1
|
Gu H, Zhang Y, Sun J, Liu L, Liu Z. Exploring the effect and mechanism of action of Jinlida granules (JLD) in the treatment of diabetes-associated cognitive impairment based on network pharmacology with experimental validation. Ann Med 2025; 57:2445181. [PMID: 39723533 DOI: 10.1080/07853890.2024.2445181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/19/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
OBJECTIVES To explore the effect and the probable mechanisms of JLD in the treatment of type 2 diabetes mellitus (T2DM) - associated cognitive impairment (TDACI). METHODS The effect of JLD in combating TDACI was assessed in T2DM model mice by conducting Morris water maze (MWM) behaviour testing. Active components and their putative targets, as well as TDACI-related targets, were collected from public databases. Protein-protein interactions (PPIs), Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses and molecular docking were then utilized to explore potential molecular network mechanisms. Finally, the main targets were verified in animal model experiments. RESULTS MWM test showed that JLD improved aspects of behaviour in T2DM model mice. JLD improved glucose intolerance, tissue insulin sensitivity, lipid metabolism and enhanced synapse-associated protein expression in hippocampus tissue. Network pharmacology revealed 185 active components, 337 targets of JLD, and 7998 TDACI related targets were obtained . PPI network analyses revealed 39 core targets. GO and KEGG analyses suggested that JLD might improve TDACI by regulating gene expression, apoptotic processes and inflammatory responses mainly via PI3K-AKT and AGE-RAGE signaling pathways. Molecular docking revealed strong binding of the main components to core targets. JLD reduced hippocampus tissue expression of the inflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-6 (IL6), core targets of treatment of TDACI. CONCLUSIONS The findings suggested that JLD has the potential to improve TDACI through multiple components, multiple targets and multiple pathways. JLD may be a promising treatment for diabetic cognitive impairment.
Collapse
Affiliation(s)
- Haiyan Gu
- Department of Hebei Provincial Key Laboratory of Basic Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
- Department of Shijiazhuang Technology Innovation Center of Precision Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
| | - Yuxin Zhang
- Department of Hebei Provincial Key Laboratory of Basic Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
- Department of Shijiazhuang Technology Innovation Center of Precision Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
| | - Jinghua Sun
- Department of Hebei Provincial Key Laboratory of Basic Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
- Department of Shijiazhuang Technology Innovation Center of Precision Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
| | - Lipeng Liu
- Department of Hebei Provincial Key Laboratory of Basic Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
- Department of Shijiazhuang Technology Innovation Center of Precision Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
| | - Zanchao Liu
- Department of Hebei Provincial Key Laboratory of Basic Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
- Department of Shijiazhuang Technology Innovation Center of Precision Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
| |
Collapse
|
2
|
Chierichetti M, Cristofani R, Crippa V, Ferrari V, Cozzi M, Casarotto E, Pramaggiore P, Cornaggia L, Patelli G, Mohamed A, Piccolella M, Galbiati M, Rusmini P, Tedesco B, Poletti A. Small heat shock protein B8: from cell functions to its involvement in diseases and potential therapeutic applications. Neural Regen Res 2025; 20:2872-2886. [PMID: 39435632 PMCID: PMC11826450 DOI: 10.4103/nrr.nrr-d-24-00517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/05/2024] [Accepted: 08/29/2024] [Indexed: 10/23/2024] Open
Abstract
Heat shock protein family B (small) member 8 (HSPB8) is a 22 kDa ubiquitously expressed protein belonging to the family of small heat shock proteins. HSPB8 is involved in various cellular mechanisms mainly related to proteotoxic stress response and in other processes such as inflammation, cell division, and migration. HSPB8 binds misfolded clients to prevent their aggregation by assisting protein refolding or degradation through chaperone-assisted selective autophagy. In line with this function, the pro-degradative activity of HSPB8 has been found protective in several neurodegenerative and neuromuscular diseases characterized by protein misfolding and aggregation. In cancer, HSPB8 has a dual role being capable of exerting either a pro- or an anti-tumoral activity depending on the pathways and factors expressed by the model of cancer under investigation. Moreover, HSPB8 exerts a protective function in different diseases by modulating the inflammatory response, which characterizes not only neurodegenerative diseases, but also other chronic or acute conditions affecting the nervous system, such as multiple sclerosis and intracerebellar hemorrhage. Of note, HSPB8 modulation may represent a therapeutic approach in other neurological conditions that develop as a secondary consequence of other diseases. This is the case of cognitive impairment related to diabetes mellitus, in which HSPB8 exerts a protective activity by assuring mitochondrial homeostasis. This review aims to summarize the diverse and multiple functions of HSPB8 in different pathological conditions, focusing on the beneficial effects of its modulation. Drug-based and alternative therapeutic approaches targeting HSPB8 and its regulated pathways will be discussed, emphasizing how new strategies for cell and tissue-specific delivery represent an avenue to advance in disease treatments.
Collapse
Affiliation(s)
- Marta Chierichetti
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Riccardo Cristofani
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Valeria Crippa
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Veronica Ferrari
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Marta Cozzi
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Elena Casarotto
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Paola Pramaggiore
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Laura Cornaggia
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Guglielmo Patelli
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Ali Mohamed
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Margherita Piccolella
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Mariarita Galbiati
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Paola Rusmini
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Barbara Tedesco
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Angelo Poletti
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
3
|
Alami M, Morvaridzadeh M, El Khayari A, Boumezough K, El Fatimy R, Khalil A, Fulop T, Berrougui H. Reducing Alzheimer's Disease Risk with SGLT2 Inhibitors: From Glycemic Control to Neuroprotection. Ageing Res Rev 2025; 108:102751. [PMID: 40204129 DOI: 10.1016/j.arr.2025.102751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 04/11/2025]
Abstract
Recent research has established a strong link between metabolic abnormalities and an increased risk of dementia. In parallel, there is growing epidemiological evidence supporting the neuroprotective effects of antidiabetic medications against cognitive impairments. Among these, sodium-glucose co-transporter (SGLT2) inhibitors have emerged as pharmacological candidates with promising potential in alleviating the burden of age-related diseases, particularly neurodegenerative diseases (NDD). SGLT2 inhibitor therapies are FDA-approved medications routinely prescribed to manage diabetes. This novel class was initially developed to address cardiovascular disorders and to reduce the risk of hypoglycemia associated with insulin-secretagogue agents. It subsequently attracted growing interest for its beneficial effects on central nervous system (CNS) disorders. However, the molecular mechanisms through which these glucose-lowering therapies mitigate cognitive decline and limit the progression of certain brain degenerative diseases remain largely unexplored. Consequently, the neuroscientific community needs further studies that gather, analyze, and critically discuss the available mechanistic evidence regarding the neuroprotective effects of SGLT2 inhibitors. This review aims to critically examine the most relevant published findings, both in vitro and in vivo, as well as human studies evaluating the impact of SGLT2 inhibitors exposure on Alzheimer's disease (AD). It seeks to integrate the current understanding of their beneficial effects at the molecular level and their role in addressing the pathophysiology and neuropathology of AD. These insights will help extend our knowledge of how SGLT2 inhibitor therapies are associated with reduced risk of dementia and thus shed light on the link between diabetes and AD.
Collapse
Affiliation(s)
- Mehdi Alami
- Sultan Moulay Sliman University, Polydisciplinary Faculty, Department of Biology. Beni Mellal, Morocco; University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service. Sherbrooke, Qc, Canada
| | - Mojgan Morvaridzadeh
- University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service. Sherbrooke, Qc, Canada
| | - Abdellatif El Khayari
- Faculty of Medical Sciences, UM6P Hospitals, Mohammed VI Polytechnic University, Ben-Guerir 43150, Morocco; Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Kaoutar Boumezough
- Sultan Moulay Sliman University, Polydisciplinary Faculty, Department of Biology. Beni Mellal, Morocco; University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service. Sherbrooke, Qc, Canada
| | - Rachid El Fatimy
- Faculty of Medical Sciences, UM6P Hospitals, Mohammed VI Polytechnic University, Ben-Guerir 43150, Morocco
| | - Abdelouahed Khalil
- University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service. Sherbrooke, Qc, Canada
| | - Tamas Fulop
- University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service. Sherbrooke, Qc, Canada
| | - Hicham Berrougui
- Sultan Moulay Sliman University, Polydisciplinary Faculty, Department of Biology. Beni Mellal, Morocco; University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service. Sherbrooke, Qc, Canada
| |
Collapse
|
4
|
Sánchez-Sánchez JL, Valenzuela PL. Lifestyle interventions in older adults with type 2 diabetes mellitus: The key for healthy ageing. J Nutr Health Aging 2025; 29:100546. [PMID: 40121957 DOI: 10.1016/j.jnha.2025.100546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Affiliation(s)
- Juan Luis Sánchez-Sánchez
- Institute of Aging, Toulouse University Hospital, Toulouse, France; IHU HealthAge, Toulouse, France.
| | - Pedro L Valenzuela
- Department of Systems Biology, University of Alcalá, Madrid, Spain; GENUD Toledo Research Group, Faculty of Sport Sciences, University of Castilla-La Mancha, Toledo, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
5
|
Tang C, Hao J, Tao F, Feng Q, Song Y, Zeng B. Association of Metformin use with risk of dementia in patients with type 2 diabetes: A systematic review and meta-analysis. Diabetes Obes Metab 2025; 27:1992-2001. [PMID: 39780315 DOI: 10.1111/dom.16192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/17/2024] [Accepted: 12/28/2024] [Indexed: 01/11/2025]
Abstract
AIM There is ongoing debate concerning the association of metformin with the risk of dementia in type 2 diabetes mellitus (T2DM). This study was conducted to evaluate the impact of metformin therapy on dementia in patients with T2DM. MATERIALS AND METHODS PubMed, Embase, Cochrane Library, Web of Science and the ClinicalTrials.gov website were searched until 9 April 2024. Cohort studies investigating the effects of metformin therapy compared with other antidiabetic drugs or no therapy in T2DM were included. The hazard ratio (HR) and the 95% confidence interval (CI) were computed using the random effects model. RESULTS Twenty cohort studies (24 individual comparisons) involving 3 463 100 participants were identified. A meta-analysis revealed that people with T2DM who take metformin are linked to a lower incidence of all-cause dementia compared to non-user (n = 17, HR = 0.76, 95% CI = 0.65-0.91, p = 0.002, I2 = 98.9%) and sulfonylureas (n = 5, HR = 0.88, 95% CI = 0.85-0.90, p < 0.001, I2 = 9.7%), but not to thiazolidinedione (n = 2, HR = 0.53, 95% CI = 0.08-3.41, p = 0.503, I2 = 92.7%). Additionally, metformin showed favourable effects in non-specified T2DM (n = 19, HR = 0.75, 95% CI = 0.64-0.89), but not in newly diagnosed T2DM (n = 5, HR = 1.01, 95% CI = 0.81-1.27). CONCLUSION Metformin might correlate with a lower dementia incidence in people with T2DM. However, it is crucial to interpret these results with caution considering the high heterogeneity.
Collapse
Affiliation(s)
- Chunbian Tang
- Medical School of Tianjin University, Tianjin, China
- Department of General Medicine, Peking University Binhai Hospital (Tianjin Fifth Central Hospital), Tianjin, China
| | - Jiayi Hao
- Medical School of Tianjin University, Tianjin, China
| | - Fengran Tao
- Office of the President, Peking University Binhai Hospital (Tianjin Fifth Central Hospital), Tianjin, China
| | - Qingguo Feng
- Medical School of Tianjin University, Tianjin, China
- Department of Emergency, Peking University Binhai Hospital (Tianjin Fifth Central Hospital), Tianjin, China
| | - Ying Song
- Medical School of Tianjin University, Tianjin, China
- Department of General Medicine, Peking University Binhai Hospital (Tianjin Fifth Central Hospital), Tianjin, China
- Office of the President, Peking University Binhai Hospital (Tianjin Fifth Central Hospital), Tianjin, China
| | - Baoqi Zeng
- Department of Emergency, Peking University Binhai Hospital (Tianjin Fifth Central Hospital), Tianjin, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| |
Collapse
|
6
|
Moseholm KF, Jensen MK, Buzkova P, Aroner SA, Fitzpatrick AL, Longstreth WT, Lopez O, Siscovick DS, Kizer JR, Ix JH, Hughes TM, Hayden KM, Nomura S, Tsai MY, McClelland R, Djoussé L, Mukamal KJ. Circulating non-esterified fatty acids, risk of dementia and cognitive decline: The cardiovascular health study and multi-ethnic study of atherosclerosis. Neurobiol Aging 2025; 148:71-79. [PMID: 39951847 DOI: 10.1016/j.neurobiolaging.2025.01.009] [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: 06/28/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/16/2025]
Abstract
Circulating non-esterified fatty acids (NEFAs) have toxic effects on a variety of organs central to cardiometabolic disease and can cross the blood-brain barrier. Whether NEFAs associate with cognitive decline or dementia remains unknown. Circulating total NEFA levels were measured in 3242 participants without dementia among older adults of the Cardiovascular Health Study (CHS) and related to adjudicated dementia over 6 years (n = 456 cases) and annually assessed cognitive decline. For confirmation, we related circulating NEFAs to cognition assessed 10 years later among 4361 participants in the Multi-Ethnic Study of Atherosclerosis (MESA). In CHS participants, each SD higher NEFA levels were associated with a hazard ratio (HR) for all-cause dementia of 1.11 (95 % CI: 1.01; 1.22). Baseline NEFA levels were also associated with more rapid decline in cognition over 6 years of follow-up. In MESA, circulating NEFA measurements were associated with lower cognitive scores measured 10 years later.'
Collapse
Affiliation(s)
- Kristine F Moseholm
- Department of Public Health, Section of Epidemiology, University of Copenhagen, Denmark.
| | - Majken K Jensen
- Department of Public Health, Section of Epidemiology, University of Copenhagen, Denmark; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Petra Buzkova
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Sarah A Aroner
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - W T Longstreth
- Department of Epidemiology, University of Washington, Seattle, WA, USA; Department of Neurology, University of Washington, Seattle, WA, USA
| | - Oscar Lopez
- Departments of Neurology and Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Jorge R Kizer
- Cardiology Section, San Francisco Veterans Affairs Health Care System, and Departments of Medicine, Epidemiology, and Biostatistics, University of California, San Francisco, CA, USA
| | - Joachim H Ix
- Division of Nephrology and Hypertension, University of California, San Diego, CA, USA
| | - Timothy M Hughes
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Kathleen M Hayden
- Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Sarah Nomura
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Michael Y Tsai
- Division of Aging, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Robyn McClelland
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Luc Djoussé
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Kenneth J Mukamal
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Division of General Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
7
|
Yu Y, Hu B, Yu XW, Cui YY, Cao XY, Ni MH, Li SN, Dai P, Sun Q, Bai XY, Tong Y, Jing XR, Yang AL, Liang SR, Du LJ, Guo S, Yan LF, Gao B, Cui GB. Dysregulated brain dynamics in the visualmotor network in type 2 diabetes patients and their relationship with cognitive impairment. Brain Res Bull 2025; 224:111313. [PMID: 40112956 DOI: 10.1016/j.brainresbull.2025.111313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
OBJECTIVE Type 2 diabetes mellitus (T2DM) is a significant risk factor for mild cognitive impairment (MCI). Here, we identified a T2DM-specific effective connectivity (EC) network, the dynamic features of which could be used to distinguish T2DM patients with MCI from healthy controls (HC) and correlation with cognitive performance. METHODS Local and multicentered T2DM patients and matched HC who underwent functional magnetic resonance imaging were recruited. Their static and dynamic effective connectivity were compared. The relationships between connectome characteristics and cognitive performance were also evaluated. RESULTS The nodes of the T2DM-related static causality network included the anterior central gyrus, tail of the parahippocampal gyrus, posterior superior temporal sulcus, posterior central parietal lobe, posterior central gyrus and V5 region of the occipital lobe. The V5 region of the visual cortex was the core node. In the multicentered dataset, compared with the HC group, the T2DM with MCI group had significantly greater fractional window and mean dwell time. Fractional windows of the state, which was dominated by the interaction of the nodes from SomMot_Network, Limbic_Network, Default_Network, in the T2DM-specific network increased with poorer cognitive performance in T2DM with MCI patients. CONCLUSION Our findings provide insights into the neurobiological mechanisms of the cognitive impairment of T2DM patients from a dynamic network perspective, which may ultimately inform more targeted and effective strategies to prevent MCI.
Collapse
Affiliation(s)
- Ying Yu
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, Shaanxi 710038, China
| | - Bo Hu
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, Shaanxi 710038, China
| | - Xin-Wen Yu
- Department of Endocrinology, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, Shaanxi 710038, 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), 569 Xinsi Road, Xi'an, Shaanxi 710038, China; Shaanxi University of Chinese Medicine, Middle Section of Century Avenue, Xian yang, 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), 569 Xinsi Road, Xi'an, Shaanxi 710038, 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), 569 Xinsi Road, Xi'an, Shaanxi 710038, 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), 569 Xinsi Road, Xi'an, Shaanxi 710038, 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), 569 Xinsi Road, Xi'an, Shaanxi 710038, 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), 569 Xinsi Road, Xi'an, Shaanxi 710038, China
| | - Xiao-Yan Bai
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, Shaanxi 710038, China; Shaanxi University of Chinese Medicine, Middle Section of Century Avenue, Xian yang, Shaanxi, China
| | - Yao Tong
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, Shaanxi 710038, China
| | - Xiao-Rui Jing
- Department of Endocrinology, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, Shaanxi 710038, China
| | - Ai-Li Yang
- Department of Endocrinology, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, Shaanxi 710038, China
| | - Sheng-Ru Liang
- Department of Endocrinology, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, Shaanxi 710038, 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), 569 Xinsi Road, Xi'an, Shaanxi 710038, China
| | - Shuo Guo
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, Shaanxi 710038, 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), 569 Xinsi Road, Xi'an, Shaanxi 710038, China.
| | - Bin Gao
- Department of Endocrinology, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, Shaanxi 710038, 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), 569 Xinsi Road, Xi'an, Shaanxi 710038, China; Shaanxi University of Chinese Medicine, Middle Section of Century Avenue, Xian yang, Shaanxi, China.
| |
Collapse
|
8
|
Espinoza SE, Broder JC, Wolfe R, Ernst ME, Shah RC, Orchard SG, Woods RL, Ryan J, Murray A. Frailty incidence by diabetes treatment regimens in older adults with diabetes mellitus in the ASPirin in Reducing Events in the Elderly Study. GeroScience 2025:10.1007/s11357-025-01598-6. [PMID: 40097879 DOI: 10.1007/s11357-025-01598-6] [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: 08/04/2024] [Accepted: 02/28/2025] [Indexed: 03/19/2025] Open
Abstract
Diabetes mellitus is a major risk factor for frailty in older adults, and studies suggest that frailty risk may differ by diabetes treatment regimen. To investigate the association between diabetes medication use and frailty, we conducted an observational cohort analysis of older adults with diabetes enrolled in the ASPirin in Reducing Events in the Elderly (ASPREE) study. Diabetes at baseline (N = 2045) was defined as self-reported diabetes, fasting blood glucose levels > 125 mg/dL, or use of diabetes medication. Diabetes medication exposure at baseline was categorized as use of metformin only (monotherapy) (N = 545), metformin combined with other diabetes medications (N = 420), other diabetes medications only (N = 200), or no diabetes medications (N = 880). Frailty was defined using a modified Fried frailty phenotype (presence of ≥ 3 of 5 criteria) and a deficit accumulation frailty index (FI, score > 0.21/1.00). Mixed effects ordinal logistic regression models revealed the odds of frailty at baseline were highest for the other diabetes medications only group, but this difference remained consistent over follow-up. After adjustment for covariates, including baseline pre-frailty, no differences in the rates of Fried or FI frailty were observed among the diabetes medication exposure groups. These findings suggest that diabetes medication exposure in older adults with diabetes does not directly impact frailty risk.
Collapse
Affiliation(s)
- Sara E Espinoza
- Center for Translational Geroscience, Diabetes and Aging Center, Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Suite B113, Los Angeles, CA, 90048, USA.
| | - Jonathan C Broder
- School of Public Health and Preventive Medicine, Monash University, 553 St Kilda Road, Melbourne, VIC, 3004, Australia
| | - Rory Wolfe
- School of Public Health and Preventive Medicine, Monash University, 553 St Kilda Road, Melbourne, VIC, 3004, Australia
| | - Michael E Ernst
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Iowa, Iowa City, IA, USA
- Department of Family Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Raj C Shah
- Department of Family and Preventive Medicine, Rush University, Chicago, IL, USA
- Rush Alzheimer's Disease Center, Rush University, Chicago, IL, USA
| | - Suzanne G Orchard
- School of Public Health and Preventive Medicine, Monash University, 553 St Kilda Road, Melbourne, VIC, 3004, Australia
| | - Robyn L Woods
- School of Public Health and Preventive Medicine, Monash University, 553 St Kilda Road, Melbourne, VIC, 3004, Australia
| | - Joanne Ryan
- School of Public Health and Preventive Medicine, Monash University, 553 St Kilda Road, Melbourne, VIC, 3004, Australia
| | - Anne Murray
- Berman Center for Outcomes & Clinical Research, Hennepin Healthcare Research Institute, Minneapolis, MN, USA
- Department of Medicine, Geriatrics Division, Hennepin Healthcare, Minneapolis, MN, USA
| |
Collapse
|
9
|
Jiao Y, Zhang X, Duan L, Cheng R, Yang N, Peng Z, Li B, Xu L, Chen W, Chen J, Liu Y, Yan H. Association of plasma zinc and copper levels with mild cognitive impairment in patients with type 2 diabetes. Front Nutr 2025; 12:1532080. [PMID: 40144573 PMCID: PMC11936807 DOI: 10.3389/fnut.2025.1532080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/27/2025] [Indexed: 03/28/2025] Open
Abstract
Background Type 2 diabetes mellitus (T2DM) is a significant risk factor for cognitive impairment. Zinc deficiency contributes to T2DM development, while copper may exacerbate diabetes through prooxidant mechanisms. Higher zinc levels may protect against copper toxicity. This study investigates the association of plasma zinc and copper levels with mild cognitive impairment (MCI) in T2DM patients. Methods T2DM patients admitted to Tongji Hospital from 2012 to 2018 were classified into MCI (n = 136) and control (n = 136) groups, matched by age (± 3 years) and gender. Conditional logistic regression was used to assess the associations between plasma zinc, copper levels and MCI. A generalized additive model (GAM) evaluated the dose-response relationship between plasma zinc, copper levels and Mini-Mental State Examination (MMSE) scores. Results The median of plasma metal levels in MCI and control groups were 831.31 μg/L and 936.29 μg/L for zinc, 932.07 μg/L and 860.47 μg/L for copper, and 0.91 and 1.11 for the zinc-to-copper (Zn/Cu) ratio. Compared to participants in the lowest tertile, the multivariable-adjusted odds ratios with 95% confidence intervals (CI) for MCI in the highest tertile were 0.33 (0.13, 0.79) for zinc, 3.56 (1.42, 8.94) for copper, and 0.37 (0.15, 0.93) for the Zn/Cu ratio. Plasma Aβ40 levels were significantly lower (p = 0.009) and plasma Aβ42/40 levels were significantly higher (p = 0.008) in MCI group compared with those in control group. Zinc concentration was positively associated with Aβ42. For per SD (327.71 μg/L) increase in plasma zinc levels, the percent change (95% CI) of Aβ42 were 2.90 (0.85, 4.99). Conclusion Higher plasma zinc levels and higher Zn/Cu ratio were associated with lower odds of MCI in T2DM patients, while higher copper levels increased the risk of MCI. This study provides insights on plasma zinc, copper, and Zn/Cu ratio and Aβ of MCI, further studies are needed to clarify the underlying mechanisms for novel therapies that could prevent or cure multiple T2DM-related cognitive impairments.
Collapse
Affiliation(s)
- Yang Jiao
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Zhang
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lian Duan
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruijie Cheng
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ning Yang
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhao Peng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ben Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Changsha Institute for Food and Drug Control, Changsha, China
| | - Lu Xu
- Xiangyang Public Inspection and Testing Center, Xiangyang, China
| | - Wenwen Chen
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingrong Chen
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanchao Liu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Yan
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Xu Z, Zhou R, Zhou X, Zhang Z, Li Q, Wang G. The current state and development trends of frailty research in diabetic patients: a bibliometric analysis. Front Med (Lausanne) 2025; 12:1529218. [PMID: 40134912 PMCID: PMC11933048 DOI: 10.3389/fmed.2025.1529218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 02/26/2025] [Indexed: 03/27/2025] Open
Abstract
Background Diabetes mellitus is a global public health issue, often leading to organ damage, complications, and disabilities. Frailty is an age-related syndrome characterized by reduced physiological reserve and increased vulnerability to stressors, significantly affecting the prognosis of older diabetic patients. The prevalence of frailty is notably higher in older adults with diabetes than in those without. Therefore, a bibliometric analysis of research on diabetes-related frailty can provide deeper insights into the current state of this field and inform future research directions. Methods This study retrieved English-language publications on diabetes-related frailty from the Web of Science Core Collection (WOS) database, covering the period from 2005 to 2023. A total of 403 articles were included in the analysis. Statistical analysis and data visualization were conducted using Microsoft Excel, R Studio, VOS viewer, and Cite Space 6.3.R1. The analysis emphasized journals, authors, keywords, country collaborations, institutional collaborations, and references to elucidate trends and knowledge structures within the field of diabetes-related frailty research. Results The number of publications on diabetes-related frailty has been steadily increasing each year, with research predominantly focused in developed countries, particularly the United States and Europe. The University of London has emerged as the institution with the highest volume of publications, while Alan J. Sinclair has been recognized as a significant contributor to this field. Key research hotspots include the complications associated with diabetes-related frailty, epidemiology, and quality of life. Additionally, a timeline analysis of references suggests that diabetic nephropathy is currently at the forefront of research in this area. Conclusion This comprehensive bibliometric analysis of diabetes-related frailty research underscores the necessity for improved international collaboration to further investigate the mechanisms underlying diabetes-related frailty and to devise more effective prevention and treatment strategies. Future research should emphasize the relationship between diabetic nephropathy and frailty, as well as the development of personalized intervention programs tailored for frail diabetic patients.
Collapse
Affiliation(s)
- Ziqi Xu
- School of Nursing, Xinxiang Medical University, Xinxiang, China
- The First People's Hospital of Shangqiu City, Shangqiu, China
| | - Rui Zhou
- School of Nursing, Xinxiang Medical University, Xinxiang, China
| | - Xinran Zhou
- School of Nursing, Xinxiang Medical University, Xinxiang, China
| | - Zhengyan Zhang
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Qiong Li
- School of Nursing, Xinxiang Medical University, Xinxiang, China
| | - Guodong Wang
- School of Nursing, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
11
|
Huang H, Zhao T, Ma W. Omega-3 polyunsaturated fatty acids attenuates cognitive impairment via the gut-brain axis in diabetes-associated cognitive dysfunction rats. Brain Behav Immun 2025; 127:147-169. [PMID: 40068791 DOI: 10.1016/j.bbi.2025.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 02/11/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
Diabetes-related cognitive dysfunction (DACD) is a comorbidity of type 2 diabetes that has a negative effect on patients' quality of life. Research has indicated that disruption of the gut microbiota (GM) may be linked to dementia with altered cognitive performance. Conversely, omega-3 polyunsaturated fatty acids (n-3 PUFAs) may reverse DACD. The present study aimed to assess the effects of an n-3 PUFA intervention and fecal microbiota transplantation (FMT) on high-fat and streptozotocin-induced DACD model rats. In DACD rats, n-3 PUFA treatment restored fasting blood glucose (FBG) levels and cognitive function, increased the expression of anti-inflammatory cytokines and downregulated the expression of proinflammatory cytokines in the cortex and colon. Additionally, the expression of the postsynaptic density protein-95 mRNA and protein varied with n-3 PUFA treatment. Treatment with n-3 PUFAs also increased the expression of tight junction proteins. Beneficial and short-chain fatty acid-producing bacteria were more abundant when rats were exposed to n-3 PUFAs. After FMT from the rats with DACD symptoms that were improved by the n-3 PUFA dietary intervention into another batch of DACD rats, we observed recovery in recipient DACD rats. These results indicated that the alleviation of DACD symptoms by n-3 PUFAs was attributed to gut microbiota remodeling.
Collapse
Affiliation(s)
- Hongying Huang
- School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China; Nanchang Institute of Disease Control and Prevention, China Railway Nanchang Bureau Group Co., Ltd., Nanchang, 330003, People's Republic of China
| | - Tong Zhao
- School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China
| | - Weiwei Ma
- School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China.
| |
Collapse
|
12
|
Sohn M, Park YH, Lim S. Effects of choline alfoscerate on cognitive function and quality of life in type 2 diabetes: A double-blind, randomized, placebo-controlled trial. Diabetes Obes Metab 2025; 27:1350-1358. [PMID: 39703111 DOI: 10.1111/dom.16131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/24/2024] [Accepted: 11/30/2024] [Indexed: 12/21/2024]
Abstract
AIMS This study evaluated the effects of choline alfoscerate on cognitive function and quality of life in T2DM patients with mild cognitive decrements. MATERIALS AND METHODS In a double-blind, randomized, placebo-controlled trial, we recruited 36 individuals with T2DM and mild cognitive impairment which was assessed by the Mini-Mental State Examination (MMSE) score of 25-28, and randomly assigned them to receive either 1200 mg/day of choline alfoscerate or a placebo. Four additional questionnaires-the 36-Item Short Form Survey, the modified Informant Questionnaire on Cognitive Decline in the Elderly, the Korean version of Activities of Daily Living, and the Patient Health Questionnaire-were investigated at 6 and 12 months and analysed via mixed-effects models for repeated measures. RESULTS The mean age of study participants was 71.8 ± 5.3 years and 69.4% women. Six-month treatment with choline alfoscerate resulted in a non-significant increase in the MMSE score from 26.2 ± 1.3 to 26.9 ± 2.0, whereas the placebo group showed a non-significant decline from 26.6 ± 1.3 to 25.9 ± 2.3, resulting in a mean difference of +1.4 between the two groups (p = 0.059). At 12 months, the mean difference increased to +1.7 with statistical significance (p < 0.001). Physical health, as measured by the SF-36 survey, was significantly better in the choline alfoscerate group than in the placebo group. CONCLUSIONS Choline alfoscerate 1200 mg once daily treatment showed marginal improvement in cognitive function in T2DM patients with mild cognitive impairment at 6 months but leading to significance at 12 months compared to placebo, suggesting its potential as an adjunct therapy for managing early cognitive decline.
Collapse
Affiliation(s)
- Minji Sohn
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Young Ho Park
- Department of Neurology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Soo Lim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| |
Collapse
|
13
|
Cai Y, Wang X, Chen X, Liu S, Cheng L, Kang Y, Lin F. Lactobacillus casei Zhang prevents hippocampal atrophy and cognitive impairment in rats with type 2 diabetes by regulating blood glucose levels. Brain Res 2025; 1850:149407. [PMID: 39706238 DOI: 10.1016/j.brainres.2024.149407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
PURPOSE Lactobacillus casei Zhang (LCZ) has health benefits, such as the ability to improve blood glucose levels in individuals with type 2 diabetes mellitus (T2DM). However, little is known about the effects of LCZ on brain structural plasticity and cognitive function in T2DM. The aims of this study were to determine whether LCZ can prevent and alleviate brain damage and memory impairment in T2DM, and to understand the mechanisms underlying the effects of LCZ in T2DM. METHODS Forty-one male Sprague-Dawley rats were divided into the saline control (CON, n = 14), T2DM (n = 14) and T2DM + LCZ (n = 13) groups. Magnetic resonance imaging (MRI) was used to evaluate alterations in brain structure among these three groups. The novel object recognition and Y-maze tests were conductedto assess cognitive function. Histological and immunohistochemical analysis, including Nissl staining, Golgi-Cox staining and glial fibrillary acidic protein immunostaining, were performed to explore the pathophysiological mechanisms underlying brain structural changes. RESULTS T2DM rats presented hyperglycemia, cognitive decline, hippocampal atrophy, and damage to hippocampal neurons and astrocytes. Compared with those in the T2DM groups, rats in the T2DM + LCZ group presented lower blood glucose levels, better cognitive function, a larger hippocampal volume, and more normal hippocampal neurons and astrocytes. There was no significant difference in these metrics between rats in the T2DM + LCZ and CON groups. CONCLUSION Hyperglycemia-induced damage to hippocampal neurons and astrocytes may lead to hippocampal atrophy and cognitive dysfunction in T2DM. LCZ can effectively prevent this damage by regulating blood glucose levels, preventing brain atrophy and cognitive impairment in T2DM rats. These findings provide a scientific basis for the clinical application of LCZ.
Collapse
Affiliation(s)
- Yue Cai
- National Center for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuxia Wang
- National Center for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xi Chen
- National Center for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Sijie Liu
- National Center for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Linlin Cheng
- National Center for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yan Kang
- National Center for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Fuchun Lin
- National Center for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
14
|
Cheng M, Qiao Y, Yang B, He M. Related factors of cognitive frailty among older adults with type 2 diabetes: a cross-sectional study. Psychogeriatrics 2025; 25:e70004. [PMID: 39935280 DOI: 10.1111/psyg.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 01/03/2025] [Accepted: 01/10/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND The correlation between diabetes and cognitive frailty is still controversial. The purpose of this study was to explore the related factors of cognitive frailty in older adults with type 2 diabetes, and to provide new ideas for realising healthy ageing of diabetic patients. METHODS The survey was conducted at Mianyang Central Hospital affiliated to University of Electronic Science and Technology of China from January 15 to June 30, 2024. The subjects were older adults with type 2 diabetes aged 60 years or older. Cognitive frailty was assessed using Fried's phenotype and Mini-Mental State Examination. Statistical analysis and plotting were conducted using SPSS 25.0 and R 4.3.3. Least absolute shrinkage and selection operator (LASSO) regression was used to reduce the dimensionality of 30 clinical features collected, and the unimportant factors were initially screened out. The important factors with non-zero coefficients identified by LASSO regression were included in a multivariate logistic regression analysis to explore the factors influencing cognitive frailty in older diabetic patients. RESULTS This study included 301 older persons with type 2 diabetes. Among them, 91 (30.2%) cases of cognitive frailty occurred. Related factors for cognitive frailty in older adults with type 2 diabetes include: age (odds ratio (OR) = 6.417, 95% CI: 1.882-21.876, P = 0.003), hypomnesia (OR = 2,985, 95%CI: 1.143-7.797, P = 0.026), depression (OR = 9.926, 95%CI: 4.117-23.934, P < 0.001), diabetic retinopathy (OR = 6.489, 95%CI: 1.969-21.384, P = 0.002), history of diabetic ketoacidosis (OR = 12.024, 95%CI: 1.724-83.872, P = 0.012). CONCLUSION The prevalence of cognitive frailty in older persons with type 2 diabetes mellitus was higher. It was closely related to age, hypomnesia, depression, diabetic retinopathy and history of diabetic ketoacidosis. Early detection of the risk and effective intervention can reduce the incidence of adverse events and improve the quality of life of patients.
Collapse
Affiliation(s)
- Min Cheng
- School of Nursing, North Sichuan Medical University, Nanchong, China
- Nursing Department of Mianyang, Central Hospital/School of Medicine Affiliated to University of Electronic Science and Technology of China, Mianyang, China
| | - Yan Qiao
- Nursing Department of Mianyang, Central Hospital/School of Medicine Affiliated to University of Electronic Science and Technology of China, Mianyang, China
| | - Bailin Yang
- Nursing Department of Mianyang, Central Hospital/School of Medicine Affiliated to University of Electronic Science and Technology of China, Mianyang, China
| | - Mei He
- Nursing Department of Mianyang, Central Hospital/School of Medicine Affiliated to University of Electronic Science and Technology of China, Mianyang, China
| |
Collapse
|
15
|
Ramakrishan P, Rajangam J, Mahinoor SS, Bisht S, Mekala S, Upadhyay DK, Solomon VR, Sabarees G, Pelluri R. Unveiling the mTOR pathway modulation by SGLT2 inhibitors: a novel approach to Alzheimer's disease in type 2 diabetes. Metab Brain Dis 2025; 40:132. [PMID: 40009301 DOI: 10.1007/s11011-025-01555-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 02/07/2025] [Indexed: 02/27/2025]
Abstract
Alzheimer's disease (AD) is a neurological condition causing cognitive deterioration, leading to severe consequences. As the global prevalence of AD increases, new treatment approaches are needed to supplement current conventional therapies, as traditional treatments are not meeting the increasing demand for alternative treatments. It is increasingly evident that treating metabolic disorders like diabetes mellitus, obesity, and AD by blocking mechanistic target of rapamycin (mTOR) signalling is advantageous. Chronic mTOR activation may cause AD's metabolic, lysosomal, and mitochondrial dysfunction, tau hyperphosphorylation, amyloid plaque development, and disruption of the blood-brain barrier through endothelial cell malfunction. Chronic glucose loss through sodium-glucose transporter 2 (SGLT2) inhibitions can restore mTOR cycling, potentially halting or slowing AD pathogenesis. Chronic activation of mTOR is implicated in pathophysiological aspects of AD, such as metabolic dysfunction, tau hyperphosphorylation, amyloid plaque formation, and disruption of the blood-brain barrier. SGLT-2 inhibitors, commonly used in treating Type 2 Diabetes, have been shown to reduce mTOR activation and restore circadian regularity, a new finding in cognitive decline and metabolic disorders. Conversely, SGLT2 inhibitors decrease oxidative damage, inflammation, insulin signaling pathways, and proliferation of endothelial cells to enhance vascular tone, flexibility, and contractility. Along with reducing the formation of plaque containing amyloid and improving brain function, neural plasticity, acetylcholinesterase (AChE) activity, damage to the brain, and cognitive decline, they also regulate the mTOR pathway in the brain. Thus, repurposing SGLT-2 inhibitors, primarily used in diabetes treatment, presents a promising avenue for changing the way that AD is managed. The purpose of this review was to focus on the mTOR signalling cascade of SGLT 2 inhibitors to AD management in Type 2 Diabetes mellitus.
Collapse
Affiliation(s)
- Prakash Ramakrishan
- Crescent School of Pharmacy, B.S. Abdur Rahman Crescent Institute of Science & Technology-BSACIST University, Chennai, 600048, India
| | - Jayaraman Rajangam
- Shri Venkateshwara College of Pharmacy, Ariyur, Pondicherry, 605102, India.
| | - Shaheedha Shabudeen Mahinoor
- Crescent School of Pharmacy, B.S.Abdur Rahman Crescent Institute of Science & Technology-BSACIST University, Chennai, 600048, India
| | - Shradha Bisht
- College of Pharmacy, Shivalik Campus, Dehradun, Uttarakhand, 248197, India
| | - Sabareesh Mekala
- Department of Pharmaceutical Sciences, School of Biotechnology and Pharmaceutical Sciences, Vignan's Foundation for Science, Technology and Research, Vadlamudi, Guntur, 522213, India
| | - Dinesh Kumar Upadhyay
- School of Pharmaceutical Sciences, Jaipur National University, Jaipur, 302017, India
| | - Viswas Raja Solomon
- Medicinal Chemistry Research Laboratory, MNR College of Pharmacy, Sangareddy, 502294, India
| | | | - Ranakishor Pelluri
- Department of Pharmacy, KL College of Pharmacy, Koneru Lakshmaiah Education Foundation (Deemed to Be University), Vaddeswaram, Guntur, 522302, India
| |
Collapse
|
16
|
Sonnino R, Ciccarelli G, Moffa S, Soldovieri L, Di Giuseppe G, Brunetti M, Cinti F, Di Piazza E, Gasbarrini A, Nista EC, Pontecorvi A, Giaccari A, Mezza T. Exploring nutraceutical approaches linking metabolic syndrome and cognitive impairment. iScience 2025; 28:111848. [PMID: 40008362 PMCID: PMC11850164 DOI: 10.1016/j.isci.2025.111848] [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] [Indexed: 02/27/2025] Open
Abstract
Metabolic syndrome (MetS) and mild cognitive impairment (MCI) are interconnected conditions sharing common pathological pathways, such as inflammation and oxidative stress, leading to the concept of "metabolic-cognitive syndrome." This highlights their mutual influence and potential overlapping therapeutic strategies. Although lifestyle modifications remain essential, nutraceutical supplementation has emerged as a promising adjunct for the prevention and management of these preclinical conditions. This review examines clinical and translational evidence on commonly used nutraceuticals targeting shared pathophysiological mechanisms of MetS and MCI. By addressing inflammation, oxidative stress, and metabolic dysfunction, these supplements may offer a valuable approach to mitigating the progression and consequences of both conditions. Understanding their efficacy could provide practical tools to complement lifestyle changes, offering a more comprehensive strategy for managing metabolic-cognitive syndrome.
Collapse
Affiliation(s)
- Rebecca Sonnino
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Center for Endocrine and Metabolic Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Gea Ciccarelli
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Center for Endocrine and Metabolic Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Simona Moffa
- Center for Endocrine and Metabolic Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Laura Soldovieri
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Center for Endocrine and Metabolic Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Gianfranco Di Giuseppe
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Center for Endocrine and Metabolic Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Michela Brunetti
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Center for Endocrine and Metabolic Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Francesca Cinti
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Center for Endocrine and Metabolic Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Eleonora Di Piazza
- Center for Endocrine and Metabolic Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Pancreas Unit, CEMAD Digestive Diseases Center, Internal Medicine and Gastroenterology Unit, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Enrico C. Nista
- Pancreas Unit, CEMAD Digestive Diseases Center, Internal Medicine and Gastroenterology Unit, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Alfredo Pontecorvi
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Giaccari
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Center for Endocrine and Metabolic Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Teresa Mezza
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Pancreas Unit, CEMAD Digestive Diseases Center, Internal Medicine and Gastroenterology Unit, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| |
Collapse
|
17
|
Liu S, Wan H, Nie S, Cao H, Liu L, Liang H, Xu H, Liu B, Chen C, Liu H, Yang Q, Li H, Kong Y, Li G, Wan Q, Zha Y, Hu Y, Xu G, Shi Y, Zhou Y, Su G, Tang Y, Gong M, Guo A, Weng J, Wu H, Hou FF, Shen J. Dipeptidyl Peptidase 4 Inhibitors vs Metformin for New-onset Dementia: A Propensity Score-matched Cohort Study. J Clin Endocrinol Metab 2025; 110:e650-e659. [PMID: 38652239 DOI: 10.1210/clinem/dgae281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/13/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Hypoglycemic pharmacotherapy interventions for alleviating the risk of dementia remain controversial, particularly regarding dipeptidyl peptidase 4 (DPP4) inhibitors vs metformin. Our objective was to investigate whether the initiation of DPP4 inhibitors, as opposed to metformin, was linked to a reduced risk of dementia. METHODS We included individuals with type 2 diabetes over 40 years old who were new users of DPP4 inhibitors or metformin in the Chinese Renal Disease Data System database between 2009 and 2020. The study employed Kaplan-Meier and Cox regression for survival analysis and the Fine and Gray model for the competing risk of death. RESULTS Following a 1:1 propensity score matching, the analysis included 3626 DPP4 inhibitor new users and an equal number of metformin new users. After adjusting for potential confounders, the utilization of DPP4 inhibitors was associated with a decreased risk of all-cause dementia compared to metformin [hazard ratio (HR) 0.63, 95% confidence interval (CI) 0.45-0.89]. Subgroup analysis revealed that the utilization of DPP4 inhibitors was associated with a reduced incidence of dementia in individuals who initiated drug therapy at the age of 60 years or older (HR 0.69, 95% CI 0.48-0.98), those without baseline macrovascular complications (HR 0.62, 95% CI 0.41-0.96), and those without baseline microvascular complications (HR 0.67, 95% CI 0.47-0.98). CONCLUSION In this real-world study, we found that DPP4 inhibitors presented an association with a lower risk of dementia in individuals with type 2 diabetes than metformin, particularly in older people and those without diabetes-related comorbidities.
Collapse
Affiliation(s)
- Siyang Liu
- Institute and Department of Endocrinology and Metabolism, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan 528308, Guangdong, China
| | - Heng Wan
- Institute and Department of Endocrinology and Metabolism, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan 528308, Guangdong, China
| | - Sheng Nie
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Huanyi Cao
- Department of Endocrinology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510000, Guangdong, China
| | - Lan Liu
- Institute and Department of Endocrinology and Metabolism, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan 528308, Guangdong, China
| | - Hua Liang
- Institute and Department of Endocrinology and Metabolism, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan 528308, Guangdong, China
| | - Hong Xu
- Department of Nephrology, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Bicheng Liu
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing 210009, China
| | - Chunbo Chen
- Department of Critical Care Medicine, Maoming People's Hospital, Maoming 525000, China
| | - Huafeng Liu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Qiongqiong Yang
- Department of Nephrology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510235, China
| | - Hua Li
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yaozhong Kong
- Department of Nephrology, The First People's Hospital of Foshan, Foshan 528000, Guangdong, China
| | - Guisen Li
- Renal Department and Institute of Nephrology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Clinical Research Center for Kidney Diseases, Chengdu 610072, China
| | - Qijun Wan
- The Second People's Hospital of Shenzhen, Shenzhen University, Shenzhen 518035, China
| | - Yan Zha
- Guizhou Provincial People's Hospital, Guizhou University, Guiyang 550002, China
| | - Ying Hu
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 313000, China
| | - Gang Xu
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yongjun Shi
- Huizhou Municipal Central Hospital, Sun Yat-Sen University, Huizhou 516003, China
| | - Yilun Zhou
- Department of Nephrology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Guobin Su
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital, The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Ying Tang
- The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Mengchun Gong
- Institute of Health Management, Southern Medical University, Guangzhou 510515, China
- DHC Technologies, Beijing 100000, China
| | - Aixin Guo
- DHC Technologies, Beijing 100000, China
| | - Jianping Weng
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Hongjiang Wu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Fan Fan Hou
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jie Shen
- Institute and Department of Endocrinology and Metabolism, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan 528308, Guangdong, China
| |
Collapse
|
18
|
Tian Y, Jing G, Yin R, Ma M, Cao W, Zhang M. Neuroprotective effects of traditional Chinese medicine Naofucong on diabetic cognitive impairment: Mechanisms involving insulin-degrading enzyme-mediated degradation of Amyloid-β and inhibition of ERK/JNK/p38 MAPK signaling pathway. Brain Res 2025; 1849:149365. [PMID: 39617284 DOI: 10.1016/j.brainres.2024.149365] [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: 10/09/2024] [Revised: 11/12/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
The increasing prevalence of diabetes and its related cognitive impairments is a significant public health concern. With limited clinical treatment options and an incomplete understanding of the underlying mechanisms, traditional Chinese medicine (TCM) Naofucong is proposed as a potential neuroprotective agent against diabetic cognitive impairment (DCI). This study aims to investigate the therapeutic mechanisms of Naofucong in DCI. We hypothesize that Naofucong may improve cognitive function in diabetic rats by modulating the extracellular regulated protein kinases (ERK)/c-Jun N-terminal kinase (JNK)/p38 mitogen-activated protein kinases (MAPK) signaling pathway, enhancing insulin-degrading enzyme (IDE) expression, reducing amyloid-beta (Aβ) deposition, decreasing phosphorylated Tau (p-Tau) levels, and alleviating oxidative stress. Diabetes was induced in specific-pathogen-free male Sprague-Dawley rats using streptozotocin, and the rats were treated with oral Naofucong for 12 weeks. We assessed cognitive function and measured neuronal damage, oxidative stress injury, and the expression levels of IDE, Aβ, amyloid precursor protein (APP), p-Tau, and components of the ERK/JNK/p38 MAPK pathway. Diabetic rats showed significant declines in cognitive function, neuronal damage, oxidative stress, low IDE expression, Aβ accumulation, high APP expression, abnormal Tau phosphorylation, and overactivation of the ERK/JNK/p38 MAPK pathway. Naofucong treatment significantly reversed these symptoms. Our findings suggest that Naofucong improves cognitive impairment in diabetic rats by inhibiting the ERK/JNK/p38 MAPK pathway, upregulating IDE, reducing Aβ deposition, suppressing APP and p-Tau expression, and alleviating neuronal damage and oxidative stress. This research provides a reference for the clinical prevention and treatment of DCI using TCM Naofucong.
Collapse
Affiliation(s)
- Yue Tian
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Guangchan Jing
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Ruiying Yin
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Mei Ma
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Weiwei Cao
- Beijing HFK Bioscience Co., LTD, Beijing 102200, China.
| | - Mengren Zhang
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
19
|
Guo X, Wang F, Zheng M, Li L, Li L, Wang J, Miao S, Ma S, Shi X. Network pharmacology and molecular docking to study the potential molecular mechanism of Qi Fu Yin for diabetic encephalopathy. J Biomol Struct Dyn 2025; 43:917-931. [PMID: 38047625 DOI: 10.1080/07391102.2023.2289038] [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: 06/06/2023] [Accepted: 08/29/2023] [Indexed: 12/05/2023]
Abstract
Diabetic encephalopathy is a chronic complication of diabetes that lacks an optimized treatment strategy. The present study sought to elucidate the potential molecular mechanism of Qi Fu Yin in improving diabetic encephalopathy through network pharmacology. The active components and target information of Qi Fu Yin were obtained from the TCMSP and Swiss target databases, while the target information of diabetic encephalopathy was sourced from Gene cards, OMIM, and Pharm Gkb databases. Enrichment analyses of KEGG and GO were conducted utilizing drug-disease common targets, while protein-protein interactions were predicted through the utilization of the STRING database platform. Subsequently, molecular docking was executed via Auto Dock Vina to authenticate the interaction between core components and core targets. The findings revealed that Qi Fu Yin exhibited 178 common targets with diabetic encephalopathy, and the enrichment analyses demonstrated that these targets were associated with lipid and atherosclerosis, AGE-RAGE signaling pathways, and other related pathways. The findings of the molecular docking indicated a favorable binding affinity between the active components of drug and the core targets, with EGF and quercetin exhibiting the most notable docking score. Additionally, the molecular dynamics simulation corroborated this high affinity. These results suggested that the active ingredients of Qi Fu Yin, including quercetin and kaempferol, may modulated the expression of genes such as IL10, TNF, EGF, and MMP2, thereby activating the AGE-RAGE signaling pathways and potentially serving as a therapeutic intervention for diabetic encephalopathy.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Xiaodi Guo
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, P. R. China
- The College of Life Sciences, Northwest University, Xi'an, P. R. China
| | - Feiyan Wang
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, P. R. China
| | - Meiling Zheng
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, P. R. China
| | - Liang Li
- Neurosurgery, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, P. R. China
| | - Long Li
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, P. R. China
| | - Jin Wang
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, P. R. China
| | - Shan Miao
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, P. R. China
| | - Shanbo Ma
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, P. R. China
| | - Xiaopeng Shi
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, P. R. China
| |
Collapse
|
20
|
Chen L, Liu R, He X, Fang J, Zhou L, Qi Z, Tao M, Yuan H, Zhou Y. Synergistically effects of n-3 PUFA and B vitamins prevent diabetic cognitive dysfunction through promoting TET2-mediated active DNA demethylation. Clin Nutr 2025; 45:111-123. [PMID: 39798222 DOI: 10.1016/j.clnu.2025.01.002] [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: 08/22/2024] [Revised: 12/11/2024] [Accepted: 01/03/2025] [Indexed: 01/15/2025]
Abstract
Diabetic cognitive dysfunction (DCD) refers to the cognitive impairment observed in individuals with diabetes. Epidemiological studies have suggested that supplementation with n-3 polyunsaturated fatty acid (PUFA) or B vitamins may prevent the development of diabetic complications. Post hoc studies indicate a potential synergistic effect of n-3 PUFA and B vitamins in preventing cognitive impairment. However, the precise effect and underlying mechanism of this combination on DCD remain unclear. In case-control study, we compared fatty acid composition of erythrocyte membrane and serum homocysteine levels between diabetic individuals with and without DCD. We found that insufficient levels of n-3 PUFA, along with elevated serum homocysteine, significantly increase the risk of developing DCD. Treatment with a combination of fish oil, folate, and vitamin B12 improved cognitive impairment and aberrant neuronal morphology in streptozotocin-induced DCD mice. Folic acid and vitamin B12 enhanced the efficiency of exogenous docosahexaenoic acid (DHA) transportation to the brain by preventing the accumulation of homocysteine and S-adenosylhomocysteine, thereby inhibiting neuronal apoptosis in diabetic brains. Furthermore, folic acid and vitamin B12 supplementation can provide sufficient 5-methylcytosine for diabetic brains by promoting DNA methylation, while increased DHA levels maintain TET-mediated active DNA demethylation in diabetic brains through enhancing TET2 function. Overall, our study provides novel insights into molecular mechanisms underlying the synergistic preventive effects of the combined supplementation with fish oil, folic acid and vitamin B12 on DCD, suggests that combining n-3 PUFA and B vitamins could be a promising strategy for preventing DCD among individuals with diabetes.
Collapse
Affiliation(s)
- Lei Chen
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong Province, China
| | - Run Liu
- School of Public Health, Qingdao University, Qingdao, Shandong Province, China
| | - Xin He
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, China
| | - Jiacheng Fang
- School of Public Health, Qingdao University, Qingdao, Shandong Province, China
| | - Liyin Zhou
- School of Public Health, Qingdao University, Qingdao, Shandong Province, China
| | - Zhongshi Qi
- School of Public Health, Qingdao University, Qingdao, Shandong Province, China
| | - Mingzhu Tao
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Haicheng Yuan
- Department of Neurology, Affiliated Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong Province, China.
| | - Yu Zhou
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong Province, China.
| |
Collapse
|
21
|
Hou Y, Chen Z, Cheng J, Li G, Yin L, Gao J. The Mechanism and Treatment of Cognitive Dysfunction in Diabetes: A Review. Exp Clin Endocrinol Diabetes 2025; 133:64-72. [PMID: 39572247 DOI: 10.1055/a-2480-7826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
Diabetes mellitus (DM) is one of the fastest growing diseases in terms of global incidence and seriously affects cognitive function. The incidence rate of cognitive dysfunction is up to 13% in diabetes patients aged 65-74 years and reaches 24% in those aged >75 years. The mechanisms and treatments of cognitive dysfunction associated with diabetes mellitus are complicated and varied. Previous studies suggest that hyperglycemia mainly contributes to cognitive dysfunction through mechanisms involving inflammation, autophagy, the microbial-gut-brain axis, brain-derived neurotrophic factors, and insulin resistance. Antidiabetic drugs such as metformin, liraglutide, and empagliflozin and other drugs such as fingolimod and melatonin can alleviate diabetes-induced cognitive dysfunction. Self-management, intermittent fasting, and repetitive transverse magnetic stimulation can also ameliorate cognitive impairment. In this review, we discuss the mechanisms linking diabetes mellitus with cognitive dysfunction and propose a potential treatment for cognitive decline associated with diabetes mellitus.
Collapse
Affiliation(s)
- Yangbo Hou
- Department of Neurology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhen Chen
- Department of Encephalopathy, Suqian Hospital of Chinese Medicine , Nanjing University of Traditional Chinese Medicine, Suqian, China
| | - Jiwei Cheng
- Department of Neurology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guoyi Li
- Department of Neurology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu Yin
- Department of Rehabilitation, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Gao
- Department of Endocrinology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
22
|
Li X, Chen B, Liu X, Ma J. Effects of exercise on cognitive function and glycated hemoglobin A1c among patients with type 2 diabetes mellitus and cognitive impairment: A systematic review and meta-analysis. Geriatr Gerontol Int 2025; 25:148-159. [PMID: 39757130 DOI: 10.1111/ggi.15061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 11/27/2024] [Accepted: 12/19/2024] [Indexed: 01/07/2025]
Abstract
AIM To evaluate the effects of exercise versus without-exercise group on global cognitive function, executive function, glycated hemoglobin A1c (HbA1c) and fasting plasma glucose in patients with type 2 diabetes mellitus and cognitive impairment. METHODS A systematic search of Cqvip, SinoMed, Wanfang Data, CINAHL, Cochrane, Embase, Pubmed, Lippincott, Web of Science and Scopus was carried out. Randomized control trials were selected. The risk of bias was evaluated using the Cochrane Risk of Bias tool. The random-effects model was used to obtain the pooled estimates. RESULTS Eight trials with 884 participants were included. Exercise could significantly improve global cognitive function (standardized mean difference 0.82, 95% CI 0.53-1.12), executive function measured by the Trail-Marking Test part B (mean difference -20.43, 95% CI -36.20, -4.66), glycated hemoglobin A1c (%; mean difference -0.58, 95% CI -0.88, -0.29) and fasting plasma glucose (mg/dL; mean difference -17.61, 95% CI -32.67, -2.54). CONCLUSIONS Exercise can improve cognitive function, glycated hemoglobin A1c and fasting plasma glucose among type 2 diabetes mellitus patients with cognitive impairment. Additional studies with higher methodological quality are expected to draw more definite conclusions. This finding could provide a reference for clinical decision-making and guide future research initiatives. Geriatr Gerontol Int 2025; 25: 148-159.
Collapse
Affiliation(s)
- Xiaoxue Li
- School of Nursing, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Bin Chen
- School of Nursing, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Xinyuan Liu
- School of Nursing, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Jingya Ma
- School of Nursing, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| |
Collapse
|
23
|
Yang Q, Luo Q, Xia W, Yao N, Wang F, Xie C, Zhang H, He Y. Study on the mechanism on Yi-guan-jian decoction alleviating cognitive dysfunction in type 2 diabetes mellitus. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119286. [PMID: 39725366 DOI: 10.1016/j.jep.2024.119286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/05/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Yi-guan-jian decoction (YGJ) is a traditional Chinese medicine prescription commonly used for treating syndromes associated with Yin deficiency in the liver and kidney, as well as Qi-obstructed in liver. AIM OF THE STUDY YGJ has shown potential alleviating cognitive dysfunction in type 2 diabetes mellitus (T2DM). However, the precise mechanisms are not yet fully understood. This study aims to reveal the mechanism by which YGJ alleviates cognitive dysfunction in T2DM. MATERIALS AND METHODS Various doses of YGJ were administered to T2DM rats with cognitive dysfunction for 8 weeks. The positive control group received a combination of metformin and memantine. Cognitive function was assessed in T2DM rats using the Morris water maze test during treatment. Changes in gut microbiota and bile acids in the intestine were evaluated, and their interactions analyzed. Additionally, this study also evaluated the expressions of inflammatory markers (IL-1β,TNF-α, IL-16, IL-18 and CRP protein), Tau protein, neurotransmitter (5-HT and GABA), and bile acid receptor (FXR, PXR, VDR, and TGR5). RESULTS YGJ significantly alleviated insulin resistance and hyperlipidemia, reduce the levels of inflammatory factors in serum and hippocampus, and decreased mortality in T2DM rats. The Morris water maze test indicated that YGJ reduced the escape latency and increased platform crossing frequency, thereby improving cognitive function in T2DM rats. Furthermore, YGJ regulated the abundance of microorganisms associated with bile acid metabolism, including Romboutsia, Bacteroides, Turicibacter, Blautia, and Ruminococcus, thus regulating bile acid metabolism in T2DM rats. Additionally, YGJ also regulated bile acid metabolism by regulating intestinal FXR, PXR, VDR and TRG5 receptors. CONCLUSION YGJ can alleviate glucose homeostasis, insulin sensitivity, lipid metabolism, neuroinflammation, cognitive function, as well as remodel intestinal flora and BA composition in CDT2DM rats, which is a potential complementary and alternative therapy for the prevention and treatment of CDT2DM. These effects may be associated that YGJ regulates the structure of intestinal flora and BA metabolism, and inhibits intestinal BA receptors FXR, PXR, TGR5, and VDR.
Collapse
Affiliation(s)
- Qiyue Yang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, PR China.
| | - Qiwei Luo
- National Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, PR China.
| | - Wenrui Xia
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, PR China.
| | - Nairong Yao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, PR China.
| | - Fang Wang
- National Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, PR China.
| | - Chunguang Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, PR China.
| | - Haiyan Zhang
- National Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, PR China.
| | - Yanan He
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China.
| |
Collapse
|
24
|
Ni W, Liu WV, Li M, Wei S, Xu X, Huang S, Zhu L, Wang J, Wen F, Zhou H. Altered brain functional network connectivity and topology in type 2 diabetes mellitus. Front Neurosci 2025; 19:1472010. [PMID: 39935840 PMCID: PMC11811103 DOI: 10.3389/fnins.2025.1472010] [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: 07/28/2024] [Accepted: 01/06/2025] [Indexed: 02/13/2025] Open
Abstract
Introduction Type 2 diabetes mellitus (T2DM) accelerates brain aging and disrupts brain functional network connectivity, though the specific mechanisms remain unclear. This study aimed to investigate T2DM-driven alterations in brain functional network connectivity and topology. Methods Eighty-five T2DM patients and 67 healthy controls (HCs) were included. All participants underwent clinical, neuropsychological, and laboratory tests, followed by MRI examinations, including resting-state functional magnetic resonance imaging (rs-fMRI) and three-dimensional high-resolution T1-weighted imaging (3D-T1WI) on a 3.0 T MRI scanner. Post-image preprocessing, brain functional networks were constructed using the Dosenbach atlas and analyzed with the DPABI-NET toolkit through graph theory. Results In T2DM patients, functional connectivity within and between the default mode network (DMN), frontal parietal network (FPN), subcortical network (SCN), ventral attention network (VAN), somatosensory network (SMN), and visual network (VN) was significantly reduced compared to HCs. Conversely, two functional connections within the VN and between the DMN and SMN were significantly increased. Global network topology analysis showed an increased shortest path length and decreased clustering coefficient, global efficiency, and local efficiency in the T2DM group. MoCA scores were negatively correlated with the shortest path length and positively correlated with global and local efficiency in the T2DM group. Node network topology analysis indicated reduced clustering coefficient, degree centrality, eigenvector centrality, and nodal efficiency in multiple nodes in the T2DM group. MoCA scores positively correlated with clustering coefficient and nodal efficiency in the bilateral precentral gyrus in the T2DM group. Discussion This study demonstrated significant abnormalities in connectivity and topology of large-scale brain functional networks in T2DM patients. These findings suggest that brain functional network connectivity and topology could serve as imaging biomarkers, providing insights into the underlying neuropathological processes associated with T2DM-related cognitive impairment.
Collapse
Affiliation(s)
- Weiwei Ni
- Physical Examination Centre, Central People's Hospital of Zhanjiang, Zhanjiang, China
| | | | - Mingrui Li
- Department of Magnetic Resonance Imaging, Zhanjiang First Hospital of Traditional Chinese Medicine, Zhanjiang, China
| | - Shouchao Wei
- Central People's Hospital of Zhanjiang, Zhanjiang Institute of Clinical Medicine, Zhanjiang, China
| | - Xuanzi Xu
- Department of Teaching and Training, Central People's Hospital of Zhanjiang, Zhanjiang, China
| | - Shutong Huang
- Department of Clinical Laboratory, Central People's Hospital of Zhanjiang, Zhanjiang, China
| | - Lanhui Zhu
- Physical Examination Centre, Central People's Hospital of Zhanjiang, Zhanjiang, China
| | - Jieru Wang
- Department of Radiology, Central People's Hospital of Zhanjiang, Zhanjiang, China
| | - Fengling Wen
- Department of Radiology, Central People's Hospital of Zhanjiang, Zhanjiang, China
| | - Hailing Zhou
- Department of Radiology, Central People's Hospital of Zhanjiang, Zhanjiang, China
| |
Collapse
|
25
|
Sjöholm Å, Bennet L, Nilsson PM. Cognitive dysfunction in diabetes - the 'forgotten' diabetes complication: a narrative review. Scand J Prim Health Care 2025:1-7. [PMID: 39876043 DOI: 10.1080/02813432.2025.2455136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/10/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND In addition to peripheral neuropathy of various kinds, diabetes can also cause central neuropathy, which among other things can manifest itself as premature cognitive dysfunction, often linked to vascular dysfunction. Although the link between diabetes and cognitive dysfunction was discovered more than 100 years ago and has important clinical implications, this diabetes complication remains relatively unknown. Recent years have seen research that has clarified cerebral insulin resistance and defective insulin signaling as examples of pathogenic factors behind this cognitive impairment in diabetes. METHOD We provide a narrative review of select and contemporary publications with relevance for the interface between diabetes/prediabetes and cognitive function. RESULTS Recently published studies show that physical activity can reverse insulin resistance in the brain as well as cognitive impairment and pathological appetite regulation. Pharmacological interventions with, for example, nasal insulin, GLP-1 receptor agonists, SGLT-2 inhibitors, or PPAR-γ agonists have also shown promising results. CONCLUSION Optimization of lifestyle factors (e.g. physical activity), as well as several pharmaceutical agents already in clinical use against diabetes, have shown promising results in improving cognitive function in diabetic patients. An important task for primary health care, where most patients with type 2 diabetes are diagnosed, treated, and followed, is to increase awareness and early detection of cognitive dysfunction in these patients for optimizing risk factor control.
Collapse
Affiliation(s)
- Åke Sjöholm
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Gävle Hospital and University of Gävle, Gävle, Sweden
| | - Louise Bennet
- Department of Clinical Sciences, Lund University, Clinical Studies Sweden, Forum South, Skåne University Hospital, Lund, Sweden
| | - Peter M Nilsson
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
26
|
Liu S, Hao J, Yu T, Tuchin VV, Li J, Li D, Zhu D. Diabetes Mellitus Impairs Blood-Brain Barrier Integrality and Microglial Reactivity. JOURNAL OF BIOPHOTONICS 2025:e202400482. [PMID: 39870511 DOI: 10.1002/jbio.202400482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/23/2024] [Accepted: 01/02/2025] [Indexed: 01/29/2025]
Abstract
Diabetes mellitus (DM), a chronic metabolic disorder that adversely affects the blood-brain barrier (BBB) and microglial function in the central nervous system (CNS), contributing to neuronal damage and neurodegenerative diseases. However, the underlying molecular mechanisms linking diabetes to BBB dysfunction and microglial dysregulation remain poorly understood. Here, we assessed the impacts of diabetes on BBB and microglial reactivity and investigated its mechanisms. We found diabetes severely disrupted the BBB integrity and microglial response to vascular injury. We also revealed a potential relationship between BBB disruption and impaired microglial function, whereby increasing BBB permeability led to a downregulation of microglial P2RY12 expression, thereby impairing microglial protection against cerebrovascular injury. Understanding these mechanisms may contribute to the developing of therapeutic strategies for diabetes-related neurological complications.
Collapse
Affiliation(s)
- Shaojun Liu
- Britton Chance Center for Biomedical Photonics-MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
- Wuhan National Laboratory for Optoelectronics-Advanced Biomedical Imaging Facility, HUST, Wuhan, China
| | - Jie Hao
- Affiliated Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Yu
- Britton Chance Center for Biomedical Photonics-MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
- Wuhan National Laboratory for Optoelectronics-Advanced Biomedical Imaging Facility, HUST, Wuhan, China
| | - Valery V Tuchin
- Institute of Physics and Science Medical Center, Saratov State University, Saratov, Russian Federation
- Laboratory of Laser Molecular Imaging and Machine Learning, Tomsk State University, Tomsk, Russian Federation
- Institute of Precision Mechanics and Control, FRS "Saratov Scientific Centre of the RAS", Saratov, Russian Federation
| | - Junming Li
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Dongyu Li
- Wuhan National Laboratory for Optoelectronics-Advanced Biomedical Imaging Facility, HUST, Wuhan, China
- School of Optical Electronic Information-Advanced Biomedical Imaging Facility, HUST, Wuhan, China
| | - Dan Zhu
- Britton Chance Center for Biomedical Photonics-MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
- Wuhan National Laboratory for Optoelectronics-Advanced Biomedical Imaging Facility, HUST, Wuhan, China
| |
Collapse
|
27
|
Zhai YJ, Li F, Lin CY, Wu F, Qiu HN, Li JB, Lin JN. The mediating role of body surface area-adjusted basal metabolic rate: effects of low muscle mass and central obesity on cognitive impairment in Chinese patients with type 2 diabetes mellitus. Front Endocrinol (Lausanne) 2025; 15:1513035. [PMID: 39926391 PMCID: PMC11802378 DOI: 10.3389/fendo.2024.1513035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/31/2024] [Indexed: 02/11/2025] Open
Abstract
Background This study investigates the relationship between basal metabolic rate (BMR), body composition, obesity indices, and cognitive impairment (CI) in middle-aged and older type 2 diabetes mellitus (T2DM) patients, assessing their potential role in CI screening. Methods A cross-sectional study included 1243 T2DM patients over 45 years old. CI was assessed using the Montreal Cognitive Assessment. BMR and body composition indices were measured through bioelectrical impedance analysis. The associations and predictions related to CI were explored using multivariable-adjusted logistic regression, restricted cubic spline (RCS) models, and receiver operating characteristic (ROC) curve analyses. Mediation analysis explored the role of BMR adjusted by body surface area (BMR/BSA) in CI risk. Results Patients with CI showed significantly lower BMR, BMR adjusted for height squared (BMR/Height²), BMR/BSA, appendicular skeletal muscle mass (ASM), and fat-free mass (FFM), alongside higher waist circumference (WC) and percentage of body fat. Logistic regression showed that participants in the fourth quartile of BMR, BMR/Height2, and BMR/BSA had approximately a 54% reduced risk of CI (odds ratio range 0.457 to 0.463). RCS analysis indicated a linear decrease in CI risk with increasing BMR metrics. ROC analysis indicated high predictive efficacy for CI with combined indicators, particularly BMR and FFM (area under the curve 0.645). Mediation analysis suggested that BMR/BSA played a significant mediating role in WC, ASM and FFM on CI risk, with a mediation proportion ranging from 45.73% to 50.87%. Conclusion Low energy expenditure assessed by BMR/BSA is an independent risk factor for increased CI risk in middle-aged and elderly T2DM patients. Central obesity, low muscle mass, and low energy expenditure significantly elevate CI risk in this population.
Collapse
Affiliation(s)
- Ya-Jie Zhai
- School of Medicine, Nankai University, Tianjin, China
- Department of Endocrinology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| | - Fang Li
- Department of Endocrinology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| | - Chen-Ying Lin
- Department of Endocrinology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
- Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
| | - Fan Wu
- Department of Endocrinology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| | - Hui-Na Qiu
- Department of Endocrinology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| | - Jing-Bo Li
- Department of Endocrinology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| | - Jing-Na Lin
- Department of Endocrinology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| |
Collapse
|
28
|
Huang CN, Chen HM, Su BY. Type 2 diabetes mellitus: A cross-sectional analysis of glycemic controls and brain health outcomes. APPLIED NEUROPSYCHOLOGY. ADULT 2025:1-8. [PMID: 39832208 DOI: 10.1080/23279095.2025.2450084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
In this cross-sectional analysis, we explored how fluctuations in glycemic levels impact executive functions and psychosocial outcomes in patients with type 2 diabetes mellitus (T2DM). The goal was to understand the relationship between glycemic control and both neuropsychological and psychosocial health. We stratified participants into well-controlled and poorly controlled groups based on glycated hemoglobin (HbA1c) levels and variability, including a healthy control group for comparison. The study consisted of neuropsychological tests and psychosocial assessments. Results indicated that the poorly controlled T2DM group experienced significant executive dysfunction and scored lower on the Tower of London, Wisconsin Card Sorting, and Digit Span Tests, reflecting a broader impact on quality of life and resilience. These findings support the importance of maintaining stable glycemic levels for better executive and psychosocial outcomes and highlight the need for regular neuropsychological and psychosocial assessments in diabetes care.
Collapse
Affiliation(s)
- Chien-Ning Huang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Hsiao-Mei Chen
- Department of Nursing, Chung Shan Medical University, Taichung, Taiwan
- Department of Nursing, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Bei-Yi Su
- Department of Psychology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Psychological Room, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
29
|
Zhang S, Wang X, Liu S, Hu C, Meng Y. Phlorizin ameliorates cognitive and behavioral impairments via the microbiota-gut-brain axis in high-fat and high-fructose diet-induced obese male mice. Brain Behav Immun 2025; 123:193-210. [PMID: 39277023 DOI: 10.1016/j.bbi.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/26/2024] [Accepted: 09/07/2024] [Indexed: 09/17/2024] Open
Abstract
The long-term high-fat, high-sugar diet exacerbates type 2 diabetes mellitus (T2DM)-related cognitive impairments. Phlorizin, a well-studied natural compound found in apples and other plants, is recognized for its bioactive properties, including modulation of glucose and lipid metabolism. Despite its established role in mitigating metabolic disorders, the neuroprotective effects of phlorizin, particularly against diabetes-related cognitive dysfunction, have not been fully elucidated. Therefore, the present study aimed to investigate the effect of dietary supplementation of phlorizin on high-fat and high-fructose diet (HFFD)-induced cognitive dysfunction and evaluate the crucial role of the microbiota-gut-brain axis. We found that dietary supplementation of phlorizin for 14 weeks effectively prevented glucolipid metabolism disorder, spatial learning impairment, and memory impairment in HFFD mice. In addition, phlorizin improved the HFFD-induced decrease in synaptic plasticity, neuroinflammation, and excessive activation of microglia in the hippocampus. Transcriptomics analysis shows that the protective effect of phlorizin on cognitive impairment was associated with increased expression of neurotransmitters and synapse-related genes in the hippocampus. Phlorizin treatment alleviated colon microbiota disturbance, mainly manifested by an increase in gut microbiota diversity and the abundance of short-chain fatty acid (SCFA)-producing bacteria. The level of microbial metabolites, including SCFA, inosine 5'-monophosphate (IMP), and D (-)-beta-hydroxybutyric acid (BHB) were also significantly increased after phlorizin treatment. Integrating multiomics analysis observed tight connections between phlorizin-regulated genes, microbiota, and metabolites. Furthermore, removal of the gut microbiota via antibiotics treatment diminished the protective effect of phlorizin against HFFD-induced cognitive impairment, underscoring the critical role of the gut microbiota in mediating cognitive behavior. Importantly, supplementation with SCFA and BHB alone mimicked the regulatory effects of phlorizin on cognitive function. Therefore, phlorizin shows promise as a potential nutritional therapy for addressing cognitive impairment associated with metabolic disorders. Further research is needed to explore its effectiveness in preventing and alleviating neurodegenerative diseases.
Collapse
Affiliation(s)
- Shuqing Zhang
- Engineering Research Center for High-Valued Utilization of Fruit Resources in Western China, Ministry of Education, National Research & Development Center of Apple Processing Technology, College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, PR China; College of Food Science and Nutritional Engineering, National Engineering Research Centre for Fruit and Vegetable Processing, Key Laboratory for Fruit and Vegetable Processing, Ministry of Agriculture, Engineering Research Centre for Fruit and Vegetable Processing, Ministry of Education, China Agricultural University, Beijing 100083, China.
| | - Xiaoyu Wang
- Engineering Research Center for High-Valued Utilization of Fruit Resources in Western China, Ministry of Education, National Research & Development Center of Apple Processing Technology, College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, PR China.
| | - Shenlin Liu
- Engineering Research Center for High-Valued Utilization of Fruit Resources in Western China, Ministry of Education, National Research & Development Center of Apple Processing Technology, College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, PR China.
| | - Chingyuan Hu
- Engineering Research Center for High-Valued Utilization of Fruit Resources in Western China, Ministry of Education, National Research & Development Center of Apple Processing Technology, College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, PR China; Department of Human Nutrition, Food and Animal Sciences, College of Tropical Agriculture and Human Resources, University of Hawaii at Manoa, 1955 East-West Road, AgSci. 415J, Honolulu, HI 96822, USA.
| | - Yonghong Meng
- Engineering Research Center for High-Valued Utilization of Fruit Resources in Western China, Ministry of Education, National Research & Development Center of Apple Processing Technology, College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, PR China.
| |
Collapse
|
30
|
Xiu L, Ge R, Lu S, Li L, Huang W, Du G, Zhang Z, Zhang J, Wan Z, Chang J. Royal Jelly and 10-Hydroxy-2-Decenoic acid activate autophagy through mTOR/ULK1 pathway to improve cognitive function in diabetic mice. J Funct Foods 2025; 124:106649. [DOI: 10.1016/j.jff.2024.106649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
|
31
|
Tu W, Xu F, Li J, Tian X, Cao L, Wang L, Qu Y. Studying targeted oxidation in diabetic cognitive dysfunction based on scientometrics analysis: research progress of natural product approaches. Front Endocrinol (Lausanne) 2024; 15:1445750. [PMID: 39758348 PMCID: PMC11695123 DOI: 10.3389/fendo.2024.1445750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 11/12/2024] [Indexed: 01/07/2025] Open
Abstract
Purpose The aim is to provide new insights for researchers studying the pathogenesis of diabetic cognitive dysfunction and promoting the wider use of natural products in their treatment. Method First, the Web of Science Core Collection was selected as the data source for a computerized literature search on oxidative stress and diabetic cognitive dysfunction (DCD). Next, Biblimetrix and VOSviewer performed statistical analysis focusing on publication countries, institutions, authors, research hotspots, and emerging directions in the field. Then, through the analysis of keywords and key articles, the forefront of the field is identified. Finally, we discussed the pathogenesis of DCD, the influence of oxidative stress on DCD and the antioxidant effect of natural products on DCD. Result 293 valid papers were obtained. Bibliometrics showed that oxidative stress, diabetes, Alzheimer's disease (AD), cognitive decline, insulin resistance and quercetin were the key words of the symbiotic network. Conclusion The antioxidant effects of natural products in improving DCD have been extensively studied in preclinical studies, providing potential for their treatment in DCD, but their evaluation in clinical trials is currently uncommon.
Collapse
Affiliation(s)
| | | | | | | | | | - Lei Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University,
Hangzhou, China
| | - Yiqian Qu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University,
Hangzhou, China
| |
Collapse
|
32
|
Schwartz SS, Herman ME, Tun MTH, Barone E, Butterfield DA. The double life of glucose metabolism: brain health, glycemic homeostasis, and your patients with type 2 diabetes. BMC Med 2024; 22:582. [PMID: 39696300 DOI: 10.1186/s12916-024-03763-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 11/11/2024] [Indexed: 12/20/2024] Open
Abstract
The maintenance of cognitive function is essential for quality of life and health outcomes in later years. Cognitive impairment, however, remains an undervalued long-term complication of type 2 diabetes by patients and providers alike. The burden of sustained hyperglycemia includes not only cognitive deficits but also the onset and progression of dementia-related conditions, including Alzheimer's disease (AD). Recent research has shown that the brain maintains an independent glucose "microsystem"-evolved to ensure the availability of fuel for brain neurons without interruption by transient hypoglycemia. When this milieu is perturbed, brain hyperglycemia, brain glucotoxicity, and brain insulin resistance can ensue and interfere with insulin signaling, a key pathway to cognitive function and neuronal integrity. This newly understood brain homeostatic system operates semi-autonomously from the systemic glucoregulatory apparatus. Large-scale clinical studies have shown that systemic dysglycemia is also strongly associated with poorer cognitive outcomes, which can be mitigated through appropriate clinical management of plasma glucose levels. Moreover, these studies demonstrated that glucose-lowering agents are not equally effective at preventing cognitive dysfunction. Glucagon-like peptide-1 (GLP-1) receptor analogs and sodium glucose cotransporter 2 inhibitors (SGLT2is) appear to afford the greatest protection; metformin and dipeptidyl peptidase 4 inhibitors (DPP-4is) also significantly improved cognitive outcomes. Sulfonylureas (SUs) and exogenous insulin, on the other hand, do not provide the same protection and may actually worsen cognitive outcomes. In the creation of a treatment plan, comorbid cognitive conditions should be considered. These efficacious treatments create a new gold standard of managing hyperglycemia-one which is consistent with the "complication-centric prescribing" mandates issued in type 2 diabetes treatment guidelines. The increasing longevity enjoyed by our populace places the onus on clinical care to play the "long game" in using targeted treatments for glucose control in patients with, or at risk for, cognitive decline to maintain cognitive wellness later in life. This article reviews critical emerging data for scientists and trialists and translates new enhancements in patient care for practitioners.
Collapse
Affiliation(s)
- Stanley S Schwartz
- University of Pennsylvania School of Medicine, 771 County Line Road, Villanova, PA, 19085, USA
| | - Mary E Herman
- Social Alchemy: Building Physician Competency Across the Globe, 5 Ave Sur #36, Antigua, Sacatepéquez, Guatemala.
| | - May Thet Hmu Tun
- Maimonides Medical Center, 4802 10th Ave, Brooklyn, NY, 11219, USA
| | - Eugenio Barone
- Sapienza University of Rome, Via Degli Equi 42, Scala A, Int. 5, 00185, Rome, Italy
| | - D Allan Butterfield
- Sanders-Brown Center On Aging, Department of Chemistry, University of Kentucky, 249 Chemistry-Physics Building, Lexington, KY, 40506-0055, USA
| |
Collapse
|
33
|
Lai H, Fan P, Wang H, Wang Z, Chen N. New perspective on central nervous system disorders: focus on mass spectrometry imaging. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:8080-8102. [PMID: 39508396 DOI: 10.1039/d4ay01205d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
An abnormally organized brain spatial network is linked to the development of various central nervous system (CNS) disorders, including neurodegenerative diseases and neuropsychiatric disorders. However, the complicated molecular mechanisms of these diseases remain unresolved, making the development of treatment strategies difficult. A novel molecular imaging technique, called mass spectrometry imaging (MSI), captures molecular information on the surface of samples in situ. With MSI, multiple compounds can be simultaneously visualized in a single experiment. The high spatial resolution enables the simultaneous visualization of the spatial distribution and relative content of various compounds. The wide application of MSI in biomedicine has facilitated extensive studies on CNS disorders in recent years. This review provides a concise overview of the processes, applications, advantages, and disadvantages, as well as mechanisms of the main types of MSI. Meanwhile, this review summarizes the main applications of MSI in studying CNS diseases, including Alzheimer's disease (AD), CNS tumors, stroke, depression, Huntington's disease (HD), and Parkinson's disease (PD). Finally, this review comprehensively discusses the synergistic application of MSI with other advanced imaging modalities, its utilization in organoid models, its integration with spatial omics techniques, and provides an outlook on its future potential in single-cell analysis.
Collapse
Affiliation(s)
- Huaqing Lai
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Pinglong Fan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Huiqin Wang
- Hunan University of Chinese Medicine, Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha 410208, Hunan, China
| | - Zhenzhen Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Naihong Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
34
|
Xia W, Yin X, Zhang Y, Ge S, Chen Y, Ma J. Gray Matter Reserve Modulates the Association between Glymphatic System Function and Cognition in Patients with Type 2 Diabetes Mellitus. Neuroendocrinology 2024; 115:48-59. [PMID: 39622216 DOI: 10.1159/000542902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 11/25/2024] [Indexed: 02/02/2025]
Abstract
INTRODUCTION The glymphatic system is regarded as a key factor in the pathogenesis of neurodegenerative diseases. Given the heightened risk of cognitive impairment in patients with type 2 diabetes mellitus (T2DM), the possible alterations in the glymphatic system in T2DM patients remain to be explored. Diffusion tensor imaging (DTI) analysis along the perivascular space (ALPS) index can be utilized to model the glymphatic system in humans. Our aim was to explore the relationship between the ALPS index and cognitive function in patients with T2DM and whether this relationship is modulated by gray matter (GM) integrity anchored by the ALPS index. METHODS All participants underwent evaluation using a comprehensive cognitive assessment scale to determine their neurocognitive status. The ALPS index was calculated based on DTI data, and the disparity in ALPS index values between patients with T2DM and healthy controls (HCs) was examined. Furthermore, multiple linear regression analysis was conducted in the T2DM group to identify the GM regions associated with the ALPS index, and the volumes of the GM partitions were extracted. The volume of GM partitions was used as the regulating variable, the ALPS index was used as the independent variable, and cognitive test scores were used as the dependent variable in our analysis. RESULTS The ALPS index differed significantly between the two groups, and the ALPS index in patients with T2DM was significantly lower than that in HCs. In addition, our analysis revealed a correlation between the ALPS index and GM volume in the insular region, consistent with the observed GM atrophy in the patient cohort. Moreover, a significant negative correlation was observed between the ALPS index in patients and performance on the Trail-Making Test-A (TMT-A), and this relationship was moderated by GM integrity. In patients with more severe GM atrophy, the ALPS index was more strongly correlated with cognitive function. CONCLUSIONS In this study, a decreased ALPS index was found in T2DM patients, indicating impaired glymphatic function in this population. Furthermore, a significant association was detected between the ALPS index and cognitive performance in T2DM patients, and this correlation was influenced by GM integrity. Therefore, the ALPS index has the potential to be used as a biomarker of cognitive impairment in diabetic patients. Further studies are needed to investigate the diagnostic and therapeutic implications of glymphatic dysfunction in T2DM patients with cognitive impairment.
Collapse
Affiliation(s)
- Wenqing Xia
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiao Yin
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yujie Zhang
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shenghui Ge
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yuchen Chen
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jianhua Ma
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
35
|
Yang J, Tan A, Li T, Chen H. Irisin alleviates the pyroptosis of β cells in T2DM by inhibiting NLRP3 inflammasome through regulating miR-19b-3p/SOCS3/STAT3 axis mediated autophagy. IUBMB Life 2024; 76:1264-1278. [PMID: 39143849 DOI: 10.1002/iub.2907] [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: 05/08/2024] [Accepted: 06/25/2024] [Indexed: 08/16/2024]
Abstract
The purpose of this study was to analyze the mechanism by which irisin affects β-cell pyroptosis in type 2 diabetes mellitus (T2DM). The in vivo T2DM model was established by raised with high-fat diet and intraperitoneally injection of streptozocin. Min6 cells were divided into four groups: negative control (NC), high glucose (HG), HG + irisin, and HG + irisin+3-MA. The cell viability was determined by CCK-8 assay. Dual-luciferase gene reporter assay was conducted to confirm the binding between miR-19b-3p and SOCS3. The expression level of FNDC5 and GSDMD was visualized using the immunofluorescence assay. The protein level of FNDC5, Beclin1, LC3II/I, NLRP3, cleaved-caspase-1, GSDMD-N, STAT3, p-STAT3, and SOCS3 was determined by Western blotting. The secretion of irisin, lactate dehydrogenase (LDH), and insulin was checked by ELISA. In vivo results showed that pathological changes in islet tissues with declined number of β cells, elevated FBG value, decreased FIN and HOMA-β value, elevated autophagy-associated proteins expressions, and activated NLRP3 signaling in T2DM mice, which were dramatically reversed by FNDC5 overexpression. Furthermore, the declined level of miR-19b-3p and p-STAT3, as well as the upregulation of SOCS3, was greatly rescued by FNDC5 overexpression. The in vitro data confirmed the binding site between SOCS3 and miR-19b-3p. SOCS3 was downregulated and p-STAT3 was upregulated in miR-19b-3p mimic-treated Min6 cells. In HG-stimulated Min6 cells, the elevated cell viability, increased production of insulin, decreased release of LDH, and inactivated NLRP3 signaling induced by irisin were abolished by miR-19b-3p inhibitor and STAT3 inhibitor. The increased level of autophagy-related proteins and activated SOCS3/STAT3 axis induced by irisin in HG-stimulated Min6 cells were abolished by miR-19b-3p inhibitor. The inhibitory effect of irisin against NLRP3 signaling in HG-stimulated Min6 cells was abrogated by 3-MA. In conclusion, irisin alleviated the pyroptosis of β cells in T2DM by inhibiting NLRP3 signaling through miR-19b-3p/SOCS3/STAT3 axis mediated autophagy.
Collapse
Affiliation(s)
- Jingjing Yang
- Department of Geriatric Medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Anjun Tan
- Department of Geriatric Medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Tianrong Li
- Department of Geriatric Medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Hewen Chen
- Department of Geriatric Medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
36
|
Lin YH, Chang HT, Wang YF, Fuh JL, Wang SJ, Chen HS, Li SR, Lin MH, Chen TJ, Hwang SJ. The association of the comorbidity status of metabolic syndrome and cognitive dysfunction with health-related quality of life. Qual Life Res 2024; 33:3421-3433. [PMID: 39269582 DOI: 10.1007/s11136-024-03784-z] [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] [Accepted: 09/02/2024] [Indexed: 09/15/2024]
Abstract
PURPOSE Both metabolic syndrome (MetS) and cognitive dysfunction impair health-related quality of life (HRQOL). This study aims to determine whether individuals experiencing both MetS and cognitive dysfunction have lower HRQOL. METHODS This cross-sectional study enrolled 567 participants who attended outpatient clinics at a medical center in northern Taiwan. MetS was diagnosed according to the modified criteria for the Asian population. Cognitive function was categorized as normal, mild cognitive dysfunction, and advanced cognitive dysfunction according to the score of the Montreal Cognitive Assessment, Taiwanese version. HRQOL was assessed using the SF-36v2® Health Survey (SF-36v2). The associations of the comorbidity status of MetS and cognitive dysfunction with HRQOL were analyzed using linear regression models, adjusting for age, sex, marital status, education level, income groups, and activities of daily living. RESULTS Out of 567 participants, 33 (5.8%) had MetS with mild cognitive dysfunction, and 34 (6.0%) had MetS with advanced cognitive dysfunction. Participants with both MetS and advanced cognitive dysfunction exhibited the lowest scores in the physical component summary and almost all scales of HRQOL. MetS exacerbated the inverse association between mild cognitive dysfunction and the mental component summary. For those with MetS, the scores on scales of role physical, bodily pain, vitality, and social functioning worsened as cognitive function deteriorated (all Ptrend<0.05). CONCLUSION As the severity of comorbidity between MetS and cognitive dysfunction varies, patients exhibited poorer performance in different aspects of HRQOL. Future research is needed to find solutions to improve HRQOL for patients with both MetS and cognitive dysfunction.
Collapse
Affiliation(s)
- Yi-Hsuan Lin
- Department of Medical Education, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Family Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Beitou Dist, Taipei City, 11217, Taiwan
| | - Hsiao-Ting Chang
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Department of Family Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Beitou Dist, Taipei City, 11217, Taiwan.
| | - Yen-Feng Wang
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jong-Ling Fuh
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shuu-Jiun Wang
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Harn-Shen Chen
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Sih-Rong Li
- Department of Family Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Beitou Dist, Taipei City, 11217, Taiwan
| | - Ming-Hwai Lin
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Family Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Beitou Dist, Taipei City, 11217, Taiwan
| | - Tzeng-Ji Chen
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Family Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Beitou Dist, Taipei City, 11217, Taiwan
- Department of Family Medicine, Taipei Veterans General Hospital Hsinchu Branch, Hsinchu County, Taiwan
- Department of Post-Baccalaureate Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Shinn-Jang Hwang
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Family Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Beitou Dist, Taipei City, 11217, Taiwan
- Department of Family Medicine, En Chu Kong Hospital, New Taipei City, Taiwan
| |
Collapse
|
37
|
Karunathilaka N, Parker C, Lazzarini PA, Chen P, Katsanos C, MacAndrew M, Finlayson K. Cognitive changes in people with diabetes with lower extremity complications compared to people with diabetes without lower extremity complications: a systematic review and meta-analysis. BMC Endocr Disord 2024; 24:258. [PMID: 39609829 PMCID: PMC11605952 DOI: 10.1186/s12902-024-01774-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 10/31/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Recent evidence suggests that diabetes-related lower-extremity complications (DRLECs) may be associated with cognitive changes in people with diabetes. However, existing literature has produced inconsistent findings, and no systematic reviews have been conducted to investigate whether DRLECs impact the cognition of people with diabetes. This systematic review evaluated existing studies that investigated cognition in people with diabetes with DRLECs and without DRLECs. METHOD Seven databases; MEDLINE, PubMed, CINAHL, EMBASE, Cochrane, PsycINFO and Web of Science were searched from inception until 22/8/2022 for studies that compared cognition in people with diabetes with and without DRLECs. Results were independently screened for eligibility and assessed for methodological quality by two authors, with key data extracted. Studies were eligible for meta-analysis if the studies reported similar cases, controls, and outcome measures. RESULTS Thirteen studies were included in the review, with eleven of medium methodological quality, one of high quality, and one of low quality. Four studies found significant differences in cognition between those with and without DRLECs, four found significant associations between diabetes-related lower-extremity complications and cognition, and five found no differences or associations. One small meta-analysis of eligible studies found that there was no statistically significant difference in cognition in people without, compared to with, peripheral neuropathy (Mean difference = -0.49; 95%CI: -1.59-0.61; N = 3; n = 215). Leave-one-out sensitivity analyses further confirmed that there was no significant difference in cognition among people with and without peripheral neuropathy (p > 0.05). CONCLUSION DRLECs may be related to cognition in people with diabetes, however, existing evidence is unclear due to variability in used methodologies that may challenge concluding the findings. Future high-quality studies investigating cognition among people with and without DRLECs are needed.
Collapse
Affiliation(s)
- Nimantha Karunathilaka
- Center for Healthcare Transformation, Queensland University of Technology, Brisbane, QLD, Australia.
- Department of Nursing and Midwifery, Faculty of Allied Health Sciences, General Sir John Kotelawala Defence University, Ratmalana, Sri Lanka.
- School of Nursing, Queensland University of Technology, Brisbane, QLD, Australia.
| | - Christina Parker
- Center for Healthcare Transformation, Queensland University of Technology, Brisbane, QLD, Australia
- School of Nursing, Queensland University of Technology, Brisbane, QLD, Australia
| | - Peter A Lazzarini
- Center for Healthcare Transformation, Queensland University of Technology, Brisbane, QLD, Australia
- School of Public Health and Social Work, Queensland University of Technology, Brisbane, QLD, Australia
- Allied Health Research Collaborative, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Pamela Chen
- Joondalup Health Campus, Ramsay Healthcare, Perth, WA, Australia
| | - Chloe Katsanos
- Podiatry Department, The Alfred, Melbourne, VIC, Australia
| | - Margaret MacAndrew
- Center for Healthcare Transformation, Queensland University of Technology, Brisbane, QLD, Australia
- School of Nursing, Queensland University of Technology, Brisbane, QLD, Australia
| | - Kathleen Finlayson
- Center for Healthcare Transformation, Queensland University of Technology, Brisbane, QLD, Australia
- School of Nursing, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
38
|
Yang Y, Peng P, Huang H, Zhao Y, Li Y, Xu X, Jiang S, Yang Y, Pan G, Wen Y, Wu D, Chen S, Feng L, Peng T, Wang J, Li Z. The triglyceride-glucose index and risk of cognitive impairment: a systematic review and meta-analysis with inclusion of two national databases. Front Neurol 2024; 15:1496871. [PMID: 39677858 PMCID: PMC11638587 DOI: 10.3389/fneur.2024.1496871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024] Open
Abstract
Background To investigate the relationship between the triglyceride and glucose (TyG) index and cognitive impairment (CI). Methods Five authoritative databases were systematically searched for potentially relevant studies on 'TyG index' and 'CI' from inception to 27 April 2024. Two representative databases from the United Kingdom and United States were also included. We used the PICOS criteria to select available articles. All data was combined to compute Odd Ratios (ORs). Results 15 studies were included in the meta-analysis (participants: 5604303). The pooled effect sizes demonstrate that individuals with a high TyG index exhibit a significantly elevated risk of CI compared to those with a low TyG index (OR = 2.16, 95%CI: 1.51; 3.08, p < 0.001). The subgroup analysis showed that inpatients with a high TyG index exhibited an increased risk of CI (OR = 4.56, 95%CI: 3.09; 6.74, p < 0.001). Furthermore, the risk of developing distinct types of CI differed significantly [CI: OR = 1.64, 95% CI: 1.29; 2.07, p < 0.001; Vascular Cognitive Impairment (VCI): OR = 5.39, 95% CI: 3.33; 8.70, p < 0.001]. Conclusion A positive correlation exists between the TyG index and risk of CI, which has potential value in optimizing CI risk stratification among elderly people, especially those hospitalized. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42023450336.
Collapse
Affiliation(s)
- Ying Yang
- Department of Neurology, Geriatric Diseases Institute of Chengdu/Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
- School of Computer Science and Technology, Chongqing University of Postsand Telecommunications, Chongqing, China
| | - Pai Peng
- School of Clinical Medicine, Jinggangshan University, Ji'an, Jiangxi Province, China
- Online Collaborative Research Center for Evidence-Based Medicine Ministry of Education, Jinggangshan University Branch, Ji'an, Jiangxi Province, China
| | - Huadong Huang
- School of Clinical Medicine, Jinggangshan University, Ji'an, Jiangxi Province, China
- Online Collaborative Research Center for Evidence-Based Medicine Ministry of Education, Jinggangshan University Branch, Ji'an, Jiangxi Province, China
| | - Yanan Zhao
- Online Collaborative Research Center for Evidence-Based Medicine Ministry of Education, Jinggangshan University Branch, Ji'an, Jiangxi Province, China
- School of Basic Medicine, Jinggangshan University, Ji'an, Jiangxi Province, China
| | - Yating Li
- Online Collaborative Research Center for Evidence-Based Medicine Ministry of Education, Jinggangshan University Branch, Ji'an, Jiangxi Province, China
- School of Basic Medicine, Jinggangshan University, Ji'an, Jiangxi Province, China
| | - Xiao Xu
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shixie Jiang
- Department of Psychiatry, University of Florida College of Medicine, Gainesville, FL, United States
| | - Yanrong Yang
- Department of Neurology, Geriatric Diseases Institute of Chengdu/Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Gaofeng Pan
- Department of Neurology, Geriatric Diseases Institute of Chengdu/Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Yanting Wen
- Department of Ultrasound Medicine, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, China
| | - Dan Wu
- School of Computer Science and Technology, Chongqing University of Postsand Telecommunications, Chongqing, China
| | - Shanping Chen
- Department of Geriatrics, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, China
| | - Lei Feng
- Department of Neurology, Geriatric Diseases Institute of Chengdu/Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Tangming Peng
- Department of Neurology, Geriatric Diseases Institute of Chengdu/Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Jiang Wang
- Online Collaborative Research Center for Evidence-Based Medicine Ministry of Education, Jinggangshan University Branch, Ji'an, Jiangxi Province, China
- School of Basic Medicine, Jinggangshan University, Ji'an, Jiangxi Province, China
| | - Zheng Li
- Department of Neurology, Geriatric Diseases Institute of Chengdu/Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| |
Collapse
|
39
|
Ge X, Zhang L, Ye M, Fei A, Wang L, Yuan A. Electroacupuncture improves diabetic cognitive impairment rats by suppressing NLRP3 inflammasome activation via induction of mitophagy. Metab Brain Dis 2024; 40:14. [PMID: 39560708 PMCID: PMC11576777 DOI: 10.1007/s11011-024-01464-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 09/15/2024] [Indexed: 11/20/2024]
Abstract
Diabetic cognitive impairment pose a significant threat to public health in our aging society. However, the underlying molecular mechanisms have not been fully elucidated, which warrants further investigation. This study aimed to investigate the effects of electroacupuncture on cognitive impairment and its associated mechanisms. The diabetes model was induced via intraperitoneal injection of streptozotocin (STZ) into Sprague-Dawley rats combined with a high-fat and high-sugar diet. The learning and memory abilities of the rats were assessed using behavioral tests. Electron microscopy and hematoxylin-eosin (H&E) staining were used to identify the histological changes of neurons in the CA1 area of the rat hippocampal CA1. An examination of related indicators was performed by Western blotting including NLRP3 inflammasome-associated proteins Caspase-1, IL-18, IL1β, NLRP3, and P62, and mitophagy-related proteins Pink1, LC3, and Parkin. After modeling, rats displayed impaired learning and memory functions. The administration of electroacupuncture treatment alleviated diabetic cognitive impairment, described as shorter escape latency and an increased frequency of platform crossings. The damaged morphological and ultrastructural changes of neurons in rat hippocampal CA1 area can be alleviated through electroacupuncture treatment. Furthermore, in-depth studies suggest that electroacupuncture treatment can suppress NLRP3 inflammasome activation through induction of Pink1, LC3 and Parkin expression. Electroacupuncture treatment can attenuate NLRP3 inflammasome activation by promoting mitophagy, eventually improving cognitive impairment in (STZ)-treated rats with a high-fat die.
Collapse
Affiliation(s)
- Xia Ge
- Department of Endocrinology, The Second Affiliated Hospital of Anhui, University of Chinese Medicine, Hefei, 230001, Anhui, China
| | - Lele Zhang
- Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Min Ye
- Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Aihua Fei
- Department of Endocrinology, The Second Affiliated Hospital of Anhui, University of Chinese Medicine, Hefei, 230001, Anhui, China
| | - Ling Wang
- Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, 100102, Anhui, China
| | - Aihong Yuan
- Department of acupuncture and rehabilitation, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui, China.
| |
Collapse
|
40
|
Lee J, Kim J, An SJ. Association of diabetes risk with changes in memory, working memory, and processing speed among older adults. Front Psychol 2024; 15:1427139. [PMID: 39600601 PMCID: PMC11588497 DOI: 10.3389/fpsyg.2024.1427139] [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: 05/03/2024] [Accepted: 09/30/2024] [Indexed: 11/29/2024] Open
Abstract
Background This study investigated the risk of diabetes by examining changes in memory, working memory, and processing speed among older adults to provide evidence on how each cognitive domain is associated with the risk of diabetes in older adults. Methods This study used Health and Retirement Study data and tracked the respondents from 2012 to 2020 (n = 5,748). The Telephone Interview for Cognitive Status-27 includes three cognitive tests (recall, seven subtraction, and counting backward tests) to assess each cognitive domain. A Cox proportional hazard regression was used to calculate the changes in the odds ratio (OR) of diabetes by increasing each cognitive function and the parameter in covariates. Results We found that the OR of diabetes decreased with increasing universal cognitive function, increasing memory, working memory, and processing speed, and that age increased the OR in all analysis models. Conclusion The findings of this study contribute to filling gaps in the literature by exploring: (a) the association between each cognitive function and the decline in diabetes risk and (b) the varying patterns of change in diabetes risk with increasing cognitive function.
Collapse
Affiliation(s)
- Jungjoo Lee
- School of Health Professions, College of Nursing and Health Professions, University of Southern Mississippi, Hattiesburg, MS, United States
| | - Junhyoung Kim
- Department of Health Behavior, School of Public Health, Texas A&M University, College Station, TX, United States
| | - Sang Joon An
- Department of Neurology, The Convergence Institute of Healthcare and Medical Science, International St. Mary’s Hospital, Catholic Kwandong University, Incheon, Republic of Korea
| |
Collapse
|
41
|
Chou PS, Lee MY, Chang WS, Chou MC, Hsu CY, Liou LM, Juan CH, Lai CL. Potential Cognitive Decline Linked to Electronegative L5 in Type 2 Diabetes: A Holo-Hilbert Spectral Analysis. Neuroendocrinology 2024; 114:1124-1138. [PMID: 39527931 DOI: 10.1159/000542360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 09/26/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Patients with type 2 diabetes mellitus (T2DM) have an increased risk of cognitive impairment. In this study, we investigated the effect of L5 - an electronegative subfraction of low-density lipoprotein cholesterol (LDL-C) - on the cognitive function of patients with T2DM. METHODS This cross-sectional study included 68 patients with T2DM: 15 with normal cognitive function, 39 with mild cognitive impairment (MCI), and 14 with Alzheimer disease (AD). Cognitive evaluation was performed using the Cognitive Abilities Screening Instrument. We developed a new method - Holo-Hilbert spectral analysis (HHSA) - for analyzing electroencephalography signals. Using HHSA, we investigated the effects of L5 on patients' neural activity. RESULTS Our findings suggested that a higher percentage of L5 in LDL-C (L5%) was independently associated with increased risks of MCI and AD in patients with T2DM. A negative correlation was observed between serum L5% and cognitive performance, particularly in the concentration subdomain, in patients with MCI. HHSA revealed that an elevated serum L5% value was correlated with an increase in low-frequency neural oscillations but a reduction in high-frequency oscillations in patients with MCI. However, no correlation was observed between L5, cognitive performance, and neural activity in patients with normal cognitive function or AD. CONCLUSION Our findings demonstrate L5 to be an efficient biomarker and electroencephalography/HHSA to be an innovative approach for assessing cognitive function in patients with T2DM. L5 may affect frontal lobe function, leading to concentration deficits. The correlation between L5 and cognitive impairment appears to vary depending on the stage of neurodegeneration.
Collapse
Affiliation(s)
- Ping-Song Chou
- Department of Neurology, Kaohsiung Medical University Gangshan Hospital, Kaohsiung, Taiwan
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Neurology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mei-Yueh Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Medical University Gangshan Hospital, Kaohsiung, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wen-Sheng Chang
- Institute of Cognitive Neuroscience, College of Health Sciences and Technology, National Central University, Taoyuan, Taiwan
| | - Mei-Chuan Chou
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chung-Yao Hsu
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Neurology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Li-Min Liou
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Neurology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chi-Hung Juan
- Institute of Cognitive Neuroscience, College of Health Sciences and Technology, National Central University, Taoyuan, Taiwan
- Cognitive Intelligence and Precision Healthcare Research Center, National Central University, Taoyuan, Taiwan
| | - Chiou-Lian Lai
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Neurology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
42
|
Moritz G, Becker JH, Ankam JV, Arcoleo K, Wysocki M, Holtzer R, Wisnivesky J, Busse PJ, Federman AD, Jariwala SP, Feldman JM. Considering different Montreal Cognitive Assessment cutoff scores for older adults with asthma. Allergy Asthma Proc 2024; 45:e72-e80. [PMID: 39517079 PMCID: PMC11572945 DOI: 10.2500/aap.2024.45.240045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Background: There is a greater prevalence of cognitive impairment among ethnic and/or racial minorities, and cognitive impairment is a barrier to asthma self-management (SM) behaviors and outcomes in older adults. Objective: The aim of this study was to examine the relationship between cognitive impairment, assessed by using the Montreal Cognitive Assessment (MoCA), and asthma SM behaviors and outcomes in a sample of predominantly Black and Latino participants. In addition, we evaluated whether using two different MoCA cutoff scores influenced the association between cognitive impairment and asthma outcomes. Methods: Baseline cross-sectional data were extracted from a longitudinal study of older adults with asthma (N = 165) ages ≥60 years. Cognition was assessed by using the MoCA. Asthma Control Questionnaire, asthma-related quality of life (AQOL), and inhaled corticosteroid (ICS) adherence were assessed by using self-report. ICS dosing was collected through chart review and inhaler technique was observed and rated. Results: Using established MoCA cutoff scores of 23 and 26 yielded 45% and 74% cognitive impairment rates, respectively. Cognitive impairment, defined by using the cutoff score of 23, was significantly associated with worse asthma control (p = 0.04) and worse ICS adherence (p = 0.01). With a cutoff score of 26, only AQOL was significantly associated with cognitive impairment (p = 0.03). Race and/or ethnicity moderated the relationship between cognitive impairment and asthma control with a MoCA cutoff score of 23, and between cognitive impairment and AQOL with a MoCA cutoff score of 26. Conclusion: Cognitive impairment in older adults with asthma is associated with important clinical outcomes, but this relationship is influenced by the cutoff score used to define cognitive impairment.
Collapse
Affiliation(s)
- Gali Moritz
- From the Ferkauf Graduate School of Psychology, Yeshiva University, Bronx, New York
| | - Jacqueline H. Becker
- Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jyoti V. Ankam
- Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kimberly Arcoleo
- College of Nursing, Michigan State University, East Lansing, Michigan
| | - Matthew Wysocki
- Division of Academic General Pediatrics, Department of Pediatrics, Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, New York
| | - Roee Holtzer
- From the Ferkauf Graduate School of Psychology, Yeshiva University, Bronx, New York
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York
| | - Juan Wisnivesky
- Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Paula J. Busse
- Division of Allergy and Immunology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alex D. Federman
- Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sunit P. Jariwala
- Division of Allergy/Immunology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York; and
| | - Jonathan M. Feldman
- From the Ferkauf Graduate School of Psychology, Yeshiva University, Bronx, New York
- Albert Einstein College of Medicine, Children’s Hospital at Montefiore, Department of Pediatrics and Department of Psychiatry and Behavioral Sciences, Division of Academic General Pediatrics, Bronx, New York
| |
Collapse
|
43
|
Zhang JM, Liu XB, Li YX, Li HJ, Fan J, Xue C, Yin YF, Zhang Y, Nong YX, Wang YN, Zheng Z, Zhong DL, Li J, Jin RJ. Characteristic Activation Pattern and Network Connectivity of Prefrontal Cortex in Patients with Type 2 Diabetes Mellitus and Major Depressive Disorder during a Verbal Fluency Task: A Functional Near-Infrared Spectroscopy Study Based on Network-Based Statistic Prediction. Neuroendocrinology 2024; 114:1112-1123. [PMID: 39471791 DOI: 10.1159/000542235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/08/2024] [Indexed: 11/01/2024]
Abstract
INTRODUCTION Type 2 diabetes mellitus (T2DM) and major depressive disorder (MDD) together occur frequently among the elderly population. However, the inconsistency in assessments and limited medical resources in the community make it challenging to identify depression in patients with T2DM. This cross-sectional study aimed to investigate the activation pattern and network connectivity of prefrontal cortex (PFC) during a verbal fluency task (VFT) in patients with T2DM and MDD using functional near-infrared spectroscopy (fNIRS). METHODS Three parallel groups (T2DM with MDD group, T2DM group, and healthy group) with 100 participants in each group were included in the study. Recruitment took place from August 1, 2020, to December 31, 2023. Due to the close association between the PFC and depressive emotions, fNIRS was used to monitor brain activation and network connectivity of PFC in all participants during a task of Chinese-language phonological VFT. Network-based statistic prediction was adopted as data analysis method. RESULTS Patients in the T2DM with MDD group showed characteristic activation pattern and network connectivity in contrast with patients with T2DM and healthy controls, including decreased activation in PFC, and decreased network connectivity of right dorsolateral prefrontal cortex (DLPFC). Furthermore, the network connectivity of the right DLPFC in patients with T2DM and MDD was negatively correlated with scores of Hamilton Depression Scale-24 (HAMD-24). CONCLUSIONS There was a distinctive activation pattern and network connectivity of the PFC in patients with T2DM and MDD. The right DLPFC could serve as a potential target for the diagnosis and intervention of MDD in patients with T2DM.
Collapse
Affiliation(s)
- Jia-Ming Zhang
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China,
- Department for Neural Function Detection and Regulation, West China Xiamen Hospital, Sichuan University, Xiamen, China,
| | - Xiao-Bo Liu
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu-Xi Li
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hui-Jing Li
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jin Fan
- School of Acupuncture Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chen Xue
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yun-Fang Yin
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuan Zhang
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu-Xuan Nong
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi-Nan Wang
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhong Zheng
- Department for Neural Function Detection and Regulation, West China Xiamen Hospital, Sichuan University, Xiamen, China
| | - Dong-Ling Zhong
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Juan Li
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rong-Jiang Jin
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
44
|
Zhang Y, Liu G, Ding H, Fan B. High expression of CNOT6L contributes to the negative development of type 2 diabetes. Sci Rep 2024; 14:24723. [PMID: 39433858 PMCID: PMC11494123 DOI: 10.1038/s41598-024-76095-5] [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: 01/13/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024] Open
Abstract
OBJECTIVE Type 2 diabetes (T2D) is a chronic metabolic disorder characterized by reduced responsiveness of body cells to insulin, leading to elevated blood sugar levels. CNOT6L is involved in glucose metabolism, insulin secretion regulation, pancreatic beta-cell proliferation, and apoptosis. These functions may be closely related to the pathogenesis of T2D. However, the exact molecular mechanisms linking CNOT6L to T2D remain unclear. Therefore, this study aims to elucidate the role of CNOT6L in T2D. METHODS The T2D datasets GSE163980 and GSE26168 profiles were downloaded from the Gene Expression Omnibusdatabase generated by GPL20115 and GPL6883.The R package limma was used to screen differentially expressed genes (DEGs). A weighted gene co-expression network analysis was performed. Construction and analysis of the protein-protein interaction (PPI) network, functional enrichment analysis, gene set enrichment analysis, and comparative toxicogenomics database (CTD) analysis were performed. Target Scan was used to screen miRNAs that regulate central DEGs. The results were verified by reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR), western blotting (WB), and blood glucose measurements in mice. RESULTS A total of 1951 DEGs were identified. GO and KEGG enrichment analysis revealed that differentially expressed genes were mainly enriched in the insulin signaling pathway, ECM-receptor interaction, and PPAR signaling pathway. Metascape analysis indicated enrichment primarily in the cAMP signaling pathway and enzyme-linked receptor protein signaling pathway. WGCNA analysis yielded 50 intersecting genes. PPI network construction and algorithm identification identified two core genes (CNOT6L and GRIN2B), among which CNOT6L gene was associated with multiple miRNAs. CTD analysis revealed associations of core genes with type 2 diabetes, diabetic complications, dyslipidemia, hyperglycemia, and inflammation. WB and RT-qPCR results showed that in different pathways, CNOT6L protein and mRNA levels were upregulated in type 2 diabetes. CONCLUSION CNOT6L is highly expressed in type 2 diabetes mellitus, and can cause diabetes complications, inflammation and other physiological processes by regulating miRNA, PPAR and other related signaling pathways, with poor prognosis. CNOT6L can be used as a potential therapeutic target for type 2 diabetes.
Collapse
Affiliation(s)
- Yuna Zhang
- Department of Endocrinology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Guihong Liu
- Department of Endocrinology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Haiyan Ding
- Department of Endocrinology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Bingge Fan
- Department of Endocrinology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China.
| |
Collapse
|
45
|
Wang Q, Yang Y. Bioinformatics analysis of effective biomarkers and immune infiltration in type 2 diabetes with cognitive impairment and aging. Sci Rep 2024; 14:23279. [PMID: 39375405 PMCID: PMC11488262 DOI: 10.1038/s41598-024-74480-8] [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: 01/26/2024] [Accepted: 09/26/2024] [Indexed: 10/09/2024] Open
Abstract
With the increasing prevalence of diabetes mellitus worldwide, type 2 diabetes mellitus (T2D) combined with cognitive impairment and aging has become one of the common and important complications of diabetes mellitus, which seriously affects the quality of life of the patients, and imposes a heavy burden on the patients' families and the society. Currently, there are no special measures for the treatment of cognitive impairment and aging in type 2 diabetes mellitus. Therefore, the search for potential biological markers of type 2 diabetes mellitus combined with cognitive impairment and aging is of great significance for future precisive treatment. We downloaded three gene expression datasets from the GEO database: GSE161355 (related to T2D with cognitive impairment and aging), GSE122063, and GSE5281 (related to Alzheimer's disease). Differentially expressed genes (DEGs) were identified, followed by gene set enrichment analysis (GSEA). A protein-protein interaction (PPI) network was constructed using the STRING database, and the top 15 hub genes were identified using the CytoHubba plugin in Cytoscape. Core genes were ultimately determined using three machine learning methods: LASSO regression, Support Vector Machine Recursive Feature Elimination (SVM-RFE), and Linear Discriminant Analysis (LDA). The diagnostic performance of these genes was assessed using ROC curve analysis and validated in an independent dataset (GSE5281). Regulatory genes related to ferroptosis were screened from the FerrDb database, and their biological functions were further explored through GO and KEGG enrichment analyses. Finally, the CIBERSORT algorithm was used to analyze immune cell infiltration, and the correlation between core genes and immune cell infiltration levels was calculated, leading to the construction of an mRNA-miRNA regulatory network. In the GSE161355 and GSE122063 datasets, 217 common DEGs were identified. GSEA analysis revealed their enrichment in the PI3K-PLC-TRK signaling pathway, TP53 regulation of metabolic genes pathway, Notch signaling pathway, among others. PPI network analysis identified 15 candidate core genes, and further selection using LASSO, LDA, and SVM-RFE machine learning algorithms resulted in 6 core genes: BCL6, TP53, HSP90AA1, CRYAB, IL1B, and DNAJB1. ROC curve analysis indicated that these genes had good diagnostic performance in the GSE161355 dataset, with TP53 and IL1B achieving an AUC of 0.9, indicating the highest predictive accuracy. BCL6, HSP90AA1, CRYAB, and DNAJB1 also had AUCs greater than 0.8, demonstrating moderate predictive accuracy. Validation in the independent dataset GSE5281 showed that these core genes also had good diagnostic performance in Alzheimer's disease samples (AUC > 0.6). Ferroptosis-related analysis revealed that IL1B and TP53 play significant roles in apoptosis and immune response. Immune cell infiltration analysis showed that IL1B is significantly positively correlated with infiltration levels of monocytes and NK cells, while TP53 is significantly negatively correlated with infiltration levels of follicular helper T cells. The construction of the miRNA-mRNA regulatory network suggested that miR-150a-5p might play a key role in the regulation of T2D-associated cognitive impairment and aging by TP53. This study, by integrating bioinformatics and machine learning methods, identified BCL6, TP53, HSP90AA1, CRYAB, IL1B, and DNAJB1 as potential diagnostic biomarkers for T2D with cognitive impairment and aging, with a particular emphasis on the significance of TP53 and IL1B in immune cell infiltration. These findings not only enhance our understanding of the molecular mechanisms linking type 2 diabetes to cognitive impairment and aging, providing new targets for early diagnosis and treatment, but also offer new directions and targets for basic research.
Collapse
Affiliation(s)
- Qin Wang
- Department of Geriatric integrative, Second Affiliated Hospital of Xinjiang Medical University, NO.38, South Lake East Road North Second Lane, Shuimogou District, Urumqi, 830063, Xinjiang, China
| | - Ye Yang
- Department of Geriatric integrative, Second Affiliated Hospital of Xinjiang Medical University, NO.38, South Lake East Road North Second Lane, Shuimogou District, Urumqi, 830063, Xinjiang, China.
| |
Collapse
|
46
|
Dove A, Wang J, Huang H, Dunk MM, Sakakibara S, Guitart-Masip M, Papenberg G, Xu W. Diabetes, Prediabetes, and Brain Aging: The Role of Healthy Lifestyle. Diabetes Care 2024; 47:1794-1802. [PMID: 39193914 PMCID: PMC11417282 DOI: 10.2337/dc24-0860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/08/2024] [Indexed: 08/29/2024]
Abstract
OBJECTIVE Diabetes is a well-known risk factor for dementia. We investigated the association between (pre)diabetes and older brain age and whether this can be attenuated by modifiable lifestyle behaviors. RESEARCH DESIGN AND METHODS The study included 31,229 dementia-free adults from the UK Biobank between the ages of 40 and 70 years. Glycemic status (normoglycemia, prediabetes, or diabetes) was ascertained based on medical history, medication use, and HbA1c measured at baseline. Information on cardiometabolic risk factors (obesity, hypertension, low HDL, and high triglycerides) and lifestyle behaviors (smoking, drinking, and physical activity) was also collected at baseline. Participants underwent up to two brain MRI scans over 11 years of follow-up. Brain age was estimated using a machine learning model based on 1,079 brain MRI phenotypes and used to calculate brain age gap (BAG; i.e., brain age minus chronological age). RESULTS At baseline, 13,518 participants (43.3%) had prediabetes and 1,149 (3.7%) had diabetes. Prediabetes (β = 0.22 [95% CI 0.10, 0.34]) and diabetes (2.01 [1.70, 2.32]) were both associated with significantly higher BAG, and diabetes was further associated with significant increase in BAG over time (0.27 [0.01, 0.53]). The association between (pre)diabetes and higher BAG was more pronounced in men and in people with two or more cardiometabolic risk factors. In joint exposure analysis, having a healthy lifestyle (i.e., no smoking, no heavy drinking, and high physical activity) significantly attenuated the diabetes-BAG association. CONCLUSIONS Diabetes and even prediabetes are associated with accelerated brain aging, especially among men and people with poor cardiometabolic health. However, a healthy lifestyle may counteract this.
Collapse
Affiliation(s)
- Abigail Dove
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Jiao Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
- Department of Epidemiology, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Huijie Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Michelle M. Dunk
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Sakura Sakakibara
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Marc Guitart-Masip
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Goran Papenberg
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Weili Xu
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| |
Collapse
|
47
|
Cheng X, Tan Y, Li H, Zhang Z, Hui S, Zhang Z, Peng W. Mechanistic Insights and Potential Therapeutic Implications of NRF2 in Diabetic Encephalopathy. Mol Neurobiol 2024; 61:8253-8278. [PMID: 38483656 DOI: 10.1007/s12035-024-04097-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/04/2024] [Indexed: 09/21/2024]
Abstract
Diabetic encephalopathy (DE) is a complication of diabetes, especially type 2 diabetes (T2D), characterized by damage in the central nervous system and cognitive impairment, which has gained global attention. Despite the extensive research aimed at enhancing our understanding of DE, the underlying mechanism of occurrence and development of DE has not been established. Mounting evidence has demonstrated a close correlation between DE and various factors, such as Alzheimer's disease-like pathological changes, insulin resistance, inflammation, and oxidative stress. Of interest, nuclear factor erythroid 2-related factor 2 (NRF2) is a transcription factor with antioxidant properties that is crucial in maintaining redox homeostasis and regulating inflammatory responses. The activation and regulatory mechanisms of NRF2 are a relatively complex process. NRF2 is involved in the regulation of multiple metabolic pathways and confers neuroprotective functions. Multiple studies have provided evidence demonstrating the significant involvement of NRF2 as a critical transcription factor in the progression of DE. Additionally, various molecules capable of activating NRF2 expression have shown potential in ameliorating DE. Therefore, it is intriguing to consider NRF2 as a potential target for the treatment of DE. In this review, we aim to shed light on the role and the possible underlying mechanism of NRF2 in DE. Furthermore, we provide an overview of the current research landscape and address the challenges associated with using NRF2 activators as potential treatment options for DE.
Collapse
Affiliation(s)
- Xin Cheng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, Hunan, 410011, People's Republic of China
- National Clinical Research Center for Mental Disorder, Changsha, 410011, China
| | - Yejun Tan
- School of Mathematics, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Hongli Li
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, Hunan, 410011, People's Republic of China
- National Clinical Research Center for Mental Disorder, Changsha, 410011, China
| | - Zhen Zhang
- YangSheng College of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Shan Hui
- Department of Geratology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, China
| | - Zheyu Zhang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, Hunan, 410011, People's Republic of China.
- National Clinical Research Center for Mental Disorder, Changsha, 410011, China.
| | - Weijun Peng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, Hunan, 410011, People's Republic of China.
- National Clinical Research Center for Mental Disorder, Changsha, 410011, China.
| |
Collapse
|
48
|
Liarakos AL, Lim JZM, Leelarathna L, Wilmot EG. The use of technology in type 2 diabetes and prediabetes: a narrative review. Diabetologia 2024; 67:2059-2074. [PMID: 38951212 PMCID: PMC11446986 DOI: 10.1007/s00125-024-06203-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/09/2024] [Indexed: 07/03/2024]
Abstract
The increasing incidence of type 2 diabetes, which represents 90% of diabetes cases globally, is a major public health concern. Improved glucose management reduces the risk of vascular complications and mortality; however, only a small proportion of the type 2 diabetes population have blood glucose levels within the recommended treatment targets. In recent years, diabetes technologies have revolutionised the care of people with type 1 diabetes, and it is becoming increasingly evident that people with type 2 diabetes can also benefit from these advances. In this review, we describe the current knowledge regarding the role of technologies for people living with type 2 diabetes and the evidence supporting their use in clinical practice. We conclude that continuous glucose monitoring systems deliver glycaemic benefits for individuals with type 2 diabetes, whether treated with insulin or non-insulin therapy; further data are required to evaluate the role of these systems in those with prediabetes (defined as impaired glucose tolerance and/or impaired fasting glucose and/or HbA1c levels between 39 mmol/mol [5.7%] and 47 mmol/mol [6.4%]). The use of insulin pumps seems to be safe and effective in people with type 2 diabetes, especially in those with an HbA1c significantly above target. Initial results from studies exploring the impact of closed-loop systems in type 2 diabetes are promising. We discuss directions for future research to fully understand the potential benefits of integrating evidence-based technology into care for people living with type 2 diabetes and prediabetes.
Collapse
Affiliation(s)
- Alexandros L Liarakos
- Department of Diabetes and Endocrinology, University Hospitals of Derby and Burton NHS Foundation Trust, Royal Derby Hospital, Derby, UK
- School of Medicine, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Jonathan Z M Lim
- Diabetes, Endocrinology and Metabolism Centre, Manchester University NHS Foundation Trust, Manchester Royal Infirmary, Manchester, UK
| | - Lalantha Leelarathna
- Diabetes, Endocrinology and Metabolism Centre, Manchester University NHS Foundation Trust, Manchester Royal Infirmary, Manchester, UK
- Department of Diabetes, Imperial College Healthcare NHS Trust, London, UK
- Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Emma G Wilmot
- Department of Diabetes and Endocrinology, University Hospitals of Derby and Burton NHS Foundation Trust, Royal Derby Hospital, Derby, UK.
- School of Medicine, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK.
| |
Collapse
|
49
|
Guan Z. Alterations in Neuronal Nicotinic Acetylcholine Receptors in the Pathogenesis of Various Cognitive Impairments. CNS Neurosci Ther 2024; 30:e70069. [PMID: 39370620 PMCID: PMC11456617 DOI: 10.1111/cns.70069] [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: 03/16/2024] [Revised: 09/02/2024] [Accepted: 09/15/2024] [Indexed: 10/08/2024] Open
Abstract
Cognitive impairment is a typical symptom of both neurodegenerative and certain other diseases. In connection with these different pathologies, the etiology and neurological and metabolic changes associated with cognitive impairment must differ. Until these characteristics and differences are understood in greater detail, pharmacological treatment of the different forms of cognitive impairment remains suboptimal. Neurotransmitter receptors, including neuronal nicotinic acetylcholine receptors (nAChRs), dopamine receptors, and glutamine receptors, play key roles in the functions and metabolisms of the brain. Among these, the role of nAChRs in the development of cognitive impairment has attracted more and more attention. The present review summarizes what is presently known concerning the structure, distribution, metabolism, and function of nAChRs, as well as their involvement in major cognitive disorders such as Alzheimer's disease, Parkinson's disease, vascular dementia, schizophrenia, and diabetes mellitus. As will be discussed, the relevant scientific literature reveals clearly that the α4β2 and α7 nAChR subtypes and/or subunits of the receptors play major roles in maintaining cognitive function and in neuroprotection of the brain. Accordingly, focusing on these as targets of drug therapy can be expected to lead to breakthroughs in the treatment of cognitive disorders such as AD and schizophrenia.
Collapse
Affiliation(s)
- Zhi‐Zhong Guan
- Department of PathologyThe Affiliated Hospital of Guizhou Medical UniversityGuiyangP.R. China
- Key Laboratory of Endemic and Ethnic DiseasesGuizhou Medical University, Ministry of Education and Provincial Key Laboratory of Medical Molecular BiologyGuiyangP.R. China
| |
Collapse
|
50
|
Shang Y, Wang S, Wei C, Xie H. Associations of Cognitive Impairment with All-Cause and Cardiovascular Mortality Among Individuals with Diabetes: A Prospective Cohort Study. J Appl Gerontol 2024; 43:1449-1460. [PMID: 38652679 DOI: 10.1177/07334648241241392] [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] [Indexed: 04/25/2024] Open
Abstract
This study explored the association between diabetes, cognitive imFpairment (CI), and mortality in a cohort of 2931 individuals aged 60 and above from the 2011 to 2014 NHANES. Mortality data was gathered through 2019, and multivariable Cox proportional hazards models were used to determine the association between diabetes, CI, and mortality adjusting for sociodemographic characteristics, lifestyle factors, and comorbidity conditions. The study spanned up to 9.17 years, observing 579 deaths, with individuals having both diabetes and CI showing the highest all-cause mortality (23.6 events per 100 patient-years). Adjusted analysis revealed a 2.34-fold higher risk of all-cause mortality for this group, surpassing those with diabetes or CI alone. These results held after a series of stratified and sensitivity analyses. In conclusion, CI was linked to higher all-cause mortality in individuals with diabetes, emphasizing the need to address cognitive dysfunction in diabetic patients.
Collapse
Affiliation(s)
- Yanchang Shang
- Department of Geriatric Neurology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Shuhui Wang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Chao Wei
- Department of Geriatric Neurology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Hengge Xie
- Department of Geriatric Neurology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|