151
|
Felisatti F, Gonneaud J, Palix C, Garnier-Crussard A, Mézenge F, Landeau B, Chocat A, Quillard A, Ferrand-Devouge E, de La Sayette V, Vivien D, Chételat G, Poisnel G. Role of Cardiovascular Risk Factors on the Association Between Physical Activity and Brain Integrity Markers in Older Adults. Neurology 2022; 98:e2023-e2035. [PMID: 35418459 PMCID: PMC9162049 DOI: 10.1212/wnl.0000000000200270] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 02/08/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Physical activity has been associated with a decreased risk for dementia, but the mechanisms underlying this association remain to be determined. Our objective was to assess whether cardiovascular risk factors mediate the association between physical activity and brain integrity markers in older adults. METHODS At baseline, participants from the Age-Well study completed a physical activity questionnaire and underwent cardiovascular risk factors collection (systolic blood pressure, body mass index [BMI], current smoker status, and high-density lipoprotein cholesterol, total cholesterol, and insulin levels) and multimodal neuroimaging (structural MRI, diffusion MRI, FDG-PET, and florbetapir PET). Multiple regressions were conducted to assess the association among physical activity, cardiovascular risk factors, and neuroimaging. Mediation analyses were performed to test whether cardiovascular risk factors mediated the associations between physical activity and neuroimaging. RESULTS A total of 134 cognitively unimpaired older adults (≥65 years) were included. Higher physical activity was associated with higher gray matter (GM) volume (β = 0.174, p = 0.030) and cerebral glucose metabolism (β = 0.247, p = 0.019) but not with amyloid deposition or white matter integrity. Higher physical activity was associated with lower insulin level and BMI but not with the other cardiovascular risk factors. Lower insulin level and BMI were related to higher GM volume but not to cerebral glucose metabolism. When controlling for insulin level and BMI, the association between physical activity and cerebral glucose metabolism remained unchanged, while the association with GM volume was lost. When insulin level and BMI were entered in the same model, only BMI remained a significant predictor of GM volume. Mediation analyses confirmed that insulin level and BMI mediated the association between physical activity and GM volume. Analyses were replicated within Alzheimer disease-sensitive regions and results remained overall similar. DISCUSSION The association between physical activity and GM volume is mediated by changes in insulin level and BMI. In contrast, the association with cerebral glucose metabolism seems to be independent from cardiovascular risk factors. Older adults engaging in physical activity experience cardiovascular benefits through the maintenance of a lower BMI and insulin level, resulting in greater structural brain integrity. This study has implications for understanding how physical activity affects brain health and may help in developing strategies to prevent or delay age-related decline. TRIAL REGISTRATION INFORMATION EudraCT: 2016-002,441-36; IDRCB: 2016-A01767-44; ClinicalTrials.gov Identifier: NCT02977819.
Collapse
Affiliation(s)
- Francesca Felisatti
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Julie Gonneaud
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Cassandre Palix
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Antoine Garnier-Crussard
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Florence Mézenge
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Brigitte Landeau
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Anne Chocat
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Anne Quillard
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Eglantine Ferrand-Devouge
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Vincent de La Sayette
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Denis Vivien
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Gaël Chételat
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Géraldine Poisnel
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| |
Collapse
|
152
|
Moayedi K, Orandi S, Ebrahimi R, Tanhapour M, Moradi M, Abbastabar M, Golestani A. A novel approach to type 3 diabetes mechanism: The interplay between noncoding RNAs and insulin signaling pathway in Alzheimer's disease. J Cell Physiol 2022; 237:2838-2861. [PMID: 35580144 DOI: 10.1002/jcp.30779] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 04/05/2022] [Accepted: 04/27/2022] [Indexed: 12/06/2022]
Abstract
Today, growing evidence indicates that patients with type 2 diabetes (T2D) are at a higher risk of developing Alzheimer's disease (AD). Indeed, AD as one of the main causes of dementia in people aged more than 65 years can be aggravated by insulin resistance (IR) and other metabolic risk factors related to T2D which are also linked to the function of the brain. Remarkably, a new term called "type 3 diabetes" has been suggested for those people who are diagnosed with AD while also showing the symptoms of IR and T2D. In this regard, the role of genetic and epigenetic changes associated with AD has been confirmed by many studies. On the other hand, it should be noted that the insulin signaling pathway is highly regulated by various mechanisms, including epigenetic factors. Among these, the role of noncoding RNAs (ncRNAs), including microRNAs and long noncoding RNAs has been comprehensively studied with respect to the pathology of AD and the most well-known underlying mechanisms. Nevertheless, the number of studies exploring the association between ncRNAs and the downstream targets of the insulin signaling pathway in the development of AD has notably increased in recent years. With this in view, the present study aimed to review the interplay between different ncRNAs and the insulin signaling pathway targets in the pathogenesis of AD to find a new approach in the field of combining biomarkers or therapeutic targets for this disease.
Collapse
Affiliation(s)
- Kiana Moayedi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shirin Orandi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reyhane Ebrahimi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Tanhapour
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mona Moradi
- Pediatric Infectious Diseases Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Maryam Abbastabar
- Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Abolfazl Golestani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
153
|
Neurobehavioral and neurobiochemical effect of atomoxetine and N-acetylcysteine in streptozotocin diabetes induced endothelial dysfunction and related dementia. Physiol Behav 2022; 249:113767. [PMID: 35245527 DOI: 10.1016/j.physbeh.2022.113767] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 01/22/2022] [Accepted: 02/28/2022] [Indexed: 11/20/2022]
Abstract
Metabolic conditions like diabetes, is a major risk factor for the development of dementia of vascular origin. This study investigates the efficacy of atomoxetine (ATX) and N-acetylcysteine (NAC) in streptozotocin (STZ) diabetes induced vascular endothelium dysfunction and related dementia. Single dose STZ (50 mg/kg i.p) was administered to Albino Wistar rats (male, 200-250 g) by dissolving in citrate buffer. Morris water maze (MWM) and attentional set shifting tests (ASST) were used to assess the spatial learning, memory, reversal learning, and executive functioning in animals. Body weight, serum glucose, serum nitrite/nitrate, vascular endothelial function, aortic superoxide anion, brains' oxidative markers (thiobarbituric acid reactive species-TBARS, reduced glutathione-GSH, superoxide dismutase-SOD, and catalase-CAT), inflammatory markers (IL-6, IL-10, TNF-α, and myeloperoxidase-MPO), acetylcholinesterase activity-AChE and histopathological changes were also assessed. Atomoxetine - ATX (2 mg kg-1/ 4 mg kg-1) and N-acetylcysteine- NAC (250 mg kg-1/ 500 mg kg-1) were used alone as well as in combination, as the treatment drugs. Donepezil (0.5 mg kg-1) was used as a positive control. STZ administered rats showed increase in serum glucose levels and decrease in body weight. Rats administered with STZ also showed reduction in learning, memory, reversal learning, executive functioning, impairment in endothelial function, increase in brains' oxidative stress, inflammation, AChE activity and histopathological changes. Administration of ATX and NAC in two different doses as well as in combination, significantly attenuated the STZ induced diabetes induced impairments in the behavioral, endothelial, biochemical parameters and histopathological changes. Co-treatment of ATX and NAC was better in comparison to the doses when given alone. Hence, STZ administration caused diabetes induced dementia of vascular origin which was attenuated by the administration of ATX and NAC. Therefore, these agents may be studied further for the assessment of their full potential in diabetes induced dementia of vascular origin conditions.
Collapse
|
154
|
Kalia V, Niedzwiecki MM, Bradner JM, Lau FK, Anderson FL, Bucher ML, Manz KE, Schlotter AP, Fuentes ZC, Pennell KD, Picard M, Walker DI, Hu WT, Jones DP, Miller GW. Cross-species metabolomic analysis of tau- and DDT-related toxicity. PNAS NEXUS 2022; 1:pgac050. [PMID: 35707205 PMCID: PMC9186048 DOI: 10.1093/pnasnexus/pgac050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 04/28/2022] [Indexed: 01/29/2023]
Abstract
Exposure to the pesticide dichlorodiphenyltrichloroethane (DDT) has been associated with increased risk of Alzheimer's disease (AD), a disease also associated with hyperphosphorylated tau (p-tau) protein aggregation. We investigated whether exposure to DDT can exacerbate tau protein toxicity in Caenorhabditiselegans using a transgenic strain that expresses human tau protein prone to aggregation by measuring changes in size, swim behavior, respiration, lifespan, learning, and metabolism. In addition, we examined the association between cerebrospinal fluid (CSF) p-tau protein-as a marker of postmortem tau burden-and global metabolism in both a human population study and in C. elegans, using the same p-tau transgenic strain. From the human population study, plasma and CSF-derived metabolic features associated with p-tau levels were related to drug, amino acid, fatty acid, and mitochondrial metabolism pathways. A total of five metabolites overlapped between plasma and C. elegans, and four between CSF and C. elegans. DDT exacerbated the inhibitory effect of p-tau protein on growth and basal respiration. In the presence of p-tau protein, DDT induced more curling and was associated with reduced levels of amino acids but increased levels of uric acid and adenosylselenohomocysteine. Our findings in C. elegans indicate that DDT exposure and p-tau aggregation both inhibit mitochondrial function and DDT exposure can exacerbate the mitochondrial inhibitory effects of p-tau aggregation. Further, biological pathways associated with exposure to DDT and p-tau protein appear to be conserved between species.
Collapse
Affiliation(s)
- Vrinda Kalia
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032 USA
| | - Megan M Niedzwiecki
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029 USA
| | - Joshua M Bradner
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032 USA
| | - Fion K Lau
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032 USA
| | - Faith L Anderson
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032 USA
| | - Meghan L Bucher
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032 USA
| | - Katherine E Manz
- School of Engineering, Brown University, Providence, RI, 02912 USA
| | - Alexa Puri Schlotter
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032 USA
| | - Zoe Coates Fuentes
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029 USA
| | - Kurt D Pennell
- School of Engineering, Brown University, Providence, RI, 02912 USA
| | - Martin Picard
- Department of Neurology, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, 10032 USA
| | - Douglas I Walker
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029 USA
| | - William T Hu
- Department of Neurology, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, 08901 USA
| | - Dean P Jones
- Division of Pulmonary, Allergy and Critical Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, 30322 USA
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032 USA
| |
Collapse
|
155
|
Linking hIAPP misfolding and aggregation with type 2 diabetes mellitus: a structural perspective. Biosci Rep 2022; 42:231205. [PMID: 35475576 PMCID: PMC9118370 DOI: 10.1042/bsr20211297] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/12/2022] [Accepted: 04/26/2022] [Indexed: 11/17/2022] Open
Abstract
There are over 40 identified human disorders that involve certain proteins folding incorrectly, accumulating in the body causing damage to cells and organs and causing disease. Type 2 Diabetes Mellitus (T2DM) is one of these protein misfolding disorders (PMDs) and involves human islet amyloid polypeptide (hIAPP) misfolding and accumulating in parts of the body, primarily in the pancreas, causing damage to islet cells and affecting glucose regulation. In this review, we have summarised our current understanding of what causes hIAPP to misfold, what conformations are found in different parts of the body with a particular focus on what is known about the structure of hIAPP and how this links to T2DM. Understanding the molecular basis behind these misfolding events is essential for understanding the role of hIAPP to develop better therapeutics since type 2 diabetes currently affects over 4.9 million people in the United Kingdom alone and is predicted to increase as our population ages.
Collapse
|
156
|
Arjunan A, Sah DK, Jung YD, Song J. Hepatic Encephalopathy and Melatonin. Antioxidants (Basel) 2022; 11:837. [PMID: 35624703 PMCID: PMC9137547 DOI: 10.3390/antiox11050837] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/06/2022] [Accepted: 04/24/2022] [Indexed: 11/25/2022] Open
Abstract
Hepatic encephalopathy (HE) is a severe metabolic syndrome linked with acute/chronic hepatic disorders. HE is also a pernicious neuropsychiatric complication associated with cognitive decline, coma, and death. Limited therapies are available to treat HE, which is formidable to oversee in the clinic. Thus, determining a novel therapeutic approach is essential. The pathogenesis of HE has not been well established. According to various scientific reports, neuropathological symptoms arise due to excessive accumulation of ammonia, which is transported to the brain via the blood-brain barrier (BBB), triggering oxidative stress and inflammation, and disturbing neuronal-glial functions. The treatment of HE involves eliminating hyperammonemia by enhancing the ammonia scavenging mechanism in systemic blood circulation. Melatonin is the sole endogenous hormone linked with HE. Melatonin as a neurohormone is a potent antioxidant that is primarily synthesized and released by the brain's pineal gland. Several HE and liver cirrhosis clinical studies have demonstrated impaired synthesis, secretion of melatonin, and circadian patterns. Melatonin can cross the BBB and is involved in various neuroprotective actions on the HE brain. Hence, we aim to elucidate how HE impairs brain functions, and elucidate the precise molecular mechanism of melatonin that reverses the HE effects on the central nervous system.
Collapse
Affiliation(s)
- Archana Arjunan
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Korea;
| | - Dhiraj Kumar Sah
- Department of Biochemistry, Chonnam National University Medical School, Hwasun 58128, Korea;
| | - Young Do Jung
- Department of Biochemistry, Chonnam National University Medical School, Hwasun 58128, Korea;
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Korea;
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, 264 Seoyangro, Hwasun 58128, Korea
| |
Collapse
|
157
|
Michailidis M, Tata DA, Moraitou D, Kavvadas D, Karachrysafi S, Papamitsou T, Vareltzis P, Papaliagkas V. Antidiabetic Drugs in the Treatment of Alzheimer's Disease. Int J Mol Sci 2022; 23:4641. [PMID: 35563031 PMCID: PMC9102472 DOI: 10.3390/ijms23094641] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023] Open
Abstract
The public health burden of type 2 diabetes mellitus and Alzheimer's disease is steadily increasing worldwide, especially in the population of older adults. Epidemiological and clinical studies suggest a possible shared pathophysiology between the two diseases and an increased risk of AD in patients with type 2 diabetes mellitus. Therefore, in recent years, there has been a substantial interest in identifying the mechanisms of action of antidiabetic drugs and their potential use in Alzheimer's disease. Human studies in patients with mild cognitive impairment and Alzheimer's disease have shown that administration of some antidiabetic medications, such as intranasal insulin, metformin, incretins, and thiazolidinediones, can improve cognition and memory. This review aims to examine the latest evidence on antidiabetic medications as a potential candidate for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Michalis Michailidis
- Laboratory of Psychology, School of Psychology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Despina A. Tata
- Laboratory of Cognitive Neuroscience, School of Psychology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (D.A.T.); (D.M.)
| | - Despina Moraitou
- Laboratory of Cognitive Neuroscience, School of Psychology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (D.A.T.); (D.M.)
| | - Dimitrios Kavvadas
- Histology and Embryology Department, Faculty of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (D.K.); (S.K.); (T.P.)
| | - Sofia Karachrysafi
- Histology and Embryology Department, Faculty of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (D.K.); (S.K.); (T.P.)
| | - Theodora Papamitsou
- Histology and Embryology Department, Faculty of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (D.K.); (S.K.); (T.P.)
| | - Patroklos Vareltzis
- Department of Chemical Engineering, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Vasileios Papaliagkas
- Department of Biomedical Sciences, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece
| |
Collapse
|
158
|
Ju Z, Shen L, Zhou M, Luo J, Yu Z, Qu C, Lei R, Lei M, Huang R. Helicobacter pylori and Alzheimer's Disease-Related Metabolic Dysfunction: Activation of TLR4/Myd88 Inflammation Pathway from p53 Perspective and a Case Study of Low-Dose Radiation Intervention. ACS Chem Neurosci 2022; 13:1065-1081. [PMID: 35312296 DOI: 10.1021/acschemneuro.2c00082] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Gut dysbiosis is observed in Alzheimer's disease (AD) and is frequently associated with AD-induced metabolic dysfunction. However, the extent and specific underlying molecular mechanisms triggered by alterations of gut microbiota composition and function mediating AD-induced metabolic dysfunction in AD remain incompletely uncovered. Here, we indicate that Helicobacter pylori (H. pylori) is abundant in AD patients with relative metabolic dysfunction. Fecal microbiota transplantation from the AD patients promoted metabolic dysfunction in mice and increased gut permeability. H. pylori increased gut permeability through activation of the TLR4/Myd88 inflammation pathway in a p53-dependent manner, leading to metabolic dysfunction. Moreover, p53 deficiency reduced bile acid concentration, leading to an increased abundance of H. pylori colonization. Overall, these data identify H. pylori as a key promoter of AD-induced metabolic dysfunction.
Collapse
Affiliation(s)
- Zhao Ju
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China
| | - Liangfang Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Meiling Zhou
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China
| | - Jinhua Luo
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China
| | - Zijian Yu
- The First Affiliated Hospital, University of South China, 69 Chuanshan Road, Hengyang, Hunan 421001, People’s Republic of China
| | - Can Qu
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China
| | - Ridan Lei
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China
| | - Mingjun Lei
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Ruixue Huang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China
| |
Collapse
|
159
|
Zhang Y, Chen D, Zhang M, Bian J, Qian S, Kou X. Treadmill training attenuate STZ-induced cognitive dysfunction in type 2 diabetic rats via modulating Grb10/IGF-R signaling. Brain Res Bull 2022; 181:12-20. [DOI: 10.1016/j.brainresbull.2022.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 12/14/2021] [Accepted: 01/18/2022] [Indexed: 11/02/2022]
|
160
|
Brain Metabolic Alterations in Alzheimer's Disease. Int J Mol Sci 2022; 23:ijms23073785. [PMID: 35409145 PMCID: PMC8998942 DOI: 10.3390/ijms23073785] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 01/27/2023] Open
Abstract
The brain is one of the most energy-consuming organs in the body. Satisfying such energy demand requires compartmentalized, cell-specific metabolic processes, known to be complementary and intimately coupled. Thus, the brain relies on thoroughly orchestrated energy-obtaining agents, processes and molecular features, such as the neurovascular unit, the astrocyte-neuron metabolic coupling, and the cellular distribution of energy substrate transporters. Importantly, early features of the aging process are determined by the progressive perturbation of certain processes responsible for adequate brain energy supply, resulting in brain hypometabolism. These age-related brain energy alterations are further worsened during the prodromal stages of neurodegenerative diseases, namely Alzheimer's disease (AD), preceding the onset of clinical symptoms, and are anatomically and functionally associated with the loss of cognitive abilities. Here, we focus on concrete neuroenergetic features such as the brain's fueling by glucose and lactate, the transporters and vascular system guaranteeing its supply, and the metabolic interactions between astrocytes and neurons, and on its neurodegenerative-related disruption. We sought to review the principles underlying the metabolic dimension of healthy and AD brains, and suggest that the integration of these concepts in the preventive, diagnostic and treatment strategies for AD is key to improving the precision of these interventions.
Collapse
|
161
|
Dysmetabolism and Neurodegeneration: Trick or Treat? Nutrients 2022; 14:nu14071425. [PMID: 35406040 PMCID: PMC9003269 DOI: 10.3390/nu14071425] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence suggests the existence of a strong link between metabolic syndrome and neurodegeneration. Indeed, epidemiologic studies have described solid associations between metabolic syndrome and neurodegeneration, whereas animal models contributed for the clarification of the mechanistic underlying the complex relationships between these conditions, having the development of an insulin resistance state a pivotal role in this relationship. Herein, we review in a concise manner the association between metabolic syndrome and neurodegeneration. We start by providing concepts regarding the role of insulin and insulin signaling pathways as well as the pathophysiological mechanisms that are in the genesis of metabolic diseases. Then, we focus on the role of insulin in the brain, with special attention to its function in the regulation of brain glucose metabolism, feeding, and cognition. Moreover, we extensively report on the association between neurodegeneration and metabolic diseases, with a particular emphasis on the evidence observed in animal models of dysmetabolism induced by hypercaloric diets. We also debate on strategies to prevent and/or delay neurodegeneration through the normalization of whole-body glucose homeostasis, particularly via the modulation of the carotid bodies, organs known to be key in connecting the periphery with the brain.
Collapse
|
162
|
Wang Y, Yang Y, Liu Y, Guo A, Zhang Y. Cognitive impairments in type 1 diabetes mellitus model mice are associated with synaptic protein disorders. Neurosci Lett 2022; 777:136587. [PMID: 35337951 DOI: 10.1016/j.neulet.2022.136587] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 01/24/2022] [Accepted: 03/21/2022] [Indexed: 11/24/2022]
Abstract
An association between type 1 diabetes mellitus (T1DM) and cognitive impairment was recently reported. However, the mechanisms by which T1DM induces cognitive impairment are still unknown. Here, we confirmed that T1DM mice induced by streptozotocin (STZ) injection had impaired working memory and spatial memory. We observed long-term potentiation (LTP) induction defects and synaptic loss in mice 20 weeks after STZ injection. We also found decreased levels of synaptic proteins, including the N-methyl-D-aspartic acid receptor (NMDAR) subunit NR2A, synaptophysin (SYP), and postsynaptic density 95 (PSD95), in the hippocampus and prefrontal cortex, revealing similarities in the alteration patterns of these synaptic proteins in aged Alzheimer's disease (AD) APP/PS1 mice and T1DM mice. Taken together, these findings expand our understanding of the mechanisms underlying T1DM-induced cognitive impairment.
Collapse
Affiliation(s)
- Yiming Wang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Yueqi Yang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Yiqiong Liu
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Angyang Guo
- Duke Kunshan University, Kunshan, Jiangsu 215316, China
| | - Yan Zhang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China.
| |
Collapse
|
163
|
Top WMC, Kooy A, Stehouwer CDA. Metformin: A Narrative Review of Its Potential Benefits for Cardiovascular Disease, Cancer and Dementia. Pharmaceuticals (Basel) 2022; 15:312. [PMID: 35337110 PMCID: PMC8951049 DOI: 10.3390/ph15030312] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 02/01/2023] Open
Abstract
The biguanide metformin has been used as first-line therapy in type 2 diabetes mellitus (T2DM) treatment for several decades. In addition to its glucose-lowering properties and its prevention of weight gain, the landmark UK Prospective Diabetes Study (UKPDS) demonstrated cardioprotective properties in obese T2DM patients. Coupled with a favorable side effect profile and low cost, metformin has become the cornerstone in the treatment of T2DM worldwide. In addition, metformin is increasingly being investigated for its potential anticancer and neuroprotective properties both in T2DM patients and non-diabetic individuals. In the meantime, new drugs with powerful cardioprotective properties have been introduced and compete with metformin for its place in the treatment of T2DM. In this review we will discuss actual insights in the various working mechanisms of metformin and the evidence for its beneficial effects on (the prevention of) cardiovascular disease, cancer and dementia. In addition to observational evidence, emphasis is placed on randomized trials and recent meta-analyses to obtain an up-to-date overview of the use of metformin in clinical practice.
Collapse
Affiliation(s)
- Wiebe M. C. Top
- Department of Intensive Care, Treant Care Group, 7909 AA Hoogeveen, The Netherlands;
| | - Adriaan Kooy
- Department of Internal Medicine, Treant Care Group, 7909 AA Hoogeveen, The Netherlands
- Bethesda Diabetes Research Center, 7909 AA Hoogeveen, The Netherlands
- Department of Internal Medicine, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Coen D. A. Stehouwer
- Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands;
| |
Collapse
|
164
|
Mani V, Arfeen M, Mohammed HA, Elsisi HA, Sajid S, Almogbel Y, Aldubayan M, Dhanasekaran M, Alhowail A. Sukkari dates seed improves type-2 diabetes mellitus-induced memory impairment by reducing blood glucose levels and enhancing brain cholinergic transmission: In vivo and molecular modeling studies. Saudi Pharm J 2022; 30:750-763. [PMID: 35812141 PMCID: PMC9257867 DOI: 10.1016/j.jsps.2022.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/25/2022] [Indexed: 11/25/2022] Open
Abstract
Cognitive decline is one of the serious complications associated with diabetes mellitus (T2DM) of type-2. In this reported work, the effect of aqueous sukkari dates seed extract (ASSE) was evaluated in T2DM-induced rats. T2DM was induced using intraperitoneal injection of nicotinamide and streptozocin (STZ) administration. The diabetic rats were then treated orally with 200 mg/kg and 400 mg/kg of dates seed extract for 30 days and results were compared with metformin-treated groups. The memory functions were assessed using three maze models. Glucose and insulin levels in the blood and acetylcholine, acetylcholinesterase brain homogenates were estimated. The results showed a significant reduction in transfer latency (TL) (p < 0.001) during the elevated plus maze (EPM) test. The novel object recognition (NOR) test revealed a longer exploration time (p > 0.05) with novel objects and a higher discrimination index (p > 0.05). The Y-maze test also showed a significant increase in the number of entries to the novel arm (p > 0.05) and the total number of entries in the trial (p > 0.01) as well as in test (p > 0.05) sessions. Reduction in blood glucose (p > 0.05) and improvement in blood insulin (p > 0.05) levels were also noted. Improvement in ACh levels (p > 0.001) with 400 mg/kg of ASSE and reduction in AChE (p > 0.001) with both doses of ASSE were also observed in the brain homogenates. The results of ASSE were found comparable with the metformin-treated rats. The estimation of phytochemical constituents displayed a significant presence of phenolic content. Further, molecular modeling studies showed ellagic acid, catechin, and epicatechin as the potential molecule interacting with GSK-3β, α-amylase, and AChE and may be responsible for observed bioactivity. In conclusion, ASSE has the ability to alleviate T2DM-related cognitive impairments.
Collapse
|
165
|
Michailidis M, Moraitou D, Tata DA, Kalinderi K, Papamitsou T, Papaliagkas V. Alzheimer's Disease as Type 3 Diabetes: Common Pathophysiological Mechanisms between Alzheimer's Disease and Type 2 Diabetes. Int J Mol Sci 2022; 23:2687. [PMID: 35269827 PMCID: PMC8910482 DOI: 10.3390/ijms23052687] [Citation(s) in RCA: 128] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/25/2022] [Accepted: 02/26/2022] [Indexed: 12/27/2022] Open
Abstract
Globally, the incidence of type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) epidemics is increasing rapidly and has huge financial and emotional costs. The purpose of the current review article is to discuss the shared pathophysiological connections between AD and T2DM. Research findings are presented to underline the vital role that insulin plays in the brain's neurotransmitters, homeostasis of energy, as well as memory capacity. The findings of this review indicate the existence of a mechanistic interplay between AD pathogenesis with T2DM and, especially, disrupted insulin signaling. AD and T2DM are interlinked with insulin resistance, neuroinflammation, oxidative stress, advanced glycosylation end products (AGEs), mitochondrial dysfunction and metabolic syndrome. Beta-amyloid, tau protein and amylin can accumulate in T2DM and AD brains. Given that the T2DM patients are not routinely evaluated in terms of their cognitive status, they are rarely treated for cognitive impairment. Similarly, AD patients are not routinely evaluated for high levels of insulin or for T2DM. Studies suggesting AD as a metabolic disease caused by insulin resistance in the brain also offer strong support for the hypothesis that AD is a type 3 diabetes.
Collapse
Affiliation(s)
- Michalis Michailidis
- Laboratory of Psychology, School of Psychology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (M.M.); (D.M.); (D.A.T.)
| | - Despina Moraitou
- Laboratory of Psychology, School of Psychology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (M.M.); (D.M.); (D.A.T.)
| | - Despina A. Tata
- Laboratory of Psychology, School of Psychology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (M.M.); (D.M.); (D.A.T.)
| | - Kallirhoe Kalinderi
- Laboratory of Medical Biology-Genetics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Theodora Papamitsou
- Histology and Embryology Department, Faculty of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Vasileios Papaliagkas
- Department of Biomedical Sciences, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece
| |
Collapse
|
166
|
Falsetti L, Viticchi G, Zaccone V, Guerrieri E, Moroncini G, Luzzi S, Silvestrini M. Shared Molecular Mechanisms among Alzheimer’s Disease, Neurovascular Unit Dysfunction and Vascular Risk Factors: A Narrative Review. Biomedicines 2022; 10:biomedicines10020439. [PMID: 35203654 PMCID: PMC8962428 DOI: 10.3390/biomedicines10020439] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/27/2022] [Accepted: 02/12/2022] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common type of dementia, affecting 24 million individuals. Clinical and epidemiological studies have found several links between vascular risk factors (VRF), neurovascular unit dysfunction (NVUd), blood-brain barrier breakdown (BBBb) and AD onset and progression in adulthood, suggesting a pathogenetic continuum between AD and vascular dementia. Shared pathways between AD, VRF, and NVUd/BBB have also been found at the molecular level, underlining the strength of this association. The present paper reviewed the literature describing commonly shared molecular pathways between adult-onset AD, VRF, and NVUd/BBBb. Current evidence suggests that VRF and NVUd/BBBb are involved in AD neurovascular and neurodegenerative pathology and share several molecular pathways. This is strongly supportive of the hypothesis that the presence of VRF can at least facilitate AD onset and progression through several mechanisms, including NVUd/BBBb. Moreover, vascular disease and several comorbidities may have a cumulative effect on VRF and worsen the clinical manifestations of AD. Early detection and correction of VRF and vascular disease by improving NVUd/BBBd could be a potential target to reduce the overall incidence and delay cognitive impairment in AD.
Collapse
Affiliation(s)
- Lorenzo Falsetti
- Internal and Subintensive Medicine Department, Azienda Ospedaliero-Universitaria “Ospedali Riuniti” di Ancona, 60100 Ancona, Italy;
- Correspondence: ; Tel.: +39-071-596-5269
| | - Giovanna Viticchi
- Neurologic Clinic, Marche Polytechnic University, 60126 Ancona, Italy; (G.V.); (S.L.); (M.S.)
| | - Vincenzo Zaccone
- Internal and Subintensive Medicine Department, Azienda Ospedaliero-Universitaria “Ospedali Riuniti” di Ancona, 60100 Ancona, Italy;
| | - Emanuele Guerrieri
- Emergency Medicine Residency Program, Università Politecnica delle Marche, 60121 Ancona, Italy;
| | | | - Simona Luzzi
- Neurologic Clinic, Marche Polytechnic University, 60126 Ancona, Italy; (G.V.); (S.L.); (M.S.)
| | - Mauro Silvestrini
- Neurologic Clinic, Marche Polytechnic University, 60126 Ancona, Italy; (G.V.); (S.L.); (M.S.)
| |
Collapse
|
167
|
Varghese SM, Joy N, John AM, George G, Chandy GM, Benjamin AI. Sweet Memories or Not? A Comparative Study on Cognitive Impairment in Diabetes Mellitus. Front Public Health 2022; 10:822062. [PMID: 35186849 PMCID: PMC8854745 DOI: 10.3389/fpubh.2022.822062] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/07/2022] [Indexed: 11/13/2022] Open
Abstract
IntroductionType 2 Diabetes Mellitus is a modern-day epidemic and dementia has been declared as a global challenge. It is, therefore, worthwhile to investigate the effect that Diabetes has on cognition. Although effective screening is routinely carried out for various complications of Diabetes, its effect on Higher Mental Functions is often overlooked.MethodologyA cross-sectional analytical study to assess Cognitive Impairment was carried out on 800 participants, 400 diabetics and 400 non-diabetics attending a tertiary care center. The Addenbrooke's Cognitive Examination- III was used, which is a validated, highly sensitive tool having a maximum score of 100. Patients with a score < /= 82 were considered to have impaired Cognition. Statistical analysis was done using SPSSv.21. Suitable statistical tests like Mann–Whitney U, t-test, ROC curve and Logistic regression analysis were done.ResultsCognitive Impairment was present in 63.8% of the diabetics when compared to only 10.8% in the non-diabetics, with an Odds Ratio-8.78 (CI-4.47–17.22). The total ACE score in diabetics [median-82 (IQR-4), mean rank-270.06] was less compared to the non-diabetic patients [median- 85 (IQR-3), mean rank-530.94] (U = 27822, p-0.001). Attention, Memory, Language, and Visuospatial domains were significantly lower in the diabetics compared to the non-diabetics. However, the fluency domain was not affected. Hypertension and the presence of macrovascular diseases were significantly associated with Cognitive Impairment (p < 0.005). Those with Cognitive dysfunction also had higher mean RBS values and longer duration of Diabetes (p-0.001). The cut-off value for RBS (to distinguish people with and without Cognitive Impairment) from ROC curve was 142.5 (AUC = 0.834, Youden's Index-0.586, p-0.001) and for duration of Diabetes was 7.5 years (AUC = 0.847, Youden's Index-0.529, p-0.001).ConclusionThis paper highlights that Cognitive Impairment exists in a very high proportion of diabetic patients in Kerala. So, it is important that we do an early assessment of cognitive function in diabetes patients and manage them prudently. Early interventions may prove to be beneficial in the long run, considering the burden of diabetes and cognitive dysfunction associated with the disease.
Collapse
Affiliation(s)
- Sangeetha Merrin Varghese
- Department of Community Medicine, Believers Church Medical College, Thiruvalla, India
- *Correspondence: Sangeetha Merrin Varghese
| | - Niva Joy
- Medical Student, Believers Church Medical College, Thiruvalla, India
| | - Anulekha Mary John
- Department of Medicine, Believers Church Medical College, Thiruvalla, India
| | - Geomcy George
- Department of Radiation Oncology, Believers Church Medical College, Thiruvalla, India
| | | | - Anoop Ivan Benjamin
- Department of Community Medicine, Believers Church Medical College, Thiruvalla, India
| |
Collapse
|
168
|
Tang AS, Oskotsky T, Havaldar S, Mantyh WG, Bicak M, Solsberg CW, Woldemariam S, Zeng B, Hu Z, Oskotsky B, Dubal D, Allen IE, Glicksberg BS, Sirota M. Deep phenotyping of Alzheimer's disease leveraging electronic medical records identifies sex-specific clinical associations. Nat Commun 2022; 13:675. [PMID: 35115528 PMCID: PMC8814236 DOI: 10.1038/s41467-022-28273-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 01/18/2022] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disorder that is still not fully understood. Sex modifies AD vulnerability, but the reasons for this are largely unknown. We utilize two independent electronic medical record (EMR) systems across 44,288 patients to perform deep clinical phenotyping and network analysis to gain insight into clinical characteristics and sex-specific clinical associations in AD. Embeddings and network representation of patient diagnoses demonstrate greater comorbidity interactions in AD in comparison to matched controls. Enrichment analysis identifies multiple known and new diagnostic, medication, and lab result associations across the whole cohort and in a sex-stratified analysis. With this data-driven method of phenotyping, we can represent AD complexity and generate hypotheses of clinical factors that can be followed-up for further diagnostic and predictive analyses, mechanistic understanding, or drug repurposing and therapeutic approaches.
Collapse
Affiliation(s)
- Alice S Tang
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA.
- Graduate Program in Bioengineering, UCSF, San Francisco, CA, USA.
- School of Medicine, UCSF, San Francisco, CA, USA.
| | - Tomiko Oskotsky
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA
- Department of Pediatrics, UCSF, San Francisco, CA, USA
| | - Shreyas Havaldar
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - William G Mantyh
- Department of Neurology, University of Minnesota School of Medicine, Minneapolis, MN, USA
| | - Mesude Bicak
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Caroline Warly Solsberg
- Pharmaceutical Sciences and Pharmacogenomics, UCSF, San Francisco, CA, USA
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
- Memory and Aging Center, UCSF, San Francisco, CA, USA
| | - Sarah Woldemariam
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA
| | - Billy Zeng
- School of Medicine, UCSF, San Francisco, CA, USA
| | - Zicheng Hu
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA
| | - Boris Oskotsky
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA
| | - Dena Dubal
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Isabel E Allen
- Department of Epidemiology and Biostatistics, UCSF, San Francisco, CA, USA
| | - Benjamin S Glicksberg
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA.
- Department of Pediatrics, UCSF, San Francisco, CA, USA.
| |
Collapse
|
169
|
Khan S, Pati S, Singh S, Akhtar M, Khare P, Khan S, Shafi S, Najmi AK. Targeting hypercoagulation to alleviate Alzheimer's disease progression in metabolic syndrome. Int J Obes (Lond) 2022; 46:245-254. [PMID: 34686782 DOI: 10.1038/s41366-021-00977-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 09/17/2021] [Accepted: 09/27/2021] [Indexed: 11/09/2022]
Abstract
INTRODUCTION Metabolic Syndrome (MetS) constitutes an important risk factor for Alzheimer's disease (AD); however, the mechanism linking these two disorders has not been completely elucidated. Hence, hypercoagulation may account for the missing hallmark connecting MetS and AD. The present review proposes how hemostatic imbalance triggered in MetS advances in the context of AD. MetS causes interruption of insulin signaling and inflammation, inciting insulin resistance in the brain. Subsequently, neuroinflammation and brain endothelial dysfunction are prompted that further intensify the exorbitant infiltration of circulating lipids and platelet aggregation, thereby causing hypercoagulable state, impairing fibrinolysis and eventually inducing prothrombic state in the brain leading to neurodegeneration. OBJECTIVE This study aims to understand the role of hypercoagulation in triggering the progression of neurodegeneration in MetS. It also offers a few interventions to prevent the progression of AD in MetS targeting hypercoagulation. METHODS Literature studies based on MetS related neurodegeneration, the impact of coagulation on aggravating obesity and AD via the mechanisms of BBB disruption, neuroinflammation, and hypofibrinolysis. CONCLUSION The present paper proposes the hypothesis that hypercoagulation might amplify MetS associated insulin resistance, neuroinflammation, BBB disruption, and amyloid beta accumulation which eventually leads to AD.
Collapse
Affiliation(s)
- Sana Khan
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Soumya Pati
- Translational Neurobiology Laboratory. Host Pathogen Interactions & Disease Modeling Group, Dept. of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, Pin-201314, UP, India
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Mohd Akhtar
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Piush Khare
- Wave Pharma Regulatory Services Limited, New Delhi, India
| | - Saba Khan
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Sadat Shafi
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
170
|
Bendicho-Lavilla C, Seoane-Viaño I, Otero-Espinar FJ, Luzardo-Álvarez A. Fighting type 2 diabetes: Formulation strategies for peptide-based therapeutics. Acta Pharm Sin B 2022; 12:621-636. [PMID: 35256935 PMCID: PMC8897023 DOI: 10.1016/j.apsb.2021.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/27/2021] [Accepted: 05/15/2021] [Indexed: 12/12/2022] Open
Abstract
Diabetes mellitus is a major health problem with increasing prevalence at a global level. The discovery of insulin in the early 1900s represented a major breakthrough in diabetes management, with further milestones being subsequently achieved with the identification of glucagon-like peptide-1 (GLP-1) and the introduction of GLP-1 receptor agonists (GLP-1 RAs) in clinical practice. Moreover, the subcutaneous delivery of biotherapeutics is a well-established route of administration generally preferred over the intravenous route due to better patient compliance and prolonged drug absorption. However, current subcutaneous formulations of GLP-1 RAs present pharmacokinetic problems that lead to adverse reactions and treatment discontinuation. In this review, we discuss the current challenges of subcutaneous administration of peptide-based therapeutics and provide an overview of the formulations available for the different routes of administration with improved bioavailability and reduced frequency of administration.
Collapse
Affiliation(s)
- Carlos Bendicho-Lavilla
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Santiago de Compostela (USC), Santiago de Compostela 15782, Spain
- Paraquasil Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela 15706, Spain
| | - Iria Seoane-Viaño
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Santiago de Compostela (USC), Santiago de Compostela 15782, Spain
- Paraquasil Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela 15706, Spain
| | - Francisco J. Otero-Espinar
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Santiago de Compostela (USC), Santiago de Compostela 15782, Spain
- Paraquasil Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela 15706, Spain
| | - Asteria Luzardo-Álvarez
- Paraquasil Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela 15706, Spain
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Sciences, Campus de Lugo, University of Santiago de Compostela, Lugo 27002, Spain
| |
Collapse
|
171
|
Malbon AJ, Sordo L, Wilson LA, Gunn-Moore D, Paraschou G, Macintyre N, Schwarz T, McGorum B, Hahn C. Alzheimer-like pathology in the parietal cortex and hippocampus of aged donkeys. Neurobiol Aging 2022; 113:7-14. [DOI: 10.1016/j.neurobiolaging.2022.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 10/19/2022]
|
172
|
Zhou HL, Premont RT, Stamler JS. The manifold roles of protein S-nitrosylation in the life of insulin. Nat Rev Endocrinol 2022; 18:111-128. [PMID: 34789923 PMCID: PMC8889587 DOI: 10.1038/s41574-021-00583-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/08/2021] [Indexed: 02/04/2023]
Abstract
Insulin, which is released by pancreatic islet β-cells in response to elevated levels of glucose in the blood, is a critical regulator of metabolism. Insulin triggers the uptake of glucose and fatty acids into the liver, adipose tissue and muscle, and promotes the storage of these nutrients in the form of glycogen and lipids. Dysregulation of insulin synthesis, secretion, transport, degradation or signal transduction all cause failure to take up and store nutrients, resulting in type 1 diabetes mellitus, type 2 diabetes mellitus and metabolic dysfunction. In this Review, we make the case that insulin signalling is intimately coupled to protein S-nitrosylation, in which nitric oxide groups are conjugated to cysteine thiols to form S-nitrosothiols, within effectors of insulin action. We discuss the role of S-nitrosylation in the life cycle of insulin, from its synthesis and secretion in pancreatic β-cells, to its signalling and degradation in target tissues. Finally, we consider how aberrant S-nitrosylation contributes to metabolic diseases, including the roles of human genetic mutations and cellular events that alter S-nitrosylation of insulin-regulating proteins. Given the growing influence of S-nitrosylation in cellular metabolism, the field of metabolic signalling could benefit from renewed focus on S-nitrosylation in type 2 diabetes mellitus and insulin-related disorders.
Collapse
Affiliation(s)
- Hua-Lin Zhou
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Richard T Premont
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Jonathan S Stamler
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA.
| |
Collapse
|
173
|
Hoyos CM, Turner A, Ireland C, Naismith SL, Duffy SL. Brain oxidative stress and cognitive function in older adults with diabetes and pre-diabetes who are at risk for dementia. Diabetes Res Clin Pract 2022; 184:109178. [PMID: 34958845 DOI: 10.1016/j.diabres.2021.109178] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/08/2021] [Accepted: 12/11/2021] [Indexed: 01/04/2023]
Abstract
AIMS Diabetes is an established risk factor for dementia. This study aimed to examine the relationship between various cognitive domains, brain oxidative stress and markers of diabetes in older adults at risk for dementia. METHODS Older adults at risk for dementia underwent comprehensive neuropsychological and medical assessment. At risk was defined as those with subjective and/or objective cognitive impairment. Pre-diabetes and type 2 diabetes were defined using American Diabetes Association definitions for fasting blood glucose and HbA1c. Brain oxidative stress as indicated by glutathione (GSH) was assessed via magnetic resonance spectroscopy in the anterior cingulate cortex. RESULTS One-hundred and forty-seven older adults completed a neuropsychological assessment and fasting blood sample with 63 also undergoing magnetic resonance spectroscopy. Those with pre-diabetes/diabetes according to FBG had impaired memory retention, set-shifting and response inhibition, compared to those with normal blood glucose. In contrast, there were no significant differences in any cognitive outcome using the HbA1c definition. Increasing glucose and HbA1c levels were associated with reduced GSH concentration in the anterior cingulate. CONCLUSIONS This study demonstrates that in older adults at risk for dementia, having pre-diabetes or diabetes is associated with impaired memory and executive dysfunction. It also highlights the potential role of oxidative stress as a pathophysiological mechanism that may underpin the link between diabetes and cognitive dysfunction.
Collapse
Affiliation(s)
- Camilla M Hoyos
- The University of Sydney, Faculty of Science, School of Psychology, Sydney, New South Wales, Australia; The University of Sydney, Brain and Mind Centre, Sydney, New South Wales, Australia; Woolcock Institute of Medical Research, Centre for Sleep and Chronobiology, The University of Sydney, Sydney, New South Wales, Australia.
| | - Ashlee Turner
- The University of Sydney, Faculty of Science, School of Psychology, Sydney, New South Wales, Australia; The University of Sydney, Brain and Mind Centre, Sydney, New South Wales, Australia
| | - Catriona Ireland
- The University of Sydney, Faculty of Science, School of Psychology, Sydney, New South Wales, Australia; The University of Sydney, Brain and Mind Centre, Sydney, New South Wales, Australia
| | - Sharon L Naismith
- The University of Sydney, Faculty of Science, School of Psychology, Sydney, New South Wales, Australia; The University of Sydney, Brain and Mind Centre, Sydney, New South Wales, Australia; The University of Sydney, Charles Perkins Centre, Sydney, New South Wales, Australia
| | - Shantel L Duffy
- The University of Sydney, Brain and Mind Centre, Sydney, New South Wales, Australia; The University of Sydney, Charles Perkins Centre, Sydney, New South Wales, Australia; The University of Sydney, Faculty of Medicine and Health, Sydney, New South Wales, Australia
| |
Collapse
|
174
|
Effects of Chronic Arginase Inhibition with Norvaline on Tau Pathology and Brain Glucose Metabolism in Alzheimer's Disease Mice. Neurochem Res 2022; 47:1255-1268. [DOI: 10.1007/s11064-021-03519-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 12/11/2021] [Accepted: 12/27/2021] [Indexed: 12/23/2022]
|
175
|
Lewis LA, Urban CM, Hashim SA. A Non-Invasive Determination of Ketosis-Induced Elimination of Chronic Daytime Somnolence in a Patient with Late-Stage Dementia (Assessed with Type 3 Diabetes): A Potential Role of Neurogenesis. J Alzheimers Dis Rep 2022; 5:827-846. [PMID: 35088033 PMCID: PMC8764628 DOI: 10.3233/adr-210315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/20/2021] [Indexed: 11/15/2022] Open
Abstract
Background The study involved a female patient diagnosed with late-stage dementia, with chronic daytime somnolence (CDS) as a prominent symptom. Objective To explore whether her dementia resulted from Type 3 diabetes, and whether it could be reversed through ketosis therapy. Methods A ketogenic diet (KD) generating low-dose 100 μM Blood Ketone Levels (BKL) enhanced by a brief Ketone Mono Ester (KME) regimen with high-dose 2-4 mM BKLs was used. Results Three sets of data describe relief (assessed by % days awake) from CDS: 1) incremental, slow, time-dependent KD plus KME-induced sigmoid curve responses which resulted in partial wakefulness (0-40% in 255 days) and complete wakefulness (40-85% in 50 days); 2) both levels of wakefulness were shown to be permanent; 3) initial permanent relief from CDS with low-dose ketosis from 6.7% to 40% took 87 days. Subsequent low-dose recovery from illness-induced CDS (6.9% to 40%) took 10 days. We deduce that the first restoration involved permanent repair, and the second energized the repaired circuits. Conclusion The results suggest a role for ketosis in the elimination of CDS with the permanent functional restoration of the awake neural circuits of the Sleep-Wake cycle. We discuss whether available evidence supports ketosis-induced bioenergetics alone or whether other mechanisms of functional renewal were the basis for the elimination of CDS. Given evidence for permanent repair, two direct links between ketosis and neurogenesis in the adult mammalian brain are discussed: Ketosis-induced 1) brain-derived neurotrophic factor, resulting in neural progenitor/stem cell proliferation, and 2) mitochondrial bioenergetics-induced stem cell biogenesis.
Collapse
Affiliation(s)
- Leslie A Lewis
- York College of the City University of New York, Jamaica, NY, USA
| | - Carl M Urban
- Department of Medicine, The Dr. James J. Rahal, Jr. Division of Infectious Diseases, New York Presbyterian/Queens, Flushing, NY, USA
| | - Sami A Hashim
- Division of Endocrinology, Mt. Sinai Morningside, New York, NY, USA
| |
Collapse
|
176
|
DURAN R, PANCUR S, BAHADORİ F. The Effect of Type 2 Diabetes Mellitus on the Development of Alzheimer’s Disease and Its Molecular Mechanism. BEZMIALEM SCIENCE 2022. [DOI: 10.14235/bas.galenos.2020.4491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
|
177
|
Role of Creatine Supplementation in Conditions Involving Mitochondrial Dysfunction: A Narrative Review. Nutrients 2022; 14:nu14030529. [PMID: 35276888 PMCID: PMC8838971 DOI: 10.3390/nu14030529] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/24/2022] [Accepted: 01/24/2022] [Indexed: 12/14/2022] Open
Abstract
Creatine monohydrate (CrM) is one of the most widely used nutritional supplements among active individuals and athletes to improve high-intensity exercise performance and training adaptations. However, research suggests that CrM supplementation may also serve as a therapeutic tool in the management of some chronic and traumatic diseases. Creatine supplementation has been reported to improve high-energy phosphate availability as well as have antioxidative, neuroprotective, anti-lactatic, and calcium-homoeostatic effects. These characteristics may have a direct impact on mitochondrion's survival and health particularly during stressful conditions such as ischemia and injury. This narrative review discusses current scientific evidence for use or supplemental CrM as a therapeutic agent during conditions associated with mitochondrial dysfunction. Based on this analysis, it appears that CrM supplementation may have a role in improving cellular bioenergetics in several mitochondrial dysfunction-related diseases, ischemic conditions, and injury pathology and thereby could provide therapeutic benefit in the management of these conditions. However, larger clinical trials are needed to explore these potential therapeutic applications before definitive conclusions can be drawn.
Collapse
|
178
|
Gupta S, Singh V, Ganesh S, Singhal NK, Sandhir R. siRNA Mediated GSK3β Knockdown Targets Insulin Signaling Pathway and Rescues Alzheimer's Disease Pathology: Evidence from In Vitro and In Vivo Studies. ACS APPLIED MATERIALS & INTERFACES 2022; 14:69-93. [PMID: 34967205 DOI: 10.1021/acsami.1c15305] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Sporadic Alzheimer's disease (sAD) is a progressive neurodegenerative disorder with dysfunctional insulin signaling and energy metabolism. Emerging evidence suggests impairments in brain insulin responsiveness, glucose utilization, and energy metabolism may be major causes of amyloid precursor protein mishandling. The support for this notion comes from the studies wherein streptozotocin (STZ) induced brain insulin resistance in rodent model resulted in sAD-like neuropathology with cognitive decline. Our previous study showed a compromised insulin signaling pathway, glucose uptake, glucose metabolism, and energy homeostasis in STZ-induced glial-neuronal coculture and in vivo model of sAD. Various components of insulin signaling pathway were examined to understand the metabolic correlation, and GSK3β was selected for gene knockdown strategy to reverse sAD pathology based on the data. In the present study, we have synthesized carboxylated graphene oxide (GO) nanosheets functionalized with PEG and subsequently with polyethylenimine (PEI) to provide attachment sites for GSK3β siRNA. Our results showed that siRNA mediated knockdown of the GSK3β gene reduced expression of amyloid pathway genes (APP and BACE1), which was further confirmed by reduced amyloid beta (Aβ) levels in the in vitro STZ-induced sAD model. GSK3β knockdown also restored insulin signaling, AMPK and Mapk3 pathway by restoring the expression of corresponding candidate genes in these pathways (IR, Glut1/3, Prkaa1/2, Mapk3, BDNF) that reflected improved cellular energy homeostasis, neuronal proliferation, differentiation, maturation, and repair. Behavioral data from Morris water maze (MWM), open field (OF), novel object recognition (NOR), Y maze, and radial arm maze (RAM) tests showed that 0.5 μg nanoformulation (GOc-PP-siRNAGSK3β) intranasally for 7 days improved spatial memory, rescued anxiety like behavior, improved visual and working memory, and rescued exploratory behavior in STZ-induced sAD rats. GSK3β silencing resulted in decreased BACE1 expression and prevented accumulation of Aβ in the cortex and hippocampus. These molecular findings with improved behavioral performances were further correlated with reduced amyloid beta (Aβ) and neurofibrillary tangle (NFTs) formation in the cortex and hippocampus of GOc-PP-siRNAGSK3β administered sAD rats. Therefore, it is conceivable from the present study that nanoparticle-mediated targeting of GSK3β in the sAD appears to be a promising strategy to reverse sAD pathology.
Collapse
Affiliation(s)
- Smriti Gupta
- Department of Biochemistry, Panjab University, Chandigarh 160014, India
| | - Vishal Singh
- National Agri-Food Biotechnology Institute, Sector 81, S.A.S. Nagar, Mohali 140306, Punjab, India
| | - Subramaniam Ganesh
- Department of Biological Science and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Nitin K Singhal
- National Agri-Food Biotechnology Institute, Sector 81, S.A.S. Nagar, Mohali 140306, Punjab, India
| | - Rajat Sandhir
- Department of Biochemistry, Panjab University, Chandigarh 160014, India
| |
Collapse
|
179
|
Gayathri S, Raghu CH, Fayaz SM. Phytotherapeutics against Alzheimer's Disease: Mechanism, Molecular Targets and Challenges for Drug Development. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 21:409-426. [PMID: 34544351 DOI: 10.2174/1871527320666210920120612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 06/24/2021] [Accepted: 08/30/2021] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease is inflating worldwide and is combatted by only a few approved drugs. At best, these drugs treat symptomatic conditions by targeting cholinesterase and N-methyl- D-aspartate receptors. Most of the clinical trials in progress are focused on developing disease-modifying agents that aim at single targets. The 'one drug-one target' approach is failing in the case of Alzheimer's disease due to its labyrinth etiopathogenesis. Traditional medicinal systems like Ayurveda use a holistic approach encompassing the legion of medicinal plants exhibiting multimodal activity. Recent advances in high-throughput technologies have catapulted the research in the arena of Ayurveda, specifically in identifying plants with potent anti-Alzheimer's disease properties and their phytochemical characterization. Nonetheless, clinical trials of very few herbal medicines are in progress. This review is a compendium of Indian plants and ayurvedic medicines against Alzheimer's disease and their paraphernalia. A record of 230 plants that are found in India with anti-Alzheimer's disease potential and about 500 phytochemicals from medicinal plants have been solicited with the hope of exploring the unexplored. Further, the molecular targets of phytochemicals isolated from commonly used medicinal plants, such as Acorus calamus, Bacopa monnieri, Convolvulus pluricaulis, Tinospora cordifolia and Withania somnifera, have been reviewed with respect to their multidimensional property, such as antioxidant, anti-inflammation, anti-aggregation, synaptic plasticity modulation, cognition, and memory-enhancing activity. In addition, the strengths and challenges in ayurvedic medicine that limit its use as mainstream therapy are discussed, and a framework for the development of herbal medicine has been proposed.
Collapse
Affiliation(s)
- S Gayathri
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka- 576104, India
| | - Chandrashekar H Raghu
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - S M Fayaz
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka- 576104, India
| |
Collapse
|
180
|
Chandarana CV, Roy S. Comprehensive Review on Neuro-degenerative Type 3 DM. Curr Diabetes Rev 2022; 18:e131221198790. [PMID: 34961464 DOI: 10.2174/1573399818666211213103624] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/28/2021] [Accepted: 10/11/2021] [Indexed: 11/22/2022]
Abstract
According to research, Alzheimer's disease (AD) is considered a metabolic illness caused by defective insulin signaling, insulin resistance, and low insulin levels in the brain. Type 3 diabetes has been postulated for AD because reduced insulin signaling has molecular and physiological consequences that are comparable to type I and type 2 diabetes mellitus, respectively. The similarities between type 2 diabetes and Alzheimer's disease suggest that these clinical trials might yield therapeutic benefits. However, it is important to note that lowering your risk of Alzheimer's dementia, whether you have diabetes or not, is still a multidimensional process involving factors like exercise, smoking, alcohol, food, and mental challenges. The current aim is to show that the relationship between T3D and AD is based on both the processing of amyloid-β (Aβ) precursor protein toxicity and the clearance of Aβ, which are the results of impaired insulin signaling. The brain's metabolism, with its high lipid content and energy needs, places excess demands on mitochondria and appears more susceptible to oxidative damage than the rest of the body. Current data suggests that increased oxidative stress relates to amyloid-β (Aβ) pathology and the onset of AD.
Collapse
Affiliation(s)
- Chandani V Chandarana
- Department of Pharmaceutical Quality Assurance, SSR College of Pharmacy, Sayli, Silvassa-396230, India
| | - Salona Roy
- SSR College of Pharmacy, Silvassa, India
| |
Collapse
|
181
|
Frank CJ, McNay EC. Breakdown of the blood-brain barrier: A mediator of increased Alzheimer's risk in patients with metabolic disorders? J Neuroendocrinol 2022; 34:e13074. [PMID: 34904299 PMCID: PMC8791015 DOI: 10.1111/jne.13074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 11/12/2021] [Accepted: 11/26/2021] [Indexed: 01/03/2023]
Abstract
Metabolic disorders (MDs), including type 1 and 2 diabetes and chronic obesity, are among the faster growing diseases globally and are a primary risk factor for Alzheimer's disease (AD). The term "type-3 diabetes" has been proposed for AD due to the interrelated cellular, metabolic, and immune features shared by diabetes, insulin resistance (IR), and the cognitive impairment and neurodegeneration found in AD. Patients with MDs and/or AD commonly exhibit altered glucose homeostasis and IR; systemic chronic inflammation encompassing all of the periphery, blood-brain barrier (BBB), and central nervous system; pathological vascular remodeling; and increased BBB permeability that allows transfusion of neurotoxic molecules from the blood to the brain. This review summarizes the components of the BBB, mechanisms through which MDs alter BBB permeability via immune and metabolic pathways, the contribution of BBB dysfunction to the manifestation and progression of AD, and current avenues of therapeutic research that address BBB permeability. In addition, issues with the translational applicability of current animal models of AD regarding BBB dysfunction and proposals for future directions of research that address the relationship between MDs, BBB dysfunction, and AD are discussed.
Collapse
Affiliation(s)
- Corey J Frank
- Behavioral Neuroscience, University at Albany, SUNY, Albany, NY, USA
| | - Ewan C McNay
- Behavioral Neuroscience, University at Albany, SUNY, Albany, NY, USA
| |
Collapse
|
182
|
Ravipati K, Chen Y, Manns JR. Reassessing Diabetes and APOE Genotype as Potential Interacting Risk Factors for Alzheimer's Disease. Am J Alzheimers Dis Other Demen 2022; 37:15333175211070912. [PMID: 35041557 PMCID: PMC10623968 DOI: 10.1177/15333175211070912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective: To assess whether diabetes alone or in association with Apolipoprotein E (APOE) ε4 genotype increases the risk of Alzheimer's Disease (AD) diagnosis. Methods: A retrospective cohort study of 33,456 participants from the National Alzheimer's Coordinating Center database. Results: Participants with one or two APOE ε4 alleles had 2.71 (CI:2.55-2.88) and 9.37 (CI:8.14-10.78) times higher odds of AD diagnosis, respectively, relative to those with zero ε4 alleles. In contrast, diabetic participants showed 1.07 (CI:0.96-1.18) times higher odds of AD relative to nondiabetics. Diabetes did not exacerbate the odds of AD in APOE ε4 carriers. APOE ε4 carriage was correlated with declines in long-term memory and verbal fluency, which were strongly correlated with conversion to AD. However, diabetes was correlated with working memory decline, which had a relatively weak correlation with AD. Conclusions: Unlike APOE ε4, there was little evidence that diabetes was a risk factor for AD.
Collapse
Affiliation(s)
- Kaushik Ravipati
- Neuroscience and Behavioral Biology, Emory University, Atlanta, GA USA
- Florida International University Herbert Wertheim College of Medicine, Miami, FL USA
| | - Yunxiao Chen
- Department of Statistics, London School of Economics and Political Science, London, UK
- Department of Psychology, Emory University, Atlanta, GA USA
| | | |
Collapse
|
183
|
In Vitro Comparative Study of Solid Lipid and PLGA Nanoparticles Designed to Facilitate Nose-to-Brain Delivery of Insulin. Int J Mol Sci 2021; 22:ijms222413258. [PMID: 34948054 PMCID: PMC8703723 DOI: 10.3390/ijms222413258] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 01/25/2023] Open
Abstract
The brain insulin metabolism alteration has been addressed as a pathophysiological factor underlying Alzheimer's disease (AD). Insulin can be beneficial in AD, but its macro-polypeptide nature negatively influences the chances of reaching the brain. The intranasal (IN) administration of therapeutics in AD suggests improved brain-targeting. Solid lipid nanoparticles (SLNs) and poly(lactic-co-glycolic acid) nanoparticles (PLGA NPs) are promising carriers to deliver the IN-administered insulin to the brain due to the enhancement of the drug permeability, which can even be improved by chitosan-coating. In the present study, uncoated and chitosan-coated insulin-loaded SLNs and PLGA NPs were formulated and characterized. The obtained NPs showed desirable physicochemical properties supporting IN applicability. The in vitro investigations revealed increased mucoadhesion, nasal diffusion, and drug release rate of both insulin-loaded nanocarriers over native insulin with the superiority of chitosan-coated SLNs. Cell-line studies on human nasal epithelial and brain endothelial cells proved the safety IN applicability of nanoparticles. Insulin-loaded nanoparticles showed improved insulin permeability through the nasal mucosa, which was promoted by chitosan-coating. However, native insulin exceeded the blood-brain barrier (BBB) permeation compared with nanoparticulate formulations. Encapsulating insulin into chitosan-coated NPs can be beneficial for ensuring structural stability, enhancing nasal absorption, followed by sustained drug release.
Collapse
|
184
|
Hadrich F, Chamkha M, Sayadi S. Protective effect of olive leaves phenolic compounds against neurodegenerative disorders: Promising alternative for Alzheimer and Parkinson diseases modulation. Food Chem Toxicol 2021; 159:112752. [PMID: 34871668 DOI: 10.1016/j.fct.2021.112752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/24/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023]
Abstract
The main objective of this work was to review literature on compounds extracted from olive tree leaves, such as simple phenols (hydroxytyrosol) and flavonoids (Apigenin, apigenin-7-O-glucoside, luteolin.) and their diverse pharmacological activities as antioxidant, antimicrobial, anti-viral, anti-obesity, anti-inflammatory and neuroprotective properties. In addition, the study discussed the key mechanisms underlying their neuroprotective effects. This study adopted an approach of collecting data through the databases provided by ScienceDirect, SCOPUS, MEDLINE, PubMed and Google Scholar. This review revealed that there was an agreement on the great impact of olive tree leaves phenolic compounds on many metabolic syndromes as well as on the most prevalent neurodegenerative diseases such as Alzheimer and Parkinson. These findings would be of great importance for the use of olive tree leaves extracts as a food supplement and/or a source of drugs for many diseases. In addition, this review would of great help to beginning researchers in the field since it would offer them a general overview of the studies undertaken in the last two decades on the topic.
Collapse
Affiliation(s)
- Fatma Hadrich
- Environmental Bioprocesses Laboratory, Center of Biotechnology of Sfax, P.O. Box 1177, 3038, Sfax, Tunisia.
| | - Mohamed Chamkha
- Environmental Bioprocesses Laboratory, Center of Biotechnology of Sfax, P.O. Box 1177, 3038, Sfax, Tunisia
| | - Sami Sayadi
- Biotechnology Program, Center of Sustainable Development, College of Arts and Sciences, Qatar University, Doha, 2713, Qatar.
| |
Collapse
|
185
|
Mittal N, Mittal R. Repurposing old molecules for new indications: Defining pillars of success from lessons in the past. Eur J Pharmacol 2021; 912:174569. [PMID: 34653378 DOI: 10.1016/j.ejphar.2021.174569] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/30/2021] [Accepted: 10/11/2021] [Indexed: 02/06/2023]
Abstract
Drug repurposing or studying existing drugs for potential therapeutic utility in newer indications has been identified as an attractive option for treating a number of diseases. Various strategies of drug repurposing include serendipitous observation of drug's unexpected effects, directing the failed investigational drugs to new indications and currently adopted systematic approach to identify, screen and develop existing drug molecules for new off-label indications. Drug repurposing is able to constructively overcome the bottleneck restraints encountered during traditional de novo drug development process in grounds of timelines, cost and resources. However, success rates of drug repurposing programs are not very impressive. Through a meticulous examination of some failed repurposing attempts we aimed to identify key factors leading to high attrition rate in such studies. Based on the fundamental elements of knowledge and evaluation, we have defined four pillars toward improving success rate in drug repurposing programs viz. sound knowledge of the repurposed drug's pharmacological characteristics (pillar 1: drug pharmacology); drug formulation considerations in new indication (pillar 2: drug formulation); evaluation in representative biological assays with translational potential (pillar 3: evaluation in biological assays); and robust clinical trial methodologies including biomarker driven approach to provide conclusive evidence of repurposed drug's efficacy in new indication (pillar 4: clinical evaluation). In addition to the pharmacological challenges, certain regulatory concerns, including lack of clear guidelines for evaluation and market exclusivity pose hurdles in the application of drug repurposing, which may however be overcome to a great extent by adopting some strategies as discussed in this review.
Collapse
Affiliation(s)
- Niti Mittal
- Dept. of Pharmacology, Postgraduate Institute of Medical Sciences, Rohtak, 124001, India.
| | - Rakesh Mittal
- Dept. of Pharmacology, Postgraduate Institute of Medical Sciences, Rohtak, 124001, India
| |
Collapse
|
186
|
Tournissac M, Leclerc M, Valentin-Escalera J, Vandal M, Bosoi CR, Planel E, Calon F. Metabolic determinants of Alzheimer's disease: A focus on thermoregulation. Ageing Res Rev 2021; 72:101462. [PMID: 34534683 DOI: 10.1016/j.arr.2021.101462] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/09/2021] [Accepted: 09/11/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a complex age-related neurodegenerative disease, associated with central and peripheral metabolic anomalies, such as impaired glucose utilization and insulin resistance. These observations led to a considerable interest not only in lifestyle-related interventions, but also in repurposing insulin and other anti-diabetic drugs to prevent or treat dementia. Body temperature is the oldest known metabolic readout and mechanisms underlying its maintenance fail in the elderly, when the incidence of AD rises. This raises the possibility that an age-associated thermoregulatory deficit contributes to energy failure underlying AD pathogenesis. Brown adipose tissue (BAT) plays a central role in thermogenesis and maintenance of body temperature. In recent years, the modulation of BAT activity has been increasingly demonstrated to regulate energy expenditure, insulin sensitivity and glucose utilization, which could also provide benefits for AD. Here, we review the evidence linking thermoregulation, BAT and insulin-related metabolic defects with AD, and we propose mechanisms through which correcting thermoregulatory impairments could slow the progression and delay the onset of AD.
Collapse
|
187
|
Sharma M, Dey CS. Role of Akt isoforms in neuronal insulin signaling and resistance. Cell Mol Life Sci 2021; 78:7873-7898. [PMID: 34724097 PMCID: PMC11073101 DOI: 10.1007/s00018-021-03993-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 02/04/2023]
Abstract
The aim of the present study was to determine the role of Akt isoforms in insulin signaling and resistance in neuronal cells. By silencing Akt isoforms individually and in pairs, in Neuro-2a and HT22 cells we observed that, in insulin-sensitive condition, Akt isoforms differentially reduced activation of AS160 and glucose uptake with Akt2 playing the major role. Under insulin-resistant condition, phosphorylation of all isoforms and glucose uptake were severely affected. Over-expression of individual isoforms in insulin-sensitive and resistant cells differentially reversed AS160 phosphorylation with concomitant reversal in glucose uptake indicating a compensatory role of Akt isoforms in controlling neuronal insulin signaling. Post-insulin stimulation Akt2 translocated to the membrane the most followed by Akt3 and Akt1, decreasing glucose uptake in the similar order in insulin-sensitive cells. None of the Akt isoforms translocated in insulin-resistant cells or high-fat-diet mediated diabetic mice brain cells. Based on our data, insulin-dependent differential translocation of Akt isoforms to the plasma membrane turns out to be the key factor in determining Akt isoform specificity. Thus, isoforms play parallel with predominant role by Akt2, and compensatory yet novel role by Akt1 and Akt3 to regulate neuronal insulin signaling, glucose uptake, and insulin-resistance.
Collapse
Affiliation(s)
- Medha Sharma
- Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi, 110016, India
| | - Chinmoy Sankar Dey
- Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|
188
|
Abstract
The Akt isoforms-AS160-GLUT4 axis is the primary axis that governs glucose homeostasis in the body. The first step on the path to insulin resistance is deregulated Akt isoforms. This could be Akt isoform expression, its phosphorylation, or improper isoform-specific redistribution to the plasma membrane in a specific tissue system. The second step is deregulated AS160 expression, its phosphorylation, improper dissociation from glucose transporter storage vesicles (GSVs), or its inability to bind to 14-3-3 proteins, thus not allowing it to execute its function. The final step is improper GLUT4 translocation and aberrant glucose uptake. These processes lead to insulin resistance in a tissue-specific way affecting the whole-body glucose homeostasis, eventually progressing to an overt diabetic phenotype. Thus, the relationship between these three key proteins and their proper regulation comes out as the defining axis of insulin signaling and -resistance. This review summarizes the role of this central axis in insulin resistance and disease in a new light.
Collapse
Affiliation(s)
- Medha Sharma
- Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi, 110016, India
| | - Chinmoy Sankar Dey
- Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|
189
|
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
|
190
|
The potential roles of excitatory-inhibitory imbalances and the repressor element-1 silencing transcription factor in aging and aging-associated diseases. Mol Cell Neurosci 2021; 117:103683. [PMID: 34775008 DOI: 10.1016/j.mcn.2021.103683] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 11/02/2021] [Accepted: 11/06/2021] [Indexed: 12/28/2022] Open
Abstract
Disruptions to the central excitatory-inhibitory (E/I) balance are thought to be related to aging and underlie a host of neural pathologies, including Alzheimer's disease. Aging may induce an increase in excitatory signaling, causing an E/I imbalance, which has been linked to shorter lifespans in mice, flies, and worms. In humans, extended longevity correlates to greater repression of genes involved in excitatory neurotransmission. The repressor element-1 silencing transcription factor (REST) is a master regulator in neural cells and is believed to be upregulated with senescent stimuli, whereupon it counters hyperexcitability, insulin/insulin-like signaling pathway activity, oxidative stress, and neurodegeneration. This review examines the putative mechanisms that distort the E/I balance with aging and neurodegeneration, and the putative roles of REST in maintaining neuronal homeostasis.
Collapse
|
191
|
Chakraborty A, Sami SA, Marma KKS. A comprehensive review on RAGE-facilitated pathological pathways connecting Alzheimer’s disease, diabetes mellitus, and cardiovascular diseases. THE EGYPTIAN JOURNAL OF INTERNAL MEDICINE 2021. [DOI: 10.1186/s43162-021-00081-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Alzheimer’s disease (AD), cardiovascular disease (CVD), and diabetes are some of the most common causes of morbidity and mortality among the aging populations and cause a heavy burden on the worldwide healthcare system. In this review, we briefly highlighted cellular inflammation-based pathways of diabetes mellitus and CVD through receptor for advanced glycation end products AGEs or RAGE leading to Alzheimer’s disease and interrelation between these vascular and metabolic disorders. The articles were retrieved from Google Scholar, Science Direct, and PubMed databases using the following terms: Alzheimer’s; AGEs; RAGE; RAGE in Alzheimer’s; AGEs in Alzheimer’s; RAGE in diabetes; RAGE related pathways of CVD; RAGE in hypertension; RAGE and RAS system; RAGE and oxidative stress.
Main body of the abstract
AD is a neurodegenerative disease characterized by cognitive dysfunction and neuronal cell death. Vascular complications like hypertension, coronary artery disease, and atherosclerosis as well as metabolic syndromes like obesity and diabetes are related to the pathophysiology of AD. RAGE plays significant role in the onset and progression of AD. Amyloid plaques and neurofibrillary tangles (NFT) are two main markers of AD that regulates via RAGE and other RAGE/ligands interactions which also induces oxidative stress and a cascade of other cellular inflammation pathways leading to AD. Though AD and diabetes are two different disorders but may be inter-linked by AGEs and RAGE. In long-term hyperglycemia, upregulated AGEs interacts with RAGE and produces reactive oxygen species which induces further inflammation and vascular complications. Aging, hypercholesterolemia, atherosclerosis, hypertension, obesity, and inflammation are some of the main risk factors for both diabetes and dementia. Chronic hypertension and coronary artery disease disrupt the functions of the blood-brain barrier and are responsible for the accumulation of senile plaques and NFTs.
Short conclusion
RAGE plays a role in the etiology of Aβ and tau hyperphosphorylation, both of which contribute to cognitive impairment. So far, targeting RAGE may provide a potential sight to develop therapies against some metabolic disorders.
Collapse
|
192
|
Christopher Kwon YI, Xie W, Zhu H, Xie J, Shinn K, Juckel N, Vince R, More SS, Lee MK. γ-Glutamyl-Transpeptidase-Resistant Glutathione Analog Attenuates Progression of Alzheimer's Disease-like Pathology and Neurodegeneration in a Mouse Model. Antioxidants (Basel) 2021; 10:antiox10111796. [PMID: 34829667 PMCID: PMC8614797 DOI: 10.3390/antiox10111796] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/26/2021] [Accepted: 11/08/2021] [Indexed: 02/01/2023] Open
Abstract
Oxidative stress in Alzheimer’s disease (AD) is mediated, in part, by the loss of glutathione (GSH). Previous studies show that γ-glutamyl transpeptidase (GGT)-resistant GSH analog, Ψ-GSH, improves brain GSH levels, reduces oxidative stress markers in brains of APP/PS1 transgenic mice, a mouse model of AD, and attenuates early memory deficits in the APP/PS1 model. Herein, we examined whether Ψ-GSH can attenuate the disease progression when administered following the onset of AD-like pathology in vivo. Cohorts of APP/PS1 mice were administered Ψ-GSH for 2 months starting at 8 month or 12 months of age. We show that Ψ-GSH treatment reduces indices of oxidative stress in older mice by restoration of enzyme glyoxalase-1 (Glo-1) activity and reduces levels of insoluble Aβ. Quantitative neuropathological analyses show that Ψ-GSH treatment significantly reduces Aβ deposition and brain inflammation in APP/PS1 mice compared to vehicle-treated mice. More importantly, Ψ-GSH treatment attenuated the progressive loss of cortical TH+ afferents and the loss of TH+ neurons in the locus coeruleus (LC). Collectively, the results show that Ψ-GSH exhibits significant antioxidant activity in aged APP/PS1 mice and chronic Ψ-GSH treatment administered after the onset of AD pathology can reverse/slow further progression of AD-like pathology and neurodegeneration in vivo.
Collapse
Affiliation(s)
- Ye In Christopher Kwon
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA; (Y.I.C.K.); (K.S.); (N.J.)
| | - Wei Xie
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (W.X.); (H.Z.); (J.X.); (R.V.)
| | - Haizhou Zhu
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (W.X.); (H.Z.); (J.X.); (R.V.)
| | - Jiashu Xie
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (W.X.); (H.Z.); (J.X.); (R.V.)
| | - Keaton Shinn
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA; (Y.I.C.K.); (K.S.); (N.J.)
| | - Nicholas Juckel
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA; (Y.I.C.K.); (K.S.); (N.J.)
| | - Robert Vince
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (W.X.); (H.Z.); (J.X.); (R.V.)
| | - Swati S. More
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (W.X.); (H.Z.); (J.X.); (R.V.)
- Correspondence: (S.S.M.); (M.K.L.)
| | - Michael K. Lee
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA; (Y.I.C.K.); (K.S.); (N.J.)
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
- Correspondence: (S.S.M.); (M.K.L.)
| |
Collapse
|
193
|
Behl T, Arora A, Sehgal A, Singh S, Sharma N, Bhatia S, Al-Harrasi A, Bungau S, Mostafavi E. Molecular and Biochemical Pathways Encompassing Diabetes Mellitus and Dementia. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:542-556. [PMID: 34758720 DOI: 10.2174/1871527320666211110115257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/25/2021] [Accepted: 09/15/2021] [Indexed: 11/22/2022]
Abstract
Diabetes mellitus is a major metabolic disorder that has now emerged as an epidemic, and it affects the brain through an array of pathways. Diabetes mellitus patients can develop pathological changes in the brain, which eventually take the shape of mild cognitive impairment progressing to Alzheimer's Disease. A number of preclinical and clinical studies demonstrate this fact, and it comes out to be those molecular pathways such as amyloidogenesis, oxidative stress, inflammation, and impaired insulin signaling are identical in diabetes mellitus and dementia. However, the critical player involved in the vicious cycle of diabetes mellitus and dementia is insulin, whose signaling, when impaired in diabetes mellitus (both type 1 and 2), leads to a decline in cognition, although other pathways are also essential contributors. Moreover, it is not only that diabetes mellitus patients indicate cognitive decline at a later stage; many Alzheimer's Disease patients also reflect symptoms of diabetes mellitus, thus creating a vicious cycle inculcating a web of complex molecular mechanisms and hence categorizing Alzheimer's Disease as 'brain diabetes'. Thus, it is practical to suggest that anti-diabetic drugs are beneficial in Alzheimer's Disease; but only smaller trials, not the larger ones, have showcased positive outcomes mainly because of the late onset of therapy. Therefore, it is extremely important to develop more of such molecules that target insulin in dementia patients along with such methods that diagnose impaired insulin signaling and the associated cognitive decline so that early therapy may be initiated and the progression of the disease be prevented.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Arpita Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Saurabh Bhatia
- Amity Institute of Pharmacy, Amity University, Haryana. India
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa. Oman
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea. Romania
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA. United States
| |
Collapse
|
194
|
Hafdi M, Hoevenaar-Blom MP, Richard E. Multi-domain interventions for the prevention of dementia and cognitive decline. Cochrane Database Syst Rev 2021; 11:CD013572. [PMID: 34748207 PMCID: PMC8574768 DOI: 10.1002/14651858.cd013572.pub2] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Dementia is a worldwide concern. Its global prevalence is increasing. Currently, no effective medical treatment exists to cure or to delay the onset of cognitive decline or dementia. Up to 40% of dementia is attributable to potentially modifiable risk factors, which has led to the notion that targeting these risk factors might reduce the incidence of cognitive decline and dementia. Since sporadic dementia is a multifactorial condition, thought to derive from multiple causes and risk factors, multi-domain interventions may be more effective for the prevention of dementia than those targeting single risk factors. OBJECTIVES To assess the effects of multi-domain interventions for the prevention of cognitive decline and dementia in older adults, including both unselected populations and populations at increased risk of cognitive decline and dementia. SEARCH METHODS We searched ALOIS, the Cochrane Dementia and Cognitive Improvement Group's register, MEDLINE (Ovid SP), Embase (Ovid SP), PsycINFO (Ovid SP), CINAHL (EBSCOhost), Web of Science Core Collection (ISI Web of Science), LILACS (BIREME), and ClinicalTrials.gov on 28 April 2021. We also reviewed citations of reference lists of included studies, landmark papers, and review papers to identify additional studies and assessed their suitability for inclusion in the review. SELECTION CRITERIA We defined a multi-domain intervention as an intervention with more than one component, pharmacological or non-pharmacological, but not consisting only of two or more drugs with the same therapeutic target. We included randomised controlled trials (RCTs) evaluating the effect of such an intervention on cognitive functioning and/or incident dementia. We accepted as control conditions any sham intervention or usual care, but not single-domain interventions intended to reduce dementia risk. We required studies to have a minimum of 400 participants and an intervention and follow-up duration of at least 12 months. DATA COLLECTION AND ANALYSIS We initially screened search results using a 'crowdsourcing' method in which members of Cochrane's citizen science platform identify RCTs. We screened the identified citations against inclusion criteria by two review authors working independently. At least two review authors also independently extracted data, assessed the risk of bias and applied the GRADE approach to assess the certainty of evidence. We defined high-certainty reviews as trials with a low risk of bias across all domains other than blinding of participants and personnel involved in administering the intervention (because lifestyle interventions are difficult to blind). Critical outcomes were incident dementia, incident mild cognitive impairment (MCI), cognitive decline measured with any validated measure, and mortality. Important outcomes included adverse events (e.g. cardiovascular events), quality of life, and activities of daily living (ADL). Where appropriate, we synthesised data in random-effects meta-analyses. We expressed treatment effects as risk ratios (RRs) and mean differences (MDs) with 95% confidence intervals (CIs). MAIN RESULTS We included nine RCTs (18.452 participants) in this review. Two studies reported incident dementia as an outcome; all nine studies reported a measure for cognitive functioning. Assessment of cognitive functioning was very heterogeneous across studies, ranging from complete neuropsychological assessments to short screening tests such as the mini-mental state examination (MMSE). The duration of the interventions varied from 12 months to 10 years. We compared multi-domain interventions against usual care or a sham intervention. Positive MDs and RRs <1 favour multi-domain interventions over control interventions. For incident dementia, there was no evidence of a difference between the multi-domain intervention group and the control group (RR 0.94, 95% CI 0.76 to 1.18; 2 studies; 7256 participants; high-certainty evidence). There was a small difference in composite Z-score for cognitive function measured with a neuropsychological test battery (NTB) (MD 0.03, 95% CI 0.01 to 0.06; 3 studies; 4617 participants; high-certainty evidence) and with the Montreal Cognitive Assessment (MoCA) scale (MD 0.76 point, 95% CI 0.05 to 1.46; 2 studies; 1554 participants), but the certainty of evidence for the MoCA was very low (due to serious risk of bias, inconsistency and indirectness) and there was no evidence of an effect on the MMSE (MD 0.02 point, 95% CI -0.06 to 0.09; 6 studies; 8697participants; moderate-certainty evidence). There was no evidence of an effect on mortality (RR 0.93, 95% CI 0.84 to 1.04; 4 studies; 11,487 participants; high-certainty evidence). There was high-certainty evidence for an interaction of the multi-domain intervention with ApoE4 status on the outcome of cognitive function measured with an NTB (carriers MD 0.14, 95% CI 0.04 to 0.25, noncarriers MD 0.04, 95% CI -0.02 to 0.10, P for interaction 0.09). There was no clear evidence for an interaction with baseline cognitive status (defined by MMSE-score) on cognitive function measured with an NTB (low baseline MMSE group MD 0.06, 95% CI 0.01 to 0.11, high baseline MMSE group MD 0.01, 95% CI -0.01 to 0.04, P for interaction 0.12), nor was there clear evidence for an effect in participants with a Cardiovascular Risk Factors, Aging, and Incidence of Dementia (CAIDE) score > 6 points (MD 0.07, 95%CI -0.00 to 0.15). AUTHORS' CONCLUSIONS We found no evidence that multi-domain interventions can prevent incident dementia based on two trials. There was a small improvement in cognitive function assessed by a NTB in the group of participants receiving a multi-domain intervention, although this effect was strongest in trials offering cognitive training within the multi-domain intervention, making it difficult to rule out a potential learning effect. Interventions were diverse in terms of their components and intensity.
Collapse
Affiliation(s)
- Melanie Hafdi
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Marieke P Hoevenaar-Blom
- Department of Public and Occupational Health, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Edo Richard
- Department of Public and Occupational Health, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Department of Neurology, Donders Institute for Brain, Behaviour and Cognition, Radboud University Nijmegen Medical Center, Nijmegen, Netherlands
| |
Collapse
|
195
|
Hobday AL, Parmar MS. The Link Between Diabetes Mellitus and Tau Hyperphosphorylation: Implications for Risk of Alzheimer's Disease. Cureus 2021; 13:e18362. [PMID: 34725612 PMCID: PMC8555851 DOI: 10.7759/cureus.18362] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
Diabetes mellitus (DM) is characterized by hyperglycemia caused by a lack of insulin, insulin resistance, or both. It is associated with the development of secondary complications resulting in several comorbidities. Recent studies have revealed an increased risk of developing cognitive dysfunction or dementia in diabetes patients. Diabetes mellitus is considered a risk factor for many neurodegenerative diseases, including Alzheimer's disease (AD). There is increasing evidence to support a link between DM and AD. Studies have shown the dysfunction of insulin signaling in the brain, resulting in increased tau protein phosphorylation (hyperphosphorylation), a hallmark and biomarker of AD pathology, leading to accumulation of neurofibrillary tangles. In DM, the insulin dysfunction in the brain is reported to alter the glycogen synthase kinase-3β (GSK-3β) activity showing to enhance tau phosphorylation. In DM and AD, GSK-3β signaling has been involved in the physiological and pathological processes, respectively. This potentially explains why DM patients have an increased risk of developing AD with disease progression and aging. Interestingly, several in vivo studies with oral antidiabetic drugs and insulin treatment in DM have improved cognitive function and decreased tau hyperphosphorylation. This article will review the relationship between DM and AD as it relates to tau pathology. More understanding of the link between DM and AD could change the approach researchers and clinicians take toward both diseases, potentially leading to new treatments and preventative strategies in the future.
Collapse
Affiliation(s)
- Amy L Hobday
- Foundational Sciences, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Clearwater, USA
| | - Mayur S Parmar
- Foundational Sciences, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Clearwater, USA
| |
Collapse
|
196
|
Disentangling Mitochondria in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms222111520. [PMID: 34768950 PMCID: PMC8583788 DOI: 10.3390/ijms222111520] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/21/2021] [Accepted: 10/22/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a major cause of dementia in older adults and is fast becoming a major societal and economic burden due to an increase in life expectancy. Age seems to be the major factor driving AD, and currently, only symptomatic treatments are available. AD has a complex etiology, although mitochondrial dysfunction, oxidative stress, inflammation, and metabolic abnormalities have been widely and deeply investigated as plausible mechanisms for its neuropathology. Aβ plaques and hyperphosphorylated tau aggregates, along with cognitive deficits and behavioral problems, are the hallmarks of the disease. Restoration of mitochondrial bioenergetics, prevention of oxidative stress, and diet and exercise seem to be effective in reducing Aβ and in ameliorating learning and memory problems. Many mitochondria-targeted antioxidants have been tested in AD and are currently in development. However, larger streamlined clinical studies are needed to provide hard evidence of benefits in AD. This review discusses the causative factors, as well as potential therapeutics employed in the treatment of AD.
Collapse
|
197
|
Nitchingham A, Milne A, Toson B, Tuch B, Agar M, Close J, Caplan G. Intranasal insulin for treatment of delirium in older hospitalised patients: study protocol for a randomised controlled trial. BMJ Open 2021; 11:e050765. [PMID: 34667006 PMCID: PMC8527126 DOI: 10.1136/bmjopen-2021-050765] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Delirium is one of the most common conditions diagnosed in hospitalised older people and is associated with numerous adverse outcomes, yet there are no proven pharmacological treatments. Recent research has identified cerebral glucose hypometabolism as a pathophysiological mechanism offering a therapeutic target in delirium. Insulin, delivered via the intranasal route, acts directly on the central nervous system and has been shown to enhance cerebral metabolism and improve cognition in patients with mild cognitive impairment and dementia. This trial will determine whether intranasal insulin can reduce the duration of delirium in older hospitalised patients. METHODS AND ANALYSIS This is a prospective randomised, placebo-controlled, double-blind study with 6 months follow-up. One hundred patients aged 65 years or older presenting to hospital with delirium admitted under geriatric medicine will be recruited. Participants will be randomised to intranasal insulin detemir or placebo administered twice daily until delirium resolves, defined as Confusion Assessment Method (CAM) negative for 2 days, or discharge from hospital. The primary outcome measure will be duration of delirium using the CAM. Secondary outcome measures will include length of hospital stay, severity of delirium, adherence to treatment, hospital complications, new admission to nursing home, mortality, use of antipsychotic medications during hospital stay and cognitive and physical function at 6 months postdischarge. ETHICS AND DISSEMINATION This trial has been approved by the South Eastern Sydney Human Research and Ethics Committee. Dissemination plans include submission to a peer-reviewed journal for publication and presentation at scientific conferences. TRIAL REGISTRATION NUMBER ACTRN12618000318280.
Collapse
Affiliation(s)
- Anita Nitchingham
- Department of Geriatric Medicine, Prince of Wales Hospital, Sydney, New South Wales, Australia
- Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Andrew Milne
- Rural Clinical School, Coffs Harbour Health Campus, University of New South Wales, Coffs Harbour, New South Wales, Australia
| | - Barbara Toson
- Flinders Centre for Epidemiology and Biostatistics, Flinders University, Adelaide, South Australia, Australia
| | - Bernard Tuch
- Department of Molecular & Translational Science, Hudson Institute, Monash University, Melbourne, Victoria, Australia
| | - Meera Agar
- Faculty of Health, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Jacqueline Close
- Department of Geriatric Medicine, Prince of Wales Hospital, Sydney, New South Wales, Australia
- Falls, Balance and Injury Research Centre, Neuroscience Research Australia, Randwick, New South Wales, Australia
| | - Gideon Caplan
- Department of Geriatric Medicine, Prince of Wales Hospital, Sydney, New South Wales, Australia
- Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
198
|
Abstract
Tumour necrosis factor (TNF) is a classical, pleiotropic pro-inflammatory cytokine. It is also the first 'adipokine' described to be produced from adipose tissue, regulated in obesity and proposed to contribute to obesity-associated metabolic disease. In this review, we provide an overview of TNF in the context of metabolic inflammation or metaflammation, its discovery as a metabolic messenger, its sites and mechanisms of action and some critical considerations for future research. Although we focus on TNF and the studies that elucidated its immunometabolic actions, we highlight a conceptual framework, generated by these studies, that is equally applicable to the complex network of pro-inflammatory signals, their biological activity and their integration with metabolic regulation, and to the field of immunometabolism more broadly.
Collapse
Affiliation(s)
- Jaswinder K Sethi
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK.
- National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service (NHS) Foundation Trust, Southampton, UK.
- Institute for Life Sciences, University of Southampton, Southampton, UK.
| | - Gökhan S Hotamisligil
- Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Harvard-MIT Broad Institute, Boston, MA, USA.
- Harvard Stem Cell Institute, Boston, MA, USA.
- The Joslin Diabetes Center, Boston, MA, USA.
| |
Collapse
|
199
|
Luna R, Talanki Manjunatha R, Bollu B, Jhaveri S, Avanthika C, Reddy N, Saha T, Gandhi F. A Comprehensive Review of Neuronal Changes in Diabetics. Cureus 2021; 13:e19142. [PMID: 34868777 PMCID: PMC8628358 DOI: 10.7759/cureus.19142] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2021] [Indexed: 12/11/2022] Open
Abstract
There has been an exponential rise in diabetes mellitus (DM) cases on a global scale. Diabetes affects almost every system of the body, and the nervous system is no exception. Although the brain is dependent on glucose, providing it with the energy required for optimal functionality, glucose also plays a key role in the regulation of oxidative stress, cell death, among others, which furthermore contribute to the pathophysiology of neurological disorders. The variety of biochemical processes engaged in this process is only matched by the multitude of clinical consequences resulting from it. The wide-ranging effects on the central and peripheral nervous system include, but are not limited to axonopathies, neurodegenerative diseases, neurovascular diseases, and general cognitive impairment. All language search was conducted on MEDLINE, COCHRANE, EMBASE, and GOOGLE SCHOLAR till September 2021. The following search strings and Medical Subject Headings (MeSH terms) were used: "Diabetes Mellitus," "CNS," "Diabetic Neuropathy," and "Insulin." We explored the literature on diabetic neuropathy, covering its epidemiology, pathophysiology with the respective molecular pathways, clinical consequences with a special focus on the central nervous system and finally, measures to prevent and treat neuronal changes. Diabetes is slowly becoming an epidemic, rapidly increasing the clinical burden on account of its wide-ranging complications. This review focuses on the neuronal changes occurring in diabetes such as the impact of hyperglycemia on brain function and structure, its association with various neurological disorders, and a few diabetes-induced peripheral neuropathic changes. It is an attempt to summarize the relevant literature about neuronal consequences of DM as treatment options available today are mostly focused on achieving better glycemic control; further research on novel treatment options to prevent or delay the progression of neuronal changes is still needed.
Collapse
Affiliation(s)
- Rudy Luna
- Neurofisiología, Instituto Nacional de Neurologia y Neurocirugia, CDMX, MEX
| | | | | | | | - Chaithanya Avanthika
- Medicine and Surgery; Pediatrics, Karnataka Institute of Medical Sciences, Hubli, IND
| | - Nikhil Reddy
- Internal Medicine, Kamineni Academy of Medical Science and Research Centre, Hyderabad, IND
| | - Tias Saha
- Internal Medicine, Diabetic Association Medical College, Faridpur, BGD
| | - Fenil Gandhi
- Medicine, Shree Krishna Hospital, Anand, IND
- Research Project Associate, Memorial Sloan Kettering Cancer Center, New York, USA
| |
Collapse
|
200
|
Hiding in Plain Sight: Modern Thiamine Deficiency. Cells 2021; 10:cells10102595. [PMID: 34685573 PMCID: PMC8533683 DOI: 10.3390/cells10102595] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022] Open
Abstract
Thiamine or vitamin B1 is an essential, water-soluble vitamin required for mitochondrial energetics—the production of adenosine triphosphate (ATP). It is a critical and rate-limiting cofactor to multiple enzymes involved in this process, including those at the entry points and at critical junctures for the glucose, fatty acid, and amino acid pathways. It has a very short half-life, limited storage capacity, and is susceptible to degradation and depletion by a number of products that epitomize modern life, including environmental and pharmaceutical chemicals. The RDA for thiamine is 1.1–1.2 mg for adult females and males, respectively. With an average diet, even a poor one, it is not difficult to meet that daily requirement, and yet, measurable thiamine deficiency has been observed across multiple patient populations with incidence rates ranging from 20% to over 90% depending upon the study. This suggests that the RDA requirement may be insufficient to meet the demands of modern living. Inasmuch as thiamine deficiency syndromes pose great risk of chronic morbidity, and if left untreated, mortality, a more comprehensive understanding thiamine chemistry, relative to energy production, modern living, and disease, may prove useful.
Collapse
|