1
|
Capatina TF, Oatu A, Babasan C, Trifu S. Translating Molecular Psychiatry: From Biomarkers to Personalized Therapies-A Narrative Review. Int J Mol Sci 2025; 26:4285. [PMID: 40362522 PMCID: PMC12072283 DOI: 10.3390/ijms26094285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/10/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
In this review, we explore the biomarkers of different psychiatric disorders, such as major depressive disorder, generalized anxiety disorder, schizophrenia, and bipolar disorder. Moreover, we show the interplay between genetic and environmental factors. Novel techniques such as genome-wide association studies (GWASs) have identified numerous risk loci and single-nucleotide polymorphisms (SNPs) implicated in these conditions, contributing to a better understanding of their mechanisms. Moreover, the impact of genetic variations on drug metabolisms, particularly through cytochrome P450 (CYP450) enzymes, highlights the importance of pharmacogenomics in optimizing psychiatric treatment. This review also explores the role of neurotransmitter regulation, immune system interactions, and metabolic pathways in psychiatric disorders. As the technology advances, integrating genetic markers into clinical practice will be crucial in advancing precision psychiatry, improving diagnostic accuracy and therapeutic interventions for individual patients.
Collapse
Affiliation(s)
| | - Anamaria Oatu
- Department of Psychiatry, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.O.); (C.B.)
| | - Casandra Babasan
- Department of Psychiatry, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.O.); (C.B.)
| | - Simona Trifu
- Department of Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
2
|
Maggiore A, Latina V, D'Erme M, Amadoro G, Coccurello R. Non-canonical pathways associated to Amyloid beta and tau protein dyshomeostasis in Alzheimer's disease: A narrative review. Ageing Res Rev 2024; 102:102578. [PMID: 39542177 DOI: 10.1016/j.arr.2024.102578] [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: 09/18/2024] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Alzheimer's Disease (AD) is the most common form of dementia among elderly people. This disease imposes a significant burden on the healthcare system, society, and economy due to the increasing global aging population. Current trials with drugs or bioactive compounds aimed at reducing cerebral Amyloid beta (Aβ) plaques and tau protein neurofibrillary tangles, which are the two main hallmarks of this devastating neurodegenerative disease, have not provided significant results in terms of their neuropathological outcomes nor met the expected clinical end-points. Ageing, genetic and environmental risk factors, along with different clinical symptoms suggest that AD is a complex and heterogeneous disorder with multiple interconnected pathological pathways rather than a single disease entity. In the present review, we highlight and discuss various non-canonical, Aβ-independent mechanisms, like gliosis, unhealthy dietary intake, lipid and sugar signaling, and cerebrovascular damage that contribute to the onset and development of AD. We emphasize that challenging the traditional "amyloid cascade hypothesis" may improve our understanding of this age-related complex syndrome and help fight the progressive cognitive decline in AD.
Collapse
Affiliation(s)
- Anna Maggiore
- Department of Biochemical Sciences, Sapienza University, P.le Aldo Moro 5, Rome 00185, Italy; Department of Brain Sciences, Imperial College, London, UK
| | - Valentina Latina
- European Brain Research Institute (EBRI), Viale Regina Elena 295, Rome 00161, Italy; Institute of Translational Pharmacology (IFT) CNR, Via Fosso del Cavaliere 100, Rome 00133, Italy
| | - Maria D'Erme
- Department of Biochemical Sciences, Sapienza University, P.le Aldo Moro 5, Rome 00185, Italy
| | - Giuseppina Amadoro
- European Brain Research Institute (EBRI), Viale Regina Elena 295, Rome 00161, Italy; Institute of Translational Pharmacology (IFT) CNR, Via Fosso del Cavaliere 100, Rome 00133, Italy.
| | - Roberto Coccurello
- Institute for Complex System (ISC) CNR, Via dei Taurini 19, Rome 00185, Italy; IRCSS Santa Lucia Foundation, European Center for Brain Research, Via Fosso del Fiorano 64-65, Rome 00143, Italy.
| |
Collapse
|
3
|
Johnson RJ, Tolan DR, Bredesen D, Nagel M, Sánchez-Lozada LG, Fini M, Burtis S, Lanaspa MA, Perlmutter D. Could Alzheimer's disease be a maladaptation of an evolutionary survival pathway mediated by intracerebral fructose and uric acid metabolism? Am J Clin Nutr 2023; 117:455-466. [PMID: 36774227 PMCID: PMC10196606 DOI: 10.1016/j.ajcnut.2023.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/21/2022] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
An important aspect of survival is to assure enough food, water, and oxygen. Here, we describe a recently discovered response that favors survival in times of scarcity, and it is initiated by either ingestion or production of fructose. Unlike glucose, which is a source for immediate energy needs, fructose metabolism results in an orchestrated response to encourage food and water intake, reduce resting metabolism, stimulate fat and glycogen accumulation, and induce insulin resistance as a means to reduce metabolism and preserve glucose supply for the brain. How this survival mechanism affects brain metabolism, which in a resting human amounts to 20% of the overall energy demand, is only beginning to be understood. Here, we review and extend a previous hypothesis that this survival mechanism has a major role in the development of Alzheimer's disease and may account for many of the early features, including cerebral glucose hypometabolism, mitochondrial dysfunction, and neuroinflammation. We propose that the pathway can be engaged in multiple ways, including diets high in sugar, high glycemic carbohydrates, and salt. In summary, we propose that Alzheimer's disease may be the consequence of a maladaptation to an evolutionary-based survival pathway and what had served to enhance survival acutely becomes injurious when engaged for extensive periods. Although more studies are needed on the role of fructose metabolism and its metabolite, uric acid, in Alzheimer's disease, we suggest that both dietary and pharmacologic trials to reduce fructose exposure or block fructose metabolism should be performed to determine whether there is potential benefit in the prevention, management, or treatment of this disease.
Collapse
Affiliation(s)
- Richard J Johnson
- Department of Medicine, Rocky Mountain VA Medical Center, Aurora, CO, USA; Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, CO, USA.
| | - Dean R Tolan
- Biology Department, Boston University, Boston, MA, USA
| | - Dale Bredesen
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Maria Nagel
- Department of Neurology, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - Laura G Sánchez-Lozada
- Department of Cardio-Renal Physiopathology, National Institute of Cardiology Ignacio Chávez, Mexico City, Mexico
| | - Mehdi Fini
- Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | | | - Miguel A Lanaspa
- Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | | |
Collapse
|
4
|
Díaz G, Lengele L, Sourdet S, Soriano G, de Souto Barreto P. Nutrients and amyloid β status in the brain: A narrative review. Ageing Res Rev 2022; 81:101728. [PMID: 36049590 DOI: 10.1016/j.arr.2022.101728] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/18/2022] [Accepted: 08/26/2022] [Indexed: 01/31/2023]
Abstract
Amyloid beta (Aβ) is a peptide and a hallmark of Alzheimer's disease (AD). Emerging evidence suggests that Aβ levels could be influenced by diet. However, the evidence is sparse and for some nutrients, controversial. The aim of this narrative review is to gather the findings of observational and clinical trials involving human participants on the relationships between nutrients and brain Aβ status. Some dietary patterns are associated to reduced levels of Aβ in the brain, such as the Mediterranean diet, ketogenic diet as well as low intake of saturated fat, high-glycemic-index food, sodium, and junk/fast food. Low Aβ status in the brain was also associated with higher density lipoproteins (HDL) cholesterol and polyunsaturated fatty acids consumption. Data on alcohol intake is not conclusive. On the contrary, high Aβ levels in the brain were related to a higher intake of total cholesterol, triglycerides, low-density lipoproteins (LDL) cholesterol, saturated fat, sucrose, and fructose. Folic acid, cobalamin, vitamin E, and vitamin D were not associated to Aβ status, while high blood concentrations of Calcium, Aluminum, Zinc, Copper, and Manganese were associated with decreased Aβ blood levels but were not associated with Aβ cerebral spinal fluid (CSF) concentrations. In conclusion, certain dietary patterns and nutrients are associated to brain Aβ status. Further research on the association between nutrients and brain Aβ status is needed in order to pave the way to use nutritional interventions as efficacious strategies to prevent Aβ disturbance and potentially AD.
Collapse
Affiliation(s)
- Gustavo Díaz
- Faculty of Medicine, Research Institute on Nutrition, Genetics, and Metabolism, Universidad El Bosque, Bogotá, Colombia; Research In Colombia Foundation, Bogotá, Colombia.
| | - Laetitia Lengele
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo Universitaire de Toulouse, France
| | - Sandrine Sourdet
- Gérontopôle, Department of Internal Medicine and Geriatrics, Toulouse University Hospital, La Cité de la Santé, Hôpital La Grave, Place Lange, Cedex 9, TSA 60033, Toulouse 31059, France
| | - Gaëlle Soriano
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo Universitaire de Toulouse, France; Gérontopôle, Department of Internal Medicine and Geriatrics, Toulouse University Hospital, La Cité de la Santé, Hôpital La Grave, Place Lange, Cedex 9, TSA 60033, Toulouse 31059, France
| | - Philipe de Souto Barreto
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo Universitaire de Toulouse, France; UPS/Inserm, CERPOP UMR1295, University of Toulouse III, Toulouse, France
| |
Collapse
|
5
|
Ullah R, Ali G, Baseer A, Irum Khan S, Akram M, Khan S, Ahmad N, Farooq U, Kanwal Nawaz N, Shaheen S, Kumari G, Ullah I. Tannic acid inhibits lipopolysaccharide-induced cognitive impairment in adult mice by targeting multiple pathological features. Int Immunopharmacol 2022; 110:108970. [DOI: 10.1016/j.intimp.2022.108970] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/05/2022] [Accepted: 06/13/2022] [Indexed: 11/16/2022]
|
6
|
Zhang X, Lei T, Wang D, Cai S, Hang Z, Yang Y, Bi W, Xiao Z, Du H. Stem cells from human exfoliated deciduous teeth relieves Alzheimer's disease symptoms in SAMP8 mice by up-regulating the PPARγ pathway. Biomed Pharmacother 2022; 152:113169. [PMID: 35689863 DOI: 10.1016/j.biopha.2022.113169] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/17/2022] [Accepted: 05/18/2022] [Indexed: 11/24/2022] Open
Abstract
The pathology of Alzheimer's disease (AD) is complex and heterogeneous, and there are currently no drugs that can stop its progression. The failure of traditional chemical small-molecule drug development showed the weakness of single target and made researchers look to cell therapy with multiple regulatory effects. Stem cells from human exfoliated deciduous teeth (SHED) are a kind of neural crest-derived mesenchymal stem cells which have broad prospects in the treatment of neurodegenerative diseases. In this study, we demonstrated the therapeutic effects of SHED in AD mice, including behavioral improvement, neuronal protection, and alleviation of neuroinflammation. Tracking experiments on SHED showed that some of the transplanted cells could enter the brain. To elucidate the role played by the majority of cells transplanted into veins, blood proteomic assays were performed. Data are available via ProteomeXchange with identifier PXD030313. Among the altered proteins, the PPAR pathway related to energy metabolism was considered to be an important signaling pathway involved in regulation through gene ontology analysis and pathway analysis. Western blot showed that the transplantation of SHED improved the glucose metabolism in AD mice by increasing the PPARγ signaling pathway. These results suggested that SHED have a potential in relieving AD pathological symptoms and improving behavioral cognition. The therapeutic mechanism of SHED is related to up-regulating PPARγ signaling pathway and reducing neuronal damage.
Collapse
Affiliation(s)
- Xiaoshuang Zhang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China
| | - Tong Lei
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China
| | - Donghui Wang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China
| | - Shanglin Cai
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China
| | - Zhongci Hang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China
| | - Yanjie Yang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China
| | - Wangyu Bi
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China
| | - Zhuangzhuang Xiao
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China
| | - Hongwu Du
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China.
| |
Collapse
|
7
|
Frausto DM, Forsyth CB, Keshavarzian A, Voigt RM. Dietary Regulation of Gut-Brain Axis in Alzheimer's Disease: Importance of Microbiota Metabolites. Front Neurosci 2021; 15:736814. [PMID: 34867153 PMCID: PMC8639879 DOI: 10.3389/fnins.2021.736814] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that impacts 45 million people worldwide and is ranked as the 6th top cause of death among all adults by the Centers for Disease Control and Prevention. While genetics is an important risk factor for the development of AD, environment and lifestyle are also contributing risk factors. One such environmental factor is diet, which has emerged as a key influencer of AD development/progression as well as cognition. Diets containing large quantities of saturated/trans-fats, refined carbohydrates, limited intake of fiber, and alcohol are associated with cognitive dysfunction while conversely diets low in saturated/trans-fats (i.e., bad fats), high mono/polyunsaturated fats (i.e., good fats), high in fiber and polyphenols are associated with better cognitive function and memory in both humans and animal models. Mechanistically, this could be the direct consequence of dietary components (lipids, vitamins, polyphenols) on the brain, but other mechanisms are also likely to be important. Diet is considered to be the single greatest factor influencing the intestinal microbiome. Diet robustly influences the types and function of micro-organisms (called microbiota) that reside in the gastrointestinal tract. Availability of different types of nutrients (from the diet) will favor or disfavor the abundance and function of certain groups of microbiota. Microbiota are highly metabolically active and produce many metabolites and other factors that can affect the brain including cognition and the development and clinical progression of AD. This review summarizes data to support a model in which microbiota metabolites influence brain function and AD.
Collapse
Affiliation(s)
- Dulce M. Frausto
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Christopher B. Forsyth
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Ali Keshavarzian
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
- Department of Physiology, Rush University Medical Center, Chicago, IL, United States
| | - Robin M. Voigt
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
8
|
Mohamed RA, Abdallah DM, El-brairy AI, Ahmed KA, El-Abhar HS. Palonosetron/Methyllycaconitine Deactivate Hippocampal Microglia 1, Inflammasome Assembly and Pyroptosis to Enhance Cognition in a Novel Model of Neuroinflammation. Molecules 2021; 26:5068. [PMID: 34443654 PMCID: PMC8401912 DOI: 10.3390/molecules26165068] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/10/2021] [Accepted: 08/17/2021] [Indexed: 12/30/2022] Open
Abstract
Since westernized diet-induced insulin resistance is a risk factor in Alzheimer's disease (AD) development, and lipopolysaccharide (LPS) coexists with amyloid β (Aβ)1-42 in these patients, our AD novel model was developed to resemble sporadic AD by injecting LPS into high fat/fructose diet (HFFD)-fed rats. The neuroprotective potential of palonosetron and/or methyllycaconitine, 5-HT3 receptor and α7 nAChR blockers, respectively, was evaluated after 8 days of daily administration in HFFD/LPS rats. All regimens improved histopathological findings and enhanced spatial memory (Morris Water Maze); however, palonosetron alone or with methyllycaconitine promoted animal performance during novel object recognition tests. In the hippocampus, all regimens reduced the expression of glial fibrillary acidic protein and skewed microglia M1 to M2 phenotype, indicated by the decreased M1 markers and the enhanced M2 related parameters. Additionally, palonosetron and its combination regimen downregulated the expression of ASC/TMS1, as well as levels of inflammasome downstream molecules and abated cleaved caspase-1, interleukin (IL)-1β, IL-18 and caspase-11. Furthermore, ACh and 5-HT were augmented after being hampered by the insult. Our study speculates that blocking 5-HT3 receptor using palonosetron overrides methyllycaconitine to combat AD-induced neuroinflammation and inflammasome cascade, as well as to restore microglial function in a HFFD/LPS novel model for sporadic AD.
Collapse
Affiliation(s)
- Reem A. Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts, 26 July Mehwar Road Intersection with Wahat Road, 6th of October City, Giza 12451, Egypt; (R.A.M.); (A.I.E.-b.)
| | - Dalaal M. Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Κasr El-Aini Str., Cairo 11562, Egypt;
| | - Amany I. El-brairy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts, 26 July Mehwar Road Intersection with Wahat Road, 6th of October City, Giza 12451, Egypt; (R.A.M.); (A.I.E.-b.)
| | - Kawkab A. Ahmed
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt;
| | - Hanan S. El-Abhar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Κasr El-Aini Str., Cairo 11562, Egypt;
| |
Collapse
|
9
|
Mohamed HE, Asker ME, Shaheen MA, Eissa RG, Younis NN. Alleviation of fructose-induced Alzheimer's disease in rats by pioglitazone and decaffeinated green coffee bean extract. J Food Biochem 2021; 45:e13715. [PMID: 33782984 DOI: 10.1111/jfbc.13715] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/13/2021] [Accepted: 03/18/2021] [Indexed: 11/28/2022]
Abstract
Increased fructose consumption is among bad nutritional habits that contribute to increased incidence of neurodegenerative diseases. We proposed that coffee, the most popular beverage worldwide, may protect against the progression of Alzheimer's disease (AD). We investigated the protective potential of decaffeinated green coffee bean extract (GCBE) and the possible potentiation of pioglitazone (PIO) effects by decaffeinated GCBE in fructose-induced AD in rats. Twenty-four rats [12-untreated and 12-pre-treated (for 4 weeks) with GCBE] consumed drinking water supplemented with 10% fructose for 18 weeks. Twelve of these rats (6-GCBE-untreated and 6-GCBE-pre-treated) were treated orally with PIO starting on the 13th week for 6 weeks. Prophylactic administration of GCBE attenuated oxidative damage (increased cortical reduced glutathione and superoxide dismutase activity), while decreased malondialdehyde. It retarded the activation of acetylcholine esterase, increased acetylcholine level in the cortex of fructose-induced AD. It also impeded the upregulation of beta-secretase-1and the accumulation of Aβ plaques that were induced by fructose drinking. With PIO therapy, GCBE showed better effects alleviating oxidative stress and Aβ extracellular plaques formation, while improving cholinergic activity, learning, and memory ability. In conclusions, the consumption of GCBE may protect against the development of AD and delay the progression of AD when given with PIO. PRACTICAL APPLICATIONS: Decaffeinated dietary supplement of green coffee bean extract attenuated the deleterious consequences of fructose-induced Alzheimer's disease in rats. It improved the antioxidant status and cortical cholinergic activity, while hindered the changes responsible for amyloid plaque formation. It also improved the impaired learning and memory. These results, if confirmed by clinical studies, may recommend the consumption of decaffeinated green coffee beans extract as dietary supplement or as a regular beverage to protect against AD in individuals with family history or early signs of AD. With pioglitazone, such dietary supplement improved pioglitazone efficacy and delayed the progression of AD.
Collapse
Affiliation(s)
- Hoda E Mohamed
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mervat E Asker
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mohamed A Shaheen
- Department of Histology and Cell Biology, Faculty of Human Medicine, Zagazig University, Zagazig, Egypt
| | - Rana G Eissa
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Nahla N Younis
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| |
Collapse
|
10
|
Qu X, Guan P, Han L, Wang Z, Huang X. Levistolide A Attenuates Alzheimer's Pathology Through Activation of the PPARγ Pathway. Neurotherapeutics 2021; 18:326-339. [PMID: 33034847 PMCID: PMC8116477 DOI: 10.1007/s13311-020-00943-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2020] [Indexed: 12/01/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by β-amyloid (Aβ) protein deposition, neurofibrillary tangle (NFT) formation, and neuronal loss in the brain. The current study was designed to investigate the potential mechanisms by which levistolide A affects the pathogenesis of AD in an amyloid precursor protein/presenilin 1 (APP/PS1) transgenic (Tg) mouse model of AD and N2a/APP695swe cells. Specifically, behavioral changes in levistolide A-treated APP/PS1 Tg mice were assessed by the nest-building and Morris water maze (MWM) tests. Levistolide A treatment clearly ameliorated memory deficits and cognitive decline in APP/PS1 Tg mice. Aβ generation and the inflammatory response in APP/PS1 Tg mouse brains were clearly reduced after long-term levistolide A application. Mechanistically, levistolide A concurrently stimulated the expression of α-secretase and decreased the generation of β- and γ-secretases. In addition, levistolide A inhibited the phosphorylation of tau in the brains of the Tg mice. Furthermore, in vitro and in vivo experiments suggested that peroxisome proliferator-activated receptor γ (PPARγ) is the key transcription factor that mediates the regulatory effects of levistolide A on the expression of α-, β-, and γ-secretases and phosphorylation of tau. Collectively, these findings show that levistolide A may be a candidate for the treatment of AD.
Collapse
Affiliation(s)
- Xiaodan Qu
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, People's Republic of China
| | - Peipei Guan
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, People's Republic of China
| | - Li Han
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, People's Republic of China.
| | - Zhanyou Wang
- Institute of Health Sciences, China Medical University, Shenyang, 110122, People's Republic of China.
| | - Xueshi Huang
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, People's Republic of China
| |
Collapse
|
11
|
Sweet but Bitter: Focus on Fructose Impact on Brain Function in Rodent Models. Nutrients 2020; 13:nu13010001. [PMID: 33374894 PMCID: PMC7821920 DOI: 10.3390/nu13010001] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022] Open
Abstract
Fructose consumption has drastically increased during the last decades due to the extensive commercial use of high-fructose corn syrup as a sweetener for beverages, snacks and baked goods. Fructose overconsumption is known to induce obesity, dyslipidemia, insulin resistance and inflammation, and its metabolism is considered partially responsible for its role in several metabolic diseases. Indeed, the primary metabolites and by-products of gut and hepatic fructolysis may impair the functions of extrahepatic tissues and organs. However, fructose itself causes an adenosine triphosphate (ATP) depletion that triggers inflammation and oxidative stress. Many studies have dealt with the effects of this sugar on various organs, while the impact of fructose on brain function is, to date, less explored, despite the relevance of this issue. Notably, fructose transporters and fructose metabolizing enzymes are present in brain cells. In addition, it has emerged that fructose consumption, even in the short term, can adversely influence brain health by promoting neuroinflammation, brain mitochondrial dysfunction and oxidative stress, as well as insulin resistance. Fructose influence on synaptic plasticity and cognition, with a major impact on critical regions for learning and memory, was also reported. In this review, we discuss emerging data about fructose effects on brain health in rodent models, with special reference to the regulation of food intake, inflammation, mitochondrial function and oxidative stress, insulin signaling and cognitive function.
Collapse
|
12
|
Yossef RR, Al-Yamany MF, Saad MA, El-Sahar AE. Neuroprotective effects of vildagliptin on drug induced Alzheimer's disease in rats with metabolic syndrome: Role of hippocampal klotho and AKT signaling pathways. Eur J Pharmacol 2020; 889:173612. [PMID: 33035520 DOI: 10.1016/j.ejphar.2020.173612] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 12/16/2022]
Abstract
Growing evidences suggest the presence of several similarities in the molecular mechanisms underlying the neurodegenerative diseases and metabolic abnormalities. Adults who develop Metabolic Syndrome (MS) are at a higher risk of developing Alzheimer's disease (AD). Pharmacological agents, like dipeptidyl peptidase-4 (DPP-4) inhibitors that increase the levels of glucagon like peptide 1 (GLP-1) and ameliorate symptoms of MS, have become an auspicious candidate as disease modifying agents in the treatment of AD. The present study investigates the beneficial effects of Vildagliptin, a DPP-4 inhibitor in counteracting cognitive decline in different models of dementia targeting the AKT, JAK/STAT signaling pathways and hippocampal Klotho expression, to judge the neuroprotective, anti-apoptotic and anti-inflammatory effects of the drug. Cognitive decline was induced by either administration of high fat high sugar (HFHS) diet for 45 days alone, or with oral administration of AlCl3 (100 mg/kg/day) for 60 days. Rats were orally administered Vildagliptin (10 mg/kg) for 60 days along with AlCl3 administration. Vildagliptin treatment improved spatial memory and activities in morris water maze (MWM) test and open field test respectively. Results revealed an increase of both hippocampal klotho and Bcl-2 expressions along with an increase in both AKT and ERK1/2 phosphorylation. In contrast, Vildagliptin treatment decreased hippocampal contents of inflammatory, apoptotic and oxidative stress biomarkers as TNF-α, caspase-3 and FOXO1 along with restoring metabolic abnormalities. A significant decrease in BAX expressions with JAK2/STAT3 inhibition was observed. These findings demonstrate that the neuroprotective role of vildagliptin is possibly via modulating Klotho protein together with AKT pathway.
Collapse
Affiliation(s)
- Rasha R Yossef
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October 6 University, Giza, Egypt.
| | - Mohamed F Al-Yamany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | | | - Ayman E El-Sahar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| |
Collapse
|
13
|
Spagnuolo MS, Pallottini V, Mazzoli A, Iannotta L, Tonini C, Morone B, Ståhlman M, Crescenzo R, Strazzullo M, Iossa S, Cigliano L. A Short‐Term Western Diet Impairs Cholesterol Homeostasis and Key Players of Beta Amyloid Metabolism in Brain of Middle Aged Rats. Mol Nutr Food Res 2020; 64:e2000541. [DOI: 10.1002/mnfr.202000541] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Indexed: 12/28/2022]
Affiliation(s)
| | - Valentina Pallottini
- Department of ScienceBiomedical and Technology Science SectionUniversity Roma Tre Rome 00146 Italy
| | - Arianna Mazzoli
- Department of BiologyUniversity of Naples Federico II Naples 80126 Italy
| | - Lucia Iannotta
- Department of BiologyUniversity of Naples Federico II Naples 80126 Italy
| | - Claudia Tonini
- Department of ScienceBiomedical and Technology Science SectionUniversity Roma Tre Rome 00146 Italy
| | - Barbara Morone
- Institute of Genetics and Biophysics “A. Buzzati‐Traverso”National Research Council Naples 80131 Italy
| | - Marcus Ståhlman
- Wallenberg LaboratoryDepartment of Molecular and Clinical MedicineSahlgrenska AcademyUniversity of Gothenburg Gothenburg 413 45 Sweden
| | | | - Maria Strazzullo
- Institute of Genetics and Biophysics “A. Buzzati‐Traverso”National Research Council Naples 80131 Italy
| | - Susanna Iossa
- Department of BiologyUniversity of Naples Federico II Naples 80126 Italy
| | - Luisa Cigliano
- Department of BiologyUniversity of Naples Federico II Naples 80126 Italy
| |
Collapse
|
14
|
Gupta S, Singhal NK, Ganesh S, Sandhir R. Extending Arms of Insulin Resistance from Diabetes to Alzheimer's Disease: Identification of Potential Therapeutic Targets. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 18:172-184. [PMID: 30430949 DOI: 10.2174/1871527317666181114163515] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/08/2018] [Accepted: 11/08/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND & OBJECTIVE Type 3 diabetes (T3D) is chronic insulin resistant state of brain which shares pathology with sporadic Alzheimer's disease (sAD). Insulin signaling is a highly conserved pathway in the living systems that orchestrate cell growth, repair, maintenance, energy homeostasis and reproduction. Although insulin is primarily studied as a key molecule in diabetes mellitus, its role has recently been implicated in the development of Alzheimer's disease (AD). Severe complications in brain of diabetic patients and metabolically compromised status is evident in brain of AD patients. Underlying shared pathology of two disorders draws a trajectory from peripheral insulin resistance to insulin unresponsiveness in the central nervous system (CNS). As insulin has a pivotal role in AD, it is not an overreach to address diabetic condition in AD brain as T3D. Insulin signaling is indispensable to nervous system and it is vital for neuronal growth, repair, and maintenance of chemical milieu at synapses. Downstream mediators of insulin signaling pathway work as a regulatory hub for aggregation and clearance of unfolded proteins like Aβ and tau. CONCLUSION In this review, we discuss the regulatory roles of insulin as a pivotal molecule in brain with the understanding of defective insulin signaling as a key pathological mechanism in sAD. This article also highlights ongoing trials of targeting insulin signaling as a therapeutic manifestation to treat diabetic condition in brain.
Collapse
Affiliation(s)
- Smriti Gupta
- Department of Biochemistry, Basic Medical Science Block II, Sector 25, Panjab University, Chandigarh 160014, India
| | - Nitin Kumar Singhal
- National Agri-Food Biotechnology Institute, Sector 81, S.A.S. Nagar, Mohali, Punjab 140306, India
| | - Subramaniam Ganesh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Rajat Sandhir
- Department of Biochemistry, Basic Medical Science Block II, Sector 25, Panjab University, Chandigarh 160014, India
| |
Collapse
|
15
|
Ohyagi Y, Miyoshi K, Nakamura N. Therapeutic Strategies for Alzheimer's Disease in the View of Diabetes Mellitus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1128:227-248. [PMID: 31062332 DOI: 10.1007/978-981-13-3540-2_11] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recently, Alzheimer's disease (AD) is understood as "diabetes of the brain" or "type 3 diabetes." Recent clinical trials of anti-amyloid β-protein (Aβ) therapies have not proved to be successful. Thus, glucose-insulin metabolism in the brain is thought to be an alternative therapeutic target. Various types of antidiabetic drugs such as insulin, thiazolidinediones, dipeptidyl peptidase-4 (DPP4) inhibitors, glucagon-like peptide-1 (GLP-1) agonists, biguanides, and others have been reported to be effective on cognitive impairment in animal models and patients with DM or AD. Here, recent reports are reviewed. While we identified apomorphine (APO) as a novel drug that promoted intracellular Aβ degradation and improved memory function in an AD mouse model, more recently, we have revealed that APO treatment improves neuronal insulin resistance and activates insulin-degrading enzyme (IDE), a major Aβ-degrading enzyme. In this context, recovery of impaired insulin signaling in AD neurons may be a promising therapeutic strategy for AD dementia.
Collapse
Affiliation(s)
- Yasumasa Ohyagi
- Department of Neurology and Geriatric Medicine, Graduate School of Medicine, Ehime University, Toon, Ehime, Japan.
| | - Katsue Miyoshi
- Long-Term Care Health Facility Cosmos, Kushiro-mutsumi, Hokkaido, Japan
| | - Norimichi Nakamura
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
16
|
Quan Q, Qian Y, Li X, Li M. Pioglitazone Reduces β Amyloid Levels via Inhibition of PPARγ Phosphorylation in a Neuronal Model of Alzheimer's Disease. Front Aging Neurosci 2019; 11:178. [PMID: 31379559 PMCID: PMC6650543 DOI: 10.3389/fnagi.2019.00178] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 07/02/2019] [Indexed: 12/28/2022] Open
Abstract
It has been demonstrated that peroxisome proliferator-activated receptor γ (PPARγ) can regulate the transcription of its target gene, insulin-degrading enzyme (IDE), and thus enhance the expression of the IDE protein. The protein can degrade β amyloid (Aβ), a core pathological product of Alzheimer’s disease (AD). PPARγ can also regulate the transcription of other target gene, β-amyloid cleavage enzyme 1 (BACE1), and thus inhibit the expression of the BACE1 protein. BACE1 can hydrolyze amyloid precursor protein (APP), the precursor of Aβ. In adipose tissue, PPARγ agonists can inhibit the phosphorylation of PPARγ by inhibiting cyclin-dependent kinase 5 (CDK5), which in turn affects the expression of target genes regulated by PPARγ. PPARγ agonists may also exert inhibitory effects on the phosphorylation of PPARγ in the brain, thereby affecting the expression of the aforementioned PPARγ target genes and reducing Aβ levels. The present study confirmed this hypothesis by showing that PPARγ agonist pioglitazone attenuated the neuronal apoptosis of primary rat hippocampal neurons induced by Aβ1–42, downregulated CDK5 expression, weakened the binding of CDK5 to PPARγ, reduced PPARγ phosphorylation, increased the expression of PPARγ and IDE, decreased the expression of BACE1, reduced APP production, and downregulated intraneuronal Aβ1–42 levels. These effects were inhibited by the PPARγ antagonist GW9662. After CDK5 silencing with CDK5 shRNA, the above effect of pioglitazone was not observed, except when upregulating the expression of PPARγ in Aβ1–42 treated neurons. In conclusion, this study demonstrated that pioglitazone could inhibit the phosphorylation of PPARγ in vitro by inhibiting CDK5 expression, which in turn affected the expression of PPARγ target genes Ide and Bace1, thereby promoting Aβ degradation and reducing Aβ production. This reduced Aβ levels in the brain, thereby exerting neuroprotective effects in an AD model.
Collapse
Affiliation(s)
- Qiankun Quan
- Department of Geriatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yihua Qian
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xi Li
- Department of Geriatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ming Li
- Department of Geriatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
17
|
Indirubin-3'-monoxime prevents aberrant activation of GSK-3β/NF-κB and alleviates high fat-high fructose induced Aβ-aggregation, gliosis and apoptosis in mice brain. Int Immunopharmacol 2019; 70:396-407. [PMID: 30856390 DOI: 10.1016/j.intimp.2019.02.053] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/13/2019] [Accepted: 02/28/2019] [Indexed: 02/05/2023]
Abstract
Deciphering the molecular mechanisms of amyloid pathology and glial cell-mediated neuroinflammation, offers a novel avenue for therapeutic intervention against neurodegeneration. Recent findings demonstrate a crucial link between activation of glycogen synthase kinase-3β (GSK-3β), amyloid deposition and a neuroinflammatory state. However, studies demonstrating the pharmacological effects of GSK-3β inhibition and the interlinked molecular mechanisms still remain elusive. The present study explores whether high fat-high fructose diet (HFFD)-induced neuropathological changes could be alleviated by indirubin-3'-monoxime (IMX), a GSK-3β inhibitor. Male Swiss albino mice (8 weeks old) were fed with normal pellet or HFFD for 60 days. HFFD mice were treated with IMX once daily for last 7 days of the experimental period. HFFD fed-mice had significant amyloid deposits in cerebral cortex and hippocampus, and protein expression analyses showed activation of GSK-3β, nuclear translocation of NF-κB p65 and upregulation of inflammatory (TNF-α, IL-6, COX-2), astrocytic (GFAP), glial surface (CD-68) and pro-apoptotic markers (Bax and caspase-3). IMX treatment promotes the inhibitory phosphorylation of GSK-3β at Ser9 and moreover, a marked reduction in the phosphorylation of IKK-β, which prevents translocation and activation of NF-κB. Protein expression studies in IMX-treated brain tissues positively correlate with the anti-neuroinflammatory effects of GSK-3β inhibition. Taken together, our results provide substantial evidence that IMX could potentially attenuate neuroinflammation in coordination with the master transcription factor-NF-κB.
Collapse
|
18
|
Sachdeva AK, Dharavath RN, Chopra K. Time-response studies on development of cognitive deficits in an experimental model of insulin resistance. Clin Nutr 2018; 38:1447-1456. [PMID: 30037709 DOI: 10.1016/j.clnu.2018.06.966] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 05/30/2018] [Accepted: 06/19/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS Alzheimer's disease is suggested to be primarily metabolic, mainly characterized by brain insulin resistance. Chronic fructose feeding results in hippocampal insulin resistance. However, variable opinion exists regarding the concentration and duration of fructose feeding to trigger insulin resistance and resultant cognitive insults. Therefore this study was planned to construct a time-response curve of the appearance of fructose-induced insulin resistance and memory insufficiencies in rats over a period of 24 weeks. Further, Pearson's correlations were drawn between indices of insulin resistance and markers of memory deficits at various time points. METHODS Male Wistar rats (6 weeks old; 155 ± 5 g) were fed with 15% fructose in normal drinking water for a period of 24 weeks. Body weight, food and water intake were weekly monitored. Fasting blood glucose, glycosylated hemoglobin (HbA1C), lipid profiling, plasma insulin, HOMA-IR index, and systolic blood pressure were estimated to confirm the manifestation of insulin resistance. Cognitive derangements were evaluated by Elevated plus maze and Morris water maze at different time points during the study. RESULTS Most of the parameters including insulin resistance became evident at the 7th week and continued until the end of study (24th week) whereas cognitive insufficiency became significantly distinct at the 20th, 22nd and 24th week. Significantly increased serum nitro-oxidative stress, inflammatory cytokines and serum homocysteine levels were intensely connected with fructose-induced neuronal deficits. CONCLUSIONS The construction of time response study reveals that the hallmark characteristics of insulin resistance appear from the 7th week of fructose feeding whereas the cognitive dysfunction appears on the 20th week and both persist till the end of the study. Fructose-induced oxidative stress and neuroinflammation plausibly impair neuronal signaling and synaptic plasticity.
Collapse
Affiliation(s)
- Anand Kamal Sachdeva
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study, Panjab University, Chandigarh, 160 014, India
| | - Ravinder Naik Dharavath
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study, Panjab University, Chandigarh, 160 014, India
| | - Kanwaljit Chopra
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study, Panjab University, Chandigarh, 160 014, India.
| |
Collapse
|
19
|
Mohamed HE, Asker ME, Younis NN, Shaheen MA, Eissa RG. Modulation of brain insulin signaling in Alzheimer’s disease: New insight on the protective role of green coffee bean extract. Nutr Neurosci 2018; 23:27-36. [DOI: 10.1080/1028415x.2018.1468535] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Hoda E. Mohamed
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mervat E. Asker
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Nahla N. Younis
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mohamed A. Shaheen
- Department of Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Rana G. Eissa
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| |
Collapse
|
20
|
Impact of Fish Oil Supplementation and Interruption of Fructose Ingestion on Glucose and Lipid Homeostasis of Rats Drinking Different Concentrations of Fructose. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4378328. [PMID: 28929113 PMCID: PMC5591931 DOI: 10.1155/2017/4378328] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 06/20/2017] [Accepted: 06/28/2017] [Indexed: 12/28/2022]
Abstract
Background. Continuous fructose consumption may cause elevation of circulating triacylglycerol. However, how much of this alteration is reverted after the removal of fructose intake is not known. We explored this question and compared the efficacy of this approach with fish oil supplementation. Methods. Male Wistar rats were divided into the following groups: control (C), fructose (F) (water intake with 10% or 30% fructose for 9 weeks), fish oil (FO), and fructose/fish oil (FFO). Fish oil was supplemented only for the last 33 days of fructose ingestion. Half of the F group remained for additional 8 weeks without fructose ingestion (FR). Results. Fructose ingestion reduced food intake to compensate for the increased energy obtained through water ingestion, independent of fructose concentration. Fish oil supplementation exerted no impact on these parameters, but the removal of fructose from water recovered both ingestion behaviors. Plasma triacylglycerol augmented significantly during the second and third weeks (both fructose groups). Fish oil supplementation did not attenuate the elevation in triacylglycerol caused by fructose intake, but the interruption of sugar consumption normalized this parameter. Conclusion. Elevation in triacylglyceridemia may be recovered by removing fructose from diet, suggesting that it is never too late to repair improper dietary habits.
Collapse
|
21
|
Kuga GK, Botezelli JD, Gaspar RC, Gomes RJ, Pauli JR, Leme JACDA. Hippocampal insulin signaling and neuroprotection mediated by physical exercise in Alzheimer´s Disease. MOTRIZ: REVISTA DE EDUCACAO FISICA 2017. [DOI: 10.1590/s1980-6574201700si0008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
22
|
Rani V, Deshmukh R, Jaswal P, Kumar P, Bariwal J. Alzheimer's disease: Is this a brain specific diabetic condition? Physiol Behav 2016; 164:259-67. [PMID: 27235734 DOI: 10.1016/j.physbeh.2016.05.041] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 05/16/2016] [Accepted: 05/22/2016] [Indexed: 01/13/2023]
Abstract
Alzheimer's disease (AD) and type 2 diabetes (T2DM) are the two major health issues affecting millions of elderly people worldwide, with major impacts in the patient's daily life. Numerous studies have demonstrated that patients with diabetes have an increased risk of developing AD compared with healthy individuals. The principal biological mechanisms that associate with the progression of diabetes and AD are not completely understood. Impaired insulin signaling, uncontrolled glucose metabolism, oxidative stress, abnormal protein processing, and the stimulation of inflammatory pathways are common features to both AD and T2DM. In recent years brain specific abnormalities in insulin and insulin like growth factor (IGF) signaling considered as a major trigger involved in the etiopathogenesis of AD, showing T2DM like milieu. This review summarizes the pathways that might link diabetes and AD and the effect of diminished insulin.
Collapse
Affiliation(s)
- Vanita Rani
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Punjab, India
| | - Rahul Deshmukh
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Punjab, India.
| | - Priya Jaswal
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Punjab, India
| | - Puneet Kumar
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Punjab, India
| | - Jitender Bariwal
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga 142001, Punjab, India
| |
Collapse
|
23
|
Quan QK, Li X, Yuan HF, Wang Y, Liu WL. Ginsenoside Rg1 inhibits high-voltage-activated calcium channel currents in hippocampal neurons of beta-amyloid peptide-exposed rat brain slices. Chin J Integr Med 2016:10.1007/s11655-015-2301-4. [PMID: 26779710 DOI: 10.1007/s11655-015-2301-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To examine whether ginsenoside Rg1 (Rg1) inhibits the high-voltage-activated calcium currents (ICa,HVA) via mitogen-activated protein kinase (MAPK) in hippocampal neurons in rat brain slices exposed to beta-amyloid peptide 25-35 (Aβ25-35). METHODS An experimental Alzheimer disease (AD) model was prepared by exposure of rat brain slices to Aβ25-35 (10 µmol/L). After treatment with Rg1 (20 µmol/L), the ICa,HVA elicited in hippocampal neurons in these rat brain slices upon depolarization from-40 to 40 mV for 200 ms was recorded by a whole-cell patch clamp to analyze the changes in the peak current density, I-V curve, activation-V curve, and inactivation-V curve. RESULTS Exposure of rat brain slices to Aβ led to a significant increase in ICa,HVA, enhancement of the voltage sensitivity of channel activation, and reduction of the voltage sensitivity of channel inactivation in neurons in the hippocampus of rat brain slices. Rg1 treatment significantly inhibited these changes. These effects of Rg1 could be effectively inhibited by the MAPK inhibitor PD98059. CONCLUSION Rg1 can inhibit Ica,HVA via MAPK in hippocampal neurons in Aβ-exposed rat brain slices.
Collapse
Affiliation(s)
- Qian-Kun Quan
- Department of Geriatrics, The Second Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, 710004, China
| | - Xi Li
- Department of Geriatrics, The Second Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, 710004, China.
| | - Hai-Feng Yuan
- Department of Encephalopathy, The Second Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, 710004, China
| | - Yi Wang
- Department of Electronic Engineering, City University of Hong Kong, Hong Kong SAR, China
| | - Wen-Li Liu
- The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, 710061, China
| |
Collapse
|
24
|
Gad ES, Zaitone SA, Moustafa YM. Pioglitazone and exenatide enhance cognition and downregulate hippocampal beta amyloid oligomer and microglia expression in insulin-resistant rats. Can J Physiol Pharmacol 2015; 94:819-28. [PMID: 27389824 DOI: 10.1139/cjpp-2015-0242] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Insulin resistance is known to be a risk factor for cognitive impairment, most likely linked to insulin signaling, microglia overactivation, and beta amyloid (Aβ) deposition in the brain. Exenatide, a long lasting glucagon-like peptide-1 (GLP-1) analogue, enhances insulin signaling and shows neuroprotective properties. Pioglitazone, a peroxisome proliferated-activated receptor-γ (PPAR-γ) agonist, was previously reported to enhance cognition through its effect on Aβ accumulation and clearance. In the present study, insulin resistance was induced in male rats by drinking fructose for 12 weeks. The effect of monotherapy with pioglitazone (10 mg·kg(-1)) and exenatide or their combination on memory dysfunction was determined and some of the probable underlying mechanisms were studied. The current results confirmed that (1) feeding male rats with fructose syrup for 12 weeks resulted in a decline of learning and memory registered in eight-arm radial maze test; (2) treatment with pioglitazone or exenatide enhanced cognition, reduced hippocampal neurodegeneration, and reduced hippocampal microglia expression and beta amyloid oligomer deposition in a manner that is equal to monotherapies. These results may give promise for the use of pioglitazone or exenatide for ameliorating the learning and memory deficits associated with insulin resistance in clinical setting.
Collapse
Affiliation(s)
- Enas S Gad
- a Medical Department at Faculty of Commerce, Suez Canal University, Ismailia, Egypt
| | - Sawsan A Zaitone
- b Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Yasser M Moustafa
- b Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
25
|
Kim B, Feldman EL. Insulin resistance as a key link for the increased risk of cognitive impairment in the metabolic syndrome. Exp Mol Med 2015; 47:e149. [PMID: 25766618 PMCID: PMC4351418 DOI: 10.1038/emm.2015.3] [Citation(s) in RCA: 235] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 11/19/2014] [Indexed: 02/07/2023] Open
Abstract
Metabolic syndrome (MetS) is a cluster of cardiovascular risk factors that includes obesity, diabetes, and dyslipidemia. Accumulating evidence implies that MetS contributes to the development and progression of Alzheimer's disease (AD); however, the factors connecting this association have not been determined. Insulin resistance (IR) is at the core of MetS and likely represent the key link between MetS and AD. In the central nervous system, insulin plays key roles in learning and memory, and AD patients exhibit impaired insulin signaling that is similar to that observed in MetS. As we face an alarming increase in obesity and T2D in all age groups, understanding the relationship between MetS and AD is vital for the identification of potential therapeutic targets. Recently, several diabetes therapies that enhance insulin signaling are being tested for a potential therapeutic benefit in AD and dementia. In this review, we will discuss MetS as a risk factor for AD, focusing on IR and the recent progress and future directions of insulin-based therapies.
Collapse
Affiliation(s)
- Bhumsoo Kim
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
26
|
Lycopene attenuates insulin signaling deficits, oxidative stress, neuroinflammation, and cognitive impairment in fructose-drinking insulin resistant rats. Neuropharmacology 2014; 86:389-96. [DOI: 10.1016/j.neuropharm.2014.07.020] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 07/17/2014] [Accepted: 07/28/2014] [Indexed: 12/25/2022]
|
27
|
Sripetchwandee J, Pipatpiboon N, Pratchayasakul W, Chattipakorn N, Chattipakorn SC. DPP-4 Inhibitor and PPARγ Agonist Restore the Loss of CA1 Dendritic Spines in Obese Insulin-resistant Rats. Arch Med Res 2014; 45:547-52. [DOI: 10.1016/j.arcmed.2014.09.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 09/12/2014] [Indexed: 02/07/2023]
|
28
|
Rojo ML, Cipriano M, Söderström I, Simonytė K, Olsson T, Fowler CJ. Effects of dietary glucose and fructose upon cannabinoid CB1 receptor functionality in the rat brain: a pilot study. Life Sci 2014; 108:116-21. [PMID: 24928368 DOI: 10.1016/j.lfs.2014.05.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 04/28/2014] [Accepted: 05/28/2014] [Indexed: 01/24/2023]
Abstract
AIMS A high consumption of fructose leads not only to peripheral changes in insulin sensitivity and vascular function, but also to central changes in several brain regions. Given the role of the endogenous cannabinoid system in the control of energy intake, we undertook a pilot study to determine whether a high fructose diet produced changes in brain CB1 receptor functionality. MAIN METHODS Male rats given access ad libitum to normal chow were given either water, glucose or fructose solutions to drink. CB1 receptor functionality was measured autoradiographically as the increase in [(35)S]GTPγS binding produced by the agonist CP55,940. KEY FINDINGS Seven regions were investigated: the prefrontal cortex, caudate-putamen, hippocampal CA1-CA3, dentate gyrus, amygdala, and dorsomedial and ventromedial hypothalami. Two-way robust Wilcoxon analyses for each brain region indicated that the dietary treatment did not produce significant main effects upon agonist-stimulated [(35)S]GTPγS binding in any of the regions, in contrast to a significant main effect upon both leptin and adiponectin levels in the blood. However, a MANCOVA of the data controlling for leptin and adiponectin as co-variables identified a significant effect of glucose and fructose treatment for five weeks upon the [(35)S]GTPγS response in the ventromedial hypothalamus, a region of importance for regulation of appetite. SIGNIFICANCE It is concluded from this pilot study that palatable solutions do not produce overt changes in brain CB1 receptor functionality, although subtle changes in discrete brain regions may occur.
Collapse
Affiliation(s)
- Maria Luisa Rojo
- Department of Public Health and Clinical Medicine, Medicine, Umeå University, SE-901 87 Umeå, Sweden
| | - Mariateresa Cipriano
- Department of Pharmacology and Clinical Neuroscience, Umeå University, SE-901 87 Umeå, Sweden
| | - Ingegerd Söderström
- Department of Public Health and Clinical Medicine, Medicine, Umeå University, SE-901 87 Umeå, Sweden
| | - Kotryna Simonytė
- Department of Public Health and Clinical Medicine, Medicine, Umeå University, SE-901 87 Umeå, Sweden
| | - Tommy Olsson
- Department of Public Health and Clinical Medicine, Medicine, Umeå University, SE-901 87 Umeå, Sweden
| | - Christopher J Fowler
- Department of Pharmacology and Clinical Neuroscience, Umeå University, SE-901 87 Umeå, Sweden.
| |
Collapse
|
29
|
Cholerton B, Baker LD, Craft S. Insulin, cognition, and dementia. Eur J Pharmacol 2013; 719:170-179. [PMID: 24070815 DOI: 10.1016/j.ejphar.2013.08.008] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 08/21/2013] [Accepted: 08/26/2013] [Indexed: 01/21/2023]
Abstract
Cognitive disorders of aging represent a serious threat to the social and economic welfare of current society. It is now widely recognized that pathology related to such conditions, particularly Alzheimer's disease, likely begins years or decades prior to the onset of clinical dementia symptoms. This revelation has led researchers to consider candidate mechanisms precipitating the cascade of neuropathological events that eventually lead to clinical Alzheimer's disease. Insulin, a hormone with potent effects in the brain, has recently received a great deal of attention for its potential beneficial and protective role in cognitive function. Insulin resistance, which refers to the reduced sensitivity of target tissues to the favorable effects of insulin, is related to multiple chronic conditions known to impact cognition and increase dementia risk. With insulin resistance-associated conditions reaching epidemic proportions, the prevalence of Alzheimer's disease and other cognitive disorders will continue to rise exponentially. Fortunately, these chronic insulin-related conditions are amenable to pharmacological intervention. As a result, novel therapeutic strategies that focus on increasing insulin sensitivity in the brain may be an important target for protecting or treating cognitive decline. The following review will highlight our current understanding of the role of insulin in brain, potential mechanisms underlying the link between insulin resistance and dementia, and current experimental therapeutic strategies aimed at improving cognitive function via modifying the brain's insulin sensitivity.
Collapse
Affiliation(s)
- Brenna Cholerton
- Department of Psychiatry and Behavioral Science, University of Washington School of Medicine, Seattle, WA, USA; Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Laura D Baker
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, Winston-Salem, NC 27157-1207, USA
| | - Suzanne Craft
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, Winston-Salem, NC 27157-1207, USA.
| |
Collapse
|
30
|
Yin QQ, Pei JJ, Xu S, Luo DZ, Dong SQ, Sun MH, You L, Sun ZJ, Liu XP. Pioglitazone improves cognitive function via increasing insulin sensitivity and strengthening antioxidant defense system in fructose-drinking insulin resistance rats. PLoS One 2013; 8:e59313. [PMID: 23527159 PMCID: PMC3603906 DOI: 10.1371/journal.pone.0059313] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 02/13/2013] [Indexed: 12/18/2022] Open
Abstract
Insulin resistance (IR) links Alzheimer’s disease (AD) with oxidative damage, cholinergic deficit, and cognitive impairment. Peroxisome proliferator-activated receptor γ (PPARγ) agonist pioglitazone previously used to treat type 2 diabetes mellitus (T2DM) has also been demonstrated to be effective in anti-inflammatory reaction and anti-oxidative stress in the animal models of AD and other neuroinflammatory diseases. Here, we investigated the effect of pioglitazone on learning and memory impairment and the molecular events that may cause it in fructose-drinking insulin resistance rats. We found that long-term fructose-drinking causes insulin resistance, oxidative stress, down-regulated activity of cholinergic system, and cognitive deficit, which could be ameliorated by pioglitazone administration. The results from the present study provide experimental evidence for using pioglitazone in the treatment of brain damage caused by insulin resistance.
Collapse
Affiliation(s)
- Qing-Qing Yin
- Department of Senile Neurology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P. R. China
| | - Jin-Jing Pei
- Department of KI-Alzheimer Disease Research Center, Karolinska Institutet, Stockholm, Sweden
| | - Song Xu
- Department of Anti-Ageing, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P. R. China
| | - Ding-Zhen Luo
- Department of Senile Neurology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P. R. China
| | - Si-Qing Dong
- Department of Senile Neurology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P. R. China
| | - Meng-Han Sun
- Department of Senile Neurology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P. R. China
| | - Li You
- Department of Central Lab, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P. R. China
| | - Zhi-Jian Sun
- Department of Anti-Ageing, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P. R. China
| | - Xue-Ping Liu
- Department of Senile Neurology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P. R. China
- Department of Anti-Ageing, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P. R. China
- * E-mail:
| |
Collapse
|
31
|
Quan Q, Wang J, Li X, Wang Y. Ginsenoside Rg1 decreases Aβ(1-42) level by upregulating PPARγ and IDE expression in the hippocampus of a rat model of Alzheimer's disease. PLoS One 2013; 8:e59155. [PMID: 23520555 PMCID: PMC3592813 DOI: 10.1371/journal.pone.0059155] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 02/11/2013] [Indexed: 02/03/2023] Open
Abstract
Background and Purpose The present study was designed to examine the effects of ginsenoside Rg1 on expression of peroxisome proliferator-activated receptor γ (PPARγ) and insulin-degrading enzyme (IDE) in the hippocampus of rat model of Alzheimer's disease (AD) to determine how ginsenoside Rg1 (Rg1) decreases Aβ levels in AD. Experimental Approach Experimental AD was induced in rats by a bilateral injection of 10 µg soluble beta-amyloid peptide 1–42 (Aβ1–42) into the CA1 region of the hippocampus, and the rats were treated with Rg1 (10 mg·kg−1, intraperitoneally) for 28 days. The Morris water maze was used to test spatial learning and memory performance. Hematoxylin-eosin staining was performed to analyze the hippocampal histopathological damage. Immunohistochemistry, western blotting, and real-time PCR were used to detect Aβ1–42, PPARγ, and insulin-degrading enzyme (IDE) expression in the hippocampus. Key Results Injection of soluble Aβ1–42 into the hippocampus led to significant dysfunction of learning and memory, hippocampal histopathological abnormalities and increased Aβ1–42 levels in the hippocampus. Rg1 treatment significantly improved learning and memory function, attenuated hippocampal histopathological abnormalities, reduced Aβ1–42 levels and increased PPARγ and IDE expression in the hippocampus; these effects of Rg1 could be effectively inhibited by GW9662, a PPARγ antagonist. Conclusions and Implications Given that PPARγ can upregulate IDE expression and IDE can degrade Aβ1–42, these results indicate that Rg1 can increase IDE expression in the hippocampus by upregulating PPARγ, leading to decreased Aβ levels, attenuated hippocampal histopathological abnormalities and improved learning and memory in a rat model of AD.
Collapse
Affiliation(s)
- QianKun Quan
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Institute of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, National Engineering Research Center of Health Care and Medical Devices, Xi'an Jiaotong University Branch, Xi'an, China
| | - Jue Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Institute of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, National Engineering Research Center of Health Care and Medical Devices, Xi'an Jiaotong University Branch, Xi'an, China
- * E-mail:
| | - Xi Li
- Department of Geriatrics, The Second Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China
| | - Yi Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Institute of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, National Engineering Research Center of Health Care and Medical Devices, Xi'an Jiaotong University Branch, Xi'an, China
| |
Collapse
|
32
|
The Components of Flemingia macrophylla Attenuate Amyloid β-Protein Accumulation by Regulating Amyloid β-Protein Metabolic Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 2012:795843. [PMID: 22719789 PMCID: PMC3376484 DOI: 10.1155/2012/795843] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 03/06/2012] [Accepted: 03/13/2012] [Indexed: 12/19/2022]
Abstract
Flemingia macrophylla (Leguminosae) is a popular traditional remedy used in Taiwan as anti-inflammatory, promoting blood circulation and antidiabetes agent. Recent study also suggested its neuroprotective activity against Alzheimer's disease. Therefore, the effects of F. macrophylla on Aβ production and degradation were studied. The effect of F. macrophylla on Aβ metabolism was detected using the cultured mouse neuroblastoma cells N2a transfected with human Swedish mutant APP (swAPP-N2a cells). The effects on Aβ degradation were evaluated on a cell-free system. An ELISA assay was applied to detect the level of Aβ1-40 and Aβ1-42. Western blots assay was employed to measure the levels of soluble amyloid precursor protein and insulin degrading enzyme (IDE). Three fractions of F. macrophylla modified Aβ accumulation by both inhibiting β-secretase and activating IDE. Three flavonoids modified Aβ accumulation by activating IDE. The activated IDE pool by the flavonoids was distinctly regulated by bacitracin (an IDE inhibitor). Furthermore, flavonoid 94-18-13 also modulates Aβ accumulation by enhancing IDE expression. In conclusion, the components of F. macrophylla possess the potential for developing new therapeutic drugs for Alzheimer's disease.
Collapse
|
33
|
Liu Y, Su Y, Sun S, Wang T, Qiao X, Run X, Liang Z. Tau phosphorylation and μ-calpain activation mediate the dexamethasone-induced inhibition on the insulin-stimulated Akt phosphorylation. PLoS One 2012; 7:e35783. [PMID: 22536436 PMCID: PMC3335002 DOI: 10.1371/journal.pone.0035783] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Accepted: 03/21/2012] [Indexed: 12/04/2022] Open
Abstract
Evidence has suggested that insulin resistance (IR) or high levels of glucocorticoids (GCs) may be linked with the pathogenesis and/or progression of Alzheimer's disease (AD). Although studies have shown that a high level of GCs results in IR, little is known about the molecular details that link GCs and IR in the context of AD. Abnormal phosphorylation of tau and activation of μ-calpain are two key events in the pathology of AD. Importantly, these two events are also related with GCs and IR. We therefore speculate that tau phosphorylation and μ-calpain activation may mediate the GCs-induced IR. Akt phosphorylation at Ser-473 (pAkt) is commonly used as a marker for assessing IR. We employed two cell lines, wild-type HEK293 cells and HEK293 cells stably expressing the longest human tau isoform (tau-441; HEK293/tau441 cells). We examined whether DEX, a synthetic GCs, induces tau phosphorylation and μ-calpain activation. If so, we examined whether the DEX-induced tau phosphorylation and μ-calpain activation mediate the DEX-induced inhibition on the insulin-stimulated Akt phosphorylation. The results showed that DEX increased tau phosphorylation and induced tau-mediated μ-calpain activation. Furthermore, pre-treatment with LiCl prevented the effects of DEX on tau phosphorylation and μ-calpain activation. Finally, both LiCl pre-treatment and calpain inhibition prevented the DEX-induced inhibition on the insulin-stimulated Akt phosphorylation. In conclusion, our study suggests that the tau phosphorylation and μ-calpain activation mediate the DEX-induced inhibition on the insulin-stimulated Akt phosphorylation.
Collapse
Affiliation(s)
- Yudong Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ying Su
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shenggang Sun
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xian Qiao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoqin Run
- Department of General Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhihou Liang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
34
|
Kim B, Feldman EL. Insulin resistance in the nervous system. Trends Endocrinol Metab 2012; 23:133-41. [PMID: 22245457 PMCID: PMC3392648 DOI: 10.1016/j.tem.2011.12.004] [Citation(s) in RCA: 227] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 12/11/2011] [Accepted: 12/13/2011] [Indexed: 12/15/2022]
Abstract
Metabolic syndrome is a cluster of cardiovascular risk factors including obesity, diabetes and dyslipidemia. Insulin resistance (IR) is at the core of metabolic syndrome. In adipose tissue and muscle, IR results in decreased insulin signaling, primarily affecting downstream phosphatidylinositol 3-kinase (PI3K)/Akt signaling. It was recently proposed that neurons can develop hyperinsulinemia-induced IR, which in turn results in injury to the peripheral and central nervous systems and is probably pathogenic in common neurological disorders such as diabetic neuropathy and Alzheimer's disease (AD). This review presents evidence indicating that, similarly to insulin-dependent metabolically active tissues such as fat and muscle, neurons also develop IR and thus cannot respond to the neurotrophic properties of insulin, resulting in neuronal injury, subsequent dysfunction and disease states.
Collapse
Affiliation(s)
- Bhumsoo Kim
- University of Michigan, Department of Neurology, Ann Arbor, MI 48109, USA.
| | | |
Collapse
|
35
|
De Angelis K, Senador DD, Mostarda C, Irigoyen MC, Morris M. Sympathetic overactivity precedes metabolic dysfunction in a fructose model of glucose intolerance in mice. Am J Physiol Regul Integr Comp Physiol 2012; 302:R950-7. [PMID: 22319048 DOI: 10.1152/ajpregu.00450.2011] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Consumption of high levels of fructose in humans and animals leads to metabolic and cardiovascular dysfunction. There are questions as to the role of the autonomic changes in the time course of fructose-induced dysfunction. C57/BL male mice were given tap water or fructose water (100 g/l) to drink for up to 2 mo. Groups were control (C), 15-day fructose (F15), and 60-day fructose (F60). Light-dark patterns of arterial pressure (AP) and heart rate (HR), and their respective variabilities were measured. Plasma glucose, lipids, insulin, leptin, resistin, adiponectin, and glucose tolerance were quantified. Fructose increased systolic AP (SAP) at 15 and 60 days during both light (F15: 123 ± 2 and F60: 118 ± 2 mmHg) and dark periods (F15: 136 ± 4 and F60: 136 ± 5 mmHg) compared with controls (light: 111 ± 2 and dark: 117 ± 2 mmHg). SAP variance (VAR) and the low-frequency component (LF) were increased in F15 (>60% and >80%) and F60 (>170% and >140%) compared with C. Cardiac sympatho-vagal balance was enhanced, while baroreflex function was attenuated in fructose groups. Metabolic parameters were unchanged in F15. However, F60 showed significant increases in plasma glucose (26%), cholesterol (44%), triglycerides (22%), insulin (95%), and leptin (63%), as well as glucose intolerance. LF of SAP was positively correlated with SAP. Plasma leptin was correlated with triglycerides, insulin, and glucose tolerance. Results show that increased sympathetic modulation of vessels and heart preceded metabolic dysfunction in fructose-consuming mice. Data suggest that changes in autonomic modulation may be an initiating mechanism underlying the cluster of symptoms associated with cardiometabolic disease.
Collapse
Affiliation(s)
- Katia De Angelis
- Laboratory of Translational Physiology, Nove de Julho University, São Paulo, Brazil
| | | | | | | | | |
Collapse
|