1
|
Ball BK, Park JH, Bergendorf AM, Proctor EA, Brubaker DK. Translational disease modeling of peripheral blood identifies type 2 diabetes biomarkers predictive of Alzheimer's disease. NPJ Syst Biol Appl 2025; 11:58. [PMID: 40442087 PMCID: PMC12122922 DOI: 10.1038/s41540-025-00539-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 05/16/2025] [Indexed: 06/02/2025] Open
Abstract
Type 2 diabetes (T2D) is a significant risk factor for Alzheimer's disease (AD). Despite multiple studies reporting this connection, the mechanism by which T2D exacerbates AD is poorly understood. It is challenging to design studies that address co-occurring and comorbid diseases, limiting the number of existing evidence bases. To address this challenge, we expanded the applications of a computational framework called Translatable Components Regression (TransComp-R), initially designed for cross-species translation modeling, to perform cross-disease modeling to identify biological programs of T2D that may exacerbate AD pathology. Using TransComp-R, we combined peripheral blood-derived T2D and AD human transcriptomic data to identify T2D principal components predictive of AD status. Our model revealed genes enriched for biological pathways associated with inflammation, metabolism, and signaling pathways from T2D principal components predictive of AD. The same T2D PC predictive of AD outcomes unveiled sex-based differences across the AD datasets. We performed a gene expression correlational analysis to identify therapeutic hypotheses tailored to the T2D-AD axis. We identified six T2D and two dementia medications that induced gene expression profiles associated with a non-T2D or non-AD state. We next assessed our blood-based T2DxAD biomarker signature in post-mortem human AD and control brain gene expression data from the hippocampus, entorhinal cortex, superior frontal gyrus, and postcentral gyrus. Using partial least squares discriminant analysis, we identified a subset of genes from our cross-disease blood-based biomarker panel that significantly separated AD and control brain samples. Finally, we validated our findings using single cell RNA-sequencing blood data of AD and healthy individuals and found erythroid cells contained the most gene expression signatures to the T2D PC. Our methodological advance in cross-disease modeling identified biological programs in T2D that may predict the future onset of AD in this population. This, paired with our therapeutic gene expression correlational analysis, also revealed alogliptin, a T2D medication that may help prevent the onset of AD in T2D patients.
Collapse
Affiliation(s)
- Brendan K Ball
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Jee Hyun Park
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Alexander M Bergendorf
- Center for Global Health & Diseases, Department of Pathology, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Elizabeth A Proctor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Penn State University, State College, PA, USA
- Penn State Neuroscience Institute, Penn State University, State College, PA, USA
- Department of Engineering Science & Mechanics, Penn State University, State College, PA, USA
| | - Douglas K Brubaker
- Center for Global Health & Diseases, Department of Pathology, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
- Blood Heart Lung Immunology Research Center, University Hospitals, Cleveland, OH, USA.
| |
Collapse
|
2
|
Yang Q, Huang D, Zhang Z, Gao H, Wu J, Zhong H, Guo X, Wang Y, Zhou H, Liu C, Duan X. Diabetes affects AD through plasma Aβ40: A Mendelian randomization study. Neuroscience 2025; 575:131-139. [PMID: 40233921 DOI: 10.1016/j.neuroscience.2025.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/05/2025] [Accepted: 04/11/2025] [Indexed: 04/17/2025]
Abstract
Amyloid and tau proteins are important proteins in the pathological changes of Alzheimer's disease (AD), while Aβ pathology and tau pathology are the most critical factors contributing to the development of AD. Some studies have shown that there is a causal relationship between AD and diabetes mellitus, but there are no studies showing a causal relationship between diabetic traits and AD biomarkers, so further exploration is needed. We first summarized and analyzed the currently published literature on the link between diabetes and AD through a systematic review. Forest plots were used to observe whether there is an association between diabetes and AD. Then a two-sample Mendelian randomization (MR) analysis based on GWAS summary statistics was performed to verify the causal relationship between diabetic traits and AD biomarkers. Based on summary statistics from the GWAS, potential causal relationships between diabetic traits and AD biomarkers were explored separately. The results of the meta-analysis part showed that diabetes can increase the risk of AD. Meanwhile, our two-sample MR results showed a significant causal relationship between diabetes and plasma Aβ40. In addition, our two-sample MR results also showed a causal relationship between increased HbA1c and plasma APLP2. Other diabetic traits may have potential effects on different AD plasma markers.
Collapse
Affiliation(s)
- Qiumin Yang
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Delong Huang
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Zhaojing Zhang
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Haiyan Gao
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Junhao Wu
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Haoshu Zhong
- Department of Hematology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaowei Guo
- School of Life Sciences, Southwest University, Chongqing, China
| | - Yiren Wang
- School of Nursing, Southwest Medical University, Luzhou, Sichuan, China
| | - Hemu Zhou
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chengzhen Liu
- Faculty of Psychology, Southwest University, Chongqing, China
| | - Xiaodong Duan
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
3
|
Manandhar S, Gurram PC, Govindula A, Kabekkodu SP, Pai KSR. Voglibose Attenuates Amyloid Beta-Induced Memory Deficits in a Rodent Model: A Potential Alzheimer's Therapy via Wnt Signaling Modulation. Mol Neurobiol 2025:10.1007/s12035-025-05047-5. [PMID: 40381169 DOI: 10.1007/s12035-025-05047-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 05/06/2025] [Indexed: 05/19/2025]
Abstract
Disruption of the Wnt signaling pathway (WSP), a highly conserved pathway essential for growth and organ development, has been proven to play a role in the pathogenesis of Alzheimer's disease (AD). This study focused on repurposing the FDA-approved drug, Voglibose to target the DKK1-LRP6 site with the goal of upregulating WSP in in vitro as well as rodent model of AD. Based on our previous computational approach, Voglibose was evaluated for the DKK1 binding, neuroprotective effects were examined using SHSY5Y cells, while WSP activation was analyzed through RTPCR in the HEK293/LRP6 cell line. Rodent model of AD was developed using intracerebroventricular administration of Aβ25-35. Male Wistar rats were randomly assigned to receive oral doses of Voglibose (1 and 10 mg/kg) for 28 days, after which behavioral assessments, biochemical analyses, RT-PCR, and histopathological evaluations were conducted. Voglibose showed significant reduction in the DKK1 binding, neuroprotective property in SHSY5Y as well as activation of WSP in LRP6 overexpressed HEK293 cells. There was a significant decrease in the island latency in rats treated with lower dose (p < 0.01) and higher dose (p < 0.05) of Voglibose when compared to the disease control rats. Similarly, in the behavioral tests, Voglibose significantly improved cognition. The deposition of amyloid plaques was found to be considerably more in the disease control rats which got reduced in the treatment groups as observed in the histopathological slides stained with Congo red. Significant alterations in mRNA levels and protein expression of glycogen synthase kinase-β (GSK-3β), β-catenin (β-cat) was observed in rat brain homogenates indicating upregulation of WSP. In conclusion, Voglibose demonstrated significant neuroprotective potential in a cell line study and showed potential cognitive benefits in a rat model of AD. Furthermore, its ability to activate WSP highlights its immense potential as AD therapeutic to enhance memory and modulate key neuroprotective mechanisms.
Collapse
Affiliation(s)
- Suman Manandhar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Prasada Chowdari Gurram
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Anusha Govindula
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - K Sreedhara Ranganath Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
4
|
Siddiqui F, Mishra P, Khanam S, Ranjan S, Alam P, Albalawi T, Khan S, Mir SS. Nano-Chaperones: Bridging Therapeutics for Amyloid Aggregation in Alzheimer's Disease and Type-2 Diabetes Mellitus. Eur J Neurosci 2025; 61:e70142. [PMID: 40384055 DOI: 10.1111/ejn.70142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 04/12/2025] [Accepted: 05/01/2025] [Indexed: 05/20/2025]
Abstract
Nano-chaperones represent an innovative therapeutic approach targeting amyloid aggregation in Alzheimer's disease (AD) and Type-2 diabetes mellitus (T2DM), two diseases linked by similar pathogenic mechanisms involving protein misfolding and insulin resistance. Current treatments primarily address symptoms, yet nano-chaperones can potentially intervene at the molecular level by mimicking natural chaperone proteins to prevent or reverse amyloid aggregation. In AD, nano-chaperones target amyloid-beta (Aβ) peptides, reducing neurotoxicity and preserving neuronal function, while in T2DM, they inhibit islet amyloid polypeptide (IAPP) aggregation, alleviating cytotoxic stress on pancreatic β-cells. These nanoparticles exhibit a dual capacity for cellular penetration and selectivity in interacting with misfolded proteins, showing promise in mitigating the shared amyloidogenic pathways of both diseases. Preclinical studies have demonstrated significant reductions in amyloid toxicity with potential applications in crossing the blood-brain barrier (BBB) to enhance central nervous system (CNS) delivery. Nano-chaperones transformative role in developing multi-targeted precision therapies for complex diseases is highlighted, underscoring their capacity to modulate disease progression through targeted biomimetic interactions. Nano-chaperone designs for clinical application focus on enhancing therapeutic efficacy and safety. This innovative approach may redefine treatment paradigms for amyloid-related diseases, offering a new frontier in personalized medicine.
Collapse
Affiliation(s)
- Faiza Siddiqui
- Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, India
| | - Pooja Mishra
- Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, India
| | - Sheeba Khanam
- Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, India
| | - Sachin Ranjan
- Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, India
| | - Pravej Alam
- Department of Biology, College of Science and Humanities, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Thamer Albalawi
- Department of Biology, College of Science and Humanities, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Salman Khan
- Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, India
| | - Snober S Mir
- Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
5
|
Alves SS, Servilha-Menezes G, Rossi L, de Oliveira JAC, Grigorio-de-Sant'Ana M, Sebollela A, da Silva-Junior RMP, Garcia-Cairasco N. Insulin signaling disruption exacerbates memory impairment and seizure susceptibility in an epilepsy model with Alzheimer's disease-like pathology. J Neural Transm (Vienna) 2025:10.1007/s00702-025-02896-1. [PMID: 39987343 DOI: 10.1007/s00702-025-02896-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 02/10/2025] [Indexed: 02/24/2025]
Abstract
Alzheimer's disease (AD) and epilepsy exhibit a complex bidirectional relationship. Curiously, diabetes as a comorbidity increases the risk of epilepsy among AD patients. Recently, we reported that the Wistar audiogenic rat (WAR) strain, a genetic model of epilepsy, displays a partial AD-like phenotype, including brain insulin resistance. We also assessed seizure susceptibility in an AD model created through intracerebroventricular injections of streptozotocin (icv-STZ), which induces AD features via brain insulin resistance. Our goal was to explore how disrupted brain insulin signaling influences AD-like features and seizure susceptibility in the WAR strain. Adult male WARs received a single intracerebroventricular injection of streptozotocin (icv-STZ) (1.5 mg/kg) or vehicle (saline). Two weeks post-injection, spatial memory was assessed using the Barnes Maze (BM) test. Three weeks later, the rats underwent an audiogenic kindling (AuK) protocol (20 acoustic stimuli, 2 per day) to evaluate seizure frequency and severity. Seizures were analyzed using the Categorized Severity Index and Racine's scale and Western blot analysis was performed on hippocampal tissue. Our findings revealed that icv-STZ significantly worsened memory performance, increased seizure frequency, and reduced seizure onset relative to vehicle. Furthermore, icv-STZ decreased Akt activation and increased Glycogen Synthase Kinase-3 (GSK3) phosphorylation, indicating disrupted insulin signaling. Notably, icv-STZ decreased tau phosphorylation without altering amyloid β precursor protein (AβPP) levels. In conclusion, a low-dose icv-STZ injection exacerbates memory deficits and seizure susceptibility in the WAR strain by disturbing downstream proteins involved in insulin signaling. This highlights the implications of brain insulin resistance in both AD and epilepsy.
Collapse
Affiliation(s)
- Suélen Santos Alves
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil
| | - Gabriel Servilha-Menezes
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Av. Dos Bandeirantes 3900, Ribeirão Preto, SP, 14049-900, Brazil
| | - Letícia Rossi
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Av. Dos Bandeirantes 3900, Ribeirão Preto, SP, 14049-900, Brazil
| | - José Antonio Cortes de Oliveira
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Av. Dos Bandeirantes 3900, Ribeirão Preto, SP, 14049-900, Brazil
| | - Mariana Grigorio-de-Sant'Ana
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Av. Dos Bandeirantes 3900, Ribeirão Preto, SP, 14049-900, Brazil
| | - Adriano Sebollela
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil
| | | | - Norberto Garcia-Cairasco
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil.
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Av. Dos Bandeirantes 3900, Ribeirão Preto, SP, 14049-900, Brazil.
| |
Collapse
|
6
|
Tel AZ, Aslan K, Yılmaz MA, Gulcin İ. A multidimensional study for design of phytochemical profiling, antioxidant potential, and enzyme inhibition effects of ışgın ( Rheum telianum) as an edible plant. Food Chem X 2025; 25:102125. [PMID: 39974533 PMCID: PMC11838142 DOI: 10.1016/j.fochx.2024.102125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 02/21/2025] Open
Abstract
The present study reveals in vitro antioxidant properties and the phytochemical content of a novel Rheum species (Rheum telianum), which grows in Southeastern Anatolia. To perform the analysis, dried leaves and seeds of the plants were ground and extracted with ethanol to obtain plant secondary metabolites and antioxidant activity. Then, dried extracts were subjected to in vitro DPPH scavenging and Cupric reducing (CUPRAC), Fe3+, and Ferric reducing antioxidant power (FRAP). In addition to the antioxidant capacity assays, quantitative phenolic, flavonoid, and secondary metabolite were determined through spectrophotometric and LC-MS/MS chromatographic methods. IC50 values showed that both leaves and the seeds of the R. telianum have high inhibitory properties over DPPH radicals with 20.79 and 5.67 μg/mL, respectively. The samples' dominant secondary metabolites were evaluated through the LC-MS/MS analysis results. The inhibition effects of both leaves and the seeds of the R. telianum extracts on acetylcholinesterase and butyrylcholinesterase, α-glycosidase and human carbonic anhydrases II isoenzyme enzymes, which associated with some global diseases including Alzheimer's disease, type-2 diabetes mellitus and glaucoma were determined. In conclusion, the extracts' contents and functional relationship and the plants' possible usage in the food, medicine, and cosmetic industries was revealed.
Collapse
Affiliation(s)
- Ahmet Zafer Tel
- Department of Agricultural Biotechnology, Faculty of Agriculture, Igdir University, 76000 Igdir, Türkiye
| | - Kubra Aslan
- Department of Chemistry, Faculty of Science, Ataturk University, 25240 Erzurum, Türkiye
| | - Mustafa Abdullah Yılmaz
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Dicle University, 21280 Diyarbakır, Türkiye
| | - İlhami Gulcin
- Department of Chemistry, Faculty of Science, Ataturk University, 25240 Erzurum, Türkiye
| |
Collapse
|
7
|
Ball BK, Hyun Park J, Proctor EA, Brubaker DK. Cross-disease modeling of peripheral blood identifies biomarkers of type 2 diabetes predictive of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.11.627991. [PMID: 39713369 PMCID: PMC11661382 DOI: 10.1101/2024.12.11.627991] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Type 2 diabetes (T2D) is a significant risk factor for Alzheimer's disease (AD). Despite multiple studies reporting this connection, the mechanism by which T2D exacerbates AD is poorly understood. It is challenging to design studies that address co-occurring and comorbid diseases, limiting the number of existing evidence bases. To address this challenge, we expanded the applications of a computational framework called Translatable Components Regression (TransComp-R), initially designed for cross-species translation modeling, to perform cross-disease modeling to identify biological programs of T2D that may exacerbate AD pathology. Using TransComp-R, we combined peripheral blood-derived T2D and AD human transcriptomic data to identify T2D principal components predictive of AD status. Our model revealed genes enriched for biological pathways associated with inflammation, metabolism, and signaling pathways from T2D principal components predictive of AD. The same T2D PC predictive of AD outcomes unveiled sex-based differences across the AD datasets. We performed a gene expression correlational analysis to identify therapeutic hypotheses tailored to the T2D-AD axis. We identified six T2D and two dementia medications that induced gene expression profiles associated with a non-T2D or non-AD state. Finally, we assessed our blood-based T2DxAD biomarker signature in post-mortem human AD and control brain gene expression data from the hippocampus, entorhinal cortex, superior frontal gyrus, and postcentral gyrus. Using partial least squares discriminant analysis, we identified a subset of genes from our cross-disease blood-based biomarker panel that significantly separated AD and control brain samples. Our methodological advance in cross-disease modeling identified biological programs in T2D that may predict the future onset of AD in this population. This, paired with our therapeutic gene expression correlational analysis, also revealed alogliptin, a T2D medication that may help prevent the onset of AD in T2D patients.
Collapse
Affiliation(s)
- Brendan K. Ball
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Jee Hyun Park
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Elizabeth A. Proctor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Penn State University, State College, PA, USA
- Center for Neural Engineering, Penn State University, State College, PA, USA
- Department of Engineering Science & Mechanics, Penn State University, State College, PA, USA
| | - Douglas K. Brubaker
- Center for Global Health & Diseases, Department of Pathology, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Blood Heart Lung Immunology Research Center, University Hospitals, Cleveland, OH, USA
| |
Collapse
|
8
|
Wang W, Zhang J, Zhang M, Zhang C, Liu H, Li W, Fan Y. Impact of diabetes mellitus on the risk of Alzheimer's disease: a mendelian randomization study. BMC Neurol 2024; 24:448. [PMID: 39558225 PMCID: PMC11571773 DOI: 10.1186/s12883-024-03955-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND The impact of diabetes on the risk of Alzheimer's disease remains uncertain. This study aimed to explore this issue from multiple perspectives by using the Mendelian randomization (MR) approach. METHODS Instrumental variables for predicting six diabetic traits (including insulin and blood glucose), eight metabolic risk factors for diabetes (including total cholesterol and blood pressure), and seven diabetic genes were extracted from their summary data. These data were derived from multiple European cohorts and included 31,684 to 810,865 subjects respectively. The two-sample MR, multivariate MR, and summary-data-based Mendelian randomization (SMR) methods were employed to determine the associations of these traits or genes with the risk of Alzheimer's disease. RESULTS The two-sample MR showed that elevated fasting insulin and total cholesterol levels were associated with an increased risk of dementia in Alzheimer's disease (P = 0.022, P = 0.041). Elevated systolic and diastolic blood pressure levels were associated with a decreased risk of dementia in Alzheimer's disease (P = 0.036, P = 0.025). The multivariate MR reported that adjusting for telomere length (a well-established biomarker of aging) did not change these findings (P < 0.05). Additionally, the two-sample MR showed that type 1 and type 2 diabetes did not affect the risk of Alzheimer's disease. The SMR also indicated that the diabetic genes did not affect the risk of this disease. CONCLUSION Multiple MR approaches concluded that fasting insulin, total cholesterol, and blood pressure, rather than diabetes, were potential metabolic variables that had an impact on the risk of Alzheimer's disease. However, aging might not be involved in these correlations.
Collapse
Affiliation(s)
- Weichao Wang
- Department of Endocrinology, Shijiazhuang People's Hospital, 365, Jianhua South Street, Yuhua District, Shijiazhuang, Hebei Province, 050011, China.
| | - Jie Zhang
- Department of Ophthalmology, Shijiazhuang Third Hospital, Shijiazhuang, Hebei Province, 050011, China
| | - Man Zhang
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei Province, 050011, China
| | - Chengyuan Zhang
- Examination Division, Medical Skills Examination and Appraisal Center, Shijiazhuang, Hebei Province, 050011, China
| | - Huanli Liu
- Teaching Division, Shijiazhuang People's Hospital, Shijiazhuang, Hebei Province, 050011, China
| | - Wanlin Li
- Teaching Division, Shijiazhuang People's Hospital, Shijiazhuang, Hebei Province, 050011, China
| | - Yimeng Fan
- Teaching Division, Shijiazhuang People's Hospital, Shijiazhuang, Hebei Province, 050011, China
| |
Collapse
|
9
|
Pujol A, Sanchis P, Tamayo MI, Godoy S, Andrés P, Speranskaya A, Espino A, Estremera A, Rigo E, Amengual GJ, Rodríguez M, Ribes JL, Gomila I, Grases F, González-Freire M, Masmiquel L. Association of Low Protein-to-Carbohydrate Energy Ratio with Cognitive Impairment in Elderly Type 2 Diabetes Patients. Nutrients 2024; 16:3888. [PMID: 39599676 PMCID: PMC11597317 DOI: 10.3390/nu16223888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND/OBJECTIVES The relationship between macronutrient intake and cognitive decline in older adults with type 2 diabetes mellitus (T2DM) remains underexplored. METHODS This cross-sectional study aimed to evaluate the association between the protein-to-carbohydrate energy ratio (%E:P) and cognitive impairment among 192 elderly T2DM patients. Cognitive function was assessed using the Montreal Cognitive Assessment (MoCA) and the Self-Administered Gerocognitive Exam (SAGE), while dietary intake data, including (%E:P), was gathered using a validated semi-quantitative food frequency questionnaire. RESULTS Participants had a mean age of 71 ± 6 years, 46.4% were female, and the median BMI was 30 ± 4 kg/m2. After adjusting for confounding variables, patients in the highest (%E:P) tertile showed significantly higher MoCA and SAGE scores compared to those in the lowest tertile (p < 0.005). We identified an optimal (%E:P) threshold of 0.375 for predicting cognitive impairment, with a sensitivity of 53% and specificity of 64%. CONCLUSIONS These findings suggest that a lower (%E:P) ratio may be a risk factor for cognitive impairment in elderly T2DM patients. Monitoring this ratio may serve as an early detection tool for cognitive deterioration. Moreover, current protein intake recommendations for older adults with T2DM may be insufficient to prevent cognitive impairment. Further research is needed to establish optimal dietary guidelines for this population.
Collapse
Affiliation(s)
- Antelm Pujol
- Vascular and Metabolic Diseases Research Group, Endocrinology Department, Son Llàtzer University Hospital, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (A.P.)
| | - Pilar Sanchis
- Vascular and Metabolic Diseases Research Group, Endocrinology Department, Son Llàtzer University Hospital, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (A.P.)
- Laboratory of Renal Lithiasis Research, Department of Chemistry, University of Balearic Islands, Research Institute of Heath Science (IUNICS), Health Research Institute of Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María I. Tamayo
- Vascular and Metabolic Diseases Research Group, Endocrinology Department, Son Llàtzer University Hospital, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (A.P.)
| | - Samantha Godoy
- Laboratory of Renal Lithiasis Research, Department of Chemistry, University of Balearic Islands, Research Institute of Heath Science (IUNICS), Health Research Institute of Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
| | - Pilar Andrés
- Neuropsychology and Cognition, Department of Psychology, Research Institute of Heath Science (IUNICS), University of Balearic Islands, Health Research Institute of Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
| | - Aleksandra Speranskaya
- Vascular and Metabolic Diseases Research Group, Endocrinology Department, Son Llàtzer University Hospital, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (A.P.)
| | - Ana Espino
- Neurology Department, Son Llàtzer University Hospital, 07198 Palma de Mallorca, Spain
| | - Ana Estremera
- Neuroradiology Department, Son Llàtzer University Hospital, 07198 Palma de Mallorca, Spain (M.R.)
| | - Elena Rigo
- Balearic Research Group on Genetic Cardiopathies, Sudden Death, and TTR Amyloidosis, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
- Neuroopthalmology Department, Son Llàtzer University Hospital, 07198 Palma de Mallorca, Spain
| | - Guillermo J. Amengual
- Neuroradiology Department, Son Llàtzer University Hospital, 07198 Palma de Mallorca, Spain (M.R.)
| | - Manuel Rodríguez
- Neuroradiology Department, Son Llàtzer University Hospital, 07198 Palma de Mallorca, Spain (M.R.)
| | - José Luis Ribes
- Clinical Analysis Department, Son Llàtzer University Hospital, 07198 Palma de Mallorca, Spain; (J.L.R.)
| | - Isabel Gomila
- Clinical Analysis Department, Son Llàtzer University Hospital, 07198 Palma de Mallorca, Spain; (J.L.R.)
- Clinical Toxicology Research Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
| | - Félix Grases
- Laboratory of Renal Lithiasis Research, Department of Chemistry, University of Balearic Islands, Research Institute of Heath Science (IUNICS), Health Research Institute of Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Marta González-Freire
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria (UFV), 28223 Madrid, Spain
| | - Lluís Masmiquel
- Vascular and Metabolic Diseases Research Group, Endocrinology Department, Son Llàtzer University Hospital, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (A.P.)
| |
Collapse
|
10
|
Seferi G, Mjønes HS, Havik M, Reiersen H, Dalen KT, Nordengen K, Morland C. Distribution of lipid droplets in hippocampal neurons and microglia: impact of diabetes and exercise. Life Sci Alliance 2024; 7:e202302239. [PMID: 39117458 PMCID: PMC11310565 DOI: 10.26508/lsa.202302239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
Neuroinflammation, aging, and neurodegenerative disorders are associated with excessive accumulation of neutral lipids in lipid droplets (LDs) in microglia. Type 2 diabetes mellitus (T2DM) may cause neuroinflammation and is a risk factor for neurodegenerative disorders. Here, we show that hippocampal pyramidal neurons contain smaller, more abundant LDs than their neighboring microglia. The density of LDs varied between pyramidal cells in adjacent subregions, with CA3 neurons containing more LDs than CA1 neurons. Within the CA3 region, a gradual increase in the LD content along the pyramidal layer from the hilus toward CA2 was observed. Interestingly, the high neuronal LD content correlated with less ramified microglial morphotypes. Using the db/db model of T2DM, we demonstrated that diabetes increased the number of LDs per microglial cell without affecting the neuronal LD density. High-intensity interval exercise induced smaller changes in the number of LDs in microglia but was not sufficient to counteract the diabetes-induced changes in LD accumulation. The changes observed in response to T2DM may contribute to the cerebral effects of T2DM and provide a mechanistic link between T2DM and neurodegenerative disorders.
Collapse
Affiliation(s)
- Gezime Seferi
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Harald S Mjønes
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Mona Havik
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Herman Reiersen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Knut Tomas Dalen
- Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kaja Nordengen
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Cecilie Morland
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
11
|
Hierro-Bujalance C, Garcia-Alloza M. Empagliflozin reduces brain pathology in Alzheimer's disease and type 2 diabetes. Neural Regen Res 2024; 19:1189-1190. [PMID: 37905858 PMCID: PMC11467955 DOI: 10.4103/1673-5374.385865] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/20/2023] [Accepted: 08/20/2023] [Indexed: 11/02/2023] Open
Affiliation(s)
- Carmen Hierro-Bujalance
- Division of Physiology, School of Medicine, Universidad de Cadiz, Cadiz, Spain
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain
- Salus Infirmorum-Universidad de Cadiz, Cadiz, Spain
| | - Monica Garcia-Alloza
- Division of Physiology, School of Medicine, Universidad de Cadiz, Cadiz, Spain
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain
| |
Collapse
|
12
|
Pujol A, Sanchis P, Tamayo MI, Nicolau J, Grases F, Espino A, Estremera A, Rigo E, Amengual GJ, Rodríguez M, Ribes JL, Gomila I, Simó-Servat O, Masmiquel L. Oral phytate supplementation on the progression of mild cognitive impairment, brain iron deposition and diabetic retinopathy in patients with type 2 diabetes: a concept paper for a randomized double blind placebo controlled trial (the PHYND trial). Front Endocrinol (Lausanne) 2024; 15:1332237. [PMID: 38872972 PMCID: PMC11169791 DOI: 10.3389/fendo.2024.1332237] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 05/14/2024] [Indexed: 06/15/2024] Open
Abstract
Type 2 diabetes mellitus has a worldwide prevalence of 10.5% in the adult population (20-79 years), and by 2045, the prevalence is expected to keep rising to one in eight adults living with diabetes. Mild cognitive impairment has a global prevalence of 19.7% in adults aged 50 years. Both conditions have shown a concerning increase in prevalence rates over the past 10 years, highlighting a growing public health challenge. Future forecasts indicate that the prevalence of dementia (no estimations done for individuals with mild cognitive impairment) is expected to nearly triple by 2050. Type 2 diabetes mellitus is a risk factor for the development of cognitive impairment, and such impairment increase the likelihood of poor glycemic/metabolic control. High phytate intake has been shown to be a protective factor against the development of cognitive impairment in observational studies. Diary phytate intake might reduce the micro- and macrovascular complications of patients with type 2 diabetes mellitus through different mechanisms. We describe the protocol of the first trial (the PHYND trial) that evaluate the effect of daily phytate supplementation over 56 weeks with a two-arm double-blind placebo-controlled study on the progression of mild cognitive impairment, cerebral iron deposition, and retinal involvement in patients with type 2 diabetes mellitus. Our hypothesis proposes that phytate, by inhibiting advanced glycation end product formation and chelating transition metals, will improve cognitive function and attenuate the progression from Mild Cognitive Impairment to dementia in individuals with type 2 diabetes mellitus and mild cognitive impairment. Additionally, we predict that phytate will reduce iron accumulation in the central nervous system, mitigate neurodegenerative changes in both the central nervous system and retina, and induce alterations in biochemical markers associated with neurodegeneration.
Collapse
Affiliation(s)
- Antelm Pujol
- Vascular and Metabolic Diseases Research Group, Endocrinology Department, Son Llàtzer University Hospital, Health Research Institute of the Balearic Islands (IdISBa), Palma de Mallorca, Spain
| | - Pilar Sanchis
- Vascular and Metabolic Diseases Research Group, Endocrinology Department, Son Llàtzer University Hospital, Health Research Institute of the Balearic Islands (IdISBa), Palma de Mallorca, Spain
- Laboratory of Renal Lithiasis Research, University of Balearic Islands, Research Institute of Health Science (IUNICS) Health Research Institute of Balearic Islands, (IdISBa), Palma de Mallorca, Spain
| | - María I. Tamayo
- Vascular and Metabolic Diseases Research Group, Endocrinology Department, Son Llàtzer University Hospital, Health Research Institute of the Balearic Islands (IdISBa), Palma de Mallorca, Spain
| | - Joana Nicolau
- Vascular and Metabolic Diseases Research Group, Endocrinology Department, Son Llàtzer University Hospital, Health Research Institute of the Balearic Islands (IdISBa), Palma de Mallorca, Spain
| | - Félix Grases
- Laboratory of Renal Lithiasis Research, University of Balearic Islands, Research Institute of Health Science (IUNICS) Health Research Institute of Balearic Islands, (IdISBa), Palma de Mallorca, Spain
| | - Ana Espino
- Neurology Department, Son Llàtzer University Hospital, Palma de Mallorca, Spain
| | - Ana Estremera
- Neuroradiology Unit, Son Llàtzer University Hospital, Palma de Mallorca, Spain
| | - Elena Rigo
- Neuroopthalmology Unit, Son Llàtzer University Hospital, Palma de Mallorca, Spain
| | | | - Manuel Rodríguez
- Neuroradiology Unit, Son Llàtzer University Hospital, Palma de Mallorca, Spain
| | - José L. Ribes
- Biochemistry Department, Son Llàtzer University Hospital, Palma de Mallorca, Spain
| | - Isabel Gomila
- Biochemistry Department, Son Llàtzer University Hospital, Palma de Mallorca, Spain
| | - Olga Simó-Servat
- Diabetes and Metabolism Research Unit, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Lluís Masmiquel
- Vascular and Metabolic Diseases Research Group, Endocrinology Department, Son Llàtzer University Hospital, Health Research Institute of the Balearic Islands (IdISBa), Palma de Mallorca, Spain
| |
Collapse
|
13
|
Liu S, Chen H, He XD, Yang XO. Glucometabolic-Related Genes as Diagnostic Biomarkers and Therapeutic Targets for Alzheimer's Disease and Type 2 Diabetes Mellitus: A Bioinformatics Analysis. Neurol Res Int 2024; 2024:5200222. [PMID: 38595695 PMCID: PMC11003797 DOI: 10.1155/2024/5200222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 01/26/2024] [Accepted: 02/24/2024] [Indexed: 04/11/2024] Open
Abstract
Background Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM) are two widespread chronic disorders characterized by shared risk factors and molecular pathways. Glucose metabolism, pivotal for cellular homeostasis and energy supply, plays a critical role in these diseases. Its disturbance has been linked to the pathogenesis of both AD and T2DM. However, a comprehensive investigation into the specific roles of glucometabolic genes in the onset and progression of AD and T2DM has yet to be conducted. Methods By analyzing microarray datasets from the Gene Expression Omnibus (GEO) repository, we identified differentially expressed glucometabolic genes (DEGs) in AD and T2DM cohorts. A range of bioinformatics tools were employed for functional annotation, pathway enrichment, protein interaction network construction, module analysis, ROC curve assessment, correlation matrix construction, gene set enrichment analysis, and gene-drug interaction mapping of these DEGs. Key genes were further validated using quantitative real-time polymerase chain reaction (qRT-PCR) in AD and T2DM murine models. Results Our investigation identified 41 glucometabolic-related DEGs, with six prominent genes (G6PD, PKM, ENO3, PFKL, PGD, and TALDO1) being common in both AD and T2DM cohorts. These genes play crucial roles in metabolic pathways including glycolysis, pentose phosphate pathway, and amino sugar metabolism. Their diagnostic potential was highlighted by area under curve (AUC) values exceeding 0.6 for AD and 0.8 for T2DM. Further analysis explored the interactions, pathway enrichments, regulatory mechanisms, and potential drug interactions of these key genes. In the AD murine model, quantitative real-time polymerase chain reaction (qRT-PCR) analysis revealed significant upregulation of G6pd, Eno3, and Taldo1. Similarly, in the T2DM murine model, elevated expression levels of G6pd, Pfkl, Eno3, and Pgd were observed. Conclusion Our rigorous research sheds light on the molecular interconnections between AD and T2DM from a glucometabolic perspective, revealing new opportunities for pharmacological innovation and therapeutic approaches. This study appears to be the first to extensively investigate glucometabolic-associated DEGs and key genes in both AD and T2DM, utilizing multiple datasets. These insights are set to enhance our understanding of the complex pathophysiology underlying these widespread chronic diseases.
Collapse
Affiliation(s)
- Shuo Liu
- The Fourth People's Hospital of Shenyang, Shenyang, Liaoning Province, China
| | - He Chen
- The Fourth People's Hospital of Shenyang, Shenyang, Liaoning Province, China
| | - Xiao-Dong He
- The Fourth People's Hospital of Shenyang, Shenyang, Liaoning Province, China
| | - Xiao-Ou Yang
- The Fourth People's Hospital of Shenyang, Shenyang, Liaoning Province, China
| |
Collapse
|
14
|
Jang H, Lee S, An S, Park Y, Kim SJ, Cheon BK, Kim JH, Kim HJ, Na DL, Kim JP, Kim K, Seo SW. Association of Glycemic Variability With Imaging Markers of Vascular Burden, β-Amyloid, Brain Atrophy, and Cognitive Impairment. Neurology 2024; 102:e207806. [PMID: 38165363 PMCID: PMC10834128 DOI: 10.1212/wnl.0000000000207806] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/27/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND AND OBJECTIVE We aimed to investigate the association between glycemic variability (GV) and neuroimaging markers of white matter hyperintensities (WMH), beta-amyloid (Aβ), brain atrophy, and cognitive impairment. METHODS This was a retrospective cohort study that included participants without dementia from a memory clinic. They all had Aβ PET, brain MRI, and standardized neuropsychological tests and had fasting glucose (FG) levels tested more than twice during the study period. We defined GV as the intraindividual visit-to-visit variability in FG levels. Multivariable linear regression and logistic regression were used to identify whether GV was associated with the presence of severe WMH and Aβ uptake with DM, mean FG levels, age, sex, hypertension, and presence of APOE4 allele as covariates. Mediation analyses were used to investigate the mediating effect of WMH and Aβ uptake on the relationship between GV and brain atrophy and cognition. RESULTS Among the 688 participants, the mean age was 72.2 years, and the proportion of female participants was 51.9%. Increase in GV was predictive of the presence of severe WMH (coefficient [95% CI] 1.032 [1.012-1.054]; p = 0.002) and increased Aβ uptake (1.005 [1.001-1.008]; p = 0.007). Both WMH and increased Aβ uptake partially mediated the relationship between GV and frontal-executive dysfunction (GV → WMH → frontal-executive; direct effect, -0.319 [-0.557 to -0.080]; indirect effect, -0.050 [-0.091 to -0.008]) and memory dysfunction (GV → Aβ → memory; direct effect, -0.182 [-0.338 to -0.026]; indirect effect, -0.067 [-0.119 to -0.015]), respectively. In addition, increased Aβ uptake completely mediated the relationship between GV and hippocampal volume (indirect effect, -1.091 [-2.078 to -0.103]) and partially mediated the relationship between GV and parietal thickness (direct effect, -0.00101 [-0.00185 to -0.00016]; indirect effect, -0.00016 [-0.00032 to -0.000002]). DISCUSSION Our findings suggest that increased GV is related to vascular and Alzheimer risk factors and neurodegenerative markers, which in turn leads to subsequent cognitive impairment. Furthermore, GV can be considered a potentially modifiable risk factor for dementia prevention.
Collapse
Affiliation(s)
- Hyemin Jang
- From the Alzheimer's Disease Convergence Research Center (H.J., S.A., Y.P., S.-J.K., B.K.C., J.H.K., H.J.K., D.L.N., J.P.K., S.W.S.), Samsung Medical Center; Department of Digital Health (H.J., S.L., K.K., S.W.S.), Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University; Department of Neurology (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center, Sungkyunkwan University School of Medicine; Neuroscience Center (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center; Happymind Clinic (D.L.N.); Biomedical Statistics Center (K.K.), Research Institute for Future Medicine, Samsung Medical Center; and Department of Data Convergence and Future Medicine (K.K.), Sungkyunkwan University School of Medicine, Seoul, Korea. Dr. Jang is currently at the Department of Neurology, Seoul National University Hospital, Korea
| | - Sungjoo Lee
- From the Alzheimer's Disease Convergence Research Center (H.J., S.A., Y.P., S.-J.K., B.K.C., J.H.K., H.J.K., D.L.N., J.P.K., S.W.S.), Samsung Medical Center; Department of Digital Health (H.J., S.L., K.K., S.W.S.), Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University; Department of Neurology (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center, Sungkyunkwan University School of Medicine; Neuroscience Center (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center; Happymind Clinic (D.L.N.); Biomedical Statistics Center (K.K.), Research Institute for Future Medicine, Samsung Medical Center; and Department of Data Convergence and Future Medicine (K.K.), Sungkyunkwan University School of Medicine, Seoul, Korea. Dr. Jang is currently at the Department of Neurology, Seoul National University Hospital, Korea
| | - Sungsik An
- From the Alzheimer's Disease Convergence Research Center (H.J., S.A., Y.P., S.-J.K., B.K.C., J.H.K., H.J.K., D.L.N., J.P.K., S.W.S.), Samsung Medical Center; Department of Digital Health (H.J., S.L., K.K., S.W.S.), Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University; Department of Neurology (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center, Sungkyunkwan University School of Medicine; Neuroscience Center (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center; Happymind Clinic (D.L.N.); Biomedical Statistics Center (K.K.), Research Institute for Future Medicine, Samsung Medical Center; and Department of Data Convergence and Future Medicine (K.K.), Sungkyunkwan University School of Medicine, Seoul, Korea. Dr. Jang is currently at the Department of Neurology, Seoul National University Hospital, Korea
| | - Yuhyun Park
- From the Alzheimer's Disease Convergence Research Center (H.J., S.A., Y.P., S.-J.K., B.K.C., J.H.K., H.J.K., D.L.N., J.P.K., S.W.S.), Samsung Medical Center; Department of Digital Health (H.J., S.L., K.K., S.W.S.), Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University; Department of Neurology (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center, Sungkyunkwan University School of Medicine; Neuroscience Center (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center; Happymind Clinic (D.L.N.); Biomedical Statistics Center (K.K.), Research Institute for Future Medicine, Samsung Medical Center; and Department of Data Convergence and Future Medicine (K.K.), Sungkyunkwan University School of Medicine, Seoul, Korea. Dr. Jang is currently at the Department of Neurology, Seoul National University Hospital, Korea
| | - Soo-Jong Kim
- From the Alzheimer's Disease Convergence Research Center (H.J., S.A., Y.P., S.-J.K., B.K.C., J.H.K., H.J.K., D.L.N., J.P.K., S.W.S.), Samsung Medical Center; Department of Digital Health (H.J., S.L., K.K., S.W.S.), Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University; Department of Neurology (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center, Sungkyunkwan University School of Medicine; Neuroscience Center (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center; Happymind Clinic (D.L.N.); Biomedical Statistics Center (K.K.), Research Institute for Future Medicine, Samsung Medical Center; and Department of Data Convergence and Future Medicine (K.K.), Sungkyunkwan University School of Medicine, Seoul, Korea. Dr. Jang is currently at the Department of Neurology, Seoul National University Hospital, Korea
| | - Bo Kyoung Cheon
- From the Alzheimer's Disease Convergence Research Center (H.J., S.A., Y.P., S.-J.K., B.K.C., J.H.K., H.J.K., D.L.N., J.P.K., S.W.S.), Samsung Medical Center; Department of Digital Health (H.J., S.L., K.K., S.W.S.), Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University; Department of Neurology (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center, Sungkyunkwan University School of Medicine; Neuroscience Center (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center; Happymind Clinic (D.L.N.); Biomedical Statistics Center (K.K.), Research Institute for Future Medicine, Samsung Medical Center; and Department of Data Convergence and Future Medicine (K.K.), Sungkyunkwan University School of Medicine, Seoul, Korea. Dr. Jang is currently at the Department of Neurology, Seoul National University Hospital, Korea
| | - Ji Hyun Kim
- From the Alzheimer's Disease Convergence Research Center (H.J., S.A., Y.P., S.-J.K., B.K.C., J.H.K., H.J.K., D.L.N., J.P.K., S.W.S.), Samsung Medical Center; Department of Digital Health (H.J., S.L., K.K., S.W.S.), Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University; Department of Neurology (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center, Sungkyunkwan University School of Medicine; Neuroscience Center (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center; Happymind Clinic (D.L.N.); Biomedical Statistics Center (K.K.), Research Institute for Future Medicine, Samsung Medical Center; and Department of Data Convergence and Future Medicine (K.K.), Sungkyunkwan University School of Medicine, Seoul, Korea. Dr. Jang is currently at the Department of Neurology, Seoul National University Hospital, Korea
| | - Hee Jin Kim
- From the Alzheimer's Disease Convergence Research Center (H.J., S.A., Y.P., S.-J.K., B.K.C., J.H.K., H.J.K., D.L.N., J.P.K., S.W.S.), Samsung Medical Center; Department of Digital Health (H.J., S.L., K.K., S.W.S.), Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University; Department of Neurology (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center, Sungkyunkwan University School of Medicine; Neuroscience Center (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center; Happymind Clinic (D.L.N.); Biomedical Statistics Center (K.K.), Research Institute for Future Medicine, Samsung Medical Center; and Department of Data Convergence and Future Medicine (K.K.), Sungkyunkwan University School of Medicine, Seoul, Korea. Dr. Jang is currently at the Department of Neurology, Seoul National University Hospital, Korea
| | - Duk L Na
- From the Alzheimer's Disease Convergence Research Center (H.J., S.A., Y.P., S.-J.K., B.K.C., J.H.K., H.J.K., D.L.N., J.P.K., S.W.S.), Samsung Medical Center; Department of Digital Health (H.J., S.L., K.K., S.W.S.), Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University; Department of Neurology (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center, Sungkyunkwan University School of Medicine; Neuroscience Center (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center; Happymind Clinic (D.L.N.); Biomedical Statistics Center (K.K.), Research Institute for Future Medicine, Samsung Medical Center; and Department of Data Convergence and Future Medicine (K.K.), Sungkyunkwan University School of Medicine, Seoul, Korea. Dr. Jang is currently at the Department of Neurology, Seoul National University Hospital, Korea
| | - Jun Pyo Kim
- From the Alzheimer's Disease Convergence Research Center (H.J., S.A., Y.P., S.-J.K., B.K.C., J.H.K., H.J.K., D.L.N., J.P.K., S.W.S.), Samsung Medical Center; Department of Digital Health (H.J., S.L., K.K., S.W.S.), Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University; Department of Neurology (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center, Sungkyunkwan University School of Medicine; Neuroscience Center (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center; Happymind Clinic (D.L.N.); Biomedical Statistics Center (K.K.), Research Institute for Future Medicine, Samsung Medical Center; and Department of Data Convergence and Future Medicine (K.K.), Sungkyunkwan University School of Medicine, Seoul, Korea. Dr. Jang is currently at the Department of Neurology, Seoul National University Hospital, Korea
| | - Kyunga Kim
- From the Alzheimer's Disease Convergence Research Center (H.J., S.A., Y.P., S.-J.K., B.K.C., J.H.K., H.J.K., D.L.N., J.P.K., S.W.S.), Samsung Medical Center; Department of Digital Health (H.J., S.L., K.K., S.W.S.), Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University; Department of Neurology (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center, Sungkyunkwan University School of Medicine; Neuroscience Center (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center; Happymind Clinic (D.L.N.); Biomedical Statistics Center (K.K.), Research Institute for Future Medicine, Samsung Medical Center; and Department of Data Convergence and Future Medicine (K.K.), Sungkyunkwan University School of Medicine, Seoul, Korea. Dr. Jang is currently at the Department of Neurology, Seoul National University Hospital, Korea
| | - Sang Won Seo
- From the Alzheimer's Disease Convergence Research Center (H.J., S.A., Y.P., S.-J.K., B.K.C., J.H.K., H.J.K., D.L.N., J.P.K., S.W.S.), Samsung Medical Center; Department of Digital Health (H.J., S.L., K.K., S.W.S.), Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University; Department of Neurology (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center, Sungkyunkwan University School of Medicine; Neuroscience Center (H.J., H.J.K., J.P.K., S.W.S.), Samsung Medical Center; Happymind Clinic (D.L.N.); Biomedical Statistics Center (K.K.), Research Institute for Future Medicine, Samsung Medical Center; and Department of Data Convergence and Future Medicine (K.K.), Sungkyunkwan University School of Medicine, Seoul, Korea. Dr. Jang is currently at the Department of Neurology, Seoul National University Hospital, Korea
| |
Collapse
|
15
|
Adem MA, Decourt B, Sabbagh MN. Pharmacological Approaches Using Diabetic Drugs Repurposed for Alzheimer's Disease. Biomedicines 2024; 12:99. [PMID: 38255204 PMCID: PMC10813018 DOI: 10.3390/biomedicines12010099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) are chronic, progressive disorders affecting the elderly, which fosters global healthcare concern with the growing aging population. Both T2DM and AD have been linked with increasing age, advanced glycosylation end products, obesity, and insulin resistance. Insulin resistance in the periphery is significant in the development of T2DM and it has been posited that insulin resistance in the brain plays a key role in AD pathogenesis, earning AD the name "type 3 diabetes". These clinical and epidemiological links between AD and T2DM have become increasingly pronounced throughout the years, and serve as a means to investigate the effects of antidiabetic therapies in AD, such as metformin, intranasal insulin, incretins, DPP4 inhibitors, PPAR-γ agonists, SGLT2 inhibitors. The majority of these drugs have shown benefit in preclinical trials, and have shown some promising results in clinical trials, with the improvement of cognitive faculties in participants with mild cognitive impairment and AD. In this review, we have summarize the benefits, risks, and conflicting data that currently exist for diabetic drugs being repurposed for the treatment of AD.
Collapse
Affiliation(s)
- Muna A. Adem
- Department of Neurology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, 350 W. Thomas Rd., Phoenix, AZ 85013, USA
| | - Boris Decourt
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| | - Marwan N. Sabbagh
- Department of Neurology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, 350 W. Thomas Rd., Phoenix, AZ 85013, USA
| |
Collapse
|
16
|
Xie H, Yu Y, Yang Y, Sun Q, Li ZY, Ni MH, Li SN, Dai P, Cui YY, Cao XY, Jiang N, Du LJ, Gao W, Bi JJ, Yan LF, Cui GB. Commonalities and distinctions between the type 2 diabetes mellitus and Alzheimer's disease: a systematic review and multimodal neuroimaging meta-analysis. Front Neurosci 2023; 17:1301778. [PMID: 38125399 PMCID: PMC10731270 DOI: 10.3389/fnins.2023.1301778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
Background Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM) are aging related diseases with high incidence. Because of the correlation of incidence rate and some possible mechanisms of comorbidity, the two diseases have been studied in combination by many researchers, and even some scholars call AD type 3 diabetes. But the relationship between the two is still controversial. Methods This study used seed-based d mapping software to conduct a meta-analysis of the whole brain resting state functional magnetic resonance imaging (rs-fMRI) study, exploring the differences in amplitude low-frequency fluctuation (ALFF) and cerebral blood flow (CBF) between patients (AD or T2DM) and healthy controls (HCs), and searching for neuroimaging evidence that can explain the relationship between the two diseases. Results The final study included 22 datasets of ALFF and 22 datasets of CBF. The results of T2DM group showed that ALFF increased in both cerebellum and left inferior temporal gyrus regions, but decreased in left middle occipital gyrus, right inferior occipital gyrus, and left anterior central gyrus regions. In the T2DM group, CBF increased in the right supplementary motor area, while decreased in the middle occipital gyrus and inferior parietal gyrus. The results of the AD group showed that the ALFF increased in the right cerebellum, right hippocampus, and right striatum, while decreased in the precuneus gyrus and right superior temporal gyrus. In the AD group, CBF in the anterior precuneus gyrus and inferior parietal gyrus decreased. Multimodal analysis within a disease showed that ALFF and CBF both decreased in the occipital lobe of the T2DM group and in the precuneus and parietal lobe of the AD group. In addition, there was a common decrease of CBF in the right middle occipital gyrus in both groups. Conclusion Based on neuroimaging evidence, we believe that T2DM and AD are two diseases with their respective characteristics of central nervous activity and cerebral perfusion. The changes in CBF between the two diseases partially overlap, which is consistent with their respective clinical characteristics and also indicates a close relationship between them. Systematic review registration PROSPERO [CRD42022370014].
Collapse
Affiliation(s)
- Hao Xie
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Ying Yu
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Yang Yang
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Qian Sun
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Ze-Yang Li
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Min-Hua Ni
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Si-Ning Li
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
- Faculty of Medical Technology, Xi’an Medical University, Xi’an, Shaanxi, China
| | - Pan Dai
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
- Faculty of Medical Technology, Xi’an Medical University, Xi’an, Shaanxi, China
| | - Yan-Yan Cui
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
- Faculty of Medical Technology, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Xin-Yu Cao
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
- Faculty of Medical Technology, Medical School of Yan’an University, Yan’an, Shaanxi, China
| | - Nan Jiang
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Li-Juan Du
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Wen Gao
- Student Brigade, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Jia-Jun Bi
- Student Brigade, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Lin-Feng Yan
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Guang-Bin Cui
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| |
Collapse
|
17
|
Kandi S, Cline EN, Rivera BM, Viola KL, Zhu J, Condello C, LeDuc RD, Klein WL, Kelleher NL, Patrie SM. Amyloid β Proteoforms Elucidated by Quantitative LC/MS in the 5xFAD Mouse Model of Alzheimer's Disease. J Proteome Res 2023; 22:3475-3488. [PMID: 37847596 PMCID: PMC10840081 DOI: 10.1021/acs.jproteome.3c00353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
Numerous Aβ proteoforms, identified in the human brain, possess differential neurotoxic and aggregation propensities. These proteoforms contribute in unknown ways to the conformations and resultant pathogenicity of oligomers, protofibrils, and fibrils in Alzheimer's disease (AD) manifestation owing to the lack of molecular-level specificity to the exact chemical composition of underlying protein products with widespread interrogating techniques, like immunoassays. We evaluated Aβ proteoform flux using quantitative top-down mass spectrometry (TDMS) in a well-studied 5xFAD mouse model of age-dependent Aβ-amyloidosis. Though the brain-derived Aβ proteoform landscape is largely occupied by Aβ1-42, 25 different forms of Aβ with differential solubility were identified. These proteoforms fall into three natural groups defined by hierarchical clustering of expression levels in the context of mouse age and proteoform solubility, with each group sharing physiochemical properties associated with either N/C-terminal truncations or both. Overall, the TDMS workflow outlined may hold tremendous potential for investigating proteoform-level relationships between insoluble fibrils and soluble Aβ, including low-molecular-weight oligomers hypothesized to serve as the key drivers of neurotoxicity. Similarly, the workflow may also help to validate the utility of AD-relevant animal models to recapitulate amyloidosis mechanisms or possibly explain disconnects observed in therapeutic efficacy in animal models vs humans.
Collapse
Affiliation(s)
- Soumya Kandi
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Erika N Cline
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Department of Neurobiology, Northwestern University, Evanston, Illinois 60208, United States
| | - Brianna M Rivera
- Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, California 94158, United States
| | - Kirsten L Viola
- Department of Neurobiology, Northwestern University, Evanston, Illinois 60208, United States
| | - Jiuhe Zhu
- Department of Neurobiology, Northwestern University, Evanston, Illinois 60208, United States
| | - Carlo Condello
- Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, California 94158, United States
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, California 94158, United States
| | - Richard D LeDuc
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - William L Klein
- Department of Neurobiology, Northwestern University, Evanston, Illinois 60208, United States
| | - Neil L Kelleher
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Steven M Patrie
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
18
|
Pelle MC, Zaffina I, Giofrè F, Pujia R, Arturi F. Potential Role of Glucagon-like Peptide-1 Receptor Agonists in the Treatment of Cognitive Decline and Dementia in Diabetes Mellitus. Int J Mol Sci 2023; 24:11301. [PMID: 37511061 PMCID: PMC10379573 DOI: 10.3390/ijms241411301] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Dementia is a permanent illness characterized by mental instability, memory loss, and cognitive decline. Many studies have demonstrated an association between diabetes and cognitive dysfunction that proceeds in three steps, namely, diabetes-associated cognitive decrements, mild cognitive impairment (MCI; both non-amnesic MCI and amnesic MCI), and dementia [both vascular dementia and Alzheimer's disease (AD)]. Based on this association, this disease has been designated as type 3 diabetes mellitus. The underlying mechanisms comprise insulin resistance, inflammation, lipid abnormalities, oxidative stress, mitochondrial dysfunction, glycated end-products and autophagy. Moreover, insulin and insulin-like growth factor-1 (IGF-1) have been demonstrated to be involved. Insulin in the brain has a neuroprotective role that alters cognitive skills and alteration of insulin signaling determines beta-amyloid (Aβ) accumulation, in turn promoting brain insulin resistance. In this complex mechanism, other triggers include hyperglycemia-induced overproduction of reactive oxygen species (ROS) and inflammatory cytokines, which result in neuroinflammation, suggesting that antidiabetic drugs may be potential treatments to protect against AD. Among these, glucagon-like peptide-1 receptor agonists (GLP-1RAs) are the most attractive antidiabetic drugs due to their actions on synaptic plasticity, cognition and cell survival. The present review summarizes the significant data concerning the underlying pathophysiological and pharmacological mechanisms between diabetes and dementia.
Collapse
Affiliation(s)
- Maria Chiara Pelle
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
| | - Isabella Zaffina
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
| | - Federica Giofrè
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
| | - Roberta Pujia
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
| | - Franco Arturi
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
- Research Center for the Prevention and Treatment of Metabolic Diseases (CR METDIS), University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
19
|
Song J. Amygdala activity and amygdala-hippocampus connectivity: Metabolic diseases, dementia, and neuropsychiatric issues. Biomed Pharmacother 2023; 162:114647. [PMID: 37011482 DOI: 10.1016/j.biopha.2023.114647] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/04/2023] Open
Abstract
With rapid aging of the population worldwide, the number of people with dementia is dramatically increasing. Some studies have emphasized that metabolic syndrome, which includes obesity and diabetes, leads to increased risks of dementia and cognitive decline. Factors such as insulin resistance, hyperglycemia, high blood pressure, dyslipidemia, and central obesity in metabolic syndrome are associated with synaptic failure, neuroinflammation, and imbalanced neurotransmitter levels, leading to the progression of dementia. Due to the positive correlation between diabetes and dementia, some studies have called it "type 3 diabetes". Recently, the number of patients with cognitive decline due to metabolic imbalances has considerably increased. In addition, recent studies have reported that neuropsychiatric issues such as anxiety, depressive behavior, and impaired attention are common factors in patients with metabolic disease and those with dementia. In the central nervous system (CNS), the amygdala is a central region that regulates emotional memory, mood disorders, anxiety, attention, and cognitive function. The connectivity of the amygdala with other brain regions, such as the hippocampus, and the activity of the amygdala contribute to diverse neuropathological and neuropsychiatric issues. Thus, this review summarizes the significant consequences of the critical roles of amygdala connectivity in both metabolic syndromes and dementia. Further studies on amygdala function in metabolic imbalance-related dementia are needed to treat neuropsychiatric problems in patients with this type of dementia.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Republic of Korea.
| |
Collapse
|
20
|
Maimaitituerxun R, Chen W, Xiang J, Xie Y, Kaminga AC, Wu XY, Chen L, Yang J, Liu A, Dai W. The use of nomogram for detecting mild cognitive impairment in patients with type 2 diabetes mellitus. J Diabetes 2023; 15:448-458. [PMID: 37057310 PMCID: PMC10172024 DOI: 10.1111/1753-0407.13384] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/09/2023] [Accepted: 03/21/2023] [Indexed: 04/15/2023] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is highly prevalent worldwide and may lead to a higher rate of cognitive dysfunction. This study aimed to develop and validate a nomogram-based model to detect mild cognitive impairment (MCI) in T2DM patients. METHODS Inpatients with T2DM in the endocrinology department of Xiangya Hospital were consecutively enrolled between March and December 2021. Well-qualified investigators conducted face-to-face interviews with participants to retrospectively collect sociodemographic characteristics, lifestyle factors, T2DM-related information, and history of depression and anxiety. Cognitive function was assessed using the Mini-Mental State Examination scale. A nomogram was developed to detect MCI based on the results of the multivariable logistic regression analysis. Calibration, discrimination, and clinical utility of the nomogram were subsequently evaluated by calibration plot, receiver operating characteristic curve, and decision curve analysis, respectively. RESULTS A total of 496 patients were included in this study. The prevalence of MCI in T2DM patients was 34.1% (95% confidence interval [CI]: 29.9%-38.3%). Age, marital status, household income, diabetes duration, diabetic retinopathy, anxiety, and depression were independently associated with MCI. Nomogram based on these factors had an area under the curve of 0.849 (95% CI: 0.815-0.883), and the threshold probability ranged from 35.0% to 85.0%. CONCLUSIONS Almost one in three T2DM patients suffered from MCI. The nomogram, based on age, marital status, household income, duration of diabetes, diabetic retinopathy, anxiety, and depression, achieved an optimal diagnosis of MCI. Therefore, it could provide a clinical basis for detecting MCI in T2DM patients.
Collapse
Affiliation(s)
- Rehanguli Maimaitituerxun
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Changsha, China
| | - Wenhang Chen
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Jingsha Xiang
- Human Resources Department, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yu Xie
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Changsha, China
| | - Atipatsa C Kaminga
- Department of Mathematics and Statistics, Mzuzu University, Mzuzu, Malawi
| | - Xin Yin Wu
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Changsha, China
| | - Letao Chen
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, China
| | - Jianzhou Yang
- Department of Preventive Medicine, Changzhi Medical College, Changzhi, China
| | - Aizhong Liu
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Changsha, China
| | - Wenjie Dai
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Changsha, China
| |
Collapse
|
21
|
Dave BP, Shah KC, Shah MB, Chorawala MR, Patel VN, Shah PA, Shah GB, Dhameliya TM. Unveiling the modulation of Nogo receptor in neuroregeneration and plasticity: Novel aspects and future horizon in a new frontier. Biochem Pharmacol 2023; 210:115461. [PMID: 36828272 DOI: 10.1016/j.bcp.2023.115461] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023]
Abstract
Neurodegenerative diseases (NDs) such as Alzheimer's, Parkinson's, Multiple Sclerosis, Hereditary Spastic Paraplegia, and Amyotrophic Lateral Sclerosis have emerged as the most dreaded diseases due to a lack of precise diagnostic tools and efficient therapies. Despite the fact that the contributing factors of NDs are still unidentified, mounting evidence indicates the possibility that genetic and cellular changes may lead to the significant production of abnormally misfolded proteins. These misfolded proteins lead to damaging effects thereby causing neurodegeneration. The association between Neurite outgrowth factor (Nogo) with neurological diseases and other peripheral diseases is coming into play. Three isoforms of Nogo have been identified Nogo-A, Nogo-B and Nogo-C. Among these, Nogo-A is mainly responsible for neurological diseases as it is localized in the CNS (Central Nervous System), whereas Nogo-B and Nogo-C are responsible for other diseases such as colitis, lung, intestinal injury, etc. Nogo-A, a membrane protein, had first been described as a CNS-specific inhibitor of axonal regeneration. Several recent studies have revealed the role of Nogo-A proteins and their receptors in modulating neurite outgrowth, branching, and precursor migration during nervous system development. It may also modulate or affect the inhibition of growth during the developmental processes of the CNS. Information about the effects of other ligands of Nogo protein on the CNS are yet to be discovered however several pieces of evidence have suggested that it may also influence the neuronal maturation of CNS and targeting Nogo-A could prove to be beneficial in several neurodegenerative diseases.
Collapse
Affiliation(s)
- Bhavarth P Dave
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Kashvi C Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Maitri B Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India.
| | - Vishvas N Patel
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Palak A Shah
- Department of Pharmacology, K. B. Institute of Pharmaceutical Education and Research, Gandhinagar 380023, Gujarat, India
| | - Gaurang B Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Tejas M Dhameliya
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad-382481, Gujarat, India
| |
Collapse
|
22
|
Al-Sayyar A, Hammad MM, Williams MR, Al-Onaizi M, Abubaker J, Alzaid F. Neurotransmitters in Type 2 Diabetes and the Control of Systemic and Central Energy Balance. Metabolites 2023; 13:384. [PMID: 36984824 PMCID: PMC10058084 DOI: 10.3390/metabo13030384] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 03/08/2023] Open
Abstract
Efficient signal transduction is important in maintaining the function of the nervous system across tissues. An intact neurotransmission process can regulate energy balance through proper communication between neurons and peripheral organs. This ensures that the right neural circuits are activated in the brain to modulate cellular energy homeostasis and systemic metabolic function. Alterations in neurotransmitters secretion can lead to imbalances in appetite, glucose metabolism, sleep, and thermogenesis. Dysregulation in dietary intake is also associated with disruption in neurotransmission and can trigger the onset of type 2 diabetes (T2D) and obesity. In this review, we highlight the various roles of neurotransmitters in regulating energy balance at the systemic level and in the central nervous system. We also address the link between neurotransmission imbalance and the development of T2D as well as perspectives across the fields of neuroscience and metabolism research.
Collapse
Affiliation(s)
| | | | | | - Mohammed Al-Onaizi
- Dasman Diabetes Institute, Kuwait City 15462, Kuwait
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait City 13110, Kuwait
| | | | - Fawaz Alzaid
- Dasman Diabetes Institute, Kuwait City 15462, Kuwait
- Institut Necker Enfants Malades-INEM, Université Paris Cité, CNRS, INSERM, F-75015 Paris, France
| |
Collapse
|
23
|
Ortiz-Zuñiga AM, Rojano Toimil A, Rahnama K, Lainez E, Raguer N, Simó-Servat O, Hernández C, Simó R, Ciudin A. Retinal sensitivity and gaze fixation evaluated by microperimetry in subjects with type 2 diabetes: two independent parameters that explore different neuronal circuits. J Endocrinol Invest 2023:10.1007/s40618-023-02046-y. [PMID: 36870015 PMCID: PMC10371889 DOI: 10.1007/s40618-023-02046-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/19/2023] [Indexed: 03/05/2023]
Abstract
BACKGROUND AND AIMS Retinal sensitivity (RS) and gaze fixation (GF) assessed by retinal microperimetry are useful and complementary tools for identifying mild cognitive impairment (MCI) in patients with type 2 diabetes (T2D). The hypothesis is that RS and GF examine different neural circuits: RS depends only on the visual pathway while GF reflects white matter complex connectivity networks. The aim of the study is to shed light to this issue by examining the relationship of these two parameters with visual evoked potentials (VEP), the current gold standard to examine the visual pathway. MATERIALS AND METHODS Consecutive T2D patients > 65 years were recruited from the outpatient clinic. Retinal microperimetry (MAIA 3rd generation) and visual evoked potentials (VEP) (Nicolet Viking ED). RS (dB), GF (BCEA63%, BCEA95%) (MAIA) and VEP (Latency P100ms, Amplitude75-100 uV) were analyzed. RESULTS Thirty three patients (45% women, 72.1 ± 4.6 years) were included. VEP parameters significantly correlated with RS but not with GF. CONCLUSIONS These results confirm that RS but not GF depends on the visual pathway, reinforcing the concept that they are complementary diagnostic tools. Used together can further increase the value of microperimetry as screening test for identifying T2D population with cognitive impairment.
Collapse
Affiliation(s)
- A M Ortiz-Zuñiga
- Department of Endocrinology and Nutrition, Hospital Universitari Vall Hebron, Pg Vall Hebron 119-129, 08035, Barcelona, Spain
- Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona (VHIR-UAB), Pg Vall Hebron 119-129, 08035, Barcelona, Spain
| | - A Rojano Toimil
- Department of Endocrinology and Nutrition, Hospital Universitari Vall Hebron, Pg Vall Hebron 119-129, 08035, Barcelona, Spain
| | - K Rahnama
- Department of Clinical Neurophysiology, Hospital Universitari Vall Hebron, Pg Vall Hebron 119-129, 08035, Barcelona, Spain
| | - E Lainez
- Department of Clinical Neurophysiology, Hospital Universitari Vall Hebron, Pg Vall Hebron 119-129, 08035, Barcelona, Spain
| | - N Raguer
- Department of Clinical Neurophysiology, Hospital Universitari Vall Hebron, Pg Vall Hebron 119-129, 08035, Barcelona, Spain
| | - O Simó-Servat
- Department of Endocrinology and Nutrition, Hospital Universitari Vall Hebron, Pg Vall Hebron 119-129, 08035, Barcelona, Spain
- Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona (VHIR-UAB), Pg Vall Hebron 119-129, 08035, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, C/Monforte de Lemos 3-5.Pabellón 11, 28029, Madrid, Spain
| | - C Hernández
- Department of Endocrinology and Nutrition, Hospital Universitari Vall Hebron, Pg Vall Hebron 119-129, 08035, Barcelona, Spain
- Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona (VHIR-UAB), Pg Vall Hebron 119-129, 08035, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, C/Monforte de Lemos 3-5.Pabellón 11, 28029, Madrid, Spain
| | - R Simó
- Department of Endocrinology and Nutrition, Hospital Universitari Vall Hebron, Pg Vall Hebron 119-129, 08035, Barcelona, Spain.
- Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona (VHIR-UAB), Pg Vall Hebron 119-129, 08035, Barcelona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, C/Monforte de Lemos 3-5.Pabellón 11, 28029, Madrid, Spain.
| | - A Ciudin
- Department of Endocrinology and Nutrition, Hospital Universitari Vall Hebron, Pg Vall Hebron 119-129, 08035, Barcelona, Spain.
- Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona (VHIR-UAB), Pg Vall Hebron 119-129, 08035, Barcelona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, C/Monforte de Lemos 3-5.Pabellón 11, 28029, Madrid, Spain.
| |
Collapse
|
24
|
Atiya A, Das Gupta D, Alsayari A, Alrouji M, Alotaibi A, Sharaf SE, Abdulmonem WA, Alorfi NM, Abdullah KM, Shamsi A. Linagliptin and Empagliflozin Inhibit Microtubule Affinity Regulatory Kinase 4: Repurposing Anti-Diabetic Drugs in Neurodegenerative Disorders Using In Silico and In Vitro Approaches. ACS OMEGA 2023; 8:6423-6430. [PMID: 36844587 PMCID: PMC9948186 DOI: 10.1021/acsomega.2c06634] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/24/2023] [Indexed: 06/18/2023]
Abstract
Type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) are significant public health burdens. Many studies have revealed the possibility of common pathophysiology between T2DM and AD. Thus, in recent years, studies deciphering the action mechanism of anti-diabetic drugs with their future use in AD and related pathologies are on high demand. Drug repurposing is a safe and effective approach owing to its low cost and time-saving attributes. Microtubule affinity regulating kinase 4 (MARK4) is a druggable target for various diseases and is found to be linked with AD and diabetes mellitus. MARK4 plays a vital role in energy metabolism and regulation and thus serves as an irrefutable target to treat T2DM. The present study was intended to identify the potent MARK4 inhibitors among FDA-approved anti-diabetic drugs. We performed structure-based virtual screening of FDA-approved drugs to identify the top hits against MARK4. We identified five FDA-approved drugs having an appreciable affinity and specificity toward the binding pocket of MARK4. Among these identified hits, two drugs, linagliptin, and empagliflozin, favorably bind to the MARK4 binding pocket, interacting with its critical residues and thus subjected to detailed analysis. All-atom detailed molecular dynamics (MD) simulations revealed the dynamics of binding of linagliptin and empagliflozin with MARK4. Kinase assay showed significant inhibition of MARK4 kinase activity in the presence of these drugs, implying them as potent MARK4 inhibitors. In conclusion, linagliptin and empagliflozin may be promising MARK4 inhibitors, which can further be exploited as potential lead molecules against MARK4-directed neurodegenerative diseases.
Collapse
Affiliation(s)
- Akhtar Atiya
- Department
of Pharmacognosy, College of Pharmacy, King
Khalid University (KKU), Guraiger St., Abha 62529, Saudi Arabia
| | - Debarati Das Gupta
- College
of Pharmacy, University of Michigan, 2428 Church Street, Ann Arbor, Michigan 48109, United States
| | - Abdulrhman Alsayari
- Department
of Pharmacognosy, College of Pharmacy, King
Khalid University (KKU), Guraiger St., Abha 62529, Saudi Arabia
- Complementary
and Alternative Medicine Unit, King Khalid
University (KKU), Guraiger St., Abha 62529, Saudi Arabia
| | - Mohammed Alrouji
- Department
of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia
| | - Abdulmajeed Alotaibi
- College
of Applied Medical Sciences, King Saud bin
Abdulaziz University for Health Sciences, Riyadh 11426, Saudi Arabia
| | - Sharaf E. Sharaf
- Pharmaceutical
Chemistry Department, College of Pharmacy, Umm Al-Qura University, Makkah 21421, Saudi Arabia
| | - Waleed Al Abdulmonem
- Department
of Pathology, College of Medicine, Qassim
University, Buraydah 52571, Saudi Arabia
| | - Nasser M. Alorfi
- Department
of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah 21421, Saudi Arabia
| | - K. M. Abdullah
- Department
of Biochemistry, Jain University, Bengaluru 560069, India
| | - Anas Shamsi
- Centre
of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates
| |
Collapse
|
25
|
Marrano N, Biondi G, Borrelli A, Rella M, Zambetta T, Di Gioia L, Caporusso M, Logroscino G, Perrini S, Giorgino F, Natalicchio A. Type 2 Diabetes and Alzheimer's Disease: The Emerging Role of Cellular Lipotoxicity. Biomolecules 2023; 13:183. [PMID: 36671568 PMCID: PMC9855893 DOI: 10.3390/biom13010183] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/06/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Type 2 diabetes (T2D) and Alzheimer's diseases (AD) represent major health issues that have reached alarming levels in the last decades. Although growing evidence demonstrates that AD is a significant comorbidity of T2D, and there is a ~1.4-2-fold increase in the risk of developing AD among T2D patients, the involvement of possible common triggers in the pathogenesis of these two diseases remains largely unknown. Of note, recent mechanistic insights suggest that lipotoxicity could represent the missing ring in the pathogenetic mechanisms linking T2D to AD. Indeed, obesity, which represents the main cause of lipotoxicity, has been recognized as a major risk factor for both pathological conditions. Lipotoxicity can lead to inflammation, insulin resistance, oxidative stress, ceramide and amyloid accumulation, endoplasmic reticulum stress, ferroptosis, and autophagy, which are shared biological events in the pathogenesis of T2D and AD. In the current review, we try to provide a critical and comprehensive view of the common molecular pathways activated by lipotoxicity in T2D and AD, attempting to summarize how these mechanisms can drive future research and open the way to new therapeutic perspectives.
Collapse
Affiliation(s)
- Nicola Marrano
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Giuseppina Biondi
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Anna Borrelli
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Martina Rella
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Tommaso Zambetta
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Ludovico Di Gioia
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Mariangela Caporusso
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Giancarlo Logroscino
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124 Bari, Italy
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari Aldo Moro at Pia Fondazione Cardinale G. Panico, 73039 Lecce, Italy
| | - Sebastio Perrini
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Francesco Giorgino
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Annalisa Natalicchio
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| |
Collapse
|
26
|
Sáiz-Vazquez O, Puente-Martínez A, Pacheco-Bonrostro J, Ubillos-Landa S. Blood pressure and Alzheimer's disease: A review of meta-analysis. Front Neurol 2023; 13:1065335. [PMID: 36712428 PMCID: PMC9874700 DOI: 10.3389/fneur.2022.1065335] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 12/21/2022] [Indexed: 01/13/2023] Open
Abstract
Background Alzheimer's disease (AD) is a neurological disorder of unknown cause, resulting in the death of brain cells. Identifying some of the modifiable risk factors for AD could be crucial for primary prevention and could lead to a reduction in the incidence of AD. Objective This study aimed to perform a meta-meta-analysis of studies in order to assess the effect of blood pressure (BP) on the diagnosis of AD. Method The search was restricted to meta-analyses assessing high systolic BP (SBP) and diastolic BP (DBP) and AD. We applied the PRISMA guidelines. Results A total of 214 studies were identified from major databases. Finally, five meta-analyses (52 studies) were analyzed in this review. Results confirm that high SBP is associated with AD. The exploration of parameters (sex, age, study design, region, and BP measurements) shows that only region significantly moderates the relationship between BP and AD. Asian people are those whose SBP levels >140 mmHg are associated with AD. BP is associated with AD in both people aged ≤65 years and those aged ≥65 years and in cross-sectional and longitudinal studies. In the case of DBP, only women are at a higher risk of AD, particularly when its levels are >90. Conclusion SBP is associated with both cerebrovascular disease and AD. Therefore, future studies should use other uncontrolled factors, such as cardiovascular diseases, diabetes, and stroke, to explain the relationship between SBP and AD.
Collapse
Affiliation(s)
- Olalla Sáiz-Vazquez
- Department of Occupational Therapy, Faculty of Health Science, University of Burgos, Burgos, Spain
| | - Alicia Puente-Martínez
- Department of Social Psychology and Anthropology, Faculty of Social Sciences, University of Salamanca (USAL), Salamanca, Spain
| | - Joaquín Pacheco-Bonrostro
- Department of Applied Economy, Faculty of Economics and Business Sciences, University of Burgos, Burgos, Spain
| | - Silvia Ubillos-Landa
- Department of Social Psychology, Faculty of Health Science, University of Burgos, Burgos, Spain,*Correspondence: Silvia Ubillos-Landa ✉
| |
Collapse
|
27
|
Wang XC, Chu CL, Li HC, Lu K, Liu CJ, Cai YF, Quan SJ, Zhang SJ. Efficacy and safety of hypoglycemic drugs in improving cognitive function in patients with Alzheimer's disease and mild cognitive impairment: A systematic review and network meta-analysis. Front Neurol 2022; 13:1018027. [PMID: 36530613 PMCID: PMC9747761 DOI: 10.3389/fneur.2022.1018027] [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: 08/12/2022] [Accepted: 11/15/2022] [Indexed: 10/21/2024] Open
Abstract
OBJECTIVE The purpose of this study was to compare the effects of oral hypoglycaemic drugs (HDs) on cognitive function and biomarkers of mild cognitive impairment (MCI) and Alzheimer's disease (AD) through a network meta-analysis of randomized controlled trials (RCTs). METHODS We conducted systematic searches for English- and Chinese-language articles in the PubMed, Medline, Embase, Cochrane Library and Google Scholar databases, with no date restrictions. We performed a network meta-analysis, which we report here according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA). The 16 studies included a total of 3,081 patients. We selected the Mini-Mental State Examination (MMSE), the Alzheimer's Disease Assessment Scale-Cognitive section (ADAS-Cog), the Alzheimer's Disease Cooperative Study Activities of Daily Living section (ADCS-ADL) and amyloid beta (Aβ) 42 as the outcome measures for analysis and comparison. RESULT We selected seven treatments and assessed the clinical trials in which they were tested against a placebo control. Of these treatments, intranasal insulin 20 IU (ITSN20), glucagon-like peptide-1 (GLP-1), and dipeptidyl peptidase 4 inhibitor (DPP-4) were associated with significantly improved MMSE scores (7 RCTs, 333 patients, 30≥MMSE score≥20: mild) compared with placebo [standardized mean difference (SMD) 1.11, 95% confidence interval (CI) (0.87, 1.35); SMD 0.75, 95% CI (0.04, 1.41); and SMD 4.08, 95% CI (3.39, 4.77), respectively]. Rosiglitazone 4 mg (RLZ4), rosiglitazone 10 mg (RLZ10), intranasal insulin 40 IU (ITSN40), and ITSN20 significantly decreased ADAS-Cog scores (11 RCTs, 4044 patients, 10 ≤ ADAS-Cog scores ≤ 30: mild and moderate) compared with placebo [SMD -1.40, 95% CI (-2.57, -0.23), SMD -3.02, 95% CI (-4.17, -1.86), SMD -0.92, 95% CI (-1.77, -0.08), SMD -1.88, 95% CI (-3.09, -0.66)]. Additionally, ITSN20 and ITSN40 significantly improved ADCS-ADL scores (2 RCTs, 208 patients, ADCS-ADL scale score ≤ 10: mild) compared with placebo [SMD 0.02, 95% CI (0.01, 0.03), and SMD 0.04, 95% CI (0.03, 0.05), respectively]. In the 16 included studies, the degree of AD was classified as mild or moderate. For mild cognitive impairment, DPP-4 performed best, but for mild to moderate impairment, ITSN40 had excellent performance. CONCLUSION Various HDs can improve the cognitive function of MCI and AD patients. Different drug regimens brought different degrees of improvement, which may be related to their dosage, duration, and mechanism of action. SYSTEMATIC REVIEW REGISTRATION www.crd.york.ac.uk/prospero.
Collapse
Affiliation(s)
- Xin-Chen Wang
- Department of Pharmaceutical Engineering, College of Food and Pharmaceutical Engineering, Zhaoqing University, Zhaoqing, China
| | - Chen-Liang Chu
- Department of Pharmaceutical Engineering, College of Food and Pharmaceutical Engineering, Zhaoqing University, Zhaoqing, China
| | - Han-Cheng Li
- Department of Pharmaceutical Engineering, College of Food and Pharmaceutical Engineering, Zhaoqing University, Zhaoqing, China
| | - Kuan Lu
- Department of Pharmaceutical Engineering, College of Food and Pharmaceutical Engineering, Zhaoqing University, Zhaoqing, China
| | - Cheng-Jiang Liu
- Department of General Medicine, Affiliated Anqing First People's Hospital of Anhui Medical University, Anqing, China
| | - Ye-Feng Cai
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shi-Jian Quan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shi-Jie Zhang
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
28
|
Lopez-Vilaret KM, Fernandez-Alvarez M, Shokri-Kojori E, Tomasi D, Cantero JL, Atienza M. Pre-diabetes is associated with altered functional connectivity density in cortical regions of the default-mode network. Front Aging Neurosci 2022; 14:1034355. [PMID: 36438011 PMCID: PMC9686287 DOI: 10.3389/fnagi.2022.1034355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/25/2022] [Indexed: 09/29/2023] Open
Abstract
Insulin resistance and glucose dysregulation are associated with patterns of regional brain hypometabolism characteristic of Alzheimer's disease (AD). As predicted by evidence linking brain glucose metabolism to brain functional connectivity, type 2 diabetes is accompanied by altered functional connectivity density (FCD) in regions highly vulnerable to AD, but whether these alterations start at earlier stages such as pre-diabetes remain to be elucidated. Here, in addition to assessing whether pre-diabetes leads to a functional reorganization of densely connected cortical areas (hubs), we will assess whether such reorganization is conditioned by sex and/or insulin resistance, and contributes to improved cognition. One hundred and forty-four cognitively unimpaired middle-aged and older adults (55-78 years, 79 females), 73 with normoglycemia and 71 with pre-diabetes, underwent resting-state fMRI scanning. We first computed FCD mapping on cortical surfaces to determine the number of short- and long-range functional connections of every vertex in the cortex, and next used hubs showing aberrant FCD as seeds for the resting-state functional connectivity (rs-FC) calculation. ANCOVAs and linear multiple regression analyses adjusted by demographic and cardiometabolic confounders using frequentist and Bayesian approaches were applied. Analyses revealed higher long-range FCD in the right precuneus of pre-diabetic females and lower short-range FCD in the left medial orbitofrontal cortex (mOFC) of pre-diabetic individuals with higher insulin resistance. Although the mOFC also showed altered rs-FC patterns with other regions of the default mode network in pre-diabetic individuals, it was FCD of the precuneus and mOFC, and not the magnitude of their rs-FC, that was associated with better planning abilities and Mini-Mental State Examination (MMSE) scores. Results suggest that being female and/or having high insulin resistance exacerbate pre-diabetes-induced alterations in the FCD of hubs of the default-mode network that are particularly vulnerable to AD pathology. These changes in brain network organization appear to be compensatory for pre-diabetic females, likely assisting them to maintain cognitive functioning at early stages of glucose dysregulation.
Collapse
Affiliation(s)
| | - Marina Fernandez-Alvarez
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Seville, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, Madrid, Spain
| | - Ehsan Shokri-Kojori
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Dardo Tomasi
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Jose L Cantero
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Seville, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, Madrid, Spain
| | - Mercedes Atienza
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Seville, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, Madrid, Spain
| |
Collapse
|
29
|
Chai YH, Zhang YP, Qiao YS, Gong HJ, Xu H, She HC, Patel I, Liu W, Stehouwer CDA, Zhou JB, Simó R. Association Between Diabetic Retinopathy, Brain Structural Abnormalities, and Cognitive Impairment for Accumulated Evidence in Observational Studies. Am J Ophthalmol 2022; 239:37-53. [PMID: 35063409 DOI: 10.1016/j.ajo.2022.01.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 01/07/2022] [Accepted: 01/10/2022] [Indexed: 12/01/2022]
Abstract
PURPOSE To evaluate the association between diabetic retinopathy (DR) and cerebral disease or cognitive impairment. DESIGN Systematic review and meta-analysis. METHOD The hypothesis was formulated prior to data collection. Cross-sectional studies and cohort studies that assessed the association between any measure of DR and cerebral small vessel disease or any type of cognitive impairment in diabetic participants were included. The data were independently extracted by two investigators. This systematic review and meta-analysis adhered to the Preferred Reporting Items for Systematic Reviews and Meta-analyses and Meta-analysis of Observational Studies in Epidemiology guidelines RESULTS: A total of 27 studies were included. The combined odds ratio of 5 cross-sectional/cohort studies that reported that the associations between DR and cerebral structural changes was 1.75 (95% confidence interval [CI]: 1.36-2.25). The combined hazard ratio of 4 cohort studies that examined the association between DR and cognitive impairment events was 1.47 (95% CI: 1.22-1.78). The combined odds ratio of 14 cross-sectional/cohort studies that examined the association between DR and different cognitive impairment events was 1.43 (95% CI: 1.06-1.93). The overall coefficient (β) of 4 studies that examined the relationship between DR and specific cognitive performance was 0.09 (95% CI: 0.00-0.18). Considering the quality of the data, we have performed subgroup analysis in studies scored >7 and studies scored ≤7, respectively, according to the Newcastle-Ottawa scale. CONCLUSION The present meta-analysis suggests that DR is associated with an increased risk of structural abnormalities in the brain and cognitive impairment. This association remained significant after adjusting for blood glucose, and the presence of hypertension, indicating that DR is an important danger signal for cerebral abnormalities.
Collapse
Affiliation(s)
- Yin-He Chai
- From the Department of Endocrinology (Y.-H.C., Y.-S.Q, H.-J.G, H.X., I.P., W.L. J.B.Z.), Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yong-Peng Zhang
- Beijing Tongren Eye Center (Y.P.Z., H.C.S.), Beijing Key Laboratory of Ophthalmology and Visual Science, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yu-Shun Qiao
- From the Department of Endocrinology (Y.-H.C., Y.-S.Q, H.-J.G, H.X., I.P., W.L. J.B.Z.), Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hong-Jian Gong
- From the Department of Endocrinology (Y.-H.C., Y.-S.Q, H.-J.G, H.X., I.P., W.L. J.B.Z.), Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hui Xu
- From the Department of Endocrinology (Y.-H.C., Y.-S.Q, H.-J.G, H.X., I.P., W.L. J.B.Z.), Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hai-Cheng She
- Beijing Tongren Eye Center (Y.P.Z., H.C.S.), Beijing Key Laboratory of Ophthalmology and Visual Science, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ikramulhaq Patel
- From the Department of Endocrinology (Y.-H.C., Y.-S.Q, H.-J.G, H.X., I.P., W.L. J.B.Z.), Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Wei Liu
- From the Department of Endocrinology (Y.-H.C., Y.-S.Q, H.-J.G, H.X., I.P., W.L. J.B.Z.), Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Coen D A Stehouwer
- Department of Internal Medicine and CARIM School for Cardiovascular Diseases (C.D.A.S.), Maastricht University Medical Center, Maastricht, the Netherlands
| | - Jian-Bo Zhou
- From the Department of Endocrinology (Y.-H.C., Y.-S.Q, H.-J.G, H.X., I.P., W.L. J.B.Z.), Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| | - Rafael Simó
- Department of Endocrinology and Nutrition (R.S.), Vall d'Hebron University Hospital, Autonomous University, Barcelona, Spain; Diabetes and Metabolism Research Unit (R.S.), Vall d'Hebron Research Institute (VHIR), Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (R.S.), Instituto de Salud Carlos III (ICSIII), Madrid, Spain
| |
Collapse
|
30
|
Patel VN, Chorawala MR, Shah MB, Shah KC, Dave BP, Shah MP, Patel TM. Emerging Pathophysiological Mechanisms Linking Diabetes Mellitus and Alzheimer’s Disease: An Old Wine in a New Bottle. J Alzheimers Dis Rep 2022; 6:349-357. [PMID: 35891636 PMCID: PMC9277673 DOI: 10.3233/adr-220021] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/30/2022] [Indexed: 01/15/2023] Open
Abstract
Type-2 diabetes mellitus (T2DM) is a chronic immuno-inflammatory and metabolic disease characterized by hyperglycemia and insulin resistance with corresponding hyperinsulinemia. On the other hand, Alzheimer’s disease (AD) is a neurodegenerative disease involving cognitive impairment, neuronal dysfunction, and memory loss. Several recently published literatures suggest a causal relationship between T2DM and AD. In this review, we have discussed several potential mechanisms underlying diabetes-induced cognitive impairment which include, abnormal insulin signaling, amyloid-β accumulation, oxidative stress, immuno-inflammation, mitochondrial dysfunction, advanced glycation end products, acetylcholinesterase and butyrylcholinesterase, advanced lipid peroxidation products, and apolipoprotein E. All these interconnected mechanisms may act either individually or synergistically which eventually leads to neurodegeneration and AD.
Collapse
Affiliation(s)
- Vishvas N. Patel
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India
| | - Mehul R. Chorawala
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India
| | - Maitri B. Shah
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India
| | - Kashvi C. Shah
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India
| | - Bhavarth P. Dave
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India
| | - Manal P. Shah
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India
| | - Tanvi M. Patel
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India
| |
Collapse
|
31
|
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
|
32
|
Ortiz GG, Huerta M, González-Usigli HA, Torres-Sánchez ED, Delgado-Lara DLC, Pacheco-Moisés FP, Mireles-Ramírez MA, Torres-Mendoza BMG, Moreno-Cih RI, Velázquez-Brizuela IE. Cognitive disorder and dementia in type 2 diabetes mellitus. World J Diabetes 2022; 13:319-337. [PMID: 35582669 PMCID: PMC9052006 DOI: 10.4239/wjd.v13.i4.319] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/14/2021] [Accepted: 03/17/2022] [Indexed: 02/06/2023] Open
Abstract
Insulin, a key pleiotropic hormone, regulates metabolism through several signaling pathways in target tissues including skeletal muscle, liver, and brain. In the brain, insulin modulates learning and memory, and impaired insulin signaling is associated with metabolic dysregulation and neurodegenerative diseases. At the receptor level, in aging and Alzheimer’s disease (AD) models, the amount of insulin receptors and their functions are decreased. Clinical and animal model studies suggest that memory improvements are due to changes in insulin levels. Furthermore, diabetes mellitus (DM) and insulin resistance are associated with age-related cognitive decline, increased levels of β-amyloid peptide, phosphorylation of tau protein; oxidative stress, pro-inflammatory cytokine production, and dyslipidemia. Recent evidence shows that deleting brain insulin receptors leads to mild obesity and insulin resistance without influencing brain size and apoptosis development. Conversely, deleting insulin-like growth factor 1 receptor (IGF-1R) affects brain size and development, and contributes to behavior changes. Insulin is synthesized locally in the brain and is released from the neurons. Here, we reviewed proposed pathophysiological hypotheses to explain increased risk of dementia in the presence of DM. Regardless of the exact sequence of events leading to neurodegeneration, there is strong evidence that mitochondrial dysfunction plays a key role in AD and DM. A triple transgenic mouse model of AD showed mitochondrial dysfunction, oxidative stress, and loss of synaptic integrity. These alterations are comparable to those induced in wild-type mice treated with sucrose, which is consistent with the proposal that mitochondrial alterations are associated with DM and contribute to AD development. Alterations in insulin/IGF-1 signaling in DM could lead to mitochondrial dysfunction and low antioxidant capacity of the cell. Thus, insulin/IGF-1 signaling is important for increased neural processing and systemic metabolism, and could be a specific target for therapeutic strategies to decrease alterations associated with age-related cognitive decline.
Collapse
Affiliation(s)
- Genaro G Ortiz
- Department of Philosophical and Methodological Disciplines, University Health Sciences Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Department of Neurology, Movement Disorders Clinic, Sub-Specialty Medical Unit, National Western Medical Center, Mexican Social Security Institute (IMSS), Guadalajara 44340, Jalisco, Mexico
| | - Miguel Huerta
- University Biomedical Research Center, University of Colima, Colima 28040, Mexico
| | - Héctor A González-Usigli
- Department of Neurology, Movement Disorders Clinic, Sub-Specialty Medical Unit, National Western Medical Center, Mexican Social Security Institute (IMSS), Guadalajara 44340, Jalisco, Mexico
| | - Erandis D Torres-Sánchez
- Department of Medical and Life Sciences, University Center of ‘La Ciénega’, University of Guadalajara, Ocotlán 47810, Jalisco, Mexico
| | - Daniela LC Delgado-Lara
- Department of Philosophical and Methodological Disciplines, University Health Sciences Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Fermín P Pacheco-Moisés
- Department of Chemistry, University Center of Exact Sciences and Engineering, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Mario A Mireles-Ramírez
- Department of Neurology, Movement Disorders Clinic, Sub-Specialty Medical Unit, National Western Medical Center, Mexican Social Security Institute (IMSS), Guadalajara 44340, Jalisco, Mexico
| | - Blanca MG Torres-Mendoza
- Department of Philosophical and Methodological Disciplines, University Health Sciences Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Division of Neurosciences, Western Biomedical Research Center, Mexican Social Security Institute (IMSS), Guadalajara 44340, Jalisco, Mexico
| | - Roxana I Moreno-Cih
- Gerontology Postgraduate Program, Public Health Department, University Health Sciences Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Irma E Velázquez-Brizuela
- Department of Philosophical and Methodological Disciplines, University Health Sciences Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
33
|
Athanasaki A, Melanis K, Tsantzali I, Stefanou MI, Ntymenou S, Paraskevas SG, Kalamatianos T, Boutati E, Lambadiari V, Voumvourakis KI, Stranjalis G, Giannopoulos S, Tsivgoulis G, Paraskevas GP. Type 2 Diabetes Mellitus as a Risk Factor for Alzheimer’s Disease: Review and Meta-Analysis. Biomedicines 2022; 10:biomedicines10040778. [PMID: 35453527 PMCID: PMC9029855 DOI: 10.3390/biomedicines10040778] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/17/2022] [Accepted: 03/25/2022] [Indexed: 01/16/2023] Open
Abstract
Alzheimer’s disease is the most common type of dementia, reaching 60–80% of case totals, and is one of the major global causes of the elderly population’s decline in functionality concerning daily life activities. Epidemiological research has already indicated that, in addition to several others metabolic factors, diabetes mellitus type 2 is a risk factor of Alzheimer’s disease. Many molecular pathways have been described, and at the same time, there are clues that suggest the connection between type 2 diabetes mellitus and Alzheimer’s disease, through specific genes, autophagy, and even inflammatory pathways. A systematic review with meta-analysis was conducted, and its main goal was to reveal the multilevel connection between these diseases.
Collapse
Affiliation(s)
- Athanasia Athanasaki
- Department of Neurology, Evangelismos Hospital, 10676 Athens, Greece; (A.A.); (S.N.)
| | - Konstantinos Melanis
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
| | - Ioanna Tsantzali
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
| | - Maria Ioanna Stefanou
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
| | - Sofia Ntymenou
- Department of Neurology, Evangelismos Hospital, 10676 Athens, Greece; (A.A.); (S.N.)
| | - Sotirios G. Paraskevas
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
| | - Theodosis Kalamatianos
- 1st Department of Neurosurgery, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 10676 Athens, Greece; (T.K.); (G.S.)
| | - Eleni Boutati
- 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (E.B.); (V.L.)
| | - Vaia Lambadiari
- 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (E.B.); (V.L.)
| | - Konstantinos I. Voumvourakis
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
| | - George Stranjalis
- 1st Department of Neurosurgery, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 10676 Athens, Greece; (T.K.); (G.S.)
| | - Sotirios Giannopoulos
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
| | - Georgios Tsivgoulis
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
| | - George P. Paraskevas
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
- Correspondence: ; Tel.: +30-2105832466
| |
Collapse
|
34
|
Faria-Pereira A, Morais VA. Synapses: The Brain's Energy-Demanding Sites. Int J Mol Sci 2022; 23:3627. [PMID: 35408993 PMCID: PMC8998888 DOI: 10.3390/ijms23073627] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/16/2022] [Accepted: 03/22/2022] [Indexed: 02/04/2023] Open
Abstract
The brain is one of the most energy-consuming organs in the mammalian body, and synaptic transmission is one of the major contributors. To meet these energetic requirements, the brain primarily uses glucose, which can be metabolized through glycolysis and/or mitochondrial oxidative phosphorylation. The relevance of these two energy production pathways in fulfilling energy at presynaptic terminals has been the subject of recent studies. In this review, we dissect the balance of glycolysis and oxidative phosphorylation to meet synaptic energy demands in both resting and stimulation conditions. Besides ATP output needs, mitochondria at synapse are also important for calcium buffering and regulation of reactive oxygen species. These two mitochondrial-associated pathways, once hampered, impact negatively on neuronal homeostasis and synaptic activity. Therefore, as mitochondria assume a critical role in synaptic homeostasis, it is becoming evident that the synaptic mitochondria population possesses a distinct functional fingerprint compared to other brain mitochondria. Ultimately, dysregulation of synaptic bioenergetics through glycolytic and mitochondrial dysfunctions is increasingly implicated in neurodegenerative disorders, as one of the first hallmarks in several of these diseases are synaptic energy deficits, followed by synapse degeneration.
Collapse
Affiliation(s)
| | - Vanessa A. Morais
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal;
| |
Collapse
|
35
|
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: 141] [Impact Index Per Article: 47.0] [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
|
36
|
Lima JEBF, Moreira NCS, Sakamoto-Hojo ET. Mechanisms underlying the pathophysiology of type 2 diabetes: From risk factors to oxidative stress, metabolic dysfunction, and hyperglycemia. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2022; 874-875:503437. [PMID: 35151421 DOI: 10.1016/j.mrgentox.2021.503437] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/08/2021] [Accepted: 12/12/2021] [Indexed: 12/17/2022]
Abstract
Type 2 diabetes (T2D) is a complex multifactorial disease that emerges from the combination of genetic and environmental factors, and obesity, lifestyle, and aging are the most relevant risk factors. Hyperglycemia is the main metabolic feature of T2D as a consequence of insulin resistance and β-cell dysfunction. Among the cellular alterations induced by hyperglycemia, the overproduction of reactive oxygen species (ROS) and consequently oxidative stress, accompanied by a reduced antioxidant response and impaired DNA repair pathways, represent essential mechanisms underlying the pathophysiology of T2D and the development of late complications. Mitochondrial dysfunction, endoplasmic reticulum (ER) stress, and inflammation are also closely correlated with insulin resistance and β-cell dysfunction. This review focus on the mechanisms by which oxidative stress, mitochondrial dysfunction, ER stress, and inflammation are involved in the pathophysiology of T2D, highlighting the importance of the antioxidant response and DNA repair mechanisms counteracting the development of the disease. Moreover, we indicate evidence on how nutritional interventions effectively improve diabetes care. Additionally, we address key molecular characteristics and signaling pathways shared between T2D and Alzheimer's disease (AD), which might probably be implicated in the risk of T2D patients to develop AD.
Collapse
Affiliation(s)
- Jessica E B F Lima
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo - USP, Ribeirão Preto, SP, Brazil
| | - Natalia C S Moreira
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo - USP, Ribeirão Preto, SP, Brazil
| | - Elza T Sakamoto-Hojo
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo - USP, Ribeirão Preto, SP, Brazil; Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
37
|
Arbo BD, Schimith LE, Goulart dos Santos M, Hort MA. Repositioning and development of new treatments for neurodegenerative diseases: Focus on neuroinflammation. Eur J Pharmacol 2022; 919:174800. [DOI: 10.1016/j.ejphar.2022.174800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/18/2022] [Accepted: 02/02/2022] [Indexed: 11/03/2022]
|
38
|
Carranza-Naval MJ, Del Marco A, Hierro-Bujalance C, Alves-Martinez P, Infante-Garcia C, Vargas-Soria M, Herrera M, Barba-Cordoba B, Atienza-Navarro I, Lubian-Lopez S, Garcia-Alloza M. Liraglutide Reduces Vascular Damage, Neuronal Loss, and Cognitive Impairment in a Mixed Murine Model of Alzheimer's Disease and Type 2 Diabetes. Front Aging Neurosci 2022; 13:741923. [PMID: 34975451 PMCID: PMC8716860 DOI: 10.3389/fnagi.2021.741923] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/20/2021] [Indexed: 12/29/2022] Open
Abstract
Alzheimer's disease is the most common form of dementia, and epidemiological studies support that type 2 diabetes (T2D) is a major contributor. The relationship between both diseases and the fact that Alzheimer's disease (AD) does not have a successful treatment support the study on antidiabetic drugs limiting or slowing down brain complications in AD. Among these, liraglutide (LRGT), a glucagon-like peptide-1 agonist, is currently being tested in patients with AD in the Evaluating Liraglutide in Alzheimer's Disease (ELAD) clinical trial. However, the effects of LRGT on brain pathology when AD and T2D coexist have not been assessed. We have administered LRGT (500 μg/kg/day) to a mixed murine model of AD and T2D (APP/PS1xdb/db mice) for 20 weeks. We have evaluated metabolic parameters as well as the effects of LRGT on learning and memory. Postmortem analysis included assessment of brain amyloid-β and tau pathologies, microglia activation, spontaneous bleeding and neuronal loss, as well as insulin and insulin-like growth factor 1 receptors. LRGT treatment reduced glucose levels in diabetic mice (db/db and APP/PS1xdb/db) after 4 weeks of treatment. LRGT also helped to maintain insulin levels after 8 weeks of treatment. While we did not detect any effects on cortical insulin or insulin-like growth factor 1 receptor m-RNA levels, LRGT significantly reduced brain atrophy in the db/db and APP/PS1xdb/db mice. LRGT treatment also rescued neuron density in the APP/PS1xdb/db mice in the proximity (p = 0.008) far from amyloid plaques (p < 0.001). LRGT reduced amyloid plaque burden in the APP/PS1 animals (p < 0.001), as well as Aβ aggregates levels (p = 0.046), and tau hyperphosphorylation (p = 0.009) in the APP/PS1xdb/db mice. Spontaneous bleeding was also ameliorated in the APP/PS1xdb/db animals (p = 0.012), and microglia burden was reduced in the proximity of amyloid plaques in the APP/PS1 and APP/PS1xdb/db mice (p < 0.001), while microglia was reduced in areas far from amyloid plaques in the db/db and APP/PS1xdb/db mice (p < 0.001). This overall improvement helped to rescue cognitive impairment in AD-T2D mice in the new object discrimination test (p < 0.001) and Morris water maze (p < 0.001). Altogether, our data support the role of LRGT in reduction of associated brain complications when T2D and AD occur simultaneously, as regularly observed in the clinical arena.
Collapse
Affiliation(s)
- Maria Jose Carranza-Naval
- Division of Physiology, School of Medicine, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cádiz (INIBICA), Cádiz, Spain.,Salus Infirmorum-Universidad de Cádiz, Cádiz, Spain
| | - Angel Del Marco
- Division of Physiology, School of Medicine, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cádiz (INIBICA), Cádiz, Spain
| | - Carmen Hierro-Bujalance
- Division of Physiology, School of Medicine, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cádiz (INIBICA), Cádiz, Spain
| | - Pilar Alves-Martinez
- Division of Physiology, School of Medicine, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cádiz (INIBICA), Cádiz, Spain
| | - Carmen Infante-Garcia
- Division of Physiology, School of Medicine, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cádiz (INIBICA), Cádiz, Spain
| | - Maria Vargas-Soria
- Division of Physiology, School of Medicine, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cádiz (INIBICA), Cádiz, Spain
| | - Marta Herrera
- Division of Physiology, School of Medicine, Universidad de Cádiz, Cádiz, Spain
| | - Belen Barba-Cordoba
- Division of Physiology, School of Medicine, Universidad de Cádiz, Cádiz, Spain
| | - Isabel Atienza-Navarro
- Division of Physiology, School of Medicine, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cádiz (INIBICA), Cádiz, Spain
| | - Simon Lubian-Lopez
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cádiz (INIBICA), Cádiz, Spain.,Section of Neonatology, Division of Pediatrics, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - Monica Garcia-Alloza
- Division of Physiology, School of Medicine, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cádiz (INIBICA), Cádiz, Spain
| |
Collapse
|
39
|
Lee CJ, Lee JY, Han K, Kim DH, Cho H, Kim KJ, Kang ES, Cha BS, Lee YH, Park S. Blood Pressure Levels and Risks of Dementia: a Nationwide Study of 4.5 Million People. Hypertension 2021; 79:218-229. [PMID: 34775786 DOI: 10.1161/hypertensionaha.121.17283] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There are inconsistent results on the impacts of controlling blood pressure (BP) on the risk of dementia. We investigated the association between BP and risk of dementia subtypes by antihypertensive treatment and comorbidities. Using the Korean National Health Insurance Service-Health Screening Database from 2009 to 2012, a total of 4 522 447 adults aged 60+ years without a history of dementia were analyzed and followed up for a mean of 5.4 years. Individuals were classified according to their baseline systolic BP (SBP) and diastolic BP; SBP 130 to <140 mm Hg and diastolic BP 80 to <90 mm Hg were used as reference groups. The risk of overall dementia and probable Alzheimer disease was significantly higher in the SBP≥160 and lower SBP groups. These U-shaped associations were consistent regardless of antihypertensive use or comorbidities. The risk of probable vascular dementia (VaD) was not higher among lower SBP groups and increased gradually as SBP increased. Although there was a linear association between SBP and the risk of probable VaD in individuals not taking antihypertensives or without comorbidities, there was a U-shaped association in individuals taking antihypertensives or with comorbidities. Patterns of association between diastolic BP and risk of probable Alzheimer disease or probable VaD were similar to those with SBP, except for the risk of probable VaD in individuals taking antihypertensives. In conclusion, risks of probable Alzheimer disease and probable VaD were different among lower BP groups. Although the risk of dementia appears higher in people with lower BP receiving antihypertensives, this finding may be affected by comorbidities.
Collapse
Affiliation(s)
- Chan Joo Lee
- Cardiology Division, Severance Cardiovascular Hospital and Cardiovascular Research Institute (C.J.L., S.P.), Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ji-Yeon Lee
- Division of Endocrinology and Metabolism (J.-Y.L., E.S.K., B.-S.C., Y.-h.L.), Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyungdo Han
- Cardiology Division, Severance Cardiovascular Hospital and Cardiovascular Research Institute (C.J.L., S.P.), Yonsei University College of Medicine, Seoul, Republic of Korea.,Department of Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea (K.H.)
| | - Da Hye Kim
- Cardiology Division, Severance Cardiovascular Hospital and Cardiovascular Research Institute (C.J.L., S.P.), Yonsei University College of Medicine, Seoul, Republic of Korea.,Department of Medical Statistics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea (D.H.K.)
| | - Hanna Cho
- Department of Neurology, Gangnam Severance Hospital (H.C.), Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kwang Joon Kim
- Division of Geriatrics (K.J.K.), Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Seok Kang
- Division of Endocrinology and Metabolism (J.-Y.L., E.S.K., B.-S.C., Y.-h.L.), Yonsei University College of Medicine, Seoul, Republic of Korea.,Institute of Endocrine Research (E.S.K., B.-S.C., Y.-h.L.), Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Bong-Soo Cha
- Division of Endocrinology and Metabolism (J.-Y.L., E.S.K., B.-S.C., Y.-h.L.), Yonsei University College of Medicine, Seoul, Republic of Korea.,Institute of Endocrine Research (E.S.K., B.-S.C., Y.-h.L.), Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong-Ho Lee
- Division of Endocrinology and Metabolism (J.-Y.L., E.S.K., B.-S.C., Y.-h.L.), Yonsei University College of Medicine, Seoul, Republic of Korea.,Institute of Endocrine Research (E.S.K., B.-S.C., Y.-h.L.), Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sungha Park
- Cardiology Division, Severance Cardiovascular Hospital and Cardiovascular Research Institute (C.J.L., S.P.), Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
40
|
Ojakäär T, Koychev I. Secondary Prevention of Dementia: Combining Risk Factors and Scalable Screening Technology. Front Neurol 2021; 12:772836. [PMID: 34867762 PMCID: PMC8634660 DOI: 10.3389/fneur.2021.772836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is the most common cause of dementia. Over a third of dementia cases are estimated to be due to potentially modifiable risk factors, thus offering opportunities for both identification of those most likely to be in early disease as well as secondary prevention. Diabetes, hypertension and chronic kidney failure have all been linked to increased risk for AD and dementia and through their high prevalence are particularly apt targets for initiatives to reduce burden of AD. This can take place through targeted interventions of cardiovascular risk factors (shown to improve cognitive outcomes) or novel disease modifying treatments in people with confirmed AD pathology. The success of this approach to secondary prevention depends on the availability of inexpensive and scalable methods for detecting preclinical and prodromal dementia states. Developments in blood-based biomarkers for Alzheimer's disease are rapidly becoming a viable such method for monitoring large at-risk groups. In addition, digital technologies for remote monitoring of cognitive and behavioral changes can add clinically relevant data to further improve personalisation of prevention strategies. This review sets the scene for this approach to secondary care of dementia through a review of the evidence for cardiovascular risk factors (diabetes, hypertension and chronic kidney disease) as major risk factors for AD. We then summarize the developments in blood-based and cognitive biomarkers that allow the detection of pathological states at the earliest possible stage. We propose that at-risk cohorts should be created based on the interaction between cardiovascular and constitutional risk factors. These cohorts can then be monitored effectively using a combination of blood-based biomarkers and digital technologies. We argue that this strategy allows for both risk factor reduction-based prevention programmes as well as for optimisation of any benefits offered by current and future disease modifying treatment through rapid identification of individuals most likely to benefit from them.
Collapse
Affiliation(s)
| | - Ivan Koychev
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
41
|
Udeh-Momoh C, Watermeyer T. Female specific risk factors for the development of Alzheimer's disease neuropathology and cognitive impairment: Call for a precision medicine approach. Ageing Res Rev 2021; 71:101459. [PMID: 34508876 DOI: 10.1016/j.arr.2021.101459] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 08/13/2021] [Accepted: 09/06/2021] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) includes a long asymptomatic stage, which precedes the formal diagnosis of dementia. AD biomarker models provide a framework for precision medicine approaches during this stage. However, such approaches have ignored the possible influence of sex on cognition and brain health, despite female sex noted as a major risk factor. Since AD-related changes may emerge in midlife, intervention efforts are being redirected around this period. Midlife coincides with several endocrinological changes, such as the menopausal transition experienced by women. In this narrative review, we discuss evidence for sex-differences in AD neuropathological burden and outline key endocrinological mechanisms for both sexes, focussing on hormonal events throughout the lifespan that may influence female susceptibility to AD neuropathology and dementia onset. We further consider common non-modifiable (genetic) and modifiable (lifestyle and health) risk factors, highlighting possible sex-dependent differential effects for the AD disease course. Finally, we evaluate the studies selected for this review demonstrating sex-differences in cognitive, pathological and health factors, summarising the state of sex differences in AD risk factors. We further provide recommendations for targeted research on female-specific risk factors, to inform personalised strategies for AD-prevention and the promotion of female brain health.
Collapse
|
42
|
Wang R, Yan W, Du M, Tao L, Liu J. The effect of physical activity interventions on cognition function in patients with diabetes: A systematic review and meta-analysis. Diabetes Metab Res Rev 2021; 37:e3443. [PMID: 33616310 PMCID: PMC8519002 DOI: 10.1002/dmrr.3443] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND In recent years, studies have revealed that cognition may be impaired by glucose metabolism disorder. Meanwhile, physical activity has been demonstrated to maintain blood glucose. This meta-analysis was conducted to assess the effect of physical activity on cognition in patients with diabetes and provide evidence for the treatment of cognition impairment among them. METHODS We searched studies published in five databases from 1 January 1984 to 29 August 2020. A random-effect or fixed-effect meta-analysis was used to estimate the pooled effect of physical activity on the change of cognition throughout intervention duration and post-intervention cognition scores by standardized mean difference (SMD) and its 95% confidence interval (CI). We used funnel plots to evaluate the publication bias, I2 statistic to evaluate the heterogeneity and did subgroup analysis stratified by sample size and follow-up time. RESULTS Five eligible studies involving 2581 patients with diabetes were included. The pooled effect of physical activity on cognition improvement in patients with diabetes was significant (SMD = 0.98, 95% CI: 0.34-1.62), while the effect on post-intervention cognition scores was not significant (SMD = 0.35, 95% CI: -0.04-0.73). In the subgroup analysis, the pooled effect was significantly higher in studies of follow-up time less than 1 year (SMD = 2.14, 95% CI: 1.63-2.64), while observing no significant effect in studies of follow-up time over 1 year (SMD = 0.10, 95% CI: -0.11-0.32). CONCLUSIONS Physical activity is beneficial to improving cognition in patients with diabetes. However, the long-term effect needs to be explored in future studies.
Collapse
Affiliation(s)
- Ruitong Wang
- School of Public HealthPeking UniversityBeijingChina
| | - Wenxin Yan
- School of Public HealthPeking UniversityBeijingChina
| | - Min Du
- School of Public HealthPeking UniversityBeijingChina
| | - Liyuan Tao
- Research Center of Clinical EpidemiologyPeking University Third HospitalBeijingChina
| | - Jue Liu
- School of Public HealthPeking UniversityBeijingChina
| |
Collapse
|
43
|
Jia Y, Liu R, Tang S, Zhang D, Wang Y, Cong L, Hou T, Ren J, Du Y. Associations of the Glycaemic Control of Diabetes with Dementia and Physical Function in Rural-Dwelling Older Chinese Adults: A Population-Based Study. Clin Interv Aging 2021; 16:1503-1513. [PMID: 34413638 PMCID: PMC8370580 DOI: 10.2147/cia.s319633] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/28/2021] [Indexed: 12/14/2022] Open
Abstract
Purpose To examine the associations of impaired fasting glucose (IFG) and glycaemic control of diabetes with dementia, global cognitive function and physical function among rural-dwelling Chinese older adults. Patients and Methods This population-based cross-sectional study included 4583 participants (age ≥65 years, 57.3% women) living in Yanlou Town, Yanggu County, western Shandong Province, China. In 2018, data were collected through interviews, clinical examinations, neuropsychological tests, and laboratory tests. Diabetes status was defined by self-reported physician-diagnosed diabetes, current use of antidiabetic agents, and fasting blood glucose tests. Global cognitive function was assessed using the Mini-Mental State Examination. Dementia was diagnosed following DSM-IV criteria, and Alzheimer's disease (AD) was diagnosed following the National Institute on Aging-Alzheimer's Association criteria. Physical function was assessed by the Short Physical Performance Battery. Data were analysed using multiple logistic and general linear regression models. Results IFG was found in 267 participants, and diabetes was diagnosed in 658 participants (257 with well-controlled diabetes, 401 with poorly controlled diabetes). Dementia was diagnosed in 166 participants (116 with AD), and physical functional impairment was found in 1973 participants. The multi-adjusted odds ratio (OR) of dementia associated with poorly controlled diabetes (vs without IFG or diabetes) was 2.41 (95% CI 1.52-3.84), and the OR of AD associated with poorly controlled diabetes was 2.32 (1.34-4.04). In addition, the adjusted OR of physical functional impairment was 1.40 (1.06-1.85) for well-controlled diabetes and 1.69 (1.35-2.12) for poorly controlled diabetes. However, IFG was not associated with cognitive or physical function. Conclusion The glycaemic control status of diabetes patients was associated with cognitive impairment and physical functional impairment.
Collapse
Affiliation(s)
- Yanhong Jia
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China.,Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Department of Neurology, Baotou Central Hospital, Baotou, Inner Mongolia, People's Republic of China
| | - Rui Liu
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Shi Tang
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China.,Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, People's Republic of China
| | - Dongming Zhang
- Department of General Surgery, Baotou Central Hospital, Baotou, Inner Mongolia, People's Republic of China
| | - Yongxiang Wang
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China.,Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, People's Republic of China
| | - Lin Cong
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China.,Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, People's Republic of China
| | - Tingting Hou
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China.,Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, People's Republic of China
| | - Juan Ren
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Yifeng Du
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China.,Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, People's Republic of China
| |
Collapse
|
44
|
Khan T, Khan S, Akhtar M, Ali J, Najmi AK. Empagliflozin nanoparticles attenuates type2 diabetes induced cognitive impairment via oxidative stress and inflammatory pathway in high fructose diet induced hyperglycemic mice. Neurochem Int 2021; 150:105158. [PMID: 34391818 DOI: 10.1016/j.neuint.2021.105158] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 01/21/2023]
Abstract
There is snowballing evidence that type 2 diabetes (T2D) predisposes to neuropathophysiological alterations including oxidative stress and triggered inflammatory responses in brain that eventually culminates into cognitive impairment.Accumulating evidences suggest that SGLT2 inhibitor can be a promising intervention for cognitive decline in T2DM. In the present paper, the potential effects of Empagliflozin (EMPA), a SGLT2 inhibitor, against T2D induced cognitive dysfunction have been explored. The effect of EMPA on array of inflammatory mediators including Interleukin-6(IL-6), Interleukin -1β (IL-1β), and Tumour necrosis factor-α(TNF-α)), neuronal proteins including glycogen synthase kinase-3β (GSK- 3β), Phosphorylated tau (p-tau), amyloid beta (Aβ) (1-40, 1-42) and altered oxidative parameters including SOD, catalase, TBARS was determined in the high fructose diet induced hyperglycaemic mice. The obtained results were compared with EMPA nanoparticles (Nps) formulated in our laboratory and found that EMPA Nps significantly showed reduced levels of inflammatory mediators and oxidative stress. Further, decrease in levels of p-tau, Aβ (1-40) and Aβ (1-42) were also observed with EMPA nanoparticles.Thus, the study has demonstrated that EMPA Nps could be a promising therapy to alleviate the progression of cognitive decline in T2D.
Collapse
Affiliation(s)
- Tahira Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia, Hamdard, New Delhi, India
| | - Sana Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia, Hamdard, New Delhi, India
| | - Mohd Akhtar
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia, Hamdard, New Delhi, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia, Hamdard, New Delhi, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia, Hamdard, New Delhi, India.
| |
Collapse
|
45
|
Peeters G, Katelekha K, Lawlor B, Demnitz N. Sex differences in the incidence and prevalence of young-onset Alzheimer's disease: A meta-analysis. Int J Geriatr Psychiatry 2021; 37:10.1002/gps.5612. [PMID: 34386999 PMCID: PMC9290036 DOI: 10.1002/gps.5612] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 08/05/2021] [Indexed: 11/07/2022]
Abstract
OBJECTIVES The lifetime incidence of Alzheimer's disease is higher in women than in men, but it remains unclear if similar sex differences exist in young-onset Alzheimer's disease (YOAD). This systematic review test the hypothesis that women have a higher prevalence and incidence of YOAD than men. METHODS We searched Pubmed and Embase (inception to 11 June 2020) for original publications of population-based observational studies with data on the prevalence and/or incidence of YOAD, defined as a medical diagnosis of Alzheimer's disease before the age of 65 years. Data on cross-sectional and/or prospective numbers, percentages, incidences, and incidence rates (in person-years) were derived from included studies. Quality assessment was done using the Nottingham Ottawa Scale. Meta-analyses were done to test the hypothesis that women have a higher prevalence and incidence of YOAD than men. RESULTS After screening of 3252 titles, 12 articles were included. The pooled prevalence was 0.4% (confidence interval [CI] = 0.1-2.1) in women and 0.2% (CI = 0-1.2) in men (six studies, relative risk [RR] = 1.54, CI = 0.69-3.44, I2 = 38%). The pooled incidence was 0.02% (CI = 0.01-0.08) in women and 0.01% (CI = 0-0.05) in men (five studies, RR = 1.50, CI = 0.91-2.48, I2 = 0%). The incidence rates per 100,000 person-years ranged from 0 to 132 in women and from 0 to 42 in men. CONCLUSIONS Given the low prevalence and wide CIs, no firm conclusions can be drawn. Large-scale studies are required to verify that women are more likely than men to develop YOAD.
Collapse
Affiliation(s)
- Geeske Peeters
- Department of Geriatric MedicineRadboud Institute of Health SciencesRadboud University Medical CentreNijmegenThe Netherlands
- Global Brain Health InstituteTrinity College Institute of NeuroscienceTrinity College DublinDublinIreland
| | - Katerine Katelekha
- Department of Geriatric MedicineRadboud Institute of Health SciencesRadboud University Medical CentreNijmegenThe Netherlands
| | - Brian Lawlor
- Global Brain Health InstituteTrinity College Institute of NeuroscienceTrinity College DublinDublinIreland
- Memory ClinicMercer's Institute for Successful AgeingSt James's HospitalDublinIreland
| | - Naiara Demnitz
- Global Brain Health InstituteTrinity College Institute of NeuroscienceTrinity College DublinDublinIreland
- Danish Research Centre for Magnetic ResonanceCentre for Functional and Diagnostic Imaging and ResearchCopenhagen University Hospital HvidovreHvidovreDenmark
| |
Collapse
|
46
|
Zuin M, Roncon L, Passaro A, Cervellati C, Zuliani G. Metabolic syndrome and the risk of late onset Alzheimer's disease: An updated review and meta-analysis. Nutr Metab Cardiovasc Dis 2021; 31:2244-2252. [PMID: 34039508 DOI: 10.1016/j.numecd.2021.03.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 01/08/2023]
Abstract
AIMS This study aims to provide an updated systematic review and meta-analysis on the risk of Alzheimer's disease (AD) in patients with metabolic syndrome (MetS) and to analyze the contribution of each MetS component on AD onset. DATA SYNTHESIS The study was performed according to the PRISMA guideline. Data were obtained searching MEDLINE, Scopus, Web of Science, and EMBASE for studies published between January 1, 2010 and July 30, 2020, evaluating the association between MetS and AD risk. A total of 255 articles were retrieved and 6 investigations (4 prospective and 2 retrospective) met the inclusion criteria. Overall, 9.788.021 patients with a mean follow-up of 4.5 years were analyzed. The pooled analysis revealed a slight increased risk of AD in MetS (hazard ratio, HR: 1.10, 95% and confidence interval, CI: 1.05-1.15). Egger's test indicated the absence of publication bias (t = 2.095 and p = 0.104). However, while analysis based on prospective studies failed to show a significant association between MetS and AD (HR: 0.80 and 95% CI: 0.61-1.05), analysis based on retrospective studies demonstrated a significant, slight increased risk (HR:1.11 and 95% CI: 1.08-1.66). With regard to MetS components, the risk was: arterial hypertension, HR: 1.05 (95% CI: 1.04-10.6); hyperglycemia/diabetes, HR: 1.19 (95% CI: 1.18-1.99); low high-density lipoprotein cholesterol (HDL-C), HR: 1.07 (95% CI: 1.06-1.07); hypertriglyceridemia, HR: 1.06 (95% CI: 1.05-1.06); and abdominal obesity, HR: 0.84 (95% CI: 0.74-0.95). CONCLUSIONS We found a significant association between MetS and AD, mainly driven by large retrospective studies. Our data also support the association of single MetS components with AD incidence, while increased waist circumference seems to have a "protective role" probably due to reverse causality.
Collapse
Affiliation(s)
- Marco Zuin
- Department of Morphology, Surgery & Experimental Medicine, University of Ferrara, Ferrara, Italy; Department of Cardiology, Santa Maria delle Misericordia Hospital, Rovigo, Italy
| | - Loris Roncon
- Department of Cardiology, Santa Maria delle Misericordia Hospital, Rovigo, Italy
| | - Angela Passaro
- Department of Cardiology, Santa Maria delle Misericordia Hospital, Rovigo, Italy
| | - Carlo Cervellati
- Department of Morphology, Surgery & Experimental Medicine, University of Ferrara, Ferrara, Italy.
| | - Giovanni Zuliani
- Department of Morphology, Surgery & Experimental Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
47
|
Role of liraglutide in Alzheimer's disease pathology. ALZHEIMERS RESEARCH & THERAPY 2021; 13:112. [PMID: 34118986 PMCID: PMC8199799 DOI: 10.1186/s13195-021-00853-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 05/31/2021] [Indexed: 12/19/2022]
Abstract
Background The described relationship between Alzheimer’s disease (AD) and type 2 diabetes (T2D) and the fact that AD has no succesful treatment has led to the study of antidiabetic drugs that may limit or slow down AD pathology. Main body Although T2D treatment has evident limitations, options are increasing including glucagon-like peptide 1 analogs. Among these, liraglutide (LRGT) is commonly used by T2D patients to improve β cell function and suppress glucagon to restore normoglycaemia. Interestingly, LRGT also counterbalances altered brain metabolism and has anti-inflammatory properties. Previous studies have reported its capacity to reduce AD pathology, including amyloid production and deposition, tau hyperphosphorylation, or neuronal and synaptic loss in animal models of AD, accompanied by cognitive improvement. Given the beneficial effects of LRGT at central level, studies in patients have been carried out, showing modest beneficial effects. At present, the ELAD trial (Evaluating Liraglutide in Alzheimer’s Disease NCT01843075) is an ongoing phase IIb study in patients with mild AD. In this minireview, we resume the outcomes of LRGT treatment in preclinical models of AD as well as the available results in patients up to date. Conclusion The effects of LRGT on animal models show significant benefits in AD pathology and cognitive impairment. While studies in patients are limited, ongoing clinical trials will probably provide more definitive conclusions on the role of LRGT in AD patients.
Collapse
|
48
|
Zhang X, Alshakhshir N, Zhao L. Glycolytic Metabolism, Brain Resilience, and Alzheimer's Disease. Front Neurosci 2021; 15:662242. [PMID: 33994936 PMCID: PMC8113697 DOI: 10.3389/fnins.2021.662242] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of age-related dementia. Despite decades of research, the etiology and pathogenesis of AD are not well understood. Brain glucose hypometabolism has long been recognized as a prominent anomaly that occurs in the preclinical stage of AD. Recent studies suggest that glycolytic metabolism, the cytoplasmic pathway of the breakdown of glucose, may play a critical role in the development of AD. Glycolysis is essential for a variety of neural activities in the brain, including energy production, synaptic transmission, and redox homeostasis. Decreased glycolytic flux has been shown to correlate with the severity of amyloid and tau pathology in both preclinical and clinical AD patients. Moreover, increased glucose accumulation found in the brains of AD patients supports the hypothesis that glycolytic deficit may be a contributor to the development of this phenotype. Brain hyperglycemia also provides a plausible explanation for the well-documented link between AD and diabetes. Humans possess three primary variants of the apolipoprotein E (ApoE) gene - ApoE∗ϵ2, ApoE∗ϵ3, and ApoE∗ϵ4 - that confer differential susceptibility to AD. Recent findings indicate that neuronal glycolysis is significantly affected by human ApoE isoforms and glycolytic robustness may serve as a major mechanism that renders an ApoE2-bearing brain more resistant against the neurodegenerative risks for AD. In addition to AD, glycolytic dysfunction has been observed in other neurodegenerative diseases, including Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis, strengthening the concept of glycolytic dysfunction as a common pathway leading to neurodegeneration. Taken together, these advances highlight a promising translational opportunity that involves targeting glycolysis to bolster brain metabolic resilience and by such to alter the course of brain aging or disease development to prevent or reduce the risks for not only AD but also other neurodegenerative diseases.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Nadine Alshakhshir
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Liqin Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
- Neuroscience Graduate Program, University of Kansas, Lawrence, KS, United States
| |
Collapse
|
49
|
Bai A, Tao L, Huang J, Tao J, Liu J. Effects of physical activity on cognitive function among patients with diabetes in China: a nationally longitudinal study. BMC Public Health 2021; 21:481. [PMID: 33706749 PMCID: PMC7948339 DOI: 10.1186/s12889-021-10537-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/01/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We aimed to examine the effect of physical activity on different cognitive domains among patients with diabetes. METHODS We used two waves of data from the Chinese Health and Retirement Longitudinal Study (CHARLS, 2013-2015), a nationally representative dataset of Chinese population aged over 45. Total physical activity scores were calculated based on the International Physical Activity Questionnaire (IPAQ). Executive function and episodic memory were used as measures of cognitive function. We conducted lagged dependent variable models to explore the association between physical activity and cognitive function in full sample as well as two different age groups (45-65, ≥65). RESULTS 862 diabetic patients were included. We found that diabetic participants who had greater level of physical activity at baseline were associated with better episodic memory function in 2 years (p < 0.05). Moreover, physical activity was significantly associated with less decline in episodic memory in fully adjusted models, and the associations were stronger among patients aged 45-65 years (p < 0.05). No statistically significant association was found between physical activity and executive function in all age groups. CONCLUSIONS Physical activity may prevent some of the potential decline in episodic memory in diabetic patients. Clinicians and public health departments should strengthen the promotion of physical activity and develop early screening tools among diabetic participants to prevent the progression of cognitive impairment.
Collapse
Affiliation(s)
- Anying Bai
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Liyuan Tao
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, China
| | - Jia Huang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine; Fujian Key Laboratory of Rehabilitation Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jing Tao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine; Fujian Key Laboratory of Rehabilitation Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jue Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191, China.
| |
Collapse
|
50
|
Moran C, Gilsanz P, Beeri MS, Whitmer RA, Lacy ME. Sex, diabetes status and cognition: findings from the study of longevity in diabetes. BMJ Open Diabetes Res Care 2021; 9:9/1/e001646. [PMID: 33509934 PMCID: PMC7845709 DOI: 10.1136/bmjdrc-2020-001646] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/02/2020] [Accepted: 12/20/2020] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Women comprise two-thirds of people with dementia, making female sex a significant dementia risk factor. Both type 1 diabetes (T1D) and type 2 diabetes (T2D) are known dementia risk factors with an increasing global incidence. Understanding whether subtle sex differences persist in cognitive function prior to dementia in the context of diabetes may help elucidate the magnitude of sex effects on dementia risk. RESEARCH DESIGN AND METHODS We examined cross-sectional data from the Study of Longevity in Diabetes (SOLID), a prospective cohort study of members of Kaiser Permanente Northern California aged 60 years and older with T1D (n=758), T2D (n=232) and without either T1D or T2D (n=247). We used factor analysis to generate summary scores of cognitive domains and used regression analyses to examine the associations between sex and cognition adjusting for sociodemographic and cardiovascular confounders. RESULTS We included 1237 participants (630 women and 607 men) with mean age 68 years. By design, the distribution of men and women in T1D, T2D and no diabetes was similar. Women had better cognitive performance than men in global cognition (β=0.21, 95% CI 0.16 to 0.26), language (β=0.08, 95% CI 0.004 to 0.15), executive function (β=0.13, 95% CI 0.05 to 0.20), episodic verbal memory (β=0.68, 95% CI 0.59 to 0.77) and attention (β=0.20, 95% CI 0.11 to 0.28) but not in episodic visual memory (β=0.006, 95% CI -0.07 to 0.09) adjusting for age and education independent of diabetes status. We did not find an interaction between sex and diabetes status for any of the cognitive outcomes. CONCLUSIONS Women in late mid-life have better cognitive performance than men in many cognitive domains independent of the presence of T1D or T2D. Further work is required to understand whether these differences change over time or in older cohorts and to understand their relationship to subsequent dementia.
Collapse
Affiliation(s)
- Chris Moran
- Academic Unit, Peninsula Clinical School, Monash University Central Clinical School, Melbourne, Victoria, Australia
- Peninsula Health, Frankston, Victoria, Australia
| | - Paola Gilsanz
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | - Michal S Beeri
- Icahn School of Medicine at Mount Sinai, New York City, New York, USA
- Joseph Sagol Neuroscience, Sheba Medical Center, Tel Hashomer, Israel
| | - Rachel A Whitmer
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
- Department of Epidemiology, University of California Davis School of Medicine, Davis, California, USA
| | - Mary E Lacy
- Department of Epidemiology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|