1
|
Acun AD, Kantar D. Modulation of oxidative stress and apoptosis by alteration of bioactive lipids in the pancreas, and effect of zinc chelation in a rat model of Alzheimer's disease. J Trace Elem Med Biol 2024; 85:127480. [PMID: 38875759 DOI: 10.1016/j.jtemb.2024.127480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/16/2024]
Abstract
INTRODUCTION Increasing epidemiological evidence highlights the association between systemic insulin resistance and Alzheimer's disease (AD). It is known that peripheral insulin resistance in the early stages of AD precedes and is a precursor to amyloid-β (Aβ) deposition. Although it is known that improving the CNS insulin sensitivity of AD patients is an important therapeutic goal and that the majority of insulin in the brain comes from the periphery, there has been little attention to the changes that occur in the pancreatic tissue of AD patients. Therefore, it is crucial to elucidate the mechanisms affecting insulin resistance in pancreatic tissue in AD. It is known that zinc (Zn2+) chelation is effective in reducing peripheral insulin resistance, cell apoptosis, cell death, and oxidative stress. OBJECTIVE It was aimed to determine the changes in bioactive lipids, amylin (AIPP), oxidative stress and apoptosis in pancreatic cells in the early stages of Alzheimer's disease. The main aim is to reveal the therapeutic effect of the Cyclo-Z agent on these changes seen in the pancreas due to AD disease. METHODS AD and ADC rats were intracerebroventricular (i.c.v.) Aβ1-42 oligomers. Cyclo-Z gavage was applied to ADC and SHC rats for 21 days. First of all, the effects of AIPP, bioactive ceramides, apoptosis and oxidative stress on the pancreatic tissue of AD group rats were evaluated. Then, the effect of Cyclo-Z treatment on these was examined. ELISA kit was used in biochemical analyses. RESULTS AIPP and ceramide (CER) levels and CER/ sphingosine-1 phosphate (S1P) ratio were increased in the pancreatic tissue of AD rats. It also increased the level of CER kinase (CERK), which is known to increase the concentration of CER 1-phosphate (C1P), which is known to be toxic to cells in the presence of excessive CER concentration. Due to the increase in CER level, it was observed that apoptosis and oxidative stress increased in the pancreatic cells of AD group rats. CONCLUSION Cyclo-Z, which has Zn2+ chelating properties, reduced AD model rats' AIPP level and oxidative stress and could prevent pancreatic apoptosis. Similar therapeutic effects were not observed in the pancreatic tissue of Cyclo-Z administered to the SH group. For this reason, it is thought that Cyclo-Z agent may have a therapeutic effect on the peripheral hyperinsulinemia observed in the early stages of AD disease and the resulting low amount of insulin transported to the brain, by protecting pancreatic cells from apoptosis and oxidative stress by regulating their bioactive metabolites.
Collapse
Affiliation(s)
- Alev Duygu Acun
- Department of Biophysics, Faculty of Medicine, Akdeniz University, Arapsuyu, Antalya 07070, Turkey.
| | - Deniz Kantar
- Department of Biophysics, Faculty of Medicine, Akdeniz University, Arapsuyu, Antalya 07070, Turkey
| |
Collapse
|
2
|
Chauhan A, Dubey S, Jain S. Association Between Type 2 Diabetes Mellitus and Alzheimer's Disease: Common Molecular Mechanism and Therapeutic Targets. Cell Biochem Funct 2024; 42:e4111. [PMID: 39228117 DOI: 10.1002/cbf.4111] [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: 06/12/2024] [Revised: 07/11/2024] [Accepted: 08/16/2024] [Indexed: 09/05/2024]
Abstract
Diabetes mellitus (DM) and Alzheimer's disease (AD) rates are rising, mirroring the global trend of an aging population. Numerous epidemiological studies have shown that those with Type 2 diabetes (T2DM) have an increased risk of developing dementia. These degenerative and progressive diseases share some risk factors. To a large extent, the amyloid cascade is responsible for AD development. Neurofibrillary tangles induce neurodegeneration and brain atrophy; this chain reaction begins with hyperphosphorylation of tau proteins caused by progressive amyloid beta (Aβ) accumulation. In addition to these processes, it seems that alterations in brain glucose metabolism and insulin signalling lead to cell death and reduced synaptic plasticity in AD, before the onset of symptoms, which may be years away. Due to the substantial evidence linking insulin resistance in the brain with AD, researchers have coined the name "Type 3 diabetes" to characterize the condition. We still know little about the processes involved, even though current animal models have helped illuminate the links between T2DM and AD. This brief overview discusses insulin and IGF-1 signalling disorders and the primary molecular pathways that may connect them. The presence of GSK-3β in AD is intriguing. These proteins' association with T2DM and pancreatic β-cell failure suggests they might be therapeutic targets for both disorders.
Collapse
Affiliation(s)
- Aparna Chauhan
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Rajasthan, India
| | - Sachin Dubey
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Rajasthan, India
| | - Smita Jain
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Rajasthan, India
| |
Collapse
|
3
|
Preeti K, Sood A, Fernandes V, Khan I, Khatri DK, Singh SB. Experimental Type 2 diabetes and lipotoxicity-associated neuroinflammation involve mitochondrial DNA-mediated cGAS/STING axis: implication of Type-1 interferon response in cognitive impairment. Mol Neurobiol 2024; 61:6217-6244. [PMID: 38285288 DOI: 10.1007/s12035-024-03933-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 01/05/2024] [Indexed: 01/30/2024]
Abstract
Type-1 IFN (interferon)-associated innate immune response is increasingly getting attention in neurodegenerative and metabolic diseases like type 2 diabetes (T2DM). However, its significance in T2DM/lipotoxicity-induced neuroglia changes and cognitive impairment is missing. The present study aims to evaluate the involvement of cGAS (cyclic GMP-AMP synthase)-STING (stimulator of interferon gene), IRF3 (interferon regulatory factor-3), TBK (TANK binding kinase)-mediated Type-1 IFN response in the diabetic brain, and lipotoxicity (palmitate-bovine serum albumin conjugate/PA-BSA)-induced changes in cells (neuro2a and BV2). T2DM was induced in C57/BL6 mice by feeding on a high-fat diet (HFD, 60% Kcal) for 16 weeks and injecting a single dose of streptozotocin (100 mg/kg, i.p) in the 12th week. Plasma biochemical parameter analysis, neurobehavioral assessment, protein expression, and quantitative polymerase chain reaction study were carried out to decipher the hypothesis. T2DM-associated metabolic and lipotoxic stress led to mitochondrial impairment causing leakage of mtDNA to the cytoplasm further commencing cGAS activation and its downstream signaling. The diseased hippocampus and cortex showed decreased expression of synaptophysin (p < 0.01) and PSD-95 (p < 0.01, p < 0.05) with increased expression of cGAS (p < 0.001), p-STING (p < 0.001), p-STAT1 (signal transducer and activator of transcription) (p < 0.01), and IFN-β (p < 0.001) compared to normal control. The IFN-β/p-STAT1-mediated microglia activation was executed employing a conditioned media approach. C-176, a selective STING inhibitor, alleviated cGAS/p-STING/IFN-β expression and proinflammatory microglia/M1-associated markers (CD16 expression, CXCL10, TNF-α, IL-1β mRNA fold change) in the diabetic brain. The present study suggests Type-1IFN response may result in neuroglia dyshomeostasis affecting normal brain function. Alleviating STING signaling has the potential to protect T2DM-associated central ailment.
Collapse
Affiliation(s)
- Kumari Preeti
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Anika Sood
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Valencia Fernandes
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Islauddin Khan
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Dharmendra Kumar Khatri
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India.
- Department of Pharmacology, Shobhaben Pratapbai Patel School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-University, Mumbai, 400056, India.
| | - Shashi Bala Singh
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India.
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India.
| |
Collapse
|
4
|
Zhang N, Zhang S, Dong X. Plant-derived bioactive compounds and their novel role in central nervous system disorder treatment via ATF4 targeting: A systematic literature review. Biomed Pharmacother 2024; 176:116811. [PMID: 38795641 DOI: 10.1016/j.biopha.2024.116811] [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: 03/30/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 05/28/2024] Open
Abstract
Central nervous system (CNS) disorders exhibit exceedingly intricate pathogenic mechanisms. Pragmatic and effective solutions remain elusive, significantly compromising human life and health. Activating transcription factor 4 (ATF4) participates in the regulation of multiple pathophysiological processes, including CNS disorders. Considering the widespread involvement of ATF4 in the pathological process of CNS disorders, the targeted regulation of ATF4 by plant-derived bioactive compounds (PDBCs) may become a viable strategy for the treatment of CNS disorders. However, the regulatory relationship between PDBCs and ATF4 remains incompletely understood. Here, we aimed to comprehensively review the studies on PDBCs targeting ATF4 to ameliorate CNS disorders, thereby offering novel directions and insights for the treatment of CNS disorders. A computerized search was conducted on PubMed, Embase, Web of Science, and Google Scholar databases to identify preclinical experiments related to PDBCs targeting ATF4 for the treatment of CNS disorders. The search timeframe was from the inception of the databases to December 2023. Two assessors conducted searches using the keywords "ATF4," "Central Nervous System," "Neurological," "Alzheimer's disease," "Parkinson's Disease," "Stroke," "Spinal Cord Injury," "Glioblastoma," "Traumatic Brain Injury," and "Spinal Cord Injury." Overall, 31 studies were included, encompassing assessments of 27 PDBCs. Combining results from in vivo and in vitro studies, we observed that these PDBCs, via ATF4 modulation, prevent the deposition of amyloid-like fibers such as Aβ, tau, and α-synuclein. They regulate ERS, reduce the release of inflammatory factors, restore mitochondrial membrane integrity to prevent oxidative stress, regulate synaptic plasticity, modulate autophagy, and engage anti-apoptotic mechanisms. Consequently, they exert neuroprotective effects in CNS disorders. Numerous PDBCs targeting ATF4 have shown potential in facilitating the restoration of CNS functionality, thereby presenting expansive prospects for the treatment of such disorders. However, future endeavors necessitate high-quality, large-scale, and comprehensive preclinical and clinical studies to further validate this therapeutic potential.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Neurology, the Seventh Clinical College of China Medical University, No. 24 Central Street, Xinfu District, Fushun, Liaoning 113000, China
| | - Shun Zhang
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao street, Heping District, Shenyang, Liaoning 110000, China
| | - Xiaoyu Dong
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao street, Heping District, Shenyang, Liaoning 110000, China.
| |
Collapse
|
5
|
Shapiro ALB, Coughlan C, Bettcher BM, Pauley ME, Kim J, Bjornstad P, Rajic B, Truong J, Bell C, Choi YJ, Walker KA, Potter H, Liese AD, Dabelea D, Whitlow CT. Biomarkers of Neurodegeneration and Alzheimer's Disease Neuropathology in Adolescents and Young Adults with Youth-Onset Type 1 or Type 2 Diabetes: A Proof-of-Concept Study. ENDOCRINES 2024; 5:197-213. [PMID: 38764894 PMCID: PMC11101213 DOI: 10.3390/endocrines5020014] [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] [Indexed: 05/21/2024] Open
Abstract
Adult-onset diabetes increases one's risk of neurodegenerative disease including Alzheimer's disease (AD); however, the risk associated with youth-onset diabetes (Y-DM) remains underexplored. We quantified plasma biomarkers of neurodegeneration and AD in participants with Y-DM from the SEARCH cohort at adolescence and young adulthood (Type 1, n = 25; Type 2, n = 25; 59% female; adolescence, age = 15 y/o [2.6]; adulthood, age = 27.4 y/o [2.2]), comparing them with controls (adolescence, n = 25, age = 14.8 y/o [2.7]; adulthood, n = 21, age = 24.9 y/o [2.8]). Plasma biomarkers, including glial fibrillary acidic protein (GFAP), neurofilament light chain protein (NfL), phosphorylated tau-181 (pTau181), and amyloid beta (Aβ40, Aβ42), were measured via Simoa. A subset of participants (n = 7; age = 27.5 y/o [5.7]) and six controls (age = 25.1 y/o [4.5]) underwent PET scans to quantify brain amyloid and tau densities in AD sensitive brain regions. Y-DM adolescents exhibited lower plasma levels of Aβ40, Aβ42, and GFAP, and higher pTau181 compared to controls (p < 0.05), a pattern persisting into adulthood (p < 0.001). All biomarkers showed significant increases from adolescence to adulthood in Y-DM (p < 0.01), though no significant differences in brain amyloid or tau were noted between Y-DM and controls in adulthood. Preliminary evidence suggests that preclinical AD neuropathology is present in young people with Y-DM, indicating a potential increased risk of neurodegenerative diseases.
Collapse
Affiliation(s)
- Allison L. B. Shapiro
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado at Anschutz (CU-Anschutz), Aurora, CO 80045, USA
- Section of Endocrinology, Department of Pediatrics, School of Medicine (SOM), CU-Anschutz, Aurora, CO 80045, USA
| | - Christina Coughlan
- University of Colorado Alzheimer’s and Cognition Center, CU-Anschutz, Aurora, CO 80045, USA
- Department of Neurology, SOM, CU-Anschutz, Aurora, CO 80045, USA
| | | | - Meghan E. Pauley
- Barbara Davis Center for Diabetes, CU-Anschutz, Aurora, CO 80045, USA
| | - Jeongchul Kim
- Radiology Informatics and Image Processing Laboratory, Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Petter Bjornstad
- Section of Endocrinology, Department of Pediatrics, School of Medicine (SOM), CU-Anschutz, Aurora, CO 80045, USA
- Division of Renal Diseases and Hypertension, Department of Medicine, SOM, CU-Anschutz, Aurora, CO 80045, USA
| | - Benjamin Rajic
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado at Anschutz (CU-Anschutz), Aurora, CO 80045, USA
| | - Jennifer Truong
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado at Anschutz (CU-Anschutz), Aurora, CO 80045, USA
| | - Christopher Bell
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado at Anschutz (CU-Anschutz), Aurora, CO 80045, USA
| | - Ye Ji Choi
- Section of Endocrinology, Department of Pediatrics, School of Medicine (SOM), CU-Anschutz, Aurora, CO 80045, USA
| | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD 20814, USA
| | - Huntington Potter
- University of Colorado Alzheimer’s and Cognition Center, CU-Anschutz, Aurora, CO 80045, USA
- Department of Neurology, SOM, CU-Anschutz, Aurora, CO 80045, USA
| | - Angela D. Liese
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Dana Dabelea
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado at Anschutz (CU-Anschutz), Aurora, CO 80045, USA
| | - Christopher T. Whitlow
- Radiology Informatics and Image Processing Laboratory, Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| |
Collapse
|
6
|
Guo B, Li QY, Liu XJ, Luo GH, Wu YJ, Nie J. Diabetes mellitus and Alzheimer's disease: Vacuolar adenosine triphosphatase as a potential link. Eur J Neurosci 2024; 59:2577-2595. [PMID: 38419188 DOI: 10.1111/ejn.16286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 03/02/2024]
Abstract
Globally, the incidence of diabetes mellitus (DM) and Alzheimer's disease (AD) is increasing year by year, causing a huge economic and social burden, and their pathogenesis and aetiology have been proven to have a certain correlation. In recent years, more and more studies have shown that vacuolar adenosine triphosphatases (v-ATPases) in eukaryotes, which are biomolecules regulating lysosomal acidification and glycolipid metabolism, play a key role in DM and AD. This article describes the role of v-ATPase in DM and AD, including its role in glycolysis, insulin secretion and insulin resistance (IR), as well as its relationship with lysosomal acidification, autophagy and β-amyloid (Aβ). In DM, v-ATPase is involved in the regulation of glucose metabolism and IR. v-ATPase is closely related to glycolysis. On the one hand, v-ATPase affects the rate of glycolysis by affecting the secretion of insulin and changing the activities of key glycolytic enzymes hexokinase (HK) and phosphofructokinase 1 (PFK-1). On the other hand, glucose is the main regulator of this enzyme, and the assembly and activity of v-ATPase depend on glucose, and glucose depletion will lead to its decomposition and inactivation. In addition, v-ATPase can also regulate free fatty acids, thereby improving IR. In AD, v-ATPase can not only improve the abnormal brain energy metabolism by affecting lysosomal acidification and autophagy but also change the deposition of Aβ by affecting the production and degradation of Aβ. Therefore, v-ATPase may be the bridge between DM and AD.
Collapse
Affiliation(s)
- Bin Guo
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Qi-Ye Li
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xue-Jia Liu
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Guo-Hui Luo
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Ya-Juan Wu
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jing Nie
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
7
|
Zhang Q, Hu S, Jin Z, Wang S, Zhang B, Zhao L. Mechanism of traditional Chinese medicine in elderly diabetes mellitus and a systematic review of its clinical application. Front Pharmacol 2024; 15:1339148. [PMID: 38510656 PMCID: PMC10953506 DOI: 10.3389/fphar.2024.1339148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/31/2024] [Indexed: 03/22/2024] Open
Abstract
Objective: Affected by aging, the elderly diabetes patients have many pathological characteristics different from the young people, including more complications, vascular aging, cognitive impairment, osteoporosis, and sarcopenia. This article will explore their pathogenesis and the mechanism of Traditional Chinese medicine (TCM) intervention, and use the method of systematic review to evaluate the clinical application of TCM in elderly diabetes. Method: Searching for randomized controlled trials (RCTs) published from January 2000 to November 2023 in the following databases: Web of Science, Pubmed, Embase, Cochrane Library, Sinomed, China National Knowledge Internet, Wanfang and VIP. They were evaluated by three subgroups of Traditional Chinese Prescription, Traditional Chinese patent medicines and Traditional Chinese medicine extracts for their common prescriptions, drugs, adverse reactions and the quality of them. Results and Conclusion: TCM has the advantages of multi-target and synergistic treatment in the treatment of elderly diabetes. However, current clinical researches have shortcomings including the inclusion of age criteria and diagnosis of subjects are unclear, imprecise research design, non-standard intervention measures, and its safety needs further exploration. In the future, the diagnosis of elderly people with diabetes needs to be further clarified. Traditional Chinese patent medicines included in the pharmacopoeia can be used to conduct more rigorous RCTs, and then gradually standardize the traditional Chinese medicine prescriptions and traditional Chinese medicine extracts, providing higher level evidence for the treatment of elderly diabetes with traditional Chinese medicine.
Collapse
Affiliation(s)
- Qiqi Zhang
- Institute of Metabolic Diseases, Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Shiwan Hu
- Institute of Metabolic Diseases, Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Zishan Jin
- Institute of Metabolic Diseases, Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Sicheng Wang
- Institute of Metabolic Diseases, Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Boxun Zhang
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Linhua Zhao
- Institute of Metabolic Diseases, Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
8
|
Yin K, Wu R. Systematic Investigation of Dose-Dependent Protein Thermal Stability Changes to Uncover the Mechanisms of the Pleiotropic Effects of Metformin. ACS Pharmacol Transl Sci 2024; 7:467-477. [PMID: 38357277 PMCID: PMC10863438 DOI: 10.1021/acsptsci.3c00298] [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: 10/24/2023] [Revised: 12/16/2023] [Accepted: 12/22/2023] [Indexed: 02/16/2024]
Abstract
Metformin is a widely used drug to treat type II diabetes. Beyond lowering blood sugar, it has been reported to have pleiotropic effects such as suppressing cancer growth and attenuating cell oxidative stress and inflammation. However, the underlying mechanisms of these effects remain to be explored. Here, we systematically study the thermal stability changes of proteins in liver cells (HepG2) induced by a wide dosage range of metformin by using the proteome integral solubility alteration (PISA) assay. The current results demonstrate that, besides the most accepted target of metformin (complex I), low concentrations of metformin (such as 0.2 μM) stabilize the complex IV subunits, suggesting its important role in the sugar-lowering effect. Low-dose metformin also results in stability alterations of ribosomal proteins, correlating with its inhibitive effect on cell proliferation. We further find that low-concentration metformin impacts mitochondrial cargo and vesicle transport, while high-concentration metformin affects cell redox responses and cell membrane protein sorting. This study provides mechanistic insights into the molecular mechanisms of lowering blood sugar and the pleiotropic effects of metformin.
Collapse
Affiliation(s)
- Kejun Yin
- School of Chemistry and Biochemistry
and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Ronghu Wu
- School of Chemistry and Biochemistry
and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
9
|
Valentin-Escalera J, Leclerc M, Calon F. High-Fat Diets in Animal Models of Alzheimer's Disease: How Can Eating Too Much Fat Increase Alzheimer's Disease Risk? J Alzheimers Dis 2024; 97:977-1005. [PMID: 38217592 PMCID: PMC10836579 DOI: 10.3233/jad-230118] [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] [Accepted: 11/15/2023] [Indexed: 01/15/2024]
Abstract
High dietary intake of saturated fatty acids is a suspected risk factor for neurodegenerative diseases, including Alzheimer's disease (AD). To decipher the causal link behind these associations, high-fat diets (HFD) have been repeatedly investigated in animal models. Preclinical studies allow full control over dietary composition, avoiding ethical concerns in clinical trials. The goal of the present article is to provide a narrative review of reports on HFD in animal models of AD. Eligibility criteria included mouse models of AD fed a HFD defined as > 35% of fat/weight and western diets containing > 1% cholesterol or > 15% sugar. MEDLINE and Embase databases were searched from 1946 to August 2022, and 32 preclinical studies were included in the review. HFD-induced obesity and metabolic disturbances such as insulin resistance and glucose intolerance have been replicated in most studies, but with methodological variability. Most studies have found an aggravating effect of HFD on brain Aβ pathology, whereas tau pathology has been much less studied, and results are more equivocal. While most reports show HFD-induced impairment on cognitive behavior, confounding factors may blur their interpretation. In summary, despite conflicting results, exposing rodents to diets highly enriched in saturated fat induces not only metabolic defects, but also cognitive impairment often accompanied by aggravated neuropathological markers, most notably Aβ burden. Although there are important variations between methods, particularly the lack of diet characterization, these studies collectively suggest that excessive intake of saturated fat should be avoided in order to lower the incidence of AD.
Collapse
Affiliation(s)
- Josue Valentin-Escalera
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l'Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain - Laboratoire International Associé (NutriNeuro France-INAF Canada)
| | - Manon Leclerc
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l'Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain - Laboratoire International Associé (NutriNeuro France-INAF Canada)
| | - Frédéric Calon
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l'Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain - Laboratoire International Associé (NutriNeuro France-INAF Canada)
| |
Collapse
|
10
|
Pradhan SP, Sahu PK, Behera A. New insights toward molecular and nanotechnological approaches to antidiabetic agents for Alzheimer's disease. Mol Cell Biochem 2023; 478:2739-2762. [PMID: 36949264 DOI: 10.1007/s11010-023-04696-1] [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: 04/20/2022] [Accepted: 02/27/2023] [Indexed: 03/24/2023]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder affecting a major class of silver citizens. The disorder shares a mutual relationship on account of its cellular and molecular pathophysiology with type-II diabetes mellitus (DM). Chronic DM increases the risk for AD. Emerging evidence recommended that resistance in insulin production develops cognitive dysfunction, which generally leads to AD. Repurposing of antidiabetic drugs can be effective in preventing and treatment of the neurodegenerative disorder. Limitations of antidiabetic drugs restrict the repurposing of the drugs for other disorders. Therefore, nanotechnological intervention plays a significant role in the treatment of neurological disorders. In this review, we discuss the common cellular and molecular pathophysiologies between AD and type-II DM, the relevance of in vivo models of type II DM in the study of AD, and the repurposing of antidiabetic drugs and the nanodelivery systems of antidiabetic drugs against AD.
Collapse
Affiliation(s)
- Sweta Priyadarshini Pradhan
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University, Campus-II, Kalinga Nagar, Bhubaneswar, Odisha, India
| | - Pratap Kumar Sahu
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University, Campus-II, Kalinga Nagar, Bhubaneswar, Odisha, India
| | - Anindita Behera
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University, Campus-II, Kalinga Nagar, Bhubaneswar, Odisha, India.
| |
Collapse
|
11
|
Veselov IM, Vinogradova DV, Maltsev AV, Shevtsov PN, Spirkova EA, Bachurin SO, Shevtsova EF. Mitochondria and Oxidative Stress as a Link between Alzheimer's Disease and Diabetes Mellitus. Int J Mol Sci 2023; 24:14450. [PMID: 37833898 PMCID: PMC10572926 DOI: 10.3390/ijms241914450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
This review is devoted to the problems of the common features linking metabolic disorders and type 2 diabetes with the development of Alzheimer's disease. The pathogenesis of Alzheimer's disease closely intersects with the mechanisms of type 2 diabetes development, and an important risk factor for both pathologies is aging. Common pathological mechanisms include both factors in the development of oxidative stress, neuroinflammation, insulin resistance, and amyloidosis, as well as impaired mitochondrial dysfunctions and increasing cell death. The currently available drugs for the treatment of type 2 diabetes and Alzheimer's disease have limited therapeutic efficacy. It is important to note that drugs used to treat Alzheimer's disease, in particular acetylcholinesterase inhibitors, show a positive therapeutic potential in the treatment of type 2 diabetes, while drugs used in the treatment of type 2 diabetes can also prevent a number of pathologies characteristic for Alzheimer's disease. A promising direction in the search for a strategy for the treatment of type 2 diabetes and Alzheimer's disease may be the creation of complex multi-target drugs that have neuroprotective potential and affect specific common targets for type 2 diabetes and Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Elena F. Shevtsova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences (IPAC RAS), Chernogolovka 142432, Russia; (I.M.V.); (A.V.M.); (P.N.S.); (E.A.S.); (S.O.B.)
| |
Collapse
|
12
|
Libard S, Alafuzoff I. Is islet amyloid polypeptide indeed expressed in the human brain? Neuropathol Appl Neurobiol 2023; 49:e12917. [PMID: 37317631 DOI: 10.1111/nan.12917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 06/07/2023] [Accepted: 06/11/2023] [Indexed: 06/16/2023]
Abstract
AIMS This study aims to study the association between pancreatic islet amyloid polypeptide (IAPP) and Alzheimer's disease neuropathological change (ADNC) in brain biopsies obtained from subjects with idiopathic normal pressure hydrocephalus (iNPH) and in post-mortem (PM) brain samples obtained from aged individuals. METHODS For the immunohistochemical (IHC) analyses, two IAPP antibodies (Abs), monoclonal and polyclonal, and Abs directed towards ADNC were applied. RESULTS The iNPH cohort included 113 subjects. Amyloid-β (Aβ) was detected in 50% and hyperphosphorylated τ (HPτ) in 47% of the cases. Concomitant pathology was seen in 32%. The PM cohort included 77 subjects. Aβ was detected in 69% and HPτ in 91% of the cases. Combined Aβ/HPτ pathology was seen in 62%. Reactivity for the monoclonal IAPP was not detected in the brain tissue in either of the cohorts. Reactivity for the polyclonal IAPP was observed in all 77 PM brain samples. CONCLUSIONS There was no specific expression of IAPP in human brain tissue; hence, an association between IAPP and ADNC is not assessable. Of note, the observed reactivity of the polyclonal IAPP Ab was not reproduced with a specific monoclonal Ab; thus, we considered the observed staining with the polyclonal Ab to be unreliable. When using IHC, several pitfalls, especially the choice of an Ab, always need to be considered. Polyclonal Abs cross-react with other epitopes and proteins, thus leading to false-positive results. This seems to be the case for the polyclonal IAPP Abs in the human brain.
Collapse
Affiliation(s)
- Sylwia Libard
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Pathology, Uppsala University Hospital, Uppsala, Sweden
| | - Irina Alafuzoff
- Department of Pathology, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
13
|
Zhai W, Zhang T, Jin Y, Huang S, Xu M, Pan J. The fibroblast growth factor system in cognitive disorders and dementia. Front Neurosci 2023; 17:1136266. [PMID: 37214403 PMCID: PMC10196031 DOI: 10.3389/fnins.2023.1136266] [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: 01/02/2023] [Accepted: 04/19/2023] [Indexed: 05/24/2023] Open
Abstract
Cognitive impairment is the core precursor to dementia and other cognitive disorders. Current hypotheses suggest that they share a common pathological basis, such as inflammation, restricted neurogenesis, neuroendocrine disorders, and the destruction of neurovascular units. Fibroblast growth factors (FGFs) are cell growth factors that play essential roles in various pathophysiological processes via paracrine or autocrine pathways. This system consists of FGFs and their receptors (FGFRs), which may hold tremendous potential to become a new biological marker in the diagnosis of dementia and other cognitive disorders, and serve as a potential target for drug development against dementia and cognitive function impairment. Here, we review the available evidence detailing the relevant pathways mediated by multiple FGFs and FGFRs, and recent studies examining their role in the pathogenesis and treatment of cognitive disorders and dementia.
Collapse
|
14
|
Franklin ZJ, Croce L, Dekeryte R, Delibegovic M, Platt B. BACE cleavage of APP does not drive the diabetic phenotype of PLB4 mice. Neurobiol Dis 2023; 182:106142. [PMID: 37137417 DOI: 10.1016/j.nbd.2023.106142] [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: 01/29/2023] [Revised: 04/26/2023] [Accepted: 04/29/2023] [Indexed: 05/05/2023] Open
Abstract
BACKGROUND Alzheimer's Disease (AD) and Type 2 Diabetes Mellitus (T2DM), two prevalent diseases related to ageing, often share common pathologies including increased inflammation, endoplasmic reticulum (ER) stress, and impaired metabolic homeostasis predominantly affecting different organs. Therefore, it was unexpected to find in a previous study that neuronal hBACE1 knock-in (PLB4 mouse) leads to both an AD- and T2DM- like phenotype. The complexity of this co-morbidity phenotype required a deeper systems approach to explore the age-related changes in AD and T2DM-like pathologies of the PLB4 mouse. Therefore, we here analysed key neuronal and metabolic tissues comparing associated pathologies to those of normal ageing. METHODS Glucose tolerance, insulin sensitivity and protein turnover were assessed in 5-h fasted 3- and 8-month-old male PLB4 and wild-type mice. Western Blot and quantitative PCR were performed to determine regulation of homeostatic and metabolic pathways in insulin-stimulated brain, liver and muscle tissue. RESULTS Neuronal hBACE1 expression caused early pathological cleavage of APP (increased monomeric Aβ (mAβ) levels at 3-months), in parallel with brain ER stress (increased phosphorylation of the translation regulation factor (p-eIF2α) and the chaperone binding immunoglobulin protein (BIP)). However, APP processing shifted over time (higher full-length APP and sAPPβ levels, alongside lower mAβ and secreted APPα at 8 months), along with increased ER stress (phosphorylated/total inositol-requiring enzyme 1α (IRE1α)) in brain and liver. Metabolically, systemic glucose intolerance was evident from 3 months, yet metabolic signalling varied greatly between tissues and ages, and was confined to the periphery (muscle insulin receptors (IR), dipeptidyl-peptidase-4 (DPP4) levels, and decreased phosphorylated protein Kinase B (p-Akt), alongside increased liver DPP4 and fibroblast growth factor 21 (FGF21)), all of which normalised to wild-type levels at 8 months. CONCLUSION Our data suggest that the murine nervous system is affected early by APP misprocessing as a result of hBACE1 introduction, which coincided with ER stress, but not IR changes, and was alleviated with age. Peripheral metabolic alterations occurred early and revealed tissue-specific (liver vs. muscle) adaptations in metabolic markers but did not correlate with neuronal APP processing. Compensatory vs. contributory neuronal mechanisms associated with hBACE1 expression at different ages may explain why mice intrinsically do not develop AD pathologies and may offer new insights for future interventions.
Collapse
Affiliation(s)
- Z J Franklin
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, Scotland, UK
| | - L Croce
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, Scotland, UK
| | - R Dekeryte
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, Scotland, UK
| | - M Delibegovic
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, Scotland, UK
| | - B Platt
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, Scotland, UK.
| |
Collapse
|
15
|
Al-Lahham R, Mendez N. Tau Loss of Function, by Deletion or Aggregation, Contributes to Peripheral Insulin Resistance. J Alzheimers Dis 2023; 95:1041-1058. [PMID: 37638441 PMCID: PMC10578286 DOI: 10.3233/jad-230392] [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] [Accepted: 07/12/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND Several epidemiological data revealed an association between Alzheimer's disease (AD) and type 2 diabetes. Researchers concentrated on brain insulin resistance with little emphasis on the link between systemic insulin resistance and AD, despite the fact that the incidence of type 2 diabetes is higher in AD patients and that impairment in insulin signaling is a risk factor for AD. OBJECTIVE The goal of this study is to determine the role of systemic insulin resistance in the pathogenesis of Alzheimer's disease by evaluating the consequences of tau loss-of-function on peripheral insulin sensitivity. METHODS Primary hepatocytes isolated from transgenic mouse models (Tau KO, P301 L) and wild type mice (C57BL/6) were evaluated for their insulin sensitivity using glucose uptake assays as well as biochemical analysis of insulin signaling markers. RESULTS Our data show that tau deletion or loss of function promotes peripheral insulin resistance as seen in primary hepatocytes isolated from Tau KO and P301 L mice, respectively. Furthermore, exposure of wild-type primary hepatocytes to sub-toxic concentrations of tau oligomers results in a dose-dependent inhibition of glucose uptake, associated with downregulation of insulin signaling. Tau oligomers-induced inactivation of insulin signaling proteins was rescued by inhibition of p38 MAPK, suggesting the involvement of p38 MAPK. CONCLUSIONS This is the first study testing tau role in peripheral insulin resistance at the cellular level using multiple transgenic mouse models. Moreover, this study suggests that tau should be functional for insulin sensitivity, therefore, any loss of function by deletion or aggregation would result in insulin resistance.
Collapse
Affiliation(s)
- Rabab Al-Lahham
- Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Nicolas Mendez
- Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
16
|
Hamzé R, Delangre E, Tolu S, Moreau M, Janel N, Bailbé D, Movassat J. Type 2 Diabetes Mellitus and Alzheimer's Disease: Shared Molecular Mechanisms and Potential Common Therapeutic Targets. Int J Mol Sci 2022; 23:ijms232315287. [PMID: 36499613 PMCID: PMC9739879 DOI: 10.3390/ijms232315287] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
The global prevalence of diabetes mellitus and Alzheimer's disease is increasing alarmingly with the aging of the population. Numerous epidemiological data suggest that there is a strong association between type 2 diabetes and an increased risk of dementia. These diseases are both degenerative and progressive and share common risk factors. The amyloid cascade plays a key role in the pathophysiology of Alzheimer's disease. The accumulation of amyloid beta peptides gradually leads to the hyperphosphorylation of tau proteins, which then form neurofibrillary tangles, resulting in neurodegeneration and cerebral atrophy. In Alzheimer's disease, apart from these processes, the alteration of glucose metabolism and insulin signaling in the brain seems to induce early neuronal loss and the impairment of synaptic plasticity, years before the clinical manifestation of the disease. The large amount of evidence on the existence of insulin resistance in the brain during Alzheimer's disease has led to the description of this disease as "type 3 diabetes". Available animal models have been valuable in the understanding of the relationships between type 2 diabetes and Alzheimer's disease, but to date, the mechanistical links are poorly understood. In this non-exhaustive review, we describe the main molecular mechanisms that may link these two diseases, with an emphasis on impaired insulin and IGF-1 signaling. We also focus on GSK3β and DYRK1A, markers of Alzheimer's disease, which are also closely associated with pancreatic β-cell dysfunction and type 2 diabetes, and thus may represent common therapeutic targets for both diseases.
Collapse
Affiliation(s)
- Rim Hamzé
- Team Biology and Pathology of the Endocrine Pancreas, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Etienne Delangre
- Team Biology and Pathology of the Endocrine Pancreas, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Stefania Tolu
- Team Biology and Pathology of the Endocrine Pancreas, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Manon Moreau
- Team Degenerative Process, Stress and Aging, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Nathalie Janel
- Team Degenerative Process, Stress and Aging, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Danielle Bailbé
- Team Biology and Pathology of the Endocrine Pancreas, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Jamileh Movassat
- Team Biology and Pathology of the Endocrine Pancreas, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
- Correspondence: ; Tel.: +33-1-57-27-77-82; Fax: +33-1-57-27-77-91
| |
Collapse
|
17
|
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
|
18
|
Hardy-Sosa A, León-Arcia K, Llibre-Guerra JJ, Berlanga-Acosta J, Baez SDLC, Guillen-Nieto G, Valdes-Sosa PA. Diagnostic Accuracy of Blood-Based Biomarker Panels: A Systematic Review. Front Aging Neurosci 2022; 14:683689. [PMID: 35360215 PMCID: PMC8963375 DOI: 10.3389/fnagi.2022.683689] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 01/24/2022] [Indexed: 01/10/2023] Open
Abstract
Background Because of high prevalence of Alzheimer's disease (AD) in low- and middle-income countries (LMICs), there is an urgent need for inexpensive and minimally invasive diagnostic tests to detect biomarkers in the earliest and asymptomatic stages of the disease. Blood-based biomarkers are predicted to have the most impact for use as a screening tool and predict the onset of AD, especially in LMICs. Furthermore, it has been suggested that panels of markers may perform better than single protein candidates. Methods Medline/Pubmed was searched to identify current relevant studies published from January 2016 to December 2020. We included all full-text articles examining blood-based biomarkers as a set of protein markers or panels to aid in AD's early diagnosis, prognosis, and characterization. Results Seventy-six articles met the inclusion criteria for systematic review. Majority of the studies reported plasma and serum as the main source for biomarker determination in blood. Protein-based biomarker panels were reported to aid in AD diagnosis and prognosis with better accuracy than individual biomarkers. Conventional (amyloid-beta and tau) and neuroinflammatory biomarkers, such as amyloid beta-42, amyloid beta-40, total tau, phosphorylated tau-181, and other tau isoforms, were the most represented. We found the combination of amyloid beta-42/amyloid beta-40 ratio and APOEε4 status to be most represented with high accuracy for predicting amyloid beta-positron emission tomography status. Conclusion Assessment of Alzheimer's disease biomarkers in blood as a non-invasive and cost-effective alternative will potentially contribute to early diagnosis and improvement of therapeutic interventions. Given the heterogeneous nature of AD, combination of markers seems to perform better in the diagnosis and prognosis of the disease than individual biomarkers.
Collapse
Affiliation(s)
- Anette Hardy-Sosa
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
- Centro de Ingeniería Genética y Biotecnología, La Habana, Cuba
| | | | | | | | - Saiyet de la C. Baez
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
- Centro de Ingeniería Genética y Biotecnología, La Habana, Cuba
| | | | - Pedro A. Valdes-Sosa
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
- Centro de Neurociencias de Cuba, La Habana, Cuba
| |
Collapse
|
19
|
Woodfield A, Porter T, Gilani I, Noordin S, Li QX, Collins S, Martins RN, Maruff P, Masters CL, Rowe CC, Villemagne VL, Dore V, Newsholme P, Laws SM, Verdile G. Insulin resistance, cognition and Alzheimer's disease biomarkers: Evidence that CSF Aβ42 moderates the association between insulin resistance and increased CSF tau levels. Neurobiol Aging 2022; 114:38-48. [DOI: 10.1016/j.neurobiolaging.2022.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/09/2022] [Accepted: 03/07/2022] [Indexed: 12/16/2022]
|
20
|
Benderradji H, Kraiem S, Courty E, Eddarkaoui S, Bourouh C, Faivre E, Rolland L, Caron E, Besegher M, Oger F, Boschetti T, Carvalho K, Thiroux B, Gauvrit T, Nicolas E, Gomez-Murcia V, Bogdanova A, Bongiovanni A, Muhr-Tailleux A, Lancel S, Bantubungi K, Sergeant N, Annicotte JS, Buée L, Vieau D, Blum D, Buée-Scherrer V. Impaired Glucose Homeostasis in a Tau Knock-In Mouse Model. Front Mol Neurosci 2022; 15:841892. [PMID: 35250480 PMCID: PMC8889017 DOI: 10.3389/fnmol.2022.841892] [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: 12/22/2021] [Accepted: 01/21/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is the leading cause of dementia. While impaired glucose homeostasis has been shown to increase AD risk and pathological loss of tau function, the latter has been suggested to contribute to the emergence of the glucose homeostasis alterations observed in AD patients. However, the links between tau impairments and glucose homeostasis, remain unclear. In this context, the present study aimed at investigating the metabolic phenotype of a new tau knock-in (KI) mouse model, expressing, at a physiological level, a human tau protein bearing the P301L mutation under the control of the endogenous mouse Mapt promoter. Metabolic investigations revealed that, while under chow diet tau KI mice do not exhibit significant metabolic impairments, male but not female tau KI animals under High-Fat Diet (HFD) exhibited higher insulinemia as well as glucose intolerance as compared to control littermates. Using immunofluorescence, tau protein was found colocalized with insulin in the β cells of pancreatic islets in both mouse (WT, KI) and human pancreas. Isolated islets from tau KI and tau knock-out mice exhibited impaired glucose-stimulated insulin secretion (GSIS), an effect recapitulated in the mouse pancreatic β-cell line (MIN6) following tau knock-down. Altogether, our data indicate that loss of tau function in tau KI mice and, particularly, dysfunction of pancreatic β cells might promote glucose homeostasis impairments and contribute to metabolic changes observed in AD.
Collapse
Affiliation(s)
- Hamza Benderradji
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Sarra Kraiem
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Emilie Courty
- Univ. Lille, INSERM, CNRS, CHU Lille, Institut Pasteur de Lille, Inserm U1283-UMR8199—EGID, Lille, France
| | - Sabiha Eddarkaoui
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Cyril Bourouh
- Univ. Lille, INSERM, CNRS, CHU Lille, Institut Pasteur de Lille, Inserm U1283-UMR8199—EGID, Lille, France
| | - Emilie Faivre
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Laure Rolland
- Univ. Lille, INSERM, CNRS, CHU Lille, Institut Pasteur de Lille, Inserm U1283-UMR8199—EGID, Lille, France
| | - Emilie Caron
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Development and Plasticity of the Neuroendocrine Brain, Lille, France
| | - Mélanie Besegher
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41—UMS 2014—PLBS, Animal Facility, Lille, France
| | - Frederik Oger
- Univ. Lille, INSERM, CNRS, CHU Lille, Institut Pasteur de Lille, Inserm U1283-UMR8199—EGID, Lille, France
| | - Theo Boschetti
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Kévin Carvalho
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Bryan Thiroux
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Thibaut Gauvrit
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Emilie Nicolas
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Victoria Gomez-Murcia
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Anna Bogdanova
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Antonino Bongiovanni
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41—UMS 2014—PLBS, BioImaging Center Lille, Lille, France
| | - Anne Muhr-Tailleux
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Steve Lancel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167—RID-AGE—Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Kadiombo Bantubungi
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Nicolas Sergeant
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Jean-Sebastien Annicotte
- Univ. Lille, INSERM, CNRS, CHU Lille, Institut Pasteur de Lille, Inserm U1283-UMR8199—EGID, Lille, France
| | - Luc Buée
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Didier Vieau
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - David Blum
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
- *Correspondence: David Blum
| | - Valérie Buée-Scherrer
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| |
Collapse
|
21
|
Lima JEBF, Moreira NCS, Sakamoto-Hojo ET. Mechanisms underlying the pathophysiology of type 2 diabetes: From risk factors to oxidative stress, metabolic dysfunction, and hyperglycemia. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2022; 874-875:503437. [PMID: 35151421 DOI: 10.1016/j.mrgentox.2021.503437] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/08/2021] [Accepted: 12/12/2021] [Indexed: 12/17/2022]
Abstract
Type 2 diabetes (T2D) is a complex multifactorial disease that emerges from the combination of genetic and environmental factors, and obesity, lifestyle, and aging are the most relevant risk factors. Hyperglycemia is the main metabolic feature of T2D as a consequence of insulin resistance and β-cell dysfunction. Among the cellular alterations induced by hyperglycemia, the overproduction of reactive oxygen species (ROS) and consequently oxidative stress, accompanied by a reduced antioxidant response and impaired DNA repair pathways, represent essential mechanisms underlying the pathophysiology of T2D and the development of late complications. Mitochondrial dysfunction, endoplasmic reticulum (ER) stress, and inflammation are also closely correlated with insulin resistance and β-cell dysfunction. This review focus on the mechanisms by which oxidative stress, mitochondrial dysfunction, ER stress, and inflammation are involved in the pathophysiology of T2D, highlighting the importance of the antioxidant response and DNA repair mechanisms counteracting the development of the disease. Moreover, we indicate evidence on how nutritional interventions effectively improve diabetes care. Additionally, we address key molecular characteristics and signaling pathways shared between T2D and Alzheimer's disease (AD), which might probably be implicated in the risk of T2D patients to develop AD.
Collapse
Affiliation(s)
- Jessica E B F Lima
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo - USP, Ribeirão Preto, SP, Brazil
| | - Natalia C S Moreira
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo - USP, Ribeirão Preto, SP, Brazil
| | - Elza T Sakamoto-Hojo
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo - USP, Ribeirão Preto, SP, Brazil; Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
22
|
Tournissac M, Leclerc M, Valentin-Escalera J, Vandal M, Bosoi CR, Planel E, Calon F. Metabolic determinants of Alzheimer's disease: A focus on thermoregulation. Ageing Res Rev 2021; 72:101462. [PMID: 34534683 DOI: 10.1016/j.arr.2021.101462] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/09/2021] [Accepted: 09/11/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a complex age-related neurodegenerative disease, associated with central and peripheral metabolic anomalies, such as impaired glucose utilization and insulin resistance. These observations led to a considerable interest not only in lifestyle-related interventions, but also in repurposing insulin and other anti-diabetic drugs to prevent or treat dementia. Body temperature is the oldest known metabolic readout and mechanisms underlying its maintenance fail in the elderly, when the incidence of AD rises. This raises the possibility that an age-associated thermoregulatory deficit contributes to energy failure underlying AD pathogenesis. Brown adipose tissue (BAT) plays a central role in thermogenesis and maintenance of body temperature. In recent years, the modulation of BAT activity has been increasingly demonstrated to regulate energy expenditure, insulin sensitivity and glucose utilization, which could also provide benefits for AD. Here, we review the evidence linking thermoregulation, BAT and insulin-related metabolic defects with AD, and we propose mechanisms through which correcting thermoregulatory impairments could slow the progression and delay the onset of AD.
Collapse
|
23
|
Lynn J, Park M, Ogunwale C, Acquaah-Mensah GK. A Tale of Two Diseases: Exploring Mechanisms Linking Diabetes Mellitus with Alzheimer's Disease. J Alzheimers Dis 2021; 85:485-501. [PMID: 34842187 DOI: 10.3233/jad-210612] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dementias, including the type associated with Alzheimer's disease (AD), are on the rise worldwide. Similarly, type 2 diabetes mellitus (T2DM) is one of the most prevalent chronic diseases globally. Although mechanisms and treatments are well-established for T2DM, there remains much to be discovered. Recent research efforts have further investigated factors involved in the etiology of AD. Previously perceived to be unrelated diseases, commonalities between T2DM and AD have more recently been observed. As a result, AD has been labeled as "type 3 diabetes". In this review, we detail the shared processes that contribute to these two diseases. Insulin resistance, the main component of the pathogenesis of T2DM, is also present in AD, causing impaired brain glucose metabolism, neurodegeneration, and cognitive impairment. Dysregulation of insulin receptors and components of the insulin signaling pathway, including protein kinase B, glycogen synthase kinase 3β, and mammalian target of rapamycin are reported in both diseases. T2DM and AD also show evidence of inflammation, oxidative stress, mitochondrial dysfunction, advanced glycation end products, and amyloid deposition. The impact that changes in neurovascular structure and genetics have on the development of these conditions is also being examined. With the discovery of factors contributing to AD, innovative treatment approaches are being explored. Investigators are evaluating the efficacy of various T2DM medications for possible use in AD, including but not limited to glucagon-like peptide-1 receptor agonists, and peroxisome proliferator-activated receptor-gamma agonists. Furthermore, there are 136 active trials involving 121 therapeutic agents targeting novel AD biomarkers. With these efforts, we are one step closer to alleviating the ravaging impact of AD on our communities.
Collapse
Affiliation(s)
- Jessica Lynn
- Massachusetts College of Pharmacy & Health Sciences (MCPHS University)/Takeda Pharmaceuticals Biopharmaceutical Industry Fellowship Program, Boston, MA, USA
| | - Mingi Park
- Massachusetts College of Pharmacy & Health Sciences (MCPHS University)/Takeda Pharmaceuticals Biopharmaceutical Industry Fellowship Program, Boston, MA, USA
| | | | - George K Acquaah-Mensah
- Massachusetts College of Pharmacy & Health Sciences (MCPHS University)/Takeda Pharmaceuticals Biopharmaceutical Industry Fellowship Program, Boston, MA, USA
| |
Collapse
|
24
|
Linking Diabetes Mellitus with Alzheimer's Disease: Bioinformatics Analysis for the Potential Pathways and Characteristic Genes. Biochem Genet 2021; 60:1049-1075. [PMID: 34779951 DOI: 10.1007/s10528-021-10154-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 11/03/2021] [Indexed: 01/22/2023]
Abstract
As the surging epidemics with significant disability, Alzheimer's disease (AD) and type II diabetes mellitus (T2DM) with microvascular complications are widely prevalent, sharing considerable similarities in putative pathomechanism. Despite a spurt of researches on the biology, knowledge about their interactive mechanisms is still rudimentary. Applying bioinformatics ways to explore the differentially co-expressed genes contributes to achieve our objectives to find new therapeutic targets. In this study, we firstly integrated gene expression omnibus datasets (GSE28146 and GSE43950) to identify differentially expressed genes. The enrichment analysis of pivotal genes, like gene ontology and pathway signaling proceeded subsequently. Besides, the related protein-protein interaction (PPI) network was then constructed. To further explain the inner connections, we ended up unearthing the biological significance of valuable targets. As a result, a set of 712, 630, 487, and 997 genes were differentially identified in T2DM with microvascular complications and AD at incipient, moderate, and severe, respectively. The enrichment analysis involving both diseases implicated the dominance of immune system, especially the noteworthy chemokine signaling. Multiple comparisons confirmed that CACNA2D3, NUMB, and IER3 were simultaneously participate in these two conditions, whose respective associations with neurological and endocrine diseases, and regulators including interacting chemicals, transcription factors, and miRNAs were analyzed. Bioinformatics analysis eventually concluded that immune-related biological functions and pathways closely link AD and T2DM with microvascular complications. Further exploration of the regulatory factors about CACNA2D3, NUMB, and IER3 in neuroendocrine field may provide us a promising direction to discover potential strategies for the comorbidity status.
Collapse
|
25
|
Zhang M, Yan W, Yu Y, Cheng J, Yi X, Guo T, Liu N, Shang J, Wang Z, Hu H, Chen L. Liraglutide ameliorates diabetes-associated cognitive dysfunction via rescuing autophagic flux. J Pharmacol Sci 2021; 147:234-244. [PMID: 34507632 DOI: 10.1016/j.jphs.2021.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/28/2021] [Accepted: 07/15/2021] [Indexed: 01/16/2023] Open
Abstract
The incidence of diabetes-associated cognitive dysfunction is increasing. However, few clinical interventions are available to prevent the disorder. Several researches have shown that liraglutide, as a glucagon-like peptide-1 analog, has protective effects on various neurodegenerative diseases, but its roles in diabetic cognitive dysfunction are rarely reported. This study aims to investigate the protective effects of liraglutide on diabetic cognitive dysfunction and its underlying mechanisms. In vivo, the effects of liraglutide treatment were investigated in a mouse model of type 2 diabetes mellitus (T2DM). In vitro, we investigated the effects of liraglutide on the high-glucose-induced rat primary neurons. The results showed that liraglutide reduced the escape latency and increased the time in effective area in the Morris water maze test, improved the damage of hippocampal and synaptic ultrastructure, and decreased the accumulation of amyloid β protein in hippocampus of T2DM mice. Furthermore, liraglutide increased the ratio of microtubule-associated protein light 1 chain Ⅱ/Ⅰ, the expression of Beclin1 protein and Lysosome-associated membrane protein 2 in vivo and vitro. Additionally, Bafilomycin A1 which can inhibit the fusion of autophagosome and lysosome partially abolished the effects of liraglutide. These findings indicate liraglutide ameliorates diabetes-associated cognitive dysfunction by rescuing autophagic flux.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Wenhui Yan
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Ye Yu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Jie Cheng
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Xinyao Yi
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Tingli Guo
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Na Liu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Jia Shang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Zhuanzhuan Wang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Hao Hu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Lina Chen
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China.
| |
Collapse
|
26
|
Quincozes-Santos A, Rosa RLD, Bobermin LD, Tureta EF, Santi L, Beys-da-Silva WO. Association between molecular markers of COVID-19 and Alzheimer's disease. J Med Virol 2021; 94:833-835. [PMID: 34647635 PMCID: PMC8662010 DOI: 10.1002/jmv.27391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/05/2021] [Accepted: 10/12/2021] [Indexed: 01/08/2023]
Affiliation(s)
- André Quincozes-Santos
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Rafael L da Rosa
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Larissa D Bobermin
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Emanuela F Tureta
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Lucélia Santi
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Walter O Beys-da-Silva
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
27
|
Shoshan-Barmatz V, Anand U, Nahon-Crystal E, Di Carlo M, Shteinfer-Kuzmine A. Adverse Effects of Metformin From Diabetes to COVID-19, Cancer, Neurodegenerative Diseases, and Aging: Is VDAC1 a Common Target? Front Physiol 2021; 12:730048. [PMID: 34671273 PMCID: PMC8521008 DOI: 10.3389/fphys.2021.730048] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022] Open
Abstract
Metformin has been used for treating diabetes mellitus since the late 1950s. In addition to its antihyperglycemic activity, it was shown to be a potential drug candidate for treating a range of other diseases that include various cancers, cardiovascular diseases, diabetic kidney disease, neurodegenerative diseases, renal diseases, obesity, inflammation, COVID-19 in diabetic patients, and aging. In this review, we focus on the important aspects of mitochondrial dysfunction in energy metabolism and cell death with their gatekeeper VDAC1 (voltage-dependent anion channel 1) as a possible metformin target, and summarize metformin's effects in several diseases and gut microbiota. We question how the same drug can act on diseases with opposite characteristics, such as increasing apoptotic cell death in cancer, while inhibiting it in neurodegenerative diseases. Interestingly, metformin's adverse effects in many diseases all show VDAC1 involvement, suggesting that it is a common factor in metformin-affecting diseases. The findings that metformin has an opposite effect on various diseases are consistent with the fact that VDAC1 controls cell life and death, supporting the idea that it is a target for metformin.
Collapse
Affiliation(s)
- Varda Shoshan-Barmatz
- Department of Life Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Uttpal Anand
- Department of Life Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | | | - Marta Di Carlo
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | - Anna Shteinfer-Kuzmine
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beersheba, Israel
| |
Collapse
|
28
|
Diabetes and Alzheimer's Disease: Might Mitochondrial Dysfunction Help Deciphering the Common Path? Antioxidants (Basel) 2021; 10:antiox10081257. [PMID: 34439505 PMCID: PMC8389322 DOI: 10.3390/antiox10081257] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 02/06/2023] Open
Abstract
A growing number of clinical and epidemiological studies support the hypothesis of a tight correlation between type 2 diabetes mellitus (T2DM) and the development risk of Alzheimer's disease (AD). Indeed, the proposed definition of Alzheimer's disease as type 3 diabetes (T3D) underlines the key role played by deranged insulin signaling to accumulation of aggregated amyloid beta (Aβ) peptides in the senile plaques of the brain. Metabolic disturbances such as hyperglycemia, peripheral hyperinsulinemia, dysregulated lipid metabolism, and chronic inflammation associated with T2DM are responsible for an inefficient transport of insulin to the brain, producing a neuronal insulin resistance that triggers an enhanced production and deposition of Aβ and concomitantly contributes to impairment in the micro-tubule-associated protein Tau, leading to neural degeneration and cognitive decline. Furthermore, the reduced antioxidant capacity observed in T2DM patients, together with the impairment of cerebral glucose metabolism and the decreased performance of mitochondrial activity, suggests the existence of a relationship between oxidative damage, mitochondrial impairment, and cognitive dysfunction that could further reinforce the common pathophysiology of T2DM and AD. In this review, we discuss the molecular mechanisms by which insulin-signaling dysregulation in T2DM can contribute to the pathogenesis and progression of AD, deepening the analysis of complex mechanisms involved in reactive oxygen species (ROS) production under oxidative stress and their possible influence in AD and T2DM. In addition, the role of current therapies as tools for prevention or treatment of damage induced by oxidative stress in T2DM and AD will be debated.
Collapse
|
29
|
Dietary-challenged mice with Alzheimer-like pathology show increased energy expenditure and reduced adipocyte hypertrophy and steatosis. Aging (Albany NY) 2021; 13:10891-10919. [PMID: 33864446 PMCID: PMC8109068 DOI: 10.18632/aging.202978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/27/2021] [Indexed: 02/06/2023]
Abstract
Alzheimer’s disease (AD) is frequently accompanied by progressing weight loss, correlating with mortality. Counter-intuitively, weight loss in old age might predict AD onset but obesity in midlife increases AD risk. Furthermore, AD is associated with diabetes-like alterations in glucose metabolism. Here, we investigated metabolic features of amyloid precursor protein overexpressing APP23 female mice modeling AD upon long-term challenge with high-sucrose (HSD) or high-fat diet (HFD). Compared to wild type littermates (WT), APP23 females were less prone to mild HSD-induced and considerable HFD-induced glucose tolerance deterioration, despite unaltered glucose tolerance during normal-control diet. Indirect calorimetry revealed increased energy expenditure and hyperactivity in APP23 females. Dietary interventions, especially HFD, had weaker effects on lean and fat mass gain, steatosis and adipocyte hypertrophy of APP23 than WT mice, as shown by 1H-magnetic-resonance-spectroscopy, histological and biochemical analyses. Proteome analysis revealed differentially regulated expression of mitochondrial proteins in APP23 livers and brains. In conclusion, hyperactivity, increased metabolic rate, and global mitochondrial dysfunction potentially add up to the development of AD-related body weight changes in APP23 females, becoming especially evident during diet-induced metabolic challenge. These findings emphasize the importance of translating this metabolic phenotyping into human research to decode the metabolic component in AD pathogenesis.
Collapse
|
30
|
Bosoi CR, Vandal M, Tournissac M, Leclerc M, Fanet H, Mitchell PL, Verreault M, Trottier J, Virgili J, Tremblay C, Lippman HR, Bajaj JS, Barbier O, Marette A, Calon F. High-Fat Diet Modulates Hepatic Amyloid β and Cerebrosterol Metabolism in the Triple Transgenic Mouse Model of Alzheimer's Disease. Hepatol Commun 2021; 5:446-460. [PMID: 33681678 PMCID: PMC7917280 DOI: 10.1002/hep4.1609] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/09/2020] [Accepted: 08/20/2020] [Indexed: 12/19/2022] Open
Abstract
Obesity and diabetes are strongly associated not only with fatty liver but also cognitive dysfunction. Moreover, their presence, particularly in midlife, is recognized as a risk factor for Alzheimer's disease (AD). AD, the most common cause of dementia, is increasingly considered as a metabolic disease, although underlying pathogenic mechanisms remain unclear. The liver plays a major role in maintaining glucose and lipid homeostasis, as well as in clearing the AD neuropathogenic factor amyloid-β (Aβ) and in metabolizing cerebrosterol, a cerebral-derived oxysterol proposed as an AD biomarker. We hypothesized that liver impairment induced by obesity contributes to AD pathogenesis. We show that the AD triple transgenic mouse model (3xTg-AD) fed a chow diet presents a hepatic phenotype similar to nontransgenic controls (NTg) at 15 months of age. A high-fat diet (HFD), started at the age of 6 months and continued for 9 months, until sacrifice, induced hepatic steatosis in NTg, but not in 3xTg-AD mice, whereas HFD did not induce changes in hepatic fatty acid oxidation, de novo lipogenesis, and gluconeogenesis. HFD-induced obesity was associated with a reduction of insulin-degrading enzyme, one of the main hepatic enzymes responsible for Aβ clearance. The hepatic rate of cerebrosterol glucuronidation was lower in obese 3xTg-AD than in nonobese controls (P < 0.05) and higher compared with obese NTg (P < 0.05), although circulating levels remained unchanged. Conclusion: Modulation of hepatic lipids, Aβ, and cerebrosterol metabolism in obese 3xTg-AD mice differs from control mice. This study sheds light on the liver-brain axis, showing that the chronic presence of NAFLD and changes in liver function affect peripheral AD features and should be considered during development of biomarkers or AD therapeutic targets.
Collapse
Affiliation(s)
- Cristina R Bosoi
- Centre De Recherche De L'institut De Cardiologie Et Pneumologie De QuébecQuébecCanada.,Faculté De MédecineUniversité LavalQuébecCanada.,Axe NeurosciencesCentre De Recherche du CHU de Québec-Université LavalQuébecCanada
| | - Milène Vandal
- Axe NeurosciencesCentre De Recherche du CHU de Québec-Université LavalQuébecCanada.,Faculté De PharmacieUniversité LavalQuébecCanada
| | - Marine Tournissac
- Axe NeurosciencesCentre De Recherche du CHU de Québec-Université LavalQuébecCanada.,Faculté De PharmacieUniversité LavalQuébecCanada.,OptiNutriBrain International Associated LaboratoryQuébecCanada
| | - Manon Leclerc
- Axe NeurosciencesCentre De Recherche du CHU de Québec-Université LavalQuébecCanada.,Faculté De PharmacieUniversité LavalQuébecCanada
| | - Hortense Fanet
- Axe NeurosciencesCentre De Recherche du CHU de Québec-Université LavalQuébecCanada.,Faculté De PharmacieUniversité LavalQuébecCanada.,OptiNutriBrain International Associated LaboratoryQuébecCanada
| | - Patricia L Mitchell
- Centre De Recherche De L'institut De Cardiologie Et Pneumologie De QuébecQuébecCanada
| | - Mélanie Verreault
- Laboratoire de Pharmacologie MoléculaireAxe Endocrinologie et NéphrologieCentre de Recherche du CHU de Québec (Pavillon CHUL)QuébecCanada
| | - Jocelyn Trottier
- Laboratoire de Pharmacologie MoléculaireAxe Endocrinologie et NéphrologieCentre de Recherche du CHU de Québec (Pavillon CHUL)QuébecCanada
| | - Jessica Virgili
- Axe NeurosciencesCentre De Recherche du CHU de Québec-Université LavalQuébecCanada.,Faculté De PharmacieUniversité LavalQuébecCanada
| | - Cynthia Tremblay
- Axe NeurosciencesCentre De Recherche du CHU de Québec-Université LavalQuébecCanada
| | - H Robert Lippman
- Central Virginia VA Health Care SystemRichmondVA.,Virginia Commonwealth UniversityRichmondVA
| | - Jasmohan S Bajaj
- Central Virginia VA Health Care SystemRichmondVA.,Virginia Commonwealth UniversityRichmondVA
| | - Olivier Barbier
- Faculté De PharmacieUniversité LavalQuébecCanada.,Laboratoire de Pharmacologie MoléculaireAxe Endocrinologie et NéphrologieCentre de Recherche du CHU de Québec (Pavillon CHUL)QuébecCanada
| | - André Marette
- Centre De Recherche De L'institut De Cardiologie Et Pneumologie De QuébecQuébecCanada.,Faculté De MédecineUniversité LavalQuébecCanada
| | - Frédéric Calon
- Axe NeurosciencesCentre De Recherche du CHU de Québec-Université LavalQuébecCanada.,Faculté De PharmacieUniversité LavalQuébecCanada.,OptiNutriBrain International Associated LaboratoryQuébecCanada
| |
Collapse
|
31
|
Zhou J, Walker RL, Gray SL, Marcum ZA, Barthold D, Bowen JD, McCormick W, McCurry SM, Larson EB, Crane PK. Glucose-Dementia Association Is Consistent Over Blood Pressure/Antihypertensive Groups. J Alzheimers Dis 2021; 80:79-90. [PMID: 33554906 DOI: 10.3233/jad-201138] [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] [Indexed: 11/15/2022]
Abstract
BACKGROUND Higher glucose levels are associated with dementia risk in people with and without diabetes. However, little is known about how this association might vary by hypertension status and antihypertensive treatment. Most studies on modifiable dementia risk factors consider each factor in isolation. OBJECTIVE To test the hypothesis that hypertension and antihypertensive treatments may modify associations between glucose levels and dementia. METHODS Analyses of data generated from a research study and clinical care of participants from a prospective cohort of dementia-free older adults, including glucose measures, diabetes and antihypertensive treatments, and blood pressure data. We defined groups based on blood pressure (hypertensive versus not, ≥140/90 mmHg versus <140/90 mmHg) and antihypertensive treatment intensity (0, 1, or ≥2 classes of antihypertensives). We used Bayesian joint models to jointly model longitudinal exposure and time to event data. RESULTS A total of 3,056 participants without diabetes treatment and 480 with diabetes treatment were included (mean age at baseline, 75.1 years; mean 7.5 years of follow-up). Higher glucose levels were associated with greater dementia risk among people without and with treated diabetes. Hazard ratios for dementia were similar across all blood pressure/antihypertensive treatment groups (omnibus p = 0.82 for people without and p = 0.59 for people with treated diabetes). CONCLUSION Hypertension and antihypertensive treatments do not appear to affect the association between glucose and dementia risk in this population-based longitudinal cohort study of community-dwelling older adults. Future studies are needed to examine this question in midlife and by specific antihypertensive treatments.
Collapse
Affiliation(s)
- Jing Zhou
- Kaiser Permanente Health Research Institute, Seattle, WA, USA
| | - Rod L Walker
- Kaiser Permanente Health Research Institute, Seattle, WA, USA
| | - Shelly L Gray
- Departments of Pharmacy, University of Washington, Seattle, WA, USA
| | - Zachary A Marcum
- Departments of Pharmacy, University of Washington, Seattle, WA, USA
| | - Douglas Barthold
- Departments of Pharmacy, University of Washington, Seattle, WA, USA
| | - James D Bowen
- Department of Neurology, Swedish Hospital Medical Center, Seattle, WA, USA
| | - Wayne McCormick
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Susan M McCurry
- Departments of Psychosocial and Community Health, University of Washington, Seattle, WA, USA
| | - Eric B Larson
- Kaiser Permanente Health Research Institute, Seattle, WA, USA
| | - Paul K Crane
- Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
32
|
Scholefield M, Unwin RD, Cooper GJ. Shared perturbations in the metallome and metabolome of Alzheimer's, Parkinson's, Huntington's, and dementia with Lewy bodies: A systematic review. Ageing Res Rev 2020; 63:101152. [PMID: 32846222 DOI: 10.1016/j.arr.2020.101152] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 08/06/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022]
Abstract
Despite differences in presentation, age-related dementing diseases such as Alzheimer's (AD), Parkinson's (PD), and Huntington's diseases (HD), and dementia with Lewy bodies (DLB) may share pathogenic processes. This review aims to systematically assemble and compare findings in various biochemical pathways across these four dementias. PubMed and Google Scholar were screened for articles reporting on brain and biofluid measurements of metals and/or metabolites in AD, PD, HD, or DLB. Articles were assessed using specific a priori-defined inclusion and exclusion criteria. Of 284 papers identified, 198 met criteria for inclusion. Although varying coverage levels of metals and metabolites across diseases and tissues made comparison of many analytes impossible, several common findings were identified: elevated glucose in both brain tissue and biofluids of AD, PD, and HD cases; increased iron and decreased copper in AD, PD and HD brain tissue; and decreased uric acid in biofluids of AD and PD cases. Other analytes were found to differ between diseases or were otherwise not covered across all conditions. These findings indicate that disturbances in glucose and purine pathways may be common to AD, PD, and HD. However, standardisation of methodologies and better coverage in some areas - notably of DLB - are necessary to validate and extend these findings.
Collapse
|
33
|
Schreyer S, Klein C, Pfeffer A, Rasińska J, Stahn L, Knuth K, Abuelnor B, Panzel AEC, Rex A, Koch S, Hemmati-Sadeghi S, Steiner B. Chia seeds as a potential cognitive booster in the APP23 Alzheimer's disease model. Sci Rep 2020; 10:18215. [PMID: 33106576 PMCID: PMC7589531 DOI: 10.1038/s41598-020-75209-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023] Open
Abstract
Glucose hypometabolism potentially contributes to Alzheimer's disease (AD) and might even represent an underlying mechanism. Here, we investigate the relationship of diet-induced metabolic stress and AD as well as the therapeutic potential of chia seeds as a modulator of glucose metabolism in the APP23 mouse model. 4-6 (pre-plaque stage, PRE) and 28-32 (advanced-plaque stage, ADV) weeks old APP23 and wild type mice received pretreatment for 12 weeks with either sucrose-rich (SRD) or control diet, followed by 8 weeks of chia seed supplementation. Although ADV APP23 mice generally showed functioning glucose homeostasis, they were more prone to SRD-induced glucose intolerance. This was accompanied by elevated corticosterone levels and mild insulin insensitivity. Chia seeds improved spatial learning deficits but not impaired cognitive flexibility, potentially mediated by amelioration of glucose tolerance, attenuation of corticosterone levels and reversal of SRD-induced elevation of pro-inflammatory cytokine levels. Since cognitive symptoms and plaque load were not aggravated by SRD-induced metabolic stress, despite enhanced neuroinflammation in the PRE group, we conclude that impairments of glucose metabolism do not represent an underlying mechanism of AD in this mouse model. Nevertheless, chia seeds might provide therapeutic potential in AD as shown by the amelioration of cognitive symptoms.
Collapse
Affiliation(s)
- Stefanie Schreyer
- Department of Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.
| | - Charlotte Klein
- Department of Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Anna Pfeffer
- Department of Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Justyna Rasińska
- Department of Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Laura Stahn
- Department of Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Karlotta Knuth
- Department of Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Basim Abuelnor
- Department of Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Alina Elisabeth Catharina Panzel
- Department of Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - André Rex
- Department of Experimental Neurology and Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Stefan Koch
- Department of Experimental Neurology and Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
- NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Shabnam Hemmati-Sadeghi
- Department of Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Barbara Steiner
- Department of Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|
34
|
Morrice N, van Aalten L, McNeilly A, McCrimmon RJ, Pearson ER, Langston R, Sutherland C. Reducing Glut2 throughout the body does not result in cognitive behaviour differences in aged male mice. BMC Res Notes 2020; 13:438. [PMID: 32938474 PMCID: PMC7493158 DOI: 10.1186/s13104-020-05276-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/09/2020] [Indexed: 11/25/2022] Open
Abstract
Objectives GLUT2 is a major facilitative glucose transporter, expressed from the SLC2A2 gene, with essential roles in the liver. Recent work in mice has shown that preventing Glut2 production in specific neuronal populations increases sugar-seeking behaviour, highlighting the importance of Slc2a2 gene expression in the brain. It implies that reduced GLUT2 in the brain, due to genetic polymorphisms or disease, impacts health through behaviour change. Defects in glucose transport in the brain are observed in conditions including type-2 diabetes and dementia. Few studies have directly examined the effect of modulating neuronal glucose transporter expression on cognitive function. The aim of this study was to investigate whether inactivating one Slc2a2 allele throughout the body had major effects on cognition. Cognitive tests to assess recognition memory, spatial working memory and anxiety were performed in Slc2a2 whole-body heterozygous mice (i.e. reduced Glut2 mRNA and protein), alongside littermates expressing normal levels of the transporter. Results No significant effects on neurological functions and cognitive capabilities were observed in mice lacking one Slc2a2 allele when fed a chow diet. This suggests that the minor variations in GLUT2 levels that occur in the human population are unlikely to influence behaviour and basic cognition.
Collapse
Affiliation(s)
- Nicola Morrice
- Division of Cellular Medicine, School of Medicine, University of Dundee, Ninewells Hospital and Medical School, James Arnott Drive, Dundee, DD1 9SY, UK
| | - Lidy van Aalten
- Division of Cellular Medicine, School of Medicine, University of Dundee, Ninewells Hospital and Medical School, James Arnott Drive, Dundee, DD1 9SY, UK
| | - Alison McNeilly
- Division of Systems Medicine, School of Medicine, University of Dundee, Ninewells Hospital and Medical School, James Arnott Drive, Dundee, DD1 9SY, UK
| | - Rory J McCrimmon
- Division of Systems Medicine, School of Medicine, University of Dundee, Ninewells Hospital and Medical School, James Arnott Drive, Dundee, DD1 9SY, UK
| | - Ewan R Pearson
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Ninewells Hospital and Medical School, James Arnott Drive, Dundee, DD1 9SY, UK
| | - Rosamund Langston
- Division of Systems Medicine, School of Medicine, University of Dundee, Ninewells Hospital and Medical School, James Arnott Drive, Dundee, DD1 9SY, UK
| | - Calum Sutherland
- Division of Cellular Medicine, School of Medicine, University of Dundee, Ninewells Hospital and Medical School, James Arnott Drive, Dundee, DD1 9SY, UK.
| |
Collapse
|
35
|
Chronic hyperglycemia impairs hippocampal neurogenesis and memory in an Alzheimer's disease mouse model. Neurobiol Aging 2020; 92:98-113. [PMID: 32417750 DOI: 10.1016/j.neurobiolaging.2020.04.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 03/25/2020] [Accepted: 04/06/2020] [Indexed: 12/30/2022]
Abstract
During aging, lifestyle-related factors shape the brain's response to insults and modulate the progression of neurodegenerative pathologies such as Alzheimer's disease (AD). This is the case for chronic hyperglycemia associated with type 2 diabetes, which reduces the brain's ability to handle the neurodegenerative burden associated with AD. However, the mechanisms behind the effects of chronic hyperglycemia in the context of AD are not fully understood. Here, we show that newly generated neurons in the hippocampal dentate gyrus of triple transgenic AD (3xTg-AD) mice present increased dendritic arborization and a number of synaptic puncta, which may constitute a compensatory mechanism allowing the animals to cope with a lower neurogenesis rate. Contrariwise, chronic hyperglycemia decreases the complexity and differentiation of 3xTg-AD newborn neurons and reduces the levels of β-catenin, a key intrinsic modulator of neuronal maturation. Moreover, synaptic facilitation is depressed in hyperglycemic 3xTg-AD mice, accompanying the defective hippocampal-dependent memory. Our data suggest that hyperglycemia evokes cellular and functional alterations that accelerate the onset of AD-related symptoms, namely memory impairment.
Collapse
|
36
|
Hong J, Kim Y, Yanpallewar S, Lin PC. The Rho/Rac Guanine Nucleotide Exchange Factor Vav1 Regulates Hif-1α and Glut-1 Expression and Glucose Uptake in the Brain. Int J Mol Sci 2020; 21:ijms21041341. [PMID: 32079227 PMCID: PMC7072975 DOI: 10.3390/ijms21041341] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/14/2020] [Accepted: 02/14/2020] [Indexed: 12/18/2022] Open
Abstract
Vav1 is a Rho/Rac (Ras-related C3 botulinum toxin substrate) guanine nucleotide exchange factor expressed in hematopoietic and endothelial cells that are involved in a wide range of cellular functions. It is also stabilized under hypoxic conditions when it regulates the accumulation of the transcription factor HIF (Hypoxia Inducible Factor)-1α, which activates the transcription of target genes to orchestrate a cellular response to low oxygen. One of the genes induced by HIF-1α is GLUT (Glucose Transporter)-1, which is the major glucose transporter expressed in vessels that supply energy to the brain. Here, we identify a role for Vav1 in providing glucose to the brain. We found that Vav1 deficiency downregulates HIF-1α and GLUT-1 levels in endothelial cells, including blood-brain barrier cells. This downregulation of GLUT-1, in turn, reduced glucose uptake to endothelial cells both in vitro and in vivo, and reduced glucose levels in the brain. Furthermore, endothelial cell-specific Vav1 knock-out in mice, which caused glucose uptake deficiency, also led to a learning delay in fear conditioning experiments. Our results suggest that Vav1 promotes learning by activating HIF-1α and GLUT-1 and thereby distributing glucose to the brain. We further demonstrate the importance of glucose transport by endothelial cells in brain functioning and reveal a potential new axis for targeting GLUT-1 deficiency syndromes and other related brain diseases.
Collapse
Affiliation(s)
- Jaewoo Hong
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA;
- Correspondence: (J.H.); (P.C.L.); Tel.: +1-301-846-6515 (J.H.); +1-301-228-4688 (P.C.L.)
| | - Yurim Kim
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA;
- Department of Biology, University of Maryland, College Park, MD 20742, USA
| | - Sudhirkumar Yanpallewar
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA;
| | - P. Charles Lin
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA;
- Correspondence: (J.H.); (P.C.L.); Tel.: +1-301-846-6515 (J.H.); +1-301-228-4688 (P.C.L.)
| |
Collapse
|
37
|
Gonçalves RA, Wijesekara N, Fraser PE, De Felice FG. Behavioral Abnormalities in Knockout and Humanized Tau Mice. Front Endocrinol (Lausanne) 2020; 11:124. [PMID: 32226410 PMCID: PMC7080660 DOI: 10.3389/fendo.2020.00124] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 02/24/2020] [Indexed: 12/30/2022] Open
Abstract
Microtubule-associated protein tau assists in stabilizing microtubules and has been particularly implicated in Alzheimer's disease (AD). Given the importance of tau to AD pathogenesis and therapies, it is important to understand non-classic physiological functions for this protein inside and outside the central nervous system (CNS). Our group has previously shown that tau ablation triggers glucose intolerance and pancreatic dysfunction in mice, suggesting that tau plays a role in peripheral metabolic regulation. Little is known about the role of tau in anxiety. Moreover, inconsistent results have been generated regarding the effects of tau deletion in memory. Here, we characterize systemic insulin resistance, anxiety-related behavior and memory in 15 to 20 weeks old Wild-Type (WT), Tau knockout (TauKO) and a distinct hTau mouse model consisting of tau knockout expressing the longest isoform (2N4R) of a non-mutant WT human Tau protein under the prion promoter (hTau). Our findings demonstrate that tau deletion leads to anxiety-related behavior, impaired contextual and cued fear memory. The presence of a human Tau transgene did not ameliorate the phenotypes observed in animals lacking the mouse tau protein and it elicited impairments in learning, memory, and peripheral insulin sensitivity. Our results suggest that tau protein plays a role in memory and anxiety-related behavior. Our findings also indicate that previously unrecognized functions for tau protein may be a complicating factor in using animal models on the TauKO background. Understanding the link between tau pathophysiology and cognitive and metabolic alterations is of great importance to establish the complete contribution of tau protein to AD pathogenesis.
Collapse
Affiliation(s)
- Rafaella Araujo Gonçalves
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Nadeeja Wijesekara
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Paul E. Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- *Correspondence: Paul E. Fraser
| | - Fernanda G. De Felice
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Psychiatry, Queen's University, Kingston, ON, Canada
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Fernanda G. De Felice
| |
Collapse
|
38
|
Sun Y, Ma C, Sun H, Wang H, Peng W, Zhou Z, Wang H, Pi C, Shi Y, He X. Metabolism: A Novel Shared Link between Diabetes Mellitus and Alzheimer's Disease. J Diabetes Res 2020; 2020:4981814. [PMID: 32083135 PMCID: PMC7011481 DOI: 10.1155/2020/4981814] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/29/2019] [Accepted: 01/14/2020] [Indexed: 12/14/2022] Open
Abstract
As a chronic metabolic disease, diabetes mellitus (DM) is broadly characterized by elevated levels of blood glucose. Novel epidemiological studies demonstrate that some diabetic patients have an increased risk of developing dementia compared with healthy individuals. Alzheimer's disease (AD) is the most frequent cause of dementia and leads to major progressive deficits in memory and cognitive function. Multiple studies have identified an increased risk for AD in some diabetic populations, but it is still unclear which diabetic patients will develop dementia and which biological characteristics can predict cognitive decline. Although few mechanistic metabolic studies have shown clear pathophysiological links between DM and AD, there are several plausible ways this may occur. Since AD has many characteristics in common with impaired insulin signaling pathways, AD can be regarded as a metabolic disease. We conclude from the published literature that the body's diabetic status under certain circumstances such as metabolic abnormalities can increase the incidence of AD by affecting glucose transport to the brain and reducing glucose metabolism. Furthermore, due to its plentiful lipid content and high energy requirement, the brain's metabolism places great demands on mitochondria. Thus, the brain may be more susceptible to oxidative damage than the rest of the body. Emerging evidence suggests that both oxidative stress and mitochondrial dysfunction are related to amyloid-β (Aβ) pathology. Protein changes in the unfolded protein response or endoplasmic reticulum stress can regulate Aβ production and are closely associated with tau protein pathology. Altogether, metabolic disorders including glucose/lipid metabolism, oxidative stress, mitochondrial dysfunction, and protein changes caused by DM are associated with an impaired insulin signal pathway. These metabolic factors could increase the prevalence of AD in diabetic patients via the promotion of Aβ pathology.
Collapse
Affiliation(s)
- Yanan Sun
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Cao Ma
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
- Department of Pathology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Hui Sun
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Huan Wang
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Wei Peng
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
- Department of Clinical Medicine, Jilin University, Changchun 130021, China
| | - Zibo Zhou
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
- Department of Clinical Medicine, Jilin University, Changchun 130021, China
| | - Hongwei Wang
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
- Department of Clinical Medicine, Jilin University, Changchun 130021, China
| | - Chenchen Pi
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
- The First Hospital, Jilin University, Changchun, Jilin 130021, China
| | - Yingai Shi
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Xu He
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| |
Collapse
|
39
|
Kumari A, Sharma R, Shrivastava N, Somvanshi P, Grover A. Bleomycin modulates amyloid aggregation in β-amyloid and hIAPP. RSC Adv 2020; 10:25929-25946. [PMID: 35518630 PMCID: PMC9055351 DOI: 10.1039/d0ra04949b] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 06/29/2020] [Indexed: 12/06/2022] Open
Abstract
Aberrant misfolding and amyloid aggregation, which result in amyloid fibrils, are frequent and critical pathological incidents in various neurodegenerative disorders. Multiple drugs or inhibitors have been investigated to avert amyloid aggregation in individual peptides, exhibiting sequence-dependent inhibition mechanisms. Establishing or inventing inhibitors capable of preventing amyloid aggregation in a wide variety of amyloid peptides is quite a daunting task. Bleomycin (BLM), a complex glycopeptide, has been widely used as an antibiotic and antitumor drug due to its ability to inhibit DNA metabolism, and as an antineoplastic, especially for solid tumors. In this study, we investigated the dual inhibitory effects of BLM on Aβ aggregation, associated with Alzheimer's disease and hIAPP, which is linked to type 2 diabetes, using both computational and experimental techniques. Combined results from drug repurposing and replica exchange molecular dynamics simulations demonstrate that BLM binds to the β-sheet region considered a hotspot for amyloid fibrils of Aβ and hIAPP. BLM was also found to be involved in β-sheet destabilization and, ultimately, in its reduction. Further, experimental validation through in vitro amyloid aggregation assays was obtained wherein the fibrillar load was decreased for the BLM-treated Aβ and hIAPP peptides in comparison to controls. For the first time, this study shows that BLM is a dual inhibitor of Aβ and hIAPP amyloid aggregation. In the future, the conformational optimization and processing of BLM may help develop various efficient sequence-dependent inhibitors against amyloid aggregation in various amyloid peptides. Bleomycin acts as a dual inhibitor against both amyloid β and human islet amyloid polypeptide by binding to the β-sheet grooves considered as the amyloids hotspot.![]()
Collapse
Affiliation(s)
- Anchala Kumari
- Department of Biotechnology
- Teri School of Advanced Studies
- New Delhi
- India
- School of Biotechnology
| | - Ritika Sharma
- School of Biotechnology
- Jawaharlal Nehru University
- New Delhi
- India
| | | | - Pallavi Somvanshi
- Department of Biotechnology
- Teri School of Advanced Studies
- New Delhi
- India
| | - Abhinav Grover
- School of Biotechnology
- Jawaharlal Nehru University
- New Delhi
- India
| |
Collapse
|
40
|
Smith PJ. Pathways of Prevention: A Scoping Review of Dietary and Exercise Interventions for Neurocognition. Brain Plast 2019; 5:3-38. [PMID: 31970058 PMCID: PMC6971820 DOI: 10.3233/bpl-190083] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease and related dementias (ADRD) represent an increasingly urgent public health concern, with an increasing number of baby boomers now at risk. Due to a lack of efficacious therapies among symptomatic older adults, an increasing emphasis has been placed on preventive measures that can curb or even prevent ADRD development among middle-aged adults. Lifestyle modification using aerobic exercise and dietary modification represents one of the primary treatment modalities used to mitigate ADRD risk, with an increasing number of trials demonstrating that exercise and dietary change, individually and together, improve neurocognitive performance among middle-aged and older adults. Despite several optimistic findings, examination of treatment changes across lifestyle interventions reveals a variable pattern of improvements, with large individual differences across trials. The present review attempts to synthesize available literature linking lifestyle modification to neurocognitive changes, outline putative mechanisms of treatment improvement, and discuss discrepant trial findings. In addition, previous mechanistic assumptions linking lifestyle to neurocognition are discussed, with a focus on potential solutions to improve our understanding of individual neurocognitive differences in response to lifestyle modification. Specific recommendations include integration of contemporary causal inference approaches for analyzing parallel mechanistic pathways and treatment-exposure interactions. Methodological recommendations include trial multiphase optimization strategy (MOST) design approaches that leverage individual differences for improved treatment outcomes.
Collapse
Affiliation(s)
- Patrick J. Smith
- Department of Psychiatry and Behavioral Sciences (Primary), Duke University Medical Center, NC, USA
- Department of Medicine (Secondary), Duke University Medical Center, NC, USA
- Department of Population Health Sciences (Secondary), Duke University, NC, USA
| |
Collapse
|
41
|
Ettcheto M, Cano A, Manzine PR, Busquets O, Verdaguer E, Castro-Torres RD, García ML, Beas-Zarate C, Olloquequi J, Auladell C, Folch J, Camins A. Epigallocatechin-3-Gallate (EGCG) Improves Cognitive Deficits Aggravated by an Obesogenic Diet Through Modulation of Unfolded Protein Response in APPswe/PS1dE9 Mice. Mol Neurobiol 2019; 57:1814-1827. [PMID: 31838720 DOI: 10.1007/s12035-019-01849-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/27/2019] [Indexed: 12/31/2022]
Abstract
Epigallocatechin-3-gallate (EGCG), a catechin found in green tea, has been previously investigated for its neuroprotective effects in vitro and in vivo. In the present study, we aimed to evaluate its possible beneficial effects in a well-established preclinical mixed model of familial Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM) based on the use of transgenic APPswe/PS1dE9 (APP/PS1) mice fed with a high fat diet (HFD). C57BL/6 wild-type (WT) and APP/PS1 mice were used in this study. APP/PS1 mice were fed with a palmitic acid-enriched HFD (APP/PS1 HFD) containing 45% of fat mainly from hydrogenated coconut oil. Intraperitoneal glucose tolerance tests (IP-GTT) and insulin tolerance tests (IP-ITT) were performed. Western blot analyses were performed to analyse protein expression, and water maze and novel object recognition test were done to evaluate the cognitive process. EGCG treatment improves peripheral parameters such as insulin sensitivity or liver insulin pathway signalling, as well as central memory deficits. It also markedly increased synaptic markers and cAMP response element binding (CREB) phosphorylation rates, as a consequence of a decrease in the unfolded protein response (UPR) activation through the reduction in the activation factor 4 (ATF4) levels and posterior downregulation of protein tyrosine phosphatase 1B (PTP1B). Moreover, EGCG significantly decreased brain amyloid β (Aβ) production and plaque burden by increasing the levels of α-secretase (ADAM10). Also, it led to a reduction in neuroinflammation, as suggested by the decrease in astrocyte reactivity and toll-like receptor 4 (TLR4) levels. Collectively, evidence suggests that chronic EGCG prevents distinct neuropathological AD-related hallmarks. This study also provides novel insights into the metabolic and neurobiological mechanisms of EGCG against cognitive loss through its effects on UPR function, suggesting that this compound may be a promising disease-modifying treatment for neurodegenerative diseases.
Collapse
Affiliation(s)
- Miren Ettcheto
- Departament of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Department of Biochemistry and Biotechnology, Faculty of Medicine and Life Science, University Rovira i Virgili, Reus, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Amanda Cano
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain.,Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
| | - Patricia R Manzine
- Department of Gerontology, Federal University of São Carlos (UFSCar), São Carlos, 13565-905, Brazil
| | - Oriol Busquets
- Departament of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Department of Biochemistry and Biotechnology, Faculty of Medicine and Life Science, University Rovira i Virgili, Reus, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Ester Verdaguer
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Rubén Dario Castro-Torres
- Departament of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Department of Biochemistry and Biotechnology, Faculty of Medicine and Life Science, University Rovira i Virgili, Reus, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain.,Department of Cellular and Molecular Biology, Neuroscience Division, C.U.C.B.A., University of Guadalajara, Sierra Mojada, Col. Independencia, Guadalajara, Jalisco, México
| | - Maria Luisa García
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain.,Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
| | - Carlos Beas-Zarate
- Department of Cellular and Molecular Biology, Neuroscience Division, C.U.C.B.A., University of Guadalajara, Sierra Mojada, Col. Independencia, Guadalajara, Jalisco, México
| | - Jordi Olloquequi
- Laboratory of Cellular and Molecular Pathology, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | - Carme Auladell
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Jaume Folch
- Department of Biochemistry and Biotechnology, Faculty of Medicine and Life Science, University Rovira i Virgili, Reus, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Antoni Camins
- Departament of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain. .,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain. .,Institute of Neuroscience, University of Barcelona, Barcelona, Spain. .,Laboratory of Cellular and Molecular Pathology, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile. .,Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Av. Joan XXIII 27/31, E-08028, Barcelona, Spain.
| |
Collapse
|
42
|
Bianchi VE, Herrera PF, Laura R. Effect of nutrition on neurodegenerative diseases. A systematic review. Nutr Neurosci 2019; 24:810-834. [PMID: 31684843 DOI: 10.1080/1028415x.2019.1681088] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Neurodegenerative diseases are characterized by the progressive functional loss of neurons in the brain, causing cognitive impairment and motoneuron disability. Although multifactorial interactions are evident, nutrition plays an essential role in the pathogenesis and evolution of these diseases. A systematic literature search was performed, and the prevalence of studies evaluated the effect of the Mediterranean diet (MeDiet), nutritional support, EPA and DHA, and vitamins on memory and cognition impairment. The data showed that malnutrition and low body mass index (BMI) is correlated with the higher development of dementia and mortality. MeDiet, nutritional support, and calorie-controlled diets play a protective effect against cognitive decline, Alzheimer's disease (AD), Parkinson disease (PD) while malnutrition and insulin resistance represent significant risk factors. Malnutrition activates also the gut-microbiota-brain axis dysfunction that exacerbate neurogenerative process. Omega-3 and -6, and the vitamins supplementation seem to be less effective in protecting neuron degeneration. Insulin activity is a prevalent factor contributing to brain health while malnutrition correlated with the higher development of dementia and mortality.
Collapse
Affiliation(s)
| | - Pomares Fredy Herrera
- Director del Centro de Telemedicina, Grupo de investigación en Atención Primaria en salud/Telesalud, Doctorado en Medicina /Neurociencias, University of Cartagena, Colombia
| | - Rizzi Laura
- Molecular Biology, School of Medicine and Surgery, University of Milano-Bicocca, Monza Brianza, Italy
| |
Collapse
|
43
|
L’îlot pancréatique : ce que nous savons 150 ans après Langerhans. BULLETIN DE L'ACADÉMIE NATIONALE DE MÉDECINE 2019. [DOI: 10.1016/j.banm.2019.06.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
44
|
Zhang S, Xue R, Hu R. The neuroprotective effect and action mechanism of polyphenols in diabetes mellitus-related cognitive dysfunction. Eur J Nutr 2019; 59:1295-1311. [PMID: 31598747 DOI: 10.1007/s00394-019-02078-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 08/10/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Diabetes mellitus (DM) is a complex and prevalent metabolic disorder worldwide. Strong evidence has emerged that DM is a risk factor for the accelerated rate of cognitive decline and the development of dementia. Though traditional pharmaceutical agents are efficient for the management of DM and DM-related cognitive decrement, long-term use of these drugs are along with undesired side effects. Therefore, tremendous studies have focused on the therapeutic benefits of natural compounds at present. Ample evidence exists to prove that polyphenols are capable to modulate diabetic neuropathy with minimal toxicity and adverse effects. PURPOSE To describe the benefits and mechanisms of polyphenols on DM-induced cognitive dysfunction. In this review, we introduce an updated overview of associations between DM and cognitive dysfunction. The risk factors as well as pathological and molecular mechanisms of DM-induced cognitive dysfunction are summarized. More importantly, many active polyphenols that possess preventive and therapeutic effects on DM-induced cognitive dysfunction and the potential signaling pathways involved in the action are highlighted. CONCLUSIONS The therapeutic effects of polyphenols on DM-related cognitive dysfunction pave a novel way for the management of diabetic encephalopathy.
Collapse
Affiliation(s)
- Shenshen Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, China.
| | - Ran Xue
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Ruizhe Hu
- School of Physical Education (Main Campus), Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
45
|
Martinez-Valbuena I, Valenti-Azcarate R, Amat-Villegas I, Riverol M, Marcilla I, de Andrea CE, Sánchez-Arias JA, Del Mar Carmona-Abellan M, Marti G, Erro ME, Martínez-Vila E, Tuñon MT, Luquin MR. Amylin as a potential link between type 2 diabetes and alzheimer disease. Ann Neurol 2019; 86:539-551. [PMID: 31376172 DOI: 10.1002/ana.25570] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Alzheimer disease (AD) is the leading cause of dementia, and although its etiology remains unclear, it seems that type 2 diabetes mellitus (T2DM) and other prediabetic states of insulin resistance could contribute to the appearance of sporadic AD. As such, we have assessed whether tau and β-amyloid (Aβ) deposits might be present in pancreatic tissue of subjects with AD, and whether amylin, an amyloidogenic protein deposited in the pancreas of T2DM patients, might accumulate in the brain of AD patients. METHODS We studied pancreatic and brain tissue from 48 individuals with no neuropathological alterations and from 87 subjects diagnosed with AD. We examined Aβ and tau accumulation in the pancreas as well as that of amylin in the brain. Moreover, we performed proximity ligation assays to ascertain whether tau and/or Aβ interact with amylin in either the pancreas or brain of these subjects. RESULTS Cytoplasmic tau and Aβ protein deposits were detected in pancreatic β cells of subjects with AD as well as in subjects with a normal neuropathological examination but with a history of T2DM and in a small cohort of control subjects without T2DM. Furthermore, we found amylin deposits in the brain of these subjects, providing histological evidence that amylin can interact with Aβ and tau in both the pancreas and hippocampus. INTERPRETATION The presence of both tau and Aβ inclusions in pancreatic β cells, and of amylin deposits in the brain, provides new evidence of a potential overlap in the mechanisms underlying the pathogenesis of T2DM and AD. ANN NEUROL 2019;86:539-551.
Collapse
Affiliation(s)
- Ivan Martinez-Valbuena
- Neurology Department, Clínica Universidad de Navarra, Pamplona, Spain
- Regenerative Therapy Laboratory, Neurosciences Division, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
- Navarra Health Research Institute, Pamplona, Spain
| | - Rafael Valenti-Azcarate
- Neurology Department, Clínica Universidad de Navarra, Pamplona, Spain
- Regenerative Therapy Laboratory, Neurosciences Division, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
- Navarra Health Research Institute, Pamplona, Spain
| | - Irene Amat-Villegas
- Navarra Health Research Institute, Pamplona, Spain
- Pathology Department, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Mario Riverol
- Neurology Department, Clínica Universidad de Navarra, Pamplona, Spain
- Navarra Health Research Institute, Pamplona, Spain
| | - Irene Marcilla
- Regenerative Therapy Laboratory, Neurosciences Division, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
- Navarra Health Research Institute, Pamplona, Spain
| | | | - Juan Antonio Sánchez-Arias
- Navarra Health Research Institute, Pamplona, Spain
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Maria Del Mar Carmona-Abellan
- Neurology Department, Clínica Universidad de Navarra, Pamplona, Spain
- Regenerative Therapy Laboratory, Neurosciences Division, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
- Navarra Health Research Institute, Pamplona, Spain
| | - Gloria Marti
- Neurology Department, Clínica Universidad de Navarra, Pamplona, Spain
- Navarra Health Research Institute, Pamplona, Spain
| | - Maria-Elena Erro
- Navarra Health Research Institute, Pamplona, Spain
- Neurology Department, Navarra Hospital Complex, Pamplona, Spain
| | - Eduardo Martínez-Vila
- Neurology Department, Clínica Universidad de Navarra, Pamplona, Spain
- Navarra Health Research Institute, Pamplona, Spain
| | - Maria-Teresa Tuñon
- Navarra Health Research Institute, Pamplona, Spain
- Pathology Department, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Maria-Rosario Luquin
- Neurology Department, Clínica Universidad de Navarra, Pamplona, Spain
- Regenerative Therapy Laboratory, Neurosciences Division, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
- Navarra Health Research Institute, Pamplona, Spain
| |
Collapse
|
46
|
Liu X, Wang W, Chen HL, Zhang HY, Zhang NX. Interplay between Alzheimer's disease and global glucose metabolism revealed by the metabolic profile alterations of pancreatic tissue and serum in APP/PS1 transgenic mice. Acta Pharmacol Sin 2019; 40:1259-1268. [PMID: 31089202 DOI: 10.1038/s41401-019-0239-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 04/11/2019] [Indexed: 12/20/2022] Open
Abstract
Increasing evidence suggests that there is a correlation between type 2 diabetes mellitus (T2D) and Alzheimer's disease (AD). Increased Aβ polypeptide production in AD patients would promote metabolic abnormalities, insulin signaling dysfunction and perturbations in glucose utilization, thus leading to the onset of T2D. However, the metabolic mechanisms underlying the interplay between AD and its diabetes-promoting effects are not fully elucidated. Particularly, systematic metabolomics analysis has not been performed for the pancreas tissues of AD subjects, which play key roles in the glucose metabolism of living systems. In the current study, we characterized the dynamic metabolic profile alterations of the serum and the pancreas of APP/PS1 double-transgenic mice (an AD mouse model) using the untargeted metabolomics approaches. Serum and pancreatic tissues of APP/PS1 transgenic mice and wild-type mice were extracted and subjected to NMR analysis to evaluate the functional state of pancreas in the progress of AD. Multivariate analysis of principal component analysis (PCA) and orthogonal partial least squares discriminant analysis (OPLS-DA) were conducted to define the global and the local (pancreas) metabolic features associated with the possible initiation of T2D in the progress of AD. Our results showed the onset of AD-induced global glucose metabolism disorders in AD mice. Hyperglycemia and its accompanying metabolic disorders including energy metabolism down-regulation and oxidative stress were observed in the serum of AD mice. Meanwhile, global disturbance of branched-chain amino acid (BCAA) metabolism was detected, and the change of BCAA (leucine) was positively correlated to the alteration of glucose. Moreover, increased level of glucose and enhanced energy metabolism were observed in the pancreas of AD mice. The results suggest that the diabetes-promoting effects accompanying the progress of AD are achieved by down-regulating the global utilization of glucose and interfering with the metabolic function of pancreas. Since T2D is a risk factor for the pathogenesis of AD, our findings suggest that targeting the glucose metabolism dysfunctions might serve as a supplementary therapeutic strategy for Alzheimer's disease.
Collapse
|
47
|
Gregosa A, Vinuesa Á, Todero MF, Pomilio C, Rossi SP, Bentivegna M, Presa J, Wenker S, Saravia F, Beauquis J. Periodic dietary restriction ameliorates amyloid pathology and cognitive impairment in PDAPP-J20 mice: Potential implication of glial autophagy. Neurobiol Dis 2019; 132:104542. [PMID: 31351172 DOI: 10.1016/j.nbd.2019.104542] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/30/2019] [Accepted: 07/22/2019] [Indexed: 12/29/2022] Open
Abstract
Dietary restriction promotes cell regeneration and stress resistance in multiple models of human diseases. One of the conditions that could potentially benefit from this strategy is Alzheimer's disease, a chronic, progressive and prevalent neurodegenerative disease. Although there are no effective pharmacological treatments for this pathology, lifestyle interventions could play therapeutic roles. Our objectives were 1) to evaluate the effects of dietary restriction on cognition, hippocampal amyloid deposition, adult neurogenesis and glial reactivity and autophagy in a mouse model of familial Alzheimer's disease, and 2) to analyze the role of glial cells mediating the effects of nutrient restriction in an in vitro model. Therefore, we established a periodic dietary restriction protocol in adult female PDAPP-J20 transgenic mice for 6 weeks. We found that dietary restriction, not involving overall caloric restriction, attenuated cognitive deficits, amyloid pathology and microglial reactivity in transgenic mice when compared with ad libitum-fed transgenic animals. Also, transgenic mice showed an increase in the astroglial positive signal for LC3, an autophagy-associated protein. In parallel, hippocampal adult neurogenesis was decreased in transgenic mice whereas dietary-restricted transgenic mice showed a neurogenic status similar to controls. In vitro experiments showed that nutrient restriction decreased astroglial and, indirectly, microglial NFκB activation in response to amyloid β peptides. Furthermore, nutrient restriction was able to preserve astroglial autophagic flux and to decrease intracellular amyloid after exposure to amyloid β peptides. Our results suggest neuroprotective effects of nutrient restriction in Alzheimer's disease, with modulation of glial activation and autophagy being potentially involved pathways.
Collapse
Affiliation(s)
- Amal Gregosa
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina; Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina; Cátedra de Anatomía e Histología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina
| | - Ángeles Vinuesa
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina; Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina
| | - María Florencia Todero
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX), CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Carlos Pomilio
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina; Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina
| | - Soledad P Rossi
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina; Cátedra de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Melisa Bentivegna
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina; Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina
| | - Jessica Presa
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina; Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina
| | - Shirley Wenker
- Fundación Instituto Leloir-IIBA, CONICET, Buenos Aires, Argentina
| | - Flavia Saravia
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina; Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina
| | - Juan Beauquis
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina; Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina.
| |
Collapse
|
48
|
Movassat J, Delangre E, Liu J, Gu Y, Janel N. Hypothesis and Theory: Circulating Alzheimer's-Related Biomarkers in Type 2 Diabetes. Insight From the Goto-Kakizaki Rat. Front Neurol 2019; 10:649. [PMID: 31293498 PMCID: PMC6606723 DOI: 10.3389/fneur.2019.00649] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 06/03/2019] [Indexed: 12/16/2022] Open
Abstract
Epidemiological data suggest an increased risk of developing Alzheimer's disease (AD) in individuals with type 2 diabetes (T2D). AD is anatomically associated with an early progressive accumulation of Aβ leading to a gradual Tau hyperphosphorylation, which constitute the main characteristics of damaged brain in AD. Apart from these processes, mounting evidence suggests that specific features of diabetes, namely impaired glucose metabolism and insulin signaling in the brain, play a key role in AD. Moreover, several studies report a potential role of Aβ and Tau in peripheral tissues such as pancreatic β cells. Thus, it appears that several biological pathways associated with diabetes overlap with AD. The link between peripheral insulin resistance and brain insulin resistance with concomitant cognitive impairment may also potentially be mediated by a liver/pancreatic/brain axis, through the excessive trafficking of neurotoxic molecules across the blood-brain barrier. Insulin resistance incites inflammation and pro-inflammatory cytokine activation modulates the homocysteine cycle in T2D patients. Elevated plasma homocysteine level is a risk factor for AD pathology and is also closely associated with metabolic syndrome. We previously demonstrated a strong association between homocysteine metabolism and insulin via cystathionine beta synthase (CBS) activity, the enzyme implicated in the first step of the trans-sulfuration pathway, in Goto-Kakizaki (GK) rats, a spontaneous model of T2D, with close similarities with human T2D. CBS activity is also correlated with DYRK1A, a serine/threonine kinase regulating brain-derived neurotrophic factor (BDNF) levels, and Tau phosphorylation, which are implicated in a wide range of disease such as T2D and AD. We hypothesized that DYRK1A, BDNF, and Tau, could be among molecular factors linking T2D to AD. In this focused review, we briefly examine the main mechanisms linking AD to T2D and provide the first evidence that certain circulating AD biomarkers are found in diabetic GK rats. We propose that the spontaneous model of T2D in GK rat could be a suitable model to investigate molecular mechanisms linking T2D to AD.
Collapse
Affiliation(s)
- Jamileh Movassat
- Univ Paris Diderot-Sorbonne Paris Cité, Laboratoire de Biologie et Pathologie du Pancréas Endocrine, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251 CNRS, Paris, France
| | - Etienne Delangre
- Univ Paris Diderot-Sorbonne Paris Cité, Laboratoire de Biologie et Pathologie du Pancréas Endocrine, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251 CNRS, Paris, France
| | - Junjun Liu
- Univ Paris Diderot-Sorbonne Paris Cité, Laboratoire de Biologie et Pathologie du Pancréas Endocrine, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251 CNRS, Paris, France
| | - YuChen Gu
- Univ Paris Diderot-Sorbonne Paris Cité, Laboratoire Processus Dégénératifs, Stress et Vieillissement, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251 CNRS, Paris, France
| | - Nathalie Janel
- Univ Paris Diderot-Sorbonne Paris Cité, Laboratoire Processus Dégénératifs, Stress et Vieillissement, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251 CNRS, Paris, France
| |
Collapse
|
49
|
Kulas JA, Franklin WF, Smith NA, Manocha GD, Puig KL, Nagamoto-Combs K, Hendrix RD, Taglialatela G, Barger SW, Combs CK. Ablation of amyloid precursor protein increases insulin-degrading enzyme levels and activity in brain and peripheral tissues. Am J Physiol Endocrinol Metab 2019; 316:E106-E120. [PMID: 30422705 PMCID: PMC6417684 DOI: 10.1152/ajpendo.00279.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The amyloid precursor protein (APP) is a type I transmembrane glycoprotein widely studied for its role as the source of β-amyloid peptide, accumulation of which is causal in at least some cases of Alzheimer's disease (AD). APP is expressed ubiquitously and is involved in diverse biological processes. Growing bodies of evidence indicate connections between AD and somatic metabolic disorders related to type 2 diabetes, and App-/- mice show alterations in glycemic regulation. We find that App-/- mice have higher levels of insulin-degrading enzyme (IDE) mRNA, protein, and activity compared with wild-type controls. This regulation of IDE by APP was widespread across numerous tissues, including liver, skeletal muscle, and brain as well as cell types within neural tissue, including neurons, astrocytes, and microglia. RNA interference-mediated knockdown of APP in the SIM-A9 microglia cell line elevated IDE levels. Fasting levels of blood insulin were lower in App-/- than App+/+ mice, but the former showed a larger increase in response to glucose. These low basal levels may enhance peripheral insulin sensitivity, as App-/- mice failed to develop impairment of glucose tolerance on a high-fat, high-sucrose ("Western") diet. Insulin levels and insulin signaling were also lower in the App-/- brain; synaptosomes prepared from App-/- hippocampus showed diminished insulin receptor phosphorylation compared with App+/+ mice when stimulated ex vivo. These findings represent a new molecular link connecting APP to metabolic homeostasis and demonstrate a novel role for APP as an upstream regulator of IDE in vivo.
Collapse
Affiliation(s)
- Joshua A Kulas
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences , Grand Forks, North Dakota
| | - Whitney F Franklin
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch , Galveston, Texas
| | - Nicholas A Smith
- Department of Pathology, University of North Dakota School of Medicine and Health Sciences , Grand Forks, North Dakota
| | - Gunjan D Manocha
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences , Grand Forks, North Dakota
| | - Kendra L Puig
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences , Grand Forks, North Dakota
| | - Kumi Nagamoto-Combs
- Department of Pathology, University of North Dakota School of Medicine and Health Sciences , Grand Forks, North Dakota
| | - Rachel D Hendrix
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences , Little Rock Arkansas
| | - Giulio Taglialatela
- Department of Neurology, University of Texas Medical Branch , Galveston, Texas
| | - Steven W Barger
- Department of Geriatrics, University of Arkansas for Medical Sciences , Little Rock Arkansas
- Geriatric Research, Education and Clinical Center, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas
| | - Colin K Combs
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences , Grand Forks, North Dakota
| |
Collapse
|
50
|
Ettcheto M, Busquets O, Camins A. Potential preventive disease-modifying pharmacological strategies to delay late onset Alzheimer's disease. Neural Regen Res 2019; 14:1721-1725. [PMID: 31169189 PMCID: PMC6585538 DOI: 10.4103/1673-5374.257513] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease that was histopathologically characterized in the brain by the presence of extracellular senile plaques made of amyloid β peptides and intracellular neurofibrillary tangles composed of hyperphosphorylated Tau protein. Over the years, AD has been classified in two subgroups: early onset or familial AD and late onset or sporadic AD. On the one hand, familial AD has been described to be the result of genetic mutations that cause, in some cases, for the overproduction of amyloid β. On the other, the cause of late onset or sporadic AD is still unclear even though several hypotheses have been proposed to explain the process of severe and progressive memory and cognitive loss. In the present review, some of the current hypotheses that try to explain the origin of late onset or sporadic AD have been summarized. Also, their potential implication in the development of new drugs for the presymptomatic treatment of late onset or sporadic AD has been considered.
Collapse
Affiliation(s)
- Miren Ettcheto
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona; Departament de Bioquímica i Biotecnologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus; Institut de Neurociències, Universitat de Barcelona, Barcelona; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Oriol Busquets
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona; Departament de Bioquímica i Biotecnologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus; Institut de Neurociències, Universitat de Barcelona, Barcelona; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Antoni Camins
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona; Institut de Neurociències, Universitat de Barcelona, Barcelona; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| |
Collapse
|