, Tianjin, China ,grid.265021.20000 0000 9792 1228Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| |
Collapse
|
121
|
Gorlova A, Svirin E, Pavlov D, Cespuglio R, Proshin A, Schroeter CA, Lesch KP, Strekalova T. Understanding the Role of Oxidative Stress, Neuroinflammation and Abnormal Myelination in Excessive Aggression Associated with Depression: Recent Input from Mechanistic Studies. Int J Mol Sci 2023; 24:915. [PMID: 36674429 PMCID: PMC9861430 DOI: 10.3390/ijms24020915] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/26/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
Aggression and deficient cognitive control problems are widespread in psychiatric disorders, including major depressive disorder (MDD). These abnormalities are known to contribute significantly to the accompanying functional impairment and the global burden of disease. Progress in the development of targeted treatments of excessive aggression and accompanying symptoms has been limited, and there exists a major unmet need to develop more efficacious treatments for depressed patients. Due to the complex nature and the clinical heterogeneity of MDD and the lack of precise knowledge regarding its pathophysiology, effective management is challenging. Nonetheless, the aetiology and pathophysiology of MDD has been the subject of extensive research and there is a vast body of the latest literature that points to new mechanisms for this disorder. Here, we overview the key mechanisms, which include neuroinflammation, oxidative stress, insulin receptor signalling and abnormal myelination. We discuss the hypotheses that have been proposed to unify these processes, as many of these pathways are integrated for the neurobiology of MDD. We also describe the current translational approaches in modelling depression, including the recent advances in stress models of MDD, and emerging novel therapies, including novel approaches to management of excessive aggression, such as anti-diabetic drugs, antioxidant treatment and herbal compositions.
Collapse
Affiliation(s)
- Anna Gorlova
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Laboratory of Cognitive Dysfunctions, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
| | - Evgeniy Svirin
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Laboratory of Cognitive Dysfunctions, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
- Neuroplast BV, 6222 NK Maastricht, The Netherlands
| | - Dmitrii Pavlov
- Hotchkiss Brain Institute, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Raymond Cespuglio
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Centre de Recherche en Neurosciences de Lyon (CRNL), 69500 Bron, France
| | - Andrey Proshin
- P.K. Anokhin Research Institute of Normal Physiology, 125315 Moscow, Russia
| | - Careen A. Schroeter
- Preventive and Environmental Medicine, Kastanienhof Clinic, 50858 Köln-Junkersdorf, Germany
| | - Klaus-Peter Lesch
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, The Netherlands
- Division of Molecular Psychiatry, Center of Mental Health, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Tatyana Strekalova
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, The Netherlands
- Division of Molecular Psychiatry, Center of Mental Health, University Hospital Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
122
|
Grant WB, Blake SM. Diet's Role in Modifying Risk of Alzheimer's Disease: History and Present Understanding. J Alzheimers Dis 2023; 96:1353-1382. [PMID: 37955087 PMCID: PMC10741367 DOI: 10.3233/jad-230418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2023] [Indexed: 11/14/2023]
Abstract
Diet is an important nonpharmacological risk-modifying factor for Alzheimer's disease (AD). The approaches used here to assess diet's role in the risk of AD include multi-country ecological studies, prospective and cross-sectional observational studies, and laboratory studies. Ecological studies have identified fat, meat, and obesity from high-energy diets as important risk factors for AD and reported that AD rates peak about 15-20 years after national dietary changes. Observational studies have compared the Western dietary pattern with those of the Dietary Approaches to Stop Hypertension (DASH), Mediterranean (MedDi), and Mediterranean-DASH Intervention for Neurodegenerative Delay (MIND) diets. Those studies identified AD risk factors including higher consumption of saturated and total fats, meat, and ultraprocessed foods and a lower risk of AD with higher consumption of fruits, legumes, nuts, omega-3 fatty acids, vegetables, and whole grains. Diet-induced factors associated with a significant risk of AD include inflammation, insulin resistance, oxidative stress, elevated homocysteine, dietary advanced glycation end products, and trimethylamine N-oxide. The molecular mechanisms by which dietary bioactive components and specific foods affect risk of AD are discussed. Given most countries' entrenched food supply systems, the upward trends of AD rates would be hard to reverse. However, for people willing and able, a low-animal product diet with plenty of anti-inflammatory, low-glycemic load foods may be helpful.
Collapse
Affiliation(s)
- William B. Grant
- Sunlight, Nutrition, and Health Research Center, San Francisco, CA, USA
| | - Steven M. Blake
- Nutritional Neuroscience, Maui Memory Clinic, Wailuku, HI, USA
| |
Collapse
|
123
|
Zhang J, Li X, Xiao J, Xiang Y, Ye F. Analysis of gene expression profiles in Alzheimer's disease patients with different lifespan: A bioinformatics study focusing on the disease heterogeneity. Front Aging Neurosci 2023; 15:1072184. [PMID: 36909942 PMCID: PMC9995587 DOI: 10.3389/fnagi.2023.1072184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/30/2023] [Indexed: 02/25/2023] Open
Abstract
Objective Alzheimer's disease (AD) as the most frequent neurodegenerative disease is featured by gradual decline of cognition and social function in the elderly. However, there have been few studies focusing on AD heterogeneity which exists both genetically and clinically, leading to the difficulties of AD researches. As one major kind of clinical heterogeneity, the lifespan of AD patients varies significantly. Aiming to investigate the potential driving factors, the current research identified the differentially expressed genes (DEGs) between longer-lived AD patients and shorter-lived ones via bioinformatics analyses. Methods Qualified datasets of gene expression profiles were identified in National Center of Biotechnology Information Gene Expression Omnibus (NCBI-GEO). The data of the temporal lobes of patients above 60 years old were used. Two groups were divided according to the lifespan: the group ≥85 years old and the group <85 years old. Then GEO2R online software and R package of Robust Rank Aggregation (RRA) were used to screen DEGs. Bioinformatic tools were adopted to identify possible pathways and construct protein-protein interaction network. Result Sixty-seven AD cases from four qualified datasets (GSE28146, GSE5281, GSE48350, and GSE36980) were included in this study. 740 DEGs were identified with 361 upregulated and 379 downregulated when compared longer-lived AD patients with shorter-lived ones. These DEGs were primarily involved in the pathways directly or indirectly associated with the regulation of neuroinflammation and cancer pathogenesis, as shown by pathway enrichment analysis. Among the DEGs, the top 15 hub genes were identified from the PPI network. Notably, the same bioinformatic procedures were conducted in 62 non-AD individuals (serving as controls of AD patients in the four included studies) with distinctly different findings from AD patients, indicating different regulatory mechanisms of lifespan between non-AD controls and AD, reconfirming the necessity of the present study. Conclusion These results shed some lights on lifespan-related regulatory mechanisms in AD patients, which also indicated that AD heterogeneity should be more taken into account in future investigations.
Collapse
Affiliation(s)
- Ji Zhang
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, China
| | - Xiaojia Li
- Department of Neurology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China.,Department of Neurology, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Jun Xiao
- Department of Neurology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China.,Department of Neurology, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Yang Xiang
- Department of Neurology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China.,Department of Neurology, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Fang Ye
- Department of Neurology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China.,Department of Neurology, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| |
Collapse
|
124
|
Ding T, Aimaiti M, Cui S, Shen J, Lu M, Wang L, Bian D. Meta-analysis of the association between dietary inflammatory index and cognitive health. Front Nutr 2023; 10:1104255. [PMID: 37081917 PMCID: PMC10111053 DOI: 10.3389/fnut.2023.1104255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/20/2023] [Indexed: 04/22/2023] Open
Abstract
Background Some studies have shown that a pro-inflammatory diet may be associated with cognitive function, but their conclusions have varied considerably. We here present a meta-analysis of the current published literature on DII score and its association with cognitive health. Methods In this meta-analysis, the PubMed, Embase, Web of Science, and Cochrane databases were searched in September 2022. The reported indexes, specifically OR, RR, and β, were extracted and analyzed using R version 3.1.0. Results A total of 636 studies in databases were identified, and 12 were included in the meta-analysis. Higher DII was associated with an increased risk of AD and MCI (OR = 1.34; 95% CI = 1.21-1.49). Meanwhile, it may also cause global function impairment (categorical: OR = 1.63; 95% CI = 1.36-1.96) and verbal fluency impairment (continuous: OR = 0.18; 95% IC = 0.08-0.42). But there was no significant association between DII and executive function (categorical: OR = 1.12; 95% IC = 0.84-1.49; continuous: OR = 0.48; 95% IC = 0.19-1.21) or episodic memory (continuous: OR = 0.56; 95% IC = 0.30-1.03). Conclusion A pro-inflammatory diet is related to AD, MCI, and the functions of some cognitive domains (specifically global function and verbal fluency). However, the current evidence on the role of diet-induced inflammation in different cognitive domains should be supported by further studies in the future.
Collapse
Affiliation(s)
- Tianze Ding
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Nutrition, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Maimaitiyusupu Aimaiti
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Nutrition, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shishuang Cui
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junhao Shen
- Department of Nutrition, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengjie Lu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Wang
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Dongsheng Bian, ; Lei Wang,
| | - Dongsheng Bian
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Clinical Nutrition, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Dongsheng Bian, ; Lei Wang,
| |
Collapse
|
125
|
Henn RE, Elzinga SE, Glass E, Parent R, Guo K, Allouch AM, Mendelson FE, Hayes J, Webber-Davis I, Murphy GG, Hur J, Feldman EL. Obesity-induced neuroinflammation and cognitive impairment in young adult versus middle-aged mice. Immun Ageing 2022; 19:67. [PMID: 36550567 PMCID: PMC9773607 DOI: 10.1186/s12979-022-00323-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Obesity rates are increasing worldwide. Obesity leads to many complications, including predisposing individuals to the development of cognitive impairment as they age. Immune dysregulation, including inflammaging (e.g., increased circulating cytokines) and immunosenescence (declining immune system function), commonly occur in obesity and aging and may impact cognitive impairment. As such, immune system changes across the lifespan may impact the effects of obesity on neuroinflammation and associated cognitive impairment. However, the role of age in obesity-induced neuroinflammation and cognitive impairment is unclear. To further define this putative relationship, the current study examined metabolic and inflammatory profiles, along with cognitive changes using a high-fat diet (HFD) mouse model of obesity. RESULTS First, HFD promoted age-related changes in hippocampal gene expression. Given this early HFD-induced aging phenotype, we fed HFD to young adult and middle-aged mice to determine the effect of age on inflammatory responses, metabolic profile, and cognitive function. As anticipated, HFD caused a dysmetabolic phenotype in both age groups. However, older age exacerbated HFD cognitive and neuroinflammatory changes, with a bi-directional regulation of hippocampal inflammatory gene expression. CONCLUSIONS Collectively, these data indicate that HFD promotes an early aging phenotype in the brain, which is suggestive of inflammaging and immunosenescence. Furthermore, age significantly compounded the impact of HFD on cognitive outcomes and on the regulation of neuroinflammatory programs in the brain.
Collapse
Affiliation(s)
- Rosemary E Henn
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sarah E Elzinga
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Emily Glass
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Molecular and Integrative Physiology, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Rachel Parent
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Molecular and Integrative Physiology, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kai Guo
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, 58202, USA
| | - Adam M Allouch
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Faye E Mendelson
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, 48109, USA
| | - John Hayes
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ian Webber-Davis
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Geoffery G Murphy
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Molecular and Integrative Physiology, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, 58202, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA.
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
126
|
Rostagno AA. Pathogenesis of Alzheimer's Disease. Int J Mol Sci 2022; 24:ijms24010107. [PMID: 36613544 PMCID: PMC9820480 DOI: 10.3390/ijms24010107] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia, accounting for 60% to 80% of all cases [...].
Collapse
Affiliation(s)
- Agueda A Rostagno
- Department of Pathology, Grossman School of Medicine, New York University, New York, NY 10016, USA
| |
Collapse
|
127
|
Pinchaud K, Hafeez Z, Auger S, Chatel JM, Chadi S, Langella P, Paoli J, Dary-Mourot A, Maguin-Gaté K, Olivier JL. Impact of Dietary Arachidonic Acid on Gut Microbiota Composition and Gut-Brain Axis in Male BALB/C Mice. Nutrients 2022; 14:nu14245338. [PMID: 36558497 PMCID: PMC9786182 DOI: 10.3390/nu14245338] [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: 11/24/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Although arachidonic acid (ARA) is the precursor of the majority of eicosanoids, its influence as a food component on health is not well known. Therefore, we investigated its impact on the gut microbiota and gut-brain axis. Groups of male BALB/c mice were fed either a standard diet containing 5% lipids (Std-ARA) or 15%-lipid diets without ARA (HL-ARA) or with 1% ARA (HL + ARA) for 9 weeks. Fatty acid profiles of all three diets were the same. The HL-ARA diet favored the growth of Bifidobacterium pseudolongum contrary to the HL + ARA diet that favored the pro-inflammatory Escherichia-Shigella genus in fecal microbiota. Dietary ARA intake induced 4- and 15-fold colic overexpression of the pro-inflammatory markers IL-1β and CD40, respectively, without affecting those of TNFα and adiponectin. In the brain, dietary ARA intake led to moderate overexpression of GFAP in the hippocampus and cortex. Both the hyperlipidic diets reduced IL-6 and IL-12 in the brain. For the first time, it was shown that dietary ARA altered the gut microbiota, led to low-grade colic inflammation, and induced astrogliosis in the brain. Further work is necessary to determine the involved mechanisms.
Collapse
Affiliation(s)
- Katleen Pinchaud
- Calbinotox (UR7488), Université de Lorraine, 54000 Nancy, France
| | - Zeeshan Hafeez
- Calbinotox (UR7488), Université de Lorraine, 54000 Nancy, France
| | - Sandrine Auger
- INRAE, Université Paris-Saclay, AgroParisTech, UMR 1319 Micalis Institute, 78352 Jouy-en-Josas, France
| | - Jean-Marc Chatel
- INRAE, Université Paris-Saclay, AgroParisTech, UMR 1319 Micalis Institute, 78352 Jouy-en-Josas, France
| | - Sead Chadi
- INRAE, Université Paris-Saclay, AgroParisTech, UMR 1319 Micalis Institute, 78352 Jouy-en-Josas, France
| | - Philippe Langella
- INRAE, Université Paris-Saclay, AgroParisTech, UMR 1319 Micalis Institute, 78352 Jouy-en-Josas, France
| | - Justine Paoli
- Calbinotox (UR7488), Université de Lorraine, 54000 Nancy, France
| | | | - Katy Maguin-Gaté
- Calbinotox (UR7488), Université de Lorraine, 54000 Nancy, France
| | - Jean Luc Olivier
- Calbinotox (UR7488), Université de Lorraine, 54000 Nancy, France
- CHRU de Nancy, Pôle des Laboratoires, Service de Biochimie-Biologie Moléculaire-Nutrition, 54000 Nancy, France
- Correspondence:
| |
Collapse
|
128
|
Hsiao HT, Ma MC, Chang HI, Lin CH, Hsu SW, Huang SH, Lee CC, Huang CW, Chang CC. Cognitive Decline Related to Diet Pattern and Nutritional Adequacy in Alzheimer's Disease Using Surface-Based Morphometry. Nutrients 2022; 14:nu14245300. [PMID: 36558459 PMCID: PMC9784891 DOI: 10.3390/nu14245300] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
Dietary pattern (DP) results in nutrition adequacy and may influence cognitive decline and cortical atrophy in Alzheimer's disease (AD). The study explored DP in 248 patients with AD. Two neurobehavioral assessments (intervals 13.4 months) and two cortical thickness measurements derived from magnetic resonance images (intervals 26.5 months) were collected as outcome measures. Reduced rank regression was used to assess the groups of DPs and a linear mixed-effect model to explore the cortical neurodegenerative patterns. At screening, underweight body mass index (BMI) was related to significant higher lipid profile, impaired cognitive function, smaller cortical thickness, lower protein DP factor loading scores and the non-spouse caregiver status. Higher mini-mental state examination (MMSE) scores were related to the DP of coffee/tea, compared to the lipid/sugar or protein DP group. The underweighted-BMI group had faster cortical thickness atrophy in the pregenual and lateral temporal cortex, while the correlations between cortical thickness degeneration and high HbA1C or low B12 and folate levels were localized in the medial and lateral prefrontal cortex. The predictive model suggested that factors related to MMSE score were related to the caregiver status. In conclusion, normal or overweight BMI, coffee/tea DP group and living with a spouse were considered as protective factors for better cognitive outcomes in patients with AD. The influence of glucose, B12 and folate on the cortical degeneration was spatially distinct from the pattern of AD degeneration.
Collapse
Affiliation(s)
- Hua-Tsen Hsiao
- Department of Nursing, National Tainan Junior College of Nursing, Tainan 700007, Taiwan
| | - Mi-Chia Ma
- Department of Statistics and Institute of Data Science, National Cheng Kung University, Tainan 701401, Taiwan
| | - Hsin-I Chang
- Department of Neurology, Cognition and Aging Center, Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan
| | - Ching-Heng Lin
- Center for Artificial Intelligence in Medicine, Chang Gung Memorial Hospital, Taoyuan 333008, Taiwan
- Bachelor Program in Artificial Intelligence, Chang Gung University, Taoyuan 333323, Taiwan
| | - Shih-Wei Hsu
- Department of Radiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan
| | - Shu-Hua Huang
- Department of Nuclear Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan
| | - Chen-Chang Lee
- Department of Radiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan
| | - Chi-Wei Huang
- Department of Neurology, Cognition and Aging Center, Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan
| | - Chiung-Chih Chang
- Department of Neurology, Cognition and Aging Center, Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan
- Correspondence:
| |
Collapse
|
129
|
Boehme M, Guzzetta KE, Wasén C, Cox LM. The gut microbiota is an emerging target for improving brain health during ageing. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2022; 4:E2. [PMID: 37179659 PMCID: PMC10174391 DOI: 10.1017/gmb.2022.11] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
The gut microbiota plays crucial roles in maintaining the health and homeostasis of its host throughout lifespan, including through its ability to impact brain function and regulate behaviour during ageing. Studies have shown that there are disparate rates of biologic ageing despite equivalencies in chronologic age, including in the development of neurodegenerative diseases, which suggests that environmental factors may play an important role in determining health outcomes in ageing. Recent evidence demonstrates that the gut microbiota may be a potential novel target to ameliorate symptoms of brain ageing and promote healthy cognition. This review highlights the current knowledge around the relationships between the gut microbiota and host brain ageing, including potential contributions to age-related neurodegenerative diseases. Furthermore, we assess key areas for which gut microbiota-based strategies may present as opportunities for intervention.
Collapse
Affiliation(s)
- Marcus Boehme
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Katherine Elizabeth Guzzetta
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Caroline Wasén
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Laura Michelle Cox
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, USA
| |
Collapse
|
130
|
Dai CL, Liu F, Iqbal K, Gong CX. Gut Microbiota and Immunotherapy for Alzheimer's Disease. Int J Mol Sci 2022; 23:15230. [PMID: 36499564 PMCID: PMC9741026 DOI: 10.3390/ijms232315230] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/08/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that eventually leads to dementia and death of the patient. Currently, no effective treatment is available that can slow or halt the progression of the disease. The gut microbiota can modulate the host immune system in the peripheral and central nervous system through the microbiota-gut-brain axis. Growing evidence indicates that gut microbiota dysbiosis plays an important role in the pathogenesis of AD, and modulation of the gut microbiota may represent a new avenue for treating AD. Immunotherapy targeting Aβ and tau has emerged as the most promising disease-modifying therapy for the treatment of AD. However, the underlying mechanism of AD immunotherapy is not known. Importantly, preclinical and clinical studies have highlighted that the gut microbiota exerts a major influence on the efficacy of cancer immunotherapy. However, the role of the gut microbiota in AD immunotherapy has not been explored. We found that immunotherapy targeting tau can modulate the gut microbiota in an AD mouse model. In this article, we focused on the crosstalk between the gut microbiota, immunity, and AD immunotherapy. We speculate that modulation of the gut microbiota induced by AD immunotherapy may partially underlie the efficacy of the treatment.
Collapse
Affiliation(s)
| | | | | | - Cheng-Xin Gong
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, NY 10314, USA
| |
Collapse
|
131
|
R Cardoso B, Machado P, Steele EM. Association between ultra-processed food consumption and cognitive performance in US older adults: a cross-sectional analysis of the NHANES 2011-2014. Eur J Nutr 2022; 61:3975-3985. [PMID: 35778619 PMCID: PMC9596521 DOI: 10.1007/s00394-022-02911-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 05/09/2022] [Indexed: 11/04/2022]
Abstract
PURPOSE This study evaluated the association between ultra-processed food (UPF) consumption and cognitive performance among older US adults. METHODS This cross-sectional study assessed 3632 participants aged 60+ years from the National Health and Nutrition Examination Survey (NHANES) 2011-14. Cognitive performance was assessed using the Consortium to Establish a Registry for Alzheimer's Disease (CERAD), Word Learning test, Animal Fluency test, and the Digit Symbol Substitution test (DSST). Dietary intake was assessed using two 24-h diet recalls. Food items were classified according to the NOVA system, a classification based on the nature, extent, and purpose of industrial food processing. Linear regression models were used to evaluate the association of dietary share of UPF (% of daily energy intake) (categorized as tertiles) and cognitive test scores, adjusting for socio-demographic variables, physical activity, smoking status, and chronic diseases (cardiovascular diseases, diabetes, and depression). Models excluding participants with pre-existing diseases were carried out to address potential reverse causality. RESULTS On average, UPF accounted for 53% of total energy intake, ranging from 33 to 70% across extreme tertiles. Inverted U-shape association between UPF consumption and Animal fluency and DSST was observed. No significant associations were observed between the UPF intake tertiles and the cognitive test results. Nonetheless, UPF consumption was significantly associated with worse performance in Animal Fluency in older adults without pre-existing diseases (P < 0.05). CONCLUSION UPF consumption was associated with worse performance in Animal Fluency among older people without pre-existing diseases. Decreasing UPF consumption may be a way to improve impaired cognition among older adults.
Collapse
Affiliation(s)
- Barbara R Cardoso
- Department of Nutrition, Dietetics and Food, Monash University, 264 Ferntree Gully Road, Notting Hill, VIC, 3168, Australia.
| | - Priscila Machado
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, VIC, Australia
| | - Euridice Martinez Steele
- Departamento de Nutrição, Faculdade de Saúde Pública, Universidade de São Paulo, Av. Dr. Arnaldo, 715, São Paulo, 01246-907, Brazil.
- Center for Epidemiological Studies in Health and Nutrition, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
132
|
Wang LY, Liu J, Peng YZ, Zhang CP, Zou W, Liu F, Zhan KB, Zhang P. Curcumin-Nicotinate Attenuates Hippocampal Synaptogenesis Dysfunction in Hyperlipidemia Rats by the BDNF/TrkB/CREB Pathway: Involving Idol/LDLR Signaling to Eliminate Aβ Deposition. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221141162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Hyperlipidemia has been demonstrated to evoke Alzheimer disease (AD) pathologies such as Amyloid-β (Aβ) deposition and synaptogenesis dysfunction in the hippocampus. Curcumin gives protection against anti-amyloid properties and synaptogenesis dysfunction. Curcumin-Nicotinate (CurTn), a new type of curcumin derivative, ameliorates cognitive impairment by rescuing autophagic flux in the CA1 hippocampus of diabetic rats. However, whether Curtn possesses an antagonistic effect on AD-related pathologies in the hippocampus induced by hyperlipidemia remains ill-defined. The present study aims to investigate whether CurTn alleviates synaptogenesis dysfunction by promoting the activation of brain-derived neurotrophic factor (BDNF)/tyrosine kinase receptor B (TrkB)/cAMP-response element binding protein (CREB) signaling and whether the underlying fundamental mechanism involves the elimination of Aβ deposition due to Idol/low-density lipoprotein receptor (LDLR) signaling in the hippocampus of high-fat diet (HFD)-induced hyperlipidemia rats. The results demonstrated that CurTn not only improved synaptogenesis dysfunction in the hippocampus of HFD rats, as evidenced by the increases in the expressions of synapse-related proteins postsynaptic density protein 95 (PSD-95), synapsin-1, and Glutamate receptor 1 (GluR1), but also activated BDNF/TrkB/CREB signaling, as evidenced by the elevation of the expressions of BDNF, pTrkB, and CREB. Moreover, CurTn modulated the Idol/LDLR pathway in the hippocampus of HFD rats, as evidenced by the decreased expression of Idol and the increased expression of LDLR. Furthermore, CurTn eliminated the deposition of Aβ, as evidenced by the reduction in the content of Aβ40 and Aβ42. These results reveal that CurTn may attenuate synaptogenesis dysfunction by activating BDNF/TrkB/CREB signaling, as the possible result of the modulation of Idol/LDLR signaling to eliminate Aβ deposition in the hippocampus of HFD rats.
Collapse
Affiliation(s)
- Lin-Yu Wang
- Department of Neurology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Jiao Liu
- Department of Neurology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Yi-Zhu Peng
- Department of Neurology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Cai-Ping Zhang
- Department of Biochemistry, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Wei Zou
- Department of Neurology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Feng Liu
- Department of Neurology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Ke-Bin Zhan
- Department of Neurology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Ping Zhang
- Department of Neurology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| |
Collapse
|
133
|
Neuffer J, González-Domínguez R, Lefèvre-Arbogast S, Low DY, Driollet B, Helmer C, Du Preez A, de Lucia C, Ruigrok SR, Altendorfer B, Aigner L, Lucassen PJ, Korosi A, Thuret S, Manach C, Pallàs M, Urpi-Sardà M, Sánchez-Pla A, Andres-Lacueva C, Samieri C. Exploration of the Gut-Brain Axis through Metabolomics Identifies Serum Propionic Acid Associated with Higher Cognitive Decline in Older Persons. Nutrients 2022; 14:4688. [PMID: 36364950 PMCID: PMC9655149 DOI: 10.3390/nu14214688] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 10/28/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
The gut microbiome is involved in nutrient metabolism and produces metabolites that, via the gut−brain axis, signal to the brain and influence cognition. Human studies have so far had limited success in identifying early metabolic alterations linked to cognitive aging, likely due to limitations in metabolite coverage or follow-ups. Older persons from the Three-City population-based cohort who had not been diagnosed with dementia at the time of blood sampling were included, and repeated measures of cognition over 12 subsequent years were collected. Using a targeted metabolomics platform, we identified 72 circulating gut-derived metabolites in a case−control study on cognitive decline, nested within the cohort (discovery n = 418; validation n = 420). Higher serum levels of propionic acid, a short-chain fatty acid, were associated with increased odds of cognitive decline (OR for 1 SD = 1.40 (95% CI 1.11, 1.75) for discovery and 1.26 (1.02, 1.55) for validation). Additional analyses suggested mediation by hypercholesterolemia and diabetes. Propionic acid strongly correlated with blood glucose (r = 0.79) and with intakes of meat and cheese (r > 0.15), but not fiber (r = 0.04), suggesting a minor role of prebiotic foods per se, but a possible link to processed foods, in which propionic acid is a common preservative. The adverse impact of propionic acid on metabolism and cognition deserves further investigation.
Collapse
Affiliation(s)
- Jeanne Neuffer
- Bordeaux Population Health Research Center, University of Bordeaux, INSERMUMR 1219, F-33000 Bordeaux, France
| | - Raúl González-Domínguez
- Nutrition, Food Science and Gastronomy Department, Food Innovation Network (XIA), Institute of Nutrition and Food Safety (INSA-UB), Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sophie Lefèvre-Arbogast
- Bordeaux Population Health Research Center, University of Bordeaux, INSERMUMR 1219, F-33000 Bordeaux, France
| | - Dorrain Y. Low
- Human Nutrition Unit, Université Clermont Auvergne, INRAEUMR1019, F-63000 Clermont Ferrand, France
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore
| | - Bénédicte Driollet
- Bordeaux Population Health Research Center, University of Bordeaux, INSERMUMR 1219, F-33000 Bordeaux, France
| | - Catherine Helmer
- Bordeaux Population Health Research Center, University of Bordeaux, INSERMUMR 1219, F-33000 Bordeaux, France
| | - Andrea Du Preez
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
| | - Chiara de Lucia
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
| | - Silvie R. Ruigrok
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
| | - Barbara Altendorfer
- Institute of Molecular Regenerative Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Paul J. Lucassen
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
- The Center for Urban Mental Health, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
| | - Aniko Korosi
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Claudine Manach
- Human Nutrition Unit, Université Clermont Auvergne, INRAEUMR1019, F-63000 Clermont Ferrand, France
| | - Mercè Pallàs
- Pharmacology Section, Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Neurociencies, University of Barcelona, 08028 Barcelona, Spain
| | - Mireia Urpi-Sardà
- Nutrition, Food Science and Gastronomy Department, Food Innovation Network (XIA), Institute of Nutrition and Food Safety (INSA-UB), Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alex Sánchez-Pla
- Nutrition, Food Science and Gastronomy Department, Food Innovation Network (XIA), Institute of Nutrition and Food Safety (INSA-UB), Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Cristina Andres-Lacueva
- Nutrition, Food Science and Gastronomy Department, Food Innovation Network (XIA), Institute of Nutrition and Food Safety (INSA-UB), Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Cécilia Samieri
- Bordeaux Population Health Research Center, University of Bordeaux, INSERMUMR 1219, F-33000 Bordeaux, France
| |
Collapse
|
134
|
Li T, Chu C, Yu L, Zhai Q, Wang S, Zhao J, Zhang H, Chen W, Tian F. Neuroprotective Effects of Bifidobacterium breve CCFM1067 in MPTP-Induced Mouse Models of Parkinson's Disease. Nutrients 2022; 14:4678. [PMID: 36364939 PMCID: PMC9655354 DOI: 10.3390/nu14214678] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/22/2022] [Accepted: 10/27/2022] [Indexed: 08/31/2023] Open
Abstract
There is mounting evidence that the microbiota-gut-brain axis (MGBA) is critical in the pathogenesis and progression of Parkinson's disease (PD), suggesting that probiotic therapy restoring gut microecology may slow down disease progression. In this study, we examined the disease-alleviating effects of Bifidobacterium breve CCFM1067, orally administered for 5 weeks in a PD mouse model. Our study shows that supplementation with the probiotic B. breve CCFM1067 protected dopaminergic neurons and suppressed glial cell hyperactivation and neuroinflammation in PD mice. In addition, the antioxidant capacity of the central nervous system was enhanced and oxidative stress was alleviated. Moreover, B. breve CCFM1067 protected the blood-brain and intestinal barriers from damage in the MPTP-induced mouse model. The results of fecal microbiota analysis showed that B. breve CCFM1067 intervention could act on the MPTP-induced microecological imbalance in the intestinal microbiota, suppressing the number of pathogenic bacteria (Escherichia-Shigella) while increasing the number of beneficial bacteria (Bifidobacterium and Akkermansia) in PD mice. In addition, the increase in short chain fatty acids (acetic and butyric acids) may explain the anti-inflammatory action of B. breve CCFM1067 in the gut or brain of the MPTP-induced PD mouse model. In conclusion, we demonstrated that the probiotic B. breve CCFM1067, which can prevent or treat PD by modulating the gut-brain axis, can be utilized as a possible new oral supplement for PD therapy.
Collapse
Affiliation(s)
- Tiantian Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Chuanqi Chu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Shunhe Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
- Department of Child Health Care, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
135
|
Coppedè F, Franzago M, Giardina E, Nigro CL, Matullo G, Moltrasio C, Nacmias B, Pileggi S, Sirchia SM, Stoccoro A, Storlazzi CT, Stuppia L, Tricarico R, Merla G. A perspective on diet, epigenetics and complex diseases: where is the field headed next? Epigenomics 2022; 14:1281-1304. [DOI: 10.2217/epi-2022-0239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Dietary factors can regulate epigenetic processes during life, modulating the intracellular pools of metabolites necessary for epigenetic reactions and regulating the activity of epigenetic enzymes. Their effects are strong during the prenatal life, when epigenetic patterns are written, allowing organogenesis. However, interactions between diet and the epigenome continue throughout life and likely contribute to the onset and progression of various complex diseases. Here, we review the contribution of dietary factors to the epigenetic changes observed in complex diseases and suggest future steps to better address this issue, focusing on neurobehavioral, neuropsychiatric and neurodegenerative disorders, cardiovascular diseases, obesity and Type 2 diabetes, cancer and inflammatory skin diseases.
Collapse
Affiliation(s)
- Fabio Coppedè
- Department of Translational Research & of New Surgical & Medical Technologies, University of Pisa, Pisa, 56126, Italy
| | - Marica Franzago
- Department of Medicine & Aging, School of Medicine & Health Sciences, “G. d'Annunzio” University of Chieti–Pescara, Chieti, 66100, Italy
- Center for Advanced Studies & Technology, “G. d'Annunzio” University of Chieti–Pescara, Chieti, 66100, Italy
| | - Emiliano Giardina
- Genomic Medicine Laboratory UILDM, IRCCS Fondazione Santa Lucia, Rome, 00179, Italy
- Department of Biomedicine & Prevention, Tor Vergata University of Rome, Rome, 00133, Italy
| | | | - Giuseppe Matullo
- Department of Medical Sciences, University of Turin, Turin, 10126, Italy
| | - Chiara Moltrasio
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, 20122, Italy
- Department of Medical Surgical & Health Sciences, University of Trieste, Trieste, 34137, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research & Child Health, University of Florence, Florence, 50139, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, 50143, Italy
| | - Silvana Pileggi
- Department of Health Sciences, Medical Genetics, University of Milan, Milan, 20142, Italy
| | - Silvia Maria Sirchia
- Department of Health Sciences, Medical Genetics, University of Milan, Milan, 20142, Italy
| | - Andrea Stoccoro
- Department of Translational Research & of New Surgical & Medical Technologies, University of Pisa, Pisa, 56126, Italy
| | | | - Liborio Stuppia
- Center for Advanced Studies & Technology, “G. d'Annunzio” University of Chieti–Pescara, Chieti, 66100, Italy
- Department of Psychological, Health & Territorial Sciences, School of Medicine & Health Sciences, “G. d'Annunzio” University of Chieti–Pescara, Chieti, 66100, Italy
| | - Rossella Tricarico
- Department of Biology & Biotechnology, University of Pavia, Pavia, 27100, Italy
| | - Giuseppe Merla
- Laboratory of Regulatory & Functional Genomics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, 71013, Italy
- Department of Molecular Medicine & Medical Biotechnology, University of Naples Federico II, Naples, 80131, Italy
| |
Collapse
|
136
|
Ren ZL, Li CX, Ma CY, Chen D, Chen JH, Xu WX, Chen CA, Cheng FF, Wang XQ. Linking Nonalcoholic Fatty Liver Disease and Brain Disease: Focusing on Bile Acid Signaling. Int J Mol Sci 2022; 23:13045. [PMID: 36361829 PMCID: PMC9654021 DOI: 10.3390/ijms232113045] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 11/01/2023] Open
Abstract
A metabolic illness known as non-alcoholic fatty liver disease (NAFLD), affects more than one-quarter of the world's population. Bile acids (BAs), as detergents involved in lipid digestion, show an abnormal metabolism in patients with NAFLD. However, BAs can affect other organs as well, such as the brain, where it has a neuroprotective effect. According to a series of studies, brain disorders may be extrahepatic manifestations of NAFLD, such as depression, changes to the cerebrovascular system, and worsening cognitive ability. Consequently, we propose that NAFLD affects the development of brain disease, through the bile acid signaling pathway. Through direct or indirect channels, BAs can send messages to the brain. Some BAs may operate directly on the central Farnesoid X receptor (FXR) and the G protein bile acid-activated receptor 1 (GPBAR1) by overcoming the blood-brain barrier (BBB). Furthermore, glucagon-like peptide-1 (GLP-1) and the fibroblast growth factor (FGF) 19 are released from the intestine FXR and GPBAR1 receptors, upon activation, both of which send signals to the brain. Inflammatory, systemic metabolic disorders in the liver and brain are regulated by the bile acid-activated receptors FXR and GPBAR1, which are potential therapeutic targets. From a bile acid viewpoint, we examine the bile acid signaling changes in NAFLD and brain disease. We also recommend the development of dual GPBAR1/FXR ligands to reduce side effects and manage NAFLD and brain disease efficiently.
Collapse
Affiliation(s)
- Zi-Lin Ren
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Chang-Xiang Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Chong-Yang Ma
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Dan Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jia-Hui Chen
- Dongzhimen Hospital, Beijing University of Traditional Chinese Medicine, Beijing 100700, China
| | - Wen-Xiu Xu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Cong-Ai Chen
- Dongzhimen Hospital, Beijing University of Traditional Chinese Medicine, Beijing 100700, China
| | - Fa-Feng Cheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xue-Qian Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
137
|
Amelioration of Age-Related Multiple Neuronal Impairments and Inflammation in High-Fat Diet-Fed Rats: The Prospective Multitargets of Geraniol. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4812993. [PMID: 36304965 PMCID: PMC9596245 DOI: 10.1155/2022/4812993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 08/31/2022] [Accepted: 09/27/2022] [Indexed: 11/18/2022]
Abstract
Neuroinflammation is documented to alter brain function as a consequence of metabolic changes linked with a high-fat diet (HFD). The primary target of this study is to see how geraniol is effective in manipulating age- and diet-associated multiple toxicity and neuroinflammation in HFD-fed rats. Sixty-four adult male Wistar rats were partitioned into two groups: Group 1 (untreated normal young and aged rats) and Group 2 (HFD-fed young and aged rats) that received HFD for 16 weeks before being orally treated with geraniol or chromax for eight weeks. The results revealed a dropping in proinflammatory cytokines (TNF-α and IL-6) and leptin while boosting adiponectin in geraniol-supplemented rats. The liver, kidney, and lipid profiles were improved in geraniol-HFD-treated groups. HFD-induced brain insulin resistance decreased insulin clearance and insulin-degrading enzyme (IDE) levels significantly after geraniol supplementation. Geraniol suppressed acetylcholinesterase (AChE) activity and alleviated oxidative stress by boosting neuronal reduced glutathione (GSH), catalase (CAT), glutathione-S-transferase (GST), and superoxide dismutase (SOD) activities. It lowered malondialdehyde concentration (TBARS), nitric oxide (NO), and xanthine oxidase (XO) and restored the structural damage to the brain tissue caused by HFD. Compared with model rats, geraniol boosted learning and memory function and ameliorated the inflammation status in the brain by lowering the protein levels of IL-1β, iNOS, NF-κBp65, and COX-2. In addition, the expression levels of inflammation-related genes (MCP-1, TNF-α, IL-6, IL-1β, and IDO-1) were lessened significantly. Remarkably, the supplementation of geraniol reversed the oxidative and inflammation changes associated with aging. It affected the redox status of young rats. In conclusion, our results exhibit the effectiveness of dietary geraniol supplementation in modifying age-related neuroinflammation and oxidative stress in rats and triggering off the use of geraniol as a noninvasive natural compound for controlling age- and diet-associated neuronal impairments and toxicity.
Collapse
|
138
|
Migliore L, Coppedè F. Gene-environment interactions in Alzheimer disease: the emerging role of epigenetics. Nat Rev Neurol 2022; 18:643-660. [PMID: 36180553 DOI: 10.1038/s41582-022-00714-w] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2022] [Indexed: 12/15/2022]
Abstract
With the exception of a few monogenic forms, Alzheimer disease (AD) has a complex aetiology that is likely to involve multiple susceptibility genes and environmental factors. The role of environmental factors is difficult to determine and, until a few years ago, the molecular mechanisms underlying gene-environment (G × E) interactions in AD were largely unknown. Here, we review evidence that has emerged over the past two decades to explain how environmental factors, such as diet, lifestyle, alcohol, smoking and pollutants, might interact with the human genome. In particular, we discuss how various environmental AD risk factors can induce epigenetic modifications of key AD-related genes and pathways and consider how epigenetic mechanisms could contribute to the effects of oxidative stress on AD onset. Studies on early-life exposures are helping to uncover critical time windows of sensitivity to epigenetic influences from environmental factors, thereby laying the foundations for future primary preventative approaches. We conclude that epigenetic modifications need to be considered when assessing G × E interactions in AD.
Collapse
Affiliation(s)
- Lucia Migliore
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy. .,Department of Laboratory Medicine, Pisa University Hospital, Pisa, Italy.
| | - Fabio Coppedè
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| |
Collapse
|
139
|
Chen L, Wang B, Liu J, Wu X, Xu X, Cao H, Ji X, Zhang P, Li X, Hou Z, Li H. Different oral and gut microbial profiles in those with Alzheimer's disease consuming anti-inflammatory diets. Front Nutr 2022; 9:974694. [PMID: 36185672 PMCID: PMC9521405 DOI: 10.3389/fnut.2022.974694] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/15/2022] [Indexed: 12/06/2022] Open
Abstract
The number of people living with Alzheimer's disease (AD) is increasing alongside with aging of the population. Systemic chronic inflammation and microbial imbalance may play an important role in the pathogenesis of AD. Inflammatory diets regulate both the host microbiomes and inflammatory status. This study aimed to explore the impact of inflammatory diets on oral-gut microbes in patients with AD and the relationship between microbes and markers of systemic inflammation. The dietary inflammatory properties and the oral and gut microorganisms were analyzed using the dietary inflammatory index (DII) and 16S RNA in 60 patients with AD. The α-diversity was not related to the DII (p > 0.05), whereas the β-diversity was different in the oral microbiomes (R2 = 0.061, p = 0.013). In the most anti-inflammatory diet group, Prevotella and Olsenella were more abundant in oral microbiomes and Alistipes, Ruminococcus, Odoribacter, and unclassified Firmicutes were in the gut microbiomes (p < 0.05). Specific oral and gut genera were associated with interleukin-6 (IL)-6, complement 3 (C3), high-sensitivity C-reactive protein (hs-CRP), IL-1β, IL-4, IL-10, IL-12, and tumor necrosis factor-α (TNF-α) (p < 0.05). In conclusion, anti-inflammatory diets seem to be associated with increased abundance of beneficial microbes, and specific oral and gut microbial composition was associated with inflammatory markers.
Collapse
Affiliation(s)
- Lili Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- The School of Nursing, Fujian Medical University, Fuzhou, China
- Fujian Provincial Hospital, Fuzhou, China
- Lili Chen
| | - Bixia Wang
- The School of Nursing, Fujian Medical University, Fuzhou, China
| | - Jinxiu Liu
- The School of Nursing, Fujian Medical University, Fuzhou, China
| | - Xiaoqi Wu
- Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xinhua Xu
- The School of Nursing, Fujian Medical University, Fuzhou, China
| | - Huizhen Cao
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Fujian Provincial Hospital, Fuzhou, China
| | - Xinli Ji
- The School of Nursing, Fujian Medical University, Fuzhou, China
| | - Ping Zhang
- The School of Nursing, Fujian Medical University, Fuzhou, China
| | - Xiuli Li
- The School of Nursing, Fujian Medical University, Fuzhou, China
| | - Zhaoyi Hou
- The School of Nursing, Fujian Medical University, Fuzhou, China
| | - Hong Li
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- The School of Nursing, Fujian Medical University, Fuzhou, China
- Fujian Provincial Hospital, Fuzhou, China
- *Correspondence: Hong Li
| |
Collapse
|
140
|
Sanderlin AH, Hayden KM, Baker LD, Craft S. Ketogenic dietary lifestyle intervention effects on sleep, cognition, and behavior in mild cognitive impairment: Study design. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12343. [PMID: 36177445 PMCID: PMC9473641 DOI: 10.1002/trc2.12343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/30/2022] [Accepted: 07/02/2022] [Indexed: 12/02/2022]
Abstract
Introduction Sleep and diet are modifiable risk factors for Alzheimer's disease (AD) that may be salient areas for the development of preventive intervention strategies against dementia in people with mild cognitive impairment (MCI). Sleep disturbances account for up to 15% of the population attributable risk for AD. Diet influences sleep quality, such that diets high in sugars, fat, and processed food affect sleep quality and cognition in older adults. The combination of poor sleep and diet health may increase risk for dementia in people with MCI, yet it is unknown how intervening on diet may influence sleep health. Methods The MCI Sleep Study assesses longitudinal changes in objective and subjective measures of sleep between two investigational diet groups in the Brain Energy for Amyloid Transformation in Alzheimer's Disease study: the modified Mediterranean ketogenic diet (MMKD) and the American Heart Association diet. Objective sleep assessments include an in-home sleep study using the WatchPAT Central Plus (Itamar Medical, Ltd) at baseline and the end of the 4-month diet intervention. Subjective sleep questionnaires include the Epworth Sleepiness Scale and Pittsburgh Sleep Quality Index. The MCI Sleep Study outcome measures include longitudinal change in cognitive performance, mood, behavior, and quality of life. Results Study recruitment is currently ongoing. We hypothesize the low-carb MMKD diet to have a beneficial impact on sleep health in individuals with MCI. Pre- and post-diet changes in sleep metrics across diet groups will be examined using mixed effects analysis of variance models. Discussion Early assessment of chronic sleep and diet behaviors may be vital in understanding when interventions are necessary and the lifestyle modifications that should accompany future AD prevention and therapy recommendations.
Collapse
Affiliation(s)
- Ashley H. Sanderlin
- Department of Internal Medicine—Gerontology and Geriatric MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Kathleen M. Hayden
- Department of Social Sciences and Health PolicyWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Laura D. Baker
- Department of Internal Medicine—Gerontology and Geriatric MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
- Department of Social Sciences and Health PolicyWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Suzanne Craft
- Department of Internal Medicine—Gerontology and Geriatric MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
141
|
Al-Ghraiybah NF, Wang J, Alkhalifa AE, Roberts AB, Raj R, Yang E, Kaddoumi A. Glial Cell-Mediated Neuroinflammation in Alzheimer's Disease. Int J Mol Sci 2022; 23:10572. [PMID: 36142483 PMCID: PMC9502483 DOI: 10.3390/ijms231810572] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/04/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder; it is the most common cause of dementia and has no treatment. It is characterized by two pathological hallmarks, the extracellular deposits of amyloid beta (Aβ) and the intraneuronal deposits of Neurofibrillary tangles (NFTs). Yet, those two hallmarks do not explain the full pathology seen with AD, suggesting the involvement of other mechanisms. Neuroinflammation could offer another explanation for the progression of the disease. This review provides an overview of recent advances on the role of the immune cells' microglia and astrocytes in neuroinflammation. In AD, microglia and astrocytes become reactive by several mechanisms leading to the release of proinflammatory cytokines that cause further neuronal damage. We then provide updates on neuroinflammation diagnostic markers and investigational therapeutics currently in clinical trials to target neuroinflammation.
Collapse
Affiliation(s)
- Nour F. Al-Ghraiybah
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Junwei Wang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Amer E. Alkhalifa
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Andrew B. Roberts
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Ruchika Raj
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Euitaek Yang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Amal Kaddoumi
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| |
Collapse
|
142
|
Cheng X, Wei Y, Qian Z, Han L. Autophagy Balances Neuroinflammation in Alzheimer's Disease. Cell Mol Neurobiol 2022; 43:1537-1549. [PMID: 35960407 DOI: 10.1007/s10571-022-01269-6] [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: 05/21/2022] [Accepted: 07/29/2022] [Indexed: 01/20/2023]
Abstract
Autophagy is a highly evolutionary conserved process that degrades cytosolic macromolecules or damaged organelles (e.g., mitochondria), as well as intracellular pathogens for energy and survival. Dysfunction of autophagy has been associated with the pathologies of Alzheimer's disease (AD), including Aβ plaques and neurofibrillary tangles. Recently, the presence of sustained immune response in the brain has been considered a new core pathology in AD. Accumulating evidence suggests that autophagy activation may suppress inflammation response through degrading inflammasomes or pro-inflammatory cytokines and improving immune system function in both clinical trials and preclinical studies. This review provides an overview of updated information on autophagy and inflammation and their potential mediators in AD. In summary, we believe that understanding the relationship between autophagy and inflammation will provide insightful knowledge for future therapeutic implications in AD.
Collapse
Affiliation(s)
- Xuehua Cheng
- Department of TCM Geriatrics, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, People's Republic of China
| | - Yong Wei
- GeneScience Pharmaceuticals CoLtd., Changchun, 130012, People's Republic of China
| | - Zijun Qian
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai, 200071, People's Republic of China
| | - Li Han
- Department of TCM Geriatrics, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
143
|
Patrakeeva VP, Shtaborov VA. Nutrition and the state of the intestinal microflora in the formation of the metabolic syndrome. OBESITY AND METABOLISM 2022. [DOI: 10.14341/omet12893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The literature review presents the results of modern studies of the relationship between diet and intestinal microbiota in the regulation of metabolic disorders. Metabolic syndrome, which is a symptom complex that combines abdominal obesity, insulin resistance, hyperglycemia, dyslipidemia and arterial hypertension, remains an important problem, being a risk factor for cardiovascular, neurodegenerative, oncological diseases and the development of type 2 diabetes mellitus. Although the pathogenesis of the metabolic syndrome has not yet been fully elucidated, it is known that visceral obesity and its associated complications, such as dyslipidemia and increased levels of pro-inflammatory cytokines, play a central role. The article presents data on the impact of the consumption of certain food products, the inclusion of plant biologically active substances (flavonoids, polyphenols, etc.) in the diet, as well as the use of elimination diets with the exclusion of carbohydrates or fats from the diet, on reducing the risk of cardiovascular accidents, levels of fasting glucose, total cholesterol, LDL, triglycerides, C-reactive protein, leptin, insulin, reduction in body weight and waist circumference, reduction in the level of circulating endotoxins and changes in the activity of immunocompetent cells. Data are presented on the possible influence of the intestinal microbiota in maintaining inflammation and the formation of degenerative changes in the body. The role of changes in the ratio of the levels of pathogenic microflora, bifidobacteria and lactobacilli in the formation of a pathological condition is shown.
Collapse
Affiliation(s)
- V. P. Patrakeeva
- N. Laverov Federal Center for Integrated Arctic Research of the Ural Branch of the Russian Academy of Sciences
| | - V. A. Shtaborov
- N. Laverov Federal Center for Integrated Arctic Research of the Ural Branch of the Russian Academy of Sciences
| |
Collapse
|
144
|
Davidson TL, Stevenson RJ. Appetitive interoception, the hippocampus and western-style diet. Rev Endocr Metab Disord 2022; 23:845-859. [PMID: 35067848 DOI: 10.1007/s11154-021-09698-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/23/2021] [Indexed: 02/06/2023]
Abstract
Obesity, Type 2 diabetes and other metabolic disorders continue to pose serious challenges to human health and well-being. An important source of these challenges is the overconsumption of saturated fats and sugar, main staples of what has been called the Western-style diet (WD). The current paper describes a theoretical model and supporting evidence that links intake of a WD to interference with a specific brain substrate that underlies processing of interoceptive signals of hunger and satiety. We review findings from rats and humans that the capacity of these signals to modulate the strength of appetitive and eating behavior depends on the functional integrity of the hippocampus and the learning memory operations it performs. Important among these operations is the use of contextual information to retrieve memories that are associated with other events. Within our framework, satiety provides an interoceptive context that informs animals that food cues and appetitive behavior will not be followed by rewarding postingestive outcomes. This serves to prevent those cues and responses from retrieving those reward memories. The findings reviewed provide evidence that consuming a WD and the high amounts of saturated fat and sugar it contains (a) is associated with the emergence of pathophysiologies to which the hippocampus appears selectively vulnerable (b) impairs hippocampal-dependent learning and memory (HDLM) and (c) weakens behavioral control by interoceptive hunger and satiety contextual stimuli. It is hypothesized that these consequences of WD intake may establish the conditions for a vicious cycle of further WD intake, obesity, and potentially cognitive decline.
Collapse
Affiliation(s)
- Terry L Davidson
- Department of Neuroscience and the Center for Neuroscience and Behavior, American University, Washington, DC, USA.
| | | |
Collapse
|
145
|
Functional Medicine Approaches to Neurodegeneration. Phys Med Rehabil Clin N Am 2022; 33:733-743. [PMID: 35989061 DOI: 10.1016/j.pmr.2022.04.011] [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: 11/24/2022]
Abstract
Neurodegenerative diseases impact more than 6 million Americans, and current predictions estimate the rates of neurodegenerative diseases will double in the next 30 years. These diseases are progressive with increasing loss of brain function throughout their course. Overtime, those suffering from neurodegenerative diseases will lose their ability to work and function efficiently in society. Families and society are burdened with skyrocketing costs to provide care for those who are unable to perform activities of daily living. There is an urgent need to develop treatment strategies to both reduce the incidence of neurodegenerative diseases and to delay the progression of the disease.
Collapse
|
146
|
Zhang Q, Jin K, Chen B, Liu R, Cheng S, Zhang Y, Lu J. Overnutrition Induced Cognitive Impairment: Insulin Resistance, Gut-Brain Axis, and Neuroinflammation. Front Neurosci 2022; 16:884579. [PMID: 35873818 PMCID: PMC9298971 DOI: 10.3389/fnins.2022.884579] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/02/2022] [Indexed: 12/11/2022] Open
Abstract
Overnutrition-related obesity has become a worldwide epidemic, and its prevalence is expected to steadily rise in the future. It is widely recognized that obesity exerts negative impacts on metabolic disorders such as type 2 diabetes mellitus (T2DM) and cardiovascular diseases. However, relatively fewer reports exist on the impairment of brain structure and function, in the form of memory and executive dysfunction, as well as neurogenerative diseases. Emerging evidence indicates that besides obesity, overnutrition diets independently induce cognitive impairments via multiple mechanisms. In this study, we reviewed the clinical and preclinical literature about the detrimental effects of obesity or high-nutrition diets on cognitive performance and cerebral structure. We mainly focused on the role of brain insulin resistance (IR), microbiota-gut-brain axis, and neuroinflammation. We concluded that before the onset of obesity, short-term exposure to high-nutrition diets already blunted central responses to insulin, altered gut microbiome composition, and activated inflammatory mediators. Overnutrition is linked with the changes in protein expression in brain insulin signaling, leading to pathological features in the brain. Microbiome alteration, bacterial endotoxin release, and gut barrier hyperpermeability also occur to trigger mental and neuronal diseases. In addition, obesity or high-nutrition diets cause chronic and low-grade systematic inflammation, which eventually spreads from the peripheral tissue to the central nervous system (CNS). Altogether, a large number of unknown but potential routes interact and contribute to obesity or diet-induced cognitive impairment. The challenge for future research is to identify effective interventions involving dietary shifts and personalized therapy targeting the underlying mechanisms to prevent and improve cognition deficits.
Collapse
Affiliation(s)
- Qin Zhang
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Kangyu Jin
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing Chen
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ripeng Liu
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China.,Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shangping Cheng
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuyan Zhang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jing Lu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, China
| |
Collapse
|
147
|
Habashy KJ, Ahmad F, Ibeh S, Mantash S, Kobeissy F, Issa H, Habis R, Tfaily A, Nabha S, Harati H, Reslan MA, Yehya Y, Barsa C, Shaito A, Zibara K, El-Yazbi AF, Kobeissy FH. Western and ketogenic diets in neurological disorders: can you tell the difference? Nutr Rev 2022; 80:1927-1941. [PMID: 35172003 DOI: 10.1093/nutrit/nuac008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
The prevalence of obesity tripled worldwide between 1975 and 2016, and it is projected that half of the US population will be overweight by 2030. The obesity pandemic is attributed, in part, to the increasing consumption of the high-fat, high-carbohydrate Western diet, which predisposes to the development of the metabolic syndrome and correlates with decreased cognitive performance. In contrast, the high-fat, low-carbohydrate ketogenic diet has potential therapeutic roles and has been used to manage intractable seizures since the early 1920s. The brain accounts for 25% of total body glucose metabolism and, as a result, is especially susceptible to changes in the types of nutrients consumed. Here, we discuss the principles of brain metabolism with a focus on the distinct effects of the Western and ketogenic diets on the progression of neurological diseases such as epilepsy, Parkinson's disease, Alzheimer's disease, and traumatic brain injury, highlighting the need to further explore the potential therapeutic effects of the ketogenic diet and the importance of standardizing dietary formulations to assure the reproducibility of clinical trials.
Collapse
Affiliation(s)
| | - Fatima Ahmad
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Stanley Ibeh
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Sarah Mantash
- PRASE and Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Fatima Kobeissy
- Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Hawraa Issa
- PRASE and Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Ralph Habis
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali Tfaily
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Comprehensive Epilepsy Program, Department of Neurology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Sanaa Nabha
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Hayat Harati
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Mohammad Amine Reslan
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Yara Yehya
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Chloe Barsa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Abdullah Shaito
- Biomedical Research Center, Department of Biomedical Sciences at College of Health Sciences, and College of Medicine, Qatar University, Doha, Qatar
| | - Kazem Zibara
- PRASE and Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt
| | - Firas H Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience, and Chemistry, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
148
|
Huang Y, Wei Y, Xu J, Wei X. A comprehensive review on the prevention and regulation of Alzheimer's disease by tea and its active ingredients. Crit Rev Food Sci Nutr 2022; 63:10560-10584. [PMID: 35647742 DOI: 10.1080/10408398.2022.2081128] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) has brought a heavy burden to society as a representative neurodegenerative disease. The etiology of AD combines multiple factors, concluding family, gender, head trauma, diseases and social psychology. There are multiple hypotheses explaining the pathogenesis of AD such as β-amyloid (Aβ) deposition and tau hyperphosphorylation, which lead to extracellular amyloid plaques and neurofibrillary tangles in neurons. The existing therapeutic drugs have several disadvantages including single target, poor curative effect, and obvious side effects. Tea contains many bioactive components, such as tea polyphenols (TPP), L-theanine (L-TH), tea pigment, tea polysaccharides and caffeine. The epidemiological investigations have shown that drinking tea can reduce the risk of AD. The mechanisms of tea active ingredients in the prevention and regulation of AD includes reducing the generation and aggregation of Aβ; inhibiting tau aggregation and hyperphosphorylation; inhibiting neuronal apoptosis and regulate neurotransmitters; relieving oxidative stress and neuroinflammation as well as the regulation of intestinal flora. This review summarizes the different signaling pathways that tea active ingredients regulate AD. Furthermore, we propose the main limitations of current research and future research directions, hoping to contribute to the development of natural functional foods based on tea active ingredients in the prevention and treatment of AD.
Collapse
Affiliation(s)
- Yi Huang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, PR China
| | - Yang Wei
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, PR China
| | - Jia Xu
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, PR China
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, PR China
| | - Xinlin Wei
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, PR China
| |
Collapse
|
149
|
Nabi M, Tabassum N. Role of Environmental Toxicants on Neurodegenerative Disorders. FRONTIERS IN TOXICOLOGY 2022; 4:837579. [PMID: 35647576 PMCID: PMC9131020 DOI: 10.3389/ftox.2022.837579] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/22/2022] [Indexed: 12/22/2022] Open
Abstract
Neurodegeneration leads to the loss of structural and functioning components of neurons over time. Various studies have related neurodegeneration to a number of degenerative disorders. Neurological repercussions of neurodegeneration can have severe impacts on the physical and mental health of patients. In the recent past, various neurodegenerative ailments such as Alzheimer’s and Parkinson’s illnesses have received global consideration owing to their global occurrence. Environmental attributes have been regarded as the main contributors to neural dysfunction-related disorders. The majority of neurological diseases are mainly related to prenatal and postnatal exposure to industrially produced environmental toxins. Some neurotoxic metals, like lead (Pb), aluminium (Al), Mercury (Hg), manganese (Mn), cadmium (Cd), and arsenic (As), and also pesticides and metal-based nanoparticles, have been implicated in Parkinson’s and Alzheimer’s disease. The contaminants are known for their ability to produce senile or amyloid plaques and neurofibrillary tangles (NFTs), which are the key features of these neurological dysfunctions. Besides, solvent exposure is also a significant contributor to neurological diseases. This study recapitulates the role of environmental neurotoxins on neurodegeneration with special emphasis on major neurodegenerative disorders such as Alzheimer’s and Parkinson’s disease.
Collapse
Affiliation(s)
- Masarat Nabi
- Department of Environmental Science, University of Kashmir, Srinagar, India
- *Correspondence: Masarat Nabi, , orcid.org/0000-0003-1677-6498; Nahida Tabassum,
| | - Nahida Tabassum
- Department of Pharmaceutical Sciences, University of Kashmir, Srinagar, India
- *Correspondence: Masarat Nabi, , orcid.org/0000-0003-1677-6498; Nahida Tabassum,
| |
Collapse
|
150
|
Mietelska-Porowska A, Domańska J, Want A, Więckowska-Gacek A, Chutorański D, Koperski M, Wojda U. Induction of Brain Insulin Resistance and Alzheimer's Molecular Changes by Western Diet. Int J Mol Sci 2022; 23:ijms23094744. [PMID: 35563135 PMCID: PMC9102094 DOI: 10.3390/ijms23094744] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 02/06/2023] Open
Abstract
The term Western diet (WD) describes the consumption of large amounts of highly processed foods, rich in simple sugars and saturated fats. Long-term WD feeding leads to insulin resistance, postulated as a risk factor for Alzheimer’s disease (AD). AD is the main cause of progressive dementia characterized by the deposition of amyloid-β (Aβ) plaques and neurofibrillary tangles consisting of the hyperphosphorylated tau (p-Tau) protein in the brain, starting from the entorhinal cortex and the hippocampus. In this study, we report that WD-derived impairment in insulin signaling induces tau and Aβ brain pathology in wild-type C57BL/6 mice, and that the entorhinal cortex is more sensitive than the hippocampus to the impairment of brain insulin signaling. In the brain areas developing WD-induced insulin resistance, we observed changes in p-Tau(Thr231) localization in neuronal subcellular compartments, indicating progressive tauopathy, and a decrease in amyloid precursor protein levels correlating with the appearance of Aβ peptides. These results suggest that WD promotes the development of AD and may be considered not only a risk factor, but also a modifiable trigger of AD.
Collapse
|