1
|
Sims SL, Frazier HN, Case SL, Lin RL, Trosper JN, Vekaria HJ, Sullivan PG, Thibault O. Variable bioenergetic sensitivity of neurons and astrocytes to insulin and extracellular glucose. NPJ METABOLIC HEALTH AND DISEASE 2024; 2:33. [PMID: 39524535 PMCID: PMC11549053 DOI: 10.1038/s44324-024-00037-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024]
Abstract
Energy flow within cellular elements of the brain is a well-orchestrated, tightly regulated process, however, details underlying these functions at the single-cell level are still poorly understood. Studying hypometabolism in aging and neurodegenerative diseases may benefit from experimentation on unicellular bioenergetics. Here, we examined energy status in neurons and astrocytes using mixed hippocampal cultures and PercevalHR, an ATP:ADP nanosensor. We assessed exposures of several compounds including KCl, glutamate, FCCP, insulin, and glucose. A mitochondrial stress test was performed, and PercevalHR's fluorescence was corrected for pH using pHrodo. Results demonstrate that PercevalHR can reliably report on the energetic status of two cell types that communicate in a mixed-culture setting. While KCl, glutamate, and FCCP showed clear changes in PercevalHR fluorescence, insulin and glucose responses were found to be more subtle and sensitive to extracellular glucose. These results may highlight mechanisms that mediate insulin sensitivity in the brain.
Collapse
Affiliation(s)
- Sophiya L. Sims
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY USA
| | - Hilaree N. Frazier
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY USA
| | - Sami L. Case
- Department of Biomedical Sciences, College of Veterinary Medicine & Biomedical Sciences, Colorado State University, Fort Collins, CO USA
| | - Ruei-Lung Lin
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY USA
| | - James N. Trosper
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY USA
| | - Hemendra J. Vekaria
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY USA
- Department of Neuroscience, University of Kentucky, Lexington, KY USA
| | - Patrick G. Sullivan
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY USA
- Department of Neuroscience, University of Kentucky, Lexington, KY USA
| | - Olivier Thibault
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY USA
| |
Collapse
|
2
|
Fonseca LM, Beeri MS, Hawks ZW, Jung L, Cleveland M, Delgado N, Bulger J, Grinspoon E, Janess K, Sliwinski MJ, Weinstock RS, Chhatwal JP, Kivisäkk P, Germine LT, Chaytor NS. ATN blood biomarkers are related to digital cognitive assessment in type 1 diabetes. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e70029. [PMID: 39445343 PMCID: PMC11497176 DOI: 10.1002/dad2.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/17/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION Associations between amyloid-tau-neurodegeneration (ATN) plasma biomarkers and cognition have not been characterized in adults with type 1 diabetes (T1D). METHODS Using data from participants in the Glycemic Variability and Fluctuations in Cognitive Status in Adults with T1D (GluCog) study (N = 114), we evaluated associations between phosphorylated tau (pTau)181, pTau217, β-amyloid 42/40 ratio, glial fibrillary acidic protein (GFAP), and neurofilament light (NfL) and self-administered digital cognitive tests, adjusting for age, sex, education, comorbidities (e.g., kidney disease), and glycemic indices. RESULTS Higher concentrations of pTau181 and GFAP were associated with slower responses on working memory tasks (pTau181: β = 0.261; p = 0.007; GFAP: β = 0.175, p = 0.036), and higher β-amyloid 42/40 ratio was associated with better vocabulary (β = 0.260, p = 0.009). Discussion Digital cognitive performance was associated with several ATN plasma biomarkers in T1D adults. Prospective studies are needed to understand the utility of these biomarkers in T1D. Highlights There is an increase in life expectancy for individuals with type 1 diabetes (T1D).Few studies investigate the relationship between T1D and neurodegeneration.We characterize the relation between ATN plasma biomarkers and cognitive function.Digital cognitive performance was associated with plasma biomarkers in T1D adults.
Collapse
Affiliation(s)
- Luciana Mascarenhas Fonseca
- Elson S. Floyd College of MedicineWashington State UniversitySpokaneWashingtonUSA
- Programa Terceira Idade (PROTER, Old Age Research Group)Department and Institute of PsychiatryUniversity of São Paulo School of MedicineSão PauloSão PauloBrazil
| | - Michal Schnaider Beeri
- Herbert and Jacqueline Krieger Klein Alzheimer's Research CenterRutgers UniversityNew BrunswickNew JerseyUSA
| | - Zoë W. Hawks
- Institute for Technology in PsychiatryMcLean HospitalBelmontMassachusettsUSA
- Department of PsychiatryHarvard Medical SchoolBostonMassachusettsUSA
| | - Lanee Jung
- Institute for Technology in PsychiatryMcLean HospitalBelmontMassachusettsUSA
- Department of PsychiatryHarvard Medical SchoolBostonMassachusettsUSA
| | - Michael Cleveland
- Department of Human DevelopmentWashington State UniversityPullmanWashingtonUSA
| | - Nikki Delgado
- Elson S. Floyd College of MedicineWashington State UniversitySpokaneWashingtonUSA
| | - Jane Bulger
- Department of MedicineSUNY Upstate Medical UniversitySyracuseNew YorkUSA
| | - Elizabeth Grinspoon
- Institute for Technology in PsychiatryMcLean HospitalBelmontMassachusettsUSA
| | - Kamille Janess
- Research Center and a HospitalJaeb Center for Health ResearchTampaFloridaUSA
| | - Martin J. Sliwinski
- Department of Human Development and Family StudiesPennsylvania State UniversityUniversity ParkState CollegePennsylvaniaUSA
- Center for Health AgingPennsylvania State UniversityUniversity ParkState CollegePennsylvaniaUSA
| | - Ruth S. Weinstock
- Department of MedicineSUNY Upstate Medical UniversitySyracuseNew YorkUSA
| | - Jasmeer P. Chhatwal
- Department of NeurologyMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
- Research Center and a HospitalBrigham and Women's HospitalBostonMassachusettsUSA
| | - Pia Kivisäkk
- Department of NeurologyMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Laura Thi Germine
- Institute for Technology in PsychiatryMcLean HospitalBelmontMassachusettsUSA
- Department of PsychiatryHarvard Medical SchoolBostonMassachusettsUSA
| | - Naomi S. Chaytor
- Elson S. Floyd College of MedicineWashington State UniversitySpokaneWashingtonUSA
| |
Collapse
|
3
|
Abedi A, Foroutan T, Mohaghegh Shalmani L, Dargahi L. Sex-dependent susceptibility to brain metabolic dysfunction and memory impairment in response to pre and postnatal high-fat diet. J Nutr Biochem 2024; 132:109675. [PMID: 38945454 DOI: 10.1016/j.jnutbio.2024.109675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/15/2024] [Accepted: 05/31/2024] [Indexed: 07/02/2024]
Abstract
The developing brain is sensitive to the impacts of early-life nutritional intake. This study investigates whether maternal high fat diet (HFD) causes glucose metabolism impairment, neuroinflammation, and memory impairment in immature and adult offspring, and whether it may be affected by postweaning diets in a sex-dependent manner in adult offspring. After weaning, female rats were fed HFD (55.9% fat) or normal chow diet (NCD; 10% fat) for 8 weeks before mating, during pregnancy, and lactation. On postnatal day 21 (PND21), the male and female offspring of both groups were split into two new groups, and NCD or HFD feeding was maintained until PND180. On PND21 and PND180, brain glucose metabolism, inflammation, and Alzheimer's pathology-related markers were by qPCR. In adult offspring, peripheral insulin resistance parameters, spatial memory performance, and brain glucose metabolism (18F-FDG-PET scan and protein levels of IDE and GLUT3) were assessed. Histological analysis was also performed on PND21 and adult offspring. On PND21, we found that maternal HFD affected transcript levels of glucose metabolism markers in both sexes. In adult offspring, more profoundly in males, postweaning HFD in combination with maternal HFD induced peripheral and brain metabolic disturbances, impaired memory performance and elevated inflammation, dementia risk markers, and neuronal loss. Our results suggest that maternal HFD affects brain glucose metabolism in the early ages of both sexes. Postweaning HFD sex-dependently causes brain metabolic dysfunction and memory impairment in later-life offspring; effects that can be worsened in combination with maternal HFD.
Collapse
Affiliation(s)
- Azam Abedi
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Tahereh Foroutan
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | - Leila Mohaghegh Shalmani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Griffen C, Cullen T, Hattersley J, Weickert MO, Dallaway A, Duncan M, Renshaw D. Effects of resistance exercise and whey protein supplementation on cognitive function in older men: secondary analysis of a randomised, double-blind, placebo-controlled trial. Exp Gerontol 2024; 193:112477. [PMID: 38844183 DOI: 10.1016/j.exger.2024.112477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 05/22/2024] [Accepted: 06/02/2024] [Indexed: 06/09/2024]
Abstract
PURPOSE Ageing is associated with cognitive decline. This study investigated the individual and combined effects of resistance exercise (RE) and whey protein supplementation (PRO) on cognitive function in older men. METHODS In a pooled-groups analysis, 36 older men (age: 67 ± 4 years) were randomised to either RE (2 x/week; n = 18) or no exercise (NE; n = 18), and either PRO (2 × 25 g/d whey protein isolate; n = 18) or control (CON, 2 × 23.75 g maltodextrin/d; n = 18). A sub-analysis was also conducted between RE + CON (n = 9) and RE + PRO (n = 9). At baseline and 12 weeks, participants completed a battery of neuropsychological tests (CANTAB; Cambridge Cognition, UK) and neurobiological, inflammatory, salivary cortisol and insulin sensitivity biomarkers were quantified. RESULTS PRO improved executive function z-score (+0.31 ± 0.08) greater than CON (+0.06 ± 0.08, P = 0.03) and there was a trend towards improved global cognitive function (P = 0.053). RE and RE + PRO did not improve any cognitive function domains (p ≥ 0.07). RE decreased tumor necrosis factor-alpha (P = 0.02) and interleukin-6 (P = 0.048) concentrations compared to NE, but changes in biomarkers did not correlate with changes in cognitive domains. Muscle strength (r = 0.34, P = 0.045) and physical function (ρ = 0.35-0.51, P < 0.05) outcomes positively correlated with cognitive function domains at baseline, but only Δskeletal muscle index correlated with Δepisodic memory (r = 0.34, P = 0.046) following the intervention. CONCLUSION In older men, PRO improved cognitive function, most notably executive functioning. RE did not improve any cognitive function domains but did decrease biomarkers of systemic inflammation. No synergistic effects were observed.
Collapse
Affiliation(s)
- Corbin Griffen
- Centre for Health and Life Sciences, Coventry University, Coventry CV1 2DS, United Kingdom; Human Metabolism Research Unit, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, United Kingdom.
| | - Tom Cullen
- Centre for Physical Activity, Sport and Exercise Sciences, Coventry University, Coventry CV1 2DS, United Kingdom
| | - John Hattersley
- Centre for Health and Life Sciences, Coventry University, Coventry CV1 2DS, United Kingdom; Human Metabolism Research Unit, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, United Kingdom; School of Engineering, University of Warwick, Coventry CV4 7HL, United Kingdom
| | - Martin O Weickert
- Centre for Health and Life Sciences, Coventry University, Coventry CV1 2DS, United Kingdom; Department of Endocrinology and Diabetes, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, United Kingdom; Warwick Medical School, University of Warwick, Coventry CV4 7AL, United Kingdom; School of Life Sciences, Faculty of Health and Life Sciences, Coventry University, Coventry CV1 2DS, United Kingdom
| | - Alexander Dallaway
- Centre for Health and Life Sciences, Coventry University, Coventry CV1 2DS, United Kingdom; Human Metabolism Research Unit, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, United Kingdom; Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, United Kingdom
| | - Michael Duncan
- Centre for Physical Activity, Sport and Exercise Sciences, Coventry University, Coventry CV1 2DS, United Kingdom; School of Life Sciences, Faculty of Health and Life Sciences, Coventry University, Coventry CV1 2DS, United Kingdom
| | - Derek Renshaw
- Centre for Health and Life Sciences, Coventry University, Coventry CV1 2DS, United Kingdom
| |
Collapse
|
5
|
Lanzillotta C, Tramutola A, Lanzillotta S, Greco V, Pagnotta S, Sanchini C, Di Angelantonio S, Forte E, Rinaldo S, Paone A, Cutruzzolà F, Cimini FA, Barchetta I, Cavallo MG, Urbani A, Butterfield DA, Di Domenico F, Paul BD, Perluigi M, Duarte JMN, Barone E. Biliverdin Reductase-A integrates insulin signaling with mitochondrial metabolism through phosphorylation of GSK3β. Redox Biol 2024; 73:103221. [PMID: 38843768 PMCID: PMC11190564 DOI: 10.1016/j.redox.2024.103221] [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: 04/28/2024] [Revised: 05/24/2024] [Accepted: 05/31/2024] [Indexed: 06/14/2024] Open
Abstract
Brain insulin resistance links the failure of energy metabolism with cognitive decline in both type 2 Diabetes Mellitus (T2D) and Alzheimer's disease (AD), although the molecular changes preceding overt brain insulin resistance remain unexplored. Abnormal biliverdin reductase-A (BVR-A) levels were observed in both T2D and AD and were associated with insulin resistance. Here, we demonstrate that reduced BVR-A levels alter insulin signaling and mitochondrial bioenergetics in the brain. Loss of BVR-A leads to IRS1 hyper-activation but dysregulates Akt-GSK3β complex in response to insulin, hindering the accumulation of pGSK3βS9 into the mitochondria. This event impairs oxidative phosphorylation and fosters the activation of the mitochondrial Unfolded Protein Response (UPRmt). Remarkably, we unveil that BVR-A is required to shuttle pGSK3βS9 into the mitochondria. Our data sheds light on the intricate interplay between insulin signaling and mitochondrial metabolism in the brain unraveling potential targets for mitigating the development of brain insulin resistance and neurodegeneration.
Collapse
Affiliation(s)
- Chiara Lanzillotta
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy
| | - Simona Lanzillotta
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy
| | - Viviana Greco
- Department of Basic Biotechnology, Perioperative and Intensive Clinics, Faculty of Medicine and Surgery, Catholic University of the Sacred Heart, L.go F.Vito 1, 00168, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A.Gemelli 8, 00168, Rome, Italy
| | - Sara Pagnotta
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy
| | - Caterina Sanchini
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, 00161, Rome, Italy
| | - Silvia Di Angelantonio
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, 00161, Rome, Italy; Department of Physiology and Pharmacology, Sapienza University of Rome, Italy
| | - Elena Forte
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy
| | - Serena Rinaldo
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy
| | - Alessio Paone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy
| | - Francesca Cutruzzolà
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy
| | | | - Ilaria Barchetta
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | | | - Andrea Urbani
- Department of Basic Biotechnology, Perioperative and Intensive Clinics, Faculty of Medicine and Surgery, Catholic University of the Sacred Heart, L.go F.Vito 1, 00168, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A.Gemelli 8, 00168, Rome, Italy
| | - D Allan Butterfield
- Sanders-Brown Center on Aging, Department of Chemistry, University of Kentucky, Lexington, KY, USA
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy
| | - Bindu D Paul
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy
| | - Joao M N Duarte
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Eugenio Barone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy.
| |
Collapse
|
6
|
Todorovic S, Simeunovic V, Prvulovic M, Dakic T, Jevdjovic T, Sokanovic S, Kanazir S, Mladenovic A. Dietary restriction alters insulin signaling pathway in the brain. Biofactors 2024; 50:450-466. [PMID: 37975613 DOI: 10.1002/biof.2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/07/2023] [Indexed: 11/19/2023]
Abstract
Insulin is known to be a key hormone in the regulation of peripheral glucose homeostasis, but beyond that, its effects on the brain are now undisputed. Impairments in insulin signaling in the brain, including changes in insulin levels, are thought to contribute significantly to declines in cognitive performance, especially during aging. As one of the most widely studied experimental interventions, dietary restriction (DR) is considered to delay the neurodegenerative processes associated with aging. Recently, however, data began to suggest that the onset and duration of a restrictive diet play a critical role in the putative beneficial outcome. Because the effects of DR on insulin signaling in the brain have been poorly studied, we decided to examine the effects of DR that differed in onset and duration: long-term DR (LTDR), medium-term DR (MTDR), and short-term DR (STDR) on the expression of proteins involved in insulin signaling in the hippocampus of 18- and 24-month-old male Wistar rats. We found that DR-induced changes in insulin levels in the brain may be independent of what happens in the periphery after restricted feeding. Significantly changed insulin content in the hippocampus, together with altered insulin signaling were found under the influence of DR, but the outcome was highly dependent on the onset and duration of DR.
Collapse
Affiliation(s)
- Smilja Todorovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Valentina Simeunovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milica Prvulovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Tamara Dakic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry "Ivan Djaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Tanja Jevdjovic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry "Ivan Djaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Srdjan Sokanovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Selma Kanazir
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Mladenovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
7
|
Liu Y, Fan L, Yang H, Wang D, Liu R, Shan T, Xia X. Ketogenic therapy towards precision medicine for brain diseases. Front Nutr 2024; 11:1266690. [PMID: 38450235 PMCID: PMC10915067 DOI: 10.3389/fnut.2024.1266690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 01/29/2024] [Indexed: 03/08/2024] Open
Abstract
Precision nutrition and nutrigenomics are emerging in the development of therapies for multiple diseases. The ketogenic diet (KD) is the most widely used clinical diet, providing high fat, low carbohydrate, and adequate protein. KD produces ketones and alters the metabolism of patients. Growing evidence suggests that KD has therapeutic effects in a wide range of neuronal diseases including epilepsy, neurodegeneration, cancer, and metabolic disorders. Although KD is considered to be a low-side-effect diet treatment, its therapeutic mechanism has not yet been fully elucidated. Also, its induced keto-response among different populations has not been elucidated. Understanding the ketone metabolism in health and disease is critical for the development of KD-associated therapeutics and synergistic therapy under any physiological background. Here, we review the current advances and known heterogeneity of the KD response and discuss the prospects for KD therapy from a precision nutrition perspective.
Collapse
Affiliation(s)
- Yang Liu
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, China
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, China
| | - Linlin Fan
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, China
| | - Haoying Yang
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, China
| | - Danli Wang
- Zhoushan People’s Hospital, Zhoushan, China
| | - Runhan Liu
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, China
| | - Tikun Shan
- Neurosurgery Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xue Xia
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, China
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
8
|
Réthelyi JM, Vincze K, Schall D, Glennon J, Berkel S. The role of insulin/IGF1 signalling in neurodevelopmental and neuropsychiatric disorders - Evidence from human neuronal cell models. Neurosci Biobehav Rev 2023; 153:105330. [PMID: 37516219 DOI: 10.1016/j.neubiorev.2023.105330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 07/15/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023]
Abstract
Insulin and insulin-like growth factor 1 (IGF1) signalling play a central role in the development and maintenance of neurons in the brain, and human neurodevelopmental as well as neuropsychiatric disorders have been linked to impaired insulin and IGF1 signalling. This review focuses on the impairments of the insulin and IGF1 signalling cascade in the context of neurodevelopmental and neuropsychiatric disorders, based on evidence from human neuronal cell models. Clear evidence was obtained for impaired insulin and IGF1 receptor downstream signalling in neurodevelopmental disorders, while the evidence for its role in neuropsychiatric disorders was less substantial. Human neuronal model systems can greatly add to our knowledge about insulin/IGF1 signalling in the brain, its role in restoring dendritic maturity, and complement results from clinical studies and animal models. Moreover, they represent a useful model for the development of new therapeutic strategies. Further research is needed to systematically investigate the exact role of the insulin/IGF1 signalling cascades in neurodevelopmental and neuropsychiatric disorders, and to elucidate the respective therapeutic implications.
Collapse
Affiliation(s)
- János M Réthelyi
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary
| | - Katalin Vincze
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary; Doctoral School of Mental Health Sciences, Semmelweis University, Budapest, Hungary
| | - Dorothea Schall
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Jeffrey Glennon
- Conway Institute of Biomedical and Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland
| | - Simone Berkel
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany; Interdisciplinary Centre of Neurosciences (IZN), Heidelberg University, Germany.
| |
Collapse
|
9
|
Wu S, Stogios N, Hahn M, Navagnanavel J, Emami Z, Chintoh A, Gerretsen P, Graff-Guerrero A, Rajji TK, Remington G, Agarwal SM. Outcomes and clinical implications of intranasal insulin on cognition in humans: A systematic review and meta-analysis. PLoS One 2023; 18:e0286887. [PMID: 37379265 DOI: 10.1371/journal.pone.0286887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/25/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND Aberrant brain insulin signaling has been posited to lie at the crossroads of several metabolic and cognitive disorders. Intranasal insulin (INI) is a non-invasive approach that allows investigation and modulation of insulin signaling in the brain while limiting peripheral side effects. OBJECTIVES The objective of this systematic review and meta-analysis is to evaluate the effects of INI on cognition in diverse patient populations and healthy individuals. METHODS MEDLINE, EMBASE, PsycINFO, and Cochrane CENTRAL were systematically searched from 2000 to July 2021. Eligible studies were randomized controlled trials that studied the effects of INI on cognition. Two independent reviewers determined study eligibility and extracted relevant descriptive and outcome data. RESULTS Twenty-nine studies (pooled N = 1,726) in healthy individuals as well as those with Alzheimer's disease (AD)/mild cognitive impairment (MCI), mental health disorders, metabolic disorders, among others, were included in the quantitative meta-analysis. Patients with AD/MCI treated with INI were more likely to show an improvement in global cognition (SMD = 0.22, 95% CI: 0.05-0.38 p = <0.00001, N = 12 studies). Among studies with healthy individuals and other patient populations, no significant effects of INI were found for global cognition. CONCLUSIONS This review demonstrates that INI may be associated with pro-cognitive benefits for global cognition, specifically for individuals with AD/MCI. Further studies are required to better understand the neurobiological mechanisms and differences in etiology to dissect the intrinsic and extrinsic factors contributing to the treatment response of INI.
Collapse
Affiliation(s)
- Sally Wu
- Schizophrenia Division, Centre for Addiction and Mental Health (CAMH), Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
| | - Nicolette Stogios
- Schizophrenia Division, Centre for Addiction and Mental Health (CAMH), Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
| | - Margaret Hahn
- Schizophrenia Division, Centre for Addiction and Mental Health (CAMH), Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, Canada
| | | | - Zahra Emami
- Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Araba Chintoh
- Schizophrenia Division, Centre for Addiction and Mental Health (CAMH), Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Philip Gerretsen
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
- Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, Canada
| | - Ariel Graff-Guerrero
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
- Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, Canada
| | - Tarek K Rajji
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
- Toronto Dementia Research Alliance, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Gary Remington
- Schizophrenia Division, Centre for Addiction and Mental Health (CAMH), Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Sri Mahavir Agarwal
- Schizophrenia Division, Centre for Addiction and Mental Health (CAMH), Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, Canada
| |
Collapse
|
10
|
Kooshki L, Zarneshan SN, Fakhri S, Moradi SZ, Echeverria J. The pivotal role of JAK/STAT and IRS/PI3K signaling pathways in neurodegenerative diseases: Mechanistic approaches to polyphenols and alkaloids. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 112:154686. [PMID: 36804755 DOI: 10.1016/j.phymed.2023.154686] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/10/2023] [Accepted: 01/28/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Neurodegenerative diseases (NDDs) are characterized by progressive neuronal dysfunctionality which results in disability and human life-threatening events. In recent decades, NDDs are on the rise. Besides, conventional drugs have not shown potential effectiveness to attenuate the complications of NDDs. So, exploring novel therapeutic agents is an urgent need to combat such disorders. Accordingly, growing evidence indicates that polyphenols and alkaloids are promising natural candidates, possessing several beneficial pharmacological effects against diseases. Considering the complex pathophysiological mechanisms behind NDDs, Janus kinase (JAK), insulin receptor substrate (IRS), phosphoinositide 3-kinase (PI3K), and signal transducer and activator of transcription (STAT) seem to play critical roles during neurodegeneration/neuroregeneration. In this line, modulation of the JAK/STAT and IRS/PI3K signaling pathways and their interconnected mediators by polyphenols/alkaloids could play pivotal roles in combating NDDs, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), stroke, aging, multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), depression and other neurological disorders. PURPOSE Thus, the present study aimed to investigate the neuroprotective roles of polyphenols/alkaloids as multi-target natural products against NDDs which are critically passing through the modulation of the JAK/STAT and IRS/PI3K signaling pathways. STUDY DESIGN AND METHODS A systematic and comprehensive review was performed to highlight the modulatory roles of polyphenols and alkaloids on the JAK/STAT and IRS/PI3K signaling pathways in NDDs, according to the PRISMA guideline, using scholarly electronic databases, including Scopus, PubMed, ScienceDirect, and associated reference lists. RESULTS In the present study 141 articles were included from a total of 1267 results. The results showed that phenolic compounds such as curcumin, epigallocatechin-3-gallate, and quercetin, and alkaloids such as berberine could be introduced as new strategies in combating NDDs through JAK/STAT and IRS/PI3K signaling pathways. This is the first systematic review that reveals the correlation between the JAK/STAT and IRS/PI3K axis which is targeted by phytochemicals in NDDs. Hence, this review highlighted promising insights into the neuroprotective potential of polyphenols and alkaloids through the JAK/STAT and IRS/PI3K signaling pathway and interconnected mediators toward neuroprotection. CONCLUSION Amongst natural products, phenolic compounds and alkaloids are multi-targeting agents with the most antioxidants and anti-inflammatory effects possessing the potential of combating NDDs with high efficacy and lower toxicity. However, additional reports are needed to prove the efficacy and possible side effects of natural products.
Collapse
Affiliation(s)
- Leila Kooshki
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran.
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran; Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Javier Echeverria
- Departamento de Ciencias del Ambiente, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile.
| |
Collapse
|
11
|
From Determining Brain Insulin Resistance in a Sporadic Alzheimer's Disease Model to Exploring the Region-Dependent Effect of Intranasal Insulin. Mol Neurobiol 2023; 60:2005-2023. [PMID: 36596966 DOI: 10.1007/s12035-022-03188-5] [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: 04/22/2022] [Accepted: 12/25/2022] [Indexed: 01/05/2023]
Abstract
Impaired response to insulin has been linked to many neurodegenerative disorders like Alzheimer's disease (AD). Animal model of sporadic AD has been developed by intracerebroventricular (icv) administration of streptozotocin (STZ), which given peripherally causes insulin resistance. Difficulty in demonstrating insulin resistance in this model led to our aim: to determine brain regional and peripheral response after intranasal (IN) administration of insulin in control and STZ-icv rats, by exploring peripheral and central metabolic parameters. One month after STZ-icv or vehicle-icv administration to 3-month-old male Wistar rats, cognitive status was determined after which rats received 2 IU of fast-acting insulin aspart intranasally (CTR + INS; STZ + INS) or saline only (CTR and STZ). Rats were sacrificed 2 h after administration and metabolic and glutamatergic parameters were measured in plasma, CSF, and the brain. Insulin and STZ increased amyloid-β concentration in plasma (CTR + INS and STZ vs CTR), while there was no effect on glucose and insulin plasma and CSF levels. INS normalized the levels of c-fos in temporal cortex of STZ + INS vs STZ (co-localized with neurons), while hypothalamic c-fos was found co-localized with the microglial marker. STZ and insulin brain region specifically altered the levels and activity of proteins involved in cell metabolism and glutamate signaling. Central changes found after INS in STZ-icv rats suggest hippocampal and cortical insulin sensitivity. Altered hypothalamic metabolic parameters of STZ-icv rats were not normalized by INS, indicating possible hypothalamic insulin insensitivity. Brain insulin sensitivity depends on the affected brain region and presence of metabolic dysfunction induced by STZ-icv administration.
Collapse
|
12
|
Abedi A, Foroutan T, Mohaghegh Shalmani L, Dargahi L. Sex-specific effects of high-fat diet on rat brain glucose metabolism and early-onset dementia symptoms. Mech Ageing Dev 2023; 211:111795. [PMID: 36828273 DOI: 10.1016/j.mad.2023.111795] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/08/2023] [Accepted: 02/21/2023] [Indexed: 02/24/2023]
Abstract
Peripheral metabolic disturbances are associated with a variety of clinical health consequences and may contribute to the development of neurocognitive disorders. This study investigates whether long-term high-fat diet (HFD) consumption changes the brain glucose metabolism and impairs memory performance in a sex-dependent manner. Male and female rats, after weaning, were fed HFD or normal chow diet (NCD) for 16 weeks. Behavioral tests for spatial memory and an 18 F-FDG-PET scan were performed. Also, the expression of brain insulin resistance markers and Alzheimer's pathology-related genes was assessed by qPCR. The Morris water maze and Y-maze results showed, respectively, that memory retrieval and spatial working memory were impaired only in HFD male rats compared to NCD controls. In addition, measuring whole brain 18 F-FDG uptake indicated a significant reduction in glucose metabolism in male but not female HFD rats. Analysis of 15 genes related to glucose metabolism and Alzheimer's pathology, in the hippocampus, showed that expression of GLUT3, IRS2, and IDE is significantly reduced in HFD male rats. Our results suggest that sex affects the HFD-induced dysregulation of brain glucose metabolism and cognitive performance.
Collapse
Affiliation(s)
- Azam Abedi
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Tahereh Foroutan
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | - Leila Mohaghegh Shalmani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Long-Term Ingestion of Sicilian Black Bee Chestnut Honey and/or D-Limonene Counteracts Brain Damage Induced by High Fat-Diet in Obese Mice. Int J Mol Sci 2023; 24:ijms24043467. [PMID: 36834882 PMCID: PMC9966634 DOI: 10.3390/ijms24043467] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/25/2023] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
Obesity is linked to neurodegeneration, which is mainly caused by inflammation and oxidative stress. We analyzed whether the long-term intake of honey and/or D-limonene, which are known for their antioxidant and anti-inflammatory actions, when ingested separately or in combination, can counteract the neurodegeneration occurring in high fat diet (HFD)-induced obesity. After 10 weeks of HFD, mice were divided into: HFD-, HFD + honey (HFD-H)-, HFD + D-limonene (HFD-L)-, HFD + honey + D-limonene (HFD-H + L)-fed groups, for another 10 weeks. Another group was fed a standard diet (STD). We analyzed the brain neurodegeneration, inflammation, oxidative stress, and gene expression of Alzheimer's disease (AD) markers. The HFD animals showed higher neuronal apoptosis, upregulation of pro-apoptotic genes Fas-L, Bim P27 and downregulation of anti-apoptotic factors BDNF and BCL2; increased gene expression of the pro-inflammatory IL-1β, IL-6 and TNF-α and elevated oxidative stress markers COX-2, iNOS, ROS and nitrite. The honey and D-limonene intake counteracted these alterations; however, they did so in a stronger manner when in combination. Genes involved in amyloid plaque processing (APP and TAU), synaptic function (Ache) and AD-related hyperphosphorylation were higher in HFD brains, and significantly downregulated in HFD-H, HFD-L and HFD-H + L. These results suggest that honey and limonene ingestion counteract obesity-related neurodegeneration and that joint consumption is more efficacious than a single administration.
Collapse
|
14
|
Bao H, Shen Y. Unmasking BACE1 in aging and age-related diseases. Trends Mol Med 2023; 29:99-111. [PMID: 36509631 DOI: 10.1016/j.molmed.2022.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/14/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022]
Abstract
The beta-site amyloid precursor protein (APP)-cleaving enzyme 1 (BACE1) has long been considered a conventional target for Alzheimer's disease (AD). Unfortunately, AD clinical trials of most BACE1 inhibitors were discontinued due to ineffective cognitive improvement or safety challenges. Recent studies investigating the involvement of BACE1 in metabolic, vascular, and immune functions have indicated a role in aging, diabetes, hypertension, and cancer. These novel BACE1 functions have helped to identify new 'druggable' targets for BACE1 against aging comorbidities. In this review, we discuss BACE1 regulation during aging, and then provide recent insights into its enzymatic and nonenzymatic involvement in aging and age-related diseases. Our study not only proposes the perspective of BACE1's actions in various systems, but also provides new directions for using BACE1 inhibitors and modulators to delay aging and to treat age-related diseases.
Collapse
Affiliation(s)
- Hong Bao
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China; Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yong Shen
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China; Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Provincial Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, 230026, China; CAS Key Laboratory of Brain Function and Disease, Division of Biological and Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China; Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
15
|
Amin AM, Mostafa H, Khojah HMJ. Insulin resistance in Alzheimer's disease: The genetics and metabolomics links. Clin Chim Acta 2023; 539:215-236. [PMID: 36566957 DOI: 10.1016/j.cca.2022.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/16/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with significant socioeconomic burden worldwide. Although genetics and environmental factors play a role, AD is highly associated with insulin resistance (IR) disorders such as metabolic syndrome (MS), obesity, and type two diabetes mellitus (T2DM). These findings highlight a shared pathogenesis. The use of metabolomics as a downstream systems' biology (omics) approach can help to identify these shared metabolic traits and assist in the early identification of at-risk groups and potentially guide therapy. Targeting the shared AD-IR metabolic trait with lifestyle interventions and pharmacological treatments may offer promising AD therapeutic approach. In this narrative review, we reviewed the literature on the AD-IR pathogenic link, the shared genetics and metabolomics biomarkers between AD and IR disorders, as well as the lifestyle interventions and pharmacological treatments which target this pathogenic link.
Collapse
Affiliation(s)
- Arwa M Amin
- Department of Clinical and Hospital Pharmacy, College of Pharmacy, Taibah University, Madinah, Saudi Arabia.
| | - Hamza Mostafa
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Food Innovation Network (XIA), Nutrition and Food Safety Research Institute (INSA), Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), 08028 Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Hani M J Khojah
- Department of Clinical and Hospital Pharmacy, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| |
Collapse
|
16
|
Deery HA, Di Paolo R, Moran C, Egan GF, Jamadar SD. Lower brain glucose metabolism in normal ageing is predominantly frontal and temporal: A systematic review and pooled effect size and activation likelihood estimates meta-analyses. Hum Brain Mapp 2022; 44:1251-1277. [PMID: 36269148 PMCID: PMC9875940 DOI: 10.1002/hbm.26119] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/29/2022] [Accepted: 10/05/2022] [Indexed: 01/31/2023] Open
Abstract
This review provides a qualitative and quantitative analysis of cerebral glucose metabolism in ageing. We undertook a systematic literature review followed by pooled effect size and activation likelihood estimates (ALE) meta-analyses. Studies were retrieved from PubMed following the PRISMA guidelines. After reviewing 635 records, 21 studies with 22 independent samples (n = 911 participants) were included in the pooled effect size analyses. Eight studies with eleven separate samples (n = 713 participants) were included in the ALE analyses. Pooled effect sizes showed significantly lower cerebral metabolic rates of glucose for older versus younger adults for the whole brain, as well as for the frontal, temporal, parietal, and occipital lobes. Among the sub-cortical structures, the caudate showed a lower metabolic rate among older adults. In sub-group analyses controlling for changes in brain volume or partial volume effects, the lower glucose metabolism among older adults in the frontal lobe remained significant, whereas confidence intervals crossed zero for the other lobes and structures. The ALE identified nine clusters of lower glucose metabolism among older adults, ranging from 200 to 2640 mm3 . The two largest clusters were in the left and right inferior frontal and superior temporal gyri and the insula. Clusters were also found in the inferior temporal junction, the anterior cingulate and caudate. Taken together, the results are consistent with research showing less efficient glucose metabolism in the ageing brain. The findings are discussed in the context of theories of cognitive ageing and are compared to those found in neurodegenerative disease.
Collapse
Affiliation(s)
- Hamish A. Deery
- Turner Institute for Brain and Mental HealthMonash UniversityMelbourneAustralia,Monash Biomedical ImagingMonash UniversityMelbourneAustralia
| | - Robert Di Paolo
- Turner Institute for Brain and Mental HealthMonash UniversityMelbourneAustralia,Monash Biomedical ImagingMonash UniversityMelbourneAustralia
| | - Chris Moran
- Peninsula Clinical School, Central Clinical SchoolMonash UniversityFrankstonVictoriaAustralia,Department of Geriatric MedicinePeninsula HealthFrankstonVictoriaAustralia
| | - Gary F. Egan
- Turner Institute for Brain and Mental HealthMonash UniversityMelbourneAustralia,Monash Biomedical ImagingMonash UniversityMelbourneAustralia,Australian Research Council Centre of Excellence for Integrative Brain FunctionMelbourneAustralia
| | - Sharna D. Jamadar
- Turner Institute for Brain and Mental HealthMonash UniversityMelbourneAustralia,Monash Biomedical ImagingMonash UniversityMelbourneAustralia,Australian Research Council Centre of Excellence for Integrative Brain FunctionMelbourneAustralia
| |
Collapse
|
17
|
Engel MG, Smith J, Mao K, Quipildor GF, Cui MH, Gulinello M, Branch CA, Gandy SE, Huffman DM. Evidence for preserved insulin responsiveness in the aging rat brain. GeroScience 2022; 44:2491-2508. [PMID: 35798912 PMCID: PMC9768080 DOI: 10.1007/s11357-022-00618-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/27/2022] [Indexed: 01/06/2023] Open
Abstract
Insulin appears to exert salutary effects in the central nervous system (CNS). Thus, brain insulin resistance has been proposed to play a role in brain aging and dementia but is conceptually complex and unlikely to fit classic definitions established in peripheral tissues. Thus, we sought to characterize brain insulin responsiveness in young (4-5 months) and old (24 months) FBN male rats using a diverse set of assays to determine the extent to which insulin effects in the CNS are impaired with age. When performing hyperinsulinemic-euglycemic clamps in rats, intracerebroventricular (ICV) infusion of insulin in old animals improved peripheral insulin sensitivity by nearly two-fold over old controls and comparable to young rats, suggesting preservation of this insulin-triggered response in aging per se (p < 0.05). We next used an imaging-based approach by comparing ICV vehicle versus insulin and performed resting state functional magnetic resonance imaging (rs-fMRI) to evaluate age- and insulin-related changes in network connectivity within the default mode network. In aging, lower connectivity between the mesial temporal (MT) region and other areas, as well as reduced MT signal complexity, was observed in old rats, which correlated with greater cognitive deficits in old. Despite these stark differences, ICV insulin failed to elicit any significant alteration to the BOLD signal in young rats, while a significant deviation of the BOLD signal was observed in older animals, characterized by augmentation in regions of the septal nucleus and hypothalamus, and reduction in thalamus and nucleus accumbens. In contrast, ex vivo stimulation of hippocampus with 10 nM insulin revealed increased Akt activation in young (p < 0.05), but not old rats. Despite similar circulating levels of insulin and IGF-1, cerebrospinal fluid concentrations of these ligands were reduced with age. Thus, these data highlight the complexity of capturing brain insulin action and demonstrate preserved or heightened brain responses to insulin with age, despite dampened canonical signaling, thereby suggesting impaired CNS input of these ligands may be a feature of reduced brain insulin action, providing further rationale for CNS replacement strategies.
Collapse
Affiliation(s)
- Matthew G Engel
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Research, Albert Einstein College of Medicine, 1300 Morris Park Ave, Golding Building Room 201, BronxBronx, NY, 10461, USA
| | - Jeremy Smith
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Kai Mao
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Research, Albert Einstein College of Medicine, 1300 Morris Park Ave, Golding Building Room 201, BronxBronx, NY, 10461, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Gabriela Farias Quipildor
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Research, Albert Einstein College of Medicine, 1300 Morris Park Ave, Golding Building Room 201, BronxBronx, NY, 10461, USA
| | - Min-Hui Cui
- Department of Radiology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Maria Gulinello
- Dominick S. Purpura Department of Neuroscience, Behavioral Core Facility, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Craig A Branch
- Department of Radiology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Samuel E Gandy
- Department of Neurology and the Mount Sinai Center for Cognitive Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry and the Mount Sinai Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Derek M Huffman
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Institute for Aging Research, Albert Einstein College of Medicine, 1300 Morris Park Ave, Golding Building Room 201, BronxBronx, NY, 10461, USA.
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
18
|
Falling Short: The Contribution of Central Insulin Receptors to Gait Dysregulation in Brain Aging. Biomedicines 2022; 10:biomedicines10081923. [PMID: 36009470 PMCID: PMC9405648 DOI: 10.3390/biomedicines10081923] [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: 06/15/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/23/2022] Open
Abstract
Insulin resistance, which manifests as a reduction of insulin receptor signaling, is known to correlate with pathological changes in peripheral tissues as well as in the brain. Central insulin resistance has been associated with impaired cognitive performance, decreased neuronal health, and reduced brain metabolism; however, the mechanisms underlying central insulin resistance and its impact on brain regions outside of those associated with cognition remain unclear. Falls are a leading cause of both fatal and non-fatal injuries in the older population. Despite this, there is a paucity of work focused on age-dependent alterations in brain regions associated with ambulatory control or potential therapeutic approaches to target these processes. Here, we discuss age-dependent alterations in central modalities that may contribute to gait dysregulation, summarize current data supporting the role of insulin signaling in the brain, and highlight key findings that suggest insulin receptor sensitivity may be preserved in the aged brain. Finally, we present novel results showing that administration of insulin to the somatosensory cortex of aged animals can alter neuronal communication, cerebral blood flow, and the motivation to ambulate, emphasizing the need for further investigations of intranasal insulin as a clinical management strategy in the older population.
Collapse
|
19
|
Schneider E, Spetter MS, Martin E, Sapey E, Yip KP, Manolopoulos KN, Tahrani AA, Thomas JM, Lee M, Hallschmid M, Rotshtein P, Dourish CT, Higgs S. The effect of intranasal insulin on appetite and mood in women with and without obesity: an experimental medicine study. Int J Obes (Lond) 2022; 46:1319-1327. [PMID: 35397638 PMCID: PMC9239904 DOI: 10.1038/s41366-022-01115-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND/OBJECTIVES Intranasal (IN) administration of insulin decreases appetite in humans, but the underlying mechanisms are unclear, and it is unknown whether IN insulin affects the food intake of women with obesity. SUBJECTS/METHODS In a double-blind, placebo-controlled, crossover design, participants (35 lean women and 17 women with obesity) were randomized to receive 160 IU/1.6 mL of IN insulin or placebo in a counterbalanced order in the post prandial state. The effects of IN insulin on cookie intake, appetite, mood, food reward, cognition and neural activity were assessed. RESULTS IN insulin in the post prandial state reduced cookie intake, appetite and food reward relative to placebo and these effects were more pronounced for women with obesity compared with lean women. IN insulin also improved mood in women with obesity. In both BMI groups, IN insulin increased neural activity in the insula when viewing food pictures. IN insulin did not affect cognitive function. CONCLUSIONS These results suggest that IN insulin decreases palatable food intake when satiated by reducing food reward and that women with obesity may be more sensitive to this effect than lean women. Further investigation of the therapeutic potential of IN insulin for weight management in women with obesity is warranted.
Collapse
Affiliation(s)
- Elizabeth Schneider
- School of Psychology, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Maartje S Spetter
- School of Psychology, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Centre for Human Brain Health, University of Birmingham, Birmingham, UK
| | - Elizabeth Martin
- School of Psychology, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Centre for Human Brain Health, University of Birmingham, Birmingham, UK
| | - Elizabeth Sapey
- Birmingham Acute Care Research Group, University of Birmingham, Birmingham, UK
| | - Kay Por Yip
- Birmingham Acute Care Research Group, University of Birmingham, Birmingham, UK
- University of Birmingham Institute of Inflammation and Ageing, Birmingham, UK
| | - Konstantinos N Manolopoulos
- Institute of Metabolism and Systems Research (IMSR), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Abd A Tahrani
- Institute of Metabolism and Systems Research (IMSR), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | | | - Michelle Lee
- Department of Psychology, Swansea University, Swansea, UK
| | - Manfred Hallschmid
- Department of Medical Psychology and Behavioural Neurobiology, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Tübingen, Germany
| | - Pia Rotshtein
- School of Psychology, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Centre for Human Brain Health, University of Birmingham, Birmingham, UK
| | - Colin T Dourish
- P1vital Ltd., Wallingford, UK
- P1vital Products Ltd, Wallingford, UK
| | - Suzanne Higgs
- School of Psychology, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
20
|
Lin RL, Frazier HN, Anderson KL, Case SL, Ghoweri AO, Thibault O. Sensitivity of the S1 neuronal calcium network to insulin and Bay-K 8644 in vivo: Relationship to gait, motivation, and aging processes. Aging Cell 2022; 21:e13661. [PMID: 35717599 PMCID: PMC9282843 DOI: 10.1111/acel.13661] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/10/2022] [Accepted: 06/05/2022] [Indexed: 01/25/2023] Open
Abstract
Neuronal hippocampal Ca2+ dysregulation is a critical component of cognitive decline in brain aging and Alzheimer's disease and is suggested to impact communication and excitability through the activation of a larger after hyperpolarization. However, few studies have tested for the presence of Ca2+ dysregulation in vivo, how it manifests, and whether it impacts network function across hundreds of neurons. Here, we tested for neuronal Ca2+ network dysregulation in vivo in the primary somatosensory cortex (S1) of anesthetized young and aged male Fisher 344 rats using single‐cell resolution techniques. Because S1 is involved in sensory discrimination and proprioception, we tested for alterations in ambulatory performance in the aged animal and investigated two potential pathways underlying these central aging‐ and Ca2+‐dependent changes. Compared to young, aged animals displayed increased overall activity and connectivity of the network as well as decreased ambulatory speed. In aged animals, intranasal insulin (INI) increased network synchronicity and ambulatory speed. Importantly, in young animals, delivery of the L‐type voltage‐gated Ca2+ channel modifier Bay‐K 8644 altered network properties, replicating some of the changes seen in the older animal. These results suggest that hippocampal Ca2+ dysregulation may be generalizable to other areas, such as S1, and might engage modalities that are associated with locomotor stability and motivation to ambulate. Further, given the safety profile of INI in the clinic and the evidence presented here showing that this central dysregulation is sensitive to insulin, we suggest that these processes can be targeted to potentially increase motivation and coordination while also reducing fall frequency with age.
Collapse
Affiliation(s)
- Ruei-Lung Lin
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Hilaree N Frazier
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Katie L Anderson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Sami L Case
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Adam O Ghoweri
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Olivier Thibault
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
21
|
Colca JR, Finck BN. Metabolic Mechanisms Connecting Alzheimer's and Parkinson's Diseases: Potential Avenues for Novel Therapeutic Approaches. Front Mol Biosci 2022; 9:929328. [PMID: 35782864 PMCID: PMC9243557 DOI: 10.3389/fmolb.2022.929328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's (AD) and Parkinson's Diseases (PD) are common neurodegenerative disorders growing in incidence and prevalence and for which there are no disease-modifying treatments. While there are considerable complexities in the presentations of these diseases, the histological pictures of these pathologies, as well as several rare genetic predispositions for each, point to the involvement of maladaptive protein processing and inflammation. Importantly, the common presentations of AD and PD are connected to aging and to dysmetabolism, including common co-diagnosis of metabolic syndrome or diabetes. Examination of anti-diabetic therapies in preclinical models and in some observational clinical studies have suggested effectiveness of the first generation insulin sensitizer pioglitazone in both AD and PD. Recently, the mitochondrial pyruvate carrier (MPC) was shown to be a previously unrecognized target of pioglitazone. New insulin sensitizers are in development that can be dosed to full engagement of this previously unappreciated mitochondrial target. Here we review molecular mechanisms that connect modification of pyruvate metabolism with known liabilities of AD and PD. The mechanisms involve modification of autophagy, inflammation, and cell differentiation in various cell types including neurons, glia, macrophages, and endothelium. These observations have implications for the understanding of the general pathology of neurodegeneration and suggest general therapeutic approaches to disease modification.
Collapse
Affiliation(s)
- Jerry R. Colca
- Metabolic Solutions Development Company, Western Michigan University, Kalamazoo, MI, United States
| | - Brian N. Finck
- Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
22
|
Mietelska-Porowska A, Domańska J, Want A, Więckowska-Gacek A, Chutorański D, Koperski M, Wojda U. Induction of Brain Insulin Resistance and Alzheimer's Molecular Changes by Western Diet. Int J Mol Sci 2022; 23:ijms23094744. [PMID: 35563135 PMCID: PMC9102094 DOI: 10.3390/ijms23094744] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 02/06/2023] Open
Abstract
The term Western diet (WD) describes the consumption of large amounts of highly processed foods, rich in simple sugars and saturated fats. Long-term WD feeding leads to insulin resistance, postulated as a risk factor for Alzheimer’s disease (AD). AD is the main cause of progressive dementia characterized by the deposition of amyloid-β (Aβ) plaques and neurofibrillary tangles consisting of the hyperphosphorylated tau (p-Tau) protein in the brain, starting from the entorhinal cortex and the hippocampus. In this study, we report that WD-derived impairment in insulin signaling induces tau and Aβ brain pathology in wild-type C57BL/6 mice, and that the entorhinal cortex is more sensitive than the hippocampus to the impairment of brain insulin signaling. In the brain areas developing WD-induced insulin resistance, we observed changes in p-Tau(Thr231) localization in neuronal subcellular compartments, indicating progressive tauopathy, and a decrease in amyloid precursor protein levels correlating with the appearance of Aβ peptides. These results suggest that WD promotes the development of AD and may be considered not only a risk factor, but also a modifiable trigger of AD.
Collapse
|
23
|
Lanz M, Janeiro MH, Milagro FI, Puerta E, Ludwig IA, Pineda-Lucena A, Ramírez MJ, Solas M. Trimethylamine N-Oxide (TMAO) drives insulin resistance and cognitive deficiencies in a senescence accelerated mouse model. Mech Ageing Dev 2022; 204:111668. [PMID: 35341897 DOI: 10.1016/j.mad.2022.111668] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/09/2022] [Accepted: 03/22/2022] [Indexed: 12/20/2022]
Abstract
It has been established that ageing is the major risk factor for cognitive deficiency and it is becoming increasingly evident that insulin resistance is another factor. Biological plausibility for a link between insulin resistance and dementia is relevant for understanding disease etiology, and to form bases for prevention efforts to decrease disease burden. In the present study, peripheral and central insulin resistance was found in SAMP8 mice (aging mouse model) accompanied by cognitive deficiencies. Furthermore, a marked peripheral inflammatory state was observed in SAMP8 mice, followed by neuroinflammation that could be due to a higher cytokine leaking into the brain across an aging-disrupted blood brain barrier. Moreover, aging-induced gut dysbiosis produces higher TMAO that could also contribute to the peripheral and central inflammatory tone as well as to the cognitive deficiencies observed in SAMP8 mice. All those alterations were reversed by DMB, a treatment that decreases TMAO levels. Data obtained from this project suggest that microbial dysbiosis and increased TMAO secretion could be a key link between aging, insulin resistance and dementia. Thus, pharmacological intervention that leads to decreased TMAO levels, such as DMB, could open a new avenue for the future treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- María Lanz
- Department of Pharmacology and Toxicology, University of Navarra, Pamplona, Spain
| | - Manuel H Janeiro
- Department of Pharmacology and Toxicology, University of Navarra, Pamplona, Spain; IdISNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Fermin I Milagro
- IdISNA, Navarra Institute for Health Research, Pamplona, Spain; Department of Nutrition, Food Science and Physiology, Centre for Nutrition Research, University of Navarra, Pamplona, Spain; CIBERobn, CIBER Fisiopatología de Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Elena Puerta
- Department of Pharmacology and Toxicology, University of Navarra, Pamplona, Spain; IdISNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Iziar A Ludwig
- Program of Molecular Therapeutics, Center for Applied Medical Research (CIMA), Universidad de Navarra, Avda. Pío XII 55, E-31008 Pamplona, Spain
| | - Antonio Pineda-Lucena
- Program of Molecular Therapeutics, Center for Applied Medical Research (CIMA), Universidad de Navarra, Avda. Pío XII 55, E-31008 Pamplona, Spain
| | - María J Ramírez
- Department of Pharmacology and Toxicology, University of Navarra, Pamplona, Spain; IdISNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Maite Solas
- Department of Pharmacology and Toxicology, University of Navarra, Pamplona, Spain; IdISNA, Navarra Institute for Health Research, Pamplona, Spain.
| |
Collapse
|
24
|
Mohseni-Moghaddam P, Ghobadian R, Khaleghzadeh-Ahangar H. Dementia in Diabetes mellitus and Atherosclerosis; Two Interrelated Systemic Diseases. Brain Res Bull 2022; 181:87-96. [DOI: 10.1016/j.brainresbull.2022.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 12/18/2021] [Accepted: 01/24/2022] [Indexed: 12/06/2022]
|
25
|
Busquets O, Carrasco M, Espinosa-Jiménez T, Ettcheto M, Verdaguer E, Auladell C, Bullò M, Camins A, Pinent M, Rodríguez-Gallego E, Folch J. GSPE pre-treatment protects against long-term cafeteria diet-induced mitochondrial and inflammatory affectations in the hippocampus of rats. Nutr Neurosci 2021; 25:2627-2637. [PMID: 34789070 DOI: 10.1080/1028415x.2021.1995118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Deregulations like the loss of sensitivity to insulin (insulin resistance) and chronic inflammation are alterations very commonly found in sporadic forms of neurodegenerative pathologies. Thus, finding strategies to protect against them, may lead to a reduction in the incidence and/or affectation of these pathologies. The grape seed-derived proanthocyanidins extract (GSPE) is a mixture of compounds highly enriched in polyphenols and flavonoids that have shown to have a wide range of therapeutic benefits due to their antioxidant and anti-inflammatory properties. OBJECTIVES This study aimed to assess the protective effects of a short pre-treatment of GSPE in the hippocampus against a prolonged feeding with cafeteria diet. METHODS GSPE was administered for 10 days followed by 12 weeks of cafeteria diet. We analyzed transcriptional activity of genes and protein expression of key mediators of neurodegeneration in brain samples. RESULTS Results indicated that GSPE was able to protect against cellular damage through the activation of AKT, as well as promote the maintenance of mitochondrial function by conserving the OXPHOS complexes and upregulating the antioxidant SOD. DISCUSSION We observed that GSPE decreased inflammatory activation as observed through the downregulation of JNK, IL6 and TNFα, just like the reduction in reactive profile of astrocytes. Overall, the data presented here offers an interesting and hopeful initial step for future long-term studies on the beneficial effects of a supplementation of common diets with polyphenol and flavonoid substances for the amelioration of typical early hallmarks of neurodegeneration.
Collapse
Affiliation(s)
- Oriol Busquets
- Department of Biochemistry and Biotechnology, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, Reus, Spain.,Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacy and Food Sciences Faculty, University of Barcelona, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Marina Carrasco
- Department of Biochemistry and Biotechnology, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, Reus, Spain.,Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacy and Food Sciences Faculty, University of Barcelona, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Triana Espinosa-Jiménez
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacy and Food Sciences Faculty, University of Barcelona, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Miren Ettcheto
- Department of Biochemistry and Biotechnology, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, Reus, Spain.,Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacy and Food Sciences Faculty, University of Barcelona, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Ester Verdaguer
- Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Cell Biology, Physiology and Immunology, Biology Faculty, University of Barcelona, Barcelona, Spain
| | - Carme Auladell
- Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Cell Biology, Physiology and Immunology, Biology Faculty, University of Barcelona, Barcelona, Spain
| | - Mònica Bullò
- Department of Biochemistry and Biotechnology, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, Reus, Spain.,Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN) dirigido por Jordi Salas en la Fundación Instituto de Investigación Sanitaria Pere Virgili, Tarragona, Spain.,Fundació Institut d'Investigació Sanitaria Pere Virgili (IISPV), Tarragona, Spain
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacy and Food Sciences Faculty, University of Barcelona, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Montserrat Pinent
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, Tarragona, Spain
| | - Esther Rodríguez-Gallego
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, Tarragona, Spain
| | - Jaume Folch
- Department of Biochemistry and Biotechnology, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, Reus, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Fundació Institut d'Investigació Sanitaria Pere Virgili (IISPV), Tarragona, Spain
| |
Collapse
|
26
|
Alzheimer's Disease and Type 2 Diabetes Mellitus: The Use of MCT Oil and a Ketogenic Diet. Int J Mol Sci 2021; 22:ijms222212310. [PMID: 34830192 PMCID: PMC8624628 DOI: 10.3390/ijms222212310] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/11/2021] [Accepted: 11/11/2021] [Indexed: 12/20/2022] Open
Abstract
Recently, type 2 diabetes mellitus (T2DM) has been reported to be strongly associated with Alzheimer’s disease (AD). This is partly due to insulin resistance in the brain. Insulin signaling and the number of insulin receptors may decline in the brain of T2DM patients, resulting in impaired synaptic formation, neuronal plasticity, and mitochondrial metabolism. In AD patients, hypometabolism of glucose in the brain is observed before the onset of symptoms. Amyloid-β accumulation, a main pathology of AD, also relates to impaired insulin action and glucose metabolism, although ketone metabolism is not affected. Therefore, the shift from glucose metabolism to ketone metabolism may be a reasonable pathway for neuronal protection. To promote ketone metabolism, medium-chain triglyceride (MCT) oil and a ketogenic diet could be introduced as an alternative source of energy in the brain of AD patients.
Collapse
|
27
|
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
|
28
|
Gadhave K, Kumar D, Uversky VN, Giri R. A multitude of signaling pathways associated with Alzheimer's disease and their roles in AD pathogenesis and therapy. Med Res Rev 2021; 41:2689-2745. [PMID: 32783388 PMCID: PMC7876169 DOI: 10.1002/med.21719] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/13/2020] [Accepted: 07/29/2020] [Indexed: 02/06/2023]
Abstract
The exact molecular mechanisms associated with Alzheimer's disease (AD) pathology continue to represent a mystery. In the past decades, comprehensive data were generated on the involvement of different signaling pathways in the AD pathogenesis. However, the utilization of signaling pathways as potential targets for the development of drugs against AD is rather limited due to the immense complexity of the brain and intricate molecular links between these pathways. Therefore, finding a correlation and cross-talk between these signaling pathways and establishing different therapeutic targets within and between those pathways are needed for better understanding of the biological events responsible for the AD-related neurodegeneration. For example, autophagy is a conservative cellular process that shows link with many other AD-related pathways and is crucial for maintenance of the correct cellular balance by degrading AD-associated pathogenic proteins. Considering the central role of autophagy in AD and its interplay with many other pathways, the finest therapeutic strategy to fight against AD is the use of autophagy as a target. As an essential step in this direction, this comprehensive review represents recent findings on the individual AD-related signaling pathways, describes key features of these pathways and their cross-talk with autophagy, represents current drug development, and introduces some of the multitarget beneficial approaches and strategies for the therapeutic intervention of AD.
Collapse
Affiliation(s)
- Kundlik Gadhave
- School of Basic Sciences, Indian Institute of Technology Mandi, Kamand, Himachal Pradesh, 175005, India
| | - Deepak Kumar
- School of Basic Sciences, Indian Institute of Technology Mandi, Kamand, Himachal Pradesh, 175005, India
| | - Vladimir N. Uversky
- Department of Molecular Medicine and Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
- Laboratory of New Methods in Biology, Institute for Biological Instrumentation, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Rajanish Giri
- School of Basic Sciences, Indian Institute of Technology Mandi, Kamand, Himachal Pradesh, 175005, India
| |
Collapse
|
29
|
Glial glucose fuels the neuronal pentose phosphate pathway for long-term memory. Cell Rep 2021; 36:109620. [PMID: 34433052 PMCID: PMC8411112 DOI: 10.1016/j.celrep.2021.109620] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 07/22/2021] [Accepted: 08/05/2021] [Indexed: 01/10/2023] Open
Abstract
Brain function relies almost solely on glucose as an energy substrate. The main model of brain metabolism proposes that glucose is taken up and converted into lactate by astrocytes to fuel the energy-demanding neuronal activity underlying plasticity and memory. Whether direct neuronal glucose uptake is required for memory formation remains elusive. We uncover, in Drosophila, a mechanism of glucose shuttling to neurons from cortex glia, an exclusively perisomatic glial subtype, upon formation of olfactory long-term memory (LTM). In vivo imaging reveals that, downstream of cholinergic activation of cortex glia, autocrine insulin signaling increases glucose concentration in glia. Glucose is then transferred from glia to the neuronal somata in the olfactory memory center to fuel the pentose phosphate pathway and allow LTM formation. In contrast, our results indicate that the increase in neuronal glucose metabolism, although crucial for LTM formation, is not routed to glycolysis. Neuronal glucose metabolism is increased upon long-term memory formation Glial cells shuttle glucose to neurons following insulin signaling activation Glucose fuels the neuronal pentose phosphate pathway
Collapse
|
30
|
Ebersole J, Rose G, Eid T, Behar K, Patrylo P. Altered hippocampal astroglial metabolism is associated with aging and preserved spatial learning and memory. Neurobiol Aging 2021; 102:188-199. [PMID: 33774381 DOI: 10.1016/j.neurobiolaging.2021.02.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 11/23/2022]
Abstract
An age-related decrease in hippocampal metabolism correlates with cognitive decline. Hippocampus-dependent learning and memory requires glutamatergic neurotransmission supported by glutamate-glutamine (GLU-GLN) cycling between neurons and astrocytes. We examined whether GLU-GLN cycling in hippocampal subregions (dentate gyrus and CA1) in Fischer 344 rats was altered with age and cognitive status. Hippocampal slices from young adult, aged cognitively-unimpaired (AU) and aged cognitively-impaired (AI) rats were incubated in artificial cerebrospinal fluid (aCSF) containing 1-13C-glucose to assess neural metabolism. Incorporation of 13C-glucose into glutamate and glutamine, measured by mass spectroscopy/liquid chromatography tandem mass spectroscopy, did not significantly differ between groups. However, when 13C-acetate, a preferential astrocytic metabolite, was used, a significant increase in 13C-labeled glutamate was observed in slices from AU rats. Taken together, the data suggest that resting state neural metabolism and GLU-GLN cycling may be preserved during aging when sufficient extracellular glucose is available, but that enhanced astroglial metabolism can occur under resting state conditions. This may be an aging-related compensatory change to maintain hippocampus-dependent cognitive function.
Collapse
Affiliation(s)
- Jeremy Ebersole
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL, USA
| | - Gregory Rose
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL, USA; Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, IL, USA; Center for Integrated Research in the Cognitive and Neural Sciences, Southern Illinois University School of Medicine, Carbondale, IL, USA
| | - Tore Eid
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Kevin Behar
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; MRRC Neurometabolism Research Laboratory, Yale University School of Medicine, New Haven, CT, USA
| | - Peter Patrylo
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL, USA; Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, IL, USA; Center for Integrated Research in the Cognitive and Neural Sciences, Southern Illinois University School of Medicine, Carbondale, IL, USA.
| |
Collapse
|
31
|
Amini Y, Saif N, Greer C, Hristov H, Isaacson R. The Role of Nutrition in Individualized Alzheimer's Risk Reduction. Curr Nutr Rep 2021; 9:55-63. [PMID: 32277428 DOI: 10.1007/s13668-020-00311-7] [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] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Decades of research suggests nutritional interventions can be an effective tool for reducing risk of Alzheimer's disease (AD), especially as part of an individualized clinical management plan. This review aims to emphasize new findings examining how specific dietary changes may delay or possibly prevent AD onset, and highlight how interventions can be adopted in clinical practice based on emerging principles of precision medicine. RECENT FINDINGS Specific dietary patterns and varied nutrient combinations can have a protective effect on brain health, promote cognitive function, and mediate the comorbidity of chronic conditions associated with increased AD risk. Individuals at risk for AD may see a greater impact of evidence-based dietary changes when initiated earlier in the AD spectrum. Depending on individual clinical profiles, incorporation of nutrition strategies is an essential component of an AD risk reduction plan in clinical practice.
Collapse
Affiliation(s)
- Yasmin Amini
- Department of Neurology, Weill Cornell Medicine & New York-Presbyterian, 428 East 72th St, Suite 500, New York, NY, 10021, USA
- Department of Sociomedical Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Nabeel Saif
- Department of Neurology, Weill Cornell Medicine & New York-Presbyterian, 428 East 72th St, Suite 500, New York, NY, 10021, USA
| | - Christine Greer
- Department of Neurology, Weill Cornell Medicine & New York-Presbyterian, 428 East 72th St, Suite 500, New York, NY, 10021, USA
| | - Hollie Hristov
- Department of Neurology, Weill Cornell Medicine & New York-Presbyterian, 428 East 72th St, Suite 500, New York, NY, 10021, USA
| | - Richard Isaacson
- Department of Neurology, Weill Cornell Medicine & New York-Presbyterian, 428 East 72th St, Suite 500, New York, NY, 10021, USA.
| |
Collapse
|
32
|
Abstract
The intranasal (IN) route enables the delivery of insulin to the central nervous system in the relative absence of systemic uptake and related peripheral side effects. Intranasally administered insulin is assumed to travel along olfactory and adjacent pathways and has been shown to rapidly accumulate in cerebrospinal fluid, indicating efficient transport to the brain. Two decades of studies in healthy humans and patients have demonstrated that IN insulin exerts functional effects on metabolism, such as reductions in food intake and body weight and improvements of glucose homeostasis, as well as cognition, ie, enhancements of memory performance both in healthy individuals and patients with mild cognitive impairment or Alzheimer's disease; these studies moreover indicate a favourable safety profile of the acute and repeated use of IN insulin. Emerging findings suggest that IN insulin also modulates neuroendocrine activity, sleep-related mechanisms, sensory perception and mood. Some, but not all studies point to sex differences in the response to IN insulin that need to be further investigated along with the impact of age. "Brain insulin resistance" is an evolving concept that posits impairments in central nervous insulin signalling as a pathophysiological factor in metabolic and cognitive disorders such as obesity, type 2 diabetes and Alzheimer's disease, and, notably, a target of interventions that rely on IN insulin. Still, the negative outcomes of longer-term IN insulin trials in individuals with obesity or Alzheimer's disease highlight the need for conceptual as well as methodological advances to translate the promising results of proof-of-concept experiments and pilot clinical trials into the successful clinical application of IN insulin.
Collapse
Affiliation(s)
- Manfred Hallschmid
- Institute of Medical Psychology and Behavioural Neurobiology, University of Tübingen, Tübingen, Germany
- German Centre for Diabetes Research (DZD), Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
| |
Collapse
|
33
|
Brain Insulin Resistance: Focus on Insulin Receptor-Mitochondria Interactions. Life (Basel) 2021; 11:life11030262. [PMID: 33810179 PMCID: PMC8005009 DOI: 10.3390/life11030262] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023] Open
Abstract
Current hypotheses implicate insulin resistance of the brain as a pathogenic factor in the development of Alzheimer’s disease and other dementias, Parkinson’s disease, type 2 diabetes, obesity, major depression, and traumatic brain injury. A variety of genetic, developmental, and metabolic abnormalities that lead to disturbances in the insulin receptor signal transduction may underlie insulin resistance. Insulin receptor substrate proteins are generally considered to be the node in the insulin signaling system that is critically involved in the development of insulin insensitivity during metabolic stress, hyperinsulinemia, and inflammation. Emerging evidence suggests that lower activation of the insulin receptor (IR) is another common, while less discussed, mechanism of insulin resistance in the brain. This review aims to discuss causes behind the diminished activation of IR in neurons, with a focus on the functional relationship between mitochondria and IR during early insulin signaling and the related roles of oxidative stress, mitochondrial hypometabolism, and glutamate excitotoxicity in the development of IR insensitivity to insulin.
Collapse
|
34
|
Frazier HN, Ghoweri AO, Sudkamp E, Johnson ES, Anderson KL, Fox G, Vatthanaphone K, Xia M, Lin RL, Hargis-Staggs KE, Porter NM, Pauly JR, Blalock EM, Thibault O. Long-Term Intranasal Insulin Aspart: A Profile of Gene Expression, Memory, and Insulin Receptors in Aged F344 Rats. J Gerontol A Biol Sci Med Sci 2021; 75:1021-1030. [PMID: 31180116 DOI: 10.1093/gerona/glz105] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Indexed: 02/06/2023] Open
Abstract
Intranasal insulin is a safe and effective method for ameliorating memory deficits associated with pathological brain aging. However, the impact of different formulations and the duration of treatment on insulin's efficacy and the cellular processes targeted by the treatment remain unclear. Here, we tested whether intranasal insulin aspart, a short-acting insulin formulation, could alleviate memory decline associated with aging and whether long-term treatment affected regulation of insulin receptors and other potential targets. Outcome variables included measures of spatial learning and memory, autoradiography and immunohistochemistry of the insulin receptor, and hippocampal microarray analyses. Aged Fischer 344 rats receiving long-term (3 months) intranasal insulin did not show significant memory enhancement on the Morris water maze task. Autoradiography results showed that long-term treatment reduced insulin binding in the thalamus but not the hippocampus. Results from hippocampal immunofluorescence revealed age-related decreases in insulin immunoreactivity that were partially offset by intranasal administration. Microarray analyses highlighted numerous insulin-sensitive genes, suggesting insulin aspart was able to enter the brain and alter hippocampal RNA expression patterns including those associated with tumor suppression. Our work provides insights into potential mechanisms of intranasal insulin and insulin resistance, and highlights the importance of treatment duration and the brain regions targeted.
Collapse
Affiliation(s)
| | | | - Emily Sudkamp
- The College of Pharmacy, University of Kentucky, Lexington, Kentucky
| | | | | | - Grant Fox
- Department of Pharmacology and Nutritional Sciences
| | | | - Mengfan Xia
- Department of Pharmacology and Nutritional Sciences
| | | | | | | | - James R Pauly
- The College of Pharmacy, University of Kentucky, Lexington, Kentucky
| | | | | |
Collapse
|
35
|
Abstract
Brain insulin signaling contributes to memory function and might be a viable target in the prevention and treatment of memory impairments including Alzheimer's disease. This short narrative review explores the potential of central nervous system (CNS) insulin administration via the intranasal pathway to improve memory performance in health and disease, with a focus on the most recent results. Proof-of-concept studies and (pilot) clinical trials in individuals with mild cognitive impairment or Alzheimer's disease indicate that acute and prolonged intranasal insulin administration enhances memory performance, and suggest that brain insulin resistance is a pathophysiological factor in Alzheimer's disease with or without concomitant metabolic dysfunction. Intranasally administered insulin is assumed to trigger improvements in synaptic plasticity and regional glucose uptake as well as alleviations of Alzheimer's disease neuropathology; additional contributions of changes in hypothalamus-pituitary-adrenocortical axis activity and sleep-related mechanisms are discussed. While intranasal insulin delivery has been conclusively demonstrated to be effective and safe, the recent outcomes of large-scale clinical studies underline the need for further investigations, which might also yield new insights into sex differences in the response to intranasal insulin and contribute to the optimization of delivery devices to grasp the full potential of intranasal insulin for Alzheimer's disease.
Collapse
Affiliation(s)
- Manfred Hallschmid
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Otfried-Müller-Str. 25, 72076, Tübingen, Germany.
- German Center for Diabetes Research (DZD), Tübingen, Germany.
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.
| |
Collapse
|
36
|
Frazier HN, Anderson KL, Ghoweri AO, Lin RL, Hawkinson TR, Popa GJ, Sompol P, Mendenhall MD, Norris CM, Thibault O. Molecular elevation of insulin receptor signaling improves memory recall in aged Fischer 344 rats. Aging Cell 2020; 19:e13220. [PMID: 32852134 PMCID: PMC7576226 DOI: 10.1111/acel.13220] [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: 05/01/2020] [Revised: 07/16/2020] [Accepted: 07/22/2020] [Indexed: 12/24/2022] Open
Abstract
As demonstrated by increased hippocampal insulin receptor density following learning in animal models and decreased insulin signaling, receptor density, and memory decline in aging and Alzheimer's diseases, numerous studies have emphasized the importance of insulin in learning and memory processes. This has been further supported by work showing that intranasal delivery of insulin can enhance insulin receptor signaling, alter cerebral blood flow, and improve memory recall. Additionally, inhibition of insulin receptor function or expression using molecular techniques has been associated with reduced learning. Here, we sought a different approach to increase insulin receptor activity without the need for administering the ligand. A constitutively active, modified human insulin receptor (IRβ) was delivered to the hippocampus of young (2 months) and aged (18 months) male Fischer 344 rats in vivo. The impact of increasing hippocampal insulin receptor expression was investigated using several outcome measures, including Morris water maze and ambulatory gait performance, immunofluorescence, immunohistochemistry, and Western immunoblotting. In aged animals, the IRβ construct was associated with enhanced performance on the Morris water maze task, suggesting that this receptor was able to improve memory recall. Additionally, in both age-groups, a reduced stride length was noted in IRβ-treated animals along with elevated hippocampal insulin receptor levels. These results provide new insights into the potential impact of increasing neuronal insulin signaling in the hippocampus of aged animals and support the efficacy of molecularly elevating insulin receptor activity in vivo in the absence of the ligand to directly study this process.
Collapse
Affiliation(s)
| | - Katie L. Anderson
- Department of Pharmacology and Nutritional SciencesLexingtonKentuckyUSA
| | - Adam O. Ghoweri
- Department of Pharmacology and Nutritional SciencesLexingtonKentuckyUSA
| | - Ruei-Lung Lin
- Department of Pharmacology and Nutritional SciencesLexingtonKentuckyUSA
| | - Tara R. Hawkinson
- Department of Pharmacology and Nutritional SciencesLexingtonKentuckyUSA
| | - Gabriel J. Popa
- Department of Molecular and Cellular BiochemistryLexingtonKentuckyUSA
| | - Pradoldej Sompol
- Sanders-Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
| | | | | | - Olivier Thibault
- Department of Pharmacology and Nutritional SciencesLexingtonKentuckyUSA
| |
Collapse
|
37
|
Argentati C, Tortorella I, Bazzucchi M, Emiliani C, Morena F, Martino S. The Other Side of Alzheimer's Disease: Influence of Metabolic Disorder Features for Novel Diagnostic Biomarkers. J Pers Med 2020; 10:E115. [PMID: 32899957 PMCID: PMC7563360 DOI: 10.3390/jpm10030115] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 02/08/2023] Open
Abstract
Nowadays, the amyloid cascade hypothesis is the dominant model to explain Alzheimer's disease (AD) pathogenesis. By this hypothesis, the inherited genetic form of AD is discriminated from the sporadic form of AD (SAD) that accounts for 85-90% of total patients. The cause of SAD is still unclear, but several studies have shed light on the involvement of environmental factors and multiple susceptibility genes, such as Apolipoprotein E and other genetic risk factors, which are key mediators in different metabolic pathways (e.g., glucose metabolism, lipid metabolism, energetic metabolism, and inflammation). Furthermore, growing clinical evidence in AD patients highlighted the presence of affected systemic organs and blood similarly to the brain. Collectively, these findings revise the canonical understating of AD pathogenesis and suggest that AD has metabolic disorder features. This review will focus on AD as a metabolic disorder and highlight the contribution of this novel understanding on the identification of new biomarkers for improving an early AD diagnosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Sabata Martino
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, 06123 Perugia, Italy; (C.A.); (I.T.); (M.B.); (C.E.); (F.M.)
| |
Collapse
|
38
|
Wetherell JL, Ripperger HS, Voegtle M, Ances BM, Balota D, Bower ES, Depp C, Eyler L, Foster ER, Head D, Hershey T, Hickman S, Kamantigue N, Klein S, Miller JP, Yingling MD, Nichols J, Nicol GE, Patterson BW, Rodebaugh TL, Shimony JS, Snyder A, Stephens M, Tate S, Uhrich ML, Wing D, Wu GF, Lenze EJ. Mindfulness, Education, and Exercise for age-related cognitive decline: Study protocol, pilot study results, and description of the baseline sample. Clin Trials 2020; 17:581-594. [PMID: 32594789 DOI: 10.1177/1740774520931864] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND/AIMS Age-related cognitive decline is a pervasive problem in our aging population. To date, no pharmacological treatments to halt or reverse cognitive decline are available. Behavioral interventions, such as physical exercise and Mindfulness-Based Stress Reduction, may reduce or reverse cognitive decline, but rigorously designed randomized controlled trials are needed to test the efficacy of such interventions. METHODS Here, we describe the design of the Mindfulness, Education, and Exercise study, an 18-month randomized controlled trial that will assess the effect of two interventions-mindfulness training plus moderate-to-vigorous intensity exercise or moderate-to-vigorous intensity exercise alone-compared with a health education control group on cognitive function in older adults. An extensive battery of biobehavioral assessments will be used to understand the mechanisms of cognitive remediation, by using structural and resting state functional magnetic resonance imaging, insulin sensitivity, inflammation, and metabolic and behavioral assessments. RESULTS We provide the results from a preliminary study (n = 29) of non-randomized pilot participants who received both the exercise and Mindfulness-Based Stress Reduction interventions. We also provide details on the recruitment and baseline characteristics of the randomized controlled trial sample (n = 585). CONCLUSION When complete, the Mindfulness, Education, and Exercise study will inform the research community on the efficacy of these widely available interventions improve cognitive functioning in older adults.
Collapse
Affiliation(s)
- Julie Loebach Wetherell
- VA San Diego Healthcare System, San Diego, CA, USA.,University of California San Diego, San Diego, CA, USA
| | - Hayley S Ripperger
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Michelle Voegtle
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Beau M Ances
- Department of Neurology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - David Balota
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Emily S Bower
- University of California San Diego, San Diego, CA, USA
| | - Colin Depp
- VA San Diego Healthcare System, San Diego, CA, USA.,University of California San Diego, San Diego, CA, USA
| | - Lisa Eyler
- University of California San Diego, San Diego, CA, USA
| | - Erin R Foster
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA.,Department of Neurology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA.,Program in Occupational Therapy, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Denise Head
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA.,Mallinckrodt Institute of Radiology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Tamara Hershey
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA.,Mallinckrodt Institute of Radiology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | | | | | - Samuel Klein
- Center for Human Nutrition, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - J Philip Miller
- Division of Biostatistics, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Michael D Yingling
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Ginger E Nicol
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Bruce W Patterson
- Center for Human Nutrition, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Thomas L Rodebaugh
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Joshua S Shimony
- Mallinckrodt Institute of Radiology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Abraham Snyder
- Mallinckrodt Institute of Radiology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Mary Stephens
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Susan Tate
- University of California San Diego, San Diego, CA, USA
| | - Mary L Uhrich
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA.,Program in Physical Therapy, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - David Wing
- University of California San Diego, San Diego, CA, USA
| | - Gregory F Wu
- Department of Neurology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA.,Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Eric J Lenze
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | | |
Collapse
|
39
|
McCulloch KA, Zhou K, Jin Y. Neuronal transcriptome analyses reveal novel neuropeptide modulators of excitation and inhibition imbalance in C. elegans. PLoS One 2020; 15:e0233991. [PMID: 32497060 PMCID: PMC7272019 DOI: 10.1371/journal.pone.0233991] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 05/16/2020] [Indexed: 01/06/2023] Open
Abstract
Neuropeptides are secreted molecules that have conserved roles modulating many processes, including mood, reproduction, and feeding. Dysregulation of neuropeptide signaling is also implicated in neurological disorders such as epilepsy. However, much is unknown about the mechanisms regulating specific neuropeptides to mediate behavior. Here, we report that the expression levels of dozens of neuropeptides are up-regulated in response to circuit activity imbalance in C. elegans. acr-2 encodes a homolog of human nicotinic receptors, and functions in the cholinergic motoneurons. A hyperactive mutation, acr-2(gf), causes an activity imbalance in the motor circuit. We performed cell-type specific transcriptomic analysis and identified genes differentially expressed in acr-2(gf), compared to wild type. The most over-represented class of genes are neuropeptides, with insulin-like-peptides (ILPs) the most affected. Moreover, up-regulation of neuropeptides occurs in motoneurons, as well as sensory neurons. In particular, the induced expression of the ILP ins-29 occurs in the BAG neurons, which were previously shown to function in gas-sensing. We also show that this up-regulation of ins-29 in acr-2(gf) animals is activity-dependent. Our genetic and molecular analyses support cooperative effects for ILPs and other neuropeptides in promoting motor circuit activity in the acr-2(gf) background. Together, this data reveals that a major transcriptional response to motor circuit dysregulation is in up-regulation of multiple neuropeptides, and suggests that BAG sensory neurons can respond to intrinsic activity states to feedback on the motor circuit.
Collapse
Affiliation(s)
- Katherine A. McCulloch
- Division of Biological Sciences, Section of Neurobiology, University of California San Diego, La Jolla, California, United States of America
| | - Kingston Zhou
- Division of Biological Sciences, Section of Neurobiology, University of California San Diego, La Jolla, California, United States of America
| | - Yishi Jin
- Division of Biological Sciences, Section of Neurobiology, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
40
|
Madhusudhanan J, Suresh G, Devanathan V. Neurodegeneration in type 2 diabetes: Alzheimer's as a case study. Brain Behav 2020; 10:e01577. [PMID: 32170854 PMCID: PMC7218246 DOI: 10.1002/brb3.1577] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 01/04/2020] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Rigorous research in the last few years has shown that in addition to the classical mechanism of neurodegeneration, certain unconventional mechanisms may also lead to neurodegenerative disease. One of them is a widely studied metabolic disorder: type 2 diabetes mellitus (T2DM). We now have a clear understanding of glucose-mediated neurodegeneration, mostly from studies in Alzheimer's disease (AD) models. AD is recognized to be significantly associated with hyperglycemia, even earning the term "type 3 diabetes." Here, we review first the pathophysiology of AD, both from the perspective of classical protein accumulation, as well as the newer T2DM-dependent mechanisms supported by findings from patients with T2DM. Secondly, we review the different pathways through which neurodegeneration is aggravated in hyperglycemic conditions taking AD as a case study. Finally, some of the current advances in AD management as a result of recent research developments in metabolic disorders-driven neurodegeneration are also discussed. METHODS Relevant literatures found from PubMed search were reviewed. RESULTS Apart from the known causes of AD, type 2 diabetes opens a new window to the AD pathology in several ways. It is a bidirectional interaction, of which, the molecular and signaling mechanisms are recently studied. This is our attempt to connect all of them to draw a complete mechanistic explanation for the neurodegeneration in T2DM. Refer to Figure 3. CONCLUSION The perspective of AD as a classical neurodegenerative disease is changing, and it is now being looked at from a zoomed-out perspective. The correlation between T2DM and AD is something observed and studied extensively. It is promising to know that there are certain advances in AD management following these studies.
Collapse
Affiliation(s)
- Jalaja Madhusudhanan
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| | - Gowthaman Suresh
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| | - Vasudharani Devanathan
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| |
Collapse
|
41
|
Rhea EM, Raber J, Banks WA. ApoE and cerebral insulin: Trafficking, receptors, and resistance. Neurobiol Dis 2020; 137:104755. [PMID: 31978603 PMCID: PMC7050417 DOI: 10.1016/j.nbd.2020.104755] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/13/2020] [Accepted: 01/20/2020] [Indexed: 12/16/2022] Open
Abstract
Central nervous system (CNS) insulin resistance is associated with Alzheimer's disease (AD). In addition, the apolipoprotein E4 (apoE4) isoform is a risk factor for AD. The connection between these two factors in relation to AD is being actively explored. We summarize this literature with a focus on the transport of insulin and apoE across the blood-brain barrier (BBB) and into the CNS, the impact of apoE and insulin on the BBB, and the interactions between apoE, insulin, and the insulin receptor once present in the CNS. We highlight how CNS insulin resistance is apparent in AD and potential ways to overcome this resistance by repurposing currently approved drugs, with apoE genotype taken into consideration as the treatment response following most interventions is apoE isoform-dependent. This review is part of a special issue focusing on apoE in AD and neurodegeneration.
Collapse
Affiliation(s)
- Elizabeth M Rhea
- Research and Development, Veterans Affairs Puget Sound Healthcare System, Seattle, WA 98108, United States of America; Department of Medicine, University of Washington, Seattle, WA 98195, United States of America.
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States of America; Departments of Neurology and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR 97239, United States of America
| | - William A Banks
- Research and Development, Veterans Affairs Puget Sound Healthcare System, Seattle, WA 98108, United States of America; Department of Medicine, University of Washington, Seattle, WA 98195, United States of America
| |
Collapse
|
42
|
Yeh SHH, Shie FS, Liu HK, Yao HH, Kao PC, Lee YH, Chen LM, Hsu SM, Chao LJ, Wu KW, Shiao YJ, Tsay HJ. A high-sucrose diet aggravates Alzheimer's disease pathology, attenuates hypothalamic leptin signaling, and impairs food-anticipatory activity in APPswe/PS1dE9 mice. Neurobiol Aging 2019; 90:60-74. [PMID: 31879131 DOI: 10.1016/j.neurobiolaging.2019.11.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 11/19/2019] [Accepted: 11/22/2019] [Indexed: 12/14/2022]
Abstract
High-fat and high-sugar diets contribute to the prevalence of type 2 diabetes and Alzheimer's disease (AD). Although the impact of high-fat diets on AD pathogenesis has been established, the effect of high-sucrose diets (HSDs) on AD pathogenesis remains unclear. This study sought to determine the impact of HSDs on AD-related pathologies. Male APPswe/PS1dE9 (APP/PS1) transgenic and wild-type mice were provided with HSD and their cognitive and hypothalamus-related noncognitive parameters, including feeding behaviors and glycemic regulation, were compared. HSD-fed APP/PS1 mice showed increased neuroinflammation, as well as increased cortical and serum levels of amyloid-β. HSD-fed APP/PS1 mice showed aggravated obesity, hyperinsulinemia, insulin resistance, and leptin resistance, but there was no induction of hyperphagia or hyperleptinemia. Leptin-induced phosphorylation of signal transducer and activator of transcription 3 in the dorsomedial and ventromedial hypothalamus was reduced in HSD-fed APP/PS1 mice, which might be associated with attenuated food-anticipatory activity, glycemic dysregulation, and AD-related noncognitive symptoms. Our study demonstrates that HSD aggravates metabolic stresses, increases AD-related pathologies, and attenuates hypothalamic leptin signaling in APP/PS1 mice.
Collapse
Affiliation(s)
| | - Feng-Shiun Shie
- Center for Neuropsychiatric Research, National Health Research Institutes, Taiwan, Miaoli, Taiwan, R.O.C
| | - Hui-Kang Liu
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan; Ph.D. Program in Clinical Drug Development of Chinese Herbal Medicine, Taipei Medical University, Taipei, Taiwan, R.O.C
| | - Heng-Hsiang Yao
- Institute of Neuroscience, School of Life Science, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | - Pei-Chen Kao
- Center for Neuropsychiatric Research, National Health Research Institutes, Taiwan, Miaoli, Taiwan, R.O.C
| | - Yi-Heng Lee
- Institute of Neuroscience, School of Life Science, National Yang-Ming University, Taipei, Taiwan, R.O.C.; Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei, Taiwan, R.O.C
| | - Li-Min Chen
- Institute of Neuroscience, School of Life Science, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | - Shu-Meng Hsu
- Center for Neuropsychiatric Research, National Health Research Institutes, Taiwan, Miaoli, Taiwan, R.O.C.; Institute of Neuroscience, School of Life Science, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | - Li-Jung Chao
- Brain Research Center, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | - Kuan-Wei Wu
- Institute of Biopharmaceutical Science, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | - Young-Ji Shiao
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan; Ph.D. Program in Clinical Drug Development of Chinese Herbal Medicine, Taipei Medical University, Taipei, Taiwan, R.O.C.; Institute of Biopharmaceutical Science, National Yang-Ming University, Taipei, Taiwan, R.O.C..
| | - Huey-Jen Tsay
- Institute of Neuroscience, School of Life Science, National Yang-Ming University, Taipei, Taiwan, R.O.C..
| |
Collapse
|
43
|
Fine JM, Stroebel BM, Faltesek KA, Terai K, Haase L, Knutzen KE, Kosyakovsky J, Bowe TJ, Fuller AK, Frey WH, Hanson LR. Intranasal delivery of low-dose insulin ameliorates motor dysfunction and dopaminergic cell death in a 6-OHDA rat model of Parkinson's Disease. Neurosci Lett 2019; 714:134567. [PMID: 31629033 DOI: 10.1016/j.neulet.2019.134567] [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: 09/04/2019] [Revised: 10/11/2019] [Accepted: 10/15/2019] [Indexed: 12/22/2022]
Abstract
Emerging evidence continues to demonstrate that disrupted insulin signaling and altered energy metabolism may play a key role underpinning pathology in neurodegenerative conditions. Intranasally administered insulin has already shown promise as a memory-enhancing therapy in patients with Alzheimer's and animal models of the disease. Intranasal drug delivery allows for direct targeting of insulin to the brain, bypassing the blood brain barrier and minimizing systemic adverse effects. In this study, we sought to expand upon previous results that show intranasal insulin may also have promise as a Parkinson's therapy. We treated 6-OHDA parkinsonian rats with a low dose (3 IU/day) of insulin and assessed apomorphine induced rotational turns, motor deficits via a horizontal ladder test, and dopaminergic cell survival via stereological counting. We found that insulin therapy substantially reduced motor dysfunction and dopaminergic cell death induced by unilateral injection of 6-OHDA. These results confirm insulin's efficacy within this model, and do so over a longer period after model induction which more closely resembles Parkinson's disease. This study also employed a lower dose than previous studies and utilizes a delivery device, which could lead to an easier transition into human clinical trials as a therapeutic for Parkinson's disease.
Collapse
Affiliation(s)
- Jared M Fine
- HealthPartners Neuroscience Center, HealthPartners Institute, 295 Phalen Blvd., Saint Paul, MN, 55130, United States.
| | - Benjamin M Stroebel
- HealthPartners Neuroscience Center, HealthPartners Institute, 295 Phalen Blvd., Saint Paul, MN, 55130, United States
| | - Katherine A Faltesek
- HealthPartners Neuroscience Center, HealthPartners Institute, 295 Phalen Blvd., Saint Paul, MN, 55130, United States
| | - Kaoru Terai
- HealthPartners Neuroscience Center, HealthPartners Institute, 295 Phalen Blvd., Saint Paul, MN, 55130, United States
| | - Lucas Haase
- HealthPartners Neuroscience Center, HealthPartners Institute, 295 Phalen Blvd., Saint Paul, MN, 55130, United States
| | - Kristin E Knutzen
- HealthPartners Neuroscience Center, HealthPartners Institute, 295 Phalen Blvd., Saint Paul, MN, 55130, United States
| | - Jacob Kosyakovsky
- HealthPartners Neuroscience Center, HealthPartners Institute, 295 Phalen Blvd., Saint Paul, MN, 55130, United States
| | - Tate J Bowe
- HealthPartners Neuroscience Center, HealthPartners Institute, 295 Phalen Blvd., Saint Paul, MN, 55130, United States
| | - Austin K Fuller
- HealthPartners Neuroscience Center, HealthPartners Institute, 295 Phalen Blvd., Saint Paul, MN, 55130, United States
| | - William H Frey
- HealthPartners Neuroscience Center, HealthPartners Institute, 295 Phalen Blvd., Saint Paul, MN, 55130, United States
| | - Leah R Hanson
- HealthPartners Neuroscience Center, HealthPartners Institute, 295 Phalen Blvd., Saint Paul, MN, 55130, United States
| |
Collapse
|