1
|
Darabi S, Gorgich EAC, Moradi F, Rustamzadeh A. Lipidopathy disrupts peripheral and central amyloid clearance in Alzheimer's disease: Where are our knowledge. IBRO Neurosci Rep 2025; 18:191-199. [PMID: 39906286 PMCID: PMC11791331 DOI: 10.1016/j.ibneur.2025.01.004] [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: 08/15/2024] [Revised: 12/09/2024] [Accepted: 01/04/2025] [Indexed: 02/06/2025] Open
Abstract
Amyloid-beta (Aβ) production is a normal physiological process, essential for neuronal function. However, an imbalance in Aβ production and clearance is the central pathological feature of Alzheimer's disease (AD), leading to the accumulation of Aβ plaques in the brain. Low-density lipoprotein receptor-related protein 1 (LRP1) plays a critical role in both the central clearance of Aβ from the brain and its peripheral transport to visceral organs. Disruptions in these processes contribute to the accumulation of Aβ in the central nervous system (CNS) and the progression of AD. Recent research emphasizes the need for a broader focus on the systemic effects of organs outside the brain, particularly in the context of AD prevention and treatment. The contribution of peripheral systems, such as the liver, in Aβ clearance, is vital, given that Aβ levels in the plasma correlate closely with those in the brain. Consequently, targeting systemic processes, rather than focusing solely on the CNS, may offer promising therapeutic approaches. Furthermore, high-density lipoprotein (HDL) facilitates the formation of lipoprotein-amyloid complexes, which are important for Aβ transport and clearance, using proteins such as apolipoproteins E and J (ApoE and ApoJ) to form complexes that help manage Aβ accumulation. On the other hand, low-density lipoprotein (LDL) facilitates Aβ efflux from the brain by binding to LRP1, promoting its clearance. Given the relationship between lipid profiles and Aβ levels, along with lipid-modifying drugs, may be effective in managing Aβ accumulation and mitigating AD progression.
Collapse
Affiliation(s)
- Shahram Darabi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-communicable Diseases, Department of Anatomical Sciences, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | | | - Fatemeh Moradi
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Auob Rustamzadeh
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-communicable Diseases, Department of Anatomical Sciences, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
2
|
Niu J, Zhu G, Zhang J. Ginseng in delaying brain aging: Progress and Perspectives. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156587. [PMID: 40049102 DOI: 10.1016/j.phymed.2025.156587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/23/2025] [Accepted: 02/26/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND The Shennong Bencao Jing (Shennong's Classic of Materia Medica) records that Panax ginseng C. A. Mey (ginseng) 'lightens the body and prolongs life'. Many investigations have documented that ginseng exerts neuroprotective effects by mitigating the aging of the brain. However, a comprehensive review of the impacts of ginseng on brain aging remains lacking. PURPOSE This study aims to review the advances in ginseng research regarding its role in delaying brain aging, focusing on its bioactive constituents, underlying mechanisms and potential side effects. The findings provide scientific pieces of evidence to support the medical utilization of ginseng in the delaying senescence and the management of aging-related diseases. METHODS This review includes studies on ginseng and brain aging in humans, retrieved from English-language research articles published between 2017 and the present in the PubMed and Web of Science databases. The work focused on ginseng, brain aging, and aging-related diseases, utilizing keywords such as "Ginseng", "Brain aging", "central nervous system", "intracellular homeostasis", "peripheral system", etc. RESULTS: Ginseng comprises a varied spectrum of biologically bioactive constituents, such as ginsenosides, Maillard reaction products, ginseng polysaccharides, volatile oils, amino acids, proteins, etc. These components work to contribute to their significant medicinal value. Based on the traditional Chinese medicine (TCM) theory that "the heart and brain are interconnected, the liver and brain are mutually supportive, the brain and spleen are related, the brain and lung are linked, and the brain and kidney work in harmony," we summarize that ginseng may sustain neural homeostasis through both central and peripheral perspectives. Additionally, the potential toxic side effects of ginseng are minimal. CONCLUSION Ginseng and its bioactive constituents exhibit considerable promise in delaying brain aging and treating neurodegenerative diseases. Future research should prioritize exploring the direct targets of ginseng and its active ingredients, and work toward establishing precise drug-target-efficacy relationships. This approach will facilitate the translation of these findings into clinically viable therapeutic approaches.
Collapse
Affiliation(s)
- Jingwen Niu
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China; Key Laboratory of Xin'an Medicine, the Ministry of Education, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Guoqi Zhu
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China; Key Laboratory of Xin'an Medicine, the Ministry of Education, Anhui University of Chinese Medicine, Hefei 230038, China.
| | - Junjie Zhang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China; Key Laboratory of Xin'an Medicine, the Ministry of Education, Anhui University of Chinese Medicine, Hefei 230038, China.
| |
Collapse
|
3
|
Sonsalla MM, Babygirija R, Johnson M, Cai S, Cole M, Yeh CY, Grunow I, Liu Y, Vertein D, Calubag MF, Trautman ME, Green CL, Rigby MJ, Puglielli L, Lamming DW. Acarbose ameliorates Western diet-induced metabolic and cognitive impairments in the 3xTg mouse model of Alzheimer's disease. GeroScience 2025; 47:1569-1591. [PMID: 39271570 PMCID: PMC11978593 DOI: 10.1007/s11357-024-01337-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
Age is the greatest risk factor for Alzheimer's disease (AD) as well as for other disorders that increase the risk of AD such as diabetes and obesity. There is growing interest in determining if interventions that promote metabolic health can prevent or delay AD. Acarbose is an anti-diabetic drug that not only improves glucose homeostasis, but also extends the lifespan of wild-type mice. Here, we test the hypothesis that acarbose will not only preserve metabolic health, but also slow or prevent AD pathology and cognitive deficits in 3xTg mice, a model of AD, fed either a Control diet or a high-fat, high-sucrose Western diet (WD). We find that acarbose decreases the body weight and adiposity of WD-fed 3xTg mice, increasing energy expenditure while also stimulating food consumption, and improves glycemic control. Both male and female WD-fed 3xTg mice have worsened cognitive deficits than Control-fed mice, and these deficits are ameliorated by acarbose treatment. Molecular and histological analysis of tau and amyloid pathology identified sex-specific effects of acarbose which are uncoupled from the dramatic improvements in cognition in females, suggesting that the benefits of acarbose on AD may be largely driven by improved metabolic health. In conclusion, our results suggest that acarbose may be a promising intervention to prevent, delay, or even treat AD, especially in individuals consuming a WD.
Collapse
Affiliation(s)
- Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Madeline Johnson
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Samuel Cai
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mari Cole
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Yang Liu
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Endocrinology and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Diana Vertein
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
| | - Mariah F Calubag
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michaela E Trautman
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Cara L Green
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michael J Rigby
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA.
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA.
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
- Endocrinology and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, WI, USA.
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
- University of Wisconsin Carbone Cancer Center, Madison, WI, 53705, USA.
- University of Wisconsin-Madison Comprehensive Diabetes Center, Madison, WI, 53705, USA.
| |
Collapse
|
4
|
Jiang Y, Zhang H, Shi J, Shan T, Liu M, Wang P, Liang X, Liang H. Nicotinamide riboside alleviates sweeteners-induced brain and cognitive impairments in immature mice. Food Funct 2025; 16:1947-1968. [PMID: 39957299 DOI: 10.1039/d4fo05553e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Abstract
The consumption of sweeteners is high around the world. Sweet beverages are one of the most important and popular sources of sweeteners. Previous studies have reported that excessive sweeteners might cause health hazards, including cognitive impairment. Nicotinamide riboside (NR), a precursor of NAD+, has been found to alleviate several cognitive impairments. However, the protective effects of NR against sweetener-induced cognitive impairment remain unclear. Hence, we evaluated the effects of sweeteners and NR (400 mg kg-1 d-1) on the brain and cognition of mice by simulating an extreme lifestyle of completely replacing water with sugar-sweetened beverage (simulated with 10% sucrose solution) or sugar-free sweet beverage (simulated with 0.05% aspartame solution) from weaning to adulthood. The results revealed that continuous exposure to sucrose or aspartame for eight weeks did not significantly cause differences in body weight but significantly induced cognitive impairments, including anxiety- and depressive-like behaviours, impairments in learning, memory and sociability. Moreover, sucrose or aspartame exposure induced neuronal injury, reduction of Nissl bodies, overactivation of the TLR4/NF-κB/NLRP3/ASC/Caspase-1 pathway and increased downstream inflammatory cytokines in mouse hippocampus, and also induced an imbalance of oxidative stress, apoptosis and autophagy, large consumptions of intracellular antioxidant factors, and overactivation of the PI3K/Akt/FOXO1 and PI3K/Akt/mTOR pathways in mouse brain. NR treatment increased NAD+ in the brain, and prevented and alleviated these impairments effectively. In summary, we found that NR supplementation protected against cognitive impairment caused by sucrose or aspartame in immature mice, which might be related to increased brain NAD+ level, relieved neuroinflammation and pyroptosis in the hippocampus, and maintained a balance of oxidative stress, apoptosis and autophagy in the brain.
Collapse
Affiliation(s)
- Yushan Jiang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Qingdao, China.
| | - Huaqi Zhang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Qingdao, China.
| | - Jing Shi
- College of continuing education, Qingdao University, Qingdao, China
| | - Tianhu Shan
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Qingdao, China.
| | - Man Liu
- Basic Medical College, Qingdao University, Qingdao, China
| | - Peng Wang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Qingdao, China.
| | - Xi Liang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Qingdao, China.
| | - Hui Liang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Qingdao, China.
| |
Collapse
|
5
|
Mikkelsen ACD, Kjærgaard K, Schapira AHV, Mookerjee RP, Thomsen KL. The liver-brain axis in metabolic dysfunction-associated steatotic liver disease. Lancet Gastroenterol Hepatol 2025; 10:248-258. [PMID: 39701123 DOI: 10.1016/s2468-1253(24)00320-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 12/21/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) affects around 30% of the global population. Studies suggest that MASLD is associated with compromised brain health and cognitive dysfunction, initiating a growing interest in exploring the liver-brain axis mechanistically within MASLD pathophysiology. With the prevalence of MASLD increasing at an alarming rate, leaving a large proportion of people potentially at risk, cognitive dysfunction in MASLD is a health challenge that requires careful consideration and awareness. This Review summarises the current literature on cognitive function in people with MASLD and discusses plausible causes for its impairment. It is likely that a multifaceted spectrum of factors works collectively to affect cognition in patients with MASLD. We describe the role of inflammation, vascular disease, and brain ageing and neurodegeneration as possible key players. This Review also highlights the need for future studies to identify the optimal test for diagnosing cognitive dysfunction in patients with MASLD, to examine the correlation between MASLD progression and the severity of cognitive dysfunction, and to evaluate whether new MASLD-targeted therapies also improve brain dysfunction.
Collapse
Affiliation(s)
- Anne Catrine Daugaard Mikkelsen
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Kristoffer Kjærgaard
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, University College London Institute of Neurology, London, UK
| | - Rajeshwar P Mookerjee
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark; Institute for Liver and Digestive Health, University College London, London, UK
| | - Karen Louise Thomsen
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Institute for Liver and Digestive Health, University College London, London, UK.
| |
Collapse
|
6
|
Qiao M, Ni J, Qing H, Qiu Y, Quan Z. Role of Peripheral NLRP3 Inflammasome in Cognitive Impairments: Insights of Non-central Factors. Mol Neurobiol 2025:10.1007/s12035-025-04779-8. [PMID: 40000575 DOI: 10.1007/s12035-025-04779-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/13/2025] [Indexed: 02/27/2025]
Abstract
Cognitive impairments are common clinical manifestation of Alzheimer's disease, vascular dementia, type 2 diabetes mellitus, and autoimmune diseases. Emerging evidence has suggested a strong correlation between peripheral chronic inflammation and cognitive impairments. For example, nearly 40% of individuals with inflammatory bowel disease also suffer from cognitive impairments. In this condition, NLRP3 inflammasome (NLRP3-I) generating pro-inflammatory cytokines like IL-1β serves as a significant effector, and its persistence exerts adverse effects to both periphery and the brain. Moreover, investigations on serum biomarkers of mild cognitive impairments have shown NLRP3-I components' upregulation, suggesting the involvement of peripheral inflammasome pathway in this disorder. Here, we systematically reviewed the current knowledge of NLRP3-I in inflammatory disease to uncover its potential role in bridging peripheral chronic inflammation and cognitive impairments. This review summarizes the molecular features and ignition process of NLRP3-I in inflammatory response. Meanwhile, various effects of NLRP3-I involved in peripheral inflammation-associated disease are also reviewed, especially its chronic disturbances to brain homeostasis and cognitive function through routes including gut-brain, liver-brain, and kidney-brain axes. In addition, current promising compounds and their targets relative to NLRP3-I are discussed in the context of cognitive impairments. Through the detailed investigation, this review highlights the critical role of peripheral NLRP3-I in the pathogenesis of cognitive disorders, and offers novel perspectives for developing effective therapeutic interventions for diseases associated with cognitive impairments. The present review outlines the current knowledge on the ignition of NLRP3-I in inflammatory disease and more importantly, emphasizes the role of peripheral NLRP3-I as a causal pathway in the development of cognitive disorders. Although major efforts to restrain cognitive decline are mainly focused on the central nervous system, it has become clear that disturbances from peripheral immune are closely associated with the dysfunctional brain. Therefore, attenuation of these inflammatory changes through inhibiting the NLRP3-I pathway in early inflammatory disease may reduce future risk of cognitive impairments, and in the meantime, considerations on such pathogenesis for combined drug therapy will be required in the clinical evaluation of cognitive disorders.
Collapse
Affiliation(s)
- Mengfan Qiao
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
- Department of Biology, Shenzhen MSU-BIT University, Shenzhen, 518172, China
| | - Yunjie Qiu
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| | - Zhenzhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| |
Collapse
|
7
|
Ma C, Wang Y. BHLHE40 regulates microglia polarization after spinal cord injury via the NF-κB pathway. Brain Res Bull 2025; 220:111139. [PMID: 39586332 DOI: 10.1016/j.brainresbull.2024.111139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 11/07/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Spinal cord injury (SCI) is a devastating disease characterized by neuroinflammation and irreversible neuronal loss. The basic helix-loop-helix family member e40 (Bhlhe40) is a stress-responsive transcription factor involved in the pathological process of inflammation. However, Bhlhe40 expression and its role in SCI are largely unknown. SCI rat models were established with an aneurysm clip and then the rats were injected with lentiviral Bhlhe40 shRNA to knock down Bhlhe40 expression. In vitro, BV2 microglia cells were stimulated with LPS and IFN-γ to promote M1 microglia polarization. The results showed that Bhlhe40 expression was significantly elevated in the injured spinal cord tissue. Bhlhe40 deficiency reduced neuroinflammation and neuronal loss, and then promoted the recovery of neurological function. Additionally, Bhlhe40 knockdown alleviated neuronal apoptosis by regulating microglia polarization. In our study, Bhlhe40 knockdown inhibited M1 microglia polarization and the secretion of pro-inflammatory factors (TNF-α, IL-1β, and IL-6). Meanwhile, the NF-κB pathway was inhibited after the Bhlhe40 knockdown in SCI rats. To further explore the functional role of Bhlhe40, we performed in vitro experiments. Bhlhe40 knockdown decreased M1 microglia polarization by inhibiting the NF-κB pathway. In conclusion, our study indicates that Bhlhe40 knockdown can alleviate the progression of SCI and its underlying mechanism in regulating macrophage polarization through the NF-κB pathway.
Collapse
Affiliation(s)
- Chao Ma
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yansong Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China; Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, Harbin Medical University, Harbin, China.
| |
Collapse
|
8
|
Agarwal P, Barnes LL, Dhana K, Liu X, Zhang Y, Beck T, Cornelis MC, Tangney C, Rajan KB. Association of MIND diet with cognitive decline among Black and White older adults. Alzheimers Dement 2024; 20:8461-8469. [PMID: 39410855 DOI: 10.1002/alz.14277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION We examined the Mediterranean-Dietary Approaches to Stop Hypertension Intervention for Neurodegenerative Delay (MIND) diet's association with cognitive decline by race among older adults in the Chicago Health and Aging Project. METHODS Five thousand two hundred fifty-nine participants (73.5 [± 6.0] years, 62% Black participants, 62% female) completed a food frequency questionnaire, and two or more cognitive assessments over 7.8 ± 4.6 years. RESULTS Overall, higher MIND diet was associated with slower cognitive decline (p for trend = 0.0025). The MIND score (range:0-15) was different between Black and White older adults(6.97 vs. 7.12, p = 0.010). Compared to the lowest tertile, among White participants, the two highest tertiles (MIND score -7: β = 0.0121 [95% confidence interval [CI]: 0.0006, 0.0237]; MIND score -8.5: β = 0.0146 [95% CI: 0.0003, 0.0260]) and among Black participants, only the highest tertile (MIND score -8.5: β = 0.0088 [95% CI: 0.0003, 0.0172]) had association with cognitive decline. Vascular and lifestyle factors attenuated the association only for Black older adults. DISCUSSION The MIND diet was associated with slower cognitive decline in Black and White older adults, but this may vary with other lifestyle and vascular factors. Further research is warranted on race-specific cultural diets considering other risk factors for cognitive decline. HIGHLIGHTS The intake of Mediterranean-Dietary Approaches to Stop Hypertension Intervention for Neurodegenerative Delay (MIND) diet components varies by race. The MIND diet may slow cognitive decline in both Black and White older adults. This association may vary with other lifestyle and vascular risk factors.
Collapse
Affiliation(s)
- Puja Agarwal
- Rush Alzheimer's Disease Center, Department of Internal Medicine, Department of Clinical Nutrition, Rush University Medical Center, Chicago, Illinois, USA
| | - Lisa L Barnes
- Rush Alzheimer's Disease Center, Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Klodian Dhana
- Rush Institute of Healthy Aging, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Xiaoran Liu
- Rush Institute of Healthy Aging, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Yanyu Zhang
- Rush Institute of Healthy Aging, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Todd Beck
- Rush Institute of Healthy Aging, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Marilyn C Cornelis
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Christy Tangney
- Department of Family and Preventive Medicine, Department of Clinical Nutrition, Rush University Medical Center, Chicago, Illinois, USA
| | - Kumar B Rajan
- Rush Institute of Healthy Aging, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
9
|
Li T, Yi J, Hu X, Wu H, Wang K, Zhou B. Association between dementia, Alzheimer's disease, and liver cancer: A Mendelian randomization analysis. J Alzheimers Dis Rep 2024; 8:1587-1595. [PMID: 40034364 PMCID: PMC11863743 DOI: 10.1177/25424823241299661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 10/21/2024] [Indexed: 03/05/2025] Open
Abstract
Background Epidemiological studies have indicated an inverse association between neurodegenerative diseases and cancer. However, the relationship between liver cancer and dementia, particularly Alzheimer's disease (AD), remains underexplored. Objective This study aimed to determine the association between dementia, specifically AD, and liver cancer using Mendelian randomization (MR) analysis. Methods We conducted a bidirectional, two-sample MR analysis using publicly available genome-wide association studies data. The inverse-variance weighted method was employed as the primary analytical approach. To detect and correct for the effects of horizontal pleiotropy, we applied three complementary methods: MR Egger, weighted median, and Maximum likelihood. Results The analysis indicated significant associations between dementia and a reduced risk of hepatocellular carcinoma (HCC) (OR: 0.87; 95%CI: 0.81-0.95; p < 0.001) and intrahepatic cholangiocarcinoma (ICC) (OR: 0.81; 95%CI: 0.72-0.92; p < 0.001). AD was significantly associated with a decreased risk of HCC (OR: 0.94; 95%CI: 0.88-0.99; p = 0.033), ICC (OR: 0.85; 95%CI: 0.78-0.93; p < 0.001), and combined hepatocellular-cholangiocarcinoma (CHC) (OR: 0.64; 95%CI: 0.43-0.93; p = 0.020). Conversely, inverse MR analyses indicated that ICC was associated with increased dementia risk (OR: 1.05; 95%CI: 1.01-1.09; p = 0.019) and CHC with increased AD risk (OR: 1.03; 95%CI: 1.00-1.04; p = 0.014). Conclusions This study suggests that dementia, particularly AD, is associated with a reduced risk of liver cancer. Conversely, liver cancer may be associated with a slightly increased risk of developing dementia and AD, although some observational studies have reported a lower risk of these conditions among cancer survivors.
Collapse
Affiliation(s)
- Tianze Li
- Hepato-Biliary-Pancreatic Surgery Division, Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Queen Mary School, Nanchang University, Nanchang, Jiangxi Province, P.R. China
| | - Jianwei Yi
- Hepato-Biliary-Pancreatic Surgery Division, Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xuliang Hu
- Hepato-Biliary-Pancreatic Surgery Division, Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Huajun Wu
- Hepato-Biliary-Pancreatic Surgery Division, Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Kai Wang
- Hepato-Biliary-Pancreatic Surgery Division, Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Engineering Research Center of Hepatobiliary Disease, Nanchang, China
| | - Binghai Zhou
- Hepato-Biliary-Pancreatic Surgery Division, Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
10
|
Wu Q, Geng Z, Lu J, Wang S, Yu Z, Wang S, Ren X, Guan S, Liu T, Zhu C. Neddylation of protein, a new strategy of protein post-translational modification for targeted treatment of central nervous system diseases. Front Neurosci 2024; 18:1467562. [PMID: 39564524 PMCID: PMC11573765 DOI: 10.3389/fnins.2024.1467562] [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: 07/20/2024] [Accepted: 10/17/2024] [Indexed: 11/21/2024] Open
Abstract
Neddylation, a type of protein post-translational modification that links the ubiquitin-like protein NEDD8 to substrate proteins, can be involved in various significant cellular processes and generate multiple biological effects. Currently, the best-characterized substrates of neddylation are the Cullin protein family, which is the core subunit of the Cullin-RING E3 ubiquitin ligase complex and controls many important biological processes by promoting ubiquitination and subsequent degradation of various key regulatory proteins. The normal or abnormal process of protein neddylation in the central nervous system can lead to a series of occurrences of normal functions and the development of diseases, providing an attractive, reasonable, and effective targeted therapeutic strategy. Therefore, this study reviews the phenomenon of neddylation in the central nervous system and summarizes the corresponding substrates. Finally, we provide a detailed description of neddylation involved in CNS diseases and treatment methods that may be used to regulate neddylation for the treatment of related diseases.
Collapse
Affiliation(s)
- Qian Wu
- Department of Neurology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ziang Geng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jun Lu
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shisong Wang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhongxue Yu
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Siqi Wang
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaolin Ren
- Department of Neurosurgery, Shenyang Red Cross Hospital, Shenyang, Liaoning, China
| | - Shu Guan
- Department of Surgical Oncology and Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Tiancong Liu
- Department of Otolaryngology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chen Zhu
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
11
|
Bano N, Khan S, Ahamad S, Kanshana JS, Dar NJ, Khan S, Nazir A, Bhat SA. Microglia and gut microbiota: A double-edged sword in Alzheimer's disease. Ageing Res Rev 2024; 101:102515. [PMID: 39321881 DOI: 10.1016/j.arr.2024.102515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/06/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
The strong association between gut microbiota (GM) and brain functions such as mood, behaviour, and cognition has been well documented. Gut-brain axis is a unique bidirectional communication system between the gut and brain, in which gut microbes play essential role in maintaining various molecular and cellular processes. GM interacts with the brain through various pathways and processes including, metabolites, vagus nerve, HPA axis, endocrine system, and immune system to maintain brain homeostasis. GM dysbiosis, or an imbalance in GM, is associated with several neurological disorders, including anxiety, depression, and Alzheimer's disease (AD). Conversely, AD is sustained by microglia-mediated neuroinflammation and neurodegeneration. Further, GM and their products also affect microglia-mediated neuroinflammation and neurodegeneration. Despite the evidence connecting GM dysbiosis and AD progression, the involvement of GM in modulating microglia-mediated neuroinflammation in AD remains elusive. Importantly, deciphering the mechanism/s by which GM regulates microglia-dependent neuroinflammation may be helpful in devising potential therapeutic strategies to mitigate AD. Herein, we review the current evidence regarding the involvement of GM dysbiosis in microglia activation and neuroinflammation in AD. We also discuss the possible mechanisms through which GM influences the functioning of microglia and its implications for therapeutic intervention. Further, we explore the potential of microbiota-targeted interventions, such as prebiotics, probiotics, faecal microbiota transplantation, etc., as a novel therapeutic strategy to mitigate neuroinflammation and AD progression. By understanding and exploring the gut-brain axis, we aspire to revolutionize the treatment of neurodegenerative disorders, many of which share a common theme of microglia-mediated neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Nargis Bano
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Sameera Khan
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India.
| | - Jitendra Singh Kanshana
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburg, PA, USA.
| | - Nawab John Dar
- CNB, SALK Institute of Biological Sciences, La Jolla, CA 92037, USA.
| | - Sumbul Khan
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Aamir Nazir
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research, New Delhi, India.
| | - Shahnawaz Ali Bhat
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
12
|
Wen W, Huang SM, Zhang B. Mechanisms Underlying Obesity-induced Aβ Accumulation in Alzheimer's Disease: A Qualitative Review. J Integr Neurosci 2024; 23:163. [PMID: 39344225 DOI: 10.31083/j.jin2309163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/27/2024] [Accepted: 03/05/2024] [Indexed: 10/01/2024] Open
Abstract
Epidemiological studies show that individuals with obesity are more likely to develop Alzheimer's disease (AD) than those who do not have obesity. However, the mechanisms underlying the relationship between obesity and AD are not entirely unclear. Here, we have reviewed and analyzed relevant articles published in the literature and found that obesity has correlation or potential increase in the levels of β-amyloid (Aβ) protein, which may explain why people with obesity are more likely to suffer from AD. Additionally, the published findings point to the roles of obesity-related metabolic disorders, such as diabetes, inflammation, oxidative stress, and imbalance in gut microbiota in Aβ accumulation caused by obesity. Therefore, in-depth experimental and clinical studies on these mechanisms in the future may help shed light on appropriate prevention and treatment strategies for AD, such as dietary changes and regular exercise to reverse or prevent obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Wei Wen
- Department of Pharmacology, College of Basic Medicine, Heilongjiang University of Chinese Medicine, 150040 Harbin, Heilongjiang, China
| | - Shu-Ming Huang
- Department of Neuroscience, Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, 150040 Harbin, Heilongjiang, China
| | - Bo Zhang
- Department of Neuroscience, Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, 150040 Harbin, Heilongjiang, China
| |
Collapse
|
13
|
Li S, Cai Y, Wang S, Luo L, Zhang Y, Huang K, Guan X. Gut microbiota: the indispensable player in neurodegenerative diseases. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:7096-7108. [PMID: 38572789 DOI: 10.1002/jsfa.13509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/26/2024] [Accepted: 04/04/2024] [Indexed: 04/05/2024]
Abstract
As one of the most urgent social and health problems in the world, neurodegenerative diseases have always been of interest to researchers. However, the pathological mechanisms and therapeutic approaches are not achieved. In addition to the established roles of oxidative stress, inflammation and immune response, changes of gut microbiota are also closely related to the pathogenesis of neurodegenerative diseases. Gut microbiota is the central player of the gut-brain axis, the dynamic bidirectional communication pathway between gut microbiota and central nervous system, and emerging insights have confirmed its indispensability in the development of neurodegenerative diseases. In this review, we discuss the complex relationship between gut microbiota and the central nervous system from the perspective of the gut-brain axis; review the mechanism of microbiota for the modulation different neurodegenerative diseases and discuss how different dietary patterns affect neurodegenerative diseases via gut microbiota; and prospect the employment of gut microbiota in the therapeutic approach to those diseases. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Sen Li
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai, China
| | - Yuwei Cai
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai, China
| | - Shuo Wang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai, China
| | - Lei Luo
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai, China
| | - Yu Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai, China
| | - Kai Huang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai, China
| | - Xiao Guan
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai, China
| |
Collapse
|
14
|
Sonsalla MM, Babygirija R, Johnson M, Cai S, Cole M, Yeh CY, Grunow I, Liu Y, Vertein D, Calubag MF, Trautman ME, Green CL, Rigby MJ, Puglielli L, Lamming DW. Acarbose ameliorates Western diet-induced metabolic and cognitive impairments in the 3xTg mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.600472. [PMID: 39005334 PMCID: PMC11244897 DOI: 10.1101/2024.06.27.600472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Age is the greatest risk factor for Alzheimer's disease (AD) as well as for other disorders that increase the risk of AD such as diabetes and obesity. There is growing interest in determining if interventions that promote metabolic health can prevent or delay AD. Acarbose is an anti-diabetic drug that not only improves glucose homeostasis, but also extends the lifespan of wild-type mice. Here, we test the hypothesis that acarbose will not only preserve metabolic health, but also slow or prevent AD pathology and cognitive deficits in 3xTg mice, a model of AD, fed either a Control diet or a high-fat, high-sucrose Western diet (WD). We find that acarbose decreases the body weight and adiposity of WD-fed 3xTg mice, increasing energy expenditure while also stimulating food consumption, and improves glycemic control. Both male and female WD-fed 3xTg mice have worsened cognitive deficits than Control-fed mice, and these deficits are ameliorated by acarbose treatment. Molecular and histological analysis of tau and amyloid pathology identified sex-specific effects of acarbose which are uncoupled from the dramatic improvements in cognition, suggesting that the benefits of acarbose on AD are largely driven by improved metabolic health. In conclusion, our results suggest that acarbose may be a promising intervention to prevent, delay, or even treat AD, especially in individuals consuming a Western diet.
Collapse
|
15
|
Tsay HJ, Gan YL, Su YH, Sun YY, Yao HH, Chen HW, Hsu YT, Hsu JTA, Wang HD, Shie FS. Reducing brain Aβ burden ameliorates high-fat diet-induced fatty liver disease in APP/PS1 mice. Biomed Pharmacother 2024; 173:116404. [PMID: 38471275 DOI: 10.1016/j.biopha.2024.116404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/18/2024] [Accepted: 03/06/2024] [Indexed: 03/14/2024] Open
Abstract
High-fat diet (HFD)-induced fatty liver disease is a deteriorating risk factor for Alzheimer's disease (AD). Mitigating fatty liver disease has been shown to attenuate AD-like pathology in animal models. However, it remains unclear whether enhancing Aβ clearance through immunotherapy would in turn attenuate HFD-induced fatty liver or whether its efficacy would be compromised by long-term exposure to HFD. Here, the therapeutic potentials of an anti-Aβ antibody, NP106, was investigated in APP/PS1 mice by HFD feeding for 44 weeks. The data demonstrate that NP106 treatment effectively reduced Aβ burden and pro-inflammatory cytokines in HFD-fed APP/PS1 mice and ameliorated HFD-aggravated cognitive impairments during the final 18 weeks of the study. The rejuvenating characteristics of microglia were evident in APP/PS1 mice with NP106 treatment, namely enhanced microglial Aβ phagocytosis and attenuated microglial lipid accumulation, which may explain the benefits of NP106. Surprisingly, NP106 also reduced HFD-induced hyperglycemia, fatty liver, liver fibrosis, and hepatic lipids, concomitant with modifications in the expressions of genes involved in hepatic lipogenesis and fatty acid oxidation. The data further reveal that brain Aβ burden and behavioral deficits were positively correlated with the severity of fatty liver disease and fasting serum glucose levels. In conclusion, our study shows for the first time that anti-Aβ immunotherapy using NP106, which alleviates AD-like disorders in APP/PS1 mice, ameliorates fatty liver disease. Minimizing AD-related pathology and symptoms may reduce the vicious interplay between central AD and peripheral fatty liver disease, thereby highlighting the importance of developing AD therapies from a systemic disease perspective.
Collapse
Affiliation(s)
- Huey-Jen Tsay
- Institute of Neuroscience, School of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yu-Ling Gan
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan, ROC
| | - Yu-Han Su
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan, ROC
| | - Yu-Yo Sun
- Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan, ROC
| | - Heng-Hsiang Yao
- Institute of Neuroscience, School of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Hui-Wen Chen
- Institute of Neuroscience, School of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Ying-Ting Hsu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan, ROC
| | - John Tsu-An Hsu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan, ROC
| | - Horng-Dar Wang
- Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Feng-Shiun Shie
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan, ROC.
| |
Collapse
|
16
|
Liu XL, Yeerlan J, Liu Z, Bai Y, Wang Q, Yan Y, Xu L, Jia C, Zhang L. Past, Present, and Future of Liver-Brain Axis in Alzheimer's Disease: A Bibliometric Review. J Alzheimers Dis 2024; 101:1267-1280. [PMID: 39302376 DOI: 10.3233/jad-240688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Background No effective drugs currently exist to cure Alzheimer's disease (AD) due to its complexity and the lack of understanding of the involved molecular signaling and pathways. The relationship between liver health and AD is now widely recognized. Still, molecular links and shared pathways between the liver and brain remain unclear, making the liver-brain axis in AD therapies a new area for exploration. However, bibliometric studies on this topic are lacking. Objective This study aims to review the liver-brain axis in AD and identify future research hotspots and trends through bibliometric analysis. Methods Articles and reviews related to AD and liver and its related diseases were searched in the Web of Science Core Collection (WoSCC) database up to 2024. Data were processed and visually analyzed using VOSviewer, CiteSpace, and Pajek. Results We collected 1,777 articles on AD and liver and its related diseases from 2,517 institutions across 80 countries. Keyword cluster analysis identified 11 clusters, with 'insulin resistance,' 'amyloid-beta,' 'apolipoprotein-E,' 'oxidative stress,' and 'inflammation' appearing most frequently, and exhibiting strong total link strength. These results indicate that these topics have been the primary focus of research on the liver-brain axis in AD. Conclusions This study is the first to comprehensively analyze the liver-brain axis in AD using bibliometric methods. The research results identify recent research frontiers and hotspots, aiding scholars in gaining a deeper understanding of the correlation between AD and the liver.
Collapse
Affiliation(s)
- Xin Lian Liu
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Pathology and Pathophysiology, Institute of Neuroscience, Chengdu Medical College, Chengdu, China
| | | | - Zhirong Liu
- Department of General Surgery, Chengdu Second People's Hospital, Chengdu, China
| | - Yang Bai
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - Qin Wang
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - YiRui Yan
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - LuKe Xu
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Cui Jia
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Pathology and Pathophysiology, Institute of Neuroscience, Chengdu Medical College, Chengdu, China
| | - LuShun Zhang
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Pathology and Pathophysiology, Institute of Neuroscience, Chengdu Medical College, Chengdu, China
| |
Collapse
|
17
|
Zheng M, Ye H, Yang X, Shen L, Dang X, Liu X, Gong Y, Wu Q, Wang L, Ge X, Fang X, Hou B, Zhang P, Tang R, Zheng K, Huang XF, Yu Y. Probiotic Clostridium butyricum ameliorates cognitive impairment in obesity via the microbiota-gut-brain axis. Brain Behav Immun 2024; 115:565-587. [PMID: 37981012 DOI: 10.1016/j.bbi.2023.11.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 11/06/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023] Open
Abstract
Obesity is a risk factor for cognitive dysfunction and neurodegenerative disease, including Alzheimer's disease (AD). The gut microbiota-brain axis is altered in obesity and linked to cognitive impairment and neurodegenerative disorders. Here, we targeted obesity-induced cognitive impairment by testing the impact of the probiotic Clostridium butyricum, which has previously shown beneficial effects on gut homeostasis and brain function. Firstly, we characterized and analyzed the gut microbial profiles of participants with obesity and the correlation between gut microbiota and cognitive scores. Then, using an obese mouse model induced by a Western-style diet (high-fat and fiber-deficient diet), the effects of Clostridium butyricum on the microbiota-gut-brain axis and hippocampal cognitive function were evaluated. Finally, fecal microbiota transplantation was performed to assess the functional link between Clostridium butyricum remodeling gut microbiota and hippocampal synaptic protein and cognitive behaviors. Our results showed that participants with obesity had gut microbiota dysbiosis characterized by an increase in phylum Proteobacteria and a decrease in Clostridium butyricum, which were closely associated with cognitive decline. In diet-induced obese mice, oral Clostridium butyricum supplementation significantly alleviated cognitive impairment, attenuated the deficit of hippocampal neurite outgrowth and synaptic ultrastructure, improved hippocampal transcriptome related to synapses and dendrites; a comparison of the effects of Clostridium butyricum in mice against human AD datasets revealed that many of the genes changes in AD were reversed by Clostridium butyricum; concurrently, Clostridium butyricum also prevented gut microbiota dysbiosis, colonic barrier impairment and inflammation, and attenuated endotoxemia. Importantly, fecal microbiota transplantation from donor-obese mice with Clostridium butyricum supplementation facilitated cognitive variables and colonic integrity compared with from donor obese mice, highlighting that Clostridium butyricum's impact on cognitive function is largely due to its ability to remodel gut microbiota. Our findings provide the first insights into the neuroprotective effects of Clostridium butyricum on obesity-associated cognitive impairments and neurodegeneration via the gut microbiota-gut-brain axis.
Collapse
Affiliation(s)
- Mingxuan Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Huaiyu Ye
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Lijun Shen
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xuemei Dang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xiaoli Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yuying Gong
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Qingyuan Wu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Li Wang
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 110033, China
| | - Xing Ge
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xiaoli Fang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Jiangsu 221004, China
| | - Benchi Hou
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 110033, China
| | - Peng Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Renxian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xu-Feng Huang
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, NSW 2522, Australia
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
18
|
Mustika S, Santosaningsih D, Handayani D, Rudijanto A. Impact of multiple different high-fat diets on metabolism, inflammatory markers, dysbiosis, and liver histology: study on NASH rat model induced diet. F1000Res 2023; 12:180. [PMID: 39229607 PMCID: PMC11369591 DOI: 10.12688/f1000research.129645.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/06/2023] [Indexed: 09/05/2024] Open
Abstract
Background The spectrum of non-alcoholic fatty liver disease (NAFLD), known as non-alcoholic steatohepatitis (NASH), can lead to advanced liver disease. It is known that a variety of diets play a significant role in the development of NAFLD/NASH. The goal of this study was to determine the most appropriate composition of diet to induce NASH in an animal model. Methods This research used Rattus norvegicus strain Wistar (n=27), which were divided into four groups and given each diet for 12 weeks: normal diet (ND, n=7), high-fat diet (HFD, n=6), western diet (WD, n=7), and high-fat-high-fructose diet (HFHFD, n=7). Subjects were monitored for changes in body weight. Blood samples were collected for biochemical analysis, including low-density lipoprotein (LDL), triglyceride, alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), hepatic lipase, tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and lipopolysaccharide (LPS). Fecal samples were taken for short-chain fatty acid (SCFA) analysis. Liver histology was assessed using NAS (NAFLD activity score). A statistical comparison test was carried out using the one-way ANOVA or Kruskal-Wallis test. Results The highest average body weight was observed in the WD group (346.14 g). Liver enzymes, LDL, triglyceride, propionic acid, and acetic acid did not show significantly differences among the groups. TNF-α, IL-6, and hepatic lipase were significant (p = 0.000; p = 0.000; p = 0.004) and the highest level recorded in the HFD group. Butyrate acid level also showed significances (p = 0.021) with the lowest concentration seen in the HFHFD group (4.77 mMol/g). Only WD and HFHFD had a NAS ≥ 5 (14% and 14%). The highest percentage of borderline NAS was found in WD (57%). Conclusions WD feeding is the most appropriate diet type to induce NASH in rats as it influences metabolic, inflammatory, dysbiosis, and liver histology of rats.
Collapse
Affiliation(s)
- Syifa Mustika
- Doctoral Program of Medical Science, Faculty of Medicine, Universitas Brawijaya, Malang, Jl. Veteran, 65145, Indonesia
| | - Dewi Santosaningsih
- Department of Clinical Microbiology, Faculty of Medicine, Universitas Brawijaya, Malang, Jl. Veteran, 65145, Indonesia
| | - Dian Handayani
- Department of Nutrition, Faculty of Health Science, Universitas Brawijaya, Malang, Jl. Veteran, 65145, Indonesia
| | - Achmad Rudijanto
- Endocrine Metabolic & Diabetes Division, Department of Internal Medicine, Faculty of Medicine, Universitas Brawijaya - Dr Saiful Anwar Hospital, Malang, Jl. Veteran, 65145, Indonesia
| |
Collapse
|
19
|
Denman CR, Park SM, Jo J. Gut-brain axis: gut dysbiosis and psychiatric disorders in Alzheimer's and Parkinson's disease. Front Neurosci 2023; 17:1268419. [PMID: 38075261 PMCID: PMC10704039 DOI: 10.3389/fnins.2023.1268419] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 10/30/2023] [Indexed: 02/17/2025] Open
Abstract
Gut dysbiosis and psychiatric symptoms are common early manifestations of Alzheimer's disease (AD) and Parkinson's disease (PD). These diseases, characterised by progressive neuron loss and pathological protein accumulation, impose debilitating effects on patients. Recently, these pathological proteins have been linked with gut dysbiosis and psychiatric disorders. The gut-brain axis links the enteric and central nervous systems, acting as a bidirectional communication pathway to influence brain function and behavior. The relationship triad between gut dysbiosis, psychiatric disorders, and neurodegeneration has been investigated in pairs; however, evidence suggests that they are all interrelated and a deeper understanding is required to unravel the nuances of neurodegenerative diseases. Therefore, this review aims to summarise the current literature on the roles of gut dysbiosis and psychiatric disorders in pathological protein-related neurodegenerative diseases. We discussed how changes in the gut environment can influence the development of psychiatric symptoms and the progression of neurodegeneration and how these features overlap in AD and PD. Moreover, research on the interplay between gut dysbiosis, psychiatric disorders, and neurodegeneration remains in its early phase. In this review, we highlighted potential therapeutic approaches aimed at mitigating gastrointestinal problems and psychiatric disorders to alter the rate of neurodegeneration. Further research to assess the molecular mechanisms underlying AD and PD pathogenesis remains crucial for developing more effective treatments and achieving earlier diagnoses. Moreover, exploring non-invasive, early preventive measures and interventions is a relatively unexplored but important avenue of research in neurodegenerative diseases.
Collapse
Affiliation(s)
- Charlotte R. Denman
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Sang Myun Park
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Republic of Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Republic of Korea
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Junghyun Jo
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Republic of Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Republic of Korea
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
| |
Collapse
|
20
|
Rudajev V, Novotny J. Cholesterol-dependent amyloid β production: space for multifarious interactions between amyloid precursor protein, secretases, and cholesterol. Cell Biosci 2023; 13:171. [PMID: 37705117 PMCID: PMC10500844 DOI: 10.1186/s13578-023-01127-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023] Open
Abstract
Amyloid β is considered a key player in the development and progression of Alzheimer's disease (AD). Many studies investigating the effect of statins on lowering cholesterol suggest that there may be a link between cholesterol levels and AD pathology. Since cholesterol is one of the most abundant lipid molecules, especially in brain tissue, it affects most membrane-related processes, including the formation of the most dangerous form of amyloid β, Aβ42. The entire Aβ production system, which includes the amyloid precursor protein (APP), β-secretase, and the complex of γ-secretase, is highly dependent on membrane cholesterol content. Moreover, cholesterol can affect amyloidogenesis in many ways. Cholesterol influences the stability and activity of secretases, but also dictates their partitioning into specific cellular compartments and cholesterol-enriched lipid rafts, where the amyloidogenic machinery is predominantly localized. The most complicated relationships have been found in the interaction between cholesterol and APP, where cholesterol affects not only APP localization but also the precise character of APP dimerization and APP processing by γ-secretase, which is important for the production of Aβ of different lengths. In this review, we describe the intricate web of interdependence between cellular cholesterol levels, cholesterol membrane distribution, and cholesterol-dependent production of Aβ, the major player in AD.
Collapse
Affiliation(s)
- Vladimir Rudajev
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Jiri Novotny
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
21
|
Agarwal P, Leurgans SE, Agrawal S, Aggarwal NT, Cherian LJ, James BD, Dhana K, Barnes LL, Bennett DA, Schneider JA. Association of Mediterranean-DASH Intervention for Neurodegenerative Delay and Mediterranean Diets With Alzheimer Disease Pathology. Neurology 2023; 100:e2259-e2268. [PMID: 36889921 PMCID: PMC10259273 DOI: 10.1212/wnl.0000000000207176] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 01/26/2023] [Indexed: 03/10/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Diet may reduce Alzheimer dementia risk and slow cognitive decline, but the understanding of the relevant neuropathologic mechanisms remains limited. The association of dietary patterns with Alzheimer disease (AD) pathology has been suggested using neuroimaging biomarkers. This study examined the association of Mediterranean-DASH Intervention for Neurodegenerative Delay (MIND) and Mediterranean dietary patterns with β-amyloid load, phosphorylated tau tangles, and global AD pathology in postmortem brain tissue of older adults. METHODS Autopsied participants of the Rush Memory and Aging Project with complete dietary information (collected through a validated food frequency questionnaire) and AD pathology data (β-amyloid load, phosphorylated tau tangles, and global AD pathology [summarized neurofibrillary tangles and neuritic and diffuse plaques]) were included in this study. Linear regression models controlled for age at death, sex, education, APOE-ε4 status, and total calories were used to investigate the dietary patterns (MIND and Mediterranean diets) and dietary components associated with AD pathology. Further effect modification was tested for APOE-ε4 status and sex. RESULTS Among our study participants (N = 581, age at death: 91.0 ± 6.3 years; mean age at first dietary assessment: 84.2 ± 5.8 years; 73% female; 6.8 ± 3.9 years of follow-up), dietary patterns were associated with lower global AD pathology (MIND: β = -0.022, p = 0.034, standardized β = -2.0; Mediterranean: β = -0.007, p = 0.039, standardized β = -2.3) and specifically less β-amyloid load (MIND: β = -0.068, p = 0.050, standardized β = -2.0; Mediterranean: β = -0.040, p = 0.004, standardized β = -2.9). The findings persisted when further adjusted for physical activity, smoking, and vascular disease burden. The associations were also retained when participants with mild cognitive impairment or dementia at the baseline dietary assessment were excluded. Those in the highest tertile of green leafy vegetables intake had less global AD pathology when compared with those in the lowest tertile (tertile 3 vs tertile 1: β = -0.115, p = 0.0038). DISCUSSION The MIND and Mediterranean diets are associated with less postmortem AD pathology, primarily β-amyloid load. Among dietary components, higher green leafy vegetable intake was associated with less AD pathology.
Collapse
Affiliation(s)
- Puja Agarwal
- From the Rush Alzheimer's Disease Center (P.A., S.E.L., N.A., B.D.J., L.L.B., D.A.B., J.A.S.), Departments of Internal Medicine (P.A., B.D.J., K.D.), Clinical Nutrition (P.A.), Neurological Sciences (S.E.L., N.T.A., L.J.C., L.L.B., D.A.B., J.A.S.) and Pathology (S.A., J.A.S.); and Rush Institute of Healthy Aging (K.D.), Rush University Medical Center, Chicago, IL.
| | - Sue E Leurgans
- From the Rush Alzheimer's Disease Center (P.A., S.E.L., N.A., B.D.J., L.L.B., D.A.B., J.A.S.), Departments of Internal Medicine (P.A., B.D.J., K.D.), Clinical Nutrition (P.A.), Neurological Sciences (S.E.L., N.T.A., L.J.C., L.L.B., D.A.B., J.A.S.) and Pathology (S.A., J.A.S.); and Rush Institute of Healthy Aging (K.D.), Rush University Medical Center, Chicago, IL
| | - Sonal Agrawal
- From the Rush Alzheimer's Disease Center (P.A., S.E.L., N.A., B.D.J., L.L.B., D.A.B., J.A.S.), Departments of Internal Medicine (P.A., B.D.J., K.D.), Clinical Nutrition (P.A.), Neurological Sciences (S.E.L., N.T.A., L.J.C., L.L.B., D.A.B., J.A.S.) and Pathology (S.A., J.A.S.); and Rush Institute of Healthy Aging (K.D.), Rush University Medical Center, Chicago, IL
| | - Neelum T Aggarwal
- From the Rush Alzheimer's Disease Center (P.A., S.E.L., N.A., B.D.J., L.L.B., D.A.B., J.A.S.), Departments of Internal Medicine (P.A., B.D.J., K.D.), Clinical Nutrition (P.A.), Neurological Sciences (S.E.L., N.T.A., L.J.C., L.L.B., D.A.B., J.A.S.) and Pathology (S.A., J.A.S.); and Rush Institute of Healthy Aging (K.D.), Rush University Medical Center, Chicago, IL
| | - Laurel J Cherian
- From the Rush Alzheimer's Disease Center (P.A., S.E.L., N.A., B.D.J., L.L.B., D.A.B., J.A.S.), Departments of Internal Medicine (P.A., B.D.J., K.D.), Clinical Nutrition (P.A.), Neurological Sciences (S.E.L., N.T.A., L.J.C., L.L.B., D.A.B., J.A.S.) and Pathology (S.A., J.A.S.); and Rush Institute of Healthy Aging (K.D.), Rush University Medical Center, Chicago, IL
| | - Bryan D James
- From the Rush Alzheimer's Disease Center (P.A., S.E.L., N.A., B.D.J., L.L.B., D.A.B., J.A.S.), Departments of Internal Medicine (P.A., B.D.J., K.D.), Clinical Nutrition (P.A.), Neurological Sciences (S.E.L., N.T.A., L.J.C., L.L.B., D.A.B., J.A.S.) and Pathology (S.A., J.A.S.); and Rush Institute of Healthy Aging (K.D.), Rush University Medical Center, Chicago, IL
| | - Klodian Dhana
- From the Rush Alzheimer's Disease Center (P.A., S.E.L., N.A., B.D.J., L.L.B., D.A.B., J.A.S.), Departments of Internal Medicine (P.A., B.D.J., K.D.), Clinical Nutrition (P.A.), Neurological Sciences (S.E.L., N.T.A., L.J.C., L.L.B., D.A.B., J.A.S.) and Pathology (S.A., J.A.S.); and Rush Institute of Healthy Aging (K.D.), Rush University Medical Center, Chicago, IL
| | - Lisa L Barnes
- From the Rush Alzheimer's Disease Center (P.A., S.E.L., N.A., B.D.J., L.L.B., D.A.B., J.A.S.), Departments of Internal Medicine (P.A., B.D.J., K.D.), Clinical Nutrition (P.A.), Neurological Sciences (S.E.L., N.T.A., L.J.C., L.L.B., D.A.B., J.A.S.) and Pathology (S.A., J.A.S.); and Rush Institute of Healthy Aging (K.D.), Rush University Medical Center, Chicago, IL
| | - David A Bennett
- From the Rush Alzheimer's Disease Center (P.A., S.E.L., N.A., B.D.J., L.L.B., D.A.B., J.A.S.), Departments of Internal Medicine (P.A., B.D.J., K.D.), Clinical Nutrition (P.A.), Neurological Sciences (S.E.L., N.T.A., L.J.C., L.L.B., D.A.B., J.A.S.) and Pathology (S.A., J.A.S.); and Rush Institute of Healthy Aging (K.D.), Rush University Medical Center, Chicago, IL
| | - Julie A Schneider
- From the Rush Alzheimer's Disease Center (P.A., S.E.L., N.A., B.D.J., L.L.B., D.A.B., J.A.S.), Departments of Internal Medicine (P.A., B.D.J., K.D.), Clinical Nutrition (P.A.), Neurological Sciences (S.E.L., N.T.A., L.J.C., L.L.B., D.A.B., J.A.S.) and Pathology (S.A., J.A.S.); and Rush Institute of Healthy Aging (K.D.), Rush University Medical Center, Chicago, IL
| |
Collapse
|
22
|
Fraile-Ramos J, Garrit A, Reig-Vilallonga J, Giménez-Llort L. Hepatic Oxi-Inflammation and Neophobia as Potential Liver-Brain Axis Targets for Alzheimer's Disease and Aging, with Strong Sensitivity to Sex, Isolation, and Obesity. Cells 2023; 12:1517. [PMID: 37296638 PMCID: PMC10252497 DOI: 10.3390/cells12111517] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/06/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Research on Alzheimer's disease (AD) has classically focused on alterations that occur in the brain and their intra- and extracellular neuropathological hallmarks. However, the oxi-inflammation hypothesis of aging may also play a role in neuroimmunoendocrine dysregulation and the disease's pathophysiology, where the liver emerges as a target organ due to its implication in regulating metabolism and supporting the immune system. In the present work, we demonstrate organ (hepatomegaly), tissue (histopathological amyloidosis), and cellular oxidative stress (decreased glutathione peroxidase and increased glutathione reductase enzymatic activities) and inflammation (increased IL-6 and TNF𝛼) as hallmarks of hepatic dysfunction in 16-month-old male and female 3xTg-AD mice at advanced stages of the disease, and as compared to age- and sex-matched non-transgenic (NTg) counterparts. Moreover, liver-brain axis alterations were found through behavioral (increased neophobia) and HPA axis correlations that were enhanced under forced isolation. In all cases, sex (male) and isolation (naturalistic and forced) were determinants of worse hepatomegaly, oxidative stress, and inflammation progression. In addition, obesity in old male NTg mice was translated into a worse steatosis grade. Further research is underway determine whether these alterations could correlate with a worse disease prognosis and to establish potential integrative system targets for AD research.
Collapse
Affiliation(s)
- Juan Fraile-Ramos
- Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Anna Garrit
- Department of Anatomy, School of Medicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Josep Reig-Vilallonga
- Department of Anatomy, School of Medicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Lydia Giménez-Llort
- Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| |
Collapse
|
23
|
Na Y, Ke L, Jie Z, Jinping W, Tao M, Jie Z, Liu Y, Yueqin Z. Amelioration of Cholesterol Rich diet-induced Impaired Cognition in AD Transgenic Mice by an LXR Agonist TO901317 Is Associated with the Activation of the LXR-β-RXR-α-ABCA1 Transmembrane Transport System and Improving the Composition of Lipid Raft. Exp Aging Res 2023; 49:214-225. [PMID: 35792710 DOI: 10.1080/0361073x.2022.2095605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 06/26/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND It has been reported that LXR agonist can inhibit Aβ generation and alleviate Aβ-induced various adverse reactions in vivo and in vitro experiments, but the mechanisms have not been clarified. The study aimed to observe the effect of LXR agonist TO901317 on the cognitive function of AD transgenic mice fed with cholesterol-rich diet (CRD), and to explore the possible mechanism. Methods: 32 male 6-month-old double transgenic AD mice were enrolled and randomly divided into 4 groups: control (normal diet) group, CRD treatment group, TO901317 treatment group and GSK2033 treatment group. After 3 month, Morris water maze was for the changes of spatial exploration and memory ability; ELISA was for detecting the production of Aβ42 in the brain; the concentration of total cholesterol (TC), low density lipoprotein (LDL) and high density lipoprotein (HDL) in serum were detected by cholesterol enzyme colorimetry; Finally, the expression of LXR-β, RXR-α, ABCA1, caveolin-1, BACE1 and APP at protein level in the brains was measured by Western blotting. RESULTS Compared with the control group, the learning, memory ability and spatial exploration ability of the mice were more significantly serious in the CRD group (P<0.05); The contents of TC and LDL in the serum and the production of Aβ42 in the brains were significantly increased (P<0.05), but HDL was remarkably decreased (P<0.05); The protein levels of LXR-β, RXR-α and ABCA1 were also significantly decreased (P<0.05); The expression of caveolin-1, APP and BACE1 were evidently increased (P<0.05). However, after treatment with TO901317, the impaired learning and memory and spatial exploration ability of the mice were significantly improved (P<0.05); The contents of TC and LDL in serum and the production of Aβ42 in the brains were significantly decreased (P<0.05), but HLD was increased (P<0.05); The protein levels of LXR-β, RXR-α, ABCA1were all significantly increased (P<0.05), while, the expression of caveolin-1, APP and BACE1 were all significantly decreased (P<0.05). All the changes were reversed by GSK2033 (P<0.05). CONCLUSIONS TO901317 attenuated the more serious impairment of spatial exploration, learning and memory in transgenic AD mice induced by CRD, and the mechanism may be that TO901317 could activate the LXR-β/RXR-α/ABCA1 transmembrane transport system, promote the cholesterol efflux, and decreased caveolin-1, APP and BACE1, further reduce Aβ42 in the brains.
Collapse
Affiliation(s)
- Yang Na
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Lin Ke
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Zhang Jie
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Wang Jinping
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Meng Tao
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Zhu Jie
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Yang Liu
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Zhou Yueqin
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| |
Collapse
|
24
|
Azbazdar Y, Poyraz YK, Ozalp O, Nazli D, Ipekgil D, Cucun G, Ozhan G. High-fat diet feeding triggers a regenerative response in the adult zebrafish brain. Mol Neurobiol 2023; 60:2486-2506. [PMID: 36670270 DOI: 10.1007/s12035-023-03210-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/04/2023] [Indexed: 01/22/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) includes a range of liver conditions ranging from excess fat accumulation to liver failure. NAFLD is strongly associated with high-fat diet (HFD) consumption that constitutes a metabolic risk factor. While HFD has been elucidated concerning its several systemic effects, there is little information about its influence on the brain at the molecular level. Here, by using a high-fat diet (HFD)-feeding of adult zebrafish, we first reveal that excess fat uptake results in weight gain and fatty liver. Prolonged exposure to HFD induces a significant increase in the expression of pro-inflammation, apoptosis, and proliferation markers in the liver and brain tissues. Immunofluorescence analyses of the brain tissues disclose stimulation of apoptosis and widespread activation of glial cell response. Moreover, glial activation is accompanied by an initial decrease in the number of neurons and their subsequent replacement in the olfactory bulb and the telencephalon. Long-term consumption of HFD causes activation of Wnt/β-catenin signaling in the brain tissues. Finally, fish fed an HFD induces anxiety, and aggressiveness and increases locomotor activity. Thus, HFD feeding leads to a non-traumatic brain injury and stimulates a regenerative response. The activation mechanisms of a regeneration response in the brain can be exploited to fight obesity and recover from non-traumatic injuries.
Collapse
Affiliation(s)
- Yagmur Azbazdar
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, 35340, Izmir, Turkey
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, CA, 90095-1662, USA
| | - Yusuf Kaan Poyraz
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, 35340, Izmir, Turkey
| | - Ozgun Ozalp
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, 35340, Izmir, Turkey
- Department of Molecular Life Sciences, University of Zurich, CH-8057, Zurich, Switzerland
| | - Dilek Nazli
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, 35340, Izmir, Turkey
| | - Dogac Ipekgil
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, 35340, Izmir, Turkey
| | - Gokhan Cucun
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, 35340, Izmir, Turkey
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), 3640 76021, Karlsruhe, Postfach, Germany
| | - Gunes Ozhan
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340, Izmir, Turkey.
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, 35340, Izmir, Turkey.
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, 35430, Izmir, Turkey.
| |
Collapse
|
25
|
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: 17] [Impact Index Per Article: 8.5] [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
|
26
|
Espinosa-Jiménez T, Cano A, Sánchez-López E, Olloquequi J, Folch J, Bulló M, Verdaguer E, Auladell C, Pont C, Muñoz-Torrero D, Parcerisas A, Camins A, Ettcheto M. A novel rhein-huprine hybrid ameliorates disease-modifying properties in preclinical mice model of Alzheimer's disease exacerbated with high fat diet. Cell Biosci 2023; 13:52. [PMID: 36895036 PMCID: PMC9999531 DOI: 10.1186/s13578-023-01000-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 02/28/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by a polyetiological origin. Despite the global burden of AD and the advances made in AD drug research and development, the cure of the disease remains elusive, since any developed drug has demonstrated effectiveness to cure AD. Strikingly, an increasing number of studies indicate a linkage between AD and type 2 diabetes mellitus (T2DM), as both diseases share some common pathophysiological features. In fact, β-secretase (BACE1) and acetylcholinesterase (AChE), two enzymes involved in both conditions, have been considered promising targets for both pathologies. In this regard, due to the multifactorial origin of these diseases, current research efforts are focusing on the development of multi-target drugs as a very promising option to derive effective treatments for both conditions. In the present study, we evaluated the effect of rhein-huprine hybrid (RHE-HUP), a synthesized BACE1 and AChE inhibitor, both considered key factors not only in AD but also in metabolic pathologies. Thus, the aim of this study is to evaluate the effects of this compound in APP/PS1 female mice, a well-established familial AD mouse model, challenged by high-fat diet (HFD) consumption to concomitantly simulate a T2DM-like condition. RESULTS Intraperitoneal treatment with RHE-HUP in APP/PS1 mice for 4 weeks reduced the main hallmarks of AD, including Tau hyperphosphorylation, Aβ42 peptide levels and plaque formation. Moreover, we found a decreased inflammatory response together with an increase in different synaptic proteins, such as drebrin 1 (DBN1) or synaptophysin, and in neurotrophic factors, especially in BDNF levels, correlated with a recovery in the number of dendritic spines, which resulted in memory improvement. Notably, the improvement observed in this model can be attributed directly to a protein regulation at central level, since no peripheral modification of those alterations induced by HFD consumption was observed. CONCLUSIONS Our results suggest that RHE-HUP could be a new candidate for the treatment of AD, even for individuals with high risk due to peripheral metabolic disturbances, given its multi-target profile which allows for the improvement of some of the most important hallmarks of the disease.
Collapse
Affiliation(s)
- Triana Espinosa-Jiménez
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain.,Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Amanda Cano
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain.,Ace Alzheimer Center Barcelona-International University of Catalunya (UIC), Barcelona, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, Barcelona, Spain
| | - Elena Sánchez-López
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, Barcelona, Spain.,Unit of Synthesis and Biomedical Applications of Peptides, IQAC-CSIC, 08034, Barcelona, Spain
| | - Jordi Olloquequi
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain.,Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Talca, Chile
| | - Jaume Folch
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Institut d'Investigació Sanitària Pere Virgili (IISPV), 43201, Reus, Spain.,Nutrition and Metabolic Health Research Group, Institute of Health Pere Virgili-IISPV, 43201, Reus, Spain
| | - Mònica Bulló
- Institut d'Investigació Sanitària Pere Virgili (IISPV), 43201, Reus, Spain.,Nutrition and Metabolic Health Research Group, Institute of Health Pere Virgili-IISPV, 43201, Reus, Spain.,CIBER Physiology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, 28029, Madrid, Spain
| | - Ester Verdaguer
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Carme Auladell
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Caterina Pont
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain
| | - Diego Muñoz-Torrero
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain.,Institute of Biomedicine (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Antoni Parcerisas
- Department of Basic Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain.,Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain. .,Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain. .,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain. .,Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Av. Joan XXIII 27/31, 08028, Barcelona, Spain.
| |
Collapse
|
27
|
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
|
28
|
Microglia and Cholesterol Handling: Implications for Alzheimer's Disease. Biomedicines 2022; 10:biomedicines10123105. [PMID: 36551857 PMCID: PMC9775660 DOI: 10.3390/biomedicines10123105] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/04/2022] Open
Abstract
Cholesterol is essential for brain function and structure, however altered cholesterol metabolism and transport are hallmarks of multiple neurodegenerative conditions, including Alzheimer's disease (AD). The well-established link between apolipoprotein E (APOE) genotype and increased AD risk highlights the importance of cholesterol and lipid transport in AD etiology. Whereas more is known about the regulation and dysregulation of cholesterol metabolism and transport in neurons and astrocytes, less is known about how microglia, the immune cells of the brain, handle cholesterol, and the subsequent implications for the ability of microglia to perform their essential functions. Evidence is emerging that a high-cholesterol environment, particularly in the context of defects in the ability to transport cholesterol (e.g., expression of the high-risk APOE4 isoform), can lead to chronic activation, increased inflammatory signaling, and reduced phagocytic capacity, which have been associated with AD pathology. In this narrative review we describe how cholesterol regulates microglia phenotype and function, and discuss what is known about the effects of statins on microglia, as well as highlighting areas of future research to advance knowledge that can lead to the development of novel therapies for the prevention and treatment of AD.
Collapse
|
29
|
Murtaj V, Penati S, Belloli S, Foti M, Coliva A, Papagna A, Gotti C, Toninelli E, Chiaffarelli R, Mantero S, Pucci S, Matteoli M, Malosio ML, Moresco RM. Brain sex-dependent alterations after prolonged high fat diet exposure in mice. Commun Biol 2022; 5:1276. [PMID: 36414721 PMCID: PMC9681749 DOI: 10.1038/s42003-022-04214-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 11/02/2022] [Indexed: 11/23/2022] Open
Abstract
We examined effects of exposing female and male mice for 33 weeks to 45% or 60% high fat diet (HFD). Males fed with either diet were more vulnerable than females, displaying higher and faster increase in body weight and more elevated cholesterol and liver enzymes levels. Higher glucose metabolism was revealed by PET in the olfactory bulbs of both sexes. However, males also displayed altered anterior cortex and cerebellum metabolism, accompanied by a more prominent brain inflammation relative to females. Although both sexes displayed reduced transcripts of neuronal and synaptic genes in anterior cortex, only males had decreased protein levels of AMPA and NMDA receptors. Oppositely, to anterior cortex, cerebellum of HFD-exposed mice displayed hypometabolism and transcriptional up-regulation of neuronal and synaptic genes. These results indicate that male brain is more susceptible to metabolic changes induced by HFD and that the anterior cortex versus cerebellum display inverse susceptibility to HFD.
Collapse
Affiliation(s)
- Valentina Murtaj
- grid.7563.70000 0001 2174 1754PhD Program in Neuroscience, University of Milano-Bicocca, Monza (MB), Italy ,grid.18887.3e0000000417581884Department of Nuclear Medicine, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy ,grid.18887.3e0000000417581884Present Address: Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Hospital and Vita Salute San Raffaele University, Milan, Italy, 20132 Milan, Italy
| | - Silvia Penati
- Institute of Neuroscience, National Research Council of Italy (CNR) c/o Humanitas Mirasole S.p.A, Via Manzoni 56, 20089 Rozzano (MI), Italy ,grid.417728.f0000 0004 1756 8807Laboratory of Pharmacology and Brain Pathology, Neuro Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano (MI), Italy ,grid.4367.60000 0001 2355 7002Present Address: Department of Pathology and Immunology, Washington Univerisity School of Medicine, St. Louis, MO 63110 USA
| | - Sara Belloli
- grid.18887.3e0000000417581884Department of Nuclear Medicine, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy ,grid.428490.30000 0004 1789 9809Institute of Molecular Bioimaging and Physiology, CNR, 20090 Segrate (MI), Italy
| | - Maria Foti
- grid.7563.70000 0001 2174 1754Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza (MB), Italy
| | - Angela Coliva
- grid.18887.3e0000000417581884Department of Nuclear Medicine, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Angela Papagna
- grid.7563.70000 0001 2174 1754Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza (MB), Italy
| | - Cecilia Gotti
- grid.5326.20000 0001 1940 4177Institute of Neuroscience, National Research Council of Italy (CNR) c/o Università di Milano-Bicocca, Via R. Follereau 3, 20854 Vedano al Lambro (MB), Italy
| | - Elisa Toninelli
- grid.18887.3e0000000417581884Department of Nuclear Medicine, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Remy Chiaffarelli
- grid.18887.3e0000000417581884Department of Nuclear Medicine, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy ,grid.7563.70000 0001 2174 1754Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza (MB), Italy ,grid.10392.390000 0001 2190 1447Present Address: Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls Universität Tübingen, 72076 Tübingen, Germany
| | - Stefano Mantero
- grid.5326.20000 0001 1940 4177Institute for Genetic and Biomedical Research, National Research Council of Italy (CNR) c/o Humanitas Mirasole S.p.A, Via Manzoni 56, 20089 Rozzano (MI), Italy ,grid.5326.20000 0001 1940 4177Present Address: DCSR, National Research Council of Italy (CNR), Via A. Corti 12, 20133 Milan, Italy
| | - Susanna Pucci
- grid.5326.20000 0001 1940 4177Institute of Neuroscience, National Research Council of Italy (CNR) c/o Università di Milano-Bicocca, Via R. Follereau 3, 20854 Vedano al Lambro (MB), Italy
| | - Michela Matteoli
- Institute of Neuroscience, National Research Council of Italy (CNR) c/o Humanitas Mirasole S.p.A, Via Manzoni 56, 20089 Rozzano (MI), Italy ,grid.417728.f0000 0004 1756 8807Laboratory of Pharmacology and Brain Pathology, Neuro Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano (MI), Italy
| | - Maria Luisa Malosio
- Institute of Neuroscience, National Research Council of Italy (CNR) c/o Humanitas Mirasole S.p.A, Via Manzoni 56, 20089 Rozzano (MI), Italy ,grid.417728.f0000 0004 1756 8807Laboratory of Pharmacology and Brain Pathology, Neuro Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano (MI), Italy
| | - Rosa Maria Moresco
- grid.18887.3e0000000417581884Department of Nuclear Medicine, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy ,grid.428490.30000 0004 1789 9809Institute of Molecular Bioimaging and Physiology, CNR, 20090 Segrate (MI), Italy ,grid.7563.70000 0001 2174 1754Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza (MB), Italy
| |
Collapse
|
30
|
Lee D, Lee VMY, Hur SK. Manipulation of the diet-microbiota-brain axis in Alzheimer's disease. Front Neurosci 2022; 16:1042865. [PMID: 36408394 PMCID: PMC9672822 DOI: 10.3389/fnins.2022.1042865] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Abstract
Several studies investigating the pathogenesis of Alzheimer's disease have identified various interdependent constituents contributing to the exacerbation of the disease, including Aβ plaque formation, tau protein hyperphosphorylation, neurofibrillary tangle accumulation, glial inflammation, and the eventual loss of proper neural plasticity. Recently, using various models and human patients, another key factor has been established as an influential determinant in brain homeostasis: the gut-brain axis. The implications of a rapidly aging population and the absence of a definitive cure for Alzheimer's disease have prompted a search for non-pharmaceutical tools, of which gut-modulatory therapies targeting the gut-brain axis have shown promise. Yet multiple recent studies examining changes in human gut flora in response to various probiotics and environmental factors are limited and difficult to generalize; whether the state of the gut microbiota in Alzheimer's disease is a cause of the disease, a result of the disease, or both through numerous feedback loops in the gut-brain axis, remains unclear. However, preliminary findings of longitudinal studies conducted over the past decades have highlighted dietary interventions, especially Mediterranean diets, as preventative measures for Alzheimer's disease by reversing neuroinflammation, modifying the intestinal and blood-brain barrier (BBB), and addressing gut dysbiosis. Conversely, the consumption of Western diets intensifies the progression of Alzheimer's disease through genetic alterations, impaired barrier function, and chronic inflammation. This review aims to support the growing body of experimental and clinical data highlighting specific probiotic strains and particular dietary components in preventing Alzheimer's disease via the gut-brain axis.
Collapse
Affiliation(s)
- Daniel Lee
- Middleton High School, Middleton, WI, United States
| | - Virginia M-Y. Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Seong Kwon Hur
- Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA, United States
| |
Collapse
|
31
|
Dietary vitamin B6 restriction aggravates neurodegeneration in mice fed a high-fat diet. Life Sci 2022; 309:121041. [DOI: 10.1016/j.lfs.2022.121041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 09/26/2022] [Accepted: 10/01/2022] [Indexed: 11/18/2022]
|
32
|
Huang Z, Lin HW(K, Zhang Q, Zong X. Targeting Alzheimer's Disease: The Critical Crosstalk between the Liver and Brain. Nutrients 2022; 14:nu14204298. [PMID: 36296980 PMCID: PMC9609624 DOI: 10.3390/nu14204298] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 01/24/2023] Open
Abstract
Alzheimer's disease (AD), an age-related neurodegenerative disorder, is currently incurable. Imbalanced amyloid-beta (Aβ) generation and clearance are thought to play a pivotal role in the pathogenesis of AD. Historically, strategies targeting Aβ clearance have typically focused on central clearance, but with limited clinical success. Recently, the contribution of peripheral systems, particularly the liver, to Aβ clearance has sparked an increased interest. In addition, AD presents pathological features similar to those of metabolic syndrome, and the critical involvement of brain energy metabolic disturbances in this disease has been recognized. More importantly, the liver may be a key regulator in these abnormalities, far beyond our past understanding. Here, we review recent animal and clinical findings indicating that liver dysfunction represents an early event in AD pathophysiology. We further propose that compromised peripheral Aβ clearance by the liver and aberrant hepatic physiological processes may contribute to AD neurodegeneration. The role of a hepatic synthesis product, fibroblast growth factor 21 (FGF21), in the management of AD is also discussed. A deeper understanding of the communication between the liver and brain may lead to new opportunities for the early diagnosis and treatment of AD.
Collapse
|
33
|
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
|
34
|
Rudajev V, Novotny J. Cholesterol as a key player in amyloid β-mediated toxicity in Alzheimer’s disease. Front Mol Neurosci 2022; 15:937056. [PMID: 36090253 PMCID: PMC9453481 DOI: 10.3389/fnmol.2022.937056] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder that is one of the most devastating and widespread diseases worldwide, mainly affecting the aging population. One of the key factors contributing to AD-related neurotoxicity is the production and aggregation of amyloid β (Aβ). Many studies have shown the ability of Aβ to bind to the cell membrane and disrupt its structure, leading to cell death. Because amyloid damage affects different parts of the brain differently, it seems likely that not only Aβ but also the nature of the membrane interface with which the amyloid interacts, helps determine the final neurotoxic effect. Because cholesterol is the dominant component of the plasma membrane, it plays an important role in Aβ-induced toxicity. Elevated cholesterol levels and their regulation by statins have been shown to be important factors influencing the progression of neurodegeneration. However, data from many studies have shown that cholesterol has both neuroprotective and aggravating effects in relation to the development of AD. In this review, we attempt to summarize recent findings on the role of cholesterol in Aβ toxicity mediated by membrane binding in the pathogenesis of AD and to consider it in the broader context of the lipid composition of cell membranes.
Collapse
|
35
|
Fanalli SL, da Silva BPM, Gomes JD, Ciconello FN, de Almeida VV, Freitas FAO, Moreira GCM, Silva-Vignato B, Afonso J, Reecy J, Koltes J, Koltes D, Regitano LCA, de Carvalho Baileiro JC, Freitas L, Coutinho LL, Fukumasu H, de Alencar SM, Luchiari Filho A, Cesar ASM. Effect of dietary soybean oil inclusion on liver-related transcription factors in a pig model for metabolic diseases. Sci Rep 2022; 12:10318. [PMID: 35725871 PMCID: PMC9209463 DOI: 10.1038/s41598-022-14069-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 05/31/2022] [Indexed: 12/21/2022] Open
Abstract
Dietary fatty acids (FA) are components of the lipids, which contribute to membrane structure, energy input, and biological functions related to cellular signaling and transcriptome regulation. However, the consumers still associate dietary FA with fat deposition and increased occurrence of metabolic diseases such as obesity and atherosclerosis. Previous studies already demonstrated that some fatty acids are linked with inflammatory response, preventing metabolic diseases. To better understand the role of dietary FA on metabolic diseases, for the first time, a study to identify key transcription factors (TF) involved in lipid metabolism and inflammatory response by transcriptome analysis from liver samples of animal models was performed. The key TF were identified by functional enrichment analysis from the list of differentially expressed genes identified in liver samples between 35 pigs fed with 1.5% or 3.0% soybean oil. The functional enrichment analysis detected TF linked to lipid homeostasis and inflammatory response, such as RXRA, EGFR, and SREBP2 precursor. These findings demonstrated that key TF related to lipid metabolism could be modulated by dietary inclusion of soybean oil. It could contribute to nutrigenomics research field that aims to elucidate dietary interventions in animal and human health, as well as to drive food technology and science.
Collapse
Affiliation(s)
- Simara Larissa Fanalli
- Faculty of Animal Science and Food Engineering, University of São Paulo, Campus Fernando Costa, Avenue Duque de Caxias Norte 225, Pirassununga, São Paulo, 13635-900, Brazil
| | - Bruna Pereira Martins da Silva
- Faculty of Animal Science and Food Engineering, University of São Paulo, Campus Fernando Costa, Avenue Duque de Caxias Norte 225, Pirassununga, São Paulo, 13635-900, Brazil
| | - Julia Dezen Gomes
- Luiz de Queiroz College of Agriculture, University of São Paulo, Avenue Pádua Dias 11, Piracicaba, São Paulo, 13418-900, Brazil
| | - Fernanda Nery Ciconello
- Luiz de Queiroz College of Agriculture, University of São Paulo, Avenue Pádua Dias 11, Piracicaba, São Paulo, 13418-900, Brazil
| | - Vivian Vezzoni de Almeida
- College of Veterinary Medicine and Animal Science, Federal University of Goiás, Nova Veneza, km 8, Campus Samambaia, Goiânia, Goiás, 74690-900, Brazil
| | - Felipe André Oliveira Freitas
- Luiz de Queiroz College of Agriculture, University of São Paulo, Avenue Pádua Dias 11, Piracicaba, São Paulo, 13418-900, Brazil
| | - Gabriel Costa Monteiro Moreira
- University of Liège, GIGA Medical Genomics, Unit of Animal Genomics, Quartier Hôpital, Avenue de l'Hôpital, 11, 4000, Liège, Belgium
| | - Bárbara Silva-Vignato
- Luiz de Queiroz College of Agriculture, University of São Paulo, Avenue Pádua Dias 11, Piracicaba, São Paulo, 13418-900, Brazil
| | - Juliana Afonso
- Embrapa Pecuária Sudeste, Km 234 s/nº, São Carlos, São Paulo, 13560-970, Brazil
| | - James Reecy
- Department of Animal Science, College of Agriculture and Life Sciences, Iowa State University, 1221, Kildee Hall, Ames, IA, 50011-3150, USA
| | - James Koltes
- Department of Animal Science, College of Agriculture and Life Sciences, Iowa State University, 1221, Kildee Hall, Ames, IA, 50011-3150, USA
| | - Dawn Koltes
- Department of Animal Science, College of Agriculture and Life Sciences, Iowa State University, 1221, Kildee Hall, Ames, IA, 50011-3150, USA
| | | | - Júlio Cesar de Carvalho Baileiro
- College of Veterinary Medicine and Animal Science, University of São Paulo, Duque de Caxias Norte, 225, Pirassununga, São Paulo, 13.635-900, Brazil
| | - Luciana Freitas
- DB Genética de Suínos, Avenue Juscelino Kubitschek de Oliveira, 2094, Patos de Minas, MG, 38.706-000, Brazil
| | - Luiz Lehmann Coutinho
- Luiz de Queiroz College of Agriculture, University of São Paulo, Avenue Pádua Dias 11, Piracicaba, São Paulo, 13418-900, Brazil
| | - Heidge Fukumasu
- Faculty of Animal Science and Food Engineering, University of São Paulo, Campus Fernando Costa, Avenue Duque de Caxias Norte 225, Pirassununga, São Paulo, 13635-900, Brazil
| | - Severino Matias de Alencar
- Luiz de Queiroz College of Agriculture, University of São Paulo, Avenue Pádua Dias 11, Piracicaba, São Paulo, 13418-900, Brazil
| | - Albino Luchiari Filho
- Luiz de Queiroz College of Agriculture, University of São Paulo, Avenue Pádua Dias 11, Piracicaba, São Paulo, 13418-900, Brazil
| | - Aline Silva Mello Cesar
- Faculty of Animal Science and Food Engineering, University of São Paulo, Campus Fernando Costa, Avenue Duque de Caxias Norte 225, Pirassununga, São Paulo, 13635-900, Brazil. .,Luiz de Queiroz College of Agriculture, University of São Paulo, Avenue Pádua Dias 11, Piracicaba, São Paulo, 13418-900, Brazil.
| |
Collapse
|
36
|
Vegas-Suárez S, Simón J, Martínez-Chantar ML, Moratalla R. Metabolic Diffusion in Neuropathologies: The Relevance of Brain-Liver Axis. Front Physiol 2022; 13:864263. [PMID: 35634148 PMCID: PMC9134112 DOI: 10.3389/fphys.2022.864263] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/25/2022] [Indexed: 11/10/2022] Open
Abstract
Chronic liver diseases include a broad group of hepatic disorders from different etiologies and with varying degrees of progression and severity. Among them, non-alcoholic fatty (NAFLD) and alcoholic (ALD) liver diseases are the most frequent forms of expression, caused by either metabolic alterations or chronic alcohol consumption. The liver is the main regulator of energy homeostasis and metabolism of potentially toxic compounds in the organism, thus hepatic disorders often promote the release of harmful substances. In this context, there is an existing interconnection between liver and brain, with the well-named brain-liver axis, in which liver pathologies lead to the promotion of neurodegenerative disorders. Alzheimer's (AD) and Parkinson's (PD) diseases are the most relevant neurological disorders worldwide. The present work highlights the relevance of the liver-related promotion of these disorders. Liver-related hyperammonemia has been related to the promotion of perturbations in nervous systems, whereas the production of ketone bodies under certain conditions may protect from developing them. The capacity of the liver of amyloid-β (Aβ) clearance is reduced under liver pathologies, contributing to the development of AD. These perturbations are even aggravated by the pro-inflammatory state that often accompanies liver diseases, leading to the named neuroinflammation. The current nourishment habits, named as Western diet (WD) and alterations in the bile acid (BA) profile, whose homeostasis is controlled by the liver, have been also related to both AD and PD, whereas the supplementation with certain compounds, has been demonstrated to alleviate the pathologies.
Collapse
Affiliation(s)
- Sergio Vegas-Suárez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain,Cajal Institute, Spanish National Research Council (CSIC), Madrid, Spain,Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERned), Carlos III Institute of Health (ISCIII), Madrid, Spain
| | - Jorge Simón
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III Institute of Health (ISCIII), Madrid, Spain
| | - María Luz Martínez-Chantar
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III Institute of Health (ISCIII), Madrid, Spain,*Correspondence: María Luz Martínez-Chantar, ; Rosario Moratalla,
| | - Rosario Moratalla
- Cajal Institute, Spanish National Research Council (CSIC), Madrid, Spain,Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERned), Carlos III Institute of Health (ISCIII), Madrid, Spain,*Correspondence: María Luz Martínez-Chantar, ; Rosario Moratalla,
| |
Collapse
|
37
|
Vila-Castelar C, Fox-Fuller JT, Guzmán-Vélez E, Schoemaker D, Quiroz YT. A cultural approach to dementia - insights from US Latino and other minoritized groups. Nat Rev Neurol 2022; 18:307-314. [PMID: 35260817 PMCID: PMC9113534 DOI: 10.1038/s41582-022-00630-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2022] [Indexed: 12/14/2022]
Abstract
Alzheimer disease and related dementias present considerable challenges to health-care and medical systems worldwide. In the USA, older Black and Latino individuals are more likely than older white individuals to have Alzheimer disease and related dementias. In this Perspective, we leverage our experience and expertise with older US Latino groups to review and discuss the need to integrate cultural factors into dementia research and care. We examine the importance of considering the effects of cultural factors on clinical presentation and diagnosis, dementia risk, clinical research and recruitment, and caregiving practices, with a focus on minoritized groups in the USA. We highlight critical gaps in the literature to stimulate future research aimed at improving the prevention and early detection of Alzheimer disease and related dementias and developing novel treatments and interventions across ethnoracially diverse populations. In addition, we briefly discuss some of our own initiatives to promote research and clinical care among Latino populations living in the USA.
Collapse
Affiliation(s)
- Clara Vila-Castelar
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Joshua T Fox-Fuller
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, USA
| | - Edmarie Guzmán-Vélez
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Dorothee Schoemaker
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yakeel T Quiroz
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
38
|
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
|
39
|
Sex Differences in Metabolic Indices and Chronic Neuroinflammation in Response to Prolonged High-Fat Diet in ApoE4 Knock-In Mice. Int J Mol Sci 2022; 23:ijms23073921. [PMID: 35409283 PMCID: PMC8999114 DOI: 10.3390/ijms23073921] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/22/2022] [Accepted: 03/30/2022] [Indexed: 12/15/2022] Open
Abstract
Late-onset Alzheimer’s disease (LOAD) likely results from combinations of risk factors that include both genetic predisposition and modifiable lifestyle factors. The E4 allele of apolipoprotein E (ApoE) is the most significant genetic risk factor for LOAD. A Western-pattern diet (WD) has been shown to strongly increase the risk of cardiovascular disease and diabetes, conditions which have been strongly linked to an increased risk for developing AD. Little is known about how the WD may contribute to, or enhance, the increased risk presented by possession of the ApoE4 allele. To model this interaction over the course of a lifetime, we exposed male and female homozygote ApoE4 knock-in mice and wild-type controls to nine months of a high-fat WD or standard chow diet. At eleven months of age, the mice were tested for glucose tolerance and then for general activity and spatial learning and memory. Postmortem analysis of liver function and neuroinflammation in the brain was also assessed. Our results suggest that behavior impairments resulted from the convergence of interacting metabolic alterations, made worse in a male ApoE4 mice group who also showed liver dysfunction, leading to a higher level of inflammatory cytokines in the brain. Interestingly, female ApoE4 mice on a WD revealed impairments in spatial learning and memory without the observed liver dysfunction or increase in inflammatory markers in the brain. These results suggest multiple direct and indirect pathways through which ApoE and diet-related factors interact. The striking sex difference in markers of chronic neuroinflammation in male ApoE4 mice fed the high-fat WD suggests a specific mechanism of interaction conferring significant enhanced LOAD risk for humans with the ApoE4 allele, which may differ between sexes. Additionally, our results suggest researchers exercise caution when designing and interpreting results of experiments employing a WD, being careful not to assume a WD impacts both sexes by the same mechanisms.
Collapse
|
40
|
Guzzardi MA, La Rosa F, Campani D, Collado MC, Monleon D, Cacciato Insilla A, Tripodi M, Zega A, Dattilo A, Brunetto MR, Maffei M, Bonino F, Iozzo P. Liver and White/Brown Fat Dystrophy Associates with Gut Microbiota and Metabolomic Alterations in 3xTg Alzheimer’s Disease Mouse Model. Metabolites 2022; 12:metabo12040278. [PMID: 35448465 PMCID: PMC9028874 DOI: 10.3390/metabo12040278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 02/04/2023] Open
Abstract
Metabolic impairments and liver and adipose depots alterations were reported in subjects with Alzheimer’s disease (AD), highlighting the role of the liver–adipose–tissue–brain axis in AD pathophysiology. The gut microbiota might play a modulating role. We investigated the alterations to the liver and white/brown adipose tissues (W/BAT) and their relationships with serum and gut metabolites and gut bacteria in a 3xTg mouse model during AD onset (adulthood) and progression (aging) and the impact of high-fat diet (HFD) and intranasal insulin (INI). Glucose metabolism (18FDG-PET), tissue radiodensity (CT), liver and W/BAT histology, BAT-thermogenic markers were analyzed. 16S-RNA sequencing and mass-spectrometry were performed in adult (8 months) and aged (14 months) 3xTg-AD mice with a high-fat or control diet. Generalized and HFD resistant deficiency of lipid accumulation in both liver and W/BAT, hypermetabolism in WAT (adulthood) and BAT (aging), abnormal cytokine–hormone profiles, and liver inflammation were observed in 3xTg mice; INI could antagonize all these alterations. Specific gut microbiota–metabolome profiles correlated with a significant disruption of the gut–microbiota–liver–adipose axis in AD mice. In conclusion, fat dystrophy in liver and adipose depots contributes to AD progression, and associates with altered profiles of the gut microbiota, which candidates as an appealing early target for preventive intervention.
Collapse
Affiliation(s)
- Maria Angela Guzzardi
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy; (F.L.R.); (M.T.); (A.Z.); (M.M.); (P.I.)
- Correspondence: ; Tel.: +39-050-3152722
| | - Federica La Rosa
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy; (F.L.R.); (M.T.); (A.Z.); (M.M.); (P.I.)
| | - Daniela Campani
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, Division of Pathology, Pisa University Hospital, 56124 Pisa, Italy; (D.C.); (A.C.I.)
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), 46980 Valencia, Spain;
| | - Daniel Monleon
- Faculty of Medicine, Health Research Institute INCLIVA/CIBERFES for Frailty and Healthy Aging, University of Valencia, 46003 Valencia, Spain;
| | - Andrea Cacciato Insilla
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, Division of Pathology, Pisa University Hospital, 56124 Pisa, Italy; (D.C.); (A.C.I.)
| | - Maria Tripodi
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy; (F.L.R.); (M.T.); (A.Z.); (M.M.); (P.I.)
| | - Alessandro Zega
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy; (F.L.R.); (M.T.); (A.Z.); (M.M.); (P.I.)
| | | | - Maurizia Rossana Brunetto
- Department of Clinical and Experimental Medicine, University of Pisa, 56124 Pisa, Italy;
- Hepatology Unit, Department of Medical Specialties, Laboratory of Molecular Genetics and Pathology of Hepatitis Viruses, Pisa University Hospital, 56124 Pisa, Italy
- Institute of Biostructure and Bioimaging (IBB), National Research Council (CNR), 80145 Napoli, Italy;
| | - Margherita Maffei
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy; (F.L.R.); (M.T.); (A.Z.); (M.M.); (P.I.)
| | - Ferruccio Bonino
- Institute of Biostructure and Bioimaging (IBB), National Research Council (CNR), 80145 Napoli, Italy;
| | - Patricia Iozzo
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy; (F.L.R.); (M.T.); (A.Z.); (M.M.); (P.I.)
| |
Collapse
|
41
|
Confettura AD, Cuboni E, Ammar MR, Jia S, Gomes GM, Yuanxiang P, Raman R, Li T, Grochowska KM, Ahrends R, Karpova A, Dityatev A, Kreutz MR. Neddylation-dependent protein degradation is a nexus between synaptic insulin resistance, neuroinflammation and Alzheimer's disease. Transl Neurodegener 2022; 11:2. [PMID: 34986876 PMCID: PMC8734066 DOI: 10.1186/s40035-021-00277-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/24/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The metabolic syndrome is a consequence of modern lifestyle that causes synaptic insulin resistance and cognitive deficits and that in interaction with a high amyloid load is an important risk factor for Alzheimer's disease. It has been proposed that neuroinflammation might be an intervening variable, but the underlying mechanisms are currently unknown. METHODS We utilized primary neurons to induce synaptic insulin resistance as well as a mouse model of high-risk aging that includes a high amyloid load, neuroinflammation, and diet-induced obesity to test hypotheses on underlying mechanisms. RESULTS We found that neddylation and subsequent activation of cullin-RING ligase complexes induced synaptic insulin resistance through ubiquitylation and degradation of the insulin-receptor substrate IRS1 that organizes synaptic insulin signaling. Accordingly, inhibition of neddylation preserved synaptic insulin signaling and rescued memory deficits in mice with a high amyloid load, which were fed with a 'western diet'. CONCLUSIONS Collectively, the data suggest that neddylation and degradation of the insulin-receptor substrate is a nodal point that links high amyloid load, neuroinflammation, and synaptic insulin resistance to cognitive decline and impaired synaptic plasticity in high-risk aging.
Collapse
Affiliation(s)
| | - Eleonora Cuboni
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Mohamed Rafeet Ammar
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Shaobo Jia
- German Center for Neurodegenerative Diseases (DZNE), 39120, Magdeburg, Germany
| | - Guilherme M Gomes
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, 39118, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Otto Von Guericke University, 39120, Magdeburg, Germany
| | - PingAn Yuanxiang
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Rajeev Raman
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Tingting Li
- Leibniz-Institut Für Analytische Wissenschaften-ISAS-e.V., 44227, Dortmund, Germany
| | - Katarzyna M Grochowska
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, 39118, Magdeburg, Germany.,Leibniz Group 'Dendritic Organelles and Synaptic Function', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Robert Ahrends
- Leibniz-Institut Für Analytische Wissenschaften-ISAS-e.V., 44227, Dortmund, Germany.,Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090, Wien, Austria
| | - Anna Karpova
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, 39118, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Otto Von Guericke University, 39120, Magdeburg, Germany
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE), 39120, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Otto Von Guericke University, 39120, Magdeburg, Germany.,Medical Faculty, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Michael R Kreutz
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, 39118, Magdeburg, Germany. .,German Center for Neurodegenerative Diseases (DZNE), 39120, Magdeburg, Germany. .,Center for Behavioral Brain Sciences, Otto Von Guericke University, 39120, Magdeburg, Germany. .,Leibniz Group 'Dendritic Organelles and Synaptic Function', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
| |
Collapse
|
42
|
Litvinenko IV, Lobzin VY. On a New Paradigm of the Development of Neurodegenerative Diseases by the Example of Alzheimer’s Disease and Parkinson’s Disease. ADVANCES IN GERONTOLOGY 2022; 12. [PMCID: PMC9774074 DOI: 10.1134/s2079057022040117] [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: 12/24/2022]
Abstract
The role of neuronal inflammation developing during the formation of amyloid plaques and Lewy bodies is investigated. The influence of various exogenous and endogenous factors on the development of neuroinflammation is established, but the role of various infectious agents in the development of this process is much less studied. Today, the existence of a universal trigger mechanism of the neurodegenerative process is obvious: a specific pathogen of a bacterial or viral nature (including long-term persistent in nervous tissue in a latent state), reactivating, penetrates into certain cerebral structures, where it is influenced by either Aβ or resident macrophages of the central nervous system, which, in turn, are activated and induce the release of proinflammatory cytokines, leading to the development of neuronal inflammation, autophagy and neurodegeneration. The reactivation of latent infection, such as herpes, in APOE4 carriers significantly increases the risk of development of Alzheimer’s disease. Class-II genes of the HLA locus (HLA II) may be related to the progression of neurodegenerative diseases. An increase in iron levels in the glia is induced by inflammation, which leads to neurodegeneration. Disruption of the homeostasis of redox-active metals, iron and copper, is an integral part of the pathogenesis of Alzheimer’s disease and Parkinson’s disease. The developing neuroinflammation leads to intensification of the processes of peroxidation, oxidation of metals and the development of ferroptosis.
Collapse
Affiliation(s)
| | - V. Yu. Lobzin
- Kirov Military Medical Academy, 194044 St. Petersburg, Russia ,Mechnikov North-Western State Medical University, 191015 St. Petersburg, Russia ,Children’s Research and Clinical Center of Infectious Diseases, 197022 St. Petersburg, Russia
| |
Collapse
|
43
|
Dominguez LJ, Veronese N, Vernuccio L, Catanese G, Inzerillo F, Salemi G, Barbagallo M. Nutrition, Physical Activity, and Other Lifestyle Factors in the Prevention of Cognitive Decline and Dementia. Nutrients 2021; 13:nu13114080. [PMID: 34836334 PMCID: PMC8624903 DOI: 10.3390/nu13114080] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 02/07/2023] Open
Abstract
Multiple factors combined are currently recognized as contributors to cognitive decline. The main independent risk factor for cognitive impairment and dementia is advanced age followed by other determinants such as genetic, socioeconomic, and environmental factors, including nutrition and physical activity. In the next decades, a rise in dementia cases is expected due largely to the aging of the world population. There are no hitherto effective pharmaceutical therapies to treat age-associated cognitive impairment and dementia, which underscores the crucial role of prevention. A relationship among diet, physical activity, and other lifestyle factors with cognitive function has been intensively studied with mounting evidence supporting the role of these determinants in the development of cognitive decline and dementia, which is a chief cause of disability globally. Several dietary patterns, foods, and nutrients have been investigated in this regard, with some encouraging and other disappointing results. This review presents the current evidence for the effects of dietary patterns, dietary components, some supplements, physical activity, sleep patterns, and social engagement on the prevention or delay of the onset of age-related cognitive decline and dementia.
Collapse
Affiliation(s)
- Ligia J. Dominguez
- Geriatric Unit, Department of Medicine, University of Palermo, 90100 Palermo, Italy; (N.V.); (L.V.); (G.C.); (F.I.); (M.B.)
- Faculty of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy
- Correspondence: ; +39-0916554828
| | - Nicola Veronese
- Geriatric Unit, Department of Medicine, University of Palermo, 90100 Palermo, Italy; (N.V.); (L.V.); (G.C.); (F.I.); (M.B.)
| | - Laura Vernuccio
- Geriatric Unit, Department of Medicine, University of Palermo, 90100 Palermo, Italy; (N.V.); (L.V.); (G.C.); (F.I.); (M.B.)
| | - Giuseppina Catanese
- Geriatric Unit, Department of Medicine, University of Palermo, 90100 Palermo, Italy; (N.V.); (L.V.); (G.C.); (F.I.); (M.B.)
| | - Flora Inzerillo
- Geriatric Unit, Department of Medicine, University of Palermo, 90100 Palermo, Italy; (N.V.); (L.V.); (G.C.); (F.I.); (M.B.)
| | - Giuseppe Salemi
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics, University of Palermo, 90100 Palermo, Italy;
- UOC of Neurology, University Hospital “Paolo Giaccone”, 90100 Palermo, Italy
| | - Mario Barbagallo
- Geriatric Unit, Department of Medicine, University of Palermo, 90100 Palermo, Italy; (N.V.); (L.V.); (G.C.); (F.I.); (M.B.)
| |
Collapse
|
44
|
Inhibition of Soluble Epoxide Hydrolase Is Protective against the Multiomic Effects of a High Glycemic Diet on Brain Microvascular Inflammation and Cognitive Dysfunction. Nutrients 2021; 13:nu13113913. [PMID: 34836168 PMCID: PMC8622784 DOI: 10.3390/nu13113913] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 12/22/2022] Open
Abstract
Diet is a modifiable risk factor for cardiovascular disease (CVD) and dementia, yet relatively little is known about the effect of a high glycemic diet (HGD) on the brain’s microvasculature. The objective of our study was to determine the molecular effects of an HGD on hippocampal microvessels and cognitive function and determine if a soluble epoxide hydrolase (sEH) inhibitor (sEHI), known to be vasculoprotective and anti-inflammatory, modulates these effects. Wild type male mice were fed a low glycemic diet (LGD, 12% sucrose/weight) or an HGD (34% sucrose/weight) with/without the sEHI, trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid (t-AUCB), for 12 weeks. Brain hippocampal microvascular gene expression was assessed by microarray and data analyzed using a multi-omic approach for differential expression of protein and non-protein-coding genes, gene networks, functional pathways, and transcription factors. Global hippocampal microvascular gene expression was fundamentally different for mice fed the HGD vs. the LGD. The HGD response was characterized by differential expression of 608 genes involved in cell signaling, neurodegeneration, metabolism, and cell adhesion/inflammation/oxidation effects reversible by t-AUCB and hence sEH inhibitor correlated with protection against Alzheimer’s dementia. Ours is the first study to demonstrate that high dietary glycemia contributes to brain hippocampal microvascular inflammation through sEH.
Collapse
|
45
|
Cerebrovascular alterations in NAFLD: Is it increasing our risk of Alzheimer's disease? Anal Biochem 2021; 636:114387. [PMID: 34537182 DOI: 10.1016/j.ab.2021.114387] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/27/2021] [Accepted: 09/15/2021] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multisystem disease, which has been classified as an emerging epidemic not only confined to liver-related morbidity and mortality. It is also becoming apparent that NAFLD is associated with moderate cerebral dysfunction and cognitive decline. A possible link between NAFLD and Alzheimer's disease (AD) has only recently been proposed due to the multiple shared genes and pathological mechanisms contributing to the development of these conditions. Although AD is a progressive neurodegenerative disease, the exact pathophysiological mechanism remains ambiguous and similarly to NAFLD, currently available pharmacological therapies have mostly failed in clinical trials. In addition to the usual suspects (inflammation, oxidative stress, blood-brain barrier alterations and ageing) that could contribute to the NAFLD-induced development and progression of AD, changes in the vasculature, cerebral perfusion and waste clearance could be the missing link between these two diseases. Here, we review the most recent literature linking NAFLD and AD, focusing on cerebrovascular alterations and the brain's clearance system as risk factors involved in the development and progression of AD, with the aim of promoting further research using neuroimaging techniques and new mechanism-based therapeutic interventions.
Collapse
|
46
|
Więckowska-Gacek A, Mietelska-Porowska A, Wydrych M, Wojda U. Western diet as a trigger of Alzheimer's disease: From metabolic syndrome and systemic inflammation to neuroinflammation and neurodegeneration. Ageing Res Rev 2021; 70:101397. [PMID: 34214643 DOI: 10.1016/j.arr.2021.101397] [Citation(s) in RCA: 156] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/10/2021] [Accepted: 06/24/2021] [Indexed: 02/06/2023]
Abstract
An excess of saturated fatty acids and simple sugars in the diet is a known environmental risk factor of Alzheimer's disease (AD) but the holistic view of the interacting processes through which such diet may contribute to AD pathogenesis is missing. We addressed this need through extensive analysis of published studies investigating the effects of western diet (WD) on AD development in humans and laboratory animals. We reviewed WD-induced systemic alterations comprising metabolic changes, induction of obesity and adipose tissue inflammation, gut microbiota dysbiosis and acceleration of systemic low-grade inflammation. Next we provide an overview of the evidence demonstrating that WD-associated systemic alterations drive impairment of the blood-brain barrier (BBB) and development of neuroinflammation paralleled by accumulation of toxic amyloid. Later these changes are followed by dysfunction of synaptic transmission, neurodegeneration and finally memory and cognitive impairment. We conclude that WD can trigger AD by acceleration of inflammaging, and that BBB impairment induced by metabolic and systemic inflammation play the central role in this process. Moreover, the concurrence of neuroinflammation and Aβ dyshomeostasis, which by reciprocal interactions drive the vicious cycle of neurodegeneration, contradicts Aβ as the primary trigger of AD. Given that in 2019 the World Health Organization recommended focusing on modifiable risk factors in AD prevention, this overview of the sequential, complex pathomechanisms initiated by WD, which can lead from peripheral disturbances to neurodegeneration, can support future prevention strategies.
Collapse
|
47
|
Duplantier SC, Gardner CD. A Critical Review of the Study of Neuroprotective Diets to Reduce Cognitive Decline. Nutrients 2021; 13:nu13072264. [PMID: 34208980 PMCID: PMC8308213 DOI: 10.3390/nu13072264] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/25/2021] [Accepted: 06/25/2021] [Indexed: 12/18/2022] Open
Abstract
Alzheimer’s disease (AD) and other dementias are now the seventh leading cause of death in the world and are projected to affect 115.4 million people by 2050. Delaying the onset of AD by just five years is estimated to reduce the cost and prevalence of the disease by half. There is no cure for AD nor any drug therapies to halt its progression once the disease begins. Lifestyle choices including diet are being seen as a viable complementary therapy to reduce cognitive decline, the hallmark of AD. Mediterranean, DASH (Dietary Approaches to Stop Hypertension), and MIND (Mediterranean-DASH Intervention for Neurodegenerative Delay) diets have biological mechanisms supporting their potential neuroprotective benefits, but the findings of study outcomes about these benefits have been inconsistent. This paper analyzed five Randomized Clinical Trials (RCTs) (from 2000 to 2021) and 27 observational studies (from 2010 to 2021) focused on the link between cognitive health and the Mediterranean/DASH/MIND diets to identify gaps and challenges that could lead to inconsistent results. These include a lack of accuracy in assessing food intake, multiple dietary pattern scoring systems, a shifting metric among studies focused on the Mediterranean diet, a lack of standards in the tools used to assess cognitive decline, and studies that were underpowered or had follow-up periods too short to detect cognitive change. Insights from these gaps and challenges are summarized in recommendations for future RCTs, including both pragmatic and explanatory RCTs.
Collapse
Affiliation(s)
- Sally C. Duplantier
- The USC Leonard School of Gerontology, University of Southern California, 3715 McClintock Ave., Los Angeles, CA 90089, USA;
| | - Christopher D. Gardner
- Stanford Prevention Research Center, Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA
- Correspondence:
| |
Collapse
|